• Keine Ergebnisse gefunden

This is the accepted manuscript of the contribution published as:

N/A
N/A
Protected

Academic year: 2022

Aktie "This is the accepted manuscript of the contribution published as:"

Copied!
28
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

This is the accepted manuscript of the contribution published as:

Bertotto, L.B., Catron, T.R., Tal, T. (2020):

Exploring interactions between xenobiotics, microbiota, and neurotoxicity in zebrafish NeuroToxicology 76 , 235 – 244

The publisher's version is available at:

http://dx.doi.org/10.1016/j.neuro.2019.11.008

(2)

1

Exploring interactions between xenobiotics, microbiota, and neurotoxicity in zebrafish 1

2

Luísa B. Bertottoa, Tara R. Catrona and Tamara Talb*. 3

4

aOak Ridge Institute for Science and Education, US EPA ORD, NHEERL, ISTD and bUS EPA 5

ORD, NHEERL, ISTD.

6 7

*Current Address; Address Correspondence: Tamara Tal, Bioanalytical Ecotoxicology 8

Department, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, 04318 9

Leipzig, Germany, tamara.tal@ufz.de, +49 341 235 1524.

10 11

ORCID 12

Luísa Bertotto (0000-0002-5457-4208), Tara Catron (0000-0003-1193-9366), Tamara Tal (0000- 13

0001-8365-9385) 14

15

Keywords: microbiome, zebrafish, developmental neurotoxicity 16

17 18

*Manuscript

Click here to view linked References

(3)

2 Abstract

19 20

Susceptibility to xenobiotic exposures is variable. One factor that might account for this is the 21

microbiome, which encompasses all microorganisms, their encoded genes, and associated 22

functions that colonize a host organism. Microbiota harbor the capacity to affect the toxicokinetics 23

and toxicodynamics of xenobiotic exposures. The neurotoxicological effects of environmental 24

chemicals may be modified by intestinal microbes via the microbiota-gut-brain axis. This is a 25

complex, bi-directional signaling pathway between intestinal microbes and the host nervous 26

system. As a model organism, zebrafish are extremely well-placed to illuminate mechanisms by 27

which microbiota modify the developmental neurotoxicity of environmental chemicals. The goal 28

of this review article is to examine the microbiota-gut-brain axis in a toxicological context, 29

specifically focusing on the strengths and weaknesses of the zebrafish model for the investigation 30

of interactions between xenobiotic agents and host-associated microbes. Previous studies 31

describing the relationship between intestinal microbes and host neurodevelopment will be 32

discussed. From a neurotoxicological perspective, studies utilizing zebrafish to assess links 33

between neurotoxicological outcomes and the microbiome are emphasized. Overall, there are 34

major gaps in our understanding the mechanisms by which microbiota interact with xenobiotics to 35

cause or modify host neurotoxicity. In this review, we demonstrate that zebrafish are an ideal 36

model system for studying the complex relationship between chemical exposures, 37

microorganisms, and host neurotoxicological outcomes.

38 39 40

(4)

3

Zebrafish as an established model for neurotoxicology studies 41

42

Zebrafish is a non-mammalian vertebrate model that is well established as an alternative model for 43

neurotoxicological studies (Peterson et al., 2008; Horzmann & Freeman, 2016). Zebrafish contain 44

a fully sequenced genome, and approximately 70-80% of their genes are homologous with human 45

counterparts (Howe et al., 2013). As opposed to commonly used animal models like mice and rats, 46

zebrafish embryos develop external to the mother, such that the developing embryo can be directly 47

exposed to xenobiotic agents (Peterson et al., 2008; Horzmann & Freeman, 2016). Zebrafish 48

organogenesis is complete by 72 hours post fertilization (hpf) (Peterson et al., 2008; Horzmann &

49

Freeman, 2016) and both embryos and larvae can be toxicologically assessed in 96-well plates or, 50

for early developmental analyses, 384-well plates, where chemical exposures can be easily 51

performed (Peterson et al., 2008; Horzmann & Freeman, 2016).

52 53

Zebrafish neurodevelopment is generally conserved compared to humans (Lee & Freeman, 2014;

54

Nishimura et al., 2015, Schmidt et al., 2012). At 6 hpf, during gastrulation, ectoderm 55

differentiation begins (Lee & Freeman, 2014; Nishimura et al., 2015). Analogous to other 56

mammalian species, zebrafish neuroectoderm develops into a neural plate and undergoes 57

neurulation (i.e. the process of folding the neural plate into the neural tube) (Nishimura et al., 2015;

58

Schmidt et al., 2012). However, neurulation occurs via a different process in zebrafish relative to 59

most vertebrates (Papan & Campos-Ortega, 1994; Buckley et al., 2013). In zebrafish, at roughly 60

12 hpf, the neural plate forms a neural keel, leading to the formation of the neural rod followed by 61

the neural tube (Papan & Campos-Ortega, 1994; Buckley et al., 2013). The forebrain 62

(diencephalon), midbrain (telencephalon), hindbrain (cerebellum), and spinal cord are apparent at 63

16 hpf (Kozol et al., 2016). By 2-3 days post fertilization (dpf), neuronal subtypes, including 64

GABAergic, catecholaminergic, serotonergic, and noradrenergic neurons, start to differentiate 65

(Lee & Freeman, 2014; Nishimura et al., 2015). Similar to mammas, zebrafish also contain 66

astrocytes, microglia, oligodendrocytes, cerebellar Purkinje cells, myelin, and motor neurons and 67

zebrafish develop a functional blood brain barrier (BBB) by 3 dpf (Fleming et al., 2013).

68

Neurotoxicological phenotypes include loss or expansion of brain ventricles, truncation of the 69

telencephalon, and neuronal necrosis (Peterson et al., 2000).

70 71

Automated behavioral tests in embryonic and larval zebrafish are widely used as a functional 72

readout of neurodevelopment in animals exposed to environmental chemicals (Figure 1) (Fraser et 73

al., 2017; Glazer et al., 2017; Dishaw et al., 2014; Chen et al., 2012; Bailey et al., 2016; Massarsky 74

et al., 2018; Cassar et al., 2018; Wolman et al., 2015). Neurotoxicological effects of xenobiotics 75

are also commonly assessed in older life stage zebrafish (Lutte et al., 2018; Anichtchik et al., 2004;

76

Pereira et al., 2012; Bencan & Levin, 2008; Massarsky et al., 2018; Pereira et al., 2012; Xu et al., 77

2016). Embryos can be assessed for the effect of xenobiotic exposures on spontaneous movement 78

(i.e. head and tail coilings), which begin as early as 17 hpf, depending on rearing temperature 79

(Kokel al., 2010). Larval zebrafish behavior can be analyzed in relation to distance travelled, time 80

spent active, or pattern of behavioral responses to stimuli like light changes (Fraser et al., 2017;

81

Glazer et al., 2017; Dishaw et al., 2014; Chen et al., 2012; Bailey et al., 2016; Massarsky et al., 82

2018), or acoustic startle (Cassar et al., 2018; Wolman et al., 2015). Furthermore, larval behavior 83

such as threat avoidance (Richendrfer & Creton, 2015; Gonzalez et al., 2016), anxiety-like 84

behavior measured through thigmotaxis (i.e. place of preference in the well) (Gonzalez et al., 85

2016), and optomotor response (Cassar et al., 2018) can also be assessed. Habituation, the most 86

(5)

4

primitive form of learning, can also be tested in larval zebrafish using an automated tracking 87

system and repeated acoustic startles (Roberts et al., 2011; Wolman et al., 2015). Adult 88

neurobehavior is also commonly assessed to investigate baseline locomotor activity (Lutte et al., 89

2018; Anichtchik et al., 2004; Pereira et al., 2012), anxiety-like behavior (Bencan & Levin, 2008;

90

Anichtchik et al., 2004; Massarsky et al., 2018), habituation and memory (Lutte et al., 2018;

91

Massarsky et al., 2018; Pereira et al., 2012), peer recognition (Massarsky et al., 2018; Fernandes 92

et al., 2015), and aversive stimulus recognition (Xu et al., 2016; Massarsky et al., 2018). For all 93

embryonic, larval, and adult behavioral tests, there is a lack of standardized testing procedures.

94 95

In addition to morphological and behavioral endpoints, molecular biology approaches are also 96

widely deployed to examine neurotoxicological outcomes in zebrafish. Targeted gene expression 97

(Massarsky et al., 2018; Pereira et al., 2012) or microarrays (Liu et al., 2015) have been usurped 98

by unbiased RNA sequencing (Xu et al., 2015; Chen et al., 2016; Zhang et al., 2017). Unless 99

sequencing approaches are applied in specific cell types isolated from transgenic lines (Hernández 100

et al., 2018, Cao et al., 2016), these approaches generally lack spatial information. In situ 101

hybridization has long been used in zebrafish to illuminate spatiotemporal gene expression in the 102

developing nervous system (Stehr et al., 2006; Wen et al., 2008; Hill et al., 2003; Kanungo et al., 103

2013). From a neurotoxicological perspective, xenobiotic-induced changes in gene expression are 104

best used for hypothesis generation that can be empirically tested by gain- or loss-of-function 105

experimentation. Historically, antisense oligonucleotide morpholinos were nearly universally 106

utilized to study neurotoxicological mechanisms of action (Bertotto et al., 2019; Chlebowski et 107

al., 2017; Tal et al., 2012). However, because of concerns about off-target effects (Eisen & Smith, 108

2008), the emerging gold standard for mechanism delineation in a toxicological context is gene 109

editing, often via clustered regularly interspaced short palindromic repeats (CRISPR) technique 110

(Zabinyakov et al., 2017; Farrar et al., 2018), although this method also introduces off-target 111

mutations (Tsai & Joung, 2016). Lastly, transgenic lines that allow for real-time visualization and 112

quantitation of electrical activity in live zebrafish have been developed and used to evaluate 113

neurotoxicity of xenobiotics (Hayashi et al., 2015; Wen et al., 2008; Hill et al., 2003; Kanungo et 114

al., 2013). Overall, there are a wealth of tools and approaches that allow for both hazard 115

identification and mechanistic research to examine the neurotoxicological effects of xenobiotic 116

exposures.

117 118

Zebrafish as an emerging model for microbiota-gut-brain-axis studies 119

120

The microbiota-gut-brain axis describes the complex bidirectional communication between 121

microorganisms that reside in the gastrointestinal (GI) tract and the host central nervous system 122

(CNS) (Figure 2). This axis monitors and integrates intestinal functions that link emotional and 123

cognitive centers in the brain to intestinal permeability, enteric reflex, entero-endocrine signaling, 124

and immune activation (Carabotti et al, 2015). Bidirectional communication occurs via vagus 125

nerve neurons that form synapses with intestinal epithelial cells and through the generation of 126

microbial products or microbial stimulation of host cytokines and chemokines that penetrate the 127

BBB and exert direct effects on the host nervous system (Figure 2) (Bravo et al, 2011; Tsavkelova 128

et al., 2000; Stephenson et al., 1947; De Vadder et al., 2014; Wikoff et al., 2009). The host 129

hypothalamus-pituitary-adrenal axis (HPA), which coordinates adaptive responses to stressors like 130

environmental chemicals and elevates systemic proinflammatory cytokines (Breit et al., 2018), 131

also plays an important role in the microbiota-gut-brain axis (Figure 2). Communication from both 132

(6)

5

vagal and HPA pathways can regulate the activity of intestinal effector cells, such as epithelial, 133

smooth muscle, interstitial, and enterochromaffin cells, as well as enteric neurons, which can also 134

be influenced by the gut microbiome. These cells are responsive to an array of molecules 135

synthesized and/or metabolized by intestinal microbiota including catecholamines, GABA, bile 136

acids, and short chain fatty acids (SCFA) (Tsavkelova et al., 2000; Stephenson et al., 1947; Bravo 137

et al., 2011; De Vadder et al., 2014). One well known host-microbiome interaction relates to the 138

neurotransmitter serotonin, of which more than 90% is produced in the gut via microbial- 139

dependent synthesis in enterochromaffin cells (Yano et al., 2015).

140 141

Animal models such as zebrafish (Rawls et al., 2004), mice (Neufeld et al., 2011), rats (Mao et 142

al., 2019), flies (Broderick et al., 2014), and nematodes (Nguyen et al., 2012) are commonly used 143

to study the microbiota-gut-brain axis (Figure 3), often by comparing hosts that lack microbes (i.e.

144

axenic) to colonized or conventionalized (i.e. axenic hosts subsequently colonized with microbes) 145

animals. In this review, the term “axenic” was selected to accurately and specifically describe 146

animals devoid of microbes rather than the more commonly used terms “germ-free” or 147

“gnotobiotic,” as not all microbes are pathogenic and animals conventionalized with known strains 148

of microbes are also considered to be gnotobiotic.

149 150

Relative to mouse and rat models that prohibitively require the maintenance of axenic breeding 151

colonies or lower order animal models such as flies and nematodes that are colonized by less 152

complex microbiota comprised of 1-30 microorganisms, zebrafish represents a powerful 153

intermediate model system where axenic offspring can be easily derived and compared to 154

colonized animals with moderate microbiota complexity (~100-200 species) (Figure 3). In 155

addition, the zebrafish GI tract is homologous with higher order vertebrates, containing a liver, 156

pancreas, gall bladder, absorptive enterocytes, goblet and enteroendocrine cells, a linearly 157

segmented intestinal tract with absorptive and secretory functions, and tight junctions and 158

microvilli in the intestinal epithelium (Goldsmith & Jobin, 2012). However, some important 159

differences exist (Figure 3). For example, zebrafish lack a stomach, lymph nodes, Peyer’s patches, 160

and splenic germinal centers (Danilova & Steiner, 2002). Diet and environment are also quite 161

different between zebrafish and humans, where intestinal microbes associated with laboratory 162

zebrafish exist at ~26-28º C as compared to human-relevant microorganisms that thrive at ~37º C 163

(Meeker & Trede, 2008). Differences in temperature, diet, and salinity all influence the complex 164

community structure of host-associated microbes that colonize zebrafish, which includes gram- 165

positive and gram-negative bacteria, protozoa, fungi, and viruses (Goldsmith & Jobin, 2012).

166 167

In both zebrafish and mammals, microbial communities become more established and diverse as 168

the organisms mature. At the phylum level, zebrafish microbiota are variable based on laboratory 169

environment and in general are dominated by Proteobacteria, Firmicutes, and Fusobacteria (Rawls 170

et al., 2006; Stephens et al., 2016). In contrast, mice and human microbiota are dominated by 171

Firmicutes and Bacteroidetes. Humans also contain a proportionally similar number of bacteria 172

that belong to the phylum Actinobacteria (Kostic et al., 2013). While some differences in 173

microbiota composition between species can be attributed to environmental factors such as 174

temperature, diet, or salinity, host factors also strongly impact the development and maturation of 175

microbial communities within each species. For example, some studies in zebrafish have shown 176

that microbial composition is influenced by host-specific selective pressures in the gut and are 177

altered even when extrinsic factors such as diet and environment remain constant (Bevins et al., 178

(7)

6

2011; Wong et al., 2012; Stephens et al., 2016). Additionally, axenic adult zebrafish transplanted 179

with mouse intestinal microbiota (primarily Firmicutes) develop microbiota that resemble 180

conventional zebrafish guts (primarily Proteobacteria) rather than conventional mouse guts (Rawls 181

et al., 2006). A similar phenomenon occurs when zebrafish microbiota are transplanted into axenic 182

mice (Rawls et al., 2006), suggesting that comparable host selection factors and signaling 183

mechanisms are present in both zebrafish and mammalian models. Importantly, the moderately 184

diverse microbiota (~100-200 species, Figure 3) within zebrafish contain functionally similar 185

enzymes and biochemical pathways compared to mammals (e.g. rodents) (Milligan-Myhre et al.

186

2011).

187 188

One major advantage of the zebrafish system relative to higher order vertebrates is that methods 189

to derive axenic embryonic zebrafish are well established (Rawls et al., 2004; Rawls et al., 2006;

190

Phelps et al., 2017; Melancon et al., 2017). Axenic status can be easily assessed by culturing media 191

samples, qRT-PCR, or metagenomic sequencing. Once axenic zebrafish are generated, they can 192

be conventionalized via simple immersion using fish facility water or specific microbial cultures 193

(Davis et al., 2016a; Davis et al., 2016b; Phelps et al., 2017), or by injection (Herbomel et al., 194

1999; Vergunst et al., 2010). It is important to note that colonization with fish facility water is 195

variable over time (Catron et al., 2019a), which can result in significant differences in community 196

structure between conventionally colonized and conventionalized control animals (Catron et al., 197

2019b; Weitekamp et al., 2019). Depending on rearing temperature and institutional animal use 198

rules, methods for rearing up zebrafish are possible without the introduction of sterile food for up 199

to ~6 dpf, as larvae rely on the yolk sac for nutrition (Dabrowski & Miller, 2018). Past ~6 dpf, 200

axenic zebrafish have been successfully reared until 10 dpf using gamma irradiated powder diet 201

(Phelps et al., 2017) and 30 dpf with the addition of microbe-free live food cultures, a labor- 202

intensive method (Melancon et al., 2017).

203 204

Regarding study design, one of the strengths of alternative models for microbiota-oriented 205

research, such as the fruit fly, the nematode worm, and the zebrafish, is the ability to easily generate 206

and maintain multiple colonization statuses of these organisms including conventionally 207

colonized, axenic, and conventionalized (Figure 3). Relative to less complex systems like 208

Drosophila and C. elegans, zebrafish is particularly advantageous because it has greater genetic 209

similarity to human/mammalian genomes, increased microbial diversity (~100-200 species) 210

(Figure 3), and more complex assays exist to investigate microbiota-gut-brain interactions.

211

Zebrafish therefore represents an ideal intermediate model system with sufficient microbiome 212

complexity, yet also allows for relatively simple modification of microbial colonization status.

213

However, the above-mentioned important disadvantages of the zebrafish model relative to mice 214

and humans should not be overlooked (e.g. GI tract structure and microbiota composition), as key 215

differences between species can likely influence toxicokinetic and toxicodynamic interactions with 216

xenobiotic agents. Overall, the ability to easily manipulate larval colonization status is a key 217

technical advantage that can be used to determine whether microbial colonization status influences 218

the developmental neurotoxicity of exposure to environmental chemicals (Figure 1). More work 219

is needed to understand how structural and potential functional differences in host-associated 220

microbes impacts microbiota interactions with the brain, particularly in the context of xenobiotics 221

exposures.

222 223

Key zebrafish microbiota-gut-brain-axis behavior studies 224

(8)

7 225

As mentioned previously, zebrafish behavior is commonly used as a functional readout of 226

neurodevelopment. The same logic can be applied to evaluate the microbiota-gut-brain axis in 227

zebrafish. In zebrafish larvae, microbiota can modulate locomotion and anxiety-related behaviors 228

(Davis et al., 2016a; Davis et al., 2016b; Phelps et al., 2017; Catron et al., 2019b). Davis et al.

229

(2016a) first reported that axenic zebrafish are hyperactive relative to colonized controls, a 230

phenotype that our laboratory has replicated (Phelps et al., 2017) and has also been observed in 231

both mammalian studies (Diaz Heijtz et al., 2011; Neufeld et al., 2011) and, more recently, in 232

Drosophila (Schretter et al., 2018). Thigmotaxis, the demonstrated preference for the edge of a 233

multiwall plate, is used as a functional measure of anxiety-like behavior in zebrafish (Kalueff &

234

Stewart, 2012). Axenic zebrafish exhibit reduced anxiety-like behavior in the thigmotaxis assay 235

(Davis et al., 2016a), although this finding was not replicated in a later study (Phelps et al., 2017).

236

The lack of a standardized method for assessing thigmotaxis in zebrafish likely explains the 237

discordant data. From a developmental perspective, Phelps et al. (2017) reported that 238

conventionalization of axenic zebrafish by 6 dpf was sufficient to block hyperactivity at 10 dpf.

239

This supports the concept that there are critical windows of nervous system development that 240

require microbial colonization to enable control-like development in zebrafish and that 241

conventionalization after these temporally distinct windows close is likely insufficient to recover 242

control-like behavior (Phelps et al., 2017). This is supported by mammalian data showing that 243

colonization of axenic mice post-weaning failed to replenish reduced serotonin levels in the CNS 244

(Clarke et al., 2013). In other words, in mice, there is a strict developmental window that requires 245

microbial colonization for control-like establishment of serotonergic signaling (Clarke et al., 246

2013). Exciting recent work has identified the bacterial enzyme xylose isomerase as critical 247

modulator of sugar metabolism in flies and that subsequent activation of host octopaminergic 248

neurons was sufficient to block axenic hyperactivity (Schretter et al., 2018). Despite these 249

advancements, the mechanism(s) by which microbial colonization influences the development and 250

function of circuits that control stereotypic behaviors in zebrafish (i.e. larval swimming responses 251

and thigmotaxis), and how these microbiome-host interactions are affected by xenobiotic 252

exposure, are unknown.

253 254

In adult zebrafish, two separate studies have similarly used behavior to observe how probiotic 255

microbes influence the host nervous system. In Davis et al. (2016b), one-month long 256

supplementation of adult zebrafish with L. plantarum (strain not specified), was shown to subtly 257

alter intestinal microbiota and cause a small but significant reduction in anxiety-like behavior in 258

the novel tank test. The same study also applied a five-day chronic unpredictable stress protocol 259

that massively restructured microbial community structure (Davis et al., 2016b). Restructuring 260

was significantly ablated via supplementation with L. plantarum, although supplementation had 261

no effect on serum cortisol levels (Davis et al., 2016b). In a similar study, Borrelli et al. (2016) 262

administered the probiotic L. rhamnosus IMC 501 for 28 days to adult zebrafish, resulting in small 263

effects on shoaling behavior and altered expression of brain derived neurotrophic factor (bdnf) and 264

genes related to serotonin metabolism and signaling such as paralogs of the tryptophan 265

hydroxylase gene (tph1a, tph1b and tph2). While tantalizing, these adult studies raise two 266

important questions. First, what are the mechanisms by which probiotic administration modifies 267

behavior? Second, do relatively small changes in behavior functionally compromise the organism?

268

Overall, although zebrafish has proven to be an essential animal model for studying host-microbe 269

(9)

8

interactions, more work is needed to establish causal links between intestinal microbes and host 270

swimming behaviors.

271 272

A framework for microbiota-xenobiotic interactions 273

274

There is growing interest in understanding the mechanisms by which microbiota interact with 275

xenobiotic agents to influence host toxicity (e.g. neurodevelopmental toxicity). This can 276

theoretically occur via toxicodynamic interactions where chemical exposures cause dysbiosis, 277

characterized by alterations in the community structure of host-associated microbiota that 278

subsequently affects the host (Figure 4). Chemical-selected microbiota can also perform chemical 279

activation or detoxification reactions (i.e. Toxicokinetic interactions) (Figure 4). Many studies in 280

larval or adult zebrafish demonstrate the utility of the model system to describe dysbiosis following 281

exposure to drugs or environmental chemicals. Recent evidence obtained in the zebrafish model 282

also demonstrates toxicokinetic interactions between chemicals and microbiota in which intestinal 283

microorganisms bioactivate or detoxify xenobiotics.

284 285

Xenobiotic exposure alters community structure of host-associated microbes (i.e. Dysbiosis) 286

287

The identification of chemical-induced dysbiosis in zebrafish is the subject of a recent review 288

(Catron et al. In Press) and will not be discussed in detail here. Briefly, in zebrafish, exposure to 289

a wide array of xenobiotic agents has been shown to disrupt the community structure of host- 290

associated microbes including pesticides (Wang et al., 2019; Zhang et al., 2018; Jin et al., 2017;

291

Oliveira et al., 2017), metals (Dahan et al., 2018, Xia et al., 2018), microplastics (Qiao at el., 2019;

292

Wan et al., 2019), and antibiotics (Nadal et al., 2018; Pindling et al., 2018). Rather than list 293

qualitative changes in specific taxa following various xenobiotic exposures, a key theory and 294

examples of concordant and discordant structural data will be discussed. First, a recent study 295

comparing the effect of six concentrations of the plasticizer Bisphenol A (BPA) or four 296

replacement chemicals (BPAF, BPB, BPF, or BPS) on community structure and developmental 297

toxicity in zebrafish was recently reported (Catron et al., 2019a). The highest concentration 298

evaluated using metagenomic sequencing was the No Observed Effect Concentration (NOEC) for 299

developmental toxicity. Not all compounds tested affected microbial structure (e.g. BPAF or BPB) 300

(Catron et al., 2019a). Interestingly, the ability of xenobiotics to restructure microbiota was 301

inversely related to their potency for developmental toxicity (Catron et al., 2019a). This illustrates 302

the principal that compounds may fail to cause dysbiosis simply because the concentrations 303

necessary to perturb community structure cannot be tolerated by the host (Catron et al., 2019a).

304

Conversely, compounds that are well tolerated by the host may be more likely to cause structural 305

dysbiosis. Because the zebrafish developmental toxicity assay is widely used for hazard 306

identification and chemical prioritization, these data suggest that chemicals with lower host 307

toxicity profiles might be more likely to simultaneously cause dysbiosis of host-associated 308

microbes.

309 310

Discordant structural data following exposure to the same chemical agent are likely because 311

microbiota are significantly affected by extrinsic factors including diet, genetics, age, or water 312

quality parameters that often vary substantially across laboratories. For example, a recent 313

publication showed that exogenous 17β-estradiol exposure (0.34–3.5 µM) failed to alter microbial 314

community structure in larval zebrafish (Catron et al., 2019b), which is in line with the theory that 315

(10)

9

the holobiont system evolved to tolerate fluctuations in endogenous hormones. However, a 316

separate study in adult zebrafish exposed to approximately 0.07 µM 17β-estradiol reported a 317

qualitative perturbation of global microbiota (Chen et al., 2018b). Perhaps more attention should 318

be given to concordant data across laboratories using the same chemical to perturb microbiota. For 319

example, exposure to the antimicrobial agent triclosan was shown to select for the gram-negative 320

Pseudomonas in zebrafish larvae (Weitekamp et al., 2019) and adults (Gaulke et al., 2016) 321

indicating that certain xenobiotic-dependent alterations in community structure are conserved at 322

multiple life stages. In addition, concordant changes in taxa following exposure to different 323

chemical stressors might also be particularly relevant. For example, >70% sequencing reads in 324

zebrafish larvae developmentally exposed to triclosan were associated with a single gram-negative 325

bacteria, Rheinheimera (Weitekamp et al., 2019). Interestingly, BPA exposure also selected for 326

both Rheinheimera and Pseudomonas in larval zebrafish (Catron et al., 2019a). This suggests that 327

certain taxa (e.g. Rheinheimera and Pseudomonas) that are either broadly resistant to xenobiotic 328

agents or are sensitive to chemical exposures yet exhibit swifter repopulation kinetics may serve 329

general markers of dysbiosis in zebrafish. Overall, while these studies report chemical-dependent 330

microbiota dysbiosis, most fail to connect changes in community structure to adverse physiological 331

outcomes in the host (i.e. Toxicodynamic interactions) and this uncertainty is a key limitation of 332

reported xenobiotic-induced structural dysbiosis in zebrafish and other model systems.

333 334

Chemical biotransformation of xenobiotics by intestinal microbiota in zebrafish (i.e.

335

Toxicokinetic interactions) 336

337

Gut microbiota carry out a multiplicity of reactions to efficiently harvest nutrients from their 338

environment. Microbial enzymes can also biotransform xenobiotics. While there are increasingly 339

more studies showing toxicodynamic effects of chemicals on the zebrafish microbiome, 340

toxicokinetic data in this model are rather limited. This topic was recently reviewed in the zebrafish 341

literature (Catron et al., In Press) and will therefore not be extensively explored in this review.

342

Briefly, intestinal microbes can perform xenobiotic reductions, hydrolysis, glucuronidation, lyase 343

reactions and nucleophilic substitutions (Rafii et al., 1997; Lee and Renwick, 1995; Laue et al., 344

2001; Peppercorn and Goldman, 1972; Takeno and Sakai, 1991; Wallace et al., 2015; Cantarel et 345

al., 2012; Sutherland, 1995; Kumano et al., 2016; Catron et al., 2019b; Weitekamp et al., 2019).

346

Catron et al. demonstrated levels of estradiol and some direct estradiol metabolites were ~3x 347

higher in axenic relative to colonized zebrafish at 10 dpf, suggesting that the microbes in this 348

system influence estradiol metabolism and exhibit toxicokinetic interactions with the host (Catron 349

et al. 2019b). Another recent paper demonstrated that colonized zebrafish contained 2.5-3x higher 350

concentrations of parent triclosan compared to axenic zebrafish (Weitekamp et al., 2019).

351

Elevations in triclosan sulfate were also generated by triclosan-selected microbiota and triclosan- 352

selected microorganisms were enriched for the ability to perform sulfonation reactions, which is 353

the first step in the biochemical pathway necessary to produce triclosan sulfate (Weitekamp et al., 354

2019). Together, these studies demonstrate that colonization status influences chemical 355

metabolism in zebrafish and that xenobiotic biotransformation profiles are likely to be chemical- 356

specific, given that consistent changes in parent concentrations were not observed in either axenic 357

or colonized zebrafish across studies (Catron et al. 2019b; Weitekamp et al., 2019). A key 358

consideration when interpreting toxicokinetic data derived from axenic, conventionally colonized, 359

and conventionalized zebrafish is that axenic animals may be compromised in their ability to 360

detoxify or metabolize chemicals. This uncertainty makes it difficult to clearly delineate host vs.

361

(11)

10

microbial effects on chemical biotransformation. Nevertheless, the comparison of xenobiotic 362

biotranformation events across zebrafish with varying colonization statuses represent a powerful 363

strategy to uncover mechanisms by which host-associated microbiota influence the kinetics of 364

xenobiotic exposures.

365 366

Using zebrafish to investigate whether microbiota modify the developmental neurotoxicity of 367

environmental chemicals 368

369

Zebrafish are an excellent model for assessing microbiota as a modifying factor for the 370

developmental neurotoxicity of environmental chemicals (Figure 3). This is in large part due to 371

three key factors. One, because zebrafish develop external to the mother, researchers can directly 372

expose the developing embryos to xenobiotic agents. Two, because the organism initially develops 373

within an acellular chorion, it is relatively simple to generate axenic zebrafish. Three, axenic and 374

colonized zebrafish exposed to chemicals can be assessed for developmental neurotoxicity using 375

a wide array of automated behavioral assays (Figure 1).

376 377

Antibiotic treatment is commonly used to perturb microbiota. In support of a microbial link to 378

neurobehavioral development in zebrafish, conventionally reared zebrafish treated with broad 379

spectrum antibiotics exhibit hyperactivity (Phelps et al., 2017). This result is in line with 380

mammalian data reporting hyperactivity (Desbonnet et al., 2010) and increased anxiety-like 381

behavior (Bercik et al., 2011) in mice exposed to antibiotics. Interestingly, antibiotic-induced 382

hyperactivity in zebrafish phenocopies the degree of locomotor activity observed in axenic 383

zebrafish (Phelps et al., 2017). From a neurotoxicology perspective, there is a single published 384

paper in zebrafish that shows a microbial-dependent behavioral effect elicited by a xenobiotic 385

exposure (Catron et al. 2019b). Light-phase dependent hypoactivity was reported in 386

conventionally colonized and conventionalized zebrafish, but not axenic animals, developmentally 387

exposed multiple concentrations of exogenous 17β-estradiol (Catron et al. 2019b). This suggests 388

that chemical-dependent hypoactivity relied on the presence of microorganisms. To our 389

knowledge, this is the first reported example showing that host-associated microbes are required 390

for the developmental neurotoxicity of a xenobiotic agent. However, despite examining microbial 391

community structure and chemical biotransformation profiles, the mechanism underlying this 392

interaction is unknown (Catron et al. 2019b). In contrast to 17β-estradiol, colonization status failed 393

to modify the effect of triclosan exposure on locomotor activity in zebrafish (Weitekamp et al., 394

2019). Collectively, the use of the zebrafish three colonization status system coupled with targeted 395

and non-targeted analytical chemistry represents a powerful approach that can reveal toxicokinetic 396

contributions of host-associated microbes to chemical toxicity.

397 398

In addition to the three colonization cohort system, two different studies investigated the ability of 399

microorganisms to reinstate control-like behavior in xenobiotic-exposed zebrafish. In a recent 400

study, adult zebrafish were exposed to triclosan and/or the probiotic L. plantarum ST-III for 90 401

days (Zang et al., 2019). Triclosan exposed fish exhibited increased speed and distance travelled, 402

and reduced time spent on the non-stimulus side of a T-maze assay (Zang et al., 2019).

403

Supplementation with L. plantarum ST-III partially restored the control-like preference for the 404

non-stimulus side of the T-maze as compared to the triclosan exposed group that did not receive 405

probiotic supplementation (Zang et al., 2019). In a separate study, adult wild-type zebrafish 406

exposed to ethanol for two weeks exhibited reduced anxiety-like behavior, demonstrated by 407

(12)

11

increased vertical exploration and time spent in the top part of the tank, using the novel tank assay 408

(Schneider et al., 2016). In this case, supplementation with a different probiotic strain of 409

Lactobacillus (i.e. L. rhamnosus GG) was not sufficient to block ethanol-dependent behavioral 410

effects (Schneider et al., 2016).

411 412

In addition to aforementioned behavioral studies, there is a single zebrafish study that correlated 413

specific microbial taxa with several transcriptional and physiological endpoints in adult zebrafish 414

exposed to a mixture of five brominated flame retardants (i.e. pentaPBDE mixture or DE-71) 415

(Chen et al., 2018a). Interestingly, the presence of Chlamydia, Thaumarchaeota, or Mycoplasma 416

was inversely correlated with intestinal serotonin levels (Chen et al., 2018a), an essential 417

neurotransmitter that is often disrupted in mood disorders. It remains to be seen whether chemical- 418

dependent alterations in serotonin synthesis and/or turnover can be causally linked to dysbiosis or 419

selection of specific taxa or to behavioral manifestations in the host.

420 421

Taken together, these studies illustrate the utility of the zebrafish model to investigate the 422

interaction between xenobiotics and the host nervous system. These studies also support the 423

concept that causal linkages between chemical exposures and behavioral and/or neurologically 424

relevant molecular endpoints are possible when using the three-colonization status zebrafish 425

experimental system. However, to truly move this field forward, more research is needed to 426

understand the fundamental mechanisms by which microbes influence neurodevelopment and 427

function and whether these same pathways are sensitive to disruption following exposure to widely 428

occurring xenobiotic agents.

429 430

Conclusion 431

432

Deconstructing the influence of intestinal microbes on the neurotoxicity of environmental 433

chemicals is an exciting and emerging field of study. Zebrafish is an excellent model with which 434

to unravel the complex relationship between xenobiotic agents, microbiota, and the host nervous 435

system. In this research domain, the major strength of the model is the ability to directly expose 436

colonized and axenic zebrafish to environmental chemicals then assess the full complexity of the 437

developing nervous system using multifaceted automated behavioral phenotyping. However, 438

major limitations of the model should not be overlooked. Zebrafish lack major GI tract organs such 439

as a stomach and lymph nodes that can affect their response to xenobiotic agents, particularly in 440

the context of the microbiota-gut-brain axis. Also critical from a toxicological perspective, 441

zebrafish microbiota is comprised of unique genera relative to mice, rats, and humans. Much more 442

work is needed to understand whether compositional differences in taxonomy result in xenobiotic 443

toxicokinetic and/or toxicodynamic alterations that are significant at the level of host physiology.

444

Overall, there are major gaps in our understanding of the interactions between environmental 445

chemicals, microbiota, and host nervous system development, function, and disease. To address 446

this, future work should expand this innovative experimental system to include colonization with 447

specific strains or communities of bacteria, a more diverse repertoire of automated behavioral 448

endpoints, and the use unbiased hypothesis-generating approaches (i.e. transcriptomics, 449

metabolomics, and/or proteomics) to ultimately illuminate novel mechanisms by which 450

xenobiotics and microbial-products converge to modulate host nervous system development and 451

function.

452

(13)

12 Figures

453 454

Figure 1: Using the zebrafish multi-colonization system to test whether microbiota affect the 455

developmental neurotoxicity of environmental chemicals. The use of conventionally colonized, 456

axenic, conventionalized, and/or monocolonized zebrafish, coupled with behavior and molecular 457

assays, can be used to uncover interactions between host-associated microbes and xenobiotics that 458

provoke developmental neurotoxicity in the host organism.

459 460

Figure 2: The microbiota-gut-brain axis. This axis allows for bidirectional communication 461

between intestinal microbiota and the host nervous system. Key elements of the pathway include 462

the vagus nerve, hypothalamus-pituitary-adrenal axis, and microbial production of bile acids, 463

neuroactive dietary metabolites, and neurotransmitters, and microbial stimulation of neuroactive 464

host-derived cytokines.

465 466

Figure 3: Strengths and weaknesses of animal models commonly used for microbiome 467

research. 1Keane et al., 2011; 2Fritz et al, 2013; 3Bedell et al., 1997; 4Kostic et al., 2013; 5Howe 468

et al., 2013; 6Patton and Zon, 2001; 7Goldsmith and Jobin, 2012; 8Rawls et al., 2004; 9Trinder et 469

al., 2017; 10Koyle et al., 2016; 11Schafer, 2005; 12Clark and Walker, 2018.

470 471

Figure 4: A framework for microbiota-xenobiotic interactions. Axenic zebrafish (Catron et al., 472

2019b; Davis et al., 2016a; Phelps et al., 2017; Weitekamp et al. 2019), mice (Diaz Heijtz et al., 473

2011; Neufeld et al., 2011), and flies (Schretter et al., 2018) exhibit hyperactivity, as do zebrafish 474

(Phelps et al., 2017) and mice (Desbonnet et al., 2010) exposed to antibiotics. Chemical exposures 475

can elicit dysbiosis of host-associated microbes. Chemical-selected microbes harbor the capacity 476

to biotranform xenobiotic agents (i.e. Toxicokinetic interaction). More work is needed to 477

understand whether chemical-induced dysbiosis or altered xenobiotic transformations cause 478

developmental toxicity in the host organism (i.e. Toxicodynamic interaction).

479 480

(14)

13 References

481 482

Anichtchik, O. V., Kaslin, J., Peitsaro, N., Scheinin, M., and Panula, P. (2004). Neurochemical 483

and behavioural changes in zebrafish Danio rerio after systemic administration of 6‐

484

hydroxydopamine and 1‐methyl‐4‐phenyl‐1, 2, 3, 6‐tetrahydropyridine. Journal of 485

Neurochemistry, 88(2), 443-453.

486 487

Bailey, J. M., Oliveri, A. N., Karbhari, N., Brooks, R. A., Amberlene, J., Janardhan, S., and Levin, 488

E. D. (2016). Persistent behavioral effects following early life exposure to retinoic acid or valproic 489

acid in zebrafish. Neurotoxicology, 52, 23-33.

490 491

Bedell, M. A., Largaespada, D. A., Jenkins, N. A., and Copeland, N. G. (1997). Mouse models of 492

human disease. Part II: recent progress and future directions. Genes and Development, 11(1), 11- 493

43.

494 495

Bencan, Z. and Levin, E. D. (2008). The role of α7 and α4β2 nicotinic receptors in the nicotine- 496

induced anxiolytic effect in zebrafish. Physiology and Behavior, 95(3), 408-412.

497 498

Bercik, P., Denou, E., Collins, J., Jackson, W., Lu, J., Jury, J., Deng, Y., Blennerhassett, P., Macri, 499

J., McCoy, K. D., Verdu, E. F. and Collins, S. C. (2011). The intestinal microbiota affects central 500

levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology, 141(2), 599- 501

609.

502 503

Bertotto, L. B., Dasgupta, S., Vliet, S., Dudley, S., Gan, J., Volz, D. C., and Schlenk, D. (2019).

504

Evaluation of the estrogen receptor alpha as a possible target of bifenthrin effects in the estrogenic 505

and dopaminergic signaling pathways in zebrafish embryos. Science of the Total Environment, 506

651, 2424-2431.

507 508

Bevins, C.L. and Salzman, N.H. (2011). The potter's wheel: the host's role in sculpting its 509

microbiota. Cell and Molecular Life Sciences, 68(22), 3675-85.

510 511

Borrelli, L., Aceto, S., Agnisola, C., De Paolo, S., Dipineto, L., Stilling, R. M., Dinan, T. G., 512

Cryan, J. F., Menna, L. F. and Fioretti, A. (2016). Probiotic modulation of the microbiota-gut- 513

brain axis and behaviour in zebrafish. Scientific Reports, 6, 30046.

514 515

Bravo, J.A., Forsythe, P., Chew, M.V., Escaravage, E., Savignac, H.M., Dinan, T.G., Bienenstock, 516

J., and Cryan, J. F. (2011). Ingestion of Lactobacillus strain regulates emotional behavior and 517

central GABA receptor expression in a mouse via the vagus nerve. Proceedings of the National 518

Academy of Sciences, 108(38), 16050-16055.

519 520

Breit, S., Kupferberg, A., Rogler, G., and Hasler, G. (2018). Vagus nerve as modulator of the 521

brain–gut axis in psychiatric and inflammatory disorders. Frontiers in Psychiatry, 9, 44.

522 523

Buckley, C. E., Ren, X., Ward, L. C., Girdler, G. C., Araya, C., Green, M. J., Clark, B. S., Link, 524

B. A. and Clarke, J. D. (2013). Mirror‐symmetric microtubule assembly and cell interactions drive 525

lumen formation in the zebrafish neural rod. The EMBO Journal, 32(1), 30-44.

526

(15)

14 527

Broderick. N.A., Buchon. N., and Lemaitre. B. (2014). Microbiota-induced changes in drosophila 528

melanogaster host gene expression and gut morphology. MBio, 5(3):e01117-14.

529 530

Cantarel, B. L., Lombard, V., and Henrissat, B. (2012). Complex carbohydrate utilization by the 531

healthy human microbiome. PLOS ONE, 7(6), e28742.

532 533

Cao, J., Navis, A., Cox, B. D., Dickson, A. L., Gemberling, M., Karra, R., Bagnat, M. and Poss, 534

K. D. (2016). Single epicardial cell transcriptome sequencing identifies Caveolin 1 as an essential 535

factor in zebrafish heart regeneration. Development, 143(2), 232-243.

536 537

Carabotti, M., Scirocco, A., Maselli, M. A., and Severi, C. (2015). The gut-brain axis: interactions 538

between enteric microbiota, central and enteric nervous systems. Annals of gastroenterology:

539

quarterly publication of the Hellenic Society of Gastroenterology, 28(2), 203.

540 541

Cassar, S., Dunn, C., Olson, A., Buck, W., Fossey, S., Ramos, M. F., Sancheti, P., Stolarik, D., 542

Britton, H., Cole, T., Bratcher, N., Huang, X., Peterson, R., Longenecker, K., and LeRoy, 543

B. (2017). From the Cover: Inhibitors of Nicotinamide Phosphoribosyltransferase Cause Retinal 544

Damage in Larval Zebrafish. Toxicological Sciences, 161(2), 300-309.

545 546

Catron, T.R., Keely, S.P., Brinkman, N.E., Zurlinden, T.J., Wood, C.E., Wright, 547

J.R., Phelps, D., Wheaton, E., Kvasnicka, A., Gaballah, S., Lamendella, R., Tal, T. (2019a). Host 548

developmental toxicity of BPA and BPA alternatives is inversely related to microbiota disruption 549

in zebrafish. Toxicological Sciences. 167(2), 468-483.

550 551

Catron, T.R., Swank, A., Wehmas, L. C., Phelps, D., Keely, S.P., Brinkman, N.E., McCord, J., 552

Singh, R., Sobus, J., Wood, C.E., Strynar, M., Wheaton, E., and Tal, T. (2019b). Microbiota alter 553

metabolism and mediate neurodevelopmental toxicity of 17β-estradiol. Scientific Reports, 9(1), 554

7064.

555 556

Catron, T.R., Gaballah, S., and Tal, T., (In Press). Using zebrafish to investigate interactions 557

between xenobiotics and microbiota. Current Pharmacology Reports.

558 559

Chen, L., Huang, C., Hu, C., Yu, K., Yang, L., and Zhou, B. (2012). Acute exposure to DE‐71:

560

Effects on locomotor behavior and developmental neurotoxicity in zebrafish 561

larvae. Environmental toxicology and chemistry, 31(10), 2338-2344.

562 563

Chen, L., Zhu, B., Guo, Y., Xu, T., Lee, J. S., Qian, P. Y., and Zhou, B. (2016). High-throughput 564

transcriptome sequencing reveals the combined effects of key e-waste contaminants, 565

decabromodiphenyl ether (BDE-209) and lead, in zebrafish larvae. EnvironmentalPollution, 214, 566

324-333.

567 568

Chen, L., Hu, C., Lai, N. L. S., Zhang, W., Hua, J., Lam, P. K., Lam, J. C. W., and Zhou, B.

569

(2018a). Acute exposure to PBDEs at an environmentally realistic concentration causes abrupt 570

changes in the gut microbiota and host health of zebrafish. Environmental Pollution, 240, 17-26.

571 572

(16)

15

Chen, L., Zhang, W., Hua, J., Hu, C., Lok-Shun Lai, N., Qian, P. Y., Lam, P. K. S., Lam, J. C.

573

W., and Zhou, B. (2018b). Dysregulation of intestinal health by environmental pollutants:

574

involvement of the estrogen receptor and aryl hydrocarbon receptor. Environmental Science and 575

Technology, 52(4), 2323-2330.

576 577

Chlebowski, A. C., Garcia, G.R., La Du, J.K., Bisson, W.H., Truong, L., Massey Simonich, S. L., 578

and Tanguay, R. L. (2017). Mechanistic investigations into the developmental toxicity of nitrated 579

and heterocyclic PAHs. Toxicological Sciences, 157(1), 246-259.

580 581

Clark, R.I. and Walker, D.W. (2018). Role of gut microbiota in aging-related health decline:

582

insights from invertebrate models. Cellular and molecular life sciences, 75(1), 93-101.

583 584

Clarke, G., Grenham, P., Fitzgerald, P., Moloney, R.D., Shanahan, F., Dinan, T.G., and Cryan, J.F.

585

(2013). The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic 586

system in a sex-dependent manner. Molecular Psychiatry, 18, 666-673.

587 588

Dabrowski, K. and Miller, M. (2018). Contested paradigm in raising zebrafish (Danio 589

rerio). Zebrafish, 15(3), 295-309.

590 591

Dahan, D., Jude, B.A., Lamendella, R., Keesing, F., and Perron, G.G. (2018). Exposure to arsenic 592

alters the microbiome of larval zebrafish. Frontiers in microbiology, 9.

593 594

Danilova, N. and Steiner, L. A. (2002). B cells develop in the zebrafish pancreas. Proceedings of 595

the National Academy of Sciences, 99(21), 13711-13716.

596 597

Davis, D.J., Bryda, E.C., Gillespie, C.H., and Ericsson, A.C. (2016a). Microbial modulation of 598

behavior and stress responses in zebrafish larvae. Behavioural brain research, 311, 219-227.

599 600

Davis, D. J., Doerr, H. M., Grzelak, A.K., Busi, S.B., Jasarevic, E., Ericsson, A.C., and Bryda, 601

E.C. (2016b). Lactobacillus plantarum attenuates anxiety-related behavior and protects against 602

stress-induced dysbiosis in adult zebrafish. Scientific reports, 6, 33726.

603 604

De Vadder, F., Kovatcheva-Datchary, P., Goncalves, D., Vinera, J., Zitoun, C., Duchampt, 605

A., Bäckhed, F., and Mithieux, G. (2014). Microbiota-generated metabolites promote metabolic 606

benefits via gut-brain neural circuits. Cell, 156(1), 84-96.

607 608

Desbonnet, L., Garrett, L., Clarke, G., Kiely, B., Cryan, J. F., and Dinan, T. G. (2010). Effects of 609

the probiotic Bifidobacterium infantis in the maternal separation model of 610

depression. Neuroscience, 170(4), 1179-1188.

611 612

Diaz Heijtz R., Wang S., Anuar F., Qian Y., Björkholm B., Samuelsson A., Hibberd M.L., 613

Forssberg H., Pettersson S. (2011). Normal gut microbiota modulates brain development and 614

behavior. Proc. Natl. Acad. Sci. U. S. A. 108, 3047–3052.

615 616

(17)

16

Dishaw, L.V., Hunter, D.L., Padnos,B., Padilla, S., and Stapleton, H.M. (2014). Developmental 617

exposure to organophosphate flame retardants elicits overt toxicity and alters behavior in early life 618

stage zebrafish (Danio rerio). Toxicological Sciences, 142(2), 445-454.

619 620

Eisen, J. S. and Smith, J. C. (2008). Controlling morpholino experiments: don't stop making 621

antisense. Development, 135(10), 1735-1743.

622 623

Farrar, M.J., Kolkman, K.E., and Fetcho, J.R. (2018). Features of the structure, development, and 624

activity of the zebrafish noradrenergic system explored in new CRISPR transgenic lines. Journal 625

of Comparative Neurology, 526(15), 2493-2508.

626 627

Fernandes, Y., Rampersad, M., and Gerlai, R. (2015). Impairment of social behaviour persists two 628

years after embryonic alcohol exposure in zebrafish: A model of fetal alcohol spectrum 629

disorders. Behavioural Brain Research, 292, 102-108.

630 631

Fleming, A., Diekmann, H., and Goldsmith, P. (2013). Functional characterization of the 632

maturation of the blood-brain-barrier in larval zebrafish. PLOS One.

633

https://doi.org/10.1371/journal.pone.0077548.

634 635

Fraser, T.W., Khezri, A., Lewandowska-Sabat, A.M., Henry, T., and Ropstad, E. (2017).

636

Endocrine disruptors affect larval zebrafish behavior: Testing potential mechanisms and 637

comparisons of behavioral sensitivity to alternative biomarkers. Aquatic Toxicology, 193, 128- 638

135.

639 640

Fritz, J.V., Desai, M. S., Shah, P., Schneider, J. G., and Wilmes, P. (2013). From meta-omics to 641

causality: experimental models for human microbiome research. Microbiome, 1(1), 14.

642 643

Gaulke, C.A., Barton, C.L., Proffitt, S., Tanguay, S.L., and Sharpton, T.J. (2016). Triclosan 644

exposure is associated with rapid restructuring of the microbiome in adult zebrafish. PLoS One.

645

11(5), e0154632.

646 647

Glazer, L., Wells, C. N., Drastal, M., Odamah, K.A., Galat, R.E., Behl, M., and Levin, E.D. (2018).

648

Developmental exposure to low concentrations of two brominated flame retardants, BDE-47 and 649

BDE-99, causes life-long behavioral alterations in zebrafish. Neurotoxicology, 66, 221-232.

650 651

Goldsmith, J. R. and Jobin, C. (2012). Think small: zebrafish as a model system of human 652

pathology. Journal of Biomedicine and Biotechnology, 2012.

653

http://dx.doi.org/10.1155/2012/817341.

654 655

Gonzalez, S. T., Remick, D., Creton, R., and Colwill, R.M. (2016). Effects of embryonic exposure 656

to polychlorinated biphenyls (PCBs) on anxiety-related behaviors in larval 657

zebrafish. Neurotoxicology, 53, 93-101.

658 659

Hayashi, L., Sheth, M., Young, A., Kruger, M., Wayman, G.A., and Coffin, A. B. (2015). The 660

effect of the aquatic contaminants bisphenol-A and PCB-95 on the zebrafish lateral 661

line. Neurotoxicology, 46, 125-136.

662

Referenzen

ÄHNLICHE DOKUMENTE

The classification of different types of pasture land using remote sensing is difficult and has 559. so far only been partly solved (Schuster et

variability in functional traits due to both standing genetic variation and evolutionary change 701. in response to changing local

A pulse repetition time of 96 ms was considered the species specific ‘attractive’ value, calculated from previously recorded males (grey area depicts mean male trait ±sd). Note

of mean and maximal canopy height, stand density, vertical heterogeneity and wood density for biomass

For neutral, recall that because all individuals share the same demographic rates, dynamics for total community abundance can be treated as a single species in the Levins model,

The particularities of both the ecosystem and the human system generate different combinations of bundles, with synergies and trade-offs among ecosystem services in space and

In summary, as plant genetic diversity, especially at low species richness, seems to increase the interaction partner redundancy in interaction networks and the diversity of

Given our derived indices of PEB, we developed linear regression models to estimate the relationship between pro-environmental behaviour and the different factors which might