developmental toxicity of domoic acid in zebrafish (danio rerio)

7
Developmental toxicity of domoic acid in zebrafish (Danio rerio ) B Jessica A. Tiedeken a , John S. Ramsdell a, * , Ann F. Ramsdell b,c a Marine Biotoxins Program, Center for Coastal Environmental Health and Biomolecular Research, NOAA, National Ocean Service, 219 Fort Johnson Rd., Charleston, SC 29412, USA b Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, SC, USA c Department of Cell and Developmental Biology and Anatomy, University of South Carolina School of Medicine and Program In Women’s Studies, University of South Carolina College of Liberal Arts, Columbia, SC, USA Received 12 April 2005; accepted 12 May 2005 Available online 2 August 2005 Abstract Domoic acid (DA) is a rigid analog of the excitatory amino acid glutamate. It is produced by the diatom genus Pseudo-nitzschia and is a potent neurotoxin in both adult and developing animals. We have used zebrafish (Danio rerio ) as a model to investigate and characterize the developmental toxicity of DA. Domoic acid was administered by microinjection to fertilized eggs at the 128- to 512-cell stages in concentrations ranging from 0.12 to 17 mg/kg (DA/egg weight). DA reduced hatching success by 40% at 0.4 mg/kg and by more than 50% at doses of 1.2 mg/kg and higher. Fifty percent of embryos treated with 1.2 mg/kg DA showed marked tonic –clonic type convulsions at 2 days post fertilization. Four days post fertilization (dpf), all embryos treated with 4.0 mg/kg DA and higher showed a complete absence of touch response reflexes. Commencing 5 dpf, rapid and constant pectoral fin movements were observed, a response which may be related to the hallmark effect in rodents of stereotypic scratching. These data indicate that zebrafish show symptoms of developmental DA toxicity as well as a similar sensitivity comparable to the effects of DA characterized in laboratory rodents. D 2005 Elsevier Inc. All rights reserved. Keywords: Domoic acid; Zebrafish; Glutamate; Development; Embryo 1. Introduction Domoic acid (DA), a structural relative to kainic acid and the neurotransmitter glutamate, activates AMPA and kainite subtypes of the glutamate receptor family resulting in excitotoxicity predominantly in brain tissues [24]. Produced by the diatom genus Pseudo-nitzschia , DA is responsible for poisonings in marine species as well as humans, known as amnesic shellfish poison (ASP). Humans become intoxicated through consumption of shellfish, which accu- mulate DA by ingesting Pseudo-nitzschia [25]. Birds and marine mammals receive effective DA doses through ingestion of contaminated, planktivirous fish [12,17, 29,37]. With prior research focusing on behavioral and neurological responses [8,23,32], the myriad of effects exhibited by these organisms following DA exposure are still being investigated. Consistent with environmental effects observed in sea lions [12], symptomatic responses to DA exposure have been repeatedly documented in mice, rats, and cynomolgus monkeys [13,32,35,34]. Various doses of DA are charac- terized by stereotypic scratching, tremors, and tonic – clonic seizures [13,23,32,35]. Along with the behavioral symp- toms, domoic acid exposure has been reported to cause prolonged neuroexcitation and extensive degeneration in brain tissues [24]. The hippocampus exhibits the most 0892-0362/$ - see front matter D 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.ntt.2005.06.013 i Disclaimer notice: The National Ocean Service (NOS) does not approve, recommend, or endorse any proprietary product or material mentioned in this publication. No reference shall be made to NOS, or to this publication furnished by NOS, in any advertising or sales promotion which would indicate or imply that NOS approves, recommends, or endorses any proprietary product or proprietary material mentioned herein or which has as its purpose any intent to cause directly or indirectly the advertised product to be used or purchased because of NOS publication. * Corresponding author. Tel.:+1 843 762 8510; fax: +1 843 762 8700. E-mail address: [email protected] (J.S. Ramsdell). Neurotoxicology and Teratology 27 (2005) 711 – 717 www.elsevier.com/locate/neutera

Upload: jessica-a-tiedeken

Post on 10-Sep-2016

218 views

Category:

Documents


2 download

TRANSCRIPT

  • Developmental toxicity of domoic

    Jessica A. Tiedeken a, John S. Ram

    Health

    Charl

    al Un

    ity of

    ge of

    05; ac

    ine 2

    monkeys [13,32,35,34]. Various doses of DA are charac-

    terized by stereotypic scratching, tremors, and tonicclonic

    seizures [13,23,32,35]. Along with the behavioral symp-

    reported to cause

    publication furnished by NOS, in any advertising or sales promotion which

    would indicate or imply that NOS approves, recommends, or endorses any

    Neurotoxicology and Teratology 27proprietary product or proprietary material mentioned herein or which has

    as its purpose any intent to cause directly or indirectly the advertised1. Introduction

    Domoic acid (DA), a structural relative to kainic acid and

    the neurotransmitter glutamate, activates AMPA and kainite

    subtypes of the glutamate receptor family resulting in

    excitotoxicity predominantly in brain tissues [24]. Produced

    by the diatom genus Pseudo-nitzschia, DA is responsible

    for poisonings in marine species as well as humans, known

    as amnesic shellfish poison (ASP). Humans become

    intoxicated through consumption of shellfish, which accu-

    mulate DA by ingesting Pseudo-nitzschia [25]. Birds and

    marine mammals receive effective DA doses through

    ingestion of contaminated, planktivirous fish [12,17,

    29,37]. With prior research focusing on behavioral and

    neurological responses [8,23,32], the myriad of effects

    exhibited by these organisms following DA exposure are

    still being investigated.

    Consistent with environmental effects observed in sea

    lions [12], symptomatic responses to DA exposure have

    been repeatedly documented in mice, rats, and cynomolgus

    i Disclaimer notice: The National Ocean Service (NOS) does not

    approve, recommend, or endorse any proprietary product or material

    mentioned in this publication. No reference shall be made to NOS, or to thisdevelopmental toxicity of DA. Domoic acid was administered by microinjection to fertilized eggs at the 128- to 512-cell stages in

    concentrations ranging from 0.12 to 17 mg/kg (DA/egg weight). DA reduced hatching success by 40% at 0.4 mg/kg and by more than 50% at

    doses of 1.2 mg/kg and higher. Fifty percent of embryos treated with 1.2 mg/kg DA showed marked tonicclonic type convulsions at 2 days

    post fertilization. Four days post fertilization (dpf), all embryos treated with 4.0 mg/kg DA and higher showed a complete absence of touch

    response reflexes. Commencing 5 dpf, rapid and constant pectoral fin movements were observed, a response which may be related to the

    hallmark effect in rodents of stereotypic scratching. These data indicate that zebrafish show symptoms of developmental DA toxicity as well

    as a similar sensitivity comparable to the effects of DA characterized in laboratory rodents.

    D 2005 Elsevier Inc. All rights reserved.

    Keywords: Domoic acid; Zebrafish; Glutamate; Development; Embryopotent neurotoxin in both adult and developing animals. We have usAbstract

    Domoic acid (DA) is a rigid analog of the excitatory amino acid glutamate. It is produced by the diatom genus Pseudo-nitzschia and is a

    ed zebrafish (Danio rerio) as a model to investigate and characterize theaMarine Biotoxins Program, Center for Coastal Environmental

    219 Fort Johnson Rd.,bDepartment of Cell Biology and Anatomy, Medic

    cDepartment of Cell and Developmental Biology and Anatomy, Univers

    University of South Carolina Colle

    Received 12 April 20

    Available onl0892-0362/$ - see front matter D 2005 Elsevier Inc. All rights reserved.

    doi:10.1016/j.ntt.2005.06.013

    product to be us

    * Corresponding author. Tel.:+1 843 762 8510; fax: +1 843 762 8700.

    E-mail address: [email protected] (J.S. Ramsdell).acid in zebrafish (Danio rerio)B

    sdell a,*, Ann F. Ramsdell b,c

    and Biomolecular Research, NOAA, National Ocean Service,

    eston, SC 29412, USA

    iversity of South Carolina, Charleston, SC, USA

    South Carolina School of Medicine and Program In Womens Studies,

    Liberal Arts, Columbia, SC, USA

    cepted 12 May 2005

    August 2005

    (2005) 711 717

    www.elsevier.com/locate/neuteratoms, domoic acid exposure has beened or purchased because of NOS publication.prolonged neuroexcitation and extensive degeneration in

    brain tissues [24]. The hippocampus exhibits the most

  • gy andamage, with DA also affecting the septum and olfactory

    bulb [6,24,28]. The hippocampal degeneration results in

    both learning and memory deficits documented in DA

    exposed humans and experimental animals [3,26,30,33].

    Most of the published studies are conducted on mature

    subjects, while the impact of DA exposure on development

    has not been fully investigated. A few researchers have

    described developmental effects of DA induced toxicity in

    laboratory rodents. Dakshinamurti et al. [9] demonstrated

    that intrauterine exposure of 0.6 mg DA/kg dam body

    weight to the developing mice offspring induced age related

    developmental neurotoxicity, with hippocampal necrosis

    observable after just 30 postnatal days. Levin et al. [19] used

    a similar exposure which lead to persisting neurobehavioral

    effects at lower DA doses (0.3 mg/kg) where overt clinical

    signs of toxicity are not present. Xi et al. [38] reported that

    neonatal rats were substantially more susceptible to DA than

    adults, which was proposed to result from insufficient renal

    clearance of toxin, allowing increased bioavailability in the

    blood. The maturation of renal function correlates with the

    decrease in susceptibility to DA as a function of neonatal

    age [10,38]. Neonatal rat exposure to DA, as passed from

    the blood to the milk of lactating mother rats, exists, but at

    levels appearing to be well below symptomatic doses [21].

    Developmental effects in oviparous species including

    frog (Xenopus sp), medaka (Oryzias latipes), and zebrafish

    (Danio rerio) have been successfully studied through

    embryo microinjection of substances ranging from aquatic

    pollutants to neuroactive biotoxins to mRNA strands

    [22,27]. This approach permits the use of minute amounts

    of rare bioactive substances, accurate dose calibration, and

    direct entry into specific tissues and cells. Microinjection of

    medaka embryos has been used to characterize several

    classes of algal toxins including ciguatoxins, brevetoxins,

    azaspiracids, and microcystins [7,11,14,15]. Developmental

    responses of the embryos are characteristic such that subtle

    differences between metabolites of the same toxin can be

    distinguished [5,4]. Because several of these classes of algal

    toxins have bioaccumulation potential, medaka microinjec-

    tion has been used to mimic the maternal transfer of algal

    toxins along with characterizing developmental effects.

    Domoic acid, in contrast, is rapidly eliminated without

    metabolism in urine and hence has little bioaccumulation

    potential [21,31]. Despite this rapid elimination, deleterious

    developmental effects may still occur in these embryo

    stages. By using microinjection, exact doses of DA can be

    attributed directly to symptoms, and consistency between

    embryos is solidified.

    To study early developmental effects of DA, the zebra-

    fish embryo (Danio rerio) was chosen as a test subject

    because of its characterized rapid development, allowing

    increased potential of effects before DA can be eliminated,

    and its importance as a genetic study model. This report

    provides an initial characterization of the developmental

    J.A. Tiedeken et al. / Neurotoxicolo712toxicity of domoic acid in zebrafish embryos and presents a

    model comparable to previously described rodent studies.2. Methods

    2.1. Zebrafish

    Fifty mixed sex wild type zebrafish (Danio rerio)

    were obtained from Tideline Aquatics (Hanahan, SC).

    Fish were kept on a 14 h light : 10 h dark cycle in an 80

    L aquarium. Water quality was maintained with both a

    Whisper 30 Power Filter and weekly 10% water changes

    of 0.06 g/L Reef Crystalsi salt mix (Aquatic Ecosys-tems, Apopka, FL). Sodium bicarbonate solution was

    added to stabilize pH levels between 7.5 and 8.5 at a

    water temperature of 28 -C. Zebrafish were fed twicedaily with TetraMin Tropical Fish Flakes (Tetra, Blacks-

    burg, VA) and afternoons before breeding, the diet was

    supplemented with live Artemia. Trays of marbles with a

    plastic plant in the center were placed on the bottom of

    the cleaned tank to collect the fertilized eggs (embryos)

    in the morning. The embryos were siphoned from the

    marbles within the first 2 h of the light cycle, separated

    and washed with sterile water.

    2.2. Sample preparation and verification

    Domoic acid (DA), along with all other reagents, was

    purchased from Sigma Chemical (St. Louis, MO). The DA

    was resuspended in sterile phosphate buffered saline (PBS)

    to a stock concentration of 10 mg/mL. The stock was diluted

    to 7.54, 2.38, 0.75, 0.24, and 0.075 Ag/AL in sterile PBS tocreate appropriate doses in 1.4 mg embryo wet weight.

    Doses were chosen in half log steps around 4.0 mg/kg, a

    common effective DA dose in developmental rodent

    exposures.

    To determine correct dosing, 2.4 nL samples of 2.38 Ag/AL DA concentrations were collected by microinjection into5 AL of PBS. These samples were taken before (n =4),between (n =2), and after (n =4) the embryo injections to

    measure consistency of injection dose. The samples were

    then diluted to a concentration of 110 pg/mL (DA/PBS) and

    confirmed by domoic acid ELISA (Biosense, Norway).

    Analysis of diluted samples produced values averaging

    100T27 pg/mL. The variance was insignificant, proving thatthe microinjections were accurate and consistent throughout

    the experiment.

    2.3. Microinjection

    Six hours post fertilization (pf), healthy embryos were

    grouped in troughs imbedded in an agarose plate as

    described in The Zebrafish Book [36]. The plates, one for

    controls and one for DA injections, were filled with 12.5%

    Hanks solution [36] until embryos were submerged and

    observed using an Olympus S2X9 microscope. A pulled (P-

    87; Sutter Instrument Co., Navato, CA) and bevelled (BV-

    d Teratology 27 (2005) 71171710; Sutter Instrument Co., Navato, CA) aluminosilicate

    filament micropipette (O.D. 1 mm; Sutter Instrument Co.,

  • embryos had hatched as well as embryos exposed to the

    lowest dose (0.12 mg/kg) of DA; 61.5% of embryos

    ogy anNavato, CA) was filled with a known DA concentration

    using a microloader pipette tip and placed in a micro-

    manipulator. A nitrogen gas pico-injector (Harvard Appa-

    ratus, PLI-90, Holliston, MA) was calibrated to consistently

    produce 2.4 nL of injection material. Starting with the sterile

    PBS group (0 Ag DA/g), up to 30 embryos were injected inorder of increasing concentration for each dose. The

    embryos were removed from the plates and transferred

    individually to wells of a sterile 24-well plate (Corning Life

    Sciences, Acton, MA) with 2 mL of sterile 12.5% Hanks

    solution, which was gradually replaced with zebrafish

    aquarium water in later stages. Twenty-four non-injected

    embryos were used as a control for the vehicle-injected

    eggs.

    2.4. Fish development monitoring

    Embryo plates were maintained at 25 -C under a 16 hlight : 8 h dark cycle. A stereomicroscope (Leica MZ 12)

    with an ocular micrometer was utilized to conduct obser-

    vations on the development of the fish embryos. Embryos

    that did not survive 24 h into the experiment were excluded

    from the study due to the possibility of outside forces

    resulting in mortality. This mortality was constant through-

    out all groups, even controls, and was determined to be

    unrelated to DA dose. Embryo stages were correlated to

    normal development at 28.5 -C using an equation asdescribed by Kimmel et al. [16]. Embryos were observed

    daily for viability, hatching, movements, physical abnor-

    malities, and heart rate (beats/min) after 2 dpf. Heart rate

    was measured (using counter and 30 s timer) in chorion until

    hatched where measurements were taken under sedation (5

    and 6 dpf). Digital images were captured using a RGB

    autoimagecam (MicroImage Video Systems Co. A209,

    Boyertown, PA) mounted onto the microscope. Images

    were enhanced using Image Pro Plus video frame grabbing

    software (Media Cybernetics, Silver Spring, MD). Obser-

    vations were concluded 6 dpf followed by euthanization

    with a lethal concentration of MS 222 (Ethyl 3-amino-

    benzoate methanesulfonate salt, Sigma Chemical, St. Louis,

    MO).

    2.5. Swimming behavior

    In a repeat experiment, hatched larvae were kept to 11

    dpf to test reflexive actions to mechanical stimuli. On

    day 5 pf the larval zebrafish touch reflexes were tested

    by gently placing a polished micropipette against the side

    of the tail. Response to this action was noted and in

    cases where burst of swimming resulted, duration was

    recorded. Those embryos that had not yet hatched 7 dpf

    were gently dechorionated using forceps to provide

    enough subjects for the swimming study. On days 8

    and 11 pf, the behavior of the swimming larvae was

    J.A. Tiedeken et al. / Neurotoxicolmeasured in a specially designed well divided into 16

    equal areas. A baseline swimming activity was measuredhatched from the 0.4 mg/kg DA treatment (Fig. 1). As the

    dose of DA increased, the percentage of hatched embryos

    continued to decrease with a low of 25% hatched at 12.6

    mg/kg. The 16.8 mg/kg treatment had 30% hatched, an

    increase resulting from fewer total viable embryos in the

    treatment group at 5 dpf.

    Most of the unhatched embryos from the higher DA

    treated groups exhibited morphological effects, including

    downward curvatures of the spinal column, swelling of the

    pericardia, and jaw underdevelopment, which reduced

    embryo survival. After dead embryos were removed (24

    h post-injection), 100% of the controls, and 75% of the

    DA dosed embryos, from 0.12 to 4.0 mg/kg, survived until

    the experiment was terminated. In the 12.6 mg/kg DA

    treatment group, 61% of the embryos survived, while in

    the 16.8 mg/kg group only 43% survived (results not

    shown). No critical time point was found to have increased

    mortality; however, unhatched embryos past 5 dpf had

    depleted yolk resources and were unable to feed while still

    in the chorion. This inability to feed caused the embryos toby counting how many partitions were crossed in 1 min,

    followed by vibrational stimulated swimming level in the

    second minute, and a response swimming level in the

    third minute. This testing protocol followed a similar one

    that Levin et al. [18] used when testing chlorpyrifos

    toxicity on larval zebrafish; however, the temperature was

    maintained at 25 -C.

    2.6. Analysis

    Dunnetts multiple comparisons tests in addition to an

    analysis of variance (JMPi statistical software, SASInstitute Inc., Cary, NC) were used to compare the responses

    of embryos at each DA treatment dose to the responses of

    both the embryos treated only with the vehicle, and the non-

    injected embryos. Morphological and behavioral differences

    were compared using the images captured with a Sony RGB

    camera (DXC-390, Sony Corporation, Japan) and captured

    with Image Pro Plus Software (Media Cybernetics, Silver

    Spring, MD).

    3. Results

    3.1. Hatch rate and viability

    Zebrafish embryos hatched sporadically between 90 to

    120 h post-fertilization at 25 -C, corresponding tocalculated stages from Kimmel et al. [16]. No significant

    difference was found between the non-injected controls

    and vehicle injected controls in any of the parameters

    measured. By day five observations, all viable control

    d Teratology 27 (2005) 711717 713become malnourished and more susceptible to fungal

    infections, resulting in mortality.

  • 0.0

    10.0

    20.0

    30.0

    40.0

    50.0

    60.0

    70.0

    80.0

    90.0

    100.0

    0.00 0.12 0.40 1.26 4.00 12.60 16.80

    Dose Concentration of DA (mg/kg)

    Perc

    enta

    ge o

    f via

    ble

    embr

    yos

    n=22 n=25

    n=26

    n=32

    n=26

    n=28 n=20

    Fig. 1. Percent of viable zebrafish embryos hatched 5 days post fertilization (dpf) with respect to treated dose. Total number of embryos viable on 5 dpf is

    expressed above group.

    J.A. Tiedeken et al. / Neurotoxicology and Teratology 27 (2005) 7117177143.2. Cardiovascular effects

    Zebrafish embryos were monitored for heart rate daily

    from 2 to 6 dpf. Heart rate increased as the heart beat

    became more prominent in both treated and nontreated

    embryos between day 2 and 3 from an average of 125T7to 177T8 beats/min, which is a normal developmentalheart rate [16]. No significant differences (95% confi-

    dence) in heart rate were observed between control and

    DA treated embryos throughout the study (results not

    shown).0

    10

    20

    30

    40

    50

    60

    70

    80

    90

    100

    0.00 0.12 0.40 1.

    Dose Concentrat

    Perc

    enta

    ge o

    f Em

    bryo

    s

    n=22

    n=26

    n=32

    n=22 n=26

    n=29

    n=25

    Fig. 2. Percent of viable embryos convulsing ( ) 2 dpf; no convulsions were obs

    touch response (swimming when gently prodded) ( ) on 5 dpf; touch response w

    viable on 2 dpf and 5 dpf is expressed above the group.3.3. Neurotoxic effects

    Neurotoxic effects, resembling tonicclonic convul-

    sions, were evident in zebrafish embryos 2 dpf. Con-

    vulsions were marked as a whole body contraction with a

    shuddering motion. Each contraction lasted approximately

    35 s, with the most frequent contractions occurring at

    the 1.26 mg/kg dose. These convulsions were present in

    37% percent of the embryos treated with 0.4 mg/kg and

    52% of the embryos treated with 1.26 mg/kg DA. This

    response then decreased to 29% in the 4.0 mg/kg26 4.00 12.60 16.80

    ion of DA (mg/kg)

    Touch response (5dpf)Convulsions (2dpf)

    n=26 n=28 n=20

    n=33

    n=27

    n=30 n=21

    erved at doses below 0.4 mg/kg. Percent of hatched embryos that exhibited

    as not observed at doses 4.0 mg/kg and higher. Total number of embryos

  • treatment and was only present in 13% of all higher

    treatments (Fig. 2). This decrease in seizure activity

    correlates to an absence of overall movement from the

    embryos in higher doses.

    3.4. Behavioral response

    3.4.1. Automatism

    The primary behavioral response observed in all DA

    treated embryos was rapid and constant pectoral fin

    movements, commencing at 5 dpf (Fig. 3). In contrast,

    the control larvae exhibited sporadic pectoral fin move-

    ments that were used to right themselves in a vertical

    position. Embryos treated with 1.26 and 4.0 mg/kg DA

    exhibited a hyperactive pectoral fin movement, even when

    still in chorion, which was insufficient to right those

    hatched into a vertical position. Those embryos injected

    with high doses (12.6 and 16.8 mg/kg) also exhibited the

    hyperactive pectoral fin behavior while the rest of the body

    elicited no movements.

    3.4.3. Swimming behavior

    A similar lack of response in higher DA treatments was

    noted in the swimming behavior study. Larvae at doses of

    1.2 mg/kg and higher exhibited an inability to swim or

    move. The few larvae that did show motility at treatments

    above 1.2 mg/kg, exhibited tail paralysis and relied on

    pectoral fin movements and body convulsions to produce

    subtle motions. Subsequently, these larvae were unable to

    cross well divisions and were excluded from the swimming

    study. Once again there was no significant difference

    between the 0.12 mg/kg DA dose and the controls; however,

    the 0.4 mg/kg dose did show a significant decrease

    ( p =0.02) in motion, as measured by divisions crossed,

    while still able to exhibit some normal swimming behaviors.

    There was a significant increase of motion between the

    baseline observation and the vibrational stimulated obser-

    vation across all groups ( p =0.001), with the 0.4 mg/kg

    embryos still exhibiting reduced motion.

    4. Discussion

    entra

    n=32

    a stere

    J.A. Tiedeken et al. / Neurotoxicology and Teratology 27 (2005) 711717 7153.4.2. Touch response

    The natural reflex of the larvae is to escape when

    touched, a commonly tested sensory response. Control

    larvae were able to sense and react to the probe, often before

    being touched, whereas doses of 4.0 mg/kg and higher

    elicited no movement, even when fish were touched

    multiple times (Fig. 2). Those fish in the 0.4 mg/kg and

    1.2 mg/kg treatment groups responded to the touch, but did

    not swim as long (avg 0.75 s) as the controls. The average

    duration of swimming response, (2.6T0.5 s) was notsignificantly different between the controls and 0.12 mg/

    kg treatment larvae.

    0.0

    10.0

    20.0

    30.0

    40.0

    50.0

    60.0

    70.0

    80.0

    90.0

    100.0

    0.00 0.12 0.40

    Dose Conc

    Perc

    enta

    ge o

    f Em

    bryo

    s

    5 dpf6 dpf

    n=22

    n=25

    n=26

    n=25

    Fig. 3. Percent of zebrafish larvae exhibiting incessant pectoral fin motions,day ( ), and maintaining these levels throughout the rest of the experiment. Total n

    one number present indicates the number that remained constant.1.26 4.00 12.60 16.80

    tion of DA (mg/kg)

    n=28

    n=20

    otypic response. This motion first appeared 5 dpf ( ), increasing on the 6thThis report provides the first examination of observable

    DA toxicity on zebrafish development. These toxic effects

    include reduced hatching, which may relate to the spinal

    deformities observed. These deformities were prevalent in

    all embryos that had not hatched by 5 dpf, however the

    precise mechanism effect of DA is unresolved. It is possible

    that DA may inhibit the hatching gland or directly cause the

    downward spinal curvatures that may inhibit the embryo

    from pushing out of the chorion. The other pronounced

    effects of DA on the zebrafish embryos include uncontrolled

    pectoral fin motions and tonicclonic like convulsions.

    n=26 n=27 n=18umber of embryos viable on 5 and 6 dpf is expressed above the group; only

  • block gene expression. Zebrafish offer additional opportu-

    nities for future toxicity studies with an increasing battery of

    gy anAll of the zebrafish embryos and larvae (6 dpf) treated

    with DA exhibited some rapid and constant pectoral fin

    movements. These movements were observable in 100% of

    embryos at 4.0 mg/kg and higher DA treatments, including

    those still in the chorion. While possibly relating to typical

    motions larvae use to position themselves, this particular

    pectoral fin motion seemed to be uncontrolled by the

    individual when compared to the control fish. An uncon-

    trolled response, or automatism, observed in rodents treated

    with DA is stereotypic scratching [32], which is comprised

    of repetitive flexionextension of the hindlimbs directed

    toward the head and neck [10]. This demonstrates that both

    zebrafish and rodents exhibit a similar automatism in

    appendages when dosed with DA.

    Around the prim-22 zebrafish developmental stage (2

    dpf), the DA treated embryos exhibited frequent tonic

    clonic type convulsions. Baraban et al. [2] have recently

    described clonus-like convulsions in zebrafish. Barabans

    study also relates neurological patterns in zebrafish seizures

    with those described in rodents and links the seizures to

    glutamate receptors. As an analog of glutamate, domoic acid

    has been shown to elicit a definitive convulsion response in

    rodents [8]. These continuous tonicclonic convulsions are

    also observed early in developmental rats, between 014

    postnatal days (PND) [10]. Convulsions observed in this

    study closely match those described by Baraban et al. [2]

    and can be related to tonicclonic convulsions expressed in

    postnatal rats.

    The dose dependency of these convulsions and autom-

    atisms also correlates well to developmental toxicity in the

    rat [10,38]. Neonatal rats show an increased (40-fold)

    sensitivity to domoic acid induced automatisms and seizures

    at PND 05 [38] which decreases nearly 10-fold in

    sensitivity between PND 0 (0.12 mg/kg) to PND 22 (1.06

    mg/kg) [10]. The lowest observable effect level of toxicity

    observed in zebrafish by egg microinjection occurs at 0.4

    mg/kg, closely relating to effects observed by intraperitoneal

    injection of PND 14 rats at that dose [10]. These

    developmental effects indicate that zebrafish can exhibit a

    rodent-comparable toxic response, both in types of

    responses and dose dependency to domoic acid.

    While physical symptoms are relatively easy to classify,

    toxin exposure during development may have effects on

    processes with symptoms manifesting later in life. Charac-

    teristically, the effects are time sensitive and dependent on a

    given developmental process as well as the expression of

    potential targets (i.e., receptors and other signaling pro-

    cesses) for toxicity. The midgestational period in rodents

    (PND-13) has been examined for domoic acid toxicity by

    various researchers [9,38]. Studies by Dakshinamurti et al.

    [9] have indicated that exposure to sublethal doses of

    domoic acid leads to changes in hippocampal structure by

    PND-10 in the offspring. A recent related study has

    indicated that persistent neurobehavioral responses occur

    J.A. Tiedeken et al. / Neurotoxicolo716as the rodents mature to juveniles and adults, demonstrated

    by maze trials [38]. Challenges of these adults with aneurobehavioral tests, along with the use of morpholino

    oligonucleotides to characterize toxicity.

    References

    [1] M. Austen, C. Dohrmann, Phenotype-first screening for the

    identification of novel drug targets, Drug. Discov. Today 10

    (2005) 275282.

    [2] S.C. Baraban, M.R. Taylor, P.A. Castro, H. Baier, Pentylenetetrazole

    induced changes in zebrafish behavior, neural activity and c-fos

    expression, Neuroscience 131 (2005) 759768.

    [3] E.C. Clayton, Y.G. Peng, L.W. Means, J.S. Ramsdell, Working

    memory deficits induced by single but not repeated exposures to

    domoic acid, Toxicon 37 (1999) 10251039.

    [4] J.R. Colman, J.S. Ramsdell, The type B brevetoxin (PbTx-3)

    adversely affects development, cardiovascular function, and survival

    in medaka (Oryzias latipes) embryos, Environ. Health Perspect. 111

    (2003) 19201925.

    [5] J.R. Colman, M.Y.B. Dechraoui, R.W. Dickey, J.S. Ramsdell,

    Characterization of the developmental toxicity of Caribbean ciguatox-

    ins in finfish embryos, Toxicon 44 (2004) 5966.

    [6] J.R. Colman, K. Nowocin, R.C. Switzer, T. Trusk, J.S. Ramsdell,

    Mapping and reconstruction of domoic acid-induced neurodegenera-

    tion in the mouse brain, Neurotoxicol Teratol 27 (2005) 753767.

    doi:10.1016/j.ntt.2005.06.009 (this issue).

    [7] J.R. Colman, M.J. Twiner, P. Hess, T. McMahon, T. Yasumoto, M.

    Satake, G.J. Doucette, J.S. Ramsdell, Teratogenic effects of azaspir-

    acid-1 identified by microinjection of Japanese medaka (Oryzias

    latipes) embryos, Toxicon 45 (2005) 881890.

    [8] K. Dakshinamurti, S. Sharma, M. Sundaram, Domoic acid induced

    seizure activity in rats, Neurosci. Lett. 127 (1991) 193197.muscarinic cholinergic antagonist, scopolamine, and the

    NMDA glutamatergic antagonist, dizocilpine, indicate that

    DA exposed animals have a compromised functional

    memory reserve. These two studies indicate that domoic

    acid exposure during development leads to silent effects

    including structural reorganization of brain tissues, neuro-

    behavioral changes and reduced capacitive reserve. This

    provides a possibility that zebrafish embryos may also

    develop symptoms from DA exposure in adulthood that are

    unable to be observed in embryo and larval stages.

    The use of zebrafish has spread to the toxicology field,

    and has been found to be a key neurotoxicological model to

    replace and enhance rodent studies [20]. We report here that

    microinjection of domoic acid into zebrafish eggs causes

    observable symptoms comparable to those found in neonatal

    rats, with close correspondence to the dose sensitivity

    characteristic for this developmental stage. Based upon this

    study, zebrafish should prove a useful species to further

    expand developmental toxicity studies in rodents. More then

    2000 isolated zebrafish mutants have been used throughout

    the medical fields for the understanding of many human

    ailments [1]. Many of these mutants have been genetically

    characterized, and phenotypes can be created early in

    development with morpholino oligonucleotides used to

    d Teratology 27 (2005) 711717[9] K. Dakshinamurti, S. Sharma, M. Sundaram, M.T. Watanabe,

    Hippocampal changes in developing postnatal mice following

  • intrauterine exposure to domoic acid, J. Neurosci. 13 (1993)

    44864495.

    [10] T.A. Doucette, S.M. Strain, G.V. Allan, C.L. Ryan, R.A.R. Tasker,

    Comparative behavioral toxicity of domoic acid and kainic acid in

    neonatal rats, Neurotoxicol. Teratol. 22 (2000) 863869.

    [11] J.S.G. Edmunds, R.A. McCarthy, J.S. Ramsdell, Ciguatoxin reduces

    larval survivability in finfish, Toxicon 37 (1999) 18271832.

    [12] F. Gulland, M. Haulena, D. Fauquier, G. Langlois, M.E. Lander, T.

    Zabka, R. Duerr, Domoic acid toxicity in California sea lions

    (Zalophus californianus): clinical signs, treatment, and survival, Vet.

    Rec. 150 (2002) 475480.

    [13] F. Iverson, J. Truelove, E. Nera, L. Tryphonas, J. Campbell, E. Lok,

    Domoic acid poisoning and mussel-associated intoxication: prelimi-

    nary investigations into the response of mice and rats to toxic mussel

    [25] T.M. Perl, L. Bedard, T. Kosatsky, J.C. Hockin, E. Todd, R.S.

    Remis, An outbreak of toxic encephalopathy caused by eating

    mussels contaminated with domoic acid, N. Engl. J. Med. 322

    (1990) 17751780.

    [26] B.F. Petrie, C. Pinsky, N.M. Standish, R. Bose, G.B. Glavin,

    Parenteral domoic acid impairs spatial learning in mice, Pharmacol.

    Biochem. Behav. 41 (1992) 211214.

    [27] H. Ro, K. Soun, E. Kim, M. Rhee, Novel vector systems optimized for

    injecting in vitro-synthesized mRNA into zebrafish embryos, Mol.

    Cells 17 (2004) 373376.

    [28] A.C. Scallet, Z. Binienda, F.A. Caputo, Domoic acid-treated cyn-

    omolgus monkeys (M. fascicularis): effects of dose on hippocampal

    neuronal and terminal degeneration, Brain Res. 627 (1993) 307313.

    [29] C.A. Scholin, F. Gulland, G.J. Doucette, S. Benson, M. Busman, F.P.

    Chavez, J. Cordaro, R. DeLong, A. De Vogelaere, J. Harvey, M.

    Haulena, K. Lefebvre, T. Lipscomb, S. Loscutoff, L.J. Lowenstine,

    R.I. Martin, P.E. Miller, W.A. McLellan, P.D.R. Moeller, C.L. Powell,

    T. Rowles, P. Silvagnl, M. Silver, T. Spraker, V. Trainer, F.M. Van

    J.A. Tiedeken et al. / Neurotoxicology and Teratology 27 (2005) 711717 717[14] C. Jacquet, V. Thermes, A. deLuze, S. Puiseux-Dao, C. Bernard, J.S.

    Joly, Effects of microcystin-LR on development of medaka embryos

    (Oryzias latipes), Toxicon 43 (2004) 141147.

    [15] K.L. Kimm-Brinson, J.S. Ramsdell, The red tide toxin, brevetoxin,

    induces embryo toxicity and developmental abnormalities, Environ.

    Health Perspect. 19 (2001) 377381.

    [16] C.B. Kimmel, W.W. Ballard, S.R. Kimmel, B. Ullmann, T.F. Schilling,

    Stages of embryonic development of the zebrafish, Dev. Dyn. 203

    (1995) 253310.

    [17] K.A. Lefebvre, C.L. Powell, M. Busman, G.J. Doucette, P.D.R.

    Moeller, J.B. Silver, P.E. Miller, M.P. Hughes, S. Singaram, M.W.

    Silver, R.S. Tjeeredema, Detection of domoic acid in northern

    anchovies and California sea lions associated with an unusual

    mortality event, Nat. Toxins 7 (1999) 8592.

    [18] E.D. Levin, H.A. Swain, S. Donerly, E. Linney, Developmental

    chlorpyrifos effects on hatchling zebrafish swimming behavior,

    Neurotoxicol. Teratol. 26 (2004) 719723.

    [19] E.D. Levin, K. Pizarro, W.G. Pang, J. Harrison, J.S. Ramsdell,

    Persisting behavioral consequences of prenatal domoic acid

    exposure in rats, Neurotoxicol. Teratol. 27 (2005) 719725.

    doi:10.1016/j.ntt.2005.06.017 (this issue).

    [20] E. Linney, L. Upchurch, S. Donerly, Zebrafish as a neurotoxicological

    model, Neurotoxicol. Teratol. 26 (2004) 709718.

    [21] J.M. Maucher, J.S. Ramsdell, Domoic acid transfer to milk: evaluation

    of a potential route of neonatal exposure, Environ. Health Perspect.

    113 (2005) 461464.

    [22] M. Mizell, E.S. Romig, The aquatic vertebrate embryo as a sentinel for

    toxins: zebrafish embryo dechorionation and perivitelline space

    microinjection, Int. J. Dev. Biol. 41 (1997) 411423.

    [23] S. Nakajima, J.L. Potvin, Neural and behavioral effects of domoic

    acid, an amnesic shellfish toxin, in the rat, Can. J. Psychol. 46 (1992)

    569581.

    [24] Y.G. Peng, T.B. Taylor, R.E. Finch, R.C. Switzer, J.S. Ramsdell,

    Neuroexcitatory and neurotoxic actions of the amnesic shellfish

    poison, domoic acid, NeuroReport 5 (1994) 981985.Dolah, Mortality of sea lions along the central California coast linked

    to a toxic diatom bloom, Nature 403 (2000) 8084.

    [30] R.J. Sutherland, J.M. Hoesing, I.Q. Whishaw, Domoic acid, an

    environmental toxin, produces hippocampal damage and severe

    memory impairment, Neurosci. Lett. 120 (1990) 221223.

    [31] C.A.M. Suzuki, S.L. Hierlihy, Renal clearance of domoic acid in the

    rat, Food Chem. Toxicol. 31 (1993) 701706.

    [32] R.A.R. Tasker, B.J. Connell, S.M. Strain, Pharmacology of systemi-

    cally administered domoic acid in mice, Can. J. Physiol. Pharm. 69

    (1991) 378382.

    [33] J.S. Teitelbaum, R.J. Zatorre, S. Carpenter, D. Gendron, A.C. Evans,

    A. Gjedde, N.R. Cashman, Neurologic sequelae of domoic acid

    intoxication due to the ingestion of contaminated mussels, N. Engl. J.

    Med. 322 (1990) 17811787.

    [34] L. Tryphonas, J. Truelove, F. Iverson, Acute parenteral neurotoxicity

    of domoic acid in cynomolgus monkeys (M. fascicularis), Toxicol.

    Pathol. 18 (1990) 297303.

    [35] L. Tryphonas, J. Truelove, E. Nera, F. Iverson, Acute neurotoxicity of

    domoic acid in the rat, Toxicol. Pathol. 18 (1990) 19.

    [36] M. Westerfield, The Zebrafish Book: Guide for the Laboratory Use of

    Zebrafish (Danio rerio), Fourth Edition, University of Oregon Press,

    Eugene, OR, 2000.

    [37] T.M. Work, A.M. Beale, L. Fritz, M.A. Quilliam, M. Silver, K. Buck,

    J.L.C. Wright, Domoic acid intoxication of brown pelicans and

    cormorants in Santa Cruz, California, in: T.J. Smayda, Y. Shimizu

    (Eds.), Toxic Phytoplankton Blooms in the Sea, Amsterdam, Elsevier,

    1993, pp. 643650.

    [38] D. Xi, Y.G. Peng, J.S. Ramsdell, Domoic acid is a potent neurotoxin to

    neonatal rats, Nat. Toxins 5 (1997) 7479.extract, Food Chem. Toxicol. 27 (1989) 377384.

    Developmental toxicity of domoic acid in zebrafish (Danio rerio)IntroductionMethodsZebrafishSample preparation and verificationMicroinjectionFish development monitoringSwimming behaviorAnalysis

    ResultsHatch rate and viabilityCardiovascular effectsNeurotoxic effectsBehavioral responseAutomatismTouch responseSwimming behavior

    DiscussionReferences