xenopus in revealing developmental toxicity and modeling

13
Review Xenopus in revealing developmental toxicity and modeling human diseases * Juanmei Gao a, b , Wanhua Shen a, * a Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China b College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China article info Article history: Received 14 June 2020 Received in revised form 1 October 2020 Accepted 9 October 2020 Available online 13 October 2020 Keywords: Xenopus Toxicity Toxicant Oocyte Embryo development Disease model abstract The Xenopus model offers many advantages for investigation of the molecular, cellular, and behavioral mechanisms underlying embryo development. Moreover, Xenopus oocytes and embryos have been extensively used to study developmental toxicity and human diseases in response to various environ- mental chemicals. This review rst summarizes recent advances in using Xenopus as a vertebrate model to study distinct types of tissue/organ development following exposure to environmental toxicants, chemical reagents, and pharmaceutical drugs. Then, the successful use of Xenopus as a model for dis- eases, including fetal alcohol spectrum disorders, autism, epilepsy, and cardiovascular disease, is reviewed. The potential application of Xenopus in genetic and chemical screening to protect against embryo decits induced by chemical toxicants and related diseases is also discussed. © 2020 Elsevier Ltd. All rights reserved. 1. Introduction The South African clawed frog has been used as one of the main vertebrate animal models since their introduction as a model or- ganism in the 1950s (Beck and Slack, 2001; Tadjuidje and Heasman, 2010). The Xenopus model is a time- and cost-efcient model for in vivo studying embryonic development and environmental toxicity. The diploid Xenopus tropicalis (X. tropicalis) and the allo- tetraploid Xenopus laevis (X. laevis) have two distinct genetic backgrounds and are widely used in developmental, biomedical, and cancer research (Buryskova et al., 2006; Davis et al., 1981; Hardwick and Philpott, 2015; Yao et al., 2017; Zhu et al., 2017 , 2018). In addition, frogs can produce massive numbers of eggs that are fertilized into tadpoles in vitro in a matter of days during all seasons (Showell and Conlon, 2009). Both embryos and adults live in the water and are independent of maternal inuence, which makes it easy for them to absorb exogenous hormones or other chemical compounds (Beck and Slack, 2001). Most importantly, Xenopus as a tetrapod is evolutionarily closer to higher vertebrates than other aquatic models in terms of organ development, physiology, and gene expression (Nenni et al., 2019). Other evolutionary similarities to mammals are also observed at the level of organ structure and function, such as lungs (Galdiero et al., 2017), heart (Blum and Ott, 2018), kidneys (Desgrange and Cereghini, 2015), lymphatics (Ny et al., 2006) and immune system (Wheeler and Brandli, 2009) (Fig. 1). Therefore, Xenopus has become an important vertebrate animal model for answering developmental questions, toxic effects, and human disease mechanisms. Many aspects of Xenopus organs can be used to investigate the mechanism of organogenesis and embryogenesis (Hoppler and Conlon, 2019). In its ease use of manipulation and molecular intervention, studies in Xenopus tissues/organs have made a sig- nicant contribution to current understanding of embryo devel- opment, including the tectal brain, eyes, heart and kidneys (Frontera et al., 2016a; Tomlinson et al., 2005). The visual retino- tectal system has been extensively used to study neurogenesis, visual circuit function and synaptic plasticity in vivo (Akerman and Cline, 2006; Faulkner et al., 2015; He et al., 2018; Lee et al., 2010; McKeown et al., 2013; Pratt and Aizenman, 2007; Shen et al., 2011 , 2014; Truszkowski et al., 2016). Free swimming and visually guided avoidance behaviors can be analyzed to measure visual-motor function, which helps in the understanding of sensory processing * This paper has been recommended for acceptance by Dr. Sarah Harmon. * Corresponding author. E-mail address: [email protected] (W. Shen). Contents lists available at ScienceDirect Environmental Pollution journal homepage: www.elsevier.com/locate/envpol https://doi.org/10.1016/j.envpol.2020.115809 0269-7491/© 2020 Elsevier Ltd. All rights reserved. Environmental Pollution 268 (2021) 115809

Upload: others

Post on 28-Feb-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

lable at ScienceDirect

Environmental Pollution 268 (2021) 115809

Contents lists avai

Environmental Pollution

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

Review

Xenopus in revealing developmental toxicity and modeling humandiseases*

Juanmei Gao a, b, Wanhua Shen a, *

a Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou,Zhejiang, 311121, Chinab College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China

a r t i c l e i n f o

Article history:Received 14 June 2020Received in revised form1 October 2020Accepted 9 October 2020Available online 13 October 2020

Keywords:XenopusToxicityToxicantOocyteEmbryo developmentDisease model

* This paper has been recommended for acceptanc* Corresponding author.

E-mail address: [email protected] (W. Shen).

https://doi.org/10.1016/j.envpol.2020.1158090269-7491/© 2020 Elsevier Ltd. All rights reserved.

a b s t r a c t

The Xenopus model offers many advantages for investigation of the molecular, cellular, and behavioralmechanisms underlying embryo development. Moreover, Xenopus oocytes and embryos have beenextensively used to study developmental toxicity and human diseases in response to various environ-mental chemicals. This review first summarizes recent advances in using Xenopus as a vertebrate modelto study distinct types of tissue/organ development following exposure to environmental toxicants,chemical reagents, and pharmaceutical drugs. Then, the successful use of Xenopus as a model for dis-eases, including fetal alcohol spectrum disorders, autism, epilepsy, and cardiovascular disease, isreviewed. The potential application of Xenopus in genetic and chemical screening to protect againstembryo deficits induced by chemical toxicants and related diseases is also discussed.

© 2020 Elsevier Ltd. All rights reserved.

1. Introduction

The South African clawed frog has been used as one of the mainvertebrate animal models since their introduction as a model or-ganism in the 1950s (Beck and Slack, 2001; Tadjuidje and Heasman,2010). The Xenopus model is a time- and cost-efficient model forin vivo studying embryonic development and environmentaltoxicity. The diploid Xenopus tropicalis (X. tropicalis) and the allo-tetraploid Xenopus laevis (X. laevis) have two distinct geneticbackgrounds and are widely used in developmental, biomedical,and cancer research (Buryskova et al., 2006; Davis et al., 1981;Hardwick and Philpott, 2015; Yao et al., 2017; Zhu et al., 2017, 2018).In addition, frogs can produce massive numbers of eggs that arefertilized into tadpoles in vitro in amatter of days during all seasons(Showell and Conlon, 2009). Both embryos and adults live in thewater and are independent of maternal influence, which makes iteasy for them to absorb exogenous hormones or other chemicalcompounds (Beck and Slack, 2001). Most importantly, Xenopus as atetrapod is evolutionarily closer to higher vertebrates than other

e by Dr. Sarah Harmon.

aquatic models in terms of organ development, physiology, andgene expression (Nenni et al., 2019). Other evolutionary similaritiesto mammals are also observed at the level of organ structure andfunction, such as lungs (Galdiero et al., 2017), heart (Blum and Ott,2018), kidneys (Desgrange and Cereghini, 2015), lymphatics (Nyet al., 2006) and immune system (Wheeler and Brandli, 2009)(Fig. 1). Therefore, Xenopus has become an important vertebrateanimal model for answering developmental questions, toxic effects,and human disease mechanisms.

Many aspects of Xenopus organs can be used to investigate themechanism of organogenesis and embryogenesis (Hoppler andConlon, 2019). In its ease use of manipulation and molecularintervention, studies in Xenopus tissues/organs have made a sig-nificant contribution to current understanding of embryo devel-opment, including the tectal brain, eyes, heart and kidneys(Frontera et al., 2016a; Tomlinson et al., 2005). The visual retino-tectal system has been extensively used to study neurogenesis,visual circuit function and synaptic plasticity in vivo (Akerman andCline, 2006; Faulkner et al., 2015; He et al., 2018; Lee et al., 2010;McKeown et al., 2013; Pratt and Aizenman, 2007; Shen et al., 2011,2014; Truszkowski et al., 2016). Free swimming and visually guidedavoidance behaviors can be analyzed to measure visual-motorfunction, which helps in the understanding of sensory processing

Fig. 1. Schematic diagram of evolutionarily conserved tissues and organs used for toxicological studies in zebrafish, Xenopus, and mouse. (A) Various tissues and organs are used fordevelopmental and toxicological studies in zebrafish, Xenopus, and mouse, such as heart, kidney, and craniofacial. The size of each organ is not proportionally scaled. A, atrium; V:ventricle; RA, right atrium; LA, left atrium; RV: right ventricle; LV: left ventricle.

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

in an intact neural circuit (He et al., 2018; Pratt and Aizenman,2007; Shen et al., 2011). The development of Xenopus pronephrickidney is similar to the amniotes metanephros, which is used toexamine nephron formation and model human kidney diseases(Cizelsky et al., 2014; Desgrange and Cereghini, 2015; Hwang et al.,2019) (Fig. 1). Amphibian metamorphosis is utilized in the researchof thyroid hormone-mediated cell apoptosis and stem cell devel-opment during vertebrate developmental transitions (Buchholz,2017; Das et al., 2002). Unlike mammals, Xenopus tadpoles havehigh regenerative potential for structures including the spinal cord(Aztekin et al., 2019; Borodinsky, 2017; Edwards-Faret et al., 2017;Munoz et al., 2015), heart (Liao et al., 2017; Marshall et al., 2019),epithelium (Frontera et al., 2016b; Kim et al., 2020), and limbs(Herrera-Rincon et al., 2018; Yokoyama et al., 2011), making Xen-opus an attractive model to uncover the mechanisms governingregeneration (Borodinsky, 2017; Phipps et al., 2020).

Exposure to environmental toxicants can cause short-termreversible or long-term irreversible damage to various tissues andorgans, leading to malformations and diseases (Fig. 2). Morerecently, several disease models, such as those for fetal alcoholspectrum disorders (FASDs), epilepsy, autism, and cardiovasculardiseases, have been successfully developed in Xenopus (Sater andMoody, 2017). Toxicant-induced developmental deficits and func-tional alterations in embryos can be characterized through a varietyof elegant techniques to gain insight into the etiology and

2

pathophysiology of human diseases. In addition, Xenopus has beensuccessfully used to identify chemically responsive factors andcharacterize the disease mechanisms of pathogenic human vari-ants. Hence, Xenopus model provides an integrative approach forunderstanding the development of organs and the mechanisms ofhuman diseases. In this review, we summarized the recent progressin using Xenopus as an animal model for developmental studies,toxicological investigations, and disease modeling, with a descrip-tion of advances in methodological applications and drugscreenings.

2. Advantages of using Xenopus as an in vivo model fortoxicological studies

The Xenopus system has been traditionally used as a develop-mental model to investigate neurogenesis, embryogenesis, organ-ogenesis, and metamorphosis (Tadjuidje and Heasman, 2010). Asamphibian species have both aquatic larval phase and adultterrestrial phase, these organisms are sensitive to water and soilpollutants, which makes them to be a unique model for monitoringenvironmental risks with low experimental costs. The Xenopus eggis a huge cell that is easily manipulated. The Xenopus oocyteexpression system is utilized to determine the properties of ionchannels and neurotransmitter transporters in single cells withoutinterfering with the complexities of brain tissues. Cell extracts have

Fig. 2. Schematic diagram of several malformations used for testing chemical toxicity in Xenopus. Representative Xenopus stages at 32, 45 and 60 are used for studying chemical-induced developmental deficits. (A) Representative defects are shown as loss of eyes, abnormal embryogenesis, and embryo malformations at the developing stage 32. (B)Representative defects are shown as pigment aggregation, pigment loss, misfolded gut, abnormal eye, heart enlargement, decreased spine length, and tail flexure at stage 45. (C)Representative defects are shown as pigment aggregation, dispersed melanosomes, increased limb length, limb deformities, abnormal metamorphosis, and growth inhibition atstage 60. The size of embryos is not proportionally scaled.

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

been used to examine the fundamental processes of developmentalbiology, genome maintenance mechanisms and chemical genetics(Cross and Powers, 2009; Hoogenboom et al., 2017; Maia et al.,2017). Other tissues in Xenopus embryos have been widely usedto study embryogenesis, toxicology and developmental diseasesusing optic tectum (Pratt and Khakhalin, 2013), intestine (Ikuzawaet al., 2006), kidneys (Hwang et al., 2019), heart (Duncan andKhokha, 2016) and eyes (Imaoka et al., 2007; Luehders et al.,2015). Combining with methodological advances, such as in vivoelectrophysiological recordings (Hiramoto and Cline, 2014;Kutsarova et al., 2016; Shen et al., 2011; Tao and Poo, 2005),genomic microarray chips (Fu et al., 2017; Huyck et al., 2015; Zhuet al., 2017), RNA deep sequencing (Paranjpe et al., 2013), electrontransfer dissociation (ETD) (McGivern et al., 2009), and in vivobioorthogonal noncanonical amino acid tagging (BONCAT) (Liuet al., 2018; Shen et al., 2014), the Xenopus system has become avaluable vertebrate model system to decipher the mechanism ofchemical toxicants-induced defects and screen potential pharma-ceutical drugs to treat the damage (Liao et al., 2020). However, thedependence on specific antibodies can also be a limitation, while itis time consuming and expensive to generate antibodies to detectXenopus proteins. The collection of large tissue samplings would bealso labor consuming.

2.1. Xenopus oocytes and egg extracts

Unfertilized Xenopus oocytes removed from female frogs havebeen broadly used to investigate endocytosis mechanisms, ionchannels, and cytological processes (Maia et al., 2017). The lowendogenous expression of ion channels in oocytes makes themamenable to expressing exogenous channel proteins or disease-related proteins and performing electrophysiological recordingsto study ion transport and channel physiology (Schmidt et al.,2009). The physiological properties of ion channels expressed inoocytes, such as NMDAR (Cruz et al., 2000), AMPAR (Chang et al.,2016), GABAR (Storustovu and Ebert, 2006), and nAChRs (van

3

Kleef et al., 2008), provide convenient tools to characterize the ef-fects of active compounds or environmental pollutants, such astoluene (Cruz et al., 1998) and ototoxic solvents (van Kleef et al.,2008) on corresponding receptors. As an in vitro model, Xenopusoocytes will continue to provide an efficient system in identifyingnovel drug targets within hundreds of potential candidates andfurther understanding the mechanism of human diseases (Zenget al., 2020).

Xenopus egg extracts are a cell-free, biologically active cyto-plasm that mimics the cellular physiological environment andcontains all cytoplasmic proteins, organelles, and other essentialcomponents. The Xenopus oocyte extract system stands out as apowerful tool in determining signaling pathway, studying genomemaintenance mechanisms (Hardwick and Philpott, 2015;Hodskinson et al., 2020; Hoogenboom et al., 2017; Hyde et al.,2016). Due to its low cost and efficient nature, the system hasbeenwidely used in screening potential activators or inhibitors andrevealing drug toxicology effects within a large number of com-pounds (Maia et al., 2017; Thorne et al., 2011). Thesemethodologiesallow researchers to build systematic tools to study chemical effectson cell signaling and screen potential anti-toxic drugs.

2.2. Xenopus embryos

Most tissues and organs of Xenopus have been extensively usedto study embryo development and toxicological studies from theembryonic to metamorphic stages (Taft et al., 2018; Zhang et al.,2020) (Fig. 1). The retinotectal visual system is a powerful modelfor examining neurotransmission in neural circuit developmentand function in response to different environmental factors(Igarashi et al., 2015; James et al., 2015; Liao et al., 2020). The visualbehavior of Xenopus tadpoles is currently used to determine visual-motor functions and neurotoxicity induced by chemical toxicants(Dong and Aizenman, 2012; Gao et al., 2016a; Shen et al., 2014).Pigment cells (melanophores in amphibians; melanocytes inmammals) are aggregated in light and dispersed in dark in the wild

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

type Xenopuswithout the influence of time of day. A complex seriesof chemical and physiological mechanisms regulates melanophorepigment distribution, making these skin cells a valuable tool tostudy pigment cell development, assess chemical toxicity andperform drug screening, as an in vivo model (Dahora et al., 2020;Iuga et al., 2009; Tomlinson et al., 2009; Wheeler and Brandli,2009). In contrast to wide type Xenopus, the transparent nature ofAlbino Xenopus tadpole skin is well suited for live tracking of neuralmorphology, cardiovascular dynamics, heart development, lym-phangiogenesis, and angiogenesis over hours to dayswith exposureto chemical toxicants (Duncan and Khokha, 2016; Hoppler andConlon, 2019; Liao et al., 2020; Ny et al., 2006).

While zebrafish shares many advantages with X. laevis in addi-tion to its well-developed genetic manipulation strategies, theirgenome may not perform same function as mammals (Lasser et al.,2019). Zebrafish genome is evolutionarily distant to humans andmake it difficult in identifying human orthologs. Moreover,X. tropicalis has a true diploid genome with substantial sharedsynteny with the human genome, which is feasible to create per-manent transgenic lines for genetic manipulations. In addition,Xenopus shares greater similarities in tissues and organs withhigher vertebrates, such as heart, kidneys, vascular, lungs andlymphatic system (Ny et al., 2006) (Fig.1). Additionally, Xenopus hasa three-chambered heart, while zebrafish has a two-chamberedheart with one atrium and one ventricle, which cannot fullymodel four-chambered human heart abnormalities (Blum and Ott,2018; Duncan and Khokha, 2016) (Fig. 1). Also, Xenopus tadpoleswill develop lungs and limbs, which are absent from zebrafish.Importantly, amphibian metamorphosis physiologically resemblesmammalian postembryonic development (Wheeler and Brandli,2009). These advantages make Xenopus a unique model to studythe impact of environmental factors on its development frommolecular and cellular to circuit and behavioral levels (Hasebe et al.,2017; Thompson and Cline, 2016; Tomlinson et al., 2005).

3. Toxicological studies using Xenopus

Accumulating evidence has shown that Xenopus embryos aresensitive to environmental chemicals, which makes Xenopus aunique model animal for cytotoxicity research (Wheeler andBrandli, 2009). Multiple tissues and organs in Xenopus have beenused to examine the developmental effects of chemical pollutants,hormones, pesticides, and other environmental factors (Table 1).Xenopus has become a good model to build links between pheno-types of malformations and changes of gene expression due toestablished molecular and cellular technologies. The frog embryoteratogenesis assay in Xenopus (FETAX) has been used for manyyears to test mortality and malformations in response to variousenvironmental factors (Fort and Mathis, 2018; Newman andDumont, 1983). Multidisciplinary techniques such as electrophysi-ology, calcium imaging, click chemistry labeling, cryo-electronmicroscopy and time-lapse imaging have been used extensivelyto study the effects of chemical toxicity on tadpole development(Bestman and Cline, 2020; Ciarleglio et al., 2015; Glasgow et al.,2019; Hasebe et al., 2017; Khakhalin et al., 2014; Liu and Haas,2011; Shen et al., 2014). Proteomic analysis using the iTRAQ tech-nique in the gonads or transcriptome-wide profile in the testis ofX. laevis has been used to examine global protein changes or m (6)amethylome profiles in response to atrazine (Chen et al., 2015).Combined with swimming behavior analysis, Xenopus can be usedas an important animal model for biosafety assessment and po-tential drug screening at an early stage of clinical trials (Maia et al.,2017; Tomlinson et al., 2005; Zhang et al., 2014), especially forscreening early event targets in cell differentiation and organo-genesis (Hwang et al., 2019). Therefore, the successful use of

4

advanced techniques in Xenopus is amenable to facilitate theresearch on developmental toxicity and the feasibility of screeningpotential drugs (Schmitt et al., 2014).

3.1. Chemical pollutants

Xenopus is a commonly used vertebrate model in the study ofthe effects of toxic compounds on embryo development and neuralcircuit function during development (Wang et al., 2018). Numerouschemical pollutants, such as benzene, methylmercury, cadmium,zinc and polychlorinated biphenyls, have been found to causeneurodevelopmental toxicity in Xenopus (Huyck et al., 2015;Mouchet et al., 2015; Rice, 2008) (Table 1).

The benzene, toluene, ethylbenzene and xylene aromatic hy-drocarbons, collectively called BTEX, are emitted from oil refineriesand gas stations and are known to cause eye inflammation, head-ache, asthma and severe diseases such as leukemia and multiplemyeloma (McKenzie et al., 2012). The developing Xenopus retino-tectal system has been used to assess chemical toxicity and screenpotential pharmaceutical drugs, which would provide us an effi-cient and convenient opportunity to treat pollutants-induced def-icits and diseases. Studies have found that para-xylene (PX) cancause developmental delay and neural apoptosis in the developingXenopus tectum through methylation and acetylation of histones(Gao et al., 2016a). Further studies have shown that PX exposureinduces deficits in neuronal structure and visual stimulation-induced excitatory compound synaptic currents without alteringneurotransmitter release probability (Liao et al., 2020). The toxiceffects of ethylbenzene and xylene are also revealed by inhibitingNMDA-induced currents (Cruz et al., 2000) and acetylcholine-induced ionic currents (van Kleef et al., 2008) in Xenopus oocytesexpressing NMDAR or a9a10 nAChR. Interestingly, the coapplica-tion of D-glucuronolactone (GA) with PX rescues all deficitsinduced by PX exposure (Gao et al., 2016a; Liao et al., 2020). Theseresults demonstrate that PX is toxic to brain development andplasticity and that GA may serve as a promising candidate to treatPX-induced defects in the developing brain.

The FETAX assay has been extensively used to assess teratogenicand toxic effects of chemical pollutants. Four aromatic ami-nesdacridine, aniline, pyridine, and quinoline have been revealedas potential environmental hazards following their exposure toX. laevis embryos (Davis et al., 1981). Polycyclic aromatic hydro-carbons (PAHs) and their derivatives have been shown to induceembryo malformations and oxidative stress at sublethal concen-trations (Buryskova et al., 2006). Alcohol ethoxylate (AE) andalcohol ethoxy sulfate (AES) cause teratogenic and toxic effects inXenopus embryos and tadpoles, in particular to the epithelia and gillcartilage (Cardellini and Ometto, 2001). Most importantly, re-searchers have used the Xenopus system to compare the toxicseverity score and find less toxic plasticizer. For example, theyexamined the developmental toxicity of citrate esters and indicatedthat triethyl 2-acetylcitrate (ATEC) could be considered an alter-native to the phthalate plasticizer of dibutyl-phthalate (DBP),which has higher toxicity in amphibian embryos (Xu and Gye,2018).

The phenotype of dispersion and aggregation of skin pigment isa good marker for monitoring chemical toxicity in water ecosys-tems. The first study to assess chemical toxicity in drinking waterwas performed in cultured Xenopus melanophores (Iuga et al.,2009). They found that some chemicals, including ammonia,arsenic, copper, mercury, pentachlorophenol, phenol, nicotine andparaquat, induce a rapid melanosome dispersion response, indi-cating that Xenopus melanophores may serve as a good toxicitysensor for screening toxicants. Environmental concentrations ofnitromusks, synthetic nitro-containing fragrances, are not

Table 1Summary of the toxicological studies performed in Xenopus tissues and organs.

Tissues/Organs Environmental factors Studies in Xenopus

Brain1,2,3,4,5

Aromatic amines; BTEX; nitromusks; pesticides; nanomaterials;methylmercury; alcohol; choline and nicotinic agonists; endosulfan;methylisothiazolinone; benomyl; DBP; CEP; triadimefon andtriadimenol; DHA; VPA; TNF-a; cis-bifenthrin and graphene oxide;thyroid hormone

(Davis et al., 1981); (Gao et al., 2016a); (Huyck et al., 2015); (Igarashiet al., 2015); (James et al., 2015); (Khakhalin et al., 2014); (Lee et al.,2010); (Li et al., 2020b); (Liao et al., 2020); (Preud’homme et al., 2015);(Shen et al., 2014); (Shi et al., 2014); (Spawn and Aizenman, 2012);(Thompson and Cline, 2016); (Titmus et al., 1999); (Yoon et al., 2003)

Embryo2,3,5,6

Benomyl; thyroid hormone; ethanol; polycyclic aromatic hydrocarbons;alcohol ethoxylate (AE) and alcohol ethoxy sulfate (AES); nitromusksatrazine; silver nanoparticles; methylmercury; rapamycin; cadmiumand zinc; regular cigarette butt (RCB), menthol (MCB) and electronic(ECB); Tributyltin (TBT) and triphenyltin (TPT); dibutyl phthalate andcitrate ester; quantum dots (QDs)

(Buryskova et al., 2006); (Cardellini and Ometto, 2001); (Chen et al.,2015); (Chou and Dietrich, 1999); (Colombo et al., 2017); (Galdieroet al., 2017); (Huyck et al., 2015); (Kim et al., 2020); (Moriyama et al.,2011); (Mouchet et al., 2015); (Parker and Rayburn, 2017); (Shi et al.,2014); (Shukrun et al., 2019); (Xu and Gye, 2018); (Yelin et al., 2007);(Yoon et al., 2003); (Yuan et al., 2011); (Zhu et al., 2018)

Eye6

Benomyl; BPA; thyroid hormone; ethanol; triphenyltin (TPT);butachlor; tire debris (TD); alcohol

(Baba et al., 2009); (Havis et al., 2006); (Li et al., 2016a); (Mantecca et al.,2007); (Peng et al., 2004); (Yelin et al., 2007); (Yoon et al., 2003); (Zhuet al., 2018)

Craniofacial3

Ethanol; thioridazine, ethanol and ICI 118,551; triadimefon; silvernanoparticles; Agþ; electronic cigarette (ECIG)

(Colombo et al., 2017); (Fainsod and Kot-Leibovich, 2018); (Fort et al.,2016); (Kennedy et al., 2017); (Li et al., 2017); (Li et al., 2020a);

Gonad7

BDE-47; atrazine; azocyclotin; boric acid; BPA; benzophenone-2; 17-ethinylestradiol (EE2); tebuconazole; triclosan

(Chen et al., 2015); (Fort et al., 2017); (Li et al., 2016b); (Haselman et al.,2016); (Hayes et al., 2010); (Pinet et al., 2019); (Poulsen et al., 2015);(Regnault et al., 2018); (Rimayi et al., 2018); (Sai et al., 2019); (Sai et al.,2020); (Tamschick et al., 2016)

Heart8,9

Chlorpyrifos; dichlorvos; diazinon; ketamine; dehydration; atrazine;thyroid hormone;

(Asouzu Johnson et al., 2019); (Guo et al., 2016); (Hawkins and Storey,2020); (Hwang et al., 2019); (Marshall et al., 2019); (Watson et al., 2014)

Intestine/gut5,6

Alcohol; thyroid hormone; aryl hydrocarbon receptor agonists; ethanol;TBBPA; nanoparticles; ZnO; polystyrene microspheres

(Bonfanti et al., 2015); (Bonfanti et al., 2020); (Colombo et al., 2017);(Hasebe et al., 2016); (Ikuzawa et al., 2006); (Ishizuya-Oka et al., 2001);(Ishizuya-Oka et al., 2014); (Hu et al., 2016); (Miller et al., 2013); (Taftet al., 2018); (Wang et al., 2017); (Yao et al., 2017); (Zhu et al., 2020);

Limb6

Dioxin; thyroid hormone; retinoic acid (Alsop et al., 2004); (Marsh-Armstrong et al., 2004); (Yokoyama et al.,2011)

Liver6

Triadimefon and triadimenol; azocyclotin; atrazine; benzophenone-2;benzo(a)pyrene and triclosan; paracetamol; tire debris organic extract(TDOE)

(Li et al., 2017); (Haselman et al., 2016); (Regnault et al., 2018); (Rimayiet al., 2018); (Saide et al., 2019); (Mantecca et al., 2007); (Zhang et al.,2020)

Skin4,6

Maltol; thyroid hormone; copper mercury; rapamycin; radiation;polystyrene nanoparticles

(Baba et al., 2009); (Carotenuto et al., 2016); (Dahora et al., 2020); (Iugaet al., 2009); (Moriyama et al., 2011); (Tussellino et al., 2015); (Yuanet al., 2011); (Zhang et al., 2014);

Spinal cord1,3,5,9

Muscarinic; thyroid hormone; aromatic amines; acridine; aniline;pyridine; quinoline; chlorpyrifos; dichlorvos; Methylisothiazolinone(MIT)

(Carotenuto et al., 2016); (Davis et al., 1981); (Marsh-Armstrong et al.,2004); (Munoz et al., 2015); (Porter and Li, 2018); (Schlosser et al.,2002); (Spawn and Aizenman, 2012); (Watson et al., 2014);

Tail3,4,9

BPA; thyroid hormone; hexabromocyclododecane (HBCD) and2,20 ,3,30 ,4,40 ,5,50 ,6-nona brominated diphenyl ether (BDE206);chlorpyrifos (CPF) and malathion (MTN); TrkA inhibitor; CuOnanoparticles; carbachol

(Bonfanti et al., 2004); (Das et al., 2002); (Helbing et al., 2003);(Iwamuro et al., 2006); (Iimura et al., 2020); (Li et al., 2020b); (Marsh-Armstrong et al., 2004); (Nations et al., 2015); (Porter and Li, 2018);(Schriks et al., 2006)

1, Behavior; 2, Mortality; 3, Malformation; 4, Metamorphosis; 5, Neurogenesis; 6, Embryogenesis; 7, Reproductive toxicity; 8, Heart dysfunction; 9, Regeneration.

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

hazardous for early developing amphibians and fish (Chou andDietrich, 1999). However, the other study using an in vivo Xen-opus model has shown that maltol, a naturally occurring flavorenhancer and fragrance agent, induces pigment aggregation in bothdura and dermal melanophores, suggesting a putative toxicologicaleffect of maltol exposure (Dahora et al., 2020). Therefore, bothexposure time and concentration of chemicals and the sites of ac-tion are important parameters to be considered for comprehen-sively evaluating the toxic effects of environmental factors.

The different impact of traditional tobacco and electronic ciga-rette on environmental toxicity can be directly assessed withexposure butt leachates to tadpoles. Evaluation of the develop-mental toxicities of three different cigarette butt leachates, regular(RCB), menthol (MCB) and electronic cigarette butt (ECB), hasshown that RCB leachates comprise the most toxic compound,while MCB leachates have higher teratogenicity (Parker andRayburn, 2017). One study has reported that craniofacial forma-tion is adversely affected by aerosol mixtures produced by e-liquidaerosolization in e-cigarettes (ECIGs), which are often advertised asa safe alternative to traditional tobacco smoking (Kennedy et al.,2017). These results confirm that the Xenopus model can be use-ful in an integrated biological hazard assessment for preliminaryscreening.

5

3.2. Pesticides

Nowadays, a variety type of pesticides, including herbicides,insecticides, fungicides, and rodenticides were proven efficient inpest control and crop productivity. However, it is known that due totheir toxicity, pesticides could have serious impacts on humanhealth as well as aquatic ecosystems (Christin et al., 2004). Thedevelopment of accurate and sensitive assessment of pesticidetoxicity is required to ensure public health. Several distinct chem-ical classes of pesticides have been primarily investigated in Xen-opus, such as chlorpyrifos (organophosphorus insecticide), atrazine(triazine herbicide), and triadimefon (conazole fungicide) (AsouzuJohnson et al., 2019; Bonfanti et al., 2004; Hayes et al., 2010;Rimayi et al., 2018; Sai et al., 2019; Watson et al., 2014; Zhang et al.,2020). However, theses pesticides exert differential toxic effects onembryogenesis, neurotransmitter release, and sexualdifferentiation.

Organophosphate pesticides, which act as a class of acetylcho-linesterase inhibitors, are used widely in agriculture to reduce in-sect populations. Chlorpyrifos (10 mM) and dichlorvos (0.1 mM),organophosphate insecticides, decrease heart rate, spine lengthand free-swimming activity of Xenopus (Bonfanti et al., 2004;Watson et al., 2014). Pyrethroid insecticide activates dopaminergic,noradrenergic, and serotonergic neurotransmitter systems, leading

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

to interference in metamorphic development (Li et al., 2020). Theherbicide atrazine, one of the most commonly applied pesticidesworldwide, affects the growth of X. laevis and may cause repro-ductive toxicity through the disruption of tight junctions andmetabolism-related signaling pathways in germ cells and turnmalefrogs into female frogs, some of which are able to mate and produceviable eggs (Carr et al., 2003; Chen et al., 2015; Hayes et al., 2010;Sai et al., 2020). However, the other studies have reported contro-versial results that atrazine in 10 ppb causes no adverse effects onmetamorphosis but affects sex differentiation by inducing femini-zation using all-ZZ-male cohorts (Oka et al., 2008). The discrepancyof these results may result from different water quality, aquariaenvironment and exposure conditions. Moreover, the most severetoxic effects of benomyl fungicide were observed in the nerve tis-sues of the Xenopus embryo, inducing deficits in neuronal prolif-eration and migration (Yoon et al., 2003). And these detrimentaleffects on the central nervous system may eventually result indeficits in swimming activity and visually mediated avoidancebehaviors (Spawn and Aizenman, 2012; Zhang et al., 2020).Furthermore, pesticides may produce sex-linked differences intoxicokinetics between male and female frogs. For instance, tri-adimefon and triadimenol exposure induces more antioxidantenzyme activities in females than in males, which result in moredamaged liver histology and thyroid hormone levels in males thanin females (Zhang et al., 2020). These studies suggest that pesticidesmay have a direct causal relationship with amphibian sex ratio andpopulation decline worldwide (Mann et al., 2009). Nevertheless,the convenience of monitoring chemical toxicity by bath exposureto Xenopus is limited by the fact that environmental exposure tohuman is mainly through ingestion or inhalation. And the detri-mental effects of chemical toxicants on embryonic tissues or organsmay not apply to adult organisms, which should be cautiouslyrelating these findings to human health.

3.3. Plastics and nanoparticles

A growing number of studies have highlighted that contami-nation from diverse microplastics (<5 mm in size) negatively af-fects the health status of aquatic species in the seas and oceansworldwide. Xenopus has become one of the most studied andsuitable organisms out of 11 amphibian species for assessing theimpact of nanomaterials on ecotoxicity on freshwater ecosystemsdue to its extensive use in embryotoxic studies (do Amaral et al.,2019).

The Xenopus system is a valuable model to observe the distri-bution of nanoparticles (NPs) in live tissues/organs and assess thetoxic effects in vivo. Xenopus tadpoles take up microplastics anddebris through the digestive tract from the surrounding water (DeFelice et al., 2018; Hu et al., 2018; Mantecca et al., 2007), whichcould decrease the food taking by absorbing more polystyreneparticles (Hu et al., 2016). Engineered aluminum oxide nano-particles Al2O3 NPs are developmentally toxic and teratogenic toX. laevis and D. rerio, shown as decreased embryo length, inducedorgan malformations, and altered heart-and liver-specific genesexpression (Ismail et al., 2019). Quantum dots (QDs) and multi-walled carbon nanotubes (MWCNTs) have lower toxic effects onembryo survival and phenotypes, which were found in the gills,lung, and intestine (Galdiero et al., 2017; Zhao et al., 2020). Unlikethe experimental limitation of cell cultures and mammalianmodels, Xenopus embryos can evaluate NP biointeractions at theintestinal barrier to determine the safety of NPs (Bacchetta et al.,2014; Bonfanti et al., 2015, 2020; Colombo et al., 2017). Forexample, positively charged silver NPs induced severe effects onembryo development by disrupting embryo tissues of the intestine(Colombo et al., 2017). Therefore, the use of X. laevis as an in vivo

6

toxicity model bridges the gap between in vitro culture assays androdent models, which allows for temporal control over the expo-sure to NPs at critical stages of embryogenesis, such as gastrulationand neurulation (Marin-Barba et al., 2018). Furthermore, Xenopusphenotypic scores with exposure to magnetite-cored NPs arehighly correlatedwith cell based in vitro assays as well as the test inmice, indicating that the Xenopus system provides a rapid andcheap identification of NP-induced toxicities (Webster et al., 2016).Nonetheless, the ability of tadpoles for taking up plastics and par-ticles is under the detection limit of the environmental particle size.The relevance between human risk assessment and detrimentaleffects on Xenopus tissues should be also carefully considered.

The toxic effects of NPs on TH-mediated amphibian meta-morphosis are dependent on the concentrations and exposuretime. Exposure to lower concentrations of CuO NPs stimulatesmetamorphosis and growth, whereas chronic exposure to 0.3 mg/LCuO elicits significant mortality and affects the rate of meta-morphosis (Nations et al., 2011, 2015). Environmentally-relevantlow concentrations (0.018 mg/L) of nanosilver particles (AgNPs)had developmental stage-specific endocrine disrupting effectsduring TH-dependent metamorphosis (Carew et al., 2015). Inparticular, microinjections of nanomaterials dispersed with Plur-onic F127 into one-to-two cell Xenopus embryos results in an activeexpulsion of nanomaterials due to the changes in membrane ac-tivity by Pluronic F127 (Holt et al., 2016). Thus, studies on X. laevisembryos allow us not only to better understand the toxicity,localization, and delivery efficacy of NPs but also to screen efficientreagents to react against the toxicity of NPs.

3.4. Endocrine-disrupting chemicals

Thyroid hormone regulates multiple aspects of neurogenesis,metamorphosis and metabolism in the early and later stages ofembryo development (Das et al., 2002; Havis et al., 2006; Marsh-Armstrong et al., 2004; Thompson and Cline, 2016). Abnormalthyrotropin-releasing hormone (TRH) secretion leads to thyroiddysfunction, which eventually leads to abnormal cell proliferationand differentiation, neuroendocrine disruption, and diseases(Wang et al., 2017). Xenopus has emerged as a powerful animalmodel for better understanding hormone balance and function dueto its high sensitivity to human reproductive hormones and thyroidhormone (TH)-dependent metamorphosis (Buchholz, 2017).

Endocrine-disrupting chemicals (EDCs) or endocrine disruptors(EDs) are exogenous chemicals or mixtures that attack the endo-crine system by interfering with endogenous hormones andconsequently cause adverse effects in reproductive developmentand sex differentiation. Some well-known EDCs, such as poly-chlorinated biphenyls (PCBs), atrazine, bisphenol A (BPA), anddibutyl phthalate (DBP), can cause feminizing effects on amphib-ians (Li et al., 2016). BPA, a major component of polycarbonateplastics, also induces various morphologic aberrations, includingscoliosis, eye dysplasia, otolith malformations and loss of pigmentsin the X. laevis tadpoles (Baba et al., 2009; Fini et al., 2009; Gibertet al., 2011; Tamschick et al., 2016). Flame retardant tetra-bromobisphenol A (TBBPA), a known TH signaling disruptor, showsvarious of neurotoxic, immunotoxic, reproductive toxic, and nu-clear receptor-mediated disruptive activities (Colnot et al., 2014;Schriks et al., 2006; Tamschick et al., 2016; Yu et al., 2019). FemaleX. tropicalis exposed to benzopyrene or triclosan EDCs at 50 ng/Ldisplayed a prediabetes state, leading to delayed metamorphosis,smaller individuals and reduced reproduction (Regnault et al.,2018).

Several studies have exerted their efforts to decipher the un-derlying mechanism for the signaling targets of EDCs. A relativelylow concentration of BPA (10�7 M) acts as an antagonist of T3

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

through suppression of TRa and TRb expression in Xenopus tailcultures (Iwamuro et al., 2006). Two EDC biocides, tributyltin (TBT)and triphenyltin (TPT), trigger severe malformations with defectiveeyes in X. tropicalis embryos by activation of the peroxisome pro-liferator activated receptor g (PPARg) (Yuan et al., 2011; Zhu et al.,2018). A recent study has shown that the environmental concen-tration of TBBPA affects intestinal development by altering cellproliferation and differentiation through Notch signaling (Zhuet al., 2020). Thus, Xenopus is considered an ideal model organ-ism for screening endocrine disruptors and understanding theintrinsic signaling pathways (Kloas et al., 1999).

3.5. Inhibitors and drugs

Xenopus has also been used to test the effects of chemicalcompounds or known reagents in treating tissues and organs,making Xenopus an excellent model to predict drug-inducedtoxicity (Maia et al., 2017). Rapamycin is commonly used as anantifungal agent, whereas it can cause developmental delay,pigmentation defects and gastrointestinal malformation in Xenopusembryogenesis (Moriyama et al., 2011). Valproic acid (VPA), ageneral histone deacetylase (HDAC) inhibitor, is one of the mostefficient antiepileptic drugs. However, it can induce neuro-developmental deficits and affect stem cell homeostasis (Gao et al.,2016b, 2020; Guo et al., 2015; James et al., 2015; Tao et al., 2015).Several other studies have tested the impacts of anti-toxic reagentson Xenopus embryos in addition to their protective effects. PXinduced deficits in the developing Xenopus tectum can be rescuedby GA, which is also beneficial to neural function and tadpolebehavior (Gao et al., 2016a; Liao et al., 2020). The effect of L-cysteinecan protect against the toxicity of acrylamide and furan, whereas itshows delayed embryo hatching within the first 24 h (Williamset al., 2014).

Currently, researchers tend to use Xenopus embryos for drugsafety assessment before performing expensive preclinical trials inmammals, such as antimalarial drug dihydroartemisinin (Longoet al., 2008; Richards and Cole, 2006; Wheeler and Brandli,2009). Decrease in glutathione levels by paracetamol, an antipy-retic and analgesic drug, has been used as an indicator ofparacetamol-induced liver injury, which helps predict the hep-atoxicity of the drug at early stages (Saide et al., 2019; Saide andWheeler, 2020). Celecoxib is a nonsteroidal anti-inflammatorydrug that is used to treat pain, fever, and inflammation by selec-tively inhibiting COX-2. However, celecoxib exposure inducesvarious malformations in Xenopus embryos as well as in humanumbilical vein endothelial cells, indicating its toxic and teratogeniceffects in developing vertebrates (Yoon et al., 2018). Ketamine, anoncompetitive inhibitor of NMDA receptors, is a commonly usedclinical anesthetic and sedative in humans. Studies have found thatketamine exposure has teratogenic effects on Xenopus embryoswith heart enlargement and ventricular shortening fraction (Guoet al., 2016). Waterborne antidepressants (amitriptyline, venlafax-ine, and sertraline) impact mRNA expression of genes related toheart, eye, brain, and bone development at environmentally rele-vant concentrations without changing Xenopus mortality(Sehonova et al., 2019). These studies provide an excellent oppor-tunity for in vivo assessing drug toxicity and screening potentialdrugs for the treatment of toxicant-induced diseases.

4. Disease modeling in Xenopus

Xenopus has served as a powerful tool for understanding humanphysiology in neurodevelopmental and biomedical research (Prattand Khakhalin, 2013; Tandon et al., 2017). Recent methodologicaladvances have allowed us to investigate genetic diseases in organ-

7

specific contexts in Xenopus, leading to a further understanding ofthe mechanisms of human diseases (Hwang et al., 2019). In the pastfew years, CRISPR/Cas9 technology has been available for screeningcandidate disease genes and create patient-specific mutations forhuman disease, including congenital heart diseases (CHD), Holt-Oram syndrome, oculocutaneous albinism type 1A, and cataractin Xenopus (Aslan et al., 2017; Bhattacharya et al., 2015; Sakaneet al., 2018; Shi et al., 2019; Viet et al., 2020). Recently, next-generation sequencing technologies have allowed researchers toidentify putative disease-causing patient variants. Using Xenopus asa model system for studying human disorders, such asWolfeHirschhorn syndrome (WHS), ciliopathies and airway dis-eases, is beneficial in understanding the mechanism of develop-mental diseases (Lasser et al., 2019; Walentek and Quigley, 2017).

More evidence has shown that environmental factors havebecome a major cause for some human diseases (Table 2). Alcoholexposure during pregnancy results in developmental defects, calledFASDs (Fainsod and Kot-Leibovich, 2018). Tumor necrosis factor a(TNF-a), a pro-inflammatory cytokine, increases the susceptibilityto pentylenetetrazol-induced seizures (Lee et al., 2010). Whilechemicals alone are sufficient to induce some disease phenotypes,environmental factors in tandem with other underlying geneticmechanisms may lead to varying degrees of severity in these dis-orders. By adopting these toxicological and developmental ap-proaches to connect environmental factors with candidate genes,we may be better equipped to determine the roles of genes in pa-tient diseases. In the following section, we mainly discuss recentstudies on chemical-induced diseasemodeling in Xenopus, focusingon the understanding of disease mechanisms and drug screening(Table 2).

4.1. Fetal alcohol spectrum disorders (FASD)

Accumulating evidence has shown that alcohol is detrimental totissue and organ development. Prenatal alcohol exposure results inFASDs, which show a series of developmental abnormalities andbirth defects (Lichtig et al., 2020; Yelin et al., 2005, 2007). Ethanolexposure in Xenopus induces similar outcomes as in humans, suchas craniofacial malformations, growth retardation, and centralnervous system deficiencies (Dubey and Saint-Jeannet, 2017;Fainsod and Kot-Leibovich, 2018; Peng et al., 2005; Shukrun et al.,2019). Frog embryos, along with those of zebrafish, have beenused to demonstrate the molecular and biochemical understandingof the etiology and pathophysiology of FASDs (Fernandes et al.,2018).

Several studies have reported that some compounds couldrescue alcohol exposure-induced defects in the developing tad-poles. Catalase and peroxiredoxin 5 can protect Xenopus embryosagainst alcohol-induced ocular malformation (Peng et al., 2004).Ethanol-induced developmental malformations can be rescued byexposure to retinol/retinaldehyde (Yelin et al., 2005) or 5-methyltetrahydrofolate (5-MTHF), the most bioactive form of folicacid, which could be beneficial for treating FASDs (Shi et al., 2014).Further research showed that delayed growth and microencephalyinduced by ethanol could be reversed by vitamin C treatmentthrough NF-kB inactivation and ROS inhibition (Peng et al., 2005).These findings indicate that Xenopus offers many tools to elucidatethe etiology of FAS-associated anomalies, which can help to identifymolecular targets for therapeutic strategies.

4.2. Seizure/epilepsy

During brain development, interruption of the excitation-to-inhibition balance will induce neurological disorders, such as epi-lepsy and autism (Preud’homme et al., 2015; Wang et al., 2018).

Table 2Selected chemical-induced diseases studied in Xenopus.

Diseases Chemicals Observations Studies in Xenopus

ASD VPA; TNF-a Increases excitation and decreasesexcitability

(Fainsod and Kot-Leibovich, 2018); (Faulkner et al., 2015);(James et al., 2015); (Lee et al., 2010)

Craniofacialdisorder

Thioridazine; ethanol; BPA, Bisphenol AF(BPAF), DBP; cyclopamine; aerosol mixtures;triadimefon

Craniofacial malformations (Arancio et al., 2019); (Deniz et al., 2017); (Dubey and Saint-Jeannet, 2017); (Groppelli et al., 2005); (Pinet et al., 2019)

Epilepsy/seizure

PTZ, methylisothiazolinone (MIT); TNF-a; kainicacid; bicuculline; picrotoxin; 4-aminopyridine;pilocarpine

Increases seizure activity andsusceptibility

(Bell et al., 2011); (Hewapathirane et al., 2008); (James et al.,2015); (Lee et al., 2010); (Spawn and Aizenman, 2012)

FASD Alcohol Short stature, microcephally and facialdysmorphogenesis

(Kot-Leibovich and Fainsod, 2009); (Shi et al., 2014); (Shukrunet al., 2019); (Yelin et al., 2005); (Yelin et al., 2007)

Metabolicsyndrome

benzo(a)pyrene or/and triclosan; retinoic acid; Induces prediabetes state (Regnault et al., 2018); (Usal et al., 2019); (Zhang et al., 2020):(Zhu et al., 2017)

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

Epilepsy is a common chronic central nervous system disease withclinical symptoms manifesting as repeated involuntary episodes.Seizures often cause aberrant and excessive neural activity, andmany antiepileptic drugs, such as phenobarbital, are intended topromote the release of GABA (g-aminobutyric acid) or enhanceGABA conduction (Bialer and White, 2010). Antiepileptic drugsprotect against seizures by regulating neuronal excitability throughNaþ and Ca2þ channels and GABAergic and glutamatergic neuro-transmission. The effects of various antiepileptic drugs wereexamined by using a Xenopus oocyte expression assay. Ethosux-imide selectively inhibits G protein-coupled inwardly rectifying Kþ

(GIRK) channels (Kobayashi et al., 2009). Carbamazepine (CBZ)produces a dose-dependent potentiation of the GABA current at aphysiological concentration in Xenopus oocytes expressing recom-binant GABAA receptors (Liu et al., 2006). Decanoic acid has beenfound to inhibit AMPAR-mediated excitatory neurotransmission,contributing to the anticonvulsant effect in Xenopus oocytes (Changet al., 2016). The oocyte screening system could facilitate researchon epilepsy mechanisms and therapeutic drug targets.

The members of the Hass lab have characterized an in vivoXenopus seizure model with bath application of several commonchemoconvulsants including pentylenetetrazol (PTZ), kainic acid,bicuculline, picrotoxin, 4-aminopyridine, or pilocarpine(Hewapathirane et al., 2008). PTZ-induced seizures in tadpoles aresimilar to the progressive nature of behavioral seizures and exhibitsimilar spike frequencies in zebrafish and rodent models, whichindicates that Xenopus is a powerful animal model for directlymonitoring seizure activity and studying the effects of seizures onneural function (James et al., 2015). VPA is a commonly prescribedantiepileptic drug with known teratogenic effects during earlydevelopment at the synaptic, cellular, circuit, and behavioral levels(Romoli et al., 2019). While tadpoles exposed to VPA have abnormalsensorimotor behaviors and increased seizure susceptibility (Jameset al., 2015), the delayed seizure onset times are due to the con-version of putrescine into GABA, which increase the frequency ofspontaneous GABAergic inhibitory postsynaptic currents. Onestudy showed that long-term developmental exposure to TNF-aresults in enhanced spontaneous excitatory synaptic transmission,increased AMPA/NMDA ratios, and decreased immature synapsesin tectal neurons, leading to increased incidences of epilepsy (Leeet al., 2010). The polyamines induced by a second PTZ exposurecan play a neuroprotective role in the developing brain (Bell et al.,2011). These findings will help researchers build a convenient andefficient Xenopus model for better understanding of the etiologyand pathology of seizure.

4.3. Autism spectrum disorder

Autism spectrum disorder (ASD) is a prevalent

8

neurodevelopmental disorder that is characterized by deficits inverbal communication, impairments in social interests and ste-reotyped behaviors (Huguet et al., 2013). Environmental factorssuch as mercury may contribute to the process of ASD and theoccurrence of epilepsy (Bozzi et al., 2018), while prenatal exposureto VPA is associated with an increased risk of ASD (Wang et al.,2018). VPA-induced changes in synaptic function, circuit forma-tion and behavior outcomes are remarkably conserved betweenvertebrate species and Xenopus models (James et al., 2015; Roulletet al., 2013). Therefore, we may take advantages of Xenopus modelto expand the understanding of the basic cellular and circuit levelchanges induced by VPA in vivo, which is difficult to be performedin other model species.

Fragile X syndrome (FXS) represents a known monogenic genecause of ASD. Lines of evidence have shown that FMR1 is highlyconserved between fruit flies, fish, amphibians, and humans (Limet al., 2005). FMR1 mRNA and protein are developmentally upre-gulated throughout Xenopus embryonic development in the optictectum (Faulkner et al., 2015; Lim et al., 2005). Loss of fragile Xmental retardation protein (FMRP) leads to a decrease in neuronalproliferation and survival but an increase in neuronal differentia-tion and deficient dendritic arbor elaboration during Xenopus em-bryonic development (Faulkner et al., 2015). FMRP knockdownusing morpholino oligos in the optic tectum results in decreasedswimming behavior and increased local protein synthesis, sug-gesting that changes at the cellular level are sufficient to cause abehavioral phenotype by enhancing synaptic inhibition (Liu andCline, 2016; Truszkowski et al., 2016). These studies demonstratepromise in the use of Xenopus as an in vivo animal model to identifythe fundamental mechanism of FXS and ASD across animal species.

4.4. Cardiovascular disease

Several studies have found that environmentally relevant con-centrations of chemicals are correlated to the development andprogress of cardiovascular diseases. The severe contamination withhigher PM10, which mainly consists of tire debris, causes cardio-vascular diseases and respiratory diseases (Mantecca et al., 2007).Most chemical compounds exert their toxic effects on animalhearts, including atrazine (Asouzu Johnson et al., 2019), ketamine(Guo et al., 2016), anti-cancer drugs (Isidori et al., 2016) andorganophosphate pesticides (Watson et al., 2014). The otherchemicals cause detrimental impacts on both vasculature andhearts, such as celecoxib, leading to haemorrhage and oedemaduring Xenopus development (Yoon et al., 2018).

Congenital heart disease (CHD) is a common birth defect withhigh rates of morbidity and mortality. CHD modeling is the mostfrequently annotated human disease ontology (DO) term in thecardiovascular disease category in the Xenbase database (Fortriede

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

et al., 2020; Nenni et al., 2019). The functions of CHD-linked genes,such as galnt11, nek2, tbx20 and bcor, have been well studied inXenopus as an efficient human disease model (Duncan and Khokha,2016). Other cardiovascular disease-related conditions, includinghypertension, long QT syndrome and atrial fibrillation, have beenthoroughly investigated by expressing cardiac channel proteins inXenopus oocytes (Hoppler and Conlon, 2019; Steffensen et al.,2015). Hence, the fast and efficient Xenopus model system cancomplement the established mouse and zebrafish model, and thusto further understand the pathophysiology underlying CHD andscreen potential drugs.

5. Future perspectives

The Xenopus model organism, commonly used in risk assess-ment and life science research, is of great significance for exploringthe mechanisms of embryo development and revealing the toxic-ities of environmental factors and related diseases. This reviewsummarized the main findings in taking advantage of Xenopus as avertebrate model to explore embryo development, neuro-developmental toxicity, and human disease modeling. Studying theeffects of chemical compounds on Xenopus tissues/organs,including those on the heart, brain, spinal cord, kidneys, intestine,eyes, and so on, will allow us to unravel the mechanisms of diseaseetiology and pathogenesis. It is surely that the usage of Xenopusoocytes and embryos wouldmake complementary advantages withthe common mouse and zebrafish model to contribute the basicscience research and biomedical application. Next steps will gathermore potential association between chemical pollutants and dis-eases, which can be used to underpin research on the relatedmechanisms of exogenous factor-induced diseases. To furthercombine with the methodological advances of genome editing,proteomics, and sequencing, the Xenopus systemwill also facilitateresearch on the pharmacological screening of potential pharma-ceutical drugs to treat chemical pollutant-induced deficits anddiseases (Sater and Moody, 2017; Tandon et al., 2017). We also lookforwardmore in-depth studies that use Xenopus systems to developmore human disease models for unveiling the functionality andmechanistic basis of environmental factors related diseases(Vismara et al., 2001). Thus, the unique advantage of Xenopus sys-tem has rightfully earned a place in assessing chemical toxicity andhuman disease modeling.

Author contributions

All authors listed havemade a substantial, direct and intellectualcontribution to the work, and approved it for publication.

Declaration of competing interest

The authors declare that they have no known competingfinancial interests or personal relationships that could haveappeared to influence the work reported in this paper.

Acknowledgements

This work was supported by the National Nature SciencesFoundation of China (NSFC 31871041 to WS).

References

Akerman, C.J., Cline, H.T., 2006. Depolarizing GABAergic conductances regulate thebalance of excitation to inhibition in the developing retinotectal circuit in vivo.J. Neurosci. 26, 5117e5130.

Alsop, D.H., Brown, S.B., van der Kraak, G.J., 2004. Dietary Retinoic Acid InducesHindlimb and Eye Deformities inXenopus laevis. Environ. Sci. Technol. 38,

9

6290e6299.Arancio, A.L., Cole, K.D., Dominguez, A.R., Cohenour, E.R., Kadie, J., Maloney, W.C.,

Cilliers, C., Schuh, S.M., 2019. Bisphenol A, Bisphenol AF, di-n-butyl phthalate,and 17b-estradiol have shared and unique dose-dependent effects on earlyembryo cleavage divisions and development in Xenopus laevis. Reprod. Toxicol.84, 65e74.

Aslan, Y., Tadjuidje, E., Zorn, A.M., Cha, S.W., 2017. High-efficiency non-mosaicCRISPR-mediated knock-in and indel mutation in F0 Xenopus. Development144, 2852e2858.

Asouzu Johnson, J., Ihunwo, A., Chimuka, L., Mbajiorgu, E.F., 2019. Cardiotoxicity inAfrican clawed frog (Xenopus laevis) sub-chronically exposed to environmen-tally relevant atrazine concentrations: implications for species survival. Aquat.Toxicol. 213, 105218.

Aztekin, C., Hiscock, T.W., Marioni, J.C., Gurdon, J.B., Simons, B.D., Jullien, J., 2019.Identification of a regeneration-organizing cell in the Xenopus tail. Science 364,653e658.

Baba, K., Okada, K., Kinoshita, T., Imaoka, S., 2009. Bisphenol A disrupts Notchsignaling by inhibiting gamma-secretase activity and causes eye dysplasia ofXenopus laevis. Toxicol. Sci. 108, 344e355.

Bacchetta, R., Moschini, E., Santo, N., Fascio, U., Del Giacco, L., Freddi, S.,Camatini, M., Mantecca, P., 2014. Evidence and uptake routes for Zinc oxidenanoparticles through the gastrointestinal barrier in Xenopus laevis. Nano-toxicology 8, 728e744.

Beck, C.W., Slack, J.M., 2001. An amphibian with ambition: a new role for Xenopusin the 21st century. Genome Biol. 2. REVIEWS1029.

Bell, M.R., Belarde, J.A., Johnson, H.F., Aizenman, C.D., 2011. A neuroprotective rolefor polyamines in a Xenopus tadpole model of epilepsy. Nat. Neurosci. 14,505e512.

Bestman, J.E., Cline, H.T., 2020. Morpholino studies in Xenopus brain development.Methods Mol. Biol. 2047, 377e395.

Bhattacharya, D., Marfo, C.A., Li, D., Lane, M., Khokha, M.K., 2015. CRISPR/Cas9: aninexpensive, efficient loss of function tool to screen human disease genes inXenopus. Dev. Biol. 408, 196e204.

Bialer, M., White, H.S., 2010. Key factors in the discovery and development of newantiepileptic drugs. Nat. Rev. Drug Discov. 9, 68e82.

Blum, M., Ott, T., 2018. Xenopus: an undervalued model organism to study andmodel human genetic disease. Cells Tissues Organs 205, 303e313.

Bonfanti, P., Colombo, A., Orsi, F., Nizzetto, I., Andrioletti, M., Bacchetta, R.,Mantecca, P., Fascio, U., Vailati, G., Vismara, C., 2004. Comparative teratogenicityof chlorpyrifos and malathion on Xenopus laevis development. Aquat. Toxicol.70, 189e200.

Bonfanti, P., Colombo, A., Saibene, M., Fiandra, L., Armenia, I., Gamberoni, F.,Gornati, R., Bernardini, G., Mantecca, P., 2020. Iron nanoparticle bio-interactionsevaluated in Xenopus laevis embryos, a model for studying the safety ofingested nanoparticles. Nanotoxicology 14, 196e213.

Bonfanti, P., Moschini, E., Saibene, M., Bacchetta, R., Rettighieri, L., Calabri, L.,Colombo, A., Mantecca, P., 2015. Do nanoparticle physico-chemical propertiesand developmental exposure window influence nano ZnO embryotoxicity inXenopus laevis? Int. J. Environ. Res. Publ. Health 12, 8828e8848.

Borodinsky, L.N., 2017. Xenopus laevis as a model organism for the study of spinalcord formation, development, function and regeneration. Front. Neural Circ. 11,90.

Bozzi, Y., Provenzano, G., Casarosa, S., 2018. Neurobiological bases of autism-epilepsy comorbidity: a focus on excitation/inhibition imbalance. Eur. J. Neu-rosci. 47, 534e548.

Buchholz, D.R., 2017. Xenopus metamorphosis as a model to study thyroid hormonereceptor function during vertebrate developmental transitions. Mol. Cell.Endocrinol. 459, 64e70.

Buryskova, B., Hilscherova, K., Blaha, L., Marsalek, B., Holoubek, I., 2006. Toxicity andmodulations of biomarkers in Xenopus laevis embryos exposed to polycyclicaromatic hydrocarbons and their N-heterocyclic derivatives. Environ. Toxicol.21, 590e598.

Cardellini, P., Ometto, L., 2001. Teratogenic and toxic effects of alcohol ethoxylateand alcohol ethoxy sulfate surfactants on Xenopus laevis embryos and tadpoles.Ecotoxicol. Environ. Saf. 48, 170e177.

Carew, A.C., Hoque, M.E., Metcalfe, C.D., Peyrot, C., Wilkinson, K.J., Helbing, C.C.,2015. Chronic sublethal exposure to silver nanoparticles disrupts thyroid hor-mone signaling during Xenopus laevis metamorphosis. Aquat. Toxicol. 159,99e108.

Carotenuto, R., Tussellino, M., Mettivier, G., Russo, P., 2016. Survival fraction andphenotype alterations of Xenopus laevis embryos at 3 Gy, 150 kV X-ray irra-diation. Biochem. Biophys. Res. Commun. 480, 580e585.

Carr, J.A., Gentles, A., Smith, E.E., Goleman, W.L., Urquidi, L.J., Thuett, K., Kendall, R.J.,Giesy, J.P., Gross, T.S., Solomon, K.R., Van Der Kraak, G., 2003. Response of larvalXenopus laevis to atrazine: assessment of growth, metamorphosis, and gonadaland laryngeal morphology. Environ. Toxicol. Chem. 22, 396e405.

Chang, P., Augustin, K., Boddum, K., Williams, S., Sun, M., Terschak, J.A.,Hardege, J.D., Chen, P.E., Walker, M.C., Williams, R.S., 2016. Seizure control bydecanoic acid through direct AMPA receptor inhibition. Brain 139, 431e443.

Chen, X., Wang, J., Zhu, H., Ding, J., Peng, Y., 2015. Proteomics analysis of Xenopuslaevis gonad tissue following chronic exposure to atrazine. Environ. Toxicol.Chem. 34, 1770e1777.

Chou, Y.J., Dietrich, D.R., 1999. Toxicity of nitromusks in early lifestages of SouthAfrican clawed frog (Xenopus laevis) and zebrafish (Danio rerio). Toxicol. Lett.111, 17e25.

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

Christin, M.S., M�enard, L., Gendron, A.D., Ruby, S., Cyr, D., Marcogliese, D.J., Rollins-Smith, L., Fournier, M., 2004. Effects of agricultural pesticides on the immunesystem of Xenopus laevis and Rana pipiens. Aquat. Toxicol. 67, 33e43.

Ciarleglio, C.M., Khakhalin, A.S., Wang, A.F., Constantino, A.C., Yip, S.P.,Aizenman, C.D., 2015. Multivariate analysis of electrophysiological diversity ofXenopus visual neurons during development and plasticity. Elife 4.

Cizelsky, W., Tata, A., Kuhl, M., Kuhl, S.J., 2014. The Wnt/JNK signaling target genealcam is required for embryonic kidney development. Development 141,2064e2074.

Colnot, T., Kacew, S., Dekant, W., 2014. Mammalian toxicology and human expo-sures to the flame retardant 2,2’,6,6’-tetrabromo-4,4’-isopropylidenediphenol(TBBPA): implications for risk assessment. Arch. Toxicol. 88, 553e573.

Colombo, A., Saibene, M., Moschini, E., Bonfanti, P., Collini, M., Kasemets, K.,Mantecca, P., 2017. Teratogenic hazard of BPEI-coated silver nanoparticles toXenopus laevis. Nanotoxicology 11, 405e418.

Cross, M.K., Powers, M.A., 2009. Learning about cancer from frogs: analysis ofmitotic spindles in Xenopus egg extracts. Dis Model Mech 2, 541e547.

Cruz, S.L., Balster, R.L., Woodward, J.J., 2000. Effects of volatile solvents on recom-binant N-methyl-D-aspartate receptors expressed in Xenopus oocytes. Br. J.Pharmacol. 131, 1303e1308.

Cruz, S.L., Mirshahi, T., Thomas, B., Balster, R.L., Woodward, J.J., 1998. Effects of theabused solvent toluene on recombinant N-methyl-D-aspartate and non-N-methyl-D-aspartate receptors expressed in Xenopus oocytes. J. Pharmacol.Exp. Therapeut. 286, 334e340.

Dahora, L.I., Fitzgerald, A., Emanuel, M., Baiges, A.F., Husain, Z., Thompson, C.K.,2020. The flavor enhancer maltol increases pigment aggregation in dermal andneural melanophores in Xenopus laevis tadpoles. Environ. Toxicol. Chem. 39,381e395.

Das, B., Schreiber, A.M., Huang, H., Brown, D.D., 2002. Multiple thyroid hormone-induced muscle growth and death programs during metamorphosis in Xen-opus laevis. Proc. Natl. Acad. Sci. U. S. A. 99, 12230e12235.

Davis, K.R., Schultz, T.W., Dumont, J.N., 1981. Toxic and teratogenic effects ofselected aromatic amines on embryos of the amphibian Xenopus laevis. Arch.Environ. Contam. Toxicol. 10, 371e391.

De Felice, B., Bacchetta, R., Santo, N., Tremolada, P., Parolini, M., 2018. Polystyrenemicroplastics did not affect body growth and swimming activity in Xenopuslaevis tadpoles. Environ. Sci. Pollut. Res. Int. 25, 34644e34651.

Deniz, E., Jonas, S., Hooper, M., Griffin, J.N., Choma, M.A., Khokha, M.K., 2017.Analysis of craniocardiac malformations in Xenopus using optical coherencetomography. Sci. Rep. 7, 42506.

Desgrange, A., Cereghini, S., 2015. Nephron patterning: lessons from Xenopus,zebrafish, and mouse studies. Cells 4, 483e499.

do Amaral, D.F., Guerra, V., Motta, A.G.C., de Melo, E.S.D., Rocha, T.L., 2019. Eco-toxicity of nanomaterials in amphibians: a critical review. Sci. Total Environ.686, 332e344.

Dong, W., Aizenman, C.D., 2012. A competition-based mechanism mediates devel-opmental refinement of tectal neuron receptive fields. J. Neurosci. 32,16872e16879.

Dubey, A., Saint-Jeannet, J.P., 2017. Modeling human craniofacial disorders in Xen-opus. Curr Pathobiol Rep 5, 79e92.

Duncan, A.R., Khokha, M.K., 2016. Xenopus as a model organism for birth defects-Congenital heart disease and heterotaxy. Semin. Cell Dev. Biol. 51, 73e79.

Edwards-Faret, G., Munoz, R., Mendez-Olivos, E.E., Lee-Liu, D., Tapia, V.S., Larrain, J.,2017. Spinal cord regeneration in Xenopus laevis. Nat. Protoc. 12, 372e389.

Fainsod, A., Kot-Leibovich, H., 2018. Xenopus embryos to study fetal alcohol syn-drome, a model for environmental teratogenesis. Biochem. Cell. Biol. 96, 77e87.

Faulkner, R.L., Wishard, T.J., Thompson, C.K., Liu, H.H., Cline, H.T., 2015. FMRP reg-ulates neurogenesis in vivo in Xenopus laevis tadpoles. eNeuro 2, e0055.

Fernandes, Y., Buckley, D.M., Eberhart, J.K., 2018. Diving into the world of alcoholteratogenesis: a review of zebrafish models of fetal alcohol spectrum disorder.Biochem. Cell. Biol. 96, 88e97.

Fini, J.B., Dolo, L., Cravedi, J.P., Demeneix, B., Zalko, D., 2009. Metabolism of theendocrine disruptor BPA by Xenopus laevis tadpoles. Ann. N. Y. Acad. Sci. 1163,394e397.

Fort, D.J., Fort, T.D., Mathis, M.B., Ball, R.W., 2016. Boric acid is reproductively toxic toadult Xenopus laevis, but not endocrine active. Toxicol. Sci. 154, 16e26.

Fort, D.J., Mathis, M., 2018. Frog embryo teratogenesis assay-Xenopus (FETAX): usein alternative preclinical safety assessment. Cold Spring Harb Protoc 2018 8.

Fort, D.J., Mathis, M.B., Pawlowski, S., Wolf, J.C., Peter, R., Champ, S., 2017. Effect oftriclosan on anuran development and growth in a larval amphibian growth anddevelopment assay. J. Appl. Toxicol. 37, 1182e1194.

Fortriede, J.D., Pells, T.J., Chu, S., Chaturvedi, P., Wang, D., Fisher, M.E., James-Zorn, C.,Wang, Y., Nenni, M.J., Burns, K.A., Lotay, V.S., Ponferrada, V.G., Karimi, K.,Zorn, A.M., Vize, P.D., 2020. Xenbase: deep integration of GEO & SRA RNA-seqand ChIP-seq data in a model organism database. Nucleic Acids Res. 48.D776ed782.

Frontera, J.L., Raices, M., Cervino, A.S., Pozzi, A.G., Paz, D.A., 2016a. Neural regen-eration dynamics of Xenopus laevis olfactory epithelium after zinc sulfate-induced damage. J. Chem. Neuroanat. 77, 1e9.

Frontera, J.L., Raices, M., Cervino, A.S., Pozzi, A.G., Paz, D.A., 2016b. Neural regen-eration dynamics of Xenopus laevis olfactory epithelium after zinc sulfate-induced damage. J. Chem. Neuroanat. 77, 1e9.

Fu, L., Das, B., Matsuura, K., Fujimoto, K., Heimeier, R.A., Shi, Y.B., 2017. Genome-wide identification of thyroid hormone receptor targets in the remodeling in-testine during Xenopus tropicalis metamorphosis. Sci. Rep. 7, 6414.

10

Galdiero, E., Falanga, A., Siciliano, A., Maselli, V., Guida, M., Carotenuto, R.,Tussellino, M., Lombardi, L., Benvenuto, G., Galdiero, S., 2017. Daphnia magnaand Xenopus laevis as in vivo models to probe toxicity and uptake of quantumdots functionalized with gH625. Int. J. Nanomed. 12, 2717e2731.

Gao, J., Liao, Y., Qiu, M., Shen, W., 2020. Wnt/beta-Catenin signaling in neural stemcell homeostasis and neurological diseases. Neuroscientist, 1073858420914509.

Gao, J., Ruan, H., Qi, X., Guo, X., Zheng, J., Liu, C., Fang, Y., Huang, M., Xu, M., Shen, W.,2016a. Increased apoptosis and abnormal visual behavior by histone modifi-cations with exposure to para-xylene in developing Xenopus. Neuroscience 331,177e185.

Gao, J., Ruan, H., Qi, X., Tao, Y., Guo, X., Shen, W., 2016b. HDAC3 but not HDAC2mediates visual experience-dependent radial Glia proliferation in the devel-oping Xenopus tectum. Front. Cell. Neurosci. 10, 221.

Gibert, Y., Sassi-Messai, S., Fini, J.B., Bernard, L., Zalko, D., Cravedi, J.P., Balaguer, P.,Andersson-Lendahl, M., Demeneix, B., Laudet, V., 2011. Bisphenol A inducesotolith malformations during vertebrate embryogenesis. BMC Dev. Biol. 11, 4.

Glasgow, S.D., McPhedrain, R., Madranges, J.F., Kennedy, T.E., Ruthazer, E.S., 2019.Approaches and limitations in the investigation of synaptic transmission andplasticity. Front. Synaptic Neurosci. 11, 20.

Groppelli, S., Pennati, R., De Bernardi, F., Menegola, E., Giavini, E., Sotgia, C., 2005.Teratogenic effects of two antifungal triazoles, triadimefon and triadimenol, onXenopus laevis development: Craniofacial defects. Aquat. Toxicol. 73, 370e381.

Guo, R., Liu, G., Du, M., Shi, Y., Jiang, P., Liu, X., Liu, L., Liu, J., Xu, Y., 2016. Early ke-tamine exposure results in cardiac enlargement and heart dysfunction inXenopus embryos. BMC Anesthesiol. 16, 23.

Guo, X., Ruan, H., Li, X., Qin, L., Tao, Y., Qi, X., Gao, J., Gan, L., Duan, S., Shen, W., 2015.Subcellular localization of class I histone deacetylases in the developing Xen-opus tectum. Front. Cell. Neurosci. 9, 510.

Hardwick, L.J., Philpott, A., 2015. An oncologists friend: how Xenopus contributes tocancer research. Dev. Biol. 408, 180e187.

Hasebe, T., Fujimoto, K., Kajita, M., Fu, L., Shi, Y.B., Ishizuya-Oka, A., 2017. Thyroidhormone-induced activation of Notch signaling is required for adult intestinalstem cell development during Xenopus laevis metamorphosis. Stem Cell. 35,1028e1039.

Hasebe, T., Fujimoto, K., Kajita, M., Ishizuya-Oka, A., 2016. Thyroid hormone acti-vates Wnt/beta-catenin signaling involved in adult epithelial developmentduring intestinal remodeling in Xenopus laevis. Cell Tiss. Res. 365, 309e318.

Haselman, J.T., Sakurai, M., Watanabe, N., Goto, Y., Onishi, Y., Ito, Y., Onoda, Y.,Kosian, P.A., Korte, J.J., Johnson, R.D., Iguchi, T., Degitz, S.J., 2016. Development ofthe Larval Amphibian Growth and Development Assay: Effects ofbenzophenone-2 exposure in Xenopus laevis from embryo to juvenile. J. Appl.Toxicol. 36, 1651e1661.

Havis, E., Le Mevel, S., Morvan Dubois, G., Shi, D.L., Scanlan, T.S., Demeneix, B.A.,Sachs, L.M., 2006. Unliganded thyroid hormone receptor is essential for Xen-opus laevis eye development. EMBO J. 25, 4943e4951.

Hawkins, L.J., Storey, K.B., 2020. MicroRNA expression in the heart of Xenopus laevisfacilitates metabolic adaptation to dehydration. Genomics 112, 3525e3536.

Hayes, T.B., Khoury, V., Narayan, A., Nazir, M., Park, A., Brown, T., Adame, L., Chan, E.,Buchholz, D., Stueve, T., Gallipeau, S., 2010. Atrazine induces complete femini-zation and chemical castration in male African clawed frogs (Xenopus laevis).Proc. Natl. Acad. Sci. U. S. A. 107, 4612e4617.

He, H.Y., Shen, W., Zheng, L., Guo, X., Cline, H.T., 2018. Excitatory synapticdysfunction cell-autonomously decreases inhibitory inputs and disruptsstructural and functional plasticity. Nat. Commun. 9, 2893.

Helbing, C.C., Werry, K., Crump, D., Domanski, D., Veldhoen, N., Bailey, C.M., 2003.Expression Profiles of Novel Thyroid Hormone-Responsive Genes and Proteinsin the Tail ofXenopus laevisTadpoles Undergoing Precocious Metamorphosis.Mol. Endocrinol. 17, 1395e1409.

Herrera-Rincon, C., Golding, A.S., Moran, K.M., Harrison, C., Martyniuk, C.J.,Guay, J.A., Zaltsman, J., Carabello, H., Kaplan, D.L., Levin, M., 2018. Brief localapplication of progesterone via a wearable bioreactor induces long-termregenerative response in adult Xenopus hindlimb. Cell Rep. 25, 1593e1609e1597.

Hewapathirane, D.S., Dunfield, D., Yen, W., Chen, S., Haas, K., 2008. In vivo imagingof seizure activity in a novel developmental seizure model. Exp. Neurol. 211,480e488.

Hiramoto, M., Cline, H.T., 2014. Optic flow instructs retinotopic map formationthrough a spatial to temporal to spatial transformation of visual information.Proc. Natl. Acad. Sci. U. S. A. 111, E5105eE5113.

Hodskinson, M.R., Bolner, A., Sato, K., Kamimae-Lanning, A.N., Rooijers, K., Witte, M.,Mahesh, M., Silhan, J., Petek, M., Williams, D.M., Kind, J., Chin, J.W., Patel, K.J.,Knipscheer, P., 2020. Alcohol-derived DNA crosslinks are repaired by twodistinct mechanisms. Nature 579, 603e608.

Holt, B.D., Shawky, J.H., Dahl, K.N., Davidson, L.A., Islam, M.F., 2016. DevelopingXenopus embryos recover by compacting and expelling single wall carbonnanotubes. J. Appl. Toxicol. 36, 579e585.

Hoogenboom, W.S., Klein Douwel, D., Knipscheer, P., 2017. Xenopus egg extract: apowerful tool to study genome maintenance mechanisms. Dev. Biol. 428,300e309.

Hoppler, S., Conlon, F.L., 2019. Xenopus: experimental access to cardiovasculardevelopment, regeneration discovery, and cardiovascular heart-defectmodeling. Cold Spring Harb Perspect Biol 12 (6).

Hu, L., Chernick, M., Hinton, D.E., Shi, H., 2018. Microplastics in small waterbodiesand tadpoles from Yangtze river delta, China. Environ. Sci. Technol. 52,8885e8893.

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

Hu, L., Su, L., Xue, Y., Mu, J., Zhu, J., Xu, J., Shi, H., 2016. Uptake, accumulation andelimination of polystyrene microspheres in tadpoles of Xenopus tropicalis.Chemosphere 164, 611e617.

Huguet, G., Ey, E., Bourgeron, T., 2013. The genetic landscapes of autism spectrumdisorders. Annu. Rev. Genom. Hum. Genet. 14, 191e213.

Huyck, R.W., Nagarkar, M., Olsen, N., Clamons, S.E., Saha, M.S., 2015. Methylmercuryexposure during early Xenopus laevis development affects cell proliferation anddeath but not neural progenitor specification. Neurotoxicol. Teratol. 47,102e113.

Hwang, W.Y., Marquez, J., Khokha, M.K., 2019. Xenopus: driving the discovery ofnovel genes in patient disease and their underlying pathological mechanismsrelevant for organogenesis. Front. Physiol. 10, 953.

Hyde, A.S., Hang, B.I., Lee, E., 2016. Reconstitution of the cytoplasmic regulation ofthe wnt signaling pathway using Xenopus egg extracts. Wnt Signaling 101e109.

Igarashi, M., Santos, R.A., Cohen-Cory, S., 2015. Impact of maternal n-3 poly-unsaturated fatty acid deficiency on dendritic arbor morphology and connec-tivity of developing Xenopus laevis central neurons in vivo. J. Neurosci. 35,6079e6092.

Iimura, A., Nishida, E., Kusakabe, M., 2020. Role of TrkA signaling during tadpole tailregeneration and early embryonic development in Xenopus laevis. Gene. Cell25, 86e99.

Ikuzawa, M., Shimizu, K., Yasumasu, S., Iuchi, I., Shi, Y.B., Ishizuya-Oka, A., 2006.Thyroid hormone-induced expression of a bZip-containing transcription factoractivates epithelial cell proliferation during Xenopus larval-to-adult intestinalremodeling. Dev. Gene. Evol. 216, 109e118.

Imaoka, S., Muraguchi, T., Kinoshita, T., 2007. Isolation of Xenopus HIF-prolyl 4-hydroxylase and rescue of a small-eye phenotype caused by Siah 2 over-expression. Biochem. Biophys. Res. Commun. 355, 419e425.

Ishizuya-Oka, A., Kajita, M., Hasebe, T., 2014. Thyroid Hormone-Regulated Wnt5a/Ror2 Signaling Is Essential for Dedifferentiation of Larval Epithelial Cells intoAdult Stem Cells in the Xenopus laevis Intestine. PLoS ONE 9, e107611.

Ishizuya-Oka, A., Ueda, S., Inokuchi, T., Amano, T., Damjanovski, S., Stolow, M.,Shi, Y.-B., 2001. Thyroid hormone-induced expression of Sonic hedgehog cor-relates with adult epithelial development during remodeling of the Xenopusstomach and intestine. Differentiation 69, 27e37.

Isidori, M., Piscitelli, C., Russo, C., Smutna, M., Blaha, L., 2016. Teratogenic effects offive anticancer drugs on Xenopus laevis embryos. Ecotoxicol. Environ. Saf. 133,90e96.

Ismail, T., Lee, H.K., Kim, C., Kim, Y., Lee, H., Kim, J.H., Kwon, S., Huh, T.L., Khang, D.,Kim, S.H., Choi, S.C., Lee, H.S., 2019. Comparative analysis of the developmentaltoxicity in Xenopus laevis and Danio rerio induced by Al2 O3 nanoparticleexposure. Environ. Toxicol. Chem. 38, 2672e2681.

Iuga, A., Lerner, E., Shedd, T.R., van der Schalie, W.H., 2009. Rapid responses of amelanophore cell line to chemical contaminants in water. J. Appl. Toxicol. 29,346e349.

Iwamuro, S., Yamada, M., Kato, M., Kikuyama, S., 2006. Effects of bisphenol A onthyroid hormone-dependent up-regulation of thyroid hormone receptor alphaand beta and down-regulation of retinoid X receptor gamma in Xenopus tailculture. Life Sci. 79, 2165e2171.

James, E.J., Gu, J., Ramirez-Vizcarrondo, C.M., Hasan, M., Truszkowski, T.L., Tan, Y.,Oupravanh, P.M., Khakhalin, A.S., Aizenman, C.D., 2015. Valproate-inducedneurodevelopmental deficits in Xenopus laevis tadpoles. J. Neurosci. 35,3218e3229.

Kennedy, A.E., Kandalam, S., Olivares-Navarrete, R., Dickinson, A.J.G., 2017. E-ciga-rette aerosol exposure can cause craniofacial defects in Xenopus laevis embryosand mammalian neural crest cells. PloS One 12, e0185729.

Khakhalin, A.S., Koren, D., Gu, J., Xu, H., Aizenman, C.D., 2014. Excitation and inhi-bition in recurrent networks mediate collision avoidance in Xenopus tadpoles.Eur. J. Neurosci. 40, 2948e2962.

Kim, H.Y., Jackson, T.R., Stuckenholz, C., Davidson, L.A., 2020. Tissue mechanicsdrives regeneration of a mucociliated epidermis on the surface of Xenopusembryonic aggregates. Nat. Commun. 11, 665.

Kloas, W., Lutz, I., Einspanier, R., 1999. Amphibians as a model to study endocrinedisruptors: II. Estrogenic activity of environmental chemicals in vitro andin vivo. Sci. Total Environ. 225, 59e68.

Kobayashi, T., Hirai, H., Iino, M., Fuse, I., Mitsumura, K., Washiyama, K., Kasai, S.,Ikeda, K., 2009. Inhibitory effects of the antiepileptic drug ethosuximide on Gprotein-activated inwardly rectifying Kþ channels. Neuropharmacology 56,499e506.

Kot-Leibovich, H., Fainsod, A., 2009. Ethanol induces embryonic malformations bycompeting for retinaldehyde dehydrogenase activity during vertebrate gastru-lation. Disease Models & Mechanisms 2, 295e305.

Kutsarova, E., Munz, M., Ruthazer, E.S., 2016. Rules for shaping neural connectionsin the developing brain. Front. Neural Circ. 10, 111.

Lasser, M., Pratt, B., Monahan, C., Kim, S.W., Lowery, L.A., 2019. The many faces ofXenopus: Xenopus laevis as a model system to study wolf-hirschhorn syn-drome. Front. Physiol. 10, 817.

Lee, R.H., Mills, E.A., Schwartz, N., Bell, M.R., Deeg, K.E., Ruthazer, E.S., Marsh-Armstrong, N., Aizenman, C.D., 2010. Neurodevelopmental effects of chronicexposure to elevated levels of pro-inflammatory cytokines in a developing vi-sual system. Neural Dev. 5, 2.

Li, S., Li, M., Gui, W., Wang, Q., Zhu, G., 2017. Disrupting effects of azocyclotin to thehypothalamo-pituitary-gonadal axis and reproduction of Xenopus laevis. Aquat.Toxicol. 185, 121e128.

Li, M., Zhu, J., Fang, H., Wang, M., Wang, Q., Zhou, B., 2020. Coexposure to

11

environmental concentrations of cis-bifenthrin and graphene oxide: adverseeffects on the nervous system during metamorphic development of Xenopuslaevis. J. Hazard Mater. 381, 120995.

Li, Y.Y., Chen, J., Qin, Z.F., 2016. Determining the optimal developmental stages ofXenopus laevis for initiating exposures to chemicals for sensitively detectingtheir feminizing effects on gonadal differentiation. Aquat. Toxicol. 179, 134e142.

Liao, S., Dong, W., Lv, L., Guo, H., Yang, J., Zhao, H., Huang, R., Yuan, Z., Chen, Y.,Feng, S., Zheng, X., Huang, J., Huang, W., Qi, X., Cai, D., 2017. Heart regenerationin adult Xenopus tropicalis after apical resection. Cell Biosci. 7, 70.

Liao, Y., Luo, Y., Ding, N., Gao, J., Wang, X., Shen, W., 2020. d-Glucuronolactone at-tenuates para-xylene-induced defects in neuronal development and plasticityin Xenopus tectum in vivo. Toxicology 430, 152341.

Lichtig, H., Artamonov, A., Polevoy, H., Reid, C.D., Bielas, S.L., Frank, D., 2020.Modeling bainbridge-ropers syndrome in Xenopus laevis embryos. Front.Physiol. 11, 75.

Lim, J.H., Luo, T., Sargent, T.D., Fallon, J.R., 2005. Developmental expression ofXenopus fragile X mental retardation-1 gene. Int. J. Dev. Biol. 49, 981e984.

Liu, H.H., Cline, H.T., 2016. Fragile X mental retardation protein is required tomaintain visual conditioning-induced behavioral plasticity by limiting localprotein synthesis. J. Neurosci. 36, 7325e7339.

Liu, H.H., McClatchy, D.B., Schiapparelli, L., Shen, W., Yates 3rd, J.R., Cline, H.T., 2018.Role of the visual experience-dependent nascent proteome in neuronal plas-ticity. Elife 7.

Liu, L., Zheng, T., Morris, M.J., Wallengren, C., Clarke, A.L., Reid, C.A., Petrou, S.,O’Brien, T.J., 2006. The mechanism of carbamazepine aggravation of absenceseizures. J. Pharmacol. Exp. Therapeut. 319, 790e798.

Liu, X.F., Haas, K., 2011. Single-cell electroporation of Xenopus tadpole tectal neu-rons. Cold Spring Harb Protoc 2011 9.

Longo, M., Zanoncelli, S., Della Torre, P., Rosa, F., Giusti, A., Colombo, P., Brughera, M.,Mazu�e, G., Olliaro, P., 2008. Investigations of the effects of the antimalarial drugdihydroartemisinin (DHA) using the Frog Embryo Teratogenesis Assay-Xenopus(FETAX). Reprod. Toxicol. 25, 433e441.

Luehders, K., Sasai, N., Davaapil, H., Kurosawa-Yoshida, M., Hiura, H., Brah, T.,Ohnuma, S., 2015. The small leucine-rich repeat secreted protein Asporin in-duces eyes in Xenopus embryos through the IGF signalling pathway. Develop-ment 142, 3351e3361.

Maia, L.A., Velloso, I., Abreu, J.G., 2017. Advances in the use of Xenopus for successfuldrug screening. Expet Opin. Drug Discov. 12, 1153e1159.

Mann, R.M., Hyne, R.V., Choung, C.B., Wilson, S.P., 2009. Amphibians and agricul-tural chemicals: review of the risks in a complex environment. Environ. Pollut.157, 2903e2927.

Mantecca, P., Gualtieri, M., Andrioletti, M., Bacchetta, R., Vismara, C., Vailati, G.,Camatini, M., 2007. Tire debris organic extract affects Xenopus development.Environ. Int. 33, 642e648.

Marin-Barba, M., Gavilan, H., Gutierrez, L., Lozano-Velasco, E., Rodriguez-Ramiro, I.,Wheeler, G.N., Morris, C.J., Morales, M.P., Ruiz, A., 2018. Unravelling themechanisms that determine the uptake and metabolism of magnetic single andmulticore nanoparticles in a Xenopus laevis model. Nanoscale 10, 690e704.

Marsh-Armstrong, N., Cai, L., Brown, D.D., 2004. Thyroid hormone controls thedevelopment of connections between the spinal cord and limbs during Xen-opus laevis metamorphosis. Proc. Natl. Acad. Sci. U. S. A. 101, 165e170.

Marshall, L.N., Vivien, C.J., Girardot, F., Pericard, L., Scerbo, P., Palmier, K.,Demeneix, B.A., Coen, L., 2019. Stage-dependent cardiac regeneration in Xen-opus is regulated by thyroid hormone availability. Proc. Natl. Acad. Sci. U. S. A.116, 3614e3623.

McGivern, J.V., Swaney, D.L., Coon, J.J., Sheets, M.D., 2009. Toward defining thephosphoproteome of Xenopus laevis embryos. Dev. Dynam. 238, 1433e1443.

McKenzie, L.M., Witter, R.Z., Newman, L.S., Adgate, J.L., 2012. Human health riskassessment of air emissions from development of unconventional natural gasresources. Sci. Total Environ. 424, 79e87.

McKeown, C.R., Sharma, P., Sharipov, H.E., Shen, W., Cline, H.T., 2013. Neurogenesisis required for behavioral recovery after injury in the visual system of Xenopuslaevis. J. Comp. Neurol. 521, 2262e2278.

Miller, T.C., Sun, G.H., Hasebe, T., Fu, L.Z., Heimeier, R.A., Das, B., Ishizuya-Oka, A.,Shi, Y.B., 2013. Tissue-specific upregulation of MDS/EVI gene transcripts in theintestine by thyroid hormone during Xenopus Metamorphosis. PLoS ONE 8,e55585.

Moriyama, Y., Ohata, Y., Mori, S., Matsukawa, S., Michiue, T., Asashima, M.,Kuroda, H., 2011. Rapamycin treatment causes developmental delay, pigmen-tation defects, and gastrointestinal malformation on Xenopus embryogenesis.Biochem. Biophys. Res. Commun. 404, 974e978.

Mouchet, F., Teaniniuraitemoana, V., Baudrimont, M., Daffe, G., Gauthier, L.,Gonzalez, P., 2015. Recovery capabilities of Xenopus laevis after exposure tocadmium and zinc. Chemosphere 139, 117e125.

Munoz, R., Edwards-Faret, G., Moreno, M., Zuniga, N., Cline, H., Larrain, J., 2015.Regeneration of Xenopus laevis spinal cord requires Sox 2/3 expressing cells.Dev. Biol. 408, 229e243.

Nations, S., Long, M., Wages, M., Maul, J.D., Theodorakis, C.W., Cobb, G.P., 2015.Subchronic and chronic developmental effects of copper oxide (CuO) nano-particles on Xenopus laevis. Chemosphere 135, 166e174.

Nations, S., Wages, M., Canas, J.E., Maul, J., Theodorakis, C., Cobb, G.P., 2011. Acuteeffects of Fe(2)O(3), TiO(2), ZnO and CuO nanomaterials on Xenopus laevis.Chemosphere 83, 1053e1061.

Nenni, M.J., Fisher, M.E., James-Zorn, C., Pells, T.J., Ponferrada, V., Chu, S.,Fortriede, J.D., Burns, K.A., Wang, Y., Lotay, V.S., Wang, D.Z., Segerdell, E.,

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

Chaturvedi, P., Karimi, K., Vize, P.D., Zorn, A.M., 2019. Xenbase: facilitating theuse of Xenopus to model human disease. Front. Physiol. 10, 154.

Newman Jr., S.M., Dumont, J.N., 1983. Thiosemicarbazide-induced osteolathyrism inmetamorphosing Xenopus laevis. J. Exp. Zool. 225, 411e421.

Ny, A., Autiero, M., Carmeliet, P., 2006. Zebrafish and Xenopus tadpoles: small an-imal models to study angiogenesis and lymphangiogenesis. Exp. Cell Res. 312,684e693.

Oka, T., Tooi, O., Mitsui, N., Miyahara, M., Ohnishi, Y., Takase, M., Kashiwagi, A.,Shinkai, T., Santo, N., Iguchi, T., 2008. Effect of atrazine on metamorphosis andsexual differentiation in Xenopus laevis. Aquat. Toxicol. 87, 215e226.

Paranjpe, S.S., Jacobi, U.G., van Heeringen, S.J., Veenstra, G.J.C., 2013. A genome-widesurvey of maternal and embryonic transcripts during Xenopus tropicalisdevelopment. BMC Genom. 14, 762.

Parker, T.T., Rayburn, J., 2017. A comparison of electronic and traditional cigarettebutt leachate on the development of Xenopus laevis embryos. Toxicol Rep 4,77e82.

Peng, Y., Kwok, K.H., Yang, P.H., Ng, S.S., Liu, J., Wong, O.G., He, M.L., Kung, H.F.,Lin, M.C., 2005. Ascorbic acid inhibits ROS production, NF-kappa B activationand prevents ethanol-induced growth retardation and microencephaly.Neuropharmacology 48, 426e434.

Peng, Y., Yang, P.H., Guo, Y., Ng, S.S., Liu, J., Fung, P.C., Tay, D., Ge, J., He, M.L.,Kung, H.F., Lin, M.C., 2004. Catalase and peroxiredoxin 5 protect Xenopus em-bryos against alcohol-induced ocular anomalies. Invest. Ophthalmol. Vis. Sci.45, 23e29.

Phipps, L.S., Marshall, L., Dorey, K., Amaya, E., 2020. Model Systems for Regenera-tion: Xenopus, vol. 147. Development.

Pinet, K., Deolanka, M., Leung, B., McLaughlin, K.A., 2019. Adaptive correction ofcraniofacial defects in pre-metamorphic Xenopus laevis tadpoles involvesthyroid hormone-independent tissue remodeling. Development 146.

Porter, N.J., Li, W.C., 2018. Muscarinic modulation of the Xenopus laevis tadpolespinal mechanosensory pathway. Brain Res. Bull. 139, 278e284.

Poulsen, R., Luong, X., Hansen, M., Styrishave, B., Hayes, T., 2015. Tebuconazoledisrupts steroidogenesis in Xenopus laevis. Aquat. Toxicol. 168, 28e37.

Pratt, K.G., Aizenman, C.D., 2007. Homeostatic regulation of intrinsic excitability andsynaptic transmission in a developing visual circuit. J. Neurosci. 27, 8268e8277.

Pratt, K.G., Khakhalin, A.S., 2013. Modeling human neurodevelopmental disordersin the Xenopus tadpole: from mechanisms to therapeutic targets. Dis ModelMech 6, 1057e1065.

Preud’homme, V., Milla, S., Gillardin, V., De Pauw, E., Denoel, M., Kestemont, P.,2015. Effects of low dose endosulfan exposure on brain neurotransmitter levelsin the African clawed frog Xenopus laevis. Chemosphere 120, 357e364.

Regnault, C., Usal, M., Veyrenc, S., Couturier, K., Batandier, C., Bulteau, A.L., Lejon, D.,Sapin, A., Combourieu, B., Chetiveaux, M., Le May, C., Lafond, T., Raveton, M.,Reynaud, S., 2018. Unexpected metabolic disorders induced by endocrine dis-ruptors in Xenopus tropicalis provide new lead for understanding amphibiandecline. Proc. Natl. Acad. Sci. U. S. A. 115, E4416eE4425.

Rice, D.C., 2008. Overview of modifiers of methylmercury neurotoxicity: chemicals,nutrients, and the social environment. Neurotoxicology 29, 761e766.

Richards, S.M., Cole, S.E., 2006. A toxicity and hazard assessment of fourteenpharmaceuticals to Xenopus laevis larvae. Ecotoxicology 15, 647e656.

Rimayi, C., Odusanya, D., Weiss, J.M., de Boer, J., Chimuka, L., Mbajiorgu, F., 2018.Effects of environmentally relevant sub-chronic atrazine concentrations onAfrican clawed frog (Xenopus laevis) survival, growth and male gonad devel-opment. Aquat. Toxicol. 199, 1e11.

Romoli, M., Mazzocchetti, P., D’Alonzo, R., Siliquini, S., Rinaldi, V.E., Verrotti, A.,Calabresi, P., Costa, C., 2019. Valproic acid and epilepsy: from molecularmechanisms to clinical evidences. Curr. Neuropharmacol. 17, 926e946.

Roullet, F.I., Lai, J.K., Foster, J.A., 2013. In utero exposure to valproic acid and autism–

a current review of clinical and animal studies. Neurotoxicol. Teratol. 36, 47e56.Sai, L., Li, Y., Zhang, Y., Zhang, J., Qu, B., Guo, Q., Han, M., Jia, Q., Yu, G., Li, K., Bo, C.,

Zhang, Y., Shao, H., Peng, C., 2020. Distinct m(6)A methylome profiles in poly(A)RNA from Xenopus laevis testis and that treated with atrazine. Chemosphere245, 125631.

Sai, L., Qu, B., Zhang, J., Liu, J., Jia, Q., Bo, C., Zhang, Y., Yu, G., Han, R., Peng, C., 2019.Analysis of long non-coding RNA involved in atrazine-induced testiculardegeneration of Xenopus laevis. Environ. Toxicol. 34, 505e512.

Saide, K., Sherwood, V., Wheeler, G.N., 2019. Paracetamol-induced liver injurymodelled in Xenopus laevis embryos. Toxicol. Lett. 302, 83e91.

Saide, K., Wheeler, G.N., 2020. In vivo assessment of drug-induced hepatotoxicityusing Xenopus embryos. Cold Spring Harb. Protoc.

Sakane, Y., Iida, M., Hasebe, T., Fujii, S., Buchholz, D.R., Ishizuya-Oka, A.,Yamamoto, T., Suzuki, K.T., 2018. Functional analysis of thyroid hormone re-ceptor beta in Xenopus tropicalis founders using CRISPR-Cas. Biol Open 7.

Sater, A.K., Moody, S.A., 2017. Using Xenopus to understand human disease anddevelopmental disorders. Genesis 55.

Schlosser, G., Koyano-Nakagawa, N., Kintner, C., 2002. Thyroid hormone promotesneurogenesis in the Xenopus spinal cord. Develop. Dynam. 225, 485e498.

Schmidt, C., Klein, C., Hollmann, M., 2009. Xenopus laevis oocytes endogenouslyexpress all subunits of the ionotropic glutamate receptor family. J. Mol. Biol.390, 182e195.

Schmitt, S.M., Gull, M., Br€andli, A.W., 2014. Engineering Xenopus embryos forphenotypic drug discovery screening. Adv. Drug Deliv. Rev. 69e70, 225e246.

Schriks, M., Zvinavashe, E., Furlow, J.D., Murk, A.J., 2006. Disruption of thyroidhormone-mediated Xenopus laevis tadpole tail tip regression by hex-abromocyclododecane (HBCD) and 2,2’,3,3’,4,4’,5,5’,6-nona brominated

12

diphenyl ether (BDE206). Chemosphere 65, 1904e1908.Sehonova, P., Hodkovicova, N., Urbanova, M., €Orn, S., Blahova, J., Svobodova, Z.,

Faldyna, M., Chloupek, P., Briedikova, K., Carlsson, G., 2019. Effects of antide-pressants with different modes of action on early life stages of fish and am-phibians. Environ. Pollut. 254, 112999.

Shen, W., Liu, H.H., Schiapparelli, L., McClatchy, D., He, H.Y., Yates 3rd, J.R., Cline, H.T.,2014. Acute synthesis of CPEB is required for plasticity of visual avoidancebehavior in Xenopus. Cell Rep. 6, 737e747.

Shen, W., McKeown, C.R., Demas, J.A., Cline, H.T., 2011. Inhibition to excitation ratioregulates visual system responses and behavior in vivo. J. Neurophysiol. 106,2285e2302.

Shi, Y., Li, J., Chen, C., Gong, M., Chen, Y., Liu, Y., Chen, J., Li, T., Song, W., 2014. 5-Mehtyltetrahydrofolate rescues alcohol-induced neural crest cell migrationabnormalities. Mol. Brain 7, 67.

Shi, Z., Xin, H., Tian, D., Lian, J., Wang, J., Liu, G., Ran, R., Shi, S., Zhang, Z., Shi, Y.,Deng, Y., Hou, C., Chen, Y., 2019. Modeling human point mutation diseases inXenopus tropicalis with a modified CRISPR/Cas9 system. FASEB (Fed. Am. Soc.Exp. Biol.) J. 33, 6962e6968.

Showell, C., Conlon, F.L., 2009. Egg collection and in vitro fertilization of the westernclawed frog Xenopus tropicalis. Cold Spring Harb Protoc 2009 9.

Shukrun, N., Shabtai, Y., Pillemer, G., Fainsod, A., 2019. Retinoic acid signalingreduction recapitulates the effects of alcohol on embryo size. Genesis 57,e23284.

Spawn, A., Aizenman, C.D., 2012. Abnormal visual processing and increased seizuresusceptibility result from developmental exposure to the biocide methyl-isothiazolinone. Neuroscience 205, 194e204.

Steffensen, A.B., Refaat, M.M., David, J.P., Mujezinovic, A., Calloe, K., Wojciak, J.,Nussbaum, R.L., Scheinman, M.M., Schmitt, N., 2015. High incidence of func-tional ion-channel abnormalities in a consecutive Long QT cohort with novelmissense genetic variants of unknown significance. Sci. Rep. 5, 10009.

Storustovu, S.I., Ebert, B., 2006. Pharmacological characterization of agonists atdelta-containing GABAA receptors: functional selectivity for extrasynaptic re-ceptors is dependent on the absence of gamma 2. J. Pharmacol. Exp. Therapeut.316, 1351e1359.

Tadjuidje, E., Heasman, J., 2010. Xenopusas an Experimental Organism, Encyclo-pedia of Life Sciences. John Wiley & Sons, Ltd.

Taft, J.D., Colonnetta, M.M., Schafer, R.E., Plick, N., Powell, W.H., 2018. Dioxinexposure alters molecular and morphological responses to thyroid hormone inXenopus laevis cultured cells and prometamorphic tadpoles. Toxicol. Sci. 161,196e206.

Tamschick, S., Rozenblut-Koscisty, B., Ogielska, M., Kekenj, D., Gajewski, F.,Kruger, A., Kloas, W., Stock, M., 2016. The plasticizer bisphenol A affects somaticand sexual development, but differently in pipid, hylid and bufonid anurans.Environ. Pollut. 216, 282e291.

Tandon, P., Conlon, F., Furlow, J.D., Horb, M.E., 2017. Expanding the genetic toolkit inXenopus: approaches and opportunities for human disease modeling. Dev. Biol.426, 325e335.

Tao, H.W., Poo, M.M., 2005. Activity-dependent matching of excitatory and inhibi-tory inputs during refinement of visual receptive fields. Neuron 45, 829e836.

Tao, Y., Ruan, H., Guo, X., Li, L., Shen, W., 2015. HDAC1 regulates the proliferation ofradial glial cells in the developing Xenopus tectum. PloS One 10, e0120118.

Thompson, C.K., Cline, H.T., 2016. Thyroid hormone acts locally to increase neuro-genesis, neuronal differentiation, and dendritic arbor elaboration in the tadpolevisual system. J. Neurosci. 36, 10356e10375.

Thorne, C.A., Lafleur, B., Lewis, M., Hanson, A.J., Jernigan, K.K., Weaver, D.C.,Huppert, K.A., Chen, T.W., Wichaidit, C., Cselenyi, C.S., Tahinci, E., Meyers, K.C.,Waskow, E., Orton, D., Salic, A., Lee, L.A., Robbins, D.J., Huppert, S.S., Lee, E., 2011.A biochemical screen for identification of small-molecule regulators of the Wntpathway using Xenopus egg extracts. J. Biomol. Screen 16, 995e1006.

Titmus, M.J., Tsai, H.J., Lima, R., Udin, S.B., 1999. Effects of choline and other nicotinicagonists on the tectum of juvenile and adult Xenopus frogs: a patch-clampstudy. Neuroscience 91, 753e769.

Tomlinson, M.L., Field, R.A., Wheeler, G.N., 2005. Xenopus as a model organism indevelopmental chemical genetic screens. Mol. Biosyst. 1, 223e228.

Tomlinson, M.L., Guan, P., Morris, R.J., Fidock, M.D., Rejzek, M., Garcia-Morales, C.,Field, R.A., Wheeler, G.N., 2009. A chemical genomic approach identifies matrixmetalloproteinases as playing an essential and specific role in Xenopus mela-nophore migration. Chem. Biol. 16, 93e104.

Truszkowski, T.L., James, E.J., Hasan, M., Wishard, T.J., Liu, Z., Pratt, K.G., Cline, H.T.,Aizenman, C.D., 2016. Fragile X mental retardation protein knockdown in thedeveloping Xenopus tadpole optic tectum results in enhanced feedforwardinhibition and behavioral deficits. Neural Dev. 11, 14.

Tussellino, M., Ronca, R., Formiggini, F., De Marco, N., Fusco, S., Netti, P.A.,Carotenuto, R., 2015. Polystyrene nanoparticles affect Xenopus laevis develop-ment. J. Nanoparticle Res. 17, 70.

Usal, M., Regnault, C., Veyrenc, S., Couturier, K., Batandier, C., Bulteau, A.L., Lejon, D.,Combourieu, B., Lafond, T., Raveton, M., Reynaud, S., 2019. Concomitant expo-sure to benzo[a]pyrene and triclosan at environmentally relevant concentra-tions induces metabolic syndrome with multigenerational consequences inSilurana (Xenopus) tropicalis. Sci. Tot. Environ. 689, 149e159.

van Kleef, R.G., Vijverberg, H.P., Westerink, R.H., 2008. Selective inhibition of humanheteromeric alpha9alpha10 nicotinic acetylcholine receptors at a low agonistconcentration by low concentrations of ototoxic organic solvents. Toxicol. Vitro22, 1568e1572.

Viet, J., Reboutier, D., Hardy, S., Lachke, S.A., Paillard, L., Gautier-Courteille, C., 2020.

J. Gao and W. Shen Environmental Pollution 268 (2021) 115809

Modeling ocular lens disease in Xenopus. Dev. Dynam. 249, 610e621.Vismara, C., Bacchetta, R., Cacciatore, B., Vailati, G., Fascio, U., 2001. Paraquat

embryotoxicity in the Xenopus laevis cleavage phase. Aquat. Toxicol. 55, 85e93.Walentek, P., Quigley, I.K., 2017. What we can learn from a tadpole about cil-

iopathies and airway diseases: using systems biology in Xenopus to study ciliaand mucociliary epithelia. Genesis 55.

Wang, R., Hausknecht, K., Shen, R.Y., Haj-Dahmane, S., 2018. Potentiation of gluta-matergic synaptic transmission onto dorsal raphe serotonergic neurons in thevalproic acid model of autism. Front. Pharmacol. 9, 1185.

Wang, Y., Li, Y., Qin, Z., Wei, W., 2017. Re-evaluation of thyroid hormone signalingantagonism of tetrabromobisphenol A for validating the T3-induced Xenopusmetamorphosis assay. J. Environ. Sci. (China) 52, 325e332.

Watson, F.L., Schmidt, H., Turman, Z.K., Hole, N., Garcia, H., Gregg, J., Tilghman, J.,Fradinger, E.A., 2014. Organophosphate pesticides induce morphological ab-normalities and decrease locomotor activity and heart rate in Danio rerio andXenopus laevis. Environ. Toxicol. Chem. 33, 1337e1345.

Webster, C.A., Di Silvio, D., Devarajan, A., Bigini, P., Micotti, E., Giudice, C.,Salmona, M., Wheeler, G.N., Sherwood, V., Bombelli, F.B., 2016. An earlydevelopmental vertebrate model for nanomaterial safety: bridging cell-basedand mammalian toxicity assessment. Nanomedicine 11, 643e656.

Wheeler, G.N., Brandli, A.W., 2009. Simple vertebrate models for chemical geneticsand drug discovery screens: lessons from zebrafish and Xenopus. Dev. Dynam.238, 1287e1308.

Williams, J.R., Rayburn, J.R., Cline, G.R., Sauterer, R., Friedman, M., 2014. Potentialprotective effect of L-cysteine against the toxicity of acrylamide and furan inexposed Xenopus laevis embryos: an interaction study. J. Agric. Food Chem. 62,7927e7938.

Xu, Y., Gye, M.C., 2018. Developmental toxicity of dibutyl phthalate and citrate esterplasticizers in Xenopus laevis embryos. Chemosphere 204, 523e534.

Yao, X., Chen, X., Zhang, Y., Li, Y., Wang, Y., Zheng, Z., Qin, Z., Zhang, Q., 2017.Optimization of the T3-induced Xenopus metamorphosis assay for detectingthyroid hormone signaling disruption of chemicals. J. Environ. Sci. (China) 52,314e324.

Yelin, R., Kot, H., Yelin, D., Fainsod, A., 2007. Early molecular effects of ethanolduring vertebrate embryogenesis. Differentiation 75, 393e403.

Yelin, R., Schyr, R.B., Kot, H., Zins, S., Frumkin, A., Pillemer, G., Fainsod, A., 2005.Ethanol exposure affects gene expression in the embryonic organizer and

13

reduces retinoic acid levels. Dev. Biol. 279, 193e204.Yokoyama, H., Maruoka, T., Ochi, H., Aruga, A., Ohgo, S., Ogino, H., Tamura, K., 2011.

Different requirement for Wnt/beta-catenin signaling in limb regeneration oflarval and adult Xenopus. PloS One 6, e21721.

Yoon, C.S., Jin, J.H., Park, J.H., Youn, H.J., Cheong, S.W., 2003. The fungicide benomylinhibits differentiation of neural tissue in the Xenopus embryo and animal capexplants. Environ. Toxicol. 18, 327e337.

Yoon, Y.H., Kim, J.Y., Bae, Y.C., Nam, S.W., Cho, H.J., Lee, S., Chung, H.Y., Lee, H.S.,Park, M.J., 2018. Evaluation of the toxic effects of celecoxib on Xenopus embryodevelopment. Biochem. Biophys. Res. Commun. 501, 329e335.

Yu, Y., Yu, Z., Chen, H., Han, Y., Xiang, M., Chen, X., Ma, R., Wang, Z., 2019. Tetra-bromobisphenol A: disposition, kinetics and toxicity in animals and humans.Environ. Pollut. 253, 909e917.

Yuan, J., Zhang, X., Yu, L., Sun, Z., Zhu, P., Wang, X., Shi, H., 2011. Stage-specificmalformations and phenotypic changes induced in embryos of amphibian(Xenopus tropicalis) by triphenyltin. Ecotoxicol. Environ. Saf. 74, 1960e1966.

Zeng, S.L., Sudlow, L.C., Berezin, M.Y., 2020. Using Xenopus oocytes in neurologicaldisease drug discovery. Expet Opin. Drug Discov. 15, 39e52.

Zhang, C., Liu, X., Wu, D., Liu, G., Tao, L., Fu, W., Hou, J., 2014. Teratogenic effects oforganic extracts from the Pearl River sediments on Xenopus laevis embryos.Environ. Toxicol. Pharmacol. 37, 202e209.

Zhang, W., Deng, Y., Chen, L., Zhang, L., Wang, Z., Liu, R., Diao, J., Zhou, Z., 2020. Effectof triadimefon and its metabolite on adult amphibians Xenopus laevis. Che-mosphere 243, 125288.

Zhao, J., Xie, G., Xu, Y., Zheng, L., Ling, J., 2020. Accumulation and toxicity of multi-walled carbon nanotubes in Xenopus tropicalis tadpoles. Chemosphere 257,127205.

Zhu, J., Hu, L., Li, L., Huang, X., Shi, H., 2017. Comparison of phenotypic and globalgene expression changes in Xenopus tropicalis embryos induced by agonists ofRAR and RXR. Toxicol. Appl. Pharmacol. 330, 40e47.

Zhu, J., Huang, X., Jiang, H., Hu, L., Michal, J.J., Jiang, Z., Shi, H., 2018. The role ofppargamma in embryonic development of Xenopus tropicalis undertriphenyltin-induced teratogenicity. Sci. Total Environ. 633, 1245e1252.

Zhu, M., Niu, Y., Li, Y., Dong, M., Li, J., Zeng, R., Qin, Z., 2020. Low concentrations oftetrabromobisphenol A disrupt Notch signaling and intestinal development inin vitro and in vivo models. Chem. Res. Toxicol. 33, 1418e1427.