draft nordic working papers - oecd nordic... · 168 ultimo 2018) & marie-louise holmer (until...

69
1 1 2 3 4 5 6 7 8 9 10 11 Draft NORDIC WORKING PAPERS The Role of Retinoids in Female and Male Reproduction Including a Scoping Effort to Identify Potential Methods for Regulatory Use This working paper has been published with financial support from the Nordic Council of Ministers. However, the contents of this working paper do not necessarily reflect the views, policies or recommendations of the Nordic Council of Ministers. Nordisk Council of Ministers Ved Stranden 18 1061 Copenhagen K www.norden.org 12

Upload: others

Post on 14-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

1

1 2 3 4 5 6 7 8 9 10 11

Draft NORDIC WORKING PAPERS

The Role of Retinoids in Female and

Male Reproduction Including a Scoping

Effort to Identify Potential Methods for

Regulatory Use

This working paper has been published with financial support from the

Nordic Council of Ministers. However, the contents of this working paper do

not necessarily reflect the views, policies or recommendations of the

Nordic Council of Ministers.

Nordisk Council of Ministers – Ved Stranden 18 – 1061 Copenhagen K –

www.norden.org

12

Page 2: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

1

13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35

The Role of Retinoids in Female and Male 36

Reproduction Including a Scoping Effort to Identify 37

Potential Methods for Regulatory Use38

Page 3: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

2

Table of contents 39

40 Table of contents ........................................................................................................................................ 2 41

1. Preface ................................................................................................................................................ 4 42

Financing and workforce ............................................................................................................................ 5 43

2. Abbreviation list .................................................................................................................................. 6 44

3. Summary ............................................................................................................................................. 7 45

4. Introduction to the retinoid system ................................................................................................... 8 46

History and general background ................................................................................................................. 8 47

Retinoid Storage, Transport and Metabolism. ........................................................................................... 9 48

Retinoid Receptors and gene regulation .................................................................................................. 11 49

Crosstalk between the retinoid signaling pathway and other endocrine signaling pathways ................. 13 50

Epigenetics and its role in the retinoid system ......................................................................................... 14 51

5. Formation of female and male reproductive organs, and the role of retinoids............................... 15 52

6. Germ cells, meiosis and the role of retinoids ................................................................................... 16 53

7. Female reproductive organ development, function and health and the role of retinoids .............. 19 54

Oogenesis .................................................................................................................................................. 19 55

Ovarian somatic cells and folliculogenesis ............................................................................................... 19 56

Ovarian steroidogenesis ........................................................................................................................... 21 57

Other female reproductive organs and pregnancy .................................................................................. 23 58

Female reproductive pathologies and retinoid involvement ................................................................... 24 59

Retinoid deficiency/excess in female reproduction ................................................................................. 25 60

Crosstalk with other signaling pathways .................................................................................................. 25 61

8. Male reproductive organ development, function and health, and the role of retinoids ................ 26 62

Testicular somatic cells and steroidogenesis ............................................................................................ 26 63

Spermatogenesis ...................................................................................................................................... 27 64

Secondary male reproductive organs ....................................................................................................... 31 65

Male reproductive pathologies ................................................................................................................ 31 66

Retinoid deficiency in male reproduction ................................................................................................ 32 67

9. Impact on female and male reproduction by chemicals acting via the retinoid system ................. 33 68

Chemicals affecting female reproduction................................................................................................. 35 69

Chemicals affecting male reproduction .................................................................................................... 36 70

10. Potential adverse outcome pathways in female and male reproduction ........................................ 39 71

Proposed AOP for in utero CYP26B1 inhibition effects on male fertility .................................................. 40 72

Proposed AOP for effects of RDH10/RALDH inhibition in the adult on male fertility .............................. 41 73

Proposed AOP for meiosis initiation, oogenesis, folliculogenesis and female fertility, focusing on Stra874 .................................................................................................................................................................. 42 75

Proposed AOP for endometriosis in the adult female, focusing on CYP26A1 .......................................... 43 76

11. Initial scoping effort: assays and endpoints for effects of chemicals on female and male 77

Page 4: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

3

reproduction via the retinoid system ....................................................................................................... 44 78

In silico methods ....................................................................................................................................... 44 79

In vitro assays and ex vivo methods ......................................................................................................... 44 80

In vivo endpoints ....................................................................................................................................... 45 81

12. Concluding remarks .......................................................................................................................... 50 82

13. References ........................................................................................................................................ 51 83

14. Appendix ........................................................................................................................................... 63 84

Assay methods and availability: ................................................................................................................ 63 85

PubMed search strategy – retinoid parameters, chemical names, female/male reproduction 86 parameters................................................................................................................................................ 64 87

88

89

Page 5: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

4

1. Preface 90

91 Terms of reference and Scope of report 92 This Nordic report represents part of the outcome of a project within the Organisation for Economic 93 Co-operation and Development (OECD) test-guideline (TG) program. This project was initiated by 94 Sweden (Helen Håkansson, Institute of Environmental Medicine, Karolinska Institutet, Sweden) and 95 coordinated by the Swedish Chemicals Agency under supervision of both a Steering committee from 96 the funding body (Nordic Co-ordination for the Development of Test Methods in Toxicology and 97 Ecotoxicology, Nord-UTTE) and by the OECD Endocrine Disrupters Testing and Assessment Advisory 98 Group. 99 100 This project builds on the ideas brought forward in the OECD Detailed Review Paper (DRP) 178 on 101 endocrine disrupter testing1 which addressed multiple aspects of the endocrine system, beyond 102 estrogens, androgens, thyroid disruption and steroidogenesis (EATS). The OECD DRP 178 suggests 103 projects in several areas, and particularly mentions assays relevant to the retinoid system, such as 104 Retinoid X Receptor (RXR) and Retinoic Acid Receptor (RAR)-reporter assays, Aryl hydrocarbon 105 Receptor (AhR) reporter assays, adipocyte differentiation, and retinoid serum levels. As identification 106 of retinoid system modulation is not included in any OECD test guideline it is urgent to cover this 107 identified knowledge gap. 108 109 The long-term aim was to develop methods that facilitate early screening, to enhance existing in vivo 110 test guidelines and to identify markers of biological effects for use in population studies, based on 111 information from retinoid biology and disturbed retinoid signalling in several organ systems. The EU 112 Commission funded development of a draft DRP on the retinoid system, that covered e.g. overall 113 biology of the retinoid system in human health and the environment, including a section on retinoic 114 acid and the reproductive system as well as a substantial annex on the retinoid system in male 115 reproduction2. However, it became apparent that the scope of the aim was too broad, and that for 116 certain aspects science was not mature enough for regulatory purposes. Consequently, the scope was 117 limited, and prioritisation was given to reproduction, as female reproduction had been identified as a 118 highly prioritized area in the recommendation from the EU Commission prioritisation workshop 20173. 119 In addition, it was decided that the annex on male reproduction should be further developed. Sweden, 120 as lead country, with support from all parties involved, decided to, as a first step of the DRP Retinoid 121 Process, publish this Nordic Working Paper. The present report will be followed up in the upcoming 122 Detailed Review Paper on retinoids, due to be published by the OECD in 2020. 123 124 Disposition 125 The report covers an overview of the retinoid system, the role of retinoids in the reproductive organs, 126 the impact of chemicals on reproduction via the retinoid system, potential adverse outcome pathways, 127 and an initial scoping effort for the possible role of chemical-induced retinoid disruption of the male 128 and female reproductive systems. In this report, the term reproduction refers to the formation and 129

1 OECD (2012), Detailed Review Paper on the State of the Science on Novel In Vitro and In Vivo Screening and Testing Methods and Endpoints for Evaluating Endocrine Disrupters, OECD Series on Testing and Assessment, No. 178, OECD Publishing, Paris, http://dx.doi.org/10.1787/9789264221352-en. 2 OECD (2017), “Detailed review paper on the retinoid system (draft)”, OECD, Paris. 3 Setting priorities for further development and validation of test methods and testing approaches for evaluating endocrine disruptors, in Brussels 2017 (Final Report European Commission Directorate-General for Environment Directorate B. (ISBN 978-92-79-83076-1), Available at https://publications.europa.eu/en/publication-detail/-/publication/6b464845-4833-11e8-be1d-01aa75ed71a1/language-en

Page 6: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

5

development of reproduction organs and their normal function. 130 131 Financing and workforce 132 The Nordic Working Group for Chemicals, Environment and Health (NKE) (in NKE Contracts 2017-003, 133 2018-026, 2019-003) and The Swedish Chemicals Agency provided the main funding for this work. 134 Furthermore, the European Commission (Framework Contract ENV.A.3/FRA/2014/0029) provided 135 funding for the draft DRP reported in 2017 (mainly written by Alice Baynes, Edwin Routledge, Sofie 136 Christiansen and Ulla Hass), parts of which has been incorporated in this report. 137 138 The main author of this Nordic report has been Dr. Charlotte Nilsson, RISE Research Institutes of 139 Sweden. The in-kind contributions from the scientific experts involved in writing and reviewing this 140 document is highly acknowledged (see list of contributors). 141 142 This draft report describes the status as of September 2019. The final report will be publicly available 143 via the NKE website4, and will describe the status as of December 2019. 144 145 146 List of contributors 147 148 EU Framework contract (2016-2017): 149 Alice Baynes & Edwin Routledge (Brunel University London); Sofie Christiansen & Ulla Hass (Technical 150 University of Denmark). 151 152 Nordic Working Paper (2018-2019): Miriam Jacobs (Public Health England); Norbert Ghyselinck & 153 Manuel Mark (Institute of Genetics and Molecular and Cellular Biology, France); Stefano Lorenzetti & 154 Alberto Mantovani (Instituto Superiore di Sanità, Italy); Taisen Iguchi (National Institute for Basic 155 Biology, Japan); Pauliina Damdimopoulou & Astrud Tuck (CLINTEC, Karolinska Institutet, Sweden); 156 Helen Håkansson (Institute of Environmental Medicine, Karolinska Institutet, Sweden); Anne-Lee 157 Gustafson & Margareta Halin Lejonklou (Swedish Chemicals Agency); Sofie Christiansen (Technical 158 University of Denmark). Nancy Baker (United States Environmental Protection Agency); Jodi Flaws 159 (University of Illinois, USA); Elise Grignard & Sharon Munn (Joint Research Center, DG Environment 160 European Commission); Peter Korytar (DG Environment European Commission); Anne Gourmelon & 161 Patience Browne (OECD Test Guidelines Programme, Environment Directorate, OECD). Contributions 162 from the OECD Retinoid EG will be added to this list. 163 164 165 Nord-UTTE steering group, subgroup NKG (Nordic Chemicals Group) (2017-2019): 166 Sjur Andersen (The Norwegian Environment Agency), Knud Ladegaard Pedersen, Henrik Tyle (until 167 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); 168 Sofie Christiansen (Technical University of Denmark); Matti Leppänen (The Finnish Environment 169 Institute); Petteri Talasniemi (The Finnish Safety and Chemicals Agency); Anne-Lee Gustafson (The 170 Swedish Chemicals Agency). 171

4 https://www.norden.org/en/organisation/nordic-working-group-chemicals-environment-and-health-nke

Page 7: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

6

2. Abbreviation list 172

173

174

Page 8: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

7

3. Summary 175

Page 9: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

8

4. Introduction to the retinoid system 176

177

178

History and general background 179 Retinoids5, a chemically related group of compounds which includes vitamin A, can exist in several 180 different forms (see Figure 1). As the name “vitamin” implies, they are essential micronutrients that 181 must be supplied from the diet, either in the form of carotenoids from vegetable sources, or retinol 182 and retinyl esters from animal sources (Al Tanoury et al. 2013). Carotenoids are responsible for the 183 bright red, yellow and orange colors of many vegetables. The liver, an organ found in all vertebrate 184 organisms, can be a rich source of retinoids and may have been used by the ancient Egyptians as a 185 cure for night blindness, which is a typical symptom of retinoid deficiency (Wolf 1996). Conversely, 186 Arctic cultures have long known to avoid eating polar bear liver, which can contain extremely high 187 levels of retinoids, to avoid adverse effects such as blurred vision, nausea, skin loss, coma and even 188 death (Rodahl and Moore 1943). Thus, history has shown that both hypovitaminosis A and 189 hypervitaminosis A can be detrimental. 190 191 The requirement for retinoids in normal physiological functions has been studied for more than a 192 century, by examining the effects of retinoid deficiency or excess in different species, and, more 193 recently, in genetically modified mice (reviewed in Clagett-Dame and Knutson 2011). Retinoids are 194 essential for vision, reproduction, embryonic development, adult growth and development, and 195 maintenance of immunity and epithelial barriers (O’Byrne and Blaner 2013). Vitamin A deficiency 196 (VAD) is the main cause of preventable blindness in the world (Bastos Maia et al. 2019), and it has 197 been hypothesized that hearing loss among humans in the developing world is due to gestational 198 VAD (Emmet and West 2014). Recently, new metabolic functions for retinoids have been discovered, 199 in e.g. lipid metabolism and insulin response (Napoli 2017, Cione et al. 2016). The importance of 200 retinoids and the retinoid signaling pathways is reflected in both the ancestry and the conservation 201 of genes and pathways among both vertebrates and invertebrates (André et al. 2014). 202 203 The most fat-soluble forms of retinoids (retinyl esters) can be stored within the body in relatively 204 high levels, which can counteract periods of low dietary retinoid dietary intake (O’Byrne and Blaner 205 2013). Despite this, retinoid deficiency is one of the most prevalent nutrient deficiencies worldwide 206 (Napoli 2017). 207 208

5 The term “retinoids” covers all analogues of vitamin A, naturally occurring as well as synthetic.

Page 10: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

9

209 210 211 212 213 214 215 216 217 Figure 1. Chemical structures of various forms of retinoid precursors (β-carotene), intermediate 218 transformation, transport form (all-trans-retinol; ROH), storage form (all-trans-retinyl ester), 219 intermediate forms also important in vision (11-cis-retinaldehyde, all-trans-retinaldehyde), and the 220 nuclear receptor-binding forms (all-trans-retinoic acid, 9-cis-retinoic acid, 13-cis-retinoic acid, 9,13-di-221 cis-retinoic acid). 222 223

Retinoid Storage, Transport and Metabolism. 224 225 Storage 226 The vast majority (approximately 80-85%) of retinoids found in the body is stored in the liver (O’Byrne 227 et al. 2005). Two main cell types have been identified as being central to metabolism and storage of 228 retinoids, namely hepatocytes and hepatic stellate cells (HSC), respectively. Hepatocytes contain 229 around 10-20% of hepatic Vitamin A and are not only involved in the initial uptake of Vitamin A, but 230 are also the site of RBP synthesis and secretion (Sauvant et al. 2001). Therefore, hepatocytes are 231 important in both the uptake and the mobilization of vitamin A into, and out of, the liver. HSC have 232 been found to contain the remainder (80-90%) of hepatic vitamin A (Blaner and Li 2015), and are 233 therefore considered the main vitamin A storage cells. Studies in mice, using radiolabeled retinyl 234 esters, have shown dietary vitamin A to be initially taken up by the hepatocytes and then rapidly 235 transferred to HSC (Blomhoff et al. 1982). 236 237 Another well-known retinoid storage organ is the eye. Retinoids has long been known to play a vital 238 role in vision and eye function. The retina and retinal pigmented epithelium (RPE) contain 11-cis and 239 all-trans-isomers of retinaldehyde, ROH, and RE (Palczewski 2012), with the RPE being the main site 240 of RE storage. In contrast, the photoreceptors of the retina contain large amounts of retinaldehyde. 241

13-cis-retinoic acid

9, 13-di-cis-retinoic acid

Page 11: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

10

RE stores are also found in a number of other organs/tissues, but at lower levels than the liver, 242 including the lung, brain, skin, muscle, kidney, spleen, white and brown adipose tissue, and testis 243 (e.g. Sertoli cells) (summarized in Blaner and Li, 2015). 244 245 Within the (mammalian) body, the most abundant forms of retinoids are ROH and RE (see figure 1). 246 RE are often found in the circulation after a meal, and are the main storage forms of retinoids in the 247 liver but can also be found in other tissues. ROH can also be found in the blood and is the main 248 circulating form of retinoids during fasting. ROH is generally bound to a number of chaperone 249 proteins, known collectively as retinol-binding proteins. Of these, retinol-binding protein (RBP) 250 transports ROH in the blood, and cellular retinol-binding protein-1 (CRBP1), CRBP2, and inter-251 photoreceptor retinoid-binding protein (IRBP) occur intracellularly (Blaner and Li 2015). Cellular 252 retinoic acid binding proteins (CRABPs) are involved in RA transport from the cytoplasm to the 253 nuclear retinoic acid receptor (RAR) and also play a role in determining intracellular RA levels by 254 controlling the amount of RA that is available for degradation (Napoli 2017). 255 256 A number of enzymes are required for the balance of storing and re-mobilizing of retinoids during 257 times of abundance and insufficiency, respectively. The most important enzymes are those 258 synthesizing RE (such as LRAT; lecithin:retinol acyltransferase) and those used to hydrolyze RE to ROH 259 (such as REH; retinyl ester hydrolase) (Figure 2). These enzymes, along with (i) the capacity to 260 accumulate RE in lipid droplets known as chylomicrons within cells and tissues and (ii) the ability to 261 re-mobilize these stores when needed, compensate for fluctuations in retinoid intake via the diet so 262 as not to negatively impact the individual. The process of remobilization of stored RE in to circulating 263 ROH seems reliant on interactions with RBP, as evidenced by studies using RBP-deficient mice 264 (Quadro et al. 1999). 265 266 Transport 267 A range of retinoid forms are found in fasting and/or non-fasting states. These include RE in 268 chylomicrons and chylomicron remnants (fat droplets exported to blood and lymph from epithelial 269 cells of the small intestine); RE in very low density lipoproteins (VLDL), low density lipoprotein (LDL) 270 and high density lipoprotein (HDL); ROH bound to RBP4 (serum retinol-binding protein type 4); RA 271 bound to albumin; and water-soluble beta-glucuronide conjugates of ROH and RA. The delivery of 272 retinoids to tissues involves many different vitamin A forms and carriers. However, ROH bound to 273 RBP4 and chylomicron RE are considered the most important (largest volume) transport pathways 274 (Laudet et al. 2015). 275 276 It is worth noting that retinoid delivery to tissues works in a similar way to vitamin D and thyroid 277 hormone (Blaner and Li 2015), by transport of large quantities of the biologically-inactive form (i.e. 278 ROH, 25-hydroxy-vitamin D, or thyroid hormone T4), and relatively low quantities of the biologically-279 active form (RA, 1,25- dihydroxy-vitamin D, or thyroid hormone T3) in the blood (Blaner and Li 2015). 280 ROH, bound to RBP4, forms a larger complex the T4 transporting protein transthyretin (TTR) in the 281 blood (Figure 2). Both TTR and RBP4 are mainly synthesized in the liver and choroid plexus, and 282 secreted into the plasma by the liver and cerebrospinal fluid, respectively (Shirakami et al. 2012). In 283 contrast, there is no specific binding protein for the transport of RA in the plasma, as it is synthesized 284 locally in target cells by SDR, ADH, RALDH enzymes (Figure 2). 285 286 Metabolism 287 RA is considered to be the most biologically important member of the retinoid family for nuclear 288 signaling (see retinoid receptor section below). However, as stated above, the most abundant forms 289 of internal circulating vitamin A are ROH and RE. Therefore, within target cells, a number of 290 conversion steps are required to transform relatively inactive forms of retinoids into the biologically 291 active RA form (reviewed in Shannon et al. 2017). Once ROH-RBP4 has been bound to STRA6 and 292 taken up into a target cell, ROH is converted to RA via a two-step oxidation process (Figure 2). The 293 first step, in which ROH is converted into retinaldehyde, is reversible and is carried out either by a 294

Page 12: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

11

short-chain dehydrogenase/reductase (SDR; a complex consisting of retinol dehydrogenase 10 295 [RDH10], and retinaldehyde reductase 3 [DHRS3]) or by cytosolic alcohol dehydrogenases (ADH) 296 (Shannon et al 2017). The majority of ROH is, in normal physiological conditions, bound to CRBP and 297 directed towards SDR rather than the more unspecific ADH (Napoli 2017). 298 299 The retinaldehyde molecule can undergo the second, irreversible, step and be further oxidized into 300 RA by retinaldehyde dehydrogenases RALDH 1, 2 or 3; (also known as aldehyde dehydrogenases 301 ALDH1A1 - ALDH1A3 (Shannon et al 2017). However, retinaldehyde can also be reduced back to ROH 302 by SDR, or, under VAD conditions, by retinol dehydrogenase 11 (RDH11) (Belyaeva et al. 2018). The 303 ratio of CRBP-ROH/retinaldehyde (holo-CRBP) to CRBP without ROH/retinaldehyde (apo-CRBP) 304 appears to signal whether retinaldehyde should be reduced back to ROH or further oxidized to RA 305 (Napoli et al. 2017). The fate of the retinaldehyde also depends on a negative feedback response to 306 available RA (Shannon et al. 2017). 307 308 RA is eventually catabolized, mainly by cytochromes P450 hydroxylases 26A1, B1 and C1 (Topletz et 309 al. 2015). A large number of other cytochrome p450 enzymes have also been shown to degrade RA in 310 vitro, although the relevance of these different CYP enzymes to normal retinoid homeostasis in vivo 311 is unclear (Laudet et al. 2015). In the adult human body, CYP26A1 is mainly expressed in the liver and 312 is responsible for more than 90% of the hepatic clearance of RA (Thatcher et al. 2010). CYP26A1 is 313 also expressed in e.g. testis, epididymis, uterus, endometrium and placenta, while CYP26B1-314 expression is more dispersed and found e.g. in the brain, testes, placenta, ovaries, endometrium. 315 (Human Protein Atlas6). In the human fetus, CYP26A1 is the form predominantly expressed in the 316 brain, whereas CYP26B1 is expressed in all tissues except the brain (Kedishvili 2013). In humans, 317 CYP26C1 is mainly expressed during embryo development, but can be expressed at low levels in 318 adult tissues, e.g. in testis (Ross and Zolfaghari 2011). 319 320 While levels of RA in tissues are governed mainly by a balance of production and metabolic 321 breakdown with RA feedback loops (Kedishvili 2013, Teletin et al. 2017). Some tissue-specific uptake 322 of RA from serum appears to take place, via a, to our knowledge, still not established mechanism 323 (Kurlandsky et al. 1995, O’Bryan and Blaner 2013). In the Kurlandsky study, performed only with male 324 rats, the contribution of serum RA to the tissue pool of RA was 0.7% for the testis, 10% for the 325 epididymis and 23% for the seminal vesicles. 326 327 The physiological role of RA in cell differentiation has led to its use in cancer treatment, including 328 reproductive organ malignancies (Siddikuzzaman et al. 2011). The treatment increases the normal 329 serum nM levels of RA up to µM levels, which over time leads to autoinduction of RA clearance via 330 upregulation of CYP26 enzymes (Jing et al. 2017). Attempts have been made to counteract therapy 331 resistance by developing e.g. CYP26 inhibitors (Nelson et al. 2013). However, side effects and low 332 potency has limited the use of these inhibitors (Jing et al. 2017). Similar efforts to maintain RA 333 homeostasis has also been observed in models of teratogenicity, and it has been speculated that the 334 teratogenic effects of RA are actually due to a prolonged local RA deficiency, caused by differential 335 induction of genes coding for enzymes producing (Raldh1-3) or breaking down (Cyp26a1 and 336 Cyp26b1) RA (Lee et al. 2012). Interestingly, maternal supplementation with low doses of RA 337 following a teratogenic insult has been shown to rescue or abrogate teratogenic effects (Lee et al. 338 2012). 339 340

Retinoid Receptors and gene regulation 341 RA acts mainly transcriptionally via specific nuclear retinoic acid receptors, RARs, which can form 342 homodimers with other RAR isomers, or heterodimers with retinoid X receptors, RXRs to influence a 343 variety of cellular processes (Germain et al. 2006). The receptor dimers bind to, and activate, the 344 retinoic acid response element (RARE), inducing transcription of the target gene in the presence of 345

6 https://www.proteinatlas.org/ accessed in May 2019

Page 13: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

12

the ligand. RARs can be activated both by RA and 9-cis-RA (Allenby et al. 1993). The endogenous. The 346 endogenous ligand for RXRs appears to be either 9,13-di-cis-RA or 9-cis-RA, formed via isomerization 347 from RA (de Lera et al. 2016). In addition, RARs have a vital role without activation by retinoids. The 348 RAR_RXR heterodimer can bind to DNA in the absence of a ligand, leading to the recruitment of a 349 corepressor complex that suppress transcription (Vilhais-Neto and Pourquié 2008). 350 351

RARs and RXRs belong to the same nuclear hormone receptor family as steroid hormones, thyroid 352 hormone and vitamin D receptors, as well as various orphan receptors and receptors activated by 353 intermediary metabolites (e.g. PPAR; peroxisome proliferator-activated receptor, LXR; liver X 354 receptor, FXR; farnesoid X receptor and PXR; pregnane X receptor) (Szanto et al. 2004). Some of 355 these nuclear receptors may also be involved in retinoid signaling responses. For example, PPARs, α, 356 β/δ and γ heterodimerize with RXR and function as transcription factors (Mangelsdorf and Evans 357 1995). RA can also serve as a ligand for PPAR β/δ, but at a much lower affinity than RAR (Al Tanoury 358 et al. 2013). 359 360 Three of retinoid receptors (RARα, RXRα and RXRβ) have widespread expression patterns, whereas 361 the others (RARβ, RARγ and RXRγ) show a more complex, tissue-specific expression. Therefore, 362 most tissues are potential targets of retinoid signalling, although different heterodimeric 363 complexes can transduce the RA signal (Rhinn and Dollé 2012). 364

In addition to the classical genomic effects, retinoic acid has been found to have a number of non- 365 genomic mechanisms such as kinase activation (Rochette-Egly 2015). 366 367

Page 14: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

13

368 369 Figure 2: Retinoid metabolism, transport and nuclear signaling. Abbreviations: alcohol 370 dehydrogenase (ADH), β-carotene (βcar), cellular retinoic acid-binding protein (CRABP), cellular 371 retinol-binding protein (CRBP), cytochrome P450 subunit 26 protein (CYP26), lecithin:retinol 372 acyltransferase (LRAT), retinoic acid (RA), retinaldehyde dehydrogenase (RALDH), retinoic acid 373 receptor (RAR), retinoic acid response element (RARE), retinol binding protein (RBP), retinyl esters 374 (RE), retinyl ester hydrolase (REH), retinol (ROH), retinoid X receptor (RXR), short-chain 375 dehydrogenase/reductase (SDR; an enzymatic complex consisting of retinol dehydrogenase RDH10 376 and retinaldehyde reductase DHRS3), stimulated by retinoic acid protein 6 (STRA6), transthyretin 377 (TTR). 378 379 380

Crosstalk between the retinoid signaling pathway and other endocrine signaling pathways 381 RXR can heterodimerize with other steroid and nuclear receptors, and functions essentially as a 382 master switch between different signaling pathways. Consequently, the retinoid signaling pathway 383 can crosstalk with other signaling pathways: in addition to the RARs, the heterodimerization partners 384 of RXRs include PPARs, TR, and VDR; the vitamin D receptor (Chambon 1996). In addition, crosstalk 385 with androgenic signaling (Long et al. 2019), as well as with xenobiotica-related receptor pathways, 386 via e.g. CAR; the constitutive androstane receptor, PXR, and AhR; the aryl hydrocarbon receptor has 387 been demonstrated. Possible responses to toxic insult include induction of xenobiotica-metabolizing 388 enzymes (Murphy et al. 2007, Shmarakov et al. 2019). 389 390 Crosstalk has also been shown on the level of regulating the expression of e.g. RA-catabolizing 391 enzymes in the CYP26 family, it has been demonstrated that in human liver cells, PPARγ agonists 392 rosiglitazone and pioglitazone induce CYP26A1 as well as the normally less abundant CYP26B1 (Tay 393 et al. 2010). The (non-endocrine) sonic hedgehog (SHH) pathway is reported to regulate expression 394 of the Cyp26a1 and Cyp26b1 genes, thereby preventing excessive RA levels during mouse embryo 395 development (El Shahawy et al. 2019). In the testicular Leydig cells, crosstalk between the retinoid 396

Page 15: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

14

signaling system and testosterone signaling has been shown to be crucial for steroidogenic cell 397 function (Jauregui et al. 2018). 398 399

Epigenetics and its role in the retinoid system 400 Epigenetic changes are changes in gene expression that a) do not involve gene sequence alterations 401 and b) may persist after the initial trigger is long gone (reviewed in e.g Villota-Salazar et al. 2016, and 402 Jacobs et al. 2017). Examples of epigenetic changes are histone modification and DNA methylation. 403 Histone deacetylation/methylation and DNA methylation are usually associated with repression of 404 gene transcription, while histone acetylation/demethylation and lack of DNA methylation instead 405 leads to transcriptional activation. 406 407 RA is known to mediate cell differentiation also via epigenetic mechanisms (reviewed in Urvalek et al. 408 2014). Epigenetic changes are also involved in, e.g., the process of transient induction of the Cyp26a1 409 gene by RA (Yuan et al. 2012). In some cases, epigenetic actions of RARs appear to be independent of 410 the ligand, i.e., RA (Laursen et al. 2012). 411 412

413

Page 16: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

15

5. Formation of female and male reproductive organs, and the role of 414

retinoids 415

416 The formation of the female and male reproductive organs is initiated very early in embryogenesis. 417 The genital ridges (the primordial gonads) are formed approximately halfway through gestation in 418 mice (Spiller et al. 2017) and during the first trimester in humans (Johansson et al. 2017). In mice, the 419 genital ridges are formed at 10.5 days post coitum (dpc), and soon begin to differentiate as testes, in 420 the presence of the Y-chromosomal gene Sry, or as ovaries, in the absence of Sry. The transcription 421 factor SRY upregulates the expression of the gene Sox9, which in turn governs the expression of 422 several male-specific genes, leading to differentiation of the bipotential somatic cell precursors into 423 Sertoli cells (Kashimada et al. 2011, Rossitto et al. 2015). In the absence of Sry, as in females, a 424 different set of genes are expressed by default, such as Wnt4 and Foxl2, leading to differentiation of 425 somatic precursor cells into female granulosa cells and subsequently to ovarian development 426 (Kashimada et al. 2011). 427 428 Degradation of endogenous RA appears to be critical for normal testis development, based on 429 observations of ovotestes and a feminized reproductive tract in male Cyp26b1-null mouse embryos, 430 which died at embryonic day (ED) 14.5-15.5 (Bowles et al. 2018). However, in an earlier study using 431 Cyp26b1-null mouse on a different genetic background, testis formation and somatic cell 432 differentiation in neonatal pups appeared essentially normal, although the testis were smaller 433 (MacLean et al. 2007). In an ex vivo model using rat fetal testis, exogenous RA could disrupt the 434 proper formation of seminiferous cords, as well as the maintenance of the testicular cell fate of the 435 somatic cells; following RA exposure, expression of ovarian-specific marker FOXL2 was observed 436 (Spade et al. 2018a). 437 438 Even in adulthood, the sexual fate of the male gonads must be maintained, probably via the active 439 presence of the transcription factor DMRT17 (Minkina et al. 2014). The role of DMRT1 in the testes 440 appears to be to prevent RA-initiated activation of specific potential feminizing genes (Foxl2 and 441 Esr2) in the Sertoli cells; without DMRT1 present, male-to-female trans-differentiation of somatic 442 cells was observed in the pre- and postnatal mouse testes (Minkina et al. 2014). 443 444 In contrast, in the female mouse embryo it seems like RA signaling is not required for correct ovarian 445 development, based on observations in mice with deletions/disruptions of either RARs or RA-446 synthesizing enzymes (Minkina et al. 2017), although earlier studies have shown that RA was 447 required in order to maintain ovarian differentiation and development in the developing fetal female 448 murine gonads (Minkina et al. 2014, Suzuki et al. 2015). Additional studies suggest that abnormal 449 endogenous RA levels may be able influence somatic cell differentiation and/or function (Bowles et 450 al. 2018). 451

452

7 DMRT1: Doublesex and mab-3 related transcription factor 1

Page 17: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

16

6. Germ cells, meiosis and the role of retinoids 453

454 In both male and female mouse embryos, a cluster of pluripotent primordial germ cell (PGC) 455 precursors arise starting 6.25 dpc at the posterior primitive streak of the embryo. Subsequently, they 456 migrate to the genital ridges (formed at 10.5 dpc), which will differentiate into testis or ovaries (see 457 section 5). 458 459 In both males and females, the haploid gametes are produced from germ cells via meiosis in the 460 gonads, following a similar process of events and requiring expression of many of the same genes. 461 However, the timing of these events is completely different. Around the same time as PGCs in fetal 462 ovaries enter prophase of the first meiotic division (ca 13 dpc in mice), PGCs in a testicular 463 environment enter quiescence; they slow down their proliferation towards mitotic arrest as GO/G1-464 arrested gonocytes (Kashimada et al. 2011). Thus, in the fetal gonad the commitment of germ cells 465 towards oogenesis involves entry into meiosis, whereas commitment to spermatogenesis involves 466 the inhibition of meiotic initiation, suppression of pluripotency and mitotic arrest. This mitotic arrest 467 is maintained until after birth (Spiller and Bowles 2015, Spiller et al. 2017) and consequently meiosis 468 initiation occurs postnatally in males. Thus, all oocytes are produced before birth, while 469 sperm(atocytes) are produced continuously during post-pubertal life in males. 470 471 Entrance into meiosis seems to be regulated by two master genes: Stra8 (“Stimulated by retinoic 472 acid, gene 8”) required for the initiation of meiosis in female fetal germ cells and Nanos2 (expressed 473 in male fetal germ cells), required to prevent Stra8 expression and meiosis initiation in male fetal 474 germ cells (Rossitto et al. 2015). Other genes, such as Fgf9, have also been implicated as being of 475 importance for proper control of meiotic entry; figure 3 is an attempt to summarize this information. 476 The Stra8 gene contains putative retinoid acid response elements (RAREs) and its expression is 477 activated by RA (Spiller et al. 2017). The Stra8 locus in non-germ cells appears to be epigenetically 478 silenced, thus, only pre-meiotic germ cells respond to RA by Stra8 induction (Spiller and Bowles 479 2015). The critical role of Stra8 is evident in Stra8-/- mice, as both females and males are infertile, 480 while heterozygotes of both sexes are fertile (Baltus et al. 2006). Stra8-/- females display smaller 481 ovaries lacking oocytes and follicles, and in the corresponding males, testes are smaller and testicular 482 germ cells severely reduced in numbers; in addition, spermatocytes undergo apoptosis before 483 reaching the leptotene and zygotene stages of meiotic prophase (Baltus et al. 2006). 484 485 Another meiosis-essential gene, the meiotic recombination component gene Rec8, is also an RA 486 target and is activated independently of Stra8: Rec8 encodes a meiosis-specific component of the 487 cohesion complex and is required for several steps in meiotic chromosomal activities (e.g. chromatid 488 cohesion and chiasmata formation) (Kuobova et al. 2014). Neither female nor male Rec8-/- mice that 489 survive to reach sexual maturity are fertile (Xu et al. 2005). 490 491

Page 18: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

17

492 493 494 Figure 3: Comparison of male and female germ cell development and meiotic progression in mice (in 495 the draft DRP, re-drawn from Dollé and Niederrether 20158). Blue indicates male-specific events, 496 red/pink indicated female-specific events and grey, events shared by both sexes. Numbers indicate 497 days post coitum (dpc). Arrows and bars indicate rough timing of gene expression, solid arrows 498 indicate direct effect, and broken arrows indicate an incompletely characterized or likely indirect 499 effect. Plus (+) or minus (-) indicate supportive/additive or inhibitory action on other genes. GC; germ 500 cell, PGC; Primordial Germ Cells, RA; retinoic acid, mitotic pro; mitotic proliferation. Summary figure 501 in the draft DRP, adapted from Clagett-Dame and Knutson 2011, Feng et al. 2014, Rossitto et al. 2015 502 and Spiller and Bowles 2015. 503 504 Retinoic acid has been implicated in controlling the onset of meiosis. The RA necessary for meiosis 505 initiation is available locally in both the fetal testis and fetal ovaries. Several sources for RA have 506 been suggested. It can diffuse into the primordial gonads from the nearby mesonephros, where, in 507 mice, retinol dehydrogenase 10 (Rdh10) and retinaldehydrogenase 2 (Raldh2) as well as Raldh3, at a 508 lower level, are expressed (Niederreither, Vermot et al. 2002, Niederreither, Fraulob et al. 2002, 509 Bowles et al. 2006, Spiller and Bowles 2015, Bowles et al. 2018). An alternative hypothesis is that RA 510 is produced by the coelomic epithelium, which covers the embryonic gonad and where RALDH2 is 511 also expressed (Niederreither et al. 1997, Teletin et al. 2017). RA-producing (retin)aldehyde 512 dehydrogenases are also present in both female and male fetal gonads in humans (Childs et al. 2011, 513 Le Bouffant et al. 2010). 514 515 The crucial difference between female and males is that the RA-catabolizing enzyme CYP26B1, which 516 is expressed in mouse fetal gonads of both sexes at 11.5 dpc, is no longer expressed in the female 517 gonad from 12.5 dpc (Bowles et al. 2006). RA would thus be degraded in the fetal testis, and thereby 518 meiotic entry is blocked (MacLean et al. 2007). In contrast, in the fetal ovary RA levels would be 519

8 Dollé P, Neiderreither K. The Retinoids: Biology, Biochemistry, and Disease. Wiley-Blackwell; 2015.

Page 19: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

18

maintained, and Stra8 and Rec8 expressed, and subsequently female gonocytes would enter into 520 meiosis I (Rossitto et al. 2015). 521 522 The importance of sufficient RA levels in the fetal ovary is implicated in a retinoid-deficient rat 523 model, where meiosis initiation was prevented (Li and Clagett-Dame 2009). As discussed in the 524 previous paragraph, RA levels are maintained in both male and female gonads alike in the absence of 525 CYP26B1. In a Cyp26b1-/- mouse model, germ cell development was studied in ovaries and testes 526 from embryos at 13.5 dpc and from neonatal pups (MacLean et al. 2007). In that study, ovarian germ 527 cells at either stage appeared to be unaffected by the lack of CYP26B1; in the embryonic Cyp26b1-/- 528 testes, some germ cells had prematurely entered meiosis while others appeared apoptotic. Increased 529 levels of RA were demonstrated in the embryonic Cyp26b1-/- testes. Germ cells were essentially 530 absent in testes from neonatal Cyp26b1-/- pups. Premature germ cell meiosis could also be observed 531 in male wildtype genital ridges cultured in the presence of a synthetic retinoid (Am580, resistant to 532 CYP26B1 metabolism), suggesting that excess RA was responsible for the effect (MacLean et al. 533 2007). It has also been shown in vitro that embryonic testes grown in the presence of RA express 534 Stra8 (Koubova et al. 2006). 535 536 The role of RA in the induction of meiosis may be facilitating (Kumar et al. 2011, Teletin et al. 2019) 537 rather than critical (Bowles et al. 2016, Spiller et al. 2017, Teletin et al. 2017). Kumar and coworkers 538 (Kumar et al. 2011) reported that meiosis occurred normally in mouse fetal ovary lacking RA due to 539 RALDH2 and RALDH3 ablations. In the post-natal male mouse, Teletin and coworkers (Teletin et al. 540 2019) showed that meiosis is not blocked, only slightly delayed, in mice lacking all RALDH isomers in 541 the seminiferous epitelium (SE) of the testis. 542

543

Page 20: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

19

7. Female reproductive organ development, function and health and the 544

role of retinoids 545

546 The ovary is the site for differentiation and release of mature oocytes for fertilization, but it is also 547 where sex hormones (necessary for follicle development, estrous cyclicity, maintenance of the 548 reproductive tract and its function) are synthesized and then released (Barnett et al. 2006). 549 550 Retinoids are now known to play an important role in the reproductive organs of females, especially 551 in sex determination (described in the previous section) and oogenesis/folliculogenesis during 552 embryogenesis but probably also in adult pathologies such as endometriosis. 553 554

Oogenesis 555 Oogenesis is the formation of haploid mature gametes from diploid PGCs, and the entire process 556 begins in utero with primary oocytes eventually arresting at prophase I (the first phase in meiosis) in 557 primordial follicles, which then remain inactive during childhood (Barnett et al. 2006, Teletin et al. 558 2017). In more detail, PGCs differentiate into oogonia upon arrival to the developing female gonad 559 and then undergo mitotic proliferation to create a stock of millions of oogonia that form germ cell 560 nests/cysts, i.e., clusters of cells connected by intercellular bridges (reviewed in Johansson et al. 561 2017). These nests break down when some of the oogonia within the nest undergo apoptosis, thus 562 breaking the connections between the cells and thereby allowing somatic cells to enter and surround 563 the germ cells, thus creating primordial follicles each containing a primary oocyte arrested in 564 prophase I. The breakdown of germ cell nests, constituting the beginning of primordial follicle 565 formation (initiating folliculogenesis, see next section), results in an oocyte number decrease. The 566 oogonia will enter meiosis I around E13.5 in mice and between 10 and 12 weeks of gestation in 567 humans (Grive and Freiman 2015). By entering meiosis I, there is a loss of pluripotency and the cells 568 can no longer divide mitotically. Thus, when all oogonia have entered the first meiotic division, no 569 further female germ cells can be produced, and the size of the ovarian reserve is fixed. 570 571 As already mentioned in the earlier section on sex determination, RA is widely accepted to play an 572 important (Bowles et al. 2016, Spiller et al. 2017, Teletin et al. 2017), although not critical (Kumar et 573 al. 2011, Teletin et al. 2019) role in the induction of meiosis. RA is believed to exerts its role by 574 activating expression of Stra8 (Spiller et al. 2017, Baltus et al. 2006) and possibly also Rec8 (Xu et al. 575 2005, Kuobova et al. 2014). However, meiosis-initiation via Stra8 induction reportedly occurs also in 576 a fetal ovary devoid of RA, as observed in Raldh2-/- mice (Kumar et al. 2011). 577 578

Ovarian somatic cells and folliculogenesis 579 As the genital ridges begin to differentiate as ovaries in female fetuses (at approx. 11 dpc in mice), 580 the gonadal somatic cells will differentiate into granulosa cells, after which meiosis will commence in 581 the oogonia (Minkina et al. 2017). The germ cell nests with the proliferating oogonia will break down 582 and somatic cells will enter and surround the germ cells, thereby forming the primordial follicles. The 583 processes of nest breakdown, germ cell death and formation of primordial follicles are similar in mice 584 and humans, but more sequentially organized in mice (Johansson et al. 2017). Nest breakdown takes 585 place around the time of birth in mice and begins during mid-gestation (around 16 weeks) in humans 586 (Grive and Freiman 2015). In addition, follicle assembly takes place prenatally in humans and 587 postnatally in rodents (Johansson et al. 2017). These differences may suggest different initiating 588 mechanisms. The primordial follicles, each of which contains an oocyte surrounded by a single layer 589 of somatic pre-granulosa cells, represent the ovarian reserve that must last for the entire 590 reproductive life in both mice and women (Grive and Freiman 2015). 591 592 Folliculogenesis, the process of follicle maturation, primarily takes place from the commencement of 593 puberty (Johansson et al. 2017). The first (primordial) follicles each consist of a single oocyte 594 surrounded by a layer of flat granulosa cells (GC). GCs progressively form several layers around the 595

Page 21: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

20

oocyte with an outer layer of androgen-producing theca cells. The GCs subsequently convert 596 androgens to estrogens. As GCs continue to proliferate, the antrum is formed. At this stage, selection 597 occurs between growing follicles, so that only one or a limited number of follicles continue growing 598 to the preovulatory stage whereas others undergo atresia. After ovulation, luteinized theca cells and 599 mural GCs produce progesterone. 600 601 602

603 604 Figure 4: Main steps of folliculogenesis (adapted from Georges et al. 2014). Folliculogenesis starts 605 with primordial follicles that are subsequently recruited to become primary follicles. Proliferating 606 granulosa cells (GC) progressively form several layers around the oocyte (secondary to late preantral 607 follicles). From this stage, a layer of theca cells surrounds the follicle, and the follicles start to 608 produce estrogens. Theca cells produce androgens, which are converted into estrogens in granulosa 609 cells. Whereas follicular growth to the secondary/preantral stage is independent of gonadotropins, 610 progression beyond this stage strictly depends on follicle stimulating hormone (FSH) stimulation. As 611 GC continue to proliferate, the antrum is formed and separates granulosa cells into two general 612 populations: cumulus and mural GCs. At this stage, selection occurs between growing follicles, and 613 only one or a limited number of follicles continue growing to the preovulatory stage whereas others 614 undergo atresia. Two layers of theca (theca interna and externa) can be distinguished around 615 selected follicles. After ovulation, which is triggered by a peak of FSH and luteinizing hormone (LH), 616 theca cells and mural granulosa cells luteinize to produce progesterone. 617 618 For several years, RA was believed to be required in order to maintain ovarian differentiation and 619 development in the developing fetal female gonads (Minkina et al. 2014, Suzuki et al. 2015). RA is 620 also believed to regulate mouse ovarian follicle development in the adult, and it has, e.g., been 621 studied studied in vitro by stimulating granulosa cell proliferation (Demczuk et al. 2016). Ex vivo, 622 using cat ovarian cortices, RA activates the development of primordial follicles into primary and 623 secondary follicles, possibly via differential MMP regulation (Fujihara et al. 2018). It has been shown 624 that murine theca cells and granulosa cells both express the enzymes necessary for conversion of 625

Page 22: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

21

retinol to retinoic acid, which consist of different forms of ADH and ALDH, and that cyp26B1 is 626 expressed in both theca and granulosa cells, but is differentially regulated after injection of equine 627 chorionic gonadotropin (eCG), an LH/FSH analogue (Kawai et al. 2016, Kawai et al. 2018). Also 628 demonstrated in vitro, ovarian de novo synthesis of RA is required for the follicular expression of the 629 LH receptor in granulosa cells of mouse ovaries and their ability to respond to the ovulatory LH surge; 630 oocytes appear to negatively regulate RA synthesis in pre-ovulatory follicles, impacting the LH 631 receptor expression in follicular somatic cells possibly via an epigenetic mechanism (Kawai et al. 632 2016, Kawai et al. 2018). Cultured human ovarian cumulus granulosa cells (obtained during oocyte 633 retrieval during the course of in vitro fertilization) produce RA from retinol in the media. RA causes 634 de-phosphorylation of connexin 43, involved in the gap junction cellular communication (GJIC) 635 between the granulosa cells in the cumulus-oocyte complex. De-phosphorylation of connexin 43 636 increases GJIC, which plays an important role in oogenesis and successful fertilization (Best et al. 637 2015). 638 639 However, at present, RA appears not to be required for ovarian granulosa cell specification, 640 differentiation or function (Minkina et al. 2017). In genetic mouse models, where all three RARs were 641 deleted in the XX somatic gonad at the time of sex determination, no significant effects were found 642 on ovarian differentiation, follicle development or female fertility. Nor did the knockout of all three 643 RA-producing aldehyde dehydrogenases (Aldh1a1-3) at the same timepoint appear to masculinize 644 the mouse ovaries. Additionally, an RAR antagonist (BMS-189453) was capable of blocking meiotic 645 initiation in the germ cells of 10.5 dpc XX gonads (from wild-type mice) in vitro but had little effect on 646 the expression of markers for either granulosa or Sertoli cells; thus, disruption of RA signaling does 647 not appear to disrupt early somatic differentiation (Minkina et al. 2017). 648 649 In murine ovaries, RBP4 is expressed before puberty but increases significantly in the peripubertal 650 period. In adult mice, RBP4 expression increased at pro-estrous and peaked at estrous and was 651 localized mainly in the granulosa and theca cells of follicles. Expression was also induced by FSH, 652 alone or in combination with LH; LH alone had no effect (Jiang et al. 2018b). 653 654

Ovarian steroidogenesis 655 Ovarian steroidogenesis, or the production of sex steroid hormones, is regulated by the pituitary 656 hormones LH and FSH; LH stimulates the ovarian thecal cells to produce androgens and FSH 657 stimulates the granulosa cells to convert these androgens to estrogens (Hannon and Flaws 2015). 658 The actual synthetic process (starting with cholesterol in ovarian follicles) is complex and requires the 659 action of several enzymes (see figure 5). 660 661 662

Page 23: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

22

663 Figure 5: Ovarian steroidogenesis is primarily conducted by the mature antral follicle and the corpus 664 luteum following ovulation. This process requires both the theca cells and granulosa cells and 665 involves the enzymatic conversion of cholesterol to 17β-estradiol and other necessary sex steroid 666 hormones. Hormones: listed in the white text boxes; steroidogenic enzymes: listed adjacent to the 667 arrows (adapted/copied from Hannon and Flaws 2015). 668 669 Ovarian steroidogenesis is reported to be regulated by RA in several systems (recently reviewed in 670 Damdimopoulou et al. 2019). For example, in rats, low levels of RA and retinol stimulate the 671 formation of progesterone (Bagavandoss and Midgley 1987). In a human ovarian surface epithelium 672 cell line, RA significantly induces production of progesterone (Papacleovoulou et al. 2009). In women, 673 retinol in plasma were associated with higher estradiol and testosterone levels (Mumford et al. 674 2016). It is likely that RA regulates either the expression or the activity (or both) of the steroidogenic 675 enzymes (Wickenheisser et al. 2005), a hypothesis that is supported by data from retinoid-deficient 676 rats (Jayaram et al. 1973). In addition, RA may modulate the pituitary gonadotropins, that in turn 677 regulate steroidogenesis (Minegishi et al. 2000) . 678 679 RA also appears to be one of the critical paracrine factors that stimulate the production of 17β-680 hydroxysteroid dehydrogenase type 2 (HSD17B2; which catalyzes the conversion of estradiol to 681 estrone) in neighboring epithelial cells, an important mechanism for local estradiol inactivation in the 682 endometrium (Taylor et al. 2015, Jiang et al. 2018a). 683 684 A connection between retinol/RA and ovarian function has been hypothesized based on the 685 observation that serum retinol levels in women vary with the stages of the estrous cycle (highest 686 levels in the proestrus and estrus phases), and that RBP4 serum levels are positively correlated with 687 that of gonadotropin (such as FSH and LH; see figure 6 for the roles of these gonadotropins in the 688 ovarian-estrous cycle) (Jiang et al. 2017). Similarly, ovarian expression of both mRNA and protein 689 levels of RBP4 in adult female mice increased at proestrus and peaked at estrus (Jiang et al. 2018b). 690 In ovaries from mice treated with FSH, increased levels of RA (and other retinoid forms) and RBP4, as 691 well as increased expression levels of RA-producing enzymes (Adh1, Aldh1a1), were observed (Jiang 692 et al. 2018b, Liu et al. 2018). Similar effects were observed in FSH-treated mouse granulosa cells, in 693 addition to increases in e.g. STRA6 (the specific receptor for RBP4) and cellular retinol binding 694 protein, suggesting that the gonadotropin FSH modulates the pathways for retinol uptake and its 695

Page 24: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

23

metabolism to RA in the mouse ovary (Liu et al. 2018). 696 697 While Stra6 knockout mice are fertile and overall healthy, this is not the case in humans with 698 mutations in the Stra6 gene (leading to the so-called Matthew-Woods syndrome9), pointing to a clear 699 species difference in functional importance of Stra6 (Szwarc et al. 2018). 700 701

Other female reproductive organs and pregnancy 702 The uterus is the key organ for reproductive processes such as blastocyst implantation and 703 placentation. In the female fetus, the Müllerian ducts differentiate into the oviduct, uterus and 704 vagina, with the resulting epithelia having distinct and separate organ-specific morphology and 705 function (Nakajima et al. 2016). At least in the mouse, RA signaling via RAR during embryo 706 development may determine the fate of Müllerian duct stroma into either uterus or vagina, as seen 707 in vitro with Müllerian ducts treated with either RA (leading to uterine epithelial differentiation) or 708 RAR inhibitors (leading to vaginal epithelial differentiation) (Nakajima et al. 2016). 709 710 The inner lining of the uterus is the endometrium, and its structure and function are regulated by the 711 ovarian sex steroid hormones estradiol and progesterone. In humans, the endometrium undergoes 712 major changes during the menstrual cycle (see figure 6). During the proliferative phase, ovarian 713 estrogens stimulate proliferation and growth, and during the secretory phase, progesterone prepares 714 the endometrium for potential implantation. Following fertilization, the implanting blastocyst first 715 attaches to the epithelial cells and then invades the endometrium by displacing the epithelial cells. It 716 will finally be embedded in the endometrial stroma where the formation of the placenta is initiated 717 (Su and Fazleabas 2015). The details of these processes can be species-specific. 718 719 RA regulates expression of matrix metalloproteinases (MMPs), which are produced by endometrial 720 stromal cells during decidualization (the process of endometrial changes that prepare for pregnancy). 721 RARs are expressed in the uterine stroma of mice and uterine epithelium of rats. In rats, RAR protein 722 expression is influenced by ovarian steroids; RAR expression increases under the influence of 723 estradiol, suggesting involvement of retinoids in growth and proliferation of endometrial epithelia. In 724 postmenopausal women taking estrogen, uterine RAR expression is reported to increase. Increased 725 RAR levels have also been observed in premenopausal women during the proliferative phase; a 726 phase which is associated with elevated estradiol levels (Sayem et al. 2018). 727 728 In the murine uterine epithelium, both mRNA and protein Cyp26a1 are expressed during the 729 blastocyst implantation period. Removing/inactivating Cyp26a1 in mice led to decreased pregnancy 730 rate, and reduced numbers of implantation sites (Han et al. 2010). It appears like the cyp26a1 731 enzyme might block the otherwise inhibitory effects of RA on implantation-related genes in the 732 endometrial epithelium during the murine embryo implantation period (Ma et al. 2012). RA 733 treatment of mouse blastocysts in vitro caused inhibition of cell proliferation and growth retardation, 734 and following implantation, the RA-treated blastocysts were also resorbed to a greater degree than 735 untreated blastocysts (Huang et al. 2005). 736 737 Via the heterodimer peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor (RXR), 738 the retinoid signaling pathway is involved in human placental processes such as the invasion of the 739 uterine epithelium by the extraembryonic trophoblast, or fetal placental, cells. Both trophoblasts and 740 decidual cells appear to be capable of synthesizing RA and (the RXR agonist ligand) 9-cis-RA (Tarrade 741 et al. 2001). At least in vitro, trophoblast invasion is inhibited by PPAR and RXR agonists, while 742 antagonists increased invasion. Functional retinoid signaling pathways have also been demonstrated 743 in human amniotic membranes (Marceau et al. 2006). 744 745

9 Matthew-Woods syndrome is characterized by anophthalmia or severe microphthalmia, and pulmonary hypoplasia or aplasia (https://www.orpha.net/consor/cgi-bin/OC_Exp.php?Expert=2470&lng=EN).

Page 25: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

24

746 RA concentration in follicular fluid (obtained during oocyte retrieval during the course of in vitro 747 fertilization) were found to be positively correlated with embryo quality (scored on day 3 after 748 fertilization). RA levels in follicular fluid also correlated moderately with plasma RA. Women with 749 endometriosis had significantly lower concentration of RA in follicular fluid and in plasma than 750 women with no endometriosis (Pauli et al. 2013). 751 752 The genital tubercle is a tissue present during embryonic development of the reproductive system; it 753 later develops into either the glans clitoridis or the glans penis in humans; RA appears to be involved 754 in its development, along with Rarb, Raldh2 and Cyp26b1 (Liu, Suzuki et al . 2012). 755 756 During preimplantation development of the blastocyst, RA signaling appear not to be involved (Rout 757 and Armant 2002). Functional retinoid signaling pathways have been demonstrated in human 758 amniotic membranes (Marceau et al. 2006). 759 760

Female reproductive pathologies and retinoid involvement 761 Endometriosis 762 The endometrium is the mucosal lining of the uterus, which changes in thickness during the 763 menstrual cycle. The endometrium consists of a single layer of glandular epithelial cells resting on a 764 stroma of connective tissue (Jiang et al. 2017). A number of retinoid-related genes (ALDH1A1, 765 ALDH1A2, CYP26A1, CRABP2, RARα, RARγ, and RXRα) is expressed in the endometrium of both 766 rodents and humans, and their expression is regulated by estrogen and/or progesterone (Jiang et al. 767 2017). As reviewed by Jiang et al. 2017, expression of these and other retinoid-related genes have 768 been noted in either stromal or epithelial cells. 769 770 Endometriosis is defined as the presence of endometrial tissue outside of the uterus, usually on the 771 ovaries, fallopian tubes, and peritoneum; a state which can cause severe pain. In addition, 772 fertilization problems have been associated with endometriosis (Taylor et al. 2015, Jiang et al. 773 2018a). 774 775 Several studies have demonstrated altered retinoid pathway signaling associated with endometriosis. 776 In human normal premenopausal endometrial tissue, mRNA expression of the RA-catabolizing 777 enzyme cyp26a1 increases substantially during the progesterone-dominated secretory phase 778 (leading to a resistance to RA signaling) when compared to the estrogen-dominated proliferative 779 phase, during which RA-synthesizing enzymes such as RALDHs were increased (Deng et al. 2003). At 780 least in vitro, RA has been shown to inhibit the decidualization of stromal cells; thus, reduced 781 concentration of RA in the endometrial tissue during the secretory phase could be necessary for a 782 successful implantation (Deng et al. 2003). In the mouse uterus, endogenous RA levels appear to be 783 controlled both by estrogen-dependent expression of RALDH enzymes and by progesterone-784 dependent expression of CYP26A1 (Fritzsche et al. 2007). Expression of CYP26A1 is down-regulated in 785 both the secretory and proliferative phases in endometrial biopsies from women with moderate or 786 severe endometriosis, when compared to healthy women (Burney et al. 2007). The availability of RA 787 may therefore be increased in the endometrial tissue of women with endometriosis. In 788 endometriosis, there is evidence for progesterone resistance, and cultured stromal fibroblasts 789 originating from endometriotic lesions have decreased ability to decidualize (which is necessary for a 790 successful blastocyst implantation) (Burney et al. 2007). 791 792 Reduced STRA6, CRBP1 and ALDH1A2 expression have been demonstrated to reduce RA in 793 endometrial stromal cells (Jiang et al. 2018a). Transcriptional activation via RA-CRABP2-RAR pathway 794 have been reported to trigger cell cycle arrest and apoptosis; thus, reduced signaling can cause 795 endometrial cells to escape apoptosis and contribute to survival of ectopic cells (Jiang et al. 2018a). 796 Since RA appears to stimulate the production of HSD17B2, which inactivates estradiol in the 797 endometrium, an abnormal RA pathway in endometriosis may explain the aberrant HSD17B2 798

Page 26: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

25

expression and the high local estradiol concentrations in endometriosis. In addition, altered retinoid 799 action may cause decreased CX43 expression and GJIC, reducing the decidualization capacity of the 800 stromal cells in endometriosis, which could contribute to progression of endometriosis lesions and 801 the associated subfertile uterine phenotype (Taylor et al. 2015, Jiang et al. 2018a). 802 803 Polycystic ovarian syndrome 804 Polycystic ovarian syndrome (PCOS) is a heterogenous disorder, characterized both by signs of 805 androgen excess and ovarian dysfunction (such as irregular ovulation and/or polycystic ovarian 806 morphology) (Escobar-Morreale 2018). Increased ovarian androgen production from theca cells, 807 elevated LH:FSH hormone ratio, and enlarged ovaries containing many antral follicles is often 808 observed; the associated arrest in follicular growth and anovulation can cause subfertility or 809 infertility (Jiang et al. 2017). 810 811 When comparing the response to RA, 9-cis-RA and retinol in cultured theca cells isolated from 812 normal-cycling women and women with PCOS, only RA led to increased testosterone production 813 (possibly via increased Cyp17 expression) in normal theca cells, while in the theca cells of PCOS 814 patients, all three (RA, retinol, and 9-cis-RA) variants have the same effect (Wickenheisser et al. 815 2005). Thus, in PCOS, theca cells may be sensitized to retinoid signaling stimulation. Moreover, 816 mRNA expression of RA-synthesizing enzymes RoDH2 and ALDH6 was increased in PCOS theca cells 817 (Wood et al. 2003), and PCOS ovaries show enhanced expression of RoDH (Marti et al. 2017). These 818 data suggest an increased rate of RA synthesis in the thecal cells of PCOS women (Jiang et al. 2017). 819 In women of reproductive age with acne and PCOS, treatment with oral isotretinoin decreased 820 ovarian volume (Acmaz et al. 2019). 821 822

Retinoid deficiency/excess in female reproduction 823 As reviewed in Clagett-Dame and Knutson 2011 (citing publications going back to the 1920’s), 824 retinoids are required for successful fertilization, implantation, placentation, embryogenesis and full-825 term pregnancy: in severely retinoid-deficient female animals, reproduction fails prior to 826 implantation, while in less severe deficiency, fertilization and implantation occur, but embryonic 827 death at mid-gestation is often observed. Retinoid deficiency has adverse effects on placenta 828 morphology (Noback and Takahashi 1978). RA can be supplemented in order to maintain normal 829 fertilization, implantation and early embryogenesis (White et al. 1998), but unless supplementation is 830 sufficiently high by 8.5 dpc, fetuses will be reabsorbed (White et al. 2000). Retinoids are also 831 required for the normal onset of meiosis in the developing embryo (discussed in detail in an earlier 832 section) and germ cells in rat embryos with severe retinoid deficiency fail to enter meiosis (an 833 accompanying decrease in ovarian Stra8 expression was observed); supplementation of small 834 amounts of RA to dams was sufficient to initiate meiosis (Li and Clagett-Dame 2009). Retinoid-835 deficient mice have a prolonged estrous cycle and also display a decreased rate of oocyte maturation 836 and number of ovulated oocytes after gonadotropin treatment (Kawai et al. 2016). 837 838 Conversely, a state of retinoid excess during pregnancy is known to cause teratogenic effects in 839 humans and animals. The pattern of birth effects, sometimes called retinoic acid syndrome, can be 840 observed in several organ systems, however does not appear to specifically target the female 841 reproductive organs (Azais-Braesco and Pascal 2000, Pennimpede et al. 2010). Retinoid excess may 842 be caused by diet or supplements, but also by administration of synthetic retinoids used for e.g. 843 treatment of severe acne. 844 845

Crosstalk with other signaling pathways 846 Via the heterodimer PPAR/RXR, the retinoid signaling pathway is involved in human placental 847 processes such as the invasion of the uterine epithelium by the extraembryonic trophoblast, or fetal 848 placental cells. Both trophoblasts and decidual cells appear to be capable of synthesizing RA and the 849 RXR agonist ligand 9-cis-RA (Tarrade et al. 2001). At least in vitro, trophoblast invasion is inhibited by 850 PPAR and RXR agonists, while antagonists increased invasion. 851

Page 27: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

26

8. Male reproductive organ development, function and health, and the 852

role of retinoids 853

854 The testis produces large numbers of gametes (sperm) throughout the reproductive age of the male 855 and is also the primary source of androgens, required for spermatogenesis and the development and 856 maintenance of male secondary sex characteristics throughout the body (Bittman 2015). 857 858 RA plays several critical roles in spermatogenesis including spermatogonia differentiation and 859 spermiation (i.e. release of spermatids into the lumen of the seminiferous tubuli) (Mark et al. 2015, 860 Teletin et al. 2017), and a facilitating role in spermatocyte meiosis (Teletin et al. 2019). In addition, 861 RA signaling appears to be necessary for proper development of the testis itself (Spade et al. 2018a). 862

863

864 865 Figure 6: The seminiferous tubule and spermatogenesis (adapted from Rato et al. 2012). The 866 seminiferous epithelium is composed of Sertoli cells and developing germ cells at different stages. 867 Leydig cells and blood vessels are located in the interstitium. The supporting Sertoli cells adhere to 868 the basement membrane where spermatogonia are also adherent. Spermatogonia type A divide and 869 develop into spermatogonia type B, which enter meiotic prophase and differentiate into primary 870 spermatocytes that undergo meiosis I to separate the homologous pairs of chromosomes and form 871 the haploid secondary spermatocytes. Meiosis II yields four equalized spermatids that migrate 872 toward the lumen where fully formed spermatozoa are finally released. Abbreviation: BTB, blood–873 testis barrier. 874 875

Testicular somatic cells and steroidogenesis 876 The bipotential somatic precursor cells in the embryonic testis develop into Sertoli cells, due to the 877 expression of the Y chromosome gene Sry (Minkina et al. 2014). During the embryonic and early 878 postnatal period, Sertoli cells divide mitotically, when the cessation of proliferation is followed by the 879 formation of tight junctions (creating the blood-testis barrier) between adjacent Sertoli cells (Nicholls 880

Page 28: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

27

et al. 2013). Sertoli cells together with the germ cells at different stages make up the seminiferous 881 tubule, which is surrounded by peritubular myoid cells and an interstitium containing the androgen-882 producing Leydig cells (Figure 6; Rato et al. 2012). The production of testosterone by Leydig cells, 883 stimulated by LH, depends on several steroidogenic enzymes, such as CYP11A1, HSD3B1 and 884 CYP17A1 (Jauregui et al. 2018). Testosterone secreted from the Leydig cells regulates the end of 885 meiosis, the establishment and maintenance of the BTB, and spermiation (Jauregui et al. 2018). 886 887 RA signaling, via RAR/RXR, appears to be necessary for development and function of the Sertoli cells 888 (Lucas et al. 2014). In primary rat Sertoli cells isolated on PND 10 and 20, RA suppressed proliferation 889 and initiated tight junction formation (Nicholls et al. 2013). Leydig cells express proteins for RA 890 synthesis, breakdown and signaling (Griswold and Hogarth 2018). Based on impairments observed in 891 VAD mice, the proper differentiation of Leydig cells appears to depend on sufficient retinoid levels 892 (Yang et al. 2018). 893 894 Fertility studies of conditional transgenic adult mice lacking functional RARα in the Leydig cells show 895 that RA signaling via RAR/RXR is required in Leydig cells for normal Leydig cell function and 896 subsequent male fertility; the knockout male mice were unable to produce pups (Jauregui et al. 897 2018). The same conditional knockouts also had changed expressions levels of steroidogenic 898 enzymes in the Leydig cells, increased BTB permeability as well as apoptotic pachytene 899 spermatocytes (Jauregui et al. 2018). The observed phenotype is similar to mice with low or no levels 900 of testosterone (Jauregui et al. 2018). Increased BTB permeability has also been shown in neonatal 901 mice after treatment with the RALDH inhibitor WIN 18,466 (Kent et al. 2016). 902 903 In cultured human fetal testes (ex vivo), addition of RA increased testosterone production and 904 expression of steroidogenic enzymes such as P450 cholesterol side-chain cleavage (P450SCC), 17α-905 hydroxylase/C17-20 lyase (P450C17), and steroidogenic acute regulatory protein (StAR) (Lambrot et 906 al. 2006). 907 908

Spermatogenesis 909 Spermatogenesis (figure 7) is the formation of haploid gametes (spermatozoa) from the diploid stem 910 spermatogonia, which includes the process of spermatogonia differentiation, meiosis, differentiation 911 of spermatids (spermiogenesis) and spermatid release (spermiation). In the mouse, after birth, the 912 mitotically quiescent gonocyte (also termed pro-spermatogonia) re-enter the cell cycle at 913 approximately PND 1-2, turn into spermatogonia at PND 3-6 as they migrate to the periphery of the 914 testis cords. Within the testis cord, the spermatogonia are flanked by Sertoli cells, and the 915 peritubular myoid cells, surrounding the outside of the cord (Teletin et al. 2017, Teletin et al. 2019). 916 During this first post-natal week, a sub-population of gonocytes differentiates directly (without 917 passing through the undifferentiated spermatogonia stages) into differentiating spermatogonia that 918 support the so-called first wave of pubertal spermatogenesis. After the first wave of spermatogenesis 919 (completed by postnatal day 35 in the mouse), subsequent waves derive from the undifferentiated 920 spermatogonia that has self renewal capacity (Yoshida et al 2006). 921

Page 29: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

28

922 Figure 7 (adapted from Mark et al. 2015): Schematic representation of adult spermatogenesis in the 923 mouse. In mouse spermatogonia in the single-cell state (AS) are considered as the true stem cells of 924 the spermatogenic lineage. In the first (proliferative) phase of spermatogenesis, As spermatogonia 925 divide to maintain a stem cell population (As) and expand the population of cells (Apr and Aal4–16; ) 926 which enter the differentiation pathway (Stages A1–A4, Int and B, collectively referred to as 927 differentiating spermatogonia (the previous stages, As – Aal16, are collectively also referred to as 928 undifferentiated spermatogonia). In the second, meiotic, phase , spermatocytes undergo 929 recombination and segregation of homologous chromosomes during the first two meiotic divisions 930 (M1 and M2) to generate step 1 spermatids (St1). The last phase, spermiogenesis, is subdivided into 931 16 steps based on morphological criteria (round spermatids, steps 1 to 8; elongating spermatids, 932 steps 9 to 16). The 12 first steps span the entire cycle of the seminiferous epithelium. Step 16 933 spermatids are released into the lumen of the seminiferous tubules as spermatozoa, during a process 934 called spermiation. 935 Abbreviations: PR, preleptotene; L, leptotene; Z, zygotene; P, pachytene; D, diplotene [the stages of 936 the first meiotic prophase]; 2S, secondary spermatocytes. 937 938 The timing of the progenitor cell commitment and subsequent differentiation and maturation of 939 spermatogonia along the seminiferous tubule is staggered, forming a spermatogenic wave (Griswold 940 2016). The cycle of the seminiferous epithelium (SE) is initiated by the precisely timed transition of A 941 (undifferentiated) spermatogonia into A1 spermatogonia, which subsequently will divide to generate 942 successively A2, A3, and A4 (differentiating) spermatogonia (Teletin et al. 2019). 943 944 The length of this wave is species-specific, e.g., 8.6 days in mice, 13 days in rats and 16 days in 945 humans (Bittman 2016). The spermatogonia continue to divide mitotically to produce cells that 946 replenish the stem cell pool and cells that undergo a series of mitotic divisions to increase the 947 number of spermatocytes, that then progress through meiotic sub-stages. In addition, it takes 8.6 948

Page 30: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

29

days for the A1 spermatogonia to become preleptotene spermatocytes (green) and enter meiosis 949 and an additional 8.6 days ×3 to form elongated spermatids ready for spermiation. The net result is 950 that once fully established, the same cell associations or the same group of cell types appear every 951 8.6 days. In the 8.6-day period between the transition of A spermatogonia to A1 spermatogonia, 952 there is a continuum of development of each cell type. 953 954 RA is considered to be critically involved in several steps of spermatogenesis: Spermatogonia 955 differentiation specifically during Aal-A1 transition, for spermiation and in regulation of the SE cycle 956 (Teletin et al. 2019). RA is indispensable in vivo to trigger the Aal – A1 transition (Teletin et al. 2019) 957 and is also required for the survival of some Aundiff spermatogonia. Just as for oocytes, RA plays a role 958 in the entry of the spermatocytes into meiosis (Raverdeau et al. 2012, Teletin et al. 2019), possibly 959 via control of replication-dependent core histone gene expression necessary for entry into S phase 960 (Chen et al. 2016). In the developing and adult testis, spermatogonia that are destined for meiosis 961 express Stra8 about a week before meiotic entry. Synthesis of both mRNA and protein Stra8 occurs 962 exclusively in two distinct phases of spermatogonia differentiation; in differentiating type A 963 spermatogonia (A1-A4), as well as in spermatocytes in the preleptotene (PR) and leptotene (L) phase 964 at meiosis entry.. At least in the mouse, RA is also required to disengage spermatozoa from the 965 Sertoli cell cytoplasm during spermiation (Spiller and Bowles 2015, Teletin et al. 2017). 966 967 During the onset of puberty in mice, sperm production is initiated by RA produced in Sertoli cells, 968 since these are the only cells in the seminiferous epithelium with RALDH activity; spermatogonia do 969 not possess the enzymatic machinery to produce RA (Ref. to be inserted). The RA produced in the 970 Sertoli cells acts in a paracrine manner on the spermatogonia, to regulate the Aal – A1 transition. 971 Spermatogenesis is blocked in juvenile male mice deficient in RDH10 in Sertoli cells and germ cells, 972 leading to a local lack of RA (Tong et al. 2013). Once the first wave of spermatogenesis has 973 progressed beyond a certain point, it appears (based on data from mice in which all three Aldh1a1 974 genes were deleted in Sertoli cells) that RALDH activity in Sertoli cells is no longer indispensable for 975 the spermatogonia differentiation to proceed. Thus, RA must be produced elsewhere, most likely by 976 RALDH2 in spermatocytes (preleptotene spermatocytes appear at every stage VIII of the cycle) and 977 spermatids. RA acts in a paracrine manner, inducing spermatogonia differentiation from the second 978 round of spermatogonia differentiation onward. The endogenous RA levels in the seminiferous 979 epithelium are tightly regulated. Pulses of increased levels of RA move along the tubule correlated 980 with the appearance of stages VIII-IX of the seminiferous cycle. In addition, the expression of the RA 981 target genes Stra8 (in spermatocytes) and Stra6 (in Sertoli cells) peak at stages VII-VIII of the SE cycle 982 (Teletin et al. 2017, Griswold and Hogarth 2018). However, at least in adult mice, ALDH enzymes 983 (ALDH1A1-3 and ALDH8A1) were not expressed in a stage-specific manner (Kent et al. 2016). In the 984 postnatal mouse testis, there appears to be other enzymes than ALDH1A isozymes that can oxidize 985 retinal to RA, since severe deficiency (obtained via genetic approaches) of the Aldh1a2 gene, 986 considered to be the major isomer involved, had no adverse effects on male fertility or health 987 (Beedle et al. 2019). In contrast, studies using human testis biopsies suggest that ALDH1A1 is the 988 predominant form that produces RA in the Sertoli cells, while ALDH1A2 has the corresponding role in 989 the developing sperm (Arnold et al. 2015). 990 991 For the regulation of spermatogonial exposure to RA, two hypotheses has been presented: 1) all 992 spermatogonia are primed to respond to RA, but the exposure to RA is periodic and tightly 993 controlled, and 2) all spermatogonia are exposed to RA but only some can respond (Busada and 994 Geyer 2016). The differential expression of RARγ between spermatogonia subpopulations could 995 explain how the As spermatogonia (with no RARγ expressed) remain undifferentiated and maintain 996 self-renewal capabilities even in the presence of high RA concentrations, which induce differentiation 997 in the (RARγ-expressing) Aal progeny populations (Teletin et al. 2017). Observations from cell-specific 998 conditional knockout studies of RA receptor isomers suggest that the RA signal in spermatogonia is 999 transduced via RAR/RXR heterodimers (Gely-Pernot et al. 2015). Another suggestion is that RA 1000 signaling also operates via Sertoli cells, as germ cell differentiation can proceed in the absence of 1001

Page 31: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

30

functional RAR and RXR isotypes in spermatogonia (Teletin et al. 2017). RA may also act via non 1002 receptor-mediated pathways, such as via kinase singaling (Busada and Geyer 2016). 1003 1004 Spermatogonia exposure to RA may also be regulated by the presence of the RA-catabolizing CYP26 1005 enzymes. All three isoforms of CYP26 family are present in the murine postnatal testis, although 1006 observations after conditional knocking-out of the A1 and/or B1 genes in germ and/or Sertoli cells 1007 reveal that CYP26B1 is the critical isoform (Hogarth et al. 2015). In the fetal testis, elimination of RA 1008 by CYP26B1 is necessary not only to avoid meiosis, but also for normal mitotic arrest of male PGCs as 1009 well as for avoiding germ cell apoptosis (Rossitto et al. 2015). CYP26B1 is initially produced by the 1010 Sertoli and Leydig cells and/or interstitial somatic cells, although after birth, Cyp26b1 transcripts are 1011 confined to the peritubular myoid cells (Rossitto et al. 2015). CYP26A1 and CYP26B1 form a catabolic 1012 barrier against any RA present in the immediate environment of the seminiferous tubules. No 1013 variations in the expression of CYP26 enzymes exist across the SE cycle, unlike several proteins 1014 involved in retinoid storage (e.g. LRAT) and RA production (RALDH1 in Sertoli cells and RALDH2 in 1015 germ cells) which are regulated in a periodic manner along the seminiferous tubule (Teletin et al. 1016 2017). 1017 1018 Differential expression of CYP26B1/A1 in their microenvironment, rather than differential expression 1019 of RARs or RXRs within stem and non-stem spermatogonia, has been suggested as the mechanism for 1020 protecting the stem spermatogonia from differentiation in response to RA signaling (Lord et al. 1021 2018). In neonatal mice, heterogeneity in terms of responsiveness to RA in the gonocytes (pro-1022 spermatogonia) was observed: 1) undifferentiated spermatogonia capable of responding to RA (due 1023 to RARγ being expressed) can either respond to low levels of RA or can only respond when higher 1024 levels of RA are added (the lack of response to low levels was hypothesized to be due to CYP26 1025 catabolism, and 2) undifferentiated spermatogonia that cannot respond to RA (due to lack of RARγ 1026 expression) and are destined to become foundational spermatogonial stem cells (Velte et al. 2019). It 1027 is also of note that RA deficiency may cause undifferentiated spermatogonia to retain their stem cell 1028 state (Agrimson et al. 2017). Thus, differential expression of RARγ and CYP26 are important for 1029 proper maintenance of spermatogonia differentiation. 1030 1031 Knockout studies in mice show that despite the critical role of RA in spermatogonia differentiation 1032 and in male germ cell meiosis, both differentiation and meiosis can occur in germ cells where RAR or 1033 RXR are absent, although a fraction of the A1 spermatogonia are adversely affected (Gely-Pernot et 1034 al. 2015). Initiation and progression of meiosis also proceeded in these mice. The conclusion was 1035 that the RA signaling pathway was not autocrine, but rather operated in Sertoli cells (Gely-Pernot et 1036 al. 2015). 1037 1038 In male mice with arrested spermatogonia differentiation, due to either vitamin A deficiency or 1039 genetic knockout of Aldh1a1 in their Sertoli cells), a single injection of RA leads to resumed Aal – A1 1040 transition, with preleptotene spermatocytes expressing Stra8 and Rec8 appearing synchronously, 8 1041 days post-RA injection, in all seminiferous tubules. The same effect is observed following treatment 1042 with the RALDH inhibitor WIN 18,446, with a subsequent RA injection (Hogarth et al. 2013). RA 1043 administration in vitamin A-deficient mice or rats results in truncation of the normal 12 (mice) or 14 1044 (rat) stages of the seminiferous epithelium cycle to only three or four stages (still enabling normal 1045 sperm production). Additionally, the normally asynchronous wave of spermatogenesis is 1046 synchronized, with near-identical timelines of the differentiation progression in all seminiferous 1047 tubules, leading to a pulsatile rather than continuous sperm production (Teletin et al. 2017). 1048 1049 Recent findings, reviewed in Teletin et al. 2019, suggest that RA is not decisive to meiotic initiation in 1050 male PGCs, but merely facilitates this process. Data indicates that meiosis is not blocked, but slightly 1051 delayed, in mice lacking all RALDH in the SE. Leptotene spermatocytes in Aldh1a1-3Ser−/− and Aldh1a1-1052 3Germ−/−;Ser−/− mutants appear 24 h later than their counterparts in control mice, but express STRA8 1053 and subsequently progress to the pachytene stage. The strong effect of BMS-204,493 on STRA8 1054

Page 32: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

31

expression and meiotic initiation is probably due to BMS-204,493 stabilizing the binding of RAR to co-1055 repressor complexes that would cause transcriptional silencing at the Stra8 locus through histone 1056 deacetylation and chromatin condensation (Teletin et al. 2019). 1057 1058 The role of Stra8 gene expression in normal spermatogenesis is supported by an observation in 1059 humans: an association was found in men with a specific single nucleotide polymorphism in their 1060 Stra8 gene with spermatogenic impairment evident as azoospermia or oligozoospermia (Lu et al. 1061 2013). 1062 1063

Secondary male reproductive organs 1064 The prostate produces slightly alkaline prostate fluid that makes up approximately 30% of the 1065 volume of semen in humans and is critical for male reproductive health (Verze et al. 2016). The 1066 retinoid signaling pathway has several functions in differentiation and maintenance of secondary 1067 male reproductive organs. Presence of RA is necessary for prostate formation from the urogenital 1068 sinus during sexual differentiation (Vezina et al. 2008, Bryant et al. 2014). Genetic deletion studies in 1069 mice have also demonstrated critical roles for RA via Rarγ in prostate and in seminal vesicles (Lohnes 1070 et al. 1993). 1071 1072 RA signaling appears to be necessary for the function of epididymis, into which sperm are released 1073 after spermiation, undergo maturation, and are stored until ejaculation (Jauregui et al. 2018). In a 1074 conditional transgenic model with a dominant negative form of RARα expressed in Leydig cells and in 1075 the epididymis, the resulting abnormal epididymis phenotype may contribute to the infertility 1076 observed in these mice (Jauregui et al. 2018). These observations support previous findings that lack 1077 of RA signaling in the epididymis results in squamous metaplastic epididymal epithelium; 1078 alternatively, the abnormal epididymal phenotype may have been due to lack of testosterone 1079 (Jauregui et al. 2018). 1080 1081

Male reproductive pathologies 1082 Impaired RA signaling may play a role in several male reproductive pathologies. The transition from 1083 PGCs to differentiating spermatogonia is impaired in the absence of RA in the testis cords (Teletin et 1084 al. 2017).PGCs that fail to differentiate into spermatogonia may be the source of carcinoma in situ, 1085 which can develop into testicular germ cell cancer in humans (Busada and Geyer 2016, Teletin et al. 1086 2017). 1087 1088 In prostate tumor tissue, RA concentrations are lower than in normal prostate tissue, and since it has 1089 been hypothesized that this is due to increased RA catabolism, inhibitors of CYP26 enzymes, called 1090 RA metabolism blocking agents (RAMBAs) have been used in the treatment of prostate cancer 1091 (Nelson et al. 2013). RAMBA therapy promotes differentiation and inhibits proliferation by increasing 1092 endogenous RA in tumors (Denis et al. 1998). Some RAMBAs also inhibit estrogen synthesis via 1093 inhibition of aromatase (CYP19) and testicular androgen synthesis via inhibition of 17,20-lyase 1094 (CYP17) (Bryson and Wagstaff 1996). The ALDH1A2 enzyme expression is also altered in prostate 1095 cancer. The expression of the Aldh1a1 gene is lower and its promoter region is hypermethylated in 1096 epithelia from malignant prostate tumors (Kim et al. 2005). 1097 1098 Cryptorchidism, or non-descended testis, is a common congenital malformation that also leads to 1099 increased risk of testicular cancer and infertility in adult life (Bay et al. 2011). Complete testicular 1100 descent is necessary for normal testicular function in adult males, and the process of testis descent is 1101 regulated by the Leydig cell hormones insulin-like peptide 3 (INSL3) and testosterone (Bay et al. 1102 2011). The spectrum of reproductive system malformations caused by VAD in rats includes 1103 cryptorchidism (See et al. 2008). Both RA levels and Stra8 expression were significantly lower in the 1104 rat cryptorchid testis (induced by in utero exposure to the anti-androgen flutamide) compared to the 1105 normal testis (Peng et al. 2016). In an in vitro system, RA upregulated the expression of the gene 1106 LGR8, which encodes for the receptor for INSL3 (Klonish et al. 2005). 1107

Page 33: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

32

1108 Retinoid deficiency in male reproduction 1109

The importance of narrowly regulated retinoid levels on male reproduction can be illustrated in 1110 animal studies measuring effects of experimentally increased or decreased levels of retinoids. 1111 Adverse effects on the male reproductive system following experimental limitations of RA have been 1112 demonstrated in multiple in vivo studies (discussed in previous sections of this report and reviewed 1113 in Clagett-Dame and Knutson 2011). In rodents maintained on a vitamin A-deficient diet, testicular 1114 degeneration with impaired spermatogenesis and a complete disappearance of all meitotic and 1115 postmeiotic cells was observed (Coward et al. 1969, Morales and Griswold 1987, van Pelt and de 1116 Rooij 1990). Spermatogenesis is arrested at the preleptotene spermatocyte stage in VAD rats and at 1117 the spermatogonia stage in VAD mice; however, spermiation failure is observed in both species 1118 under VAD conditions (Ghyselinck et al. 2006). Vitamin A deficiency also leads to replacement of 1119 normal glandular epithelium in the epididymis, prostate and seminal vesicles, by a stratified 1120 squamous keratinizing epithelium, resulting in inhibited seminal fluid production (Wiseman et al. 1121 2017). The RBP4-/- mouse model phenotype is similar to VAD, including the inability to mobilize 1122 hepatic retinoid stores into circulation, progressive testicular degeneration together with 1123 spermatogonia differentiation arrest and failure of spermiation (Ghyselinck et al. 2006). 1124 1125 In human populations with insufficient circulating retinoids, symptoms such as blindness is well-1126 known, but there appears to be no information on reproductive parameters such as sperm 1127 production or fertility (Hogarth and Griswold 2010). However, observations made on testicular tissue 1128 samples obtained in different clinical situations, suggest a correlation between adverse reproductive 1129 parameters and an affected retinoid system. A decrease in RA and 13-cis-RA was measured in testis 1130 tissue from men with abnormal sperm production undergoing scrotal surgery (Nya-Ngatchou et al. 1131 2013). In another study, lower levels of the RA-producing RALDH1A2 enzyme in testicular tissue were 1132 associated with male infertility (Amory et al. 2017). 1133 1134 1135

Page 34: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

33

9. Impact on female and male reproduction by chemicals acting via the 1136

retinoid system 1137

1138 Exposure to chemicals early in life may affect both female and male reproductive organs and their 1139 functions, most likely via a large number of mechanisms, with effects via estrogen and/or androgen 1140 pathways being most frequently discussed. A testicular dysgenesis syndrome (Skakkebaek et al. 1141 2001) as well as an ovarian dysgenesis syndrome (Johansson et al. 2017) have been described. Both 1142 dysgenesis syndromes are defined as early (fetal) alterations in testicular or ovarian structure or 1143 function that cause an impairment of reproductive parameters in adulthood. One of the mentioned 1144 fetal alterations in the ovarian dysgenesis syndrome was disruption of the RA-dependent meiosis 1145 initiation (Johansson et al. 2017). Also for embryonic males, it has been suggested that testicular 1146 toxicity could result from disruption of local RA homeostasis or signaling (Spade et al. 2018b). In the 1147 adult organism, chemicals may interfere with e.g. normal sperm production (Sharpe 2010) and 1148 normal function and morphology of female reproductive tissue. 1149 1150 Some observed effects and mechanisms by which chemicals could interfere with the retinoid 1151 pathway are summarized in table 1 (and also in e.g. Nilsson and Håkansson 2002, Novak et al 2008, 1152 and Shmarakov 2015). In brief, chemicals have been shown to deplete tissue retinoid levels by 1153 affecting retinoid metabolism, also after in utero exposure. Chemicals can also cause activation or 1154 inactivation of retinoid receptors. As described in this report, the retinoid pathway is important, or 1155 even critical, in several aspects of both female and male reproduction and during fetal development. 1156 It is thus conceivable that chemicals capable of interfering with the retinoid pathway may, as a result, 1157 cause adverse effects on reproductive parameters, assuming that such chemicals reach target cells 1158 during critical windows. 1159 1160 Under the course of this project, very few articles have been retrieved that focus on effects of 1161 contaminants on the female reproductive system while simultaneously looking at retinoid 1162 parameters such as e.g. RA-synthesizing/metabolizing enzymes. For similar studies on the male 1163 reproductive system, there are a slightly higher number of available references. There are also 1164 publications were chemicals have had adverse effects on reproductive parameters while 1165 simultaneously causing decreased liver retinoid stores; however, no retinoid-related parameters 1166 were measured in the reproductive organs (see e.g. van der Ven et al. 2008 and 2009 in table 1). 1167 While no conclusions on any causal role of retinoid perturbation in the adverse reproductive effects 1168 can be made from such studies, causality cannot be excluded. It should be noted that chemical-1169 induced decreases of hepatic retinoid levels appear to be accompanied by decreased testicular levels 1170 (Håkansson et al. 1991). 1171 1172 While most studies use animals, there is human data as well. E.g., in the 1960’s, it was demonstrated 1173 that the compound WIN 18,446 could reversibly inhibit spermatogenesis in men (Heller et al. 1961). 1174 More recently, the same compound was shown to inhibit the conversion of retinal to RA, most likely 1175 by inhibiting ALDH1A2 (Paik et al. 2014), and this finding has led researchers to suggest inhibition of 1176 retinoid metabolism as an approach to male contraception (Hogarth et al. 2011). In males, treatment 1177 with 13-cis-RA (used in the treatment of e.g. acne) can improve sperm production (Cinar et al. 2016, 1178 Amory et al. 2017). 1179 1180 1181

Page 35: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

34

Table 1: Examples of chemicals interfering with the retinoid system in different models. 1182 Chemical(s) Model system Endpoint Observed effect Reference 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)

Male rats, single dose

Retinoid levels in several organs, including testes and epididymis

↓Retinyl esters in liver, testes, epididymis ↑Retinyl esters in kidney

Håkansson et al. 1991

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)

Pregnant rats and PND 7 pups, single gestational dose

Retinoid levels in liver, lung and kidney

↓Retinyl esters in maternal and perinatal liver and lung ↑Retinyl esters in maternal and perinatal kidney

Kransler et al. 2007

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)

Male rats, single dose

RA and retinyl ester levels and LRAT expression in liver and kidney

↑ RA in liver and kidney ↓Retinyl esters in liver ↑Retinyl esters in kidney ↑ LRAT in kidney

Hoegberg et al. 2003

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)

Male rats, single dose

RA metabolism in microsomes from liver, lungs, kidneys, testes

↑ RA metabolism in all investigated tissues

Fiorella et al. 1995

Technical pentabromodiphenyl ether mixture

Female and male rats (enhanced 28d TG407 study)

Retinoid levels in liver ↓Retinyl esters in liver van der Ven et al. 2008a

Hexabromocyclodecane Female and male rats (enhanced 1-gen TG415 study)

Retinoid levels in liver ↓Retinyl esters in liver van der Ven et al. 2009b

Fluconazole Mice (exposed during pregnancy)

mRNa induction of CYP26A1 and CYP26B1 Organ development (not repro)

Upregulation of CYP26A1 and CYP26B1 Abnormal branchial arch development.

Tiboni et al. 2009

Organochlorine pesticides (chlordane, dieldring, aldrin, endrin, endosulfan)

Reporter cell lines (RARE & RARα, β, γ) CYP26A1

Activation of RARs Induction of CYP26A1

Activation was observed. CYP26A1 induction was observed.

Lemaire et al. 2004

543 environmental chemicals

Yeast cells transfected with the human RARγ

RARγ agonistic activity 85 of the 543 chemicals had RARγ agonistic effects (especially monoalkylphenols and styrene dimers).

Kamata et al. 2008

309 environmental chemicals

HepG2 cells transfected with cis/trans-reporter transcription units for RARα, β, γ

Transcription factor activity

Max responders were lindane, oxadiazon and imazalil

Martin et al. 2010

28 environmental and other compounds

Human uterus and prostate cytosol

Chemical displacement; inhibition of 3H-RA binding to cytosol

A number of the chemicals could displace 3H-RA. MEHP most potent.

Paganetto et al. 2000

PPARγ agonists rosiglitazone and pioglitazone

HepG2 cells mRNA induction of CYP26B1 and CYP26A1

mRNA induction of CYP26B1 and (to a lesser extent) CYP26A1

Tay et al. 2010

Phenobarbital Human hepatocytes

mRNA induction of CYP26B1

Weak induction of CYP26B1 Finkelstein et al. 2006

Tributyl tin (TBT) Human and mice ovarian theca cells

Cholesterol homeostasis via RXR pathway

Impaired cholesterol homeostasis Pu et al. 2019

a Also observed in males: decreased weight of epididymis, seminal vesicles and prostate, sperm head deformities; in 1183 females: induced adrenal activity of the steroidogenic CYP17 enzyme 1184 b Also observed in males: decreased weight of testis 1185 1186 It should be pointed out that effects of chemicals on retinoid homeostasis can be indirect, since some 1187 metabolic enzymes are used both for detoxification and for retinoid homeostasis, such as 1188 RALDH/ALDH class 1 (Alnouti and Klaassen 2008). Induction of Aldh isomers (as studied in mouse 1189 livers) after in vivo administration of several activators of different nuclear receptors (such as the 1190 constitutive androstane receptor CAR, pregnane X receptor PXR, PPAR, AhR) was isomer- and 1191 activator-specific (Alnouti and Klaassen 2008). The well-known induction of cytochrome P450 1192 enzymes via AhR after exposure to dioxins and dioxin-like polychlorinated biphenyls (PCBs) may be 1193 relevant since some of these P450 enzymes are believed to be involved in either synthesis or 1194 oxidation of RA (Murphy et al. 2007). If the end result is altered RA concentrations in fetal or adult 1195 reproductive organs, correct development and/or function of these organs could be compromised. 1196 1197

Page 36: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

35

Since RXR can heterodimerize with several other nuclear receptors, it is also conceivable (and has 1198 indeed been shown; see e.g. Tarrade et al. 2001) that any interference with the retinoid system can 1199 also have effects on other signaling pathways, via e.g. PPAR, pregnane X receptor (PXR), constitutive 1200 androstane receptor (CAR) and vitamin D receptor (VDR). Also, interactions with AhR signaling 1201 pathways on several levels have been shown (Murphy et al. 2007, Vezina et al. 2009). From a 1202 functional/endpoint perspective, crosstalk with the other endocrine systems (estrogen, androgen, 1203 thyroid) is important to consider. 1204 1205 1206

Chemicals affecting female reproduction 1207 1208 Analgesics: 1209 Exposure to the analgesic chemicals acetaminophen (a.k.a. paracetamol) and indomethacin 1210 appeared to give rise to delayed entry into meiosis in female rats after in utero exposure, with 1211 changes in ovarian Stra8 levels reflecting this delay (Dean et al. 2016). Other effects, not currently 1212 linked to Stra8 or RA, included decreased fetal ovarian germ cell numbers and (in adult females 1213 exposed in utero) reduced ovarian size and female fertility (measured as number of pups per litter) 1214 were observed (Dean et al. 2016). 1215 However, in a mouse study with in utero exposure to paracetamol, ovarian Stra8 levels were not 1216 affected, while in the same females, germ cell numbers decreased (not a Stra8-related effect) (Holm 1217 et al. 2016). The diverging results may be due to e.g. differences in species and/or dose or time of 1218 exposure (350 mg/kg on days 13.5-21 in the Dean study (Dean et al. 2016); 50 or 150 mg/kg on days 1219 7 until birth in the Holm study (Holm et al. 2016)). 1220 1221 Bisphenol A (BPA): 1222 In utero exposure to BPA in mice has been associated with a delay in meiotic prophase I, 1223 hypothesized to be due to decreased expression of Stra8 in the fetal oocytes (Zhang et al. 2012). In 1224 the same mice at postnatal day three, increased BPA exposure levels were associated with more 1225 oocytes in germ cell cysts and fewer oocytes in primordial follicles (Zhang et al. 2012). One suggested 1226 mechanism for how BPA affects Stra8 expression is increased DNA methylation, meaning that the 1227 effect of BPA appears to be independent of any direct effect on retinoid metabolism or homeostasis, 1228 although it exerts (one of) its effects on (the retinoid-regulated gene) Stra8 (Zhang et al. 2012). 1229 Not all data support an effect of BPA on Stra8. In human fetal oocytes cultured in BPA-containing 1230 media, the expression pattern of Stra8 was similar to that in control cultures (Brieno-Enriquez et al. 1231 2012. In fetal ovaries originating from pregnant mice exposed to BPA, the observed increase in Stra8 1232 expression did not differ from that in unexposed mice (Lawson et al. 2011). 1233 1234 Phthalates: 1235 Only a few publications were retrieved in which retinoid-related endpoints were investigated 1236 following DEHP exposure. DEHP caused a delay in meiosis in mouse fetal germ cells following in utero 1237 exposure; the concomitant decrease in mRNA and protein expression of Stra8 was suggested to be 1238 related (Zhang et al. 2015). The DNA methylation level of Stra8 in oocytes of the F1 generation was 1239 increased as a result of DEHP exposure, and these changes were inherited by the F2 generation 1240 (Zhang et al. 2015). 1241 DEHP may also exert effects via PPAR-RXR. In vitro, the DEHP metabolite monoethylhexyl phthalate 1242 (MEHP) suppressed aromatase, the rate-limiting enzyme for conversion of testosterone to estradiol, 1243 expression via activation of PPAR-RXR heterodimers in rat ovarian granulosa cells (Lovekamp-Swan et 1244 al. 2003). 1245 Phthalates can target many aspects of ovarian development and normal function (reviewed by 1246 Hannon and Flaws 2015); the exact mechanisms are unknown. 1247 1248 1249

Page 37: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

36

Organotins: 1250 In marine gastropods, tributyltin, used in antifouling paints for ships and fishing nets, induces 1251 imposex (the development of male genitals in females) via binding RXR and thereby activating the 1252 RXR-RAR heterodimer (Nishikawa et al. 2004). 1253 In human placental choriocarcinoma cells, trialkyltins stimulate human chorionic gonadotropin 1254 production and aromatase activity by acting as RXR agonists (Nakanishi et al. 2005). 1255 The RXR signaling pathway may be involved in the observed increase in progesterone production in 1256 human placental cells (in vitro) following organotin exposure (Macejova et al. 2016). 1257 No RXR activation was observed in rodent or human placenta tissue in which ng-mg/kg levels of 1258 organotin levels were present (de Araújo et al. 2018). 1259 In ovarian theca cells from humans, mice and other mammalian species, TBT stimulated cholesterol 1260 extracellular efflux via the RXR pathway (Pu et al. 2019). 1261 1262 R115866: 1263 This triazole-containing molecule is a CYP26 inhibitor (more potent than liarozole and largely without 1264 the inhibitory ability of liarozole on the CYP-dependent formation of estradiol and testosterone) 1265 (Stoppie et al. 2000). R115866 administration leads to increased endogenous levels of RA, with 1266 subsequent RA-like effects such as inhibition of vaginal keratinization in estrogen-stimulated rats 1267 (Stoppie et al. 2000). 1268 1269 Isotretinoin/13-cis-RA: 1270 This pharmaceutical retinoid, used for e.g. treatment of acne has been found to lead to reduced 1271 antral follicle count, ovarian volume and levels of anti-Müllerian hormone (AMH; a marker of ovarian 1272 follicle number) in humans, suggesting negative effects on the ovarian reserve (Aksoy et al. 2015). 1273 Similar effects have been observed in rats (Abali et al. 2013). The effects appear to be transient both 1274 in humans (Cinar et al. 2016) and in rats (Korkmaz et al. 2017) once treatment ceases. Isotretinoin 1275 administered to rats at doses up to five times higher than clinical doses used for acne treatment 1276 reportedly had no adverse effects on fertility, conception rate, gestation or parturition, as 1277 summarized in an US FDA Pharmacology Review of isotretinoin10. The ICH reproductive toxicology 1278 guideline for registrations of pharmaceuticals for human use does not require histopathological 1279 examination of ovaries in reproductive toxicity studies and therefore it is not possible to draw any 1280 firm conclusions on possible effects of isotretinoin on ovarian volume or follicle count from the US 1281 FDA summary of this study. 1282 1283

Chemicals affecting male reproduction 1284

1285 Thiocarbamate herbicides: 1286 Molinate, a known testicular toxicant in the rat, is also an inhibitor of RALDH and has been shown to 1287 inhibit the conversion of retinal to RA; decreased testicular levels of RA were observed in rats dosed 1288 with molinate (Zuno-Floriano et al. 2012). 1289 1290 Conazoles: 1291 Conazole fungicides are triazole-based compounds used both in agriculture and pharmacologically, 1292 and which exert their fungicidal effects via broad inhibition of CYP enzymes. In vertebrates, effects 1293 include inhibition of CYPs involved in vertebrate sex steroid synthesis as well as hepatic xenobiotic 1294 and hepatic metabolism (Chen et al. 2009, Tonk et al. 2015). Reprotoxic effects have been observed 1295 following in vivo exposure (Vickery et al. 1985, Taxvig et al. 2007). 1296 In the pharmaceutical industry, structurally related compounds such as liarozole and talarozole have 1297 been considered for treatment of some cancers and dermatological diseases (Stevison et al. 2017). 1298 One mechanism of action of these compounds is the blocking of RA catabolism via inhibition of 1299 10 https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/021951Orig1s000PharmR.pdf, downloaded Sep 4, 2018.

Page 38: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

37

CYP26. Transient increased testicular RA levels have been observed in mice given talarozole (Stevison 1300 et al. 2017). In VAD mice administered liarozole after a dose of RA, the RA-induced proliferative 1301 effects on A spermatogonia was less than in VAD mice given RA but not liarozole (Gaemers et al. 1302 1997). However, to date, no data are available indicating causal relationships between conazole 1303 exposure, altered retinoid synthesis, and impaired male reproduction 1304 1305 Bisphenol A: 1306 In mice, neonatal exposure to bisphenol A has been shown to cause decrease in the number of 1307 sperm and damage to the motility and morphology of sperm in adult mice, and the effect was, to 1308 some extent, ameliorated by vitamin A supplementation and aggravated under vitamin A-deficient 1309 conditions (Nakahashi et al. 2001, Aikawa et al. 2004). The authors suggested that the development 1310 and functional differentiation of the reproductive tracts and the gonads may be controlled by a 1311 balance between levels of estrogen and retinoids. 1312 In mouse embryonic stem cells, bisphenol A was found to upregulate the expression of Stra8 1313 (amongst other genes) in a manner that appeared to be consistent with a feminizing effect, but 1314 bisphenol A seemed to act via a non-RA and non-RAR mechanism (Aoki et al. 2012). 1315 1316 Triorganotin compounds: 1317 Triorganotins are considered endocrine-disrupting compounds and have been shown to bind RXR 1318 (Ref to be inserted). Macejova et al. (2016) reviewed effects of triorganotin compounds on male 1319 reproductive organs in rats, but it was unclear if these effects were the result of a disrupted retinoid 1320 pathway. 1321 1322 Phthalates: 1323 Some phthalate esters are anti-androgenic and thus considered potential causing agents in the 1324 “testicular dysgenesis syndrome” (Skakkebæck et al. 2001). These compounds have now been shown 1325 to also interfere with RA synthesis, both in vitro (Chen and Reese 2016) and ex vivo (Spade et al. 1326 2018b). It was suggested (in Chen and Reese 2016) that a dual mechanism of both anti-androgen and 1327 retinoid disruption may lead to developmental effects in humans and rodents. 1328 1329 Hexachlorocyclohexane: (the gamma isomer is known under the name of lindane) 1330 Lindane administration caused atrophy of the epididymidis and seminal vesicles, along with 1331 decreased sperm count in the epididymis and reduced activities of steroidogenic enzymes, in VAD 1332 rats but not in rats given sufficient amounts of retinoids in their diet (Pius et al. 1990). 1333 1334 BMS-189453: 1335 BMS-189453 is a synthetic retinoid RARα, β and γ antagonist. Following oral administration to rats 1336 and rabbits for up to one month, BMS-189453 caused testicular degeneration and atrophy (Schulze 1337 et al. 2001). In later studies using lower doses, this compound was shown to reversibly inhibit 1338 spermatogenesis in rats, without other adverse testicular effects (Chung et al. 2016). 1339 1340 WIN 18,446: 1341 While the BMS synthetic retinoid above inhibits RA signaling, WIN 18,466 (a bisdichloroacetyl 1342 diamine) acts by lowering local RA concentration via inhibition of RALDH2 (Kogan et al. 2014). WIN 1343 18,466 administration to neonatal mice caused meiotic defects in spermatocytes (Kent et al. 2016). 1344 In WIN 18,466-treated rodents, the progression of progenitor cells from A spermatogonia into A1 1345 spermatogonia is blocked (Griswold and Hogarth 2018). 1346 1347 R115866: 1348 This triazole CYP26 inhibitor was capable of mimicking the effects of RA in an in vitro dog testis 1349 model, such as upregulation of e.g. Stra8, an effect that was also observed at protein level 1350 (Kasimanickam and Kasimanickam 2014). 1351 1352

Page 39: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

38

Ro 23-2895: 1353 High doses of the synthetic retinoid Ro 23-2895 administered to rats caused testicular degeneration, 1354 decreased testicular weight, delayed sperm release and retention, and 1355 disorganization/desquamination of the tubular epithelium accompanied by reduced numbers of 1356 mature elongated spermatids (Bosakowski et al. 1991). These effects resemble those caused by 1357 vitamin A deficiency. In a parallel study plasma and testis retinol levels were lower compared to 1358 controls, suggesting that Ro 23-2895 caused testicular degeneration by interfering with normal 1359 retinoid homeostasis (Bosakowski et al. 1991). 1360 1361 Isotetinoin/13-cis-RA: 1362 Results from a pilot study suggest that treatment with 13-cis-RA (used for e.g. acne treatment) can 1363 improve sperm production (Amory et al. 2017). 1364 1365 1366

Page 40: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

39

10. Potential adverse outcome pathways in female and male reproduction 1367

1368 Adverse Outcome Pathways (AOPs) form a framework for organizing data on the relationships 1369 between a molecular initiating event (MIE) induced by the interaction of a stressor (i.e. a chemical) 1370 with a molecular target and the resulting sequence of events (KE), leading to an adverse outcome 1371 (AO). AOPs can establish the rationale for the use of particular assays or in vivo endpoints, and/or 1372 highlight the need for the development of assays or exploring existing test guidelines to cover one or 1373 more components of an AOP. 1374 1375 Currently, there are no AOPs published focusing on the disruption of the retinoid pathway in the AOP 1376 Wiki database11, with the exception of one AOP under development, linking RALDH inhibition with 1377 visual impairment in fish (not open for citing). In the scientific literature, attempts to build AOPs and 1378 other frameworks to understand RA-dependent effects on embryogenesis have been published 1379 (Tonk et al. 2015, Baker et al. 2018, Battistoni et al. 2019, Di Renzo et al. 2019, Piersma et al. 201912), 1380 with disruption of CYP26 enzymes and RALDH2 suggested to be important KE. Previous efforts have 1381 not focused specifically on the reproduction system per se, even though embryo development of 1382 several other organ systems is discussed. Below, drafts of such potential AOPs are described. 1383 1384 Since there is substantial crosstalk between the retinoid system and other nuclear hormone systems 1385 (see section 4), there are additional AOPs that may be involved with retinoid pathway effects on 1386 reproduction., For example, activation of PPARα may impair steroidogenesis, and could lead to 1387 impaired fertility in males (AOP1813). RXR is a PPAR heterodimer partner, and it is possible that RXR 1388 may be involved in this AOP. 1389 1390 1391

11 https://aopwiki.org/aops; accessed in September 2019. 12 https://cfpub.epa.gov/si/si_public_record_Report.cfm?dirEntryId=345956&Lab=NCCT 13 https://aopwiki.org/aops/18

Page 41: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

40

Proposed AOP for in utero CYP26B1 inhibition effects on male fertility 1392 As described in sections 5 and 6, temporally and spatially regulated RA concentrations in the fetal 1393 gonads appear to play a role both in the proper development of the testis and in the differentiation 1394 of PGCs into either oocytes or spermatogonia. In the male fetal testis, perturbed RA catabolism 1395 caused by inhibition of CYP26B1 (putative MIE) could disrupt the normal RA signaling pathway, which 1396 could lead to the AO impaired male fertility (Figure 10). 1397 1398 Following inhibition of the CYP26B1 enzyme or complete knockout of the cyp26b1 gene in the male 1399 mouse fetus, aberrant meiotic and apoptotic GC are observed in the testis (Bowles et al. 2006, 1400 MacLean et al. 2007, Teletin et al. 2017), and virtually no germ cells are present in testes from 1401 neonatal pups. In vitro, CYP26 inhibition prevents the normal mitotic arrest of the male GC and 1402 induces their death by apoptosis (Teletin et al. 2017). Male cyp26b1-/- homozygote 13.5 dpc mouse 1403 embryos display a mild ovotestis phenotype, with an “ovarian component” at the anterior end of the 1404 gonad which is where RA levels are expected to be higher due to the connection to the mesonephric 1405 tubules at this end (Bowles et al. 2018). In addition, abnormal development of the Leydig cells and of 1406 the Müllerian and Wolffian ducts was observed in the same male mouse embryos (Bowles et al. 1407 2018). Thus, it is possible that the (observed) effects on the germ cells and on testis development 1408 may lead to (possible) adverse effects on spermatogenesis and malfunctioning testes, which could 1409 eventually lead to (possible) impaired male fertility (see figure 10 below). 1410 1411 1412 1413

1414 1415 Figure 8: Proposed AOP for how CYP26B1 inhibition in the fetal male testis may lead to impaired 1416 male infertility. Studies looking at CYP26B1 inhibition were using deletion of the gene rather than 1417 chemical inhibition of the enzyme. 1418 1419 1420

Page 42: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

41

Proposed AOP for effects of RDH10/RALDH inhibition in the adult on male fertility 1421 Based on the critical roles of RA in spermatogenesis in the adult male described in section 8, it seems 1422 possible that chemicals that inhibits the enzymes that produce RA (putative MIE), via decreased RA 1423 levels, may cause adverse effects on the spermatogenic process leading to the AO impaired male 1424 fertility (see figure 11 below). 1425 1426 In juvenile male mice deficient in RDH10 in Sertoli cells and germ cells (leading to a local lack of RA, 1427 observed via effects Stra8 expression levels and by using a RA-responsive reporter cell system), 1428 spermatogenesis is blocked (Tong et al. 2013). Also in juvenile mice, the differentiation of A aligned 1429 to A1 spermatogonia has been shown to be dependent on RA synthesis by RALDH in Sertoli cells 1430 (Raverdeau et al. 2012). At least in the mouse, RA is also required to disengage spermatozoa from 1431 the Sertoli cell cytoplasm during spermiation (Spiller and Bowles 2015, Teletin et al. 2017). 1432 1433 1434 1435

1436 1437 1438 Figure 9: Proposed AOP for how RDH10 or RALDH inhibition in the adult male testis may lead to 1439 impaired male infertility. Studies looking at RDH10 or RALDH inhibition were using deletion of the 1440 genes rather than chemical inhibition of the enzymes. 1441 1442 1443 Although no such data have been retrieved, it also seems possible that decreased RA testis levels via 1444 chemically-induced CYP26B1 inhibition (putative MIE) could lead to similar adverse effects on 1445 spermatogenesis. 1446 1447

Page 43: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

42

Proposed AOP for meiosis initiation, oogenesis, folliculogenesis and female fertility, 1448

focusing on Stra8 1449 The role for RA for meiosis initiation, oogenesis and folliculogenesis in the female fetus is discussed 1450 in section 7. In female fetal offspring of VAD rats, a delay or failure of meiosis initiation was observed 1451 in oogonia (Li and Clagett-Dame 2009). Although RA levels were not measured, the low Stra8 1452 expression suggest lower than normal RA levels in the ovaries. The critical role of Stra8 in meiosis, 1453 oogenesis and follicular development is also evident in Stra8-/- female mice, which are infertile and 1454 have smaller ovaries with no oocytes or follicles (heterozygotes are fertile) (Baltus et al. 2006). 1455 Animal experiments with BPA and DEHP have connected lack of increased Stra8 expression with 1456 impaired female fertility; however, the effects of BPA (Zhang et al. 2012) and DEHP (Zhang et al. 1457 2015) appear to by-pass the retinoid system and instead affect Stra8 expression via epigenetic 1458 mechanisms. 1459 1460 1461 1462

1463 1464 1465 Figure 10: Proposed AOP for absence of ovarian Stra8 induction in utero possibly leading to impaired 1466 female fertility. 1467 1468 1469

Page 44: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

43

Proposed AOP for endometriosis in the adult female, focusing on CYP26A1 1470 In endometrial tissue, RA appears to inhibit the decidualization of stromal cells, which is a 1471 prerequisite for blastocyst implantation after fertilization; thus, reduced concentration of RA in the 1472 endometrial tissue could be necessary for successful implantation (Deng et al. 2013). As discussed in 1473 section 7, the RA-catabolizing enzyme in the endometrium is CYP26A1, and its expression increases 1474 during the endometrial secretory phase (lowering the RA levels) when compared to the proliferative 1475 phase, during which RA-synthesizing enzymes such as RALDHs were increased (Deng et al. 2003). 1476 Expression of CYP26A1 is down-regulated in both the secretory and proliferative phases in 1477 endometrial biopsies from women with moderate or severe endometriosis, when compared to 1478 healthy women (Burney et al. 2007). The availability of RA may therefore be increased in the 1479 endometrial tissue of women with endometriosis. 1480 1481 1482 1483

1484 1485 Figure 11: Proposed AOP for lack of CYP26A1 increase in the adult female endometrium possibly 1486 leading to impaired female fertility. 1487 1488

Page 45: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

44

11. Initial scoping effort: assays and endpoints for effects of chemicals on 1489

female and male reproduction via the retinoid system 1490

1491 Several existing OECD test guidelines measure endpoints relevant to the estrogen, androgen, 1492 steroidogenesis, and thyroid hormone pathways, but currently, no OECD test guidelines include 1493 endpoints specifically indicative of retinoid system modulation. Some reproductive parameters 1494 already included in existing test guidelines, would also provide information on adverse effects 1495 stemming from retinoid disruption. However, due to extensive cross-talk with other pathways, there 1496 is no endpoint identified that will give information specifically on disruption of retinoid signaling. The 1497 information assembled in this report regarding the role of retinoids in female and male reproduction 1498 indicates possible in vitro/ex vivo assays that could associate adverse reproductive outcomes to 1499 disruption of retinoid signaling. 1500 1501 Some of the proposed assays are suitable for screening larger numbers of chemicals (e.g. in silico/in 1502 vitro, OECD Conceptual Framework [CF] levels 1, 2), while in vivo test methods address more 1503 complex mechanistic and organ system effects (CF level 3, 4, 5). One other option could be to include 1504 ex vivo models. In order to correctly interpret positive results in lower-tier models, the ability of a 1505 substance to reach reproductive target cells/tissues during the relevant developmental window must 1506 be evaluated. 1507 1508

In silico methods 1509 In silico methods, such as molecular docking models and quantitative structure-activity relationship 1510 (QSAR) models for binding to e.g. CYP26, RALDH, Stra8, RAR and RXR, could, if available, be 1511 integrated into CF Level 1. A quantitative AOP has been developed for craniofacial malformations 1512 caused by azole fungicides, acting as CYP26 inhibitors (Battistoni et al. 2019). Also, homology 1513 modeling and computational docking simulations for CYP26A1 and CYP26B1 have been performed to 1514 identify inhibitors for these enzymes (Foti et al. 2016). Several studies describe QSAR models for 1515 binding to and activating e.g. Pregnane X receptor (PXR; an RXRα-heterodimerizing partner) and 1516 CYP3A4 (Rosenberg et al. 2017b), thyroperoxidase (TPO; Rosenberg et al. 2017a) and AhR (Klimenko 1517 et al. 2019). Similar models for e.g. RARs and RXRs would be valuable, and should be developed. 1518 1519

In vitro assays and ex vivo methods 1520 Temporal and spatial regulation of local RA is critical for normal reproductive development. Thus, in 1521 addition to nuclear receptor activation assays, assays measuring expression and/or activity of the 1522 enzymes involved in the metabolism of RA can be considered as candidate in vitro assays (Piersma et 1523 al. 2017; see also section 10). 1524 1525 Considering the complex RA modulation of both female and male reproduction, a panel of in vitro 1526 assays with the following endpoints is suggested: 1527 1528

CYP26A1, CYP26B1 (induction, inhibition) 1529

RALDH (induction, inhibition) 1530

STRA8 (induction) 1531

RAR/RXR transactivation 1532 1533 The proposed top candidate enzyme for fetal exposure to chemicals is the RA-catalyzing CYP26B1 1534 enzyme. Inhibition of CYP26B1 in the fetal testis (leading to increased RA levels) would cause serious 1535 adverse effects on normal development of testicular somatic and germ cells, while induction of the 1536 CYP26B1 enzyme in the female fetus could potentially lead to decreased RA tissue levels and 1537 subsequent adverse effects on initiation of germ cell meiosis. For evaluating adult females, CYP26A1 1538 is the more relevant enzyme, considering its role in the endometrium. 1539 1540

Page 46: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

45

Inhibition of the RA-synthesizing RALDH enzymes could decrease RA levels, and assays measuring 1541 RALDH inhibition may therefore be considered for screening purposes. In the postnatal male, RALDH 1542 inhibition and the subsequent decrease in RA levels would interfere with spermatogenesis. 1543 1544 Stra8 is regulated by RA, and is expressed only in germ cells (Oulad-Abdelghani et al. 1996, Mark et 1545 al. 2008). Stra8 is required for germ cells to enter meiosis (Baltus et al. 2006). Therefore, an in vitro 1546 screening assay for Stra8 expression could be used to identify substances that, through disturbed RA 1547 signaling, affect meiosis initiation. 1548 1549 The different RAR and RXR transcription factors mediate the retinoid signal when activated by RA or 1550 9-cis-RA (RXRs can also act as a silent partner, i.e. no RXR ligand is necessary for RXR to bind and 1551 active genes). Activation or inactivation of these transcription factors by chemicals could lead to 1552 different effects on the cellular level, which may be translated to effects on tissue/organ level. 1553 1554 For any in vitro assay, the choice of cellular system is important in terms of both relevance and 1555 practicality. Ovarian stem cell test systems have been described, in which pre-meiotic oogonia in the 1556 post-natal ovary are used (Bhartiya and Patel 2018). This type of cells could theoretically be used to 1557 evaluate meiotic initiation in vitro. For screening large numbers of chemicals, gonadal cell or tissue 1558 models (preferably of human origin) would probably be necessary. Given the sex-specific effects of 1559 RA pathways, using two model systems in parallel (ovarian/follicular and testicular) is recommended. 1560 Any in vitro model should be characterized in terms of expression and function of e.g. CYP26, RoDH, 1561 and RALDH). 1562 1563 Ex vivo model systems consisting of rodent follicular cells or whole rodent neonatal ovaries (Hannon 1564 and Flaws 2015) could generate valuable information on chemical disruption of the possible 1565 association between RA and the formation and maturation of follicles (Minkina et al. 2017, 1566 Damdimopoulou et al. 2019). Only pre-meoitic germ cells, which can be obtained from fetal rodent 1567 ovaries and cultured in vitro (Liu, Paczkowski et al. 2012) can provide information on chemical effects 1568 on RA-mediated meiotic induction. Both human and rat fetal testis cultures have been used to study 1569 the effects of e.g. hormone disrupting chemicals on the development of human fetal testis (Lambrot 1570 et al. 2006, Spade et al. 2018b). 1571 1572

In vivo endpoints 1573 Expression measurements of e.g. CYP26, RoDH, RALDH, and Stra8 using immunohistochemistry or in 1574 situ hybridization could be performed on tissues from in vivo studies, for obtaining information on 1575 the mechanistic level. 1576 1577 The detailed histopathological (morphology) examination of reproductive organs/organ structures 1578 that are part of animal studies performed according to OECD reproductive development guideline 1579 studies could add valuable information when evaluating a possible retinoid-disrupting impact of the 1580 test chemical on these organs (Table 2 and 3; CF Level 3 or 4). Serum levels of AMH, as a proxy for 1581 follicle counting, could potentially be evaluated. 1582 1583

Page 47: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

Table 2. Current OECD TG endpoints which might capture possible retinoid effects on mammalian female reproductive health, at CF level 4 and 5 (adapted 1584 from the draft DRP). Also indicated are limitations and data gaps. 1585 1586

Retinoid effects OECD Test Guidelines (TG) Comments

Reproductive development (female rodents)

Oogenesis,

follicular count

Extended one-generation reproductive toxicity study (TG 443); 2-Generation reproduction toxicity study (TG 416).

TG 443: Quantitative (most sensitive). Histopathological examination should be aimed at detecting a quantitative evaluation of primordial and small growing follicles, as well as corpora lutea, in F1 females. TG 416: only qualitative (limited sensitivity). A quantitative evaluation of primordial follicles should be conducted for F1 females.

Oestrus cycles Extended one-generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update).

TG 443: Vaginal smears should be examined daily for all F1 females in cohort 1A, after the onset of vaginal patency, until the first cornified smear is recorded, in order to determine the time interval between these two events. Oestrous cycles for all F1 females in cohort 1A should also be monitored for a period of two weeks, commencing around PND 75. TG 421/422: Not included in the offspring and also not possible due to, termination of the offspring on PND 14, i.e. before puberty. TG 416: Oestrous cycle length and normality are evaluated in F1 females by vaginal smears prior to mating, and optionally during mating, until evidence of mating is found.

Vagina, uterus

with cervix, and

ovaries

Extended one-generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update).

TG 443: Uterus (with oviducts and cervix), ovaries will be weighed (F1). Full histopathology is performed for all high-dose and control F1 animals. All litters should be represented by at least 1 pup per sex. Organs and tissues demonstrating treatment-related changes and all gross lesions should also be examined in all animals in the lower dose groups to aid in determining a NOAEL. TG 421/422: Not included and not possible due to termination on PND 14 (F1). TG 416: Vagina, uterus with cervix, and ovaries (preserved for histopathology (parental F1 animals) determining a NOAEL. TG 421/422: Not included and not possible due to termination on PND 14, although follicular counts can be made at this age (F1). TG 416: Vagina, uterus with cervix, and ovaries (preserved for histopathology (parental F1 animals).

Page 48: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

Adult exposure (female rodents)

Vagina, uterus with cervix and

ovaries (histopathology)

Extended one-generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update).

TG 443: Uterus (with oviducts and cervix), ovaries will be weighed. Full histopathology is performed for all high- dose and control P animals. Organs demonstrating treatment-related changes should also be examined in all animals at the lower dose groups to aid in determining a NOAEL. Additionally, reproductive organs of all animals suspected of reduced fertility should be subjected to histopathological evaluation. TG 421/422: optional: paired ovaries (wet weight) and uterus (including cervix) in females (P). TG 416: Examination of the ovaries of the P animals is optional.

Oestrus cycles Extended one-generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update).

TG 443: Normally the assessment of oestrous cyclicity (by vaginal cytology) will start at the beginning of the treatment period and continue until confirmation of mating or the end of the 2-week mating period. TGs 421/422: Oestrous cycles should be monitored before treatment to select study females with regular cyclicity. Vaginal smears should also be monitored daily from the beginning of the treatment period until evidence of mating. TG 416: Oestrous cycle length and normality are evaluated in P females by vaginal smears prior to mating, and optionally during mating, until evidence of mating is found.

1587 1588

Page 49: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

Table 3. Current OECD TG endpoints which might capture possible retinoid effects on mammalian male reproductive health, at CF level 4 and 5. Also indicated 1589 are limitations and data gaps (adapted from the draft DRP). 1590 1591

Retinoid effects OECD Test Guidelines (TG) Comments

Reproductive development (male rodents)

Hypospadias, dysgenesis of

external reproductive

organs, cryptorchidism

Extended one-generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update); Prenatal Developmental Toxicity Study (TG 414).

TG443: Most sensitive design due to 20 litters per group, 2 or more males per litter and termination after puberty. TG 421/422: Limited sensitivity due to limited group size (n~8-10) and termination before puberty (hypospadia should be detectable at birth). TG416: Sensitive design due to 20 litters per group and termination after puberty, but only 1 male per litter decreases the sensitivity compared to TG443. TG 414: Sensitive design due to 20 litters per group, but limited/unknown sensitivity as the offspring is terminated before birth (although e.g. hypospadia should still be detectable).

Testes development (weight and

histopathology)

Extended one-generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update).

TG443: Most sensitive design due to 20 litters per group and 2 adult male offspring per litter. TG 421/422: Not included but could be assessed at termination on PND 14. Limited sensitivity due to limited group size (n~8-10) and termination well before puberty. TG416: Sensitive design due to 20 litters per group and termination after puberty, but only 1 male per litter decreases the sensitivity compared to TG443.

Page 50: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

Spermatogenesis (sperm quality and testicular

histology

Extended one-generation reproductive

toxicity study (TG 443);

Reproductive screening test (TG 421);

Combined 28- day/reproductive screening

assay (TG 422);

2-Generation reproduction toxicity study (TG

416 most recent update).

TG443: sensitive design due to 20 litters per group, 1 adult male offspring per litter examined. TG 421/422: Not included and not possible due to termination on PND 14. TG416: Sensitive design due to 20 litters per group, 1 adult male offspring per litter examined (only included in the most recent update in 2001).

Adult exposure (male rodents)

Spermatogenesis (sperm quality,

testis weight and testicular

histopathology)

Extended one- generation reproductive toxicity study (TG 443); Reproductive screening test (TG 421); Combined 28- day/reproductive screening assay (TG 422); 2-Generation reproduction toxicity study (TG 416 most recent update);, Repeated Dose 90-day Oral Toxicity Study in Rodents (TG 408); Repeated Dose 28-Day Oral Toxicity Study in Rodents (TG 407).

TG 443: Sperm parameters assessed in around 20 parental males unless there is existing data to show that sperm parameters are unaffected in a 90-day study. TG 421/422: Limited sensitivity due to limited group size (n~8-10) and assessment of sperm quality is not required; TG416: Sperm parameters assessed in around 20 parental males (sperm quality only included in the most recent update in 2001). TG 408, 90-day study: Limited sensitivity due to limited group size (n~10) and assessment of sperm quality is not required. TG407, 28 day study: Very limited sensitivity due to limited group size (n~5), assessment of epididymal sperm parameters are optional.

1592

1593

Page 51: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

50

12. Concluding remarks 1594

1595 Potential endpoints for studying effects on female and male reproduction caused by chemical 1596 exposure, possibly affecting the retinoid pathway, have been identified and is described in this 1597 report. 1598 1599 In silico methods, such as QSAR and molecular docking models for e.g. CYP26, RALDH, Stra8, RAR, 1600 RXR could be integrated into OECD CF Level 1. A small number of in silico tools for retinoid signaling 1601 pathway (all focusing on CYP26) have been identified (Battistoni et al. 2019, Foti et al. 2016). 1602 Multiple such tools exist for other signaling pathways (Rosenberg et al. 2017b, Rosenberg et al. 1603 2017a, Klimenko et al. 2019), which suggests that they could be developed also for the retinoid 1604 pathway. 1605 1606 For in vitro assays, there are commercial as well as governmental/academic assays that measure 1607 retinoid-relevant endpoints such as RAR/RXR activation (see section 14). For others (e.g. CYP26, 1608 Stra8, RALDH), assays suitable for integration into OECD CF Level 2 remain to be developed (see 1609 section 14 for more information). The CYP26 and RALDH isomers appear to be the best candidates at 1610 present, since they play a critical role in regulating RA concentrations in several reproductive tissues 1611 in both males and females. Also, CYP26 isomer expression and activity appears to be well studied in 1612 many labs, suggesting that these assays might be mature enough for further development. However, 1613 a more comprehensive analysis has to be performed in order to identify the most suitable candidate 1614 assay. 1615 1616 Regarding in vivo models (current mammalian OECD TG studies), no retinoid-relevant endpoints that 1617 could be added to already existing test methods has been identified, especially for TG443 which has 1618 the most sensitive design. One exception could be to evaluate the value of addition of ovary 1619 histopathology of pups in TGs 421/422. Certain already existing endpoints could be informative 1620 regarding retinoid disruption. For females ovaries and follicles are of particular interest, and for 1621 males sperm parameters. However, we have not identified any endpoints that would provide 1622 information specifically on retinoid disruption, since the control of male and female reproduction 1623 involves many other pathways. 1624 1625 Several RA-targeted drugs have been demonstrated to affect reproductive function while also 1626 interfering with retinoid homeostasis. However, no single chemical compound has been identified 1627 that is known to affect female or male reproduction specifically via perturbation of the retinoid 1628 pathway(s). This means that validation of the potential endpoints or assays described in this report, 1629 with regard specifically to retinoid disruption, will be challenging. 1630 1631

1632

Page 52: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

51

13. References 1633

1634 Abali, R., Yuksel, M. A., Aktas, C., Celik, C., Guzel, S., Erfan, G., & Sahin, O. (2013). Decreased ovarian reserve in 1635

female Sprague–Dawley rats induced by isotretinoin (retinoic acid) exposure. Reproductive 1636 BioMedicine Online, 27(2), 184-191. doi:10.1016/j.rbmo.2013.04.010 1637

Acmaz, G., Cmar, L., Acmaz, B., Aksoy, H., Kafadar, Y. T., Madendag, Y., . . . Muderris, I. (2019). The Effects of Oral 1638 Isotretinoin in Women with Acne and Polycystic Ovary Syndrome. BioMed Research International, 2019, 5. 1639 doi:10.1155/2019/2513067 1640

Agrimson, K. S., Oatley, M. J., Mitchell, D., Oatley, J. M., Griswold, M. D., & Hogarth, C. A. (2017). Retinoic acid 1641 deficiency leads to an increase in spermatogonial stem number in the neonatal mouse testis, but excess 1642 retinoic acid results in no change. Developmental Biology, 432(2), 229-236. 1643 doi:https://doi.org/10.1016/j.ydbio.2017.10.002 1644

Aikawa, H., Koyama, S., Matsuda, M., Nakahashi, K., Akazome, Y., & Mori, T. (2004). Relief effect of vitamin A on the 1645 decreased motility of sperm and the increased incidence of malformed sperm in mice exposed neonatally 1646 to bisphenol A. Cell and Tissue Research, 315(1), 119-124. doi:10.1007/s00441-003-0806-1 1647

Aksoy, H., Cinar, L., Acmaz, G., Aksoy, U., Aydin, T., Vurdem, U. E., . . . Kartal, D. (2015). The Effect of Isotretinoin on 1648 Ovarian Reserve Based on Hormonal Parameters, Ovarian Volume, and Antral Follicle Count in Women 1649 with Acne. Gynecologic and Obstetric Investigation, 79(2), 78-82. doi:10.1159/000371551 1650

Al Tanoury, Z., Piskunov, A., & Rochette-Egly, C. (2013). Vitamin A and retinoid signaling: genomic and 1651 nongenomic effects. Journal of Lipid Research, 54(7), 1761-1775. doi:10.1194/jlr.R030833 1652

Allenby, G., Bocquel, M. T., Saunders, M., Kazmer, S., Speck, J., Rosenberger, M., . . . Chambon, P. (1993). Retinoic 1653 acid receptors and retinoid X receptors: interactions with endogenous retinoic acids. Proceedings of the 1654 National Academy of Sciences of the United States of America, 90(1), 30. doi:10.1073/pnas.90.1.30 1655

Alnouti, Y., & Klaassen, C. D. (2008). Tissue Distribution, Ontogeny, and Regulation of Aldehyde Dehydrogenase 1656 (Aldh) Enzymes mRNA by Prototypical Microsomal Enzyme Inducers in Mice. Toxicological Sciences, 1657 101(1), 51-64. doi:10.1093/toxsci/kfm280 1658

Amory, J. K., Ostrowski, K. A., Gannon, J. R., Berkseth, K., Stevison, F., Isoherranen, N., . . . Walsh, T. (2017). 1659 Isotretinoin administration improves sperm production in men with infertility from 1660 oligoasthenozoospermia: a pilot study. Andrology, 5(6), 1115-1123. doi:10.1111/andr.12420 1661

André, A., Ruivo, R., Gesto, M., Castro, L. F. C., & Santos, M. M. (2014). Retinoid metabolism in invertebrates: When 1662 evolution meets endocrine disruption. General and Comparative Endocrinology, 208, 134-145. 1663 doi:https://doi.org/10.1016/j.ygcen.2014.08.005 1664

Aoki, T., & Takada, T. (2012). Bisphenol A modulates germ cell differentiation and retinoic acid signaling in mouse 1665 ES cells. Reproductive Toxicology, 34(3), 463-470. doi:https://doi.org/10.1016/j.reprotox.2012.06.001 1666

Arnold, S. L., Kent, T., Hogarth, C. A., Schlatt, S., Prasad, B., Haenisch, M., . . . Isoherranen, N. (2015). Importance of 1667 ALDH1A enzymes in determining human testicular retinoic acid concentrations. Journal of Lipid Research, 1668 56(2), 342-357. doi:10.1194/jlr.M054718 1669

Azaïs-Braesco, V., & Pascal, G. (2000). Vitamin A in pregnancy: requirements and safety limits. The American 1670 Journal of Clinical Nutrition, 71(5), 1325S-1333S. doi:10.1093/ajcn/71.5.1325s 1671

Bagavandoss, P., & Midgley, A. R. J. (1987). Lack of Difference Between Retinoic Acid and Retinol in Stimulating 1672 Progesterone Production by Luteinizing Granulosa Cells in Vitro. Endocrinology, 121(1), 420-428. 1673 doi:10.1210/endo-121-1-420 1674

Baker, N., Boobis, A., Burgoon, L., Carney, E., Currie, R., Fritsche, E., . . . Daston, G. (2018). Building a developmental 1675 toxicity ontology. Birth Defects Res. doi:10.1002/bdr2.1189 1676

Balmer, J. E., & Blomhoff, R. (2002). Gene expression regulation by retinoic acid. Journal of Lipid Research, 43(11), 1677 1773-1808. doi:10.1194/jlr.R100015-JLR200 1678

Baltus, A. E., Menke, D. B., Hu, Y.-C., Goodheart, M. L., Carpenter, A. E., de Rooij, D. G., & Page, D. C. (2006). In germ 1679 cells of mouse embryonic ovaries, the decision to enter meiosis precedes premeiotic DNA replication. 1680 Nature Genetics, 38, 1430. doi:10.1038/ng1919 1681

Barnett, K. R., Schilling, C., Greenfeld, C. R., Tomic, D., & Flaws, J. A. (2006). Ovarian follicle development and 1682 transgenic mouse models. Human Reproduction Update, 12(5), 537-555. doi:10.1093/humupd/dml022 1683

Bastos Maia, S., Rolland Souza, A. S., Costa Caminha, M. d. F., Lins da Silva, S., Callou Cruz, R. d. S. B. L., Carvalho 1684 Dos Santos, C., & Batista Filho, M. (2019). Vitamin A and Pregnancy: A Narrative Review. Nutrients, 11(3), 1685 681. doi:10.3390/nu11030681 1686

Battistoni, M., Di Renzo, F., Menegola, E., & Bois, F. Y. (2019). Quantitative AOP based teratogenicity prediction for 1687 mixtures of azole fungicides. Computational Toxicology, 11, 72-81. 1688 doi:https://doi.org/10.1016/j.comtox.2019.03.004 1689

Bay, K., Main, K. M., Toppari, J., & Skakkebæk, N. E. (2011). Testicular descent: INSL3, testosterone, genes and the 1690

Page 53: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

52

intrauterine milieu. Nature Reviews Urology, 8, 187. doi:10.1038/nrurol.2011.23 1691 Beedle, M.-T., Stevison, F., Zhong, G., Topping, T., Hogarth, C., Isoherranen, N., & Griswold, M. D. (2018). Sources of 1692

all-trans retinal oxidation independent of the aldehyde dehydrogenase 1A isozymes exist in the postnatal 1693 testis†. Biology of Reproduction, 100(2), 547-560. doi:10.1093/biolre/ioy200 1694

Belyaeva, O. V., Wu, L., Shmarakov, I., Nelson, P. S., & Kedishvili, N. Y. (2018). Retinol dehydrogenase 11 is essential 1695 for the maintenance of retinol homeostasis in liver and testis in mice. The Journal of biological chemistry, 1696 293(18), 6996-7007. doi:10.1074/jbc.RA117.001646 1697

Best, M. W., Wu, J., Pauli, S. A., Kane, M. A., Pierzchalski, K., Session, D. R., . . . Sidell, N. (2015). A role for retinoids in 1698 human oocyte fertilization: regulation of connexin 43 by retinoic acid in cumulus granulosa cells. 1699 Molecular Human Reproduction, 21(6), 527-534. doi:10.1093/molehr/gav017 1700

Bhartiya, D., & Patel, H. (2018). Ovarian stem cells—resolving controversies. Journal of Assisted Reproduction and 1701 Genetics, 35(3), 393-398. doi:10.1007/s10815-017-1080-6 1702

Bittman, E. L. (2015). Timing in the Testis. Journal of Biological Rhythms, 31(1), 12-36. 1703 doi:10.1177/0748730415618297 1704

Blaner, W. S., & Li, Y. (2015). Vitamin A Metabolism, Storage and Tissue Delivery Mechanisms. In P. Dollé & K. 1705 Neiderreither (Eds.), The Retinoids: Biology, Biochemistry and Disease (pp. 1-34). 1706

Blomhoff, R., Helgerud, P., Rasmussen, M., Berg, T., & Norum, K. R. (1982). In vivo uptake of chylomicron 1707 [3H]retinyl ester by rat liver: evidence for retinol transfer from parenchymal to nonparenchymal cells. 1708 Proceedings of the National Academy of Sciences of the United States of America, 79(23), 7326. 1709 doi:10.1073/pnas.79.23.7326 1710

Bosakowski, T., Levin, A. A., & Durham, S. K. (1991). Time course of testicular degeneration in rats induced by a 1711 synthetic retinoid (Ro 23-2895) and evidence for induction of hypovitaminosis A in the testes. Toxicology, 1712 66(1), 105-118. doi:10.1016/0300-483X(91)90182-Z 1713

Bowles, J., Feng, C.-W., Ineson, J., Miles, K., Spiller, C. M., Harley, V. R., . . . Koopman, P. (2018). Retinoic Acid 1714 Antagonizes Testis Development in Mice. Cell Reports, 24(5), 1330-1341. 1715 doi:https://doi.org/10.1016/j.celrep.2018.06.111 1716

Bowles, J., Feng, C.-W., Miles, K., Ineson, J., Spiller, C., & Koopman, P. (2016). ALDH1A1 provides a source of 1717 meiosis-inducing retinoic acid in mouse fetal ovaries. Nature Communications, 7, 10845. 1718 doi:10.1038/ncomms10845 1719

Bowles, J., Knight, D., Smith, C., Wilhelm, D., Richman, J., Mamiya, S., . . . Koopman, P. (2006). Retinoid Signaling 1720 Determines Germ Cell Fate in Mice. Science, 312(5773), 596. 1721

Brieño-Enríquez, M. A., Reig-Viader, R., Cabero, L., Toran, N., Martínez, F., Roig, I., & Garcia Caldés, M. (2012). Gene 1722 expression is altered after bisphenol A exposure in human fetal oocytes in vitro. MHR: Basic science of 1723 reproductive medicine, 18(4), 171-183. doi:10.1093/molehr/gar074 1724

Bruner-Tran, K. L., & Osteen, K. G. (2011). Developmental Exposure to TCDD Reduces Fertility and Negatively 1725 Affects Pregnancy Outcomes across Multiple Generations. Reproductive toxicology (Elmsford, N.Y.), 31(3), 1726 344-350. doi:10.1016/j.reprotox.2010.10.003 1727

Bryant, S. L., Francis, J. C., Lokody, I. B., Wang, H., Risbridger, G. P., Loveland, K. L., & Swain, A. (2014). Sex specific 1728 retinoic acid signaling is required for the initiation of urogenital sinus bud development. Developmental 1729 Biology, 395(2), 209-217. doi:10.1016/j.ydbio.2014.09.016 1730

Bryson, H. M., & Wagstaff, A. J. (1996). Liarozole. Drugs & Aging, 9(6), 478-484. doi:10.2165/00002512-199609060-1731 00010 1732

Burney, R. O., Talbi, S., Hamilton, A. E., Vo, K. C., Nyegaard, M., Nezhat, C. R., . . . Giudice, L. C. (2007). Gene 1733 Expression Analysis of Endometrium Reveals Progesterone Resistance and Candidate Susceptibility Genes 1734 in Women with Endometriosis. Endocrinology, 148(8), 3814-3826. doi:10.1210/en.2006-1692 1735

Busada, J. T., & Geyer, C. B. (2016). The Role of Retinoic Acid (RA) in Spermatogonial Differentiation1. Biology of 1736 Reproduction, 94(1), 10, 11-10-10, 11-10. doi:10.1095/biolreprod.115.135145 1737

Chambon, P. (1996). A decade of molecular biology of retinoic acid receptors. FASEB journal : official publication of 1738 the Federation of American Societies for Experimental Biology, 10(9), 940. doi:10.1096/fasebj.10.9.8801176 1739

Chen, P.-J., Padgett, W. T., Moore, T., Winnik, W., Lambert, G. R., Thai, S.-F., . . . Nesnow, S. (2009). Three conazoles 1740 increase hepatic microsomal retinoic acid metabolism and decrease mouse hepatic retinoic acid levels in 1741 vivo. Toxicology and Applied Pharmacology, 234(2), 143-155. 1742 doi:https://doi.org/10.1016/j.taap.2008.10.004 1743

Chen, Y., Ma, L., Hogarth, C., Wei, G., Griswold, M. D., & Tong, M.-H. (2016). Retinoid signaling controls 1744 spermatogonial differentiation by regulating expression of replication-dependent core histone genes. 1745 Development, 143(9), 1502. doi:10.1242/dev.135939 1746

Chen, Y., & Reese, D. H. (2013). A screen for disruptors of the retinol (vitamin A) signaling pathway. Birth Defects 1747 Res B Dev Reprod Toxicol, 98(3), 276-282. doi:10.1002/bdrb.21062 1748

Childs, A. J., Cowan, G., Kinnell, H. L., Anderson, R. A., & Saunders, P. T. K. (2011). Retinoic Acid Signalling and the 1749

Page 54: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

53

Control of Meiotic Entry in the Human Fetal Gonad. PLOS ONE, 6(6), e20249. 1750 doi:10.1371/journal.pone.0020249 1751

Chlapek, P., Slavikova, V., Mazanek, P., Sterba, J., & Veselska, R. (2018). Why Differentiation Therapy Sometimes 1752 Fails: Molecular Mechanisms of Resistance to Retinoids. International Journal of Molecular Sciences, 19(1), 1753 132. doi:10.3390/ijms19010132 1754

Chung, S. S. W., Wang, X., & Wolgemuth, D. J. (2016). Prolonged Oral Administration of a Pan-Retinoic Acid 1755 Receptor Antagonist Inhibits Spermatogenesis in Mice With a Rapid Recovery and Changes in the 1756 Expression of Influx and Efflux Transporters. Endocrinology, 157(4), 1601-1612. doi:10.1210/en.2015-1675 1757

Çinar, L., Kartal, D., Ergin, C., Aksoy, H., Karadag, M. A., Aydin, T., . . . Borlu, M. (2016). The effect of systemic 1758 isotretinoin on male fertility. Cutaneous and Ocular Toxicology, 35(4), 296-299. 1759 doi:10.3109/15569527.2015.1119839 1760

Cione, E., Maria Cristina, C., Roberto, C., Mariarita, P., Attilio, P., & Giuseppe, G. (2016). Vitamin A and Diabesity: 1761 New Insight for Drug Discovery. Mini-Reviews in Medicinal Chemistry, 16(9), 738-742. 1762 doi:http://dx.doi.org/10.2174/1389557515666150709112822 1763

Clagett-Dame, M., & Knutson, D. (2011). Vitamin A in reproduction and development. Nutrients, 3(4), 385-428. 1764 doi:10.3390/nu3040385 1765

Clermont, Y. (1963). The cycle of the seminiferous epithelium in man. The American journal of anatomy, 112, 35. 1766 Coward, W. A., Howell, J. M., Thompson, J. N., & Pitt, G. A. J. (1969). The retinol requirements of rats for 1767

spermatogenesis and vision. British Journal of Nutrition, 23(3), 619-626. doi:10.1079/BJN19690069 1768 de Araújo, J. F. P., Podratz, P. L., Merlo, E., Sarmento, I. V., da Costa, C. S., Niño, O. M. S., . . . Graceli, J. B. (2018). 1769

Organotin Exposure and Vertebrate Reproduction: A Review. Frontiers in Endocrinology, 9(64). 1770 doi:10.3389/fendo.2018.00064 1771

de Lera, Á. R., Krezel, W., & Rühl, R. (2016). An Endogenous Mammalian Retinoid X Receptor Ligand, At Last! 1772 ChemMedChem, 11(10), 1027-1037. doi:10.1002/cmdc.201600105 1773

Dean, A., van den Driesche, S., Wang, Y., McKinnell, C., Macpherson, S., Eddie, S. L., . . . Sharpe, R. M. (2016). 1774 Analgesic exposure in pregnant rats affects fetal germ cell development with inter-generational 1775 reproductive consequences. Scientific Reports, 6, 19789. doi:10.1038/srep19789 1776

Demczuk, M., Huang, H., White, C., & Kipp, J. L. (2016). Retinoic Acid Regulates Calcium Signaling to Promote 1777 Mouse Ovarian Granulosa Cell Proliferation1. Biology of Reproduction, 95(3), 70, 71-10-70, 71-10. 1778 doi:10.1095/biolreprod.115.136986 1779

Deng, L., Shipley, G. L., Loose-Mitchell, D. S., Stancel, G. M., Broaddus, R., Pickar, J. H., & Davies, P. J. A. (2003). 1780 Coordinate Regulation of the Production and Signaling of Retinoic Acid by Estrogen in the Human 1781 Endometrium. The Journal of Clinical Endocrinology & Metabolism, 88(5), 2157-2163. doi:10.1210/jc.2002-1782 021844 1783

Denis, L., Debruyne, F., De Porre, P., & Bruynseels, J. (1998). Early clinical experience with liarozole 1784 (Liazal<sup>&#x2122;</sup>) in patients with progressive prostate cancer. European Journal of Cancer, 1785 34(4), 469-475. doi:10.1016/S0959-8049(97)10120-4 1786

Di Renzo, F., Metruccio, F., Battistoni, M., Moretto, A., & Menegola, E. (2019). Relative potency ranking of azoles 1787 altering craniofacial morphogenesis in rats: An in vitro data modelling approach. Food and Chemical 1788 Toxicology, 123, 553-560. doi:https://doi.org/10.1016/j.fct.2018.12.004 1789

El Shahawy, M., Reibring, C.-G., Hallberg, K., Neben, C. L., Marangoni, P., Harfe, B. D., . . . Gritli-Linde, A. (2019). 1790 Sonic Hedgehog Signaling Is Required for Cyp26 Expression during Embryonic Development. 1791 International Journal of Molecular Sciences, 20(9), 2275. 1792

Elabbas, L. E., Esteban, J., Barber, X., Hamscher, G., Nau, H., Bowers, W. J., . . . Håkansson, H. (2014). In Utero and 1793 Lactational Exposure to a Mixture of Environmental Contaminants Detected in Canadian Arctic Human 1794 Populations Alters Retinoid Levels in Rat Offspring with Low Margins of Exposure. Journal of Toxicology 1795 and Environmental Health, Part A, 77(5), 223-245. doi:10.1080/15287394.2013.861776 1796

Emmett, S. D., & West, K. P., Jr. (2014). Gestational vitamin A deficiency: a novel cause of sensorineural hearing loss 1797 in the developing world? Medical hypotheses, 82(1), 6-10. doi:10.1016/j.mehy.2013.09.028 1798

Escobar-Morreale, H. (2018). Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nature 1799 Reviews Endocrinology, 14(5), 270-284. doi:10.1038/nrendo.2018.24 1800

Feng, Y.-M., Liang, G.-J., Pan, B., Qin, X.-S., Zhang, X.-F., Chen, C.-L., . . . Shen, W. (2014). Notch pathway regulates 1801 female germ cell meiosis progression and early oogenesis events in fetal mouse. Cell cycle (Georgetown, 1802 Tex.), 13(5), 782-791. doi:10.4161/cc.27708 1803

Finkelstein, D., Lamba, V., Assem, M., Rengelshausen, J., Yasuda, K., Strom, S., & Schuetz, E. (2006). ADME 1804 transcriptome in Hispanic versus White donor livers: Evidence of a globally enhanced NR1I3 (CAR, 1805 constitutive androstane receptor) gene signature in Hispanics. Xenobiotica, 36(10-11), 989-1012. 1806 doi:10.1080/00498250600861769 1807

Fiorella, P. D., Olson, J. R., & Napoli, J. L. (1995). 2,3,7,8-Tetrachlorodibenzo-p-dioxin Induces Diverse Retinoic Acid 1808

Page 55: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

54

Metabolites in Multiple Tissues of the Sprague-Dawley Rat. Toxicology and Applied Pharmacology, 134(2), 1809 222-228. doi:https://doi.org/10.1006/taap.1995.1187 1810

Foti, R. S., Diaz, P., & Douguet, D. (2016). Comparison of the ligand binding site of CYP2C8 with CYP26A1 and 1811 CYP26B1: a structural basis for the identification of new inhibitors of the retinoic acid hydroxylases. 1812 Journal of Enzyme Inhibition and Medicinal Chemistry, 31(sup2), 148-161. 1813 doi:10.1080/14756366.2016.1193734 1814

Fritzsche, B., Vermot, J., Neumann, U., Schmidt, A., Schweigert, F. J., Dollé, P., & Rühl, R. (2007). Regulation of 1815 expression of the retinoic acid metabolizing enzyme CYP26A1 in uteri of ovariectomized mice after 1816 treatment with ovarian steroid hormones. Molecular Reproduction and Development, 74(2), 258-264. 1817 doi:10.1002/mrd.20526 1818

Fujihara, M., Yamamizu, K., Comizzoli, P., Wildt, D. E., & Songsasen, N. (2018). Retinoic acid promotes in vitro 1819 follicle activation in the cat ovary by regulating expression of matrix metalloproteinase 9. PLOS ONE, 1820 13(8), e0202759. doi:10.1371/journal.pone.0202759 1821

Gaemers, I. C., van Pelt, A. M. M., van der Saag, P. T., Hoogerbrugge, J. W., Themmen, A. P. N., & de Rooij, D. G. 1822 (1997). Effect of Retinoid Status on the Messenger Ribonucleic Acid Expression of Nuclear Retinoid 1823 Receptors α, β, and γ, and Retinoid X Receptors α, β, and γ in the Mouse Testis*. Endocrinology, 138(4), 1824 1544-1551. doi:10.1210/endo.138.4.5051 1825

Gely-Pernot, A., Raverdeau, M., Teletin, M., Vernet, N., Féret, B., Klopfenstein, M., . . . Ghyselinck, N. B. (2015). 1826 Retinoic Acid Receptors Control Spermatogonia Cell-Fate and Induce Expression of the SALL4A 1827 Transcription Factor. PLOS Genetics, 11(10), e1005501. doi:10.1371/journal.pgen.1005501 1828

Georges, A., Auguste, A., Bessière, L., Vanet, A., Todeschini, A.-L., & Veitia, R. A. (2014). FOXL2: a central 1829 transcription factor of the ovary. 52(1), R17. doi:10.1530/jme-13-0159 1830

Germain, P., Chambon, P., Eichele, G., Evans, R. M., Lazar, M. A., Leid, M., . . . Gronemeyer, H. (2006). International 1831 Union of Pharmacology. LX. Retinoic Acid Receptors. Pharmacological Reviews, 58(4), 712. 1832 doi:10.1124/pr.58.4.4 1833

Ghyselinck, N. B., Vernet, N., Dennefeld, C., Giese, N., Nau, H., Chambon, P., . . . Mark, M. (2006). Retinoids and 1834 spermatogenesis: Lessons from mutant mice lacking the plasma retinol binding protein. Developmental 1835 Dynamics, 235(6), 1608-1622. doi:10.1002/dvdy.20795 1836

Griswold, M. D. (2016). Spermatogenesis: The Commitment to Meiosis. Physiological reviews, 96(1), 1-17. 1837 doi:10.1152/physrev.00013.2015 1838

Griswold, M. D., & Hogarth, C. (2018). Beyond stem cells: Commitment of progenitor cells to meiosis. Stem Cell 1839 Research, 27, 169-171. doi:https://doi.org/10.1016/j.scr.2018.01.032 1840

Grive, K. J., & Freiman, R. N. (2015). The developmental origins of the mammalian ovarian reserve. Development, 1841 142(15), 2554. 1842

Han, B. C., Xia, H. F., Sun, J., Yang, Y., & Peng, J. P. (2010). Retinoic acid‐metabolizing enzyme cytochrome P450 1843 26a1 (cyp26a1) is essential for implantation: Functional study of its role in early pregnancy. Journal of 1844 Cellular Physiology, 223(2), 471-479. doi:10.1002/jcp.22056 1845

Hannon, P. R., & Flaws, J. A. (2015). The Effects of Phthalates on the Ovary. Frontiers in Endocrinology, 6, 8. 1846 doi:10.3389/fendo.2015.00008 1847

Heller, C. G., Moore, D. J., & Paulsen, C. A. (1961). Suppression of spermatogenesis and chronic toxicity in men by a 1848 new series of bis(dichloroacetyl) diamines. Toxicology and Applied Pharmacology, 3(1), 1-11. 1849 doi:https://doi.org/10.1016/0041-008X(61)90002-3 1850

Hoegberg, P., Schmidt, C. K., Nau, H., Catharine Ross, A., Zolfaghari, R., Fletcher, N., . . . Håkansson, H. (2003). 1851 2,3,7,8-Tetrachlorodibenzo-p-dioxin induces lecithin: retinol acyltransferase transcription in the rat 1852 kidney. Chemico-Biological Interactions, 145(1), 1-16. doi:https://doi.org/10.1016/S0009-2797(02)00157-6 1853

Hogarth, C. A., Amory, J. K., & Griswold, M. D. (2011). Inhibiting vitamin A metabolism as an approach to male 1854 contraception. Trends Endocrinol Metab, 22(4), 136-144. doi:10.1016/j.tem.2011.01.001 1855

Hogarth, C. A., Evanoff, R., Mitchell, D., Kent, T., Small, C., Amory, J. K., & Griswold, M. D. (2013). Turning a 1856 spermatogenic wave into a tsunami: synchronizing murine spermatogenesis using WIN 18,446. Biology of 1857 Reproduction, 88(2), 40-40. doi:10.1095/biolreprod.112.105346 1858

Hogarth, C. A., Evans, E., Onken, J., Kent, T., Mitchell, D., Petkovich, M., & Griswold, M. D. (2015). CYP26 Enzymes 1859 Are Necessary Within the Postnatal Seminiferous Epithelium for Normal Murine Spermatogenesis1. 1860 Biology of Reproduction, 93(1). doi:10.1095/biolreprod.115.129718 1861

Hogarth, C. A., & Griswold, M. D. (2010). The key role of vitamin A in spermatogenesis. J Clin Invest, 120(4), 956-1862 962. doi:10.1172/JCI41303 1863

Holm, J. B., Mazaud-Guittot, S., Danneskiold-Samsøe, N. B., Chalmey, C., Jensen, B., Nørregård, M. M., . . . 1864 Kristensen, D. M. (2016). Intrauterine Exposure to Paracetamol and Aniline Impairs Female Reproductive 1865 Development by Reducing Follicle Reserves and Fertility. Toxicological Sciences, 150(1), 178-189. 1866 doi:10.1093/toxsci/kfv332 1867

Page 56: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

55

Huang, F.-J., Hsuuw, Y.-D., Lan, K.-C., Kang, H.-Y., Chang, S.-Y., Hsu, Y.-C., & Huang, K.-E. (2005). Adverse effects of 1868 retinoic acid on embryo development and the selective expression of retinoic acid receptors in mouse 1869 blastocysts. Human Reproduction, 21(1), 202-209. doi:10.1093/humrep/dei286 1870

Håkansson, H., Manzoor, E., & Ahlborg, U. G. (1991). Interaction between dietary vitamin A and single oral doses of 1871 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the TCDD-induced toxicity and on the vitamin A status in 1872 the rat. Journal of nutritional science and vitaminology, 37(3), 239. doi:10.3177/jnsv.37.239 1873

Jacobs, M. N., Marczylo, E. L., Guerrero-Bosagna, C., & Rüegg, J. (2017). Marked for Life: Epigenetic Effects of 1874 Endocrine Disrupting Chemicals. Annual Review of Environment and Resources, 42(1), 105-160. 1875 doi:10.1146/annurev-environ-102016-061111 1876

Jana, K., Douglas, B. M., Qing, Z., Blanche, C., Michael, D. G., & David, C. P. (2006). Retinoic acid regulates sex-1877 specific timing of meiotic initiation in mice. Proceedings of the National Academy of Sciences of the United 1878 States of America, 103(8), 2474. doi:10.1073/pnas.0510813103 1879

Jauregui, E. J., Mitchell, D., Topping, T., Hogarth, C. A., & Griswold, M. D. (2018). Retinoic acid receptor signaling is 1880 necessary in steroidogenic cells for normal spermatogenesis and epididymal function. Development. 1881

Jayaram, M., Murthy, S. K., & Ganguly, J. (1973). Effect of vitamin A deprivation on the cholesterol side-chain 1882 cleavage enzyme activity of testes and ovaries of rats (Short Communication). Biochemical Journal, 1883 136(1), 221-223. 1884

Jiang, Y., Chen, L., Taylor, R. N., Li, C., & Zhou, X. (2018a). Physiological and pathological implications of retinoid 1885 action in the endometrium. J Endocrinol, 236(3), R169-R188. doi:10.1530/JOE-17-0544 1886

Jiang, Y., Li, C., Chen, L., Wang, F., & Zhou, X. (2017). Potential role of retinoids in ovarian physiology and 1887 pathogenesis of polycystic ovary syndrome. Clin Chim Acta, 469, 87-93. doi:10.1016/j.cca.2017.03.025 1888

Jiang, Y., Zhao, Y., Chen, S., Chen, L., Li, C., & Zhou, X. (2018b). Regulation by FSH of the dynamic expression of 1889 retinol-binding protein 4 in the mouse ovary. Reproductive Biology and Endocrinology, 16(1), 25. 1890 doi:10.1186/s12958-018-0348-8 1891

Jing, J., Nelson, C., Paik, J., Shirasaka, Y., Amory, J. K., & Isoherranen, N. (2017). Physiologically Based 1892 Pharmacokinetic Model of All-trans-Retinoic Acid with Application to Cancer Populations and Drug 1893 Interactions. The Journal of pharmacology and experimental therapeutics, 361(2), 246-258. 1894 doi:10.1124/jpet.117.240523 1895

Johansson, H. K. L., Svingen, T., Fowler, P. A., Vinggaard, A. M., & Boberg, J. (2017). Environmental influences on 1896 ovarian dysgenesis - developmental windows sensitive to chemical exposures. Nat Rev Endocrinol, 13(7), 1897 400-414. doi:10.1038/nrendo.2017.36 1898

Jørgensen, A., Nielsen, J. E., Perlman, S., Lundvall, L., Mitchell, R. T., Juul, A., & Rajpert-De Meyts, E. (2015). Ex vivo 1899 culture of human fetal gonads: manipulation of meiosis signalling by retinoic acid treatment disrupts 1900 testis development. Human Reproduction, 30(10), 2351-2363. doi:10.1093/humrep/dev194 1901

Kamata, R., Shiraishi, F., Nishikawa, J.-i., Yonemoto, J., & Shiraishi, H. (2008). Screening and detection of the in vitro 1902 agonistic activity of xenobiotics on the retinoic acid receptor. Toxicology in Vitro, 22(4), 1050-1061. 1903 doi:https://doi.org/10.1016/j.tiv.2008.01.002 1904

Kane, M., Folias, A., Wang, C., & Napoli, J. (2008). Quantitative Profiling of Endogenous Retinoic Acid in Vivo and in 1905 Vitro by Tandem Mass Spectrometry. Analytical Chemistry, 80(5), 1702-1708. doi:10.1021/ac702030f 1906

Kashimada, K., Svingen, T., Feng, C.-W., Pelosi, E., Bagheri-Fam, S., Harley, V. R., . . . Koopman, P. (2011). 1907 Antagonistic regulation of Cyp26b1 by transcription factors SOX9/SF1 and FOXL2 during gonadal 1908 development in mice. FASEB journal : official publication of the Federation of American Societies for 1909 Experimental Biology, 25(10), 3561-3569. doi:10.1096/fj.11-184333 1910

Kasimanickam, V., & Kasimanickam, R. (2014). Exogenous Retinoic Acid and Cytochrome P450 26B1 Inhibitor 1911 Modulate Meiosis‐Associated Genes Expression in Canine Testis, an In Vitro Model. Reproduction in 1912 Domestic Animals, 49(2), 315-323. doi:10.1111/rda.12276 1913

Kawai, T., Richards, J. S., & Shimada, M. (2018). The Cell Type–Specific Expression of Lhcgr in Mouse Ovarian Cells: 1914 Evidence for a DNA-Demethylation–Dependent Mechanism. Endocrinology, 159(5), 2062-2074. 1915 doi:10.1210/en.2018-00117 1916

Kawai, T., Yanaka, N., Richards, J. S., & Shimada, M. (2016). De Novo-Synthesized Retinoic Acid in Ovarian Antral 1917 Follicles Enhances FSH-Mediated Ovarian Follicular Cell Differentiation and Female Fertility. 1918 Endocrinology, 157(5), 2160-2172. doi:10.1210/en.2015-2064 1919

Kedishvili, N. Y. (2013). Enzymology of retinoic acid biosynthesis and degradation. Journal of Lipid Research, 54(7), 1920 1744-1760. doi:10.1194/jlr.R037028 1921

Kent, T., Arnold, S. L., Fasnacht, R., Rowsey, R., Mitchell, D., Hogarth, C. A., . . . Griswold, M. D. (2016). ALDH Enzyme 1922 Expression Is Independent of the Spermatogenic Cycle, and Their Inhibition Causes Misregulation of 1923 Murine Spermatogenic Processes1. Biology of Reproduction, 94(1). doi:10.1095/biolreprod.115.131458 1924

Kim, H., Lapointe, J., Kaygusuz, G., Ong, D. E., Li, C., van de Rijn, M., . . . Pollack, J. R. (2005). The retinoic acid 1925 synthesis gene ALDH1a2 is a candidate tumor suppressor in prostate cancer. Cancer research, 65(18), 1926

Page 57: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

56

8118. doi:10.1158/0008-5472.CAN-04-4562 1927 Klimenko, K., Rosenberg, S. A., Dybdahl, M., Wedebye, E. B., & Nikolov, N. G. (2019). QSAR modelling of a large 1928

imbalanced aryl hydrocarbon activation dataset by rational and random sampling and screening of 1929 80,086 REACH pre-registered and/or registered substances.(Research Article)(quantitative structure-1930 activity relationship)(Report). PLOS ONE, 14(3), e0213848. doi:10.1371/journal.pone.0213848 1931

Klonisch, T., Muller-Huesmann, H., Riedel, M., Kehlen, A., Bialek, J., Radestock, Y., . . . Hombach-Klonisch, S. (2005). 1932 INSL3 in the benign hyperplastic and neoplastic human prostate gland. Int J Oncol, 27(2), 307-315. 1933

Kogan, P., & Wald, M. (2014). Male Contraception: History and Development. Urologic Clinics of North America, 1934 41(1), 145-161. doi:https://doi.org/10.1016/j.ucl.2013.08.012 1935

Korkmaz, E., Cetinkaya, N., Oz, M., Ozgu, E., Bas, S., Isikalan, M., . . . Gungor, T. (2017). The Possibly Reversible 1936 Isotretinoin Effect of Decreased Ovarian Reserve in Sprague-Dawley Albinos: Part I, Biochemical Analyses. 1937 Gynecologic and Obstetric Investigation, 82(1), 72-77. 1938

Koubova, J., Hu, Y.-C., Bhattacharyya, T., Soh, Y. Q. S., Gill, M. E., Goodheart, M. L., . . . Page, D. C. (2014). Retinoic 1939 acid activates two pathways required for meiosis in mice. PLOS Genetics, 10(8), e1004541-e1004541. 1940 doi:10.1371/journal.pgen.1004541 1941

Kransler, K. M., Tonucci, D. A., McGarrigle, B. P., Napoli, J. L., & Olson, J. R. (2007). Gestational exposure to 2,3,7,8-1942 tetrachlorodibenzo-p-dioxin alters retinoid homeostasis in maternal and perinatal tissues of the 1943 Holtzman rat. Toxicology and Applied Pharmacology, 224(1), 29-38. 1944 doi:https://doi.org/10.1016/j.taap.2007.06.006 1945

Kumar, S., Chatzi, C., Brade, T., Cunningham, T. J., Zhao, X., & Duester, G. (2011). Sex-specific timing of meiotic 1946 initiation is regulated by Cyp26b1 independent of retinoic acid signalling. Nature Communications, 2, 1947 151-151. doi:10.1038/ncomms1136 1948

Kurlandsky, S. B., Gamble, M. V., Ramakrishnan, R., & Blaner, W. S. (1995). Plasma Delivery of Retinoic Acid to 1949 Tissues in the Rat. Journal of Biological Chemistry, 270(30), 17850-17857. doi:10.1074/jbc.270.30.17850 1950

Lambrot, R., Coffigny, H., Pairault, C., Donnadieu, A.-C., Frydman, R., Habert, R., & Rouiller-Fabre, V. (2006). Use of 1951 Organ Culture to Study the Human Fetal Testis Development: Effect of Retinoic Acid. The Journal of 1952 Clinical Endocrinology & Metabolism, 91(7), 2696-2703. doi:10.1210/jc.2005-2113 1953

Laudet, V., Zieger, E., & Schubert, M. (2015). Evolution of the Retinoic Acid Signaling Pathway. In P. Dollé & K. 1954 Neiderreither (Eds.), The Retinoids: Biology, Biochemistry and Disease (pp. 75-90). 1955

Laursen, K. B., Wong, P.-M., & Gudas, L. J. (2012). Epigenetic regulation by RARα maintains ligand-independent 1956 transcriptional activity. Nucleic acids research, 40(1), 102-115. doi:10.1093/nar/gkr637 1957

Lawson, C., Gieske, M., Murdoch, B., Ye, P., Li, Y., Hassold, T., & Hunt, P. A. (2011). Gene Expression in the Fetal 1958 Mouse Ovary Is Altered by Exposure to Low Doses of Bisphenol A1. Biology of Reproduction, 84(1), 79-86. 1959 doi:10.1095/biolreprod.110.084814 1960

Le Bouffant, R., Guerquin, M. J., Duquenne, C., Frydman, N., Coffigny, H., Rouiller-Fabre, V., . . . Livera, G. (2010). 1961 Meiosis initiation in the human ovary requires intrinsic retinoic acid synthesis. Human Reproduction, 1962 25(10), 2579-2590. doi:10.1093/humrep/deq195 1963

Lee, L. M. Y., Leung, C.-Y., Tang, W. W. C., Choi, H.-L., Leung, Y.-C., McCaffery, P. J., . . . Shum, A. S. W. (2012). A 1964 paradoxical teratogenic mechanism for retinoic acid. Proceedings of the National Academy of Sciences of 1965 the United States of America, 109(34), 13668-13673. doi:10.1073/pnas.1200872109 1966

Lemaire, G., Balaguer, P., Michel, S., & Rahmani, R. (2005). Activation of retinoic acid receptor-dependent 1967 transcription by organochlorine pesticides. Toxicology and Applied Pharmacology, 202(1), 38-49. 1968 doi:10.1016/j.taap.2004.06.004 1969

Li, H., & Clagett-Dame, M. (2009). Vitamin A Deficiency Blocks the Initiation of Meiosis of Germ Cells in the 1970 Developing Rat Ovary In Vivo1. Biology of Reproduction, 81(5), 996-1001. 1971 doi:10.1095/biolreprod.109.078808 1972

Liu, C., Paczkowski, M., Othman, M., & Yao, H. H.-C. (2012). Investigating the Origins of Somatic Cell Populations in 1973 the Perinatal Mouse Ovaries Using Genetic Lineage Tracing and Immunohistochemistry. In W.-Y. Chan & 1974 L. A. Blomberg (Eds.), Germline Development: Methods and Protocols (pp. 211-221). New York, NY: 1975 Springer New York. 1976

Liu, L., Suzuki, K., Nakagata, N., Mihara, K., Matsumaru, D., Ogino, Y., . . . Yamada, G. (2012). Retinoic acid signaling 1977 regulates sonic hedgehog and bone morphogenetic protein signalings during genital tubercle 1978 development. Birth defects research. Part B, Developmental and reproductive toxicology, 95(1), 79-88. 1979 doi:10.1002/bdrb.20344 1980

Liu, Z., Sun, Y., Jiang, Y., Qian, Y., Chen, S., Gao, S., . . . Zhou, X. (2018). Follicle-stimulating hormone (FSH) promotes 1981 retinol uptake and metabolism in the mouse ovary. Reproductive biology and endocrinology : RB&E, 16(1), 1982 52-52. doi:10.1186/s12958-018-0371-9 1983

Lohnes, D., Kastner, P., Dierich, A., Mark, M., Lemeur, M., & Chambon, P. (1993). Function of retinoic acid receptor γ 1984 in the mouse. Cell, 73(4), 643-658. doi:10.1016/0092-8674(93)90246-M 1985

Page 58: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

57

Long, M. D., Singh, P. K., Russell, J. R., Llimos, G., Rosario, S., Rizvi, A., . . . Campbell, M. J. (2019). The miR-96 and 1986 RARγ signaling axis governs androgen signaling and prostate cancer progression. Oncogene, 38(3), 421-1987 444. doi:10.1038/s41388-018-0450-6 1988

Lord, T., Oatley, M. J., & Oatley, J. M. (2018). Testicular Architecture Is Critical for Mediation of Retinoic Acid 1989 Responsiveness by Undifferentiated Spermatogonial Subtypes in the Mouse. Stem cell reports, 10(2), 538-1990 552. doi:10.1016/j.stemcr.2018.01.003 1991

Lovekamp-Swan, T., Jetten, A. M., & Davis, B. J. (2003). Dual activation of PPARα and PPARγ by mono-(2-1992 ethylhexyl) phthalate in rat ovarian granulosa cells. Molecular and Cellular Endocrinology, 201(1), 133-141. 1993 doi:https://doi.org/10.1016/S0303-7207(02)00423-9 1994

Lu, C., Xu, M., Wang, Y., Qin, Y., Du, G., Wu, W., . . . Wang, X. (2013). Genetic variants in meiotic program initiation 1995 pathway genes are associated with spermatogenic impairment in a Han Chinese population. PLOS ONE, 1996 8(1), e53443-e53443. doi:10.1371/journal.pone.0053443 1997

Lucas, T. F., Nascimento, A. R., Pisolato, R., Pimenta, M. T., Lazari, M. F. M., & Porto, C. S. (2014). Receptors and 1998 signaling pathways involved in proliferation and differentiation of Sertoli cells. Spermatogenesis, 4, 1999 e28138-e28138. doi:10.4161/spmg.28138 2000

Ma, J.-j., Han, B.-c., Yang, Y., & Peng, J.-p. (2012). Retinoic acid synthesis and metabolism are concurrent in the 2001 mouse uterus during peri-implantation. Cell and Tissue Research, 350(3), 525-537. doi:10.1007/s00441-2002 012-1507-4 2003

Macejova, D., Toporova, L., & Brtko, J. (2016). The role of retinoic acid receptors and their cognate ligands in 2004 reproduction in a context of triorganotin based endocrine disrupting chemicals. Endocr Regul, 50(3), 154-2005 164. doi:10.1515/enr-2016-0018 2006

Maclean, G., Li, H., Metzger, D., Chambon, P., & Petkovich, M. (2007). Apoptotic Extinction of Germ Cells in Testes 2007 of Cyp26b1 Knockout Mice. Endocrinology, 148(10), 4560-4567. doi:10.1210/en.2007-0492 2008

Mangelsdorf, D. J., & Evans, R. M. (1995). The RXR heterodimers and orphan receptors (Vol. 83, pp. 841-850). 2009 Marceau, G., Gallot, D., Borel, V., Lémery, D., Dastugue, B., Dechelotte, P., & Sapin, V. (2006). Molecular and 2010

metabolic retinoid pathways in human amniotic membranes. Biochemical and Biophysical Research 2011 Communications, 346(4), 1207-1216. doi:https://doi.org/10.1016/j.bbrc.2006.06.024 2012

Mark, M., Jacobs, H., Oulad-Abdelghani, M., Dennefeld, C., Féret, B., Vernet, N., . . . Ghyselinck, N. B. (2008). STRA8-2013 deficient spermatocytes initiate, but fail to complete, meiosis and undergo premature chromosome 2014 condensation. Journal of Cell Science, 121(19), 3233. 2015

Mark, M., Teletin, M., Vernet, N., & Ghyselinck, N. B. (2015). Role of retinoic acid receptor (RAR) signaling in post-2016 natal male germ cell differentiation. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, 2017 1849(2), 84-93. doi:https://doi.org/10.1016/j.bbagrm.2014.05.019 2018

Marti, N., Galván, J. A., Pandey, A. V., Trippel, M., Tapia, C., Müller, M., . . . Flück, C. E. (2017). Genes and proteins of 2019 the alternative steroid backdoor pathway for dihydrotestosterone synthesis are expressed in the human 2020 ovary and seem enhanced in the polycystic ovary syndrome. Molecular and Cellular Endocrinology, 441, 2021 116-123. doi:https://doi.org/10.1016/j.mce.2016.07.029 2022

Martin, M. T., Dix, D. J., Judson, R. S., Kavlock, R. J., Reif, D. M., Richard, A. M., . . . Houck, K. A. (2010). Impact of 2023 Environmental Chemicals on Key Transcription Regulators and Correlation to Toxicity End Points within 2024 EPA’s ToxCast Program. Chemical Research in Toxicology, 23(3), 578-590. doi:10.1021/tx900325g 2025

Minegishi, T., Hirakawa, T., Kishi, H., Abe, K., Tano, M., Abe, Y., & Miyamoto, K. (2000). The mechanisms of retinoic 2026 acid-induced regulation on the follicle-stimulating hormone receptor in rat granulosa cells. Biochimica et 2027 Biophysica Acta (BBA) - Molecular Cell Research, 1495(3), 203-211. doi:https://doi.org/10.1016/S0167-2028 4889(00)00003-3 2029

Minkina, A., Lindeman, R. E., Gearhart, M. D., Chassot, A. A., Chaboissier, M. C., Ghyselinck, N. B., . . . Zarkower, D. 2030 (2017). Retinoic acid signaling is dispensable for somatic development and function in the mammalian 2031 ovary. Dev Biol, 424(2), 208-220. doi:10.1016/j.ydbio.2017.02.015 2032

Minkina, A., Matson, Clinton K., Lindeman, Robin E., Ghyselinck, Norbert B., Bardwell, Vivian J., & Zarkower, D. 2033 (2014). DMRT1 Protects Male Gonadal Cells from Retinoid-Dependent Sexual Transdifferentiation. 2034 Developmental Cell, 29(5), 511-520. doi:10.1016/j.devcel.2014.04.017 2035

Morales, C. R., & Griswold, M. D. (1987). Retinol Induces Stage Synchronization in Seminiferous Tubules of Vitamin 2036 A Deficient Ratsa. Annals of the New York Academy of Sciences, 513(1), 292-293. doi:10.1111/j.1749-2037 6632.1987.tb25018.x 2038

Mumford, S. L., Browne, R. W., Schliep, K. C., Schmelzer, J., Plowden, T. C., Michels, K. A., . . . Schisterman, E. F. 2039 (2016). Serum Antioxidants Are Associated with Serum Reproductive Hormones and Ovulation among 2040 Healthy Women. The Journal of Nutrition, 146(1), 98-106. doi:10.3945/jn.115.217620 2041

Murphy, K. A., Quadro, L., & White, L. A. (2007). The Intersection Between the Aryl Hydrocarbon Receptor (AhR)‐ 2042 and Retinoic Acid‐Signaling Pathways Vitamins & Hormones (Vol. 75, pp. 33-67): Academic Press. 2043

Nakahashi, K., Matsuda, M., & Mori, T. (2001). Vitamin A Insufficiency Accelerates the Decrease in the Number of 2044

Page 59: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

58

Sperm Induced by an Environmental Disruptor, Bisphenol A, in Neonatal Mice. Zoological Science, 18(6), 2045 819-821. doi:10.2108/zsj.18.819 2046

Nakajima, T., Iguchi, T., & Sato, T. (2016). Retinoic acid signaling determines the fate of uterine stroma in the 2047 mouse Müllerian duct. Proceedings of the National Academy of Sciences, 113(50), 14354-14359. 2048 doi:10.1073/pnas.1608808113 2049

Nakanishi, T., Nishikawa, J.-i., Hiromori, Y., Yokoyama, H., Koyanagi, M., Takasuga, S., . . . Tanaka, K. (2005). 2050 Trialkyltin Compounds Bind Retinoid X Receptor to Alter Human Placental Endocrine Functions. 2051 Molecular Endocrinology, 19(10), 2502-2516. doi:10.1210/me.2004-0397 2052

Napoli, J. L. (2012). Physiological insights into all-trans-retinoic acid biosynthesis. BBA - Molecular and Cell Biology 2053 of Lipids, 1821(1), 152-167. doi:10.1016/j.bbalip.2011.05.004 2054

Napoli, J. L. (2017). Cellular retinoid binding-proteins, CRBP, CRABP, FABP5: Effects on retinoid metabolism, 2055 function and related diseases. Pharmacology & therapeutics, 173, 19-33. 2056 doi:10.1016/j.pharmthera.2017.01.004 2057

Nelson, C. H., Buttrick, B. R., & Isoherranen, N. (2013). Therapeutic potential of the inhibition of the retinoic acid 2058 hydroxylases CYP26A1 and CYP26B1 by xenobiotics. Current topics in medicinal chemistry, 13(12), 1402-2059 1428. 2060

Nicholls, P. K., Harrison, C. A., Rainczuk, K. E., Wayne Vogl, A., & Stanton, P. G. (2013). Retinoic acid promotes 2061 Sertoli cell differentiation and antagonises activin-induced proliferation. Molecular and Cellular 2062 Endocrinology, 377(1-2), 33-43. doi:10.1016/j.mce.2013.06.034 2063

Niederreither, K., Fraulob, V., Garnier, J.-M., Chambon, P., & Dollé, P. (2002). Differential expression of retinoic acid-2064 synthesizing (RALDH) enzymes during fetal development and organ differentiation in the mouse. 2065 Mechanisms of Development, 110(1), 165-171. doi:https://doi.org/10.1016/S0925-4773(01)00561-5 2066

Niederreither, K., McCaffery, P., Dräger, U. C., Chambon, P., & Dollé, P. (1997). Restricted expression and retinoic 2067 acid-induced downregulation of the retinaldehyde dehydrogenase type 2 (RALDH-2) gene during mouse 2068 development. Mechanisms of Development, 62(1), 67-78. doi:https://doi.org/10.1016/S0925-2069 4773(96)00653-3 2070

Niederreither, K., Vermot, J., Fraulob, V., Chambon, P., & Dolle, P. (2002). Retinaldehyde dehydrogenase 2 2071 (RALDH2)- independent patterns of retinoic acid synthesis in the mouse embryo. Proceedings of the 2072 National Academy of Sciences of the United States of America, 99(25), 16111-16116. 2073 doi:10.1073/pnas.252626599 2074

Nilsson, C. B., & Håkansson, H. (2002). The Retinoid Signaling System — A Target in Dioxin Toxicity. Critical 2075 Reviews in Toxicology, 32(3), 211-232. doi:10.1080/20024091064228 2076

Nishikawa, J.-i., Mamiya, S., Kanayama, T., Nishikawa, T., Shiraishi, F., & Horiguchi, T. (2004). Involvement of the 2077 Retinoid X Receptor in the Development of Imposex Caused by Organotins in Gastropods. Environmental 2078 Science & Technology, 38(23), 6271-6276. doi:10.1021/es049593u 2079

Noback, C. R., & Takahashi, Y. I. (1978). Micromorphology of the placenta of rats reared on marginal vitamin-A-2080 deficient diet. Cells Tissues Organs, 102(2), 195-202. doi:10.1159/000145637 2081

Novak, J., Benisek, M., & Hilscherova, K. (2008). Disruption of retinoid transport, metabolism and signaling by 2082 environmental pollutants. Environ Int, 34(6), 898-913. doi:10.1016/j.envint.2007.12.024 2083

Nya-Ngatchou, J. J., Arnold, S. L. M., Walsh, T. J., Muller, C. H., Page, S. T., Isoherranen, N., & Amory, J. K. (2013). 2084 Intratesticular 13-cis retinoic acid is lower in men with abnormal semen analyses: a pilot study. Andrology, 2085 1(2), 325-331. doi:10.1111/j.2047-2927.2012.00033.x 2086

O'Byrne, S. M., & Blaner, W. S. (2013). Retinol and retinyl esters: biochemistry and physiology. Journal of Lipid 2087 Research, 54(7), 1731-1743. doi:10.1194/jlr.R037648 2088

O'Byrne, S. M., Wongsiriroj, N., Libien, J., Vogel, S., Goldberg, I. J., Baehr, W., . . . Blaner, W. S. (2005). Retinoid 2089 absorption and storage is impaired in mice lacking lecithin:retinol acyltransferase (LRAT). The Journal of 2090 biological chemistry, 280(42), 35647-35657. doi:10.1074/jbc.M507924200 2091

Oulad-Abdelghani, M., Bouillet, P., Décimo, D., Gansmuller, A., Heyberger, S., Dollé, P., . . . Chambon, P. (1996). 2092 Characterization of a premeiotic germ cell-specific cytoplasmic protein encoded by Stra8, a novel retinoic 2093 acid-responsive gene. The Journal of Cell Biology, 135(2), 469. 2094

Paganetto, G., Campi, F., Varani, K., Piffanelli, A., Giovannini, G., & Borea, P. A. (2000). Endocrine-Disrupting Agents 2095 on Healthy Human Tissues. Pharmacology & Toxicology, 86(1), 24-29. doi:10.1034/j.1600-2096 0773.2000.pto860105.x 2097

Paik, J., Haenisch, M., Muller, C. H., Goldstein, A. S., Arnold, S., Isoherranen, N., . . . Amory, J. K. (2014). Inhibition of 2098 Retinoic Acid Biosynthesis by the Bisdichloroacetyldiamine WIN 18,446 Markedly Suppresses 2099 Spermatogenesis and Alters Retinoid Metabolism in Mice. Journal of Biological Chemistry, 289(21), 2100 15104-15117. doi:10.1074/jbc.M113.540211 2101

Palczewski, K. (2012). Chemistry and biology of vision. The Journal of biological chemistry, 287(3), 1612. 2102 doi:10.1074/jbc.R111.301150 2103

Page 60: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

59

Papacleovoulou, G., Edmondson, R. J., Critchley, H. O. D., Hillier, S. G., & Mason, J. I. (2009). 3β-Hydroxysteroid 2104 dehydrogenases and pre-receptor steroid metabolism in the human ovarian surface epithelium. 2105 Molecular and Cellular Endocrinology, 301(1), 65-73. doi:https://doi.org/10.1016/j.mce.2008.08.010 2106

Pauli, S. A., Session, D. R., Shang, W., Easley, K., Wieser, F., Taylor, R. N., . . . Sidell, N. (2013). Analysis of follicular 2107 fluid retinoids in women undergoing in vitro fertilization: retinoic acid influences embryo quality and is 2108 reduced in women with endometriosis. Reproductive sciences (Thousand Oaks, Calif.), 20(9), 1116-1124. 2109 doi:10.1177/1933719113477487 2110

Peng, J., Shen, L., Chen, J., Cao, X., Zhou, Y., Weng, H., . . . Wei, G. (2016). New discovery of cryptorchidism: 2111 Decreased retinoic acid in testicle. Saudi pharmaceutical journal : SPJ : the official publication of the Saudi 2112 Pharmaceutical Society, 24(3), 279-285. doi:10.1016/j.jsps.2016.04.020 2113

Pennimpede, T., Cameron Don, A., MacLean Glenn, A., Li, H., Abu-Abed, S., & Petkovich, M. (2010). The role of 2114 CYP26 enzymes in defining appropriate retinoic acid exposure during embryogenesis. Birth Defects 2115 Research Part A: Clinical and Molecular Teratology, 88(10), 883-894. doi:10.1002/bdra.20709 2116

Piersma, A. H., Hessel, E. V., & Staal, Y. C. (2017). Retinoic acid in developmental toxicology: Teratogen, morphogen 2117 and biomarker. Reprod Toxicol, 72, 53-61. doi:10.1016/j.reprotox.2017.05.014 2118

Pius, J., Shivanandappa, T., & Krishnakumari, M. K. (1990). Protective role of vitamin A in the male reproductive 2119 toxicity of Hexachlorocyclohexane (HCH) in the rat. Reproductive Toxicology, 4(4), 325-330. 2120 doi:10.1016/0890-6238(90)90045-W 2121

Pu, Y., Pearl, S., Gingrich, J., Jing, J., Martin, D., Murga-Zamalloa, C., & Veiga-Lopez, A. (2019). Multispecies study: 2122 low-dose tributyltin impairs ovarian theca cell cholesterol homeostasis through the RXR pathway in five 2123 mammalian species including humans. Archives of Toxicology, 93(6), 1665-1677. doi:10.1007/s00204-019-2124 02449-y 2125

Quadro, L., Blaner, W. S., Salchow, D. J., Vogel, S., Piantedosi, R., Gouras, P., . . . Gottesman, M. E. (1999). Impaired 2126 retinal function and vitamin A availability in mice lacking retinol‐binding protein. EMBO Journal, 18(17), 2127 4633-4644. doi:10.1093/emboj/18.17.4633 2128

Rato, L., Alves, M. G., Socorro, S., Duarte, A. I., Cavaco, J. E., & Oliveira, P. F. (2012). Metabolic regulation is 2129 important for spermatogenesis. Nature Reviews Urology, 9, 330. doi:10.1038/nrurol.2012.77 2130

Raverdeau, M., Gely-Pernot, A., Féret, B., Dennefeld, C., Benoit, G., Davidson, I., . . . Ghyselinck, N. B. (2012). Retinoic 2131 acid induces Sertoli cell paracrine signals for spermatogonia differentiation but cell autonomously drives 2132 spermatocyte meiosis. Proceedings of the National Academy of Sciences, 109(41), 16582. 2133 doi:10.1073/pnas.1214936109 2134

Rhinn, M., & Dollé, P. (2012). Retinoic acid signalling during development. Development (Cambridge, England), 2135 139(5), 843. doi:10.1242/dev.065938 2136

Rochette-Egly, C. (2015). Retinoic acid signaling and mouse embryonic stem cell differentiation: Cross talk 2137 between genomic and non-genomic effects of RA. Biochimica et Biophysica Acta (BBA) - Molecular and 2138 Cell Biology of Lipids, 1851(1), 66-75. doi:https://doi.org/10.1016/j.bbalip.2014.04.003 2139

Rodahl, K., & Moore, T. (1943). The vitamin A content and toxicity of bear and seal liver. The Biochemical journal, 2140 37(2), 166-168. doi:10.1042/bj0370166 2141

Rosenberg, S. A., Watt, E. D., Judson, R. S., Simmons, S. O., Paul Friedman, K., Dybdahl, M., . . . Wedebye, E. B. 2142 (2017a). QSAR models for thyroperoxidase inhibition and screening of U.S. and EU chemical inventories. 2143 Computational Toxicology, 4, 11-21. doi:https://doi.org/10.1016/j.comtox.2017.07.006 2144

Rosenberg, S. A., Xia, M., Huang, R., Nikolov, N. G., Wedebye, E. B., & Dybdahl, M. (2017b). QSAR development and 2145 profiling of 72,524 REACH substances for PXR activation and CYP3A4 induction. Computational 2146 Toxicology, 1, 39-48. doi:https://doi.org/10.1016/j.comtox.2017.01.001 2147

Ross, A. C., & Zolfaghari, R. (2011). Cytochrome P450s in the regulation of cellular retinoic acid metabolism. 2148 Annual review of nutrition, 31, 65-87. doi:10.1146/annurev-nutr-072610-145127 2149

Rossitto, M., Philibert, P., Poulat, F., & Boizet-Bonhoure, B. (2015). Molecular events and signalling pathways of 2150 male germ cell differentiation in mouse. Seminars in Cell & Developmental Biology, 45, 84-93. 2151 doi:https://doi.org/10.1016/j.semcdb.2015.09.014 2152

Rout, U. K., & Armant, D. R. (2002). Expression of genes for alcohol and aldehyde metabolizing enzymes in mouse 2153 oocytes and preimplantation embryos. Reproductive Toxicology, 16(3), 253-258. 2154 doi:https://doi.org/10.1016/S0890-6238(02)00022-9 2155

Sauvant, P., Sapin, V., Alexandre-Gouabau, M.-C., Dodeman, I., Delpal, S., Quadro, L., . . . S-Braesco, V. (2001). 2156 Retinol mobilization from cultured rat hepatic stellate cells does not require retinol binding protein 2157 synthesis and secretion. International Journal of Biochemistry and Cell Biology, 33(10), 1000-1012. 2158 doi:10.1016/S1357-2725(01)00066-8 2159

Sayem, A. S. M., Giribabu, N., Karim, K., Si, L. K., Muniandy, S., & Salleh, N. (2018). Differential expression of the 2160 receptors for thyroid hormone, thyroid stimulating hormone, vitamin D and retinoic acid and extracellular 2161 signal-regulated kinase in uterus of rats under influence of sex-steroids. Biomedicine & Pharmacotherapy, 2162

Page 61: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

60

100, 132-141. doi:https://doi.org/10.1016/j.biopha.2018.02.008 2163 Schmidt, C. K., Hoegberg, P., Fletcher, N., Nilsson, C. B., Trossvik, C., Håkansson, H., & Nau, H. (2003). 2,3,7,8-2164

Tetrachlorodibenzo-p-dioxin (TCDD) alters the endogenous metabolism of all-trans-retinoic acid in the 2165 rat. Archives of Toxicology, 77(7), 371-383. doi:10.1007/s00204-003-0457-8 2166

Schulze, G. E., Clay, R. J., Mezza, L. E., Bregman, C. L., Buroker, R. A., & Frantz, J. D. (2001). BMS-189453, a Novel 2167 Retinoid Receptor Antagonist, Is a Potent Testicular Toxin. Toxicological Sciences, 59(2), 297-308. 2168 doi:10.1093/toxsci/59.2.297 2169

See, A. W.-M., Kaiser, M. E., White, J. C., & Clagett-Dame, M. (2008). A nutritional model of late embryonic vitamin 2170 A deficiency produces defects in organogenesis at a high penetrance and reveals new roles for the 2171 vitamin in skeletal development. Developmental Biology, 316(2), 171-190. 2172 doi:https://doi.org/10.1016/j.ydbio.2007.10.018 2173

Shannon, S. R., Moise, A. R., & Trainor, P. A. (2017). New insights and changing paradigms in the regulation of 2174 vitamin A metabolism in development. Wiley interdisciplinary reviews. Developmental biology, 6(3), 2175 10.1002/wdev.1264. doi:10.1002/wdev.264 2176

Sharpe, R. M. (2010). Environmental/lifestyle effects on spermatogenesis. Philosophical transactions of the Royal 2177 Society of London. Series B, Biological sciences, 365(1546), 1697-1712. doi:10.1098/rstb.2009.0206 2178

Shirakami, Y., Lee, S.-A., Clugston, R. D., & Blaner, W. S. (2012). Hepatic metabolism of retinoids and disease 2179 associations. BBA - Molecular and Cell Biology of Lipids, 1821(1), 124-136. 2180 doi:10.1016/j.bbalip.2011.06.023 2181

Shmarakov, I. O. (2015). Retinoid-xenobiotic interactions: the Ying and the Yang. Hepatobiliary Surg Nutr, 4(4), 2182 243-267. doi:10.3978/j.issn.2304-3881.2015.05.05 2183

Shmarakov, I. O., Lee, Y. J., Jiang, H., & Blaner, W. S. (2019). Constitutive androstane receptor mediates PCB-2184 induced disruption of retinoid homeostasis. Toxicology and Applied Pharmacology, 381, 114731. 2185 doi:https://doi.org/10.1016/j.taap.2019.114731 2186

Siddikuzzaman, Guruvayoorappan, C., & Berlin Grace, V. M. (2011). All Trans Retinoic Acid and Cancer. 2187 Immunopharmacology and Immunotoxicology, 33(2), 241-249. doi:10.3109/08923973.2010.521507 2188

Skakkebæk, N. E., Rajpert-De Meyts, E., & Main, K. M. (2001). Testicular dysgenesis syndrome: an increasingly 2189 common developmental disorder with environmental aspects: Opinion. Human Reproduction, 16(5), 972-2190 978. doi:10.1093/humrep/16.5.972 2191

Spade, D. J., Dere, E., Hall, S. J., Schorl, C., Freiman, R. N., & Boekelheide, K. (2018a). All-Trans Retinoic Acid Disrupts 2192 Development in Ex Vivo Cultured Fetal Rat Testes. I: Altered Seminiferous Cord Maturation and Testicular 2193 Cell Fate. Toxicological Sciences, 167(2), 546-558. doi:10.1093/toxsci/kfy260 2194

Spade, D. J., Hall, S. J., Wortzel, J. D., Reyes, G., & Boekelheide, K. (2018b). All-trans Retinoic Acid Disrupts 2195 Development in Ex Vivo Cultured Fetal Rat Testes. II: Modulation of Mono-(2-ethylhexyl) Phthalate 2196 Toxicity. Toxicological Sciences, 168(1), 149-159. doi:10.1093/toxsci/kfy283 2197

Spiller, C., Koopman, P., & Bowles, J. (2017). Sex Determination in the Mammalian Germline. Annu Rev Genet, 51, 2198 265-285. doi:10.1146/annurev-genet-120215-035449 2199

Spiller, C. M., & Bowles, J. (2015). Sex determination in mammalian germ cells. Asian J Androl, 17(3), 427-432. 2200 doi:10.4103/1008-682X.150037 2201

Stevison, F., Hogarth, C., Tripathy, S., Kent, T., & Isoherranen, N. (2017). Inhibition of the all trans-retinoic acid 2202 hydroxylases CYP26A1 and CYP26B1 results in dynamic, tissue-specific changes in endogenous atRA 2203 signaling. Drug Metabolism and Disposition. 2204

Stoppie, P., Borgers, M., Borghgraef, P., Dillen, L., Goossens, J., Sanz, G., . . . Van Wauwe, J. (2000). R115866 inhibits 2205 all-trans-retinoic acid metabolism and exerts retinoidal effects in rodents. The Journal of pharmacology 2206 and experimental therapeutics, 293(1), 304. 2207

Su, R.-W., & Fazleabas, A. T. (2015). Implantation and Establishment of Pregnancy in Human and Nonhuman 2208 Primates. Advances in anatomy, embryology, and cell biology, 216, 189-213. doi:10.1007/978-3-319-2209 15856-3_10 2210

Suzuki, H., Kanai-Azuma, M., & Kanai, Y. (2015). From Sex Determination to Initial Folliculogenesis in Mammalian 2211 Ovaries: Morphogenetic Waves along the Anteroposterior and Dorsoventral Axes. Sex Dev, 9(4), 190-204. 2212 doi:10.1159/000440689 2213

Szanto, A., Narkar, V., Shen, Q., Uray, I. P., Davies, P. J. A., & Nagy, L. (2004). Retinoid X receptors: X-ploring their 2214 (patho)physiological functions. Cell death and differentiation, 11 Suppl 2, S126. 2215

Szwarc, M., Lan, H., William, E. G., Lisa, D. W., Qianxing, M., Ramakrishna, K., . . . John, P. L. (2018). Retinoid signaling 2216 controlled by SRC-2 in decidualization revealed by transcriptomics. REPRODUCTION, 156(5), 387-395. 2217 doi:10.1530/REP-18-0282 2218

Tarrade, A., Schoonjans, K., Pavan, L., Auwerx, J., Rochette-Egly, C. c., Evain-Brion, D. l., & Fournier, T. (2001). 2219 PPARγ/RXRα Heterodimers Control Human Trophoblast Invasion. The Journal of Clinical Endocrinology & 2220 Metabolism, 86(10), 5017-5024. doi:10.1210/jcem.86.10.7924 2221

Page 62: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

61

Taxvig, C., Hass, U., Axelstad, M., Dalgaard, M., Boberg, J., Andeasen, H. R., & Vinggaard, A. M. (2007). Endocrine-2222 Disrupting Activities In Vivo of the Fungicides Tebuconazole and Epoxiconazole. Toxicological Sciences, 2223 100(2), 464-473. doi:10.1093/toxsci/kfm227 2224

Tay, S., Dickmann, L., Dixit, V., & Isoherranen, N. (2010). A comparison of the roles of peroxisome proliferator-2225 activated receptor and retinoic acid receptor on CYP26 regulation. Molecular pharmacology, 77(2), 218-2226 227. doi:10.1124/mol.109.059071 2227

Taylor, R. N., Kane, M. A., & Sidell, N. (2015). Pathogenesis of Endometriosis: Roles of Retinoids and Inflammatory 2228 Pathways. Semin Reprod Med, 33(4), 246-256. doi:10.1055/s-0035-1554920 2229

Teletin, M., Vernet, N., Ghyselinck, N. B., & Mark, M. (2017). Roles of Retinoic Acid in Germ Cell Differentiation. Curr 2230 Top Dev Biol, 125, 191-225. doi:10.1016/bs.ctdb.2016.11.013 2231

Teletin, M., Vernet, N., Yu, J., Klopfenstein, M., Jones, J. W., Féret, B., . . . Mark, M. (2019). Two functionally 2232 redundant sources of retinoic acid secure spermatogonia differentiation in the seminiferous epithelium. 2233 Development, 146(1), dev170225. doi:10.1242/dev.170225 2234

Thatcher, J. E., Zelter, A., & Isoherranen, N. (2010). The relative importance of CYP26A1 in hepatic clearance of all-2235 trans retinoic acid. Biochemical pharmacology, 80(6), 903-912. doi:10.1016/j.bcp.2010.05.023 2236

Theodosiou, M., Laudet, V., & Schubert, M. (2010). From carrot to clinic: an overview of the retinoic acid signaling 2237 pathway. Cellular and Molecular Life Sciences, 67(9), 1423-1445. doi:10.1007/s00018-010-0268-z 2238

Tiboni, G. M., Marotta, F., & Carletti, E. (2009). Fluconazole alters CYP26 gene expression in mouse embryos. 2239 Reproductive Toxicology, 27(2), 199-202. doi:https://doi.org/10.1016/j.reprotox.2009.01.001 2240

Tong, M.-H., Yang, Q.-E., Davis, J. C., & Griswold, M. D. (2013). Retinol dehydrogenase 10 is indispensible for 2241 spermatogenesis in juvenile males. Proceedings of the National Academy of Sciences of the United States 2242 of America, 110(2), 543-548. doi:10.1073/pnas.1214883110 2243

Tonk, E. C. M., Pennings, J. L. A., & Piersma, A. H. (2015). An adverse outcome pathway framework for neural tube 2244 and axial defects mediated by modulation of retinoic acid homeostasis. Reproductive Toxicology, 55, 104-2245 113. doi:https://doi.org/10.1016/j.reprotox.2014.10.008 2246

Topletz, A. R., Tripathy, S., Foti, R. S., Shimshoni, J. A., Nelson, W. L., & Isoherranen, N. (2015). Induction of CYP26A1 2247 by metabolites of retinoic acid: evidence that CYP26A1 is an important enzyme in the elimination of 2248 active retinoids. Molecular pharmacology, 87(3), 430-441. doi:10.1124/mol.114.096784 2249

Urvalek, A., Laursen, K. B., & Gudas, L. J. (2014). The roles of retinoic acid and retinoic acid receptors in inducing 2250 epigenetic changes. Sub-cellular biochemistry, 70, 129-149. doi:10.1007/978-94-017-9050-5_7 2251

van Der Ven, L. T. M., van de Kuil, T., Leonards, P. E. G., Slob, W., Lilienthal, H., Litens, S., . . . Piersma, A. H. (2009). 2252 Endocrine effects of hexabromocyclododecane (HBCD) in a one-generation reproduction study in Wistar 2253 rats. Toxicology Letters, 185(1), 51-62. doi:10.1016/j.toxlet.2008.12.003 2254

van Der Ven, L. T. M., van de Kuil, T., Verhoef, A., Leonards, P. E. G., Slob, W., Cantón, R. F., . . . Vos, J. G. (2008). A 2255 28-day oral dose toxicity study enhanced to detect endocrine effects of a purified technical 2256 pentabromodiphenyl ether (pentaBDE) mixture in Wistar rats. Toxicology, 245(1), 109-122. 2257 doi:10.1016/j.tox.2007.12.016 2258

Van Pelt, A. M. M., & De Rooij, D. G. (1990). The Origin of the Synchronization of the Seminiferous Epithelium in 2259 Vitamin A-Deficient Rats after Vitamin A Replacement. Biology of Reproduction, 42(4), 677-682. 2260 doi:10.1095/biolreprod42.4.677 2261

Velte, E. K., Niedenberger, B. A., Serra, N. D., Singh, A., Roa-Delacruz, L., Hermann, B. P., & Geyer, C. B. (2019). 2262 Differential RA responsiveness directs formation of functionally-distinct spermatogonial populations at 2263 the initiation of spermatogenesis in the mouse. Development (Cambridge, England), 146(12). 2264 doi:10.1242/dev.173088 2265

Verze, P., Cai, T., & Lorenzetti, S. (2016). The role of the prostate in male fertility, health and disease. Nature 2266 Reviews Urology, 13, 379. doi:10.1038/nrurol.2016.89 2267

Vezina Chad, M., Allgeier Sarah, H., Fritz Wayne, A., Moore Robert, W., Strerath, M., Bushman, W., & Peterson 2268 Richard, E. (2008). Retinoic acid induces prostatic bud formation. Developmental Dynamics, 237(5), 1321-2269 1333. doi:10.1002/dvdy.21526 2270

White, J. C., Highland, M., & Clagett-Dame, M. (2000). Abnormal development of the sinuatrial venous valve and 2271 posterior hindbrain may contribute to late fetal resorption of vitamin A-deficient rat embryos. Teratology, 2272 62(6), 374-384. doi:10.1002/1096-9926(200012)62:6<374::AID-TERA4>3.0.CO;2-5 2273

White, J. C., Shankar, V. N., Highland, M., Epstein, M. L., DeLuca, H. F., & Clagett-Dame, M. (1998). Defects in 2274 embryonic hindbrain development and fetal resorption resulting from vitamin A deficiency in the rat are 2275 prevented by feeding pharmacological levels of all-&lt;em&gt;trans-&lt;/em&gt;retinoic acid. 2276 Proceedings of the National Academy of Sciences, 95(23), 13459. 2277

Wickenheisser, J. K., Nelson-DeGrave, V. L., Hendricks, K. L., Legro, R. S., Strauss, J. F., & McAllister, J. M. (2005). 2278 Retinoids and retinol differentially regulate steroid biosynthesis in ovarian theca cells isolated from 2279 normal cycling women and women with polycystic ovary syndrome. J Clin Endocrinol Metab, 90. 2280

Page 63: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

62

doi:10.1210/jc.2005-0330 2281 Vickery, B., Burns, J., Zaneveld, L., Goodpasture, J., & Bergström, K. (1985). Orally administered ketoconazole 2282

rapidly appears in seminal plasma and suppresses sperm motility. Advances in Contraception, 1(4), 341-2283 353. doi:10.1007/BF01849310 2284

Vilhais-Neto, G. C., & Pourquié, O. (2008). Retinoic acid. Current Biology, 18(7), 550-552. 2285 doi:10.1016/j.cub.2008.03.032 2286

Villota-Salazar, N. A., Mendoza-Mendoza, A., & González-Prieto, J. M. (2016). Epigenetics: from the past to the 2287 present. Frontiers in Life Science, 9(4), 347-370. doi:10.1080/21553769.2016.1249033 2288

Wiseman, E. M., Bar-El Dadon, S., & Reifen, R. (2017). The vicious cycle of vitamin a deficiency: A review. Crit Rev 2289 Food Sci Nutr, 57(17), 3703-3714. doi:10.1080/10408398.2016.1160362 2290

Wolf, G. (1996). A history of vitamin A and retinoids. The FASEB Journal, 10(9), 1102-1107. 2291 doi:10.1096/fasebj.10.9.8801174 2292

Wood, J. R., Nelson, V. L., Ho, C., Jansen, E., Wang, C. Y., Urbanek, M., . . . Strauss, J. F. (2003). The Molecular 2293 Phenotype of Polycystic Ovary Syndrome (PCOS) Theca Cells and New Candidate PCOS Genes Defined by 2294 Microarray Analysis. Journal of Biological Chemistry, 278(29), 26380-26390. doi:10.1074/jbc.M300688200 2295

Xu, H., Beasley, M. D., Warren, W. D., van der Horst, G. T. J., & McKay, M. J. (2005). Absence of Mouse REC8 2296 Cohesin Promotes Synapsis of Sister Chromatids in Meiosis. Developmental Cell, 8(6), 949-961. 2297 doi:10.1016/j.devcel.2005.03.018 2298

Yang, Y., Luo, J., Yu, D., Zhang, T., Lin, Q., Li, Q., . . . Huang, Y. (2018). Vitamin A Promotes Leydig Cell Differentiation 2299 via Alcohol Dehydrogenase 1. Frontiers in Endocrinology, 9, 644-644. doi:10.3389/fendo.2018.00644 2300

Yashiro, K., Zhao, X., Uehara, M., Yamashita, K., Nishijima, M., Nishino, J., . . . Hamada, H. (2004). Regulation of 2301 Retinoic Acid Distribution Is Required for Proximodistal Patterning and Outgrowth of the Developing 2302 Mouse Limb. Developmental Cell, 6(3), 411-422. doi:https://doi.org/10.1016/S1534-5807(04)00062-0 2303

Yoshida, S., Sukeno, M., Nakagawa, T., Ohbo, K., Nagamatsu, G., Suda, T., & Nabeshima, Y.-i. (2006). The first round 2304 of mouse spermatogenesis is a distinctive program that lacks the self-renewing spermatogonia stage. 2305 Development, 133(8), 1495. doi:10.1242/dev.02316 2306

Yuan, W., Wu, T., Fu, H., Dai, C., Wu, H., Liu, N., . . . Zhu, B. (2012). Dense Chromatin Activates Polycomb Repressive 2307 Complex 2 to Regulate H3 Lysine 27 Methylation. Science, 337(6097), 971. doi:10.1126/science.1225237 2308

Zhang, H.-Q., Zhang, X.-F., Zhang, L.-J., Chao, H.-H., Pan, B., Feng, Y.-M., . . . Shen, W. (2012). Fetal exposure to 2309 bisphenol A affects the primordial follicle formation by inhibiting the meiotic progression of oocytes. 2310 Molecular Biology Reports, 39(5), 5651-5657. doi:10.1007/s11033-011-1372-3 2311

Zhang, X.-F., Zhang, T., Han, Z., Liu, J.-C., Liu, Y.-P., Ma, J.-Y., . . . Shen, W. (2015). Transgenerational inheritance of 2312 ovarian development deficiency induced by maternal diethylhexyl phthalate exposure. Reproduction, 2313 Fertility and Development, 27(8), 1213-undefined. doi:10.1071/RD14113 2314

Zolfaghari, R., Mattie, F. J., Wei, C.-H., Chisholm, D. R., Whiting, A., & Ross, A. C. (2019). CYP26A1 gene promoter is 2315 a useful tool for reporting RAR-mediated retinoid activity. Analytical Biochemistry, 577, 98-109. 2316 doi:https://doi.org/10.1016/j.ab.2019.04.022 2317

Zuno-Floriano, F. G., Holstege, D., Hengel, M. J., Gaikwad, N. W., Aldana-Madrid, M. L., & Miller, M. G. (2012). 2318 Determination of Vitamin A and its Metabolites in Rat Testis: Possible Involvement of Vitamin A in 2319 Testicular Toxicity Caused by Molinate. Bulletin of Environmental Contamination and Toxicology, 88(6), 2320 1038-1042. doi:10.1007/s00128-012-0612-0 2321

2322

Page 64: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

63

14. Appendix 2323

2324

Assay methods and availability: 2325 2326 RA levels: 2327 Can be measured in tissues (organs, blood) or cells with e.g. liquid chromatography methods coupled 2328 to ultraviolet or mass spectrometric detection (a multitude of publications reviewed in the D-DRP). 2329 Can also be measured with ELISA kits (available for several species) 2330 https://www.mybiosource.com 2331 CYP 26: 2332 The different isomers can be studied on mRNA and protein level using commercially available probes 2333 and antibodies (see e.g. Topletz et al. 2012). 2334 ELISA kits for CYP26A1, B1 and C1 for several species are available. 2335 https://www.mybiosource.com 2336 Enzyme activity can be measured in microsomes as conversion of RA to its metabolites (Thatcher et 2337 al. 2010 analyzed human liver microsomes). 2338 RoDH 10: 2339 ELISA kits for several species are commercially available. 2340 https://www.mybiosource.com 2341 Stra8: 2342 mRNA and protein levels can be measured (Mark et al. 2008). 2343 cDNA and antibodies for multiple species are commercially available, as are ELISA kits 2344 https://www.mybiosource.com, https://www.labome.com, https://www.biocompare.com 2345 RARs, RXRs: 2346 Several assays are commercially available, many use HepG2 cells: 2347 http://alttox.org/application-of-medium-and-high-throughput-screening-for-in-vitro-toxicology-in-2348 the-pharmaceutical-industry/ 2349 http://www.attagene.com (used in ToxCast™) 2350 https://ncats.nih.gov/tox21/projects/assays 2351 AhR: 2352 AhR mRNA induction or activation can be measured in e.g. human HepG2 cells; commercially 2353 available. 2354 http://www.attagene.com (Attagene used in ToxCast™) 2355 https://ncats.nih.gov/tox21/projects/assays (Tox21 used in ToxCast™) 2356 CYP1A1: 2357 CYP1A1 activity can be measured in microsomes 2358 A multitude of assays (mRNA, protein, activity) are commercially available. 2359

2360 A description of a cell-based method (using mouse pluripotent P19 cells) for identifying chemicals 2361 that disrupt RA signaling pathways was published by Chen and Reese in 2013. It has been used to 2362 test phthalate esters (Chen and Reese 2016). 2363 2364 2365

Page 65: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

64

PubMed search strategy – retinoid parameters, chemical names, female/male 2366

reproduction parameters 2367 2368 Retinoid parameters were combined with chemical names (in Search table 1 below) and with either 2369 female or male reproduction parameters (in Search tables 2 and 3 below). No cutoffs (for e.g. year of 2370 publication) were used. The searches yielded several hundred of hits. However, only a small 2371 percentage was deemed relevant. In fact, a larger number of relevant articles was found via non-2372 structured searches (using e.g. references in articles, citing articles, etc). 2373 2374 Retinoid parameters: (retinoic OR retinoid OR retinol OR retinyl OR retinal). Inclusion of the term 2375 “vitamin A” rendered the exact same number of hits as when this term was excluded. 2376 2377 The selected chemicals are a combination of a) chemicals handpicked by Nancy Baker and coworkers 2378 at EPA, from ToxCast™ and other sources; b) ToxCast™ chemicals with activity on RARs/RXRs and 2379 CYP1A1 (information found in tables in the D-DRP); c) CYP26 inhibitors found via text mining 2380 (information found in table in the D-DRP); and finally, d) chemicals classified as Persistent Organic 2381 Pollutants by the Stockholm Convention. 2382 2383 Search table 1 (chemicals) 2384

"ser 2–7" (methyl 3-(1H-imidazol-1-yl)-2,2-dimethyl-3-(4-(naphthalen-2-ylamino)phenyl)propanoate

1-(6-tert-Butyl-1,1-dimethyl-2,3-dihydro-1H-inden-4-yl)ethanone

1,2,3-Trichlorobenzene

1,3,5-Triisopropylbenzene

2,2-dimethyl-3-(4-(naphthalen-2- ylamino)phenyl)-3-(1,2,4)triazol- 1-yl-propionic acid methyl ester

2,4,5-Trichlorophenol

2,4,6-Tris(tert-butyl)phenol

2,4-Bis(1-methyl-1-phenylethyl)phenol

2,6-Di-tert-butyl-4-ethylphenol

2,6-Di-tert-butyl-4-methoxyphenol

2-Mercaptobenzothiazole

2-Naphthylamine

3-(6-(2-dimethylamino-1- imidazol-1-yl-butyl)naphthalen-2- yloxy)-2,2-dimethyl-propionic acid

4,4'-Sulfonylbis[2-(prop-2-en-1-yl)phenol]

4-Nonylphenol

4-Nonylphenol, branched

4'-octyl-4-biphenylcarboxylic acid

AC-41848 hydrate

Acitretin

Adapalene

Aldrin

all/trans-Retinoic acid

Alpha-hexachlorocyclohexane

AM580

AR7

Azinphos-methyl

Bensulide

Benzyl alcohol

Beta-hexachlorocyclohexane

Bifenazate

Page 66: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

65

BMS 493

BMS-189453

BMS-195614

Butylated hydroxytoluene

CD437

Chlordane

Chlordecone

Chlorthal-dimethyl

Citric acid

Clorophene

Coumaphos

CP-532623

DDT OR Dichlorodiphenyltrichloroethane

Dieldrin

diethylaminobenzaldehyde

Difenoconazole

Diniconazole

Dioxin OR dibenzofuran

Dodecyl gallate

Dysprosium(III) chloride

Econazole nitrate

Endosulfan

Endrin

EPN OR ethyl phenylphosphonothioate

Esfenvalerate

Fludioxonil

Gentian Violet

Heptachlor

Hexabromobiphenyl

Hexabromocyclododecane

Hexabromodiphenyl ether and heptabromodiphenyl ether

Hexabromodiphenyl ether OR heptabromodiphenyl ether

Hexachlorbenzene

Hexachlorobutadiene

Imazalil

Imidazole

Isoniazid

Isopentyl benzoate

LE 135

Liarozole

Lindane OR gamma-hexachlorocyclohexane

Mepanipyrim

Methoprene acid

methyl 3-(4-(6-bromopyridin-3- ylamino)phenyl)-3-(1H-imidazol- 1-yl)-2,2-dimethylpropanoate

Mirex

N,N-Dimethylformamide

Nicotine

Page 67: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

66

N-Phenyl-1,4-benzenediamine

Organotin

Pentachlorobenzene

Pentachlorphenol

Perfluorooctanesulfonic acid OR perfluorooctanesulfonyl fluoride

Phosalone

polychlorinated AND (naphtalene OR naphtalenes OR napthalene OR napthalenes)

Polychlorinated biphenyls OR PCBs

Polychlorinated AND (dibenzo-p-dioxins OR dibenzofurans)

Prochloraz

Pyraclostrobin

R115866 OR rambazole

R116010

SR271425

Symclosene

Talarozole

Tetrabromodiphenyl ether and pentabromodiphenyl ether

Tetrabromodiphenyl ether OR pentabromodiphenyl ether

Tetrabutyltin

Toxaphene

Tributyltin benzoate

Tributyltin chloride

Tributyltin methacrylate

Triflumizole

Triphenyltin hydroxide

Triticonazole

TTNPB

2385 2386

Page 68: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

67

2387 Search table 2 (female reproduction parameters) 2388 “female genitalia”

“female gonad”

“Female reproduction”

“Granulosa cells”

“Polycystic ovarian syndrome”

“Theca cells”

Abortion

Cervix OR cervical

Corpus Luteum

Decidua OR decidualization

Eclampsia

Embryo OR embryonic

Endometriosis

Endometrium OR endometrial

Estrous

Fallopian

Fertility OR infertility

Fetus OR fetal

Folliculogenesis

Gestational

Lactation

Litter

Luteal

Maternal

Menopause

Menstrual OR menstruation

Oocyte

Oogenesis

Oogonia

Ovary OR Ovarian

Ovulation

Placenta OR placental

Pregnancy OR pregnant

Prenatal OR postnatal

Puberty

Trimester

Uterus OR uterine

Vagina OR vaginal

2389 2390

Page 69: Draft NORDIC WORKING PAPERS - OECD Nordic... · 168 ultimo 2018) & Marie-Louise Holmer (until primo 2018 (The Danish Environmental Protection Agency); ... 182 must be supplied from

68

Search table 3 (male reproduction parameters) 2391

Aspermia Asthenozoospermia Azoospermia Blood-Testis Barrier Cryptorchidism Ejaculation epididymis Epididymitis erectile fertilization Hemospermia Hypospadia impotence Leydig male Infertility Oligospermia Orchitis penile prostate prostatic Prostatitis Rete Testis scrotum Seminiferous Sertoli Sperm Spermatid Spermatocele Spermatocyte Spermatogenesis Spermatogonia Spermatozoa Testes Testicular Testis

2392