non invasive high resolution

18
Author's personal copy Aquatic Toxicology 86 (2008) 20–37 Non invasive high resolution in vivo imaging of -naphthylisothiocyanate (ANIT) induced hepatobiliary toxicity in STII medaka Ron Hardman , Seth Kullman, Bonny Yuen, David E. Hinton Duke University, Nicholas School of the Environment and Earth Sciences, Durham, NC 27708, United States Received 27 June 2007; received in revised form 13 September 2007; accepted 21 September 2007 Abstract A novel transparent stock of medaka (Oryzias latipes; STII), homozygous recessive for all four pigments (iridophores, xanthophores, leucophores, melanophores), permits transcutaneous, high resolution (<1 m) imaging of internal organs and tissues in living individuals. We applied this model to in vivo investigation of -naphthylisothiocyanate (ANIT) induced hepatobiliary toxicity. Distinct phenotypic responses to ANIT involving all aspects of intrahepatic biliary passageways (IHBPs), particularly bile preductular epithelial cells (BPDECs), associated with transitional passage- ways between canaliculi and bile ductules, were observed. Alterations included: attenuation/dilation of bile canaliculi, bile preductular lesions, hydropic vacuolation of hepatocytes and BPDECs, mild BPDEC hypertrophy, and biliary epithelial cell (BEC) hyperplasia. Ex vivo histologi- cal, immunohistochemical, and ultrastructural studies were employed to aid in interpretation of, and verify, in vivo findings. 3D reconstructions from in vivo investigations provided quantitative morphometric and volumetric evaluation of ANIT exposed and untreated livers. The findings presented show for the first time in vivo evaluation of toxicity in the STII medaka hepatobiliary system, and, in conjunction with prior in vivo work characterizing normalcy, advance our comparative understanding of this lower vertebrate hepatobiliary system and its response to toxic insult. © 2007 Elsevier B.V. All rights reserved. Keywords: Fish; Toxicology; Hepatobiliary; Liver; Toxicity; Medaka; ANIT; Biliary; Biliary toxicity; -Naphthylisothiocyanate; Hepatotoxicity; Piscine liver 1. Introduction Bile synthesis and transport, performed by the hepatobiliary system, are essential life functions; fundamental to the elimi- nation of metabolic byproducts, and vital to the assimilation of lipid soluble nutrients (e.g. vitamins A, K, E, triacylglycerols) (Arias, 1988; Boyer, 1996a; Trauner and Boyer, 2003; and oth- ers). Impairment or inhibition of bile synthesis and transport (cholestasis), a common response of the mammalian hepato- biliary system to xenobiotic insult, results in morbidity and Abbreviations: ANIT, -naphthylisothiocyanate; BEC, biliary epithelial cell; BPDEC, bile preductular epithelial cell; BPD, bile preductule; DIC, dif- ferential interference microscopy; ERM, embryo rearing medium; DMSO, dimethylsulfoxide; FITC, fluorescein isothiocyanate; HV, hydropic vacuolation; LSCM, laser scanning confocal microscopy; PCNA, proliferating cell nuclear antigen; TEM, transmission electron microscopy; REACH, Registration, Eval- uation, Authorization and Restriction of Chemicals. Corresponding author at: Environmental Sciences and Policy Division, Nicholas School of the Environment and Earth Sciences, LSRC A333, Box 90328, Durham, NC 27708-0328, United States. Tel.: +1 919 741 0621. E-mail address: [email protected] (R. Hardman). mortality; the result of systemic accumulation of endogenous & exogenous compounds and their metabolites (Alpini et al., 2002b; Arias, 1988; Arrese et al., 1998; Boyer, 1996b; Groothuis and Meijer, 1996; Trauner et al., 1998, 2000; Wolkoff and Cohen, 2003). The majority of our understanding of hepatobiliary trans- port, and vertebrate biliary disease and toxicity, has been derived from mammalian liver studies (Alpini et al., 2002a; Bove et al., 2000; Boyer, 1996a,b; Chignard et al., 2001; and others). We know comparatively less about the piscine biliary system, though we are gaining greater insight into piscine hepatobiliary struc- ture/function relationships (Ballatori et al., 1999, 2000; Boyer et al., 1976a,b; Hampton et al., 1988, 1989; Hardman et al., 2007b; Hinton et al., 1987, 2001; Rocha et al., 1997, 2001). Because our understanding of the piscine biliary system has lagged, par- ticularly in a comparative sense, our ability to interpret and communicate biliary disease and toxicity in piscine species has been limited. By example, cholestasis (impaired/inhibited bile transport) has never been described in fish, a fact more represen- tative of our lack of understanding (investigation), as opposed to the lack of occurrence of this response in piscine systems. 0166-445X/$ – see front matter © 2007 Elsevier B.V. All rights reserved. doi:10.1016/j.aquatox.2007.09.014

Upload: ronchardman

Post on 01-Nov-2014

658 views

Category:

Education


4 download

DESCRIPTION

Non invasive high resolution in vivo imaging of alpha-naphthylisothiocyanate (ANIT) induced hepatobiliary toxicity in STII medaka Ron Hardman∗, Seth Kullman, Bonny Yuen, David E. Hinton Duke University, Nicholas School of the Environment and Earth Sciences, Durham, NC 27708, United States

TRANSCRIPT

Page 1: Non invasive high resolution

Author's personal copy

Aquatic Toxicology 86 (2008) 20–37

Non invasive high resolution in vivo imaging of �-naphthylisothiocyanate(ANIT) induced hepatobiliary toxicity in STII medaka

Ron Hardman ∗, Seth Kullman, Bonny Yuen, David E. HintonDuke University, Nicholas School of the Environment and Earth Sciences, Durham, NC 27708, United States

Received 27 June 2007; received in revised form 13 September 2007; accepted 21 September 2007

Abstract

A novel transparent stock of medaka (Oryzias latipes; STII), homozygous recessive for all four pigments (iridophores, xanthophores, leucophores,melanophores), permits transcutaneous, high resolution (<1 �m) imaging of internal organs and tissues in living individuals. We applied this modelto in vivo investigation of � -naphthylisothiocyanate (ANIT) induced hepatobiliary toxicity. Distinct phenotypic responses to ANIT involving allaspects of intrahepatic biliary passageways (IHBPs), particularly bile preductular epithelial cells (BPDECs), associated with transitional passage-ways between canaliculi and bile ductules, were observed. Alterations included: attenuation/dilation of bile canaliculi, bile preductular lesions,hydropic vacuolation of hepatocytes and BPDECs, mild BPDEC hypertrophy, and biliary epithelial cell (BEC) hyperplasia. Ex vivo histologi-cal, immunohistochemical, and ultrastructural studies were employed to aid in interpretation of, and verify, in vivo findings. 3D reconstructionsfrom in vivo investigations provided quantitative morphometric and volumetric evaluation of ANIT exposed and untreated livers. The findingspresented show for the first time in vivo evaluation of toxicity in the STII medaka hepatobiliary system, and, in conjunction with prior in vivowork characterizing normalcy, advance our comparative understanding of this lower vertebrate hepatobiliary system and its response to toxicinsult.© 2007 Elsevier B.V. All rights reserved.

Keywords: Fish; Toxicology; Hepatobiliary; Liver; Toxicity; Medaka; ANIT; Biliary; Biliary toxicity; �-Naphthylisothiocyanate; Hepatotoxicity; Piscine liver

1. Introduction

Bile synthesis and transport, performed by the hepatobiliarysystem, are essential life functions; fundamental to the elimi-nation of metabolic byproducts, and vital to the assimilation oflipid soluble nutrients (e.g. vitamins A, K, E, triacylglycerols)(Arias, 1988; Boyer, 1996a; Trauner and Boyer, 2003; and oth-ers). Impairment or inhibition of bile synthesis and transport(cholestasis), a common response of the mammalian hepato-biliary system to xenobiotic insult, results in morbidity and

Abbreviations: ANIT, �-naphthylisothiocyanate; BEC, biliary epithelialcell; BPDEC, bile preductular epithelial cell; BPD, bile preductule; DIC, dif-ferential interference microscopy; ERM, embryo rearing medium; DMSO,dimethylsulfoxide; FITC, fluorescein isothiocyanate; HV, hydropic vacuolation;LSCM, laser scanning confocal microscopy; PCNA, proliferating cell nuclearantigen; TEM, transmission electron microscopy; REACH, Registration, Eval-uation, Authorization and Restriction of Chemicals.

∗ Corresponding author at: Environmental Sciences and Policy Division,Nicholas School of the Environment and Earth Sciences, LSRC A333, Box90328, Durham, NC 27708-0328, United States. Tel.: +1 919 741 0621.

E-mail address: [email protected] (R. Hardman).

mortality; the result of systemic accumulation of endogenous& exogenous compounds and their metabolites (Alpini et al.,2002b; Arias, 1988; Arrese et al., 1998; Boyer, 1996b; Groothuisand Meijer, 1996; Trauner et al., 1998, 2000; Wolkoff and Cohen,2003).

The majority of our understanding of hepatobiliary trans-port, and vertebrate biliary disease and toxicity, has been derivedfrom mammalian liver studies (Alpini et al., 2002a; Bove et al.,2000; Boyer, 1996a,b; Chignard et al., 2001; and others). Weknow comparatively less about the piscine biliary system, thoughwe are gaining greater insight into piscine hepatobiliary struc-ture/function relationships (Ballatori et al., 1999, 2000; Boyer etal., 1976a,b; Hampton et al., 1988, 1989; Hardman et al., 2007b;Hinton et al., 1987, 2001; Rocha et al., 1997, 2001). Becauseour understanding of the piscine biliary system has lagged, par-ticularly in a comparative sense, our ability to interpret andcommunicate biliary disease and toxicity in piscine species hasbeen limited. By example, cholestasis (impaired/inhibited biletransport) has never been described in fish, a fact more represen-tative of our lack of understanding (investigation), as opposedto the lack of occurrence of this response in piscine systems.

0166-445X/$ – see front matter © 2007 Elsevier B.V. All rights reserved.doi:10.1016/j.aquatox.2007.09.014

Page 2: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 21

Relevant to the findings presented here is a brief synopsis ofwhat is known about the piscine biliary system. Previous studiesfrom this laboratory have shown the hepatobiliary systemsof channel catfish (Ictalurus punctatus), trout (Oncorhynchusmykiss), and medaka (Oryzias latipes) to exhibit numeroustransitional biliary passageways, termed bile preductules,between hepatocellular canaliculi and biliary epithelial cell(BEC) delimited bile ductules (Hampton et al., 1988; Hardmanet al., 2007b; Okihiro and Hinton, 2000). These transitionalbiliary passageways, first described in the mammalian liver bySteiner and Carruthers (1961), are anatomically associated withperi-portal canals of Hering and oval cells in the mammalianliver (Fausto, 2000; Fausto and Campbell, 2003; Golding et al.,1996; Theise et al., 1999).

More recent in vivo investigations in STII medaka thatelucidated structure/function relationships in both 2 and 3dimensional contexts revealed medaka livers to be replete withbile preductular epithelial cells (BPDECs), and the transitionalbiliary passageways (bile preductules, BPDs) associated withthem (Hardman et al., 2007a,b). These investigations revealedthat the intrahepatic biliary system in medaka is largely an inter-connected network of equidiameter (1–2 �m) canaliculi and bilepreductules, organized through a polyhedral (hexagonal) struc-tural motif, that occupies the majority of the liver corpus (∼95%)uniformly. Larger bile ductules and ducts were predominantlyfound in the hilar and peri-hilar region of the liver, and it follows,an arborizing biliary tree (as described in mammals) was absent,seen only in the rudimentary branching of intrahepatic ductsfrom the hilar hepatic duct. From prior investigations we rec-ognized injury to BPDECs may serve to distort bile preductularlumina and result in transient or longer alterations to intrahep-atic bile flow, and that attention to BPDECs/BPDs, and theirrelationship to the interconnected intrahepatic biliary network,is essential to understanding the spectrum of responses of thepiscine hepatobiliary system to xenobiotics that target this organsystem.

With a better comparative understanding of the medaka hep-atobiliary system established in prior studies, and normalcycharacterized, we were then able to investigate response ofthe hepatobiliary system to xenobiotics in vivo. To do sowe used �-naphthylisothiocyanate (ANIT), a well describedhepatotoxicant that induces hallmark responses in the mam-malian biliary system, namely: cytotoxicity in biliary epitheliumof bile ductules and ducts, cholestasis (Hill and Roth, 1998;Orsler et al., 1999; Waters et al., 2002; Woolley et al., 1979),and biliary tree arborization (biliary epithelial cell hyperpla-sia) (Alpini et al., 1992; Connolly et al., 1988; Masyuk etal., 2003). Because biliary toxicity and cholestasis are poorlyunderstood in piscine species, and because we now under-stand the medaka hepatobiliary system to be more similarto mammalian liver architecture than previously considered(Hardman et al., 2007b), we considered ANIT a good toxicantfor investigation of comparative hepatology. The goals of theseinvestigations were to determine the cellular targets of ANIT inmedaka and to characterize toxic response, relative to what isknown regarding ANIT induced hepatotoxicity in mammalianliver.

2. Materials and methods

2.1. STII medaka

For decades various color mutant strains of medaka (O.latipes), acquired from natural and commercially available pop-ulations, have been maintained in the Laboratory of FreshwaterFish Stocks at Nagoya University, Japan. Cross breeding fromthese stocks was used to produce a stable “transparent” strain ofmedaka. See-through (STII) medaka are homozygous recessivefor all four pigments (iridophores, leucophores, xanthophores,melanophores). Exhibiting no expression of leucophores andmelanophores, and minimal expression of xanthophores andiridiophores, STII medaka are essentially transparent through-out their life cycle (Wakamatsu et al., 2001), and allow highresolution (<1 �m) non invasive in vivo imaging of internalorgans and tissues at the subcellular level (Fig. 1) (Hardmanet al., 2007a,b; Hinton et al., 2004). Our STII medaka colony,maintained at Duke University since 2002, was first culturedwith stock obtained from Prof. Y. Wakamatsu (Nagoya Uni-versity). Medaka were housed in a charcoal filtrated, UVtreated re-circulating system (City of Durham, NC, water) main-tained at 25 ± 0.5 ◦C. Water chemistries were maintained at: pH(7.0–7.4), dissolved oxygen (6–7 ppm), ammonia (0–0.5 ppm),nitrite (0–0.5 ppm) and nitrate (0–10 ppm). A diel cycle of16:8 h light:dark was employed. Medaka larvae were fed ground(pressed through a 60 �m sieve) Otohime � diet (Ashby Aquat-ics, West Chester, PA) via an automatic feeder seven times perday. Because Otohime � has been shown to be free of estro-genic complications (Inudo et al., 2004), we considered it anoptimal fish food. In addition, all brood stock fish diets were sup-plemented daily with Artemia nauplia (hatched brine shrimp).Egg clusters, collected daily, were cleaned in embryo rearingmedium (ERM), and individual fertilized eggs were separatedand maintained in ERM at 25 ◦C. Unconsumed diet, detritusand associated algal material were removed from rearing andbrood stock tanks daily. Care and maintenance of medaka were inaccordance with protocols approved by the Institutional AnimalCare and Use Committee (IACUC; A117-07-04; A141-06-04;A173-03-05).

2.2. Xenobiotic exposures

Studies were designed to evaluate hepatobiliary struc-ture/function during the onset, progression, and recovery fromANIT exposure. Multiple cohorts of STII medaka (10–30 fish)were exposed to the reference toxicant �-naphthylisothiocyanate(ANIT) to target hepatocytes and biliary epithelia for toxicresponse. Acute and chronic aqueous bath exposures werecarried out from 3 to 60 days post fertilization (dpf). Con-trols consisted of untreated medaka, and medaka treated withdimethyl sulfoxide (DMSO; ANIT solvent). All exposures werecarried out in 750 ml wide-bottom glass rearing beakers at25 ◦C, using a 16 h light/8 h dark cycle. Aqueous bath expo-sure medium consisted of ERM:de-ionized water (1:3). Acuteexposures: medaka cohorts were reared in aqueous baths thatwere given a single aliquot of ANIT to achieve exposure con-

Page 3: Non invasive high resolution

Author's personal copy

22 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

Fig. 1. Non invasive in vivo imaging of hepatic parenchyma and blood to bile transport. (A) Brightfield microscopy, STII medaka, 30 dpf, left lateral view. Theliver (L), gall bladder (GB) and associated organs are observable through the abdominal wall. (A1) Widefield fluorescence microscopy of region of interest (graysquare) in frame A, illustrating in vivo imaging of �-Bodipy C5 phosphocholine fluorescence (green) transport through intrahepatic biliary passageways (IHBPs)of the liver, and gall bladder. (B1) Confocal DIC image, single optical section. STII medaka, 9 dpf. Two rows of hepatocytes in longitudinal section characterizeparenchymal architecture (muralium). Hepatic nuclei (HN). Red blood cells in circulation through sinusoids (S/r) appear as stacked ovate structures. (B2) Same as B1,illustrating concentrative transport of �-Bodipy C5 phosphocholine (FITC, green fluorescence) from sinusoids (S/r) to IHBPs. Imaged acquired in vivo 30 min postadministration of fluorophore in aqueous bath. (B3) Composite of B1 (DIC) and B2 (FITC) localizing fluorophore transport to the area between apical membranes ofadjacent hepatocytes. (C) Surface map of region of interest (white square) in B2 illustrating concentration of the fluorophore in IHBPs. (D) Semi-quantitative analysisof fluorescence profile across an 18.3 �m section (white rectangle in frame C) of the parenchyma from sinusoid (S) to IHBP, showed an increase in fluorescence,from sinusoid to canaliculus (IHBP), of ∼20-fold.

centrations ranging from 0.25 �M to 10 �M ANIT. Chronicexposures: medaka were serially exposed every 3 days or onceweekly (static renewal) for the duration of study using the sameconcentrations given for acute exposure. At given time pointsduring exposure regimes (e.g. at 5 min, 15 min. . . 3, 6, 12, 24,48, 72 and 96 h, and day 7, 10, 20, 30, 40 and 60 post expo-sure) subpopulations of medaka were removed from a cohortfor in vivo and ex vivo studies (histological, immunohistochem-ical, and transmission electron microscopy) of the hepatobiliarysystem.

Adult STII medaka (>90 dpf) were anesthetized with0.1 g/L tricane methanesulfonate (MS-222) and twice injected(intraperitoneal; IP) with 200 mg/kg ANIT, with a 7-day depu-ration period between injections. Livers were removed from

anesthetized fish 7 days after the 2nd ANIT injection, placed in4% paraformaldehyde overnight at 4 ◦C, dehydrated in gradedethanol solutions, cleared with xylene, and paraffin embeddedat 60 ◦C. A 200 mg/kg ANIT dose was employed because: ourpreliminary studies in medaka showed 200 mg/kg ANIT to bethe LD50 (96 h) for IP injection; an LD50 of 200 mg/kg of ANIThas been reported in rats following oral exposure (McLean andRees, 1958); and biliary epithelial cell proliferation has beenobserved in rats treated with a single oral dose of 150 mg/kg ofANIT (Kossor et al., 1995). While embryo, larval and juvenilemedaka were subjected to waterborne ANIT exposures, IP injec-tions were employed in adult fish for comparison to publishedANIT studies in rodent animal models, and to investigate thesensitivity of medaka to both routes of exposure.

Page 4: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 23

2.3. In vivo imaging

Prior studies (Fig. 1) described the utility of fluorophoressuch as �-Bodipy C5 phosphocholine and fluorescein isothio-cyanate in elucidating the intra- and extrahepatic biliary systemof STII medaka, and the application of exogenous fluorophoresfor in vivo evaluation of blood to bile transport (Hardman et al.,2007a,b). Briefly, fluorescent probes were administered to con-trol and ANIT treated STII medaka, via aqueous bath, priorto in vivo imaging to aid in elucidation of biological struc-ture/function and interpretation of normalcy and toxic response.Time points for in vivo investigations varied from 10 min to 2 hpost fluorophore administration, dependent on the fluorescentprobe employed, and the portion of the hepatobiliary systembeing studied. By example; Bodipy C5 HPC and fluoresceinisothiocyanate accumulation in the hepatic parenchyma was firstobserved at ∼10 min post fluorophore exposure, with saturation(peak fluorescence; proxy for equilibrium of fluorophore uptakeand excretion) of the fluorophores in the hepatic parenchymaoccurring at ∼40 min. In contrast, Bodipy C5 ceramide satura-tion was commonly observed at ∼70 min. The majority of in vivoobservations were made between 15 and 50 min post fluorophoreexposure. In addition to the use of exogenous fluorophores, aut-ofluorescence was also employed for in vivo elucidation of celland tissue morphology, and xenobiotic response.

After fluorophore exposure medaka embryos, larvae andjuveniles (treated and untreated), at various stages of devel-opment, and at the time-points described, were sedated with10 �M tricaine-methane sulfonate (MS-222) in accordance withIACUC approved animal protocols. Once sedated, medaka weremounted in a solution of de-ionized water:ERM (3:1) on depres-sion well glass slides, oriented in the desired the anatomicalposition, and glass slides sealed with a cover slip. Medaka werethen imaged live with brightfield, widefield and/or laser scan-ning confocal fluorescence microscopy (LSCM). With widefieldand LSCM salient features of the organ system such as canali-culi, space of Disse, endothelial cells, biliary epithelial cells,red blood cells, and hepatocytes and their nuclei, were clearlyresolved. Confocal stacks from in vivo imaging (LSCM) of thehepatobiliary system were used for 3D reconstructions, and fromthese, architectural, morphometric and volumetric analyses weremade.

2.4. Imaging systems

Confocal fluorescence microscopy was performed on a Zeiss510 Meta system with Zeiss LSM 5 Axiovision image acquisi-tion software, Argon and HeNe laser, Carl Zeiss C-apochromatic40×/1.2, and C-apochromatic 10×/0.45. Widefield fluores-cence microscopy was performed on a Zeiss Axioskoppwith DAPI/TRITC/FITC filter cube set. Excitation/emissionparameters for widefield microscopy were: DAPI/UV (Ex360–380 nm/Em All Vis >400 nm); FITC (Ex 450-490 nm/Em515–565 nm); TRITC (Ex 528–552 nm/Em 578–632 nm). Forbrightfield microscopy a Nikon SZM 1500 dissecting micro-scope with a Nikon DXM 1200 digital capture system wasemployed. Software used: EclipseNet (Nikon, USA), Adobe

Table 1Fluorescent probes employed for in vivo investigations

Probe Exposure concentration

7 BR: 7-benzyloxyresorufin 10–50 �M�-Bodipy C5-HPC [BODIPY® 581/591 C5-HPC

(2-(4,4-difluoro-5-(4-phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza-s-indacene-3-pentanoyl)-1-hexadecanoyl-sn-glycero-3-phosphocholine)

30 nM–10 �M

Bodipy FL C5-ceramide[N-(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-pentanoyl)sphingosine]

500 nM–5 �M

FITC: fluorescein isothiocyanate 1 nM–50 �MDAPI [4′,6-diamidino-2-phenylindole,

dihydrochloride], *1 mM solution in DMSO0.3–3.0 �M

YO-PRO®-1 iodide (491/509) 1 �M

Photoshop (Adobe, Inc.), Amira 3D (Mercury Computer Sys-tems, Berlin), ImageJ (V1.32j), IP Lab software (Scanalytics,Inc., version 3.55), and Zeiss Image Browser (Carl Zeiss). Alltransmission electron microscopy (TEM) was performed at theLaboratory for Advanced Electron and Light Optical Methods(LAELOM), College of Veterinary Medicine, North CarolinaState University. For TEM investigations individual medakawere anesthetized and fixed in 4F:1G fixative (4% formalde-hyde and 1% glutaraldehyde in a monobasic phosphate bufferwith a final pH of 7.2–7.4 and a final osmolality of 176 mosmol).Following processing and embedment thin sections (Spurr resinembedded) were made and examined using a FEI/Philips EM208S Transmission Electron Microscope.

2.5. Fluorescent probes

Fluorescent probes employed are listed in Table 1. Fluo-rophores were acquired through Invitrogen/Molecular Probes(Carlsbad, CA). All fluorescent probes were administered toSTII medaka via aqueous bath at the exposure concentrationranges given, under dark conditions, at room temperature.

2.6. Chemicals

�-Naphthyliosthiocyanate (Sigma, N4525), tricaine-methane sulfonate (Sigma, E10521), dimethyl sulfoxide(DMSO) (Sigma, 276855), and Pronase (streptococcalprotease, Sigma).

2.7. Immunohistochemistry

Cytokeratins were localized on paraffin sections of medakaliver using mouse pan-cytokeratin (AE1/AE3, 1:200 dilution)antibody (Zymed, CA, USA) and visualized with the DAKOEnvisionTM+System (Dako, CA, USA). In brief; after de-waxing and rehydration of paraffin sections, heat inducedepitope retrieval was carried out prior to immuno-labeling.Endogenous peroxidase activity was blocked by incubating sec-tions with 0.03% hydrogen peroxide and non-specific labeling

Page 5: Non invasive high resolution

Author's personal copy

24 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

was reduced by blocking sections with 5% normal goat serum for20 min. Sections were incubated with AE1/AE3 (0.01 M phos-phate buffer saline at pH 7.4, 0.1% Tween 20, 0.1% sodium azideand 1% BSA) at 4 ◦C overnight. After rinsing in PBS sectionswere incubated with secondary antibody (peroxidase labeledpolymer conjugated goat anti-mouse immunoglobulin (IgG))at room temperature for 30 min. Sections were rinsed a sec-ond time with PBS, visualized with diaminobenzidine (DAB),counterstained with Harris’ hematoxylin and observed/imagedwith brightfield microscopy. Negative controls were pre-pared by substituting primary antibody with non immuneserum.

After fixation of anaesthetized medaka in 10% formalin for24 h whole mount paraffin sections were prepared and assayedwith proliferating cell nuclear antigen (PCNA; Biogenics, SanRamon, CA) by the histopathology laboratory in the College ofVeterinary Medicine at North Carolina State University. As apositive control, PCNA labeling of the gut was evaluated.

2.8. Statistics

Differences in fluorescence intensity in digital image cap-tures were analyzed statistically using ImageJ (NIH, (V1.32j)and Statview software (SAS institute, Cary, NC). Two wayANOVA with Fisher’s T-test was employed to assess statisticallysignificant differences in fluorescence intensity. Backgroundfluorescence and autofluorescence were accounted for in statis-tical analyses. Descriptive statistics were used for volumetricand morphometric analyses. Pearson’s correlation coefficientwas used for comparison of calculated versus measured mor-phometric values in vivo. Equality of variance F-test wasused for assessment of blood to bile transport; temporalevaluation of fluorescence intensities across sinusoid, hepato-cytic cytosol and canalicular spaces. All quantitative analyseswere performed on unaltered (no deconvolution), or normal-ized, single optical sections from in vivo confocal imagecaptures.

3. Results

3.1. Overview

Exposure of medaka to ANIT resulted in distinct concentra-tion dependent responses, summarized in Fig. 2. These included:(1) canalicular attenuation and dilation in response to 1–3 �Macute aqueous ANIT exposure; (2) bile preductular lesionsin response to 2–5 �M chronic ANIT exposure; (3) hydropicvacuolation, at ANIT concentrations of 2–8 �M ANIT, whichresulted in a distinct “pebbling” of the liver when evaluatedin vivo; and (4) chronic passive hepatic congestion, an endstage response of the liver associated with high mortality, at6–8 �M ANIT. In vivo observations were correlated with exvivo histological and electron microscopic studies to aid ininterpretation of in vivo findings and to verify affected celltypes. These responses are described in detail in the followingsections.

Fig. 2. Overview: responses of the medaka hepatobiliary system to aqueousANIT.

3.2. Canalicular attenuation and dilation and bilepreductular lesions

Medaka cohorts exposed to 1–5 �M aqueous ANIT exhibiteddistinct alterations to the intrahepatic biliary system, to includecanalicular attenuation and dilation (acute exposure), and bilepreductular lesions (chronic exposure). In vivo investigations inacutely exposed medaka revealed a simultaneous attenuationand dilation of bile canaliculi, first observed 4 h post ANITexposure (1–3 �M, Fig. 3). Canalicular attenuation/dilationappeared relatively uniformly throughout the parenchyma, andpersisted for 96–120 h post acute exposure. Both attenuated anddilated canaliculi occurred in close spatial proximity (e.g. within20 �m); where one bile segment, or canaliculus, was observedto be attenuated, the adjacent connected branching bile segmentwas observed to be dilated. Dilated canaliculi, which rangedbetween ∼3 and 4 �m, were found to be up to ∼3 times normaldiameter (1.3 ± 0.4 �m). Attenuated canaliculi were distinct,appearing as fine sinuous passageways 0.4–0.8 �m in diameter(Fig. 3A and B).

Where acute exposure to 1–3 �M aqueous ANIT resulted incanalicular dilation/attenuation, chronic exposures to 2–5 �Maqueous ANIT (>3 days) resulted in foci of alteration thatappeared more consistent with changes to bile preductule(BPD) structural integrity. Where normal (control) canali-culi and bile preductules appeared as equidiameter tubularpassageways (1.3 ± 0.4 �m), altered bile preductules exhib-ited marked irregularities in lumen morphology and increasedlumen diameter (Fig. 4B, C, E and F), suggesting loss ofintegrity of hepatocellular/bile preductular epithelial cell junc-tions. Foci of alteration were frequently associated with changesto bile preductular epithelia cell (BPDEC) morphology. Trans-mission electron micrographs (TEM) of livers from medakacohorts exposed to 2–5 �M aqueous ANIT typically showedBPDECs with an increased cytosolic area, cytosolic vacuo-lation, and altered cell membrane integrity (Fig. 4E and F).When lesions were reconstructed and examined in 3 dimen-sions (Fig. 4D and E) altered BPDs were often found toterminate in blind ends, unconnected to surrounding canali-culi/bile preductules. It follows that the majority of lesions

Page 6: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 25

Fig. 3. Phenotypic responses of the medaka hepatobiliary system: canalicular attenuation and dilation. (A and B) STII medaka, 24 dpf, 2.5 �M aqueous ANIT(48 h, acute exposure): LSCM in vivo imaging illustrating dilated (black arrowheads) and attenuated bile canaliculi (white arrowheads). IHBPs (green fluorescence)elucidated with fluorescein isothiocyanate (FITC). Epithelium is largely non-fluorescent, aside from weak fluorescence of surrounding hepatocellular cytosol andnucleus (HN, gray arrowhead). Frame B is same as frame A, in a different plane of section in the liver. Example diameters of IHBPs are given. (C) STII medaka control,30 dpf, showing typical morphological appearance of IHBPs in vivo. Normalcy finds canaliculi and bile preductules equidiameter throughout the liver, averaging1.3 �m (±0.3 �m), also see blue arrowhead in frame (B). (D) TEM, STII medaka, 30 dpf, 48 h post exposure to 1 �M ANIT. Ultrastructure suggested hepatocellularswelling (HN, hepatocyte nuclei) to be associated with canalicular attenuation/dilation (white and black arrowheads). Normal morphological appearance of a bilepreductular epithelial cell indicated by green arrowhead.

appeared to be localized to BPDEC/hepatocellular junctionalcomplexes (bile preductules); unique morphological complexesdescribed by Hampton et al. (1988) and Hardman et al.(2007b).

BPD lesions were observed in all fish chronically exposedto 2–5 �M aqueous ANIT. Onset of lesions appeared approx-imately 48 h post ANIT exposure, and a higher incidence oflesions was associated with increased exposure duration. Byexample; at 10 days of chronic ANIT exposure approximately10% of bile preductular complexes (transitional biliary passage-ways) appeared to be affected. BPD lesions persisted for theduration of chronic exposure studies (e.g. out to 60 days), andfoci of alteration appeared randomly distributed in the liversexamined. BPD lesions, like canalicular attenuation/dilation,were observed to be a reversible form of injury, as medakawithdrawn from chronic exposures (2–5 �M aqueous ANIT)

exhibited no such phenotype when allowed to recover for 7 daysin an ANIT free bath.

3.3. Hydropic vacuolation

Acute and chronic aqueous exposures to 2–8 �M ANITresulted in a distinct “pebbling” of the liver, a phenotypicresponse consistently evident by 24 h post ANIT exposure(Fig. 5), which was not observed in DMSO or untreatedmedaka, nor readily apparent at lower or higher ANIT expo-sure concentrations. Non invasive in vivo imaging (Fig. 5Cand E) revealed intracellular ovate structures within hepato-cytes and biliary epithelia, which, when viewed at the organlevel of organization (Fig. 5A and B), manifested as a “peb-bling” of the hepatic parenchyma. This phenotype was observedwith the aid of autofluorescence (Fig. 5A and B) and/or

Page 7: Non invasive high resolution

Author's personal copy

26 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

Fig. 4. Phenotypic responses of the medaka hepatobiliary system: bile preductular lesions. (A) STII medaka control, 23 dpf: LSCM, in vivo, single optical section.Sinusoids (S), bile preductules (BPD), hepatocellular nuclei (HN). Parenchyma elucidated with Bodipy C5 Ceramide. (B) STII medaka, 30 dpf: LSCM, in vivo, singleoptical section, 10 days chronic exposure to 2.5 �M aqueous ANIT illustrating BDP lesions (orange arrowheads). White arrows indicate BPDECs. Sinusoid with redblood cell (S/r). (C) STII medaka, 28 dpf: LSCM, in vivo, single optical section, 3 days chronic exposure to 5 �M ANIT, illustrating appearance of dilated BPDs(gray arrowheads) and mild BDPEC cytomegaly (white arrows). (D) STII medaka control, 30 dpf: example of 3D reconstruction of IHBPs (green) and surroundingsinusoids (S, red). (E) STII medaka, 30 dpf, 10 days chronic exposure to 2.5 �M ANIT, reconstruction of terminal BPD lesion (IHBPs). (F) TEM: STII medaka, 26dpf, 5 �M ANIT, illustrating alterations to BPDECs associated with BPD lesions. Inset illustrates normal BPDEC morphology, scale bar = 2 �m.

differential interference microscopy (DIC) alone (Fig. 5E).The application of exogenous fluorophores was not neces-sary for elucidating/imaging this phenotypic response (Fig. 5Aand B).

ANIT exposed medaka exhibiting a “pebbling” phenotypewere treated with the nuclear stain DAPI (via aqueous bath)to investigate whether intracellular ovate structures were asso-ciated with nuclei. Ovate structures did not label with DAPI,were distinguishable from hepatocyte and biliary epithelial cellnuclei, and were localized to the cytosol of affected cell types(Fig. 5C).

Ultrastructural investigations (TEM) revealed the alterationsto be membrane-less cytosolic structures ranging in diameterfrom 2–10 �m, which contained moderate to dense granularinfiltrates of low electron density (Fig. 5D); features con-sistent with hydropic vacuolation (symptomatic cell injuryand swelling, the result of the intracellular accumulation ofwater). Hydropic vacuoles (HV) occurred in both hepato-cytes and BPDECs (Fig. 5D and E), and were observedto displace, and in some instances wrap themselves around,cell nuclei. Vacuoles appeared as early as 6 h post ANITexposure, and were consistently marked by 24 h post expo-sure.

Formation of HVs was concentration dependent, withincreasing prevalence of vacuolation associated with increasing

aqueous ANIT concentrations from 1–6 �M. HV was observedto be most prevalent in the livers of medaka exposed to 4–6 �MANIT, and occurred throughout the hepatic parenchyma, affect-ing ∼95% of the area of livers examined (Fig. 5B). Vacuoleswere first observed in response to 1 �M ANIT, and of low preva-lence, affecting ∼10% of observed areas of the liver. Vacuoleswere also of lower prevalence at ANIT concentrations exceeding6 �M (ranging from 5 to 20% of the area of the liver examined).Hence, a concentration dependent increase in HV was observedfrom 1 to 6 �M ANIT, and decrease observed from 6 to 10 �M.Medaka exhibiting this phenotypic response showed no overtsigns of impaired health, exhibiting normal swimming and feed-ing behavior. HV was not observed in the livers of DMSO controlmedaka.

In the majority of livers studied hepatocytes and biliaryepithelia affected by HV appeared throughout the liver corpus,with no apparent zonation; an observation enabled by the abilityto observe/image internal liver structure at various depths (e.g.up to 200 �m from the liver surface with confocal microscopy).Hydropic vacuoles were found to be a reversible form of ANITinduced cell injury; not observed in any cell type followingrecovery from ANIT exposure. By example, medaka exposedto 3–6 �M ANIT for 3 days, and subsequently reared in anANIT free bath for 7 days, showed no signs of HV in vivo or inultrastructure (TEM) studies.

Page 8: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 27

Fig. 5. Phenotypic responses of the medaka hepatobiliary system: hydropic vacuolation. (A) STII medaka control, 20 dpf, ventral view, widefield fluorescencemicroscopy, autofluorescence (DAPI/UV excitation). Inset scale bar = 100 �m. Gill (GI), ventral aorta (VA), heart atrium (Ha), heart ventricle (Hv), liver (L), gallbladder (GB), gut (Gt). (B) Pebbling phenotype: STII medaka, 20 dpf, ventral view, 4 �M ANIT, 48 h of exposure, widefield fluorescence microscopy (autoflu-orescence; FITC-DAPI/UV composite). Inset scale bar = 20 �m. (C) STII medaka, 29 dpf, 4 �M ANIT, 48 h of exposure. DAPI labeling (blue) differentiatingintracellular ovate structures from nuclei. (D) TEM: STII medaka, 28 dpf, 4 �M ANIT, 24 h of exposure. Hydropic vacuolation in hepatocytes (black arrowheads)and bile preductular epithelia (inset). (E) STII medaka, 29 dpf, 4 �M ANIT, 24 h of exposure: In vivo confocal image (DIC and TRITC composite) of YO-PRO-1labeling (green). Hydropic vacuoles (black arrowhead).

3.4. In vivo investigation of apoptosis

Preliminary studies suggest it is possible to detect cells withcompromised cell membranes in vivo. While these observa-tions are not conclusive, they merit mention. Putative apoptotic,or necrotic cells with compromised cell membranes weredetected/imaged in vivo using confocal microscopy and the flu-orescent probes SYTO® 16 green and YO-PRO-1, both of whichare established in vitro nucleic acid stains used for detection ofapoptosis (the cationic fluorophores only enter cells with com-promised cell membrane integrity) (Al-Gubory, 2005; Santos etal., 2006; Wlodkowic et al., 2007). Of these, YO-PRO-1 wasthe fluorophore of choice for in vivo studies (Fig. 5E). Cellsincorporating YO-PRO-1 were observed in the livers of medakaexposed to 3–6 �M ANIT as early as 6 h post exposure. In thefields of liver observed in vivo, ∼3% of cells fluoresced as aresult of YO-PRO-1 incorporation, indicative of cells with com-

promised membrane integrity. YO-PRO-1 fluorescent cells werenot observed in DMSO controls, and were only observed in tan-dem with the development of hydropic vacuoles, typically within24–72 h post ANIT exposure. Due to the staining characteristicsof YO-PRO-1, which primarily labeled nuclei, and the fact thatlabeled cells also exhibited loss of normal cell morphology, itwas difficult to differentiate affected cell types.

3.5. Biliary epithelial cell proliferation

In vivo and immunohistochemical analyses (AE1/AE3 andPCNA), used to assess proliferation of BPDECs, BECs (cellslining bile ductules and ducts) and hepatocytes in response toANIT, suggest biliary epithelial cells are early responders toANIT, with hepatocytic changes occurring subsequently (Fig. 6).BEC hyperplasia was first observed at 5 days of chronic expo-sure to 2–6 �M ANIT, and remained apparent out to 60 days

Page 9: Non invasive high resolution

Author's personal copy

28 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

Fig. 6. Phenotypic responses of the medaka hepatobiliary system: biliary and bile preductular epithelial cells (A and A1) AE1/AE3 immunohistochemistry: STIImedaka DMSO control, ∼9 months of age. (A2) AE1/AE3 immunohistochemistry: STII medaka, ∼9 months of age, 72 h post injection 200 mg/kg ANIT, illustratingdenser BEC AE1/AE3 labeling and BEC cytomegaly. (B) TEM: STII medaka, 31 dpf, 5 days post exposure to 1 �M ANIT, illustrating BPDEC alterations (redarrowheads) and associated BPDs (orange arrows). Inset: normal BPD and BPDEC (arrowhead) morphology, 26 dpf, scale bar = 2 �m. (B1) TEM: STII medaka,34 dpf, 5 �M ANIT, 24 h of acute exposure, illustrating BEC cytotoxicity (red arrowhead) and associated dilated bile passageway (black arrowhead). Lipid vesicles(gray arrowhead), hydropic vacuolation (white arrowhead). Inset (scale bar = 5 �m) illustrates normal appearance of BECs (gray arrowheads) and associated bilepassageway. (C) LSCM, in vivo, single optical section, STII medaka, 80 dpf, 60 days 2.5 �M ANIT exposure. Under chronic exposure BPDECs (red arrowheads)appeared enlarged and more numerous (contiguously) per unit area of liver examined, as compared to controls. (C1) STII medaka control illustrating normal invivo appearance of BPDs/BPDECs (red arrowheads). Hepatocytes (white arrowhead), sinusoid (gray arrowhead). (D) STII medaka, DMSO control, 55 dpf: six ofeight control livers exhibited no or minimal PCNA staining throughout the liver (L). Gall bladder (GB), Gut (Gt). (D1) STII medaka, 55 dpf, 2.5 �M ANIT, 30 daysof chronic exposure: ANIT treated medaka exhibited stronger and more prevalent PCNA staining in the hilar and peri-hilar regions of the liver (circled areas in Dand D1).

during chronic exposure studies. The majority of proliferatingcell nuclear antigen (PCNA) positive cells were localized to thehilar and peri-hilar region of the livers examined, suggestingBECs were the most responsive (hyperplastic) cell type (Fig. 6Dand D1). PCNA staining was occasionally observed in more dis-tal regions of the liver in small cuboidal biliary epithelia of bileductules/ducts (see Fig. 7A for these cell types), though thisobservation was infrequent, and encountered in livers of maturemedaka, older than 120 days (not shown in figures). PCNA pos-itive hepatocytes, BECs, and BPDECs were not evident in acuteANIT exposures, or chronic exposures exceeding 6 �M ANIT.While PCNA positive BPDECs were not apparent in any expo-sure regime, it is possible that BPDECs, if labeled, were not

clearly resolved in immunohistochemical preparations due totheir small size (3–8 �m), and relatively low density comparedto hepatocytes and biliary epithelial cells (per unit volume ofliver). While BPDEC proliferation was not evidenced by PCNAanalyses, qualitative in vivo observations suggested changes toBPDECs consistent with mild BPDEC cytomegaly, and hyper-plasia. In chronically exposed medaka BPDECs consistentlyappeared, in vivo, more numerous, contiguous, and larger, asopposed to control livers (Fig. 6C).

Immunohistochemical studies with the pan cytokeratin stainAE1/AE3, used to detect in biliary epithelial and hepatocyticcytokeratins, also suggest BECs to be early responders to ANIT.In adult medaka injected with 200 mg/kg ANIT, BECs exhibited

Page 10: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 29

Fig. 7. Bile duct cystic vacuolation in response to aqueous ANIT. (A) STII medaka control, 40 dpf, H&E, showing normal morphology of an intrahepatic bile ductand associated biliary epithelial cells (square and enlarged inset). (B) STII medaka, 40 dpf, H&E, 40 days of chronic exposure to 3 �M ANIT. Cystic formations(black arrowhead) were observed in bile ducts of the liver hilus. Cysts often contained inspissated bile or proteinaceous material. An enlarged vessel filled with redblood cells (gray arrowhead), likely an early branch of the hepatic portal vein, can be seen below and left of the biliary cyst.

cytomegaly, as compared to DMSO (vehicle/solvent) controls(Fig. 6A, A1 and A2). BECs lining bile ductules and ducts inANIT treated livers also appeared to label more heavily withthe AE1/AE3 cytokeratin antibodies, suggesting greater density,and perhaps, proliferation of these cell types. Modest vascularendothelial staining with AE1/AE3 was also observed, thoughwith less consistency and intensity.

3.6. ANIT associated biliary cystic vacuolation

Chronic exposures to 3 �M ANIT resulted in cystic vacuola-tion of larger bile ducts of the liver hilus, with accompaniedhepatocellular changes; responses consistent with spongiosishepatis, a probable degenerative lesion (Brown-Peterson et al.,1999) (Fig. 7B). In histological preparations (H&E) cysts weredistinct, ranged from 40 to 100 �m in diameter, and often con-tained material consistent with inspissated bile or proteinaceousmaterial. Evident in the same livers were smaller hepatocellu-lar vacuoles consistent with hydropic vacuolation (Section 3.3).Cystic formations of this nature were encountered only duringchronic exposures, and were not observed in livers of acutelyexposed medaka (<3 days), regardless of ANIT concentration.

3.7. Hepatobiliary transport

STII medaka exposed to 0.25– 8 �M aqueous ANIT weretreated with the fluorophores FITC and Bodipy C5 Ceramideand examined for altered hepatobiliary transport at 6, 8, 12,24, 48, and 96 h (Fig. 1, Fig. 8). In vivo confocal image cap-tures (single optical sections) were measured (random repeatedmeasures) for fluorescence intensity in sinusoid, cytosol andcanalicular spaces. No impairment of blood to bile transport wasdetected for either fluorophore in response to 0.25–6 �M aque-ous ANIT exposures (Fig. 8). Loss of hepatobiliary transportfunction (for both FITC and Bodipy C5 Ceramide) was onlyobserved in vivo at ANIT exposure concentrations exceeding6 �M, ANIT concentrations also associated with vasodilationand decreased cardiac output (discussed following).

3.8. Volumetric studies on ANIT induced biliary changes

While altered blood to bile transport was not evident via quan-titative in vivo studies with FITC and Bodipy C5 Ceramide,volumetric analyses (which can be considered a proxy forintrahepatic bile flow) from 3D in vivo investigations suggest dif-ferences in intrahepatic biliary volume in ANIT treated versusuntreated medaka. Quantitative morphometric and volumetricanalyses were performed on 3D reconstructions from two ANITtreated medaka and three controls (Table 2). Volumetric indicesof an individual medaka (40 dpf) from a cohort exposed to2.5 �M ANIT for 30 days suggest a reduction in canalicularlumen volume of ∼50% (Table 2). A second evaluation of anindividual medaka from a cohort exposed to 1 �M ANIT for 48 hvolumetric indices suggested an increase in intrahepatic biliaryvolume. Relative to liver volume, biliary volume was found tobe 1.18%, versus a mean of 0.95% (±0.08) in control livers(Table 2). In both cases volumes of vasculature, parenchymaand hepatocellular space, relative to total liver volume exam-ined, remained well conserved across ANIT treated and controlfish, from 8 to 40 dpf, indices which serve as controls, and whichcan be used to assess precision across individual 3D volumetricstudies (Table 2).

3.9. Cardiovascular changes

Aqueous ANIT concentrations of 4–8 �M resulted in phe-notypic changes consistent with passive hepatic congestion. Invivo investigations revealed a marked cardiovascular response,where all liver vasculature, afferent and efferent vessels, showedincreasing dilation in response to increasing ANIT concentra-tions (Fig. 9). Dilation of sinusoids, hepatic vein, and hepaticportal vein, were all observed. Sinusoid diameter was observedto increase ∼2-fold at ANIT concentrations approaching theLC100 (8 �M). Where control sinusoids averaged 7.4 �m indiameter, sinusoid diameter averaged 15.3 �m (±4.1, n = 18) at8 �M ANIT, 48 h post exposure (Fig. 9). Vasodilation of hepaticvasculature was observed in tandem with a concentration depen-

Page 11: Non invasive high resolution

Author's personal copy

30 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

Fig. 8. In vivo evaluations of altered hepatobiliary transport function in response to ANIT. (A) STII medaka control, 40 dpf, LSCM, single optical section, illustratingnormal appearance of FITC (green fluorescence) transport from blood to bile, through IHBPs of the liver (see also Fig. 1). (B) STII medaka, 40 dpf, LSCM, singleoptical section of medaka liver 48 h post exposure to 6 �M ANIT, illustrating reduced fluorescence of FITC in IHBPs and hepatocellular cytosol. Note increasedFITC fluorescence in blood plasma (gray arrowhead), but not in hepatocyte cytosol, and minimal fluorescence in IHBPs. Such an altered fluorescence profile wouldbe consistent with decreased hepatocellular uptake of fluorophore from blood plasma. (C) STII medaka, 20 dpf, quantitative analysis of blood to bile transport. Thestatistical means of fluorescence intensity (n = 30) are given. No statistically significant difference in fluorophore transport was observed between controls and ANITtreated animals (in vivo), at ANIT concentrations below 6 �M.

dent decrease in heart rate and motility, observed in all medaka(n = 18) exposed to 4 �M to 8 �M ANIT (acute and chronicexposures). By example: where control heart rates averaged 134(±9) beats per minute (bpm), medaka exposed to 8 �M ANITexhibited heart rates of 118 (±12) bpm at 6 h post exposure, 73(±13) bpm at 24 h post exposure, and 61 (± 7) bpm at 48 h postexposure. Hence, the magnitude of both the vasodilation andheart rate responses were concentration (ANIT) dependent, andappeared to be coupled.

4. Discussion

Results of this study suggest cells of medaka intrahepaticbiliary system respond to ANIT, and that responses, both cellu-lar and system level, are similar to those described in rodents(Alpini et al., 2001; Carpenter-Deyo et al., 1991; Connollyet al., 1988; Hill et al., 1999; Kossor et al., 1993; Lesage etal., 2001; Orsler et al., 1999; Waters et al., 2001). Changesobserved in vivo, and confirmed with ex-vivo analyses (histolog-

Table 2Volumetric indices from 3D reconstructions: ANIT treated vs. untreated medaka

Compartment Control (%) ANIT treated (%)

8 dpf 12 dpf 30 dpf Mean (%) (S.D.): 8–12 dpf 2.5 �M ANIT 30 dpf 1 �M ANIT 30 dpf

IHBPs 1.03 0.86 1.01 0.97 (0.09) 0.50 1.18Vasculature 6.33 8.60 7.60 7.51 (1.14) 7.30 7.92Parenchyma 93.67 91.40 92.40 92.49 (1.14) 92.70 92.08Hepatocellular 92.64 90.54 91.39 91.52 (1.06) 92.20 92.15

Volumetric comparisons between two ANIT treated and three untreated medaka. Individual indices, statistical mean and standard deviation (S.D.) of volumetricanalyses from control (untreated) livers at 8, 12, and 30 dpf are given. Indices are % volumes relative to volume of liver examined.

Page 12: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 31

Fig. 9. Phenotypic responses of the medaka hepatobiliary system: passive hepatic congestion. (A) STII medaka control (DMSO), 20 dpf, 24 h post exposure,widefield fluorescence (DAPI/UV autofluorescence). Vasculature (V) appears non fluorescent (dark), epithelia of hepatic parenchyma appears light gray. Inset scalebar = 100 �m. Ventral aorta (Va), heart ventricle (Hv), heart atrium (Ha), liver (L). (B &C) STII medaka, 18 dpf, 4 �M ANIT, 24 h post exposure: widefield fluorescence(DAPI/UV autofluorescence) illustrating moderate dilation of hepatic vasculature (V). Inset scale bar = 100 �m. Gill (Gl), sinus venosus (SV), gall bladder (GB). (C)Widefield fluorescence (DAPI/TRITC composite) illustrating FITC (white arrows) in transport through the hepatic parenchyma (gray arrowhead), in the presenceof ANIT induced vasodilation (white arrowhead, also indicating red cells in circulation (S/r)). (D) STII medaka, 17 dpf, 8 �M ANIT, 24 h, widefield microscopy(DAPI/UV). (E) STII medaka, 24 dpf, 8 �M ANIT, 48 h post exposure: In vivo LSCM confirmed dilation of intrahepatic vasculature occurred uniformly throughoutthe liver. Vasculature lumena (white arrowheads), replete with red blood cells, appears dark gray, parenchyma fluoresces green (Bodipy C5 ceramide). (F) TEM: STIImedaka, 8 �M ANIT, 20 dpf, 48 h post exposure: intrahepatic vessel showing abnormal (attenuated) endothelial cell membrane morphology (gray arrowhead), andvasodilation (V). Endothelial cell nucleus (black arrowhead). Graph: ANIT dose-duration relationship of heart rate to sinusoid diameter. Indices are mean, ±S.E.

ical, immunohistochemical, ultrastructural), were; attenuationand dilation of bile canaliculi, bile preductular lesions, hydropicvacuolation of hepatocytes and BPDECs, BPDEC cytomegaly,and hyperplasia of BECs in the hilar and peri-hilar region ofthe liver. While in vivo evaluations revealed no alterations tobile transport, volumetric analyses of 3D reconstructions fromANIT treated medaka suggest a reduction in intrahepatic bil-iary passageway volume at 2.5 �M ANIT (cholestasis?), and anincrease in intrahepatic biliary passageway volume (choleresis?)at 1 �M ANIT, with no changes to other volumetric liver indices(Table 2).

4.1. Canalicular attenuation and dilation

Canalicular dilation/attenuation, frequently observed inresponse to 1–3 �M aqueous ANIT, and evaluated both in vivoand via 3D reconstructions, appeared to be more consistent withan adaptive response of the intrahepatic biliary system, as noclear alteration to overall canalicular integrity or loss of func-tion was observed in association with this change, nor weremortality or morbidity observed. Apical hepatocyte membraneintegrity in dilated canaliculi appeared to be maintained, andcanalicular lumens appeared uniform and smooth, equidiame-ter in dimension, as observed in the hepatocytes (canaliculi) ofnormal livers. In short, altered canalicular lumen diameter wasthe only observed change as compared to untreated livers. Thistype of canalicular response (e.g. mosaic of normal and abnor-mal canaliculi; either dilated canaliculi, or in some instances,collapsed canaliculi of reduced diameter) is often observed dur-ing cholestasis in the mammalian liver (Arias, 1988; Thung andGerber, 1992).

Of interest, canalicular dilation/attenuation was only dis-tinct in vivo. Were in vivo observations not made, it isquestionable whether this phenotypic response would havebeen detected simply employing histological or ultrastructuralstudies alone. Even knowing what to look for from in vivoobservations, attenuated/dilated canaliculi could not be clearlydiscerned in histological preparations. In fixed tissue sectionscanaliculi were often indistinct. It is also possible that tissueprocessing may alter canalicular lumen diameter. Hence, evenwhen resolved/identified, histological observations of canaliculimay have proven inaccurate. Likewise, canalicular attenua-tion/dilation may have gone undetected in TEM investigationswere the response not previously well recognized in vivo(Fig. 3). Hence, in vivo observations were not only importantto elucidation of this phenotypic response, but elucidated thischange in ways ex vivo techniques were not, we found, wellsuited.

What may explain canalicular dilation/attenuation? Bear-ing in mind that hepatocytes have been observed to contributeto 1 to 3 canaliculi, in both mammals and medaka (Arias,1988; Hardman et al., 2007b; Motta, 1975), canalicular atten-uation/dilation could result from two sources: (1) a contractileregulatory problem at the pericanalicular region of hepatocytesinvolving cytoskeletal and tight junctional elements, or (2) it mayresult from the swelling of hepatocytes (or BPDECs, while atten-uation/dilation of intrahepatic biliary passageways appeared tobe largely associated with canaliculi, bile preductules cannot beruled out). In the latter case, hepatocellular swelling could pre-clude or diminish bile flow via reduction of canalicular lumendiameter. If bile secretory rates remained unchanged, canalicu-lar attenuation would force bile to take alternate routes through

Page 13: Non invasive high resolution

Author's personal copy

32 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

the interconnected intrahepatic biliary network, which, in theevent bile volume is conserved, would necessitate the occur-rence of dilated canaliculi in other portions of the intrahepaticcanaliculo-preductular network. Dilated canaliculi may reflectlocally redirected bile from attenuated passageways (i.e. alter-nate routes of flow). This structural change may comprise anadaptive/functional response of the liver to maintain bile secre-tory/transport functions in the presence of impaired bile flow,and may illustrate one of the underlying attributes of an intercon-nected, polyhedral based, canaliculo-bile preductular network;in that it provides alternate routes of bile flow for continu-ous secretion of hepatocellular bile (Hardman et al., 2007a,b).Because diminished/precluded canalicular transport of bile mayresult in hepatocellular toxicity by reducing or inhibiting elimi-nation of potentially toxic cytosolic solutes (from intracellular tocanalicular lumen), an interconnected network of canaliculi andpreductules would allow alternate/multiple routes of elimination(transport) of bile solutes away from hepatocytes for export to thegall bladder and elimination via the gut, circumventing potentialhepatotoxicity (bile retention) in the event of impaired/precludedbile flow in portions of intrahepatic biliary passageways. Hence,because hepatocytes synthesize and secret bile, and may con-tribute bile to one to three individual canaliculi, it is possiblethat simultaneous attenuation/dilation of canaliculi is reflectiveof an adaptive response of the liver to maintain flow of total bilevolume (homeostasis of bile synthesis and secretion).

While attributing canalicular dilation/attenuation to swollenhepatocytes is not definitively supported by the findings pre-sented, it can be hypothesized this is a feasible mechanismby which lumen attenuation/dilation could be occurring. Com-panion TEMs showing altered hepatocytes, in conjunction withassociated dilated/attenuated canaliculi, support this conjecture(Figs. 3–6 Figs. 3D, 4F, 5D and 6B, B1), and studies in rodentsdescribe ANIT induced cytotoxicity in hepatocytes, and biliaryepithelium (Alpini et al., 1992; Connolly et al., 1988; Leonardet al., 1981; Lesage et al., 2001). Because in vivo techniquesfocused on elucidation of IHBPs via application of FITC andBodipy HPC fluorophores (excellent for elucidation of intra-and extrahepatic bile passageways, not optimal for elucidat-ing cell cytosol or organelles), swollen hepatocytes and BECscould not be consistently evaluated in vivo in livers exhibit-ing dilated/attenuated canaliculi. It is possible to co-administerBodipy C5 ceramide (good for elucidating intracellular mor-phology) with either of the aforementioned fluorophores tosimultaneously elucidate both intrahepatic bile passageways andcell morphology in vivo. However, we were not able to ade-quately refine this in vivo procedure during the course of theANIT studies. In summary, results from in vivo and ex vivo anal-yses suggest hepatocellular swelling may be responsible for thedilated/attenuated canaliculi observed, a phenotypic responseto ANIT that merits further study in medaka and other piscinespecies, particularly in regard to the effect of xenobiotics on bilesynthesis and transport.

An alternate hypothesis is that ANIT induced cytoskeletalderangements may be responsible for the altered canalicu-lar morphology (e.g. constriction/dilation regulation). Becausecanaliculi are contractile structures, the function of which is

governed by cytoskeletal proteins and actin filaments in thepericanalicular region of hepatocytes, it is possible the canalic-ular changes observed may be attributable to ANIT induced(either direct or indirect) cytoskeletal alterations in hepatocytes.Cytoskeletal derangements resulting from ANIT exposure havebeen observed in isolated rat hepatocyte couplets (Orsler et al.,1999), and in vivo (Furuta et al., 2004; Kan and Coleman,1986; Lowe et al., 1985). Hence, mammalian studies suggestANIT induced cytoskeletal alterations a plausible mechanism bywhich the observed canalicular attenuation/dilation in medakamay manifest. We have to date not fully explored ANIT associ-ated cytoskeletal changes in medaka, though future studies aimto incorporate assessment of cytoskeletal integrity in medakahepatocytes and biliary epithelia pre and post ANIT exposure.

While canalicular dilation/attenuation was distinct, there didnot appear to be any loss of canalicular (bile) transport ofthe fluorophores Bodipy FL C5 Ceramide, �-Bodipy C5-HPCor fluorescein isothiocyanate associated with this phenotypicresponse. The only qualitative and quantitative changes inbile transport observed were, perhaps not surprisingly, a mod-est increase in fluorescence in dilated canaliculi (a proxy forincreased bile volume), and decrease in fluorescence in attenu-ated canaliculi (a proxy for decreased bile volume). While theselocal changes were apparent, overall transport of fluorophoresfrom IHBPs to extrahepatic bile ducts and gall bladder did notappear qualitatively or quantitatively different than that observedin control fish. Hence, in vivo investigations into hepatobiliarytransport suggest the intrahepatic biliary system, due to its inter-connected network of canaliculi and bile preductules, was ableto maintain overall bile transport from intrahepatic to extrahep-atic bile passageways at the time canalicular attenuation/dilationwas observed.

4.2. Bile preductular epithelial cell toxicity

The cause of bile preductular (BPD) lesions is perhapsmore clear. In vivo and ex vivo investigations suggest theobserved BPD lesions are likely attributable to BPDECswelling/cytotoxicity, as opposed to hepatocellular injury. BPDlesions (as opposed to attenuated/dilated canaliculi) werecommonly associated with changes in BPDEC morphology,observed in vivo, and in TEM investigations (Figs. 4 and 5Figs. 4C, F and 5D). 3D reconstructions also help affirm thehypothesis that BDPEC cytotoxicity contributed to altered bilepreductule morphology (Fig. 4D and E), and may in part beresponsible for observed alterations to bile flow/transport (dis-cussion follows). Hence, in vivo and ex vivo findings togethersuggest BPDEC toxicity to be a likely candidate for the BPDlesions observed.

Relevant to BPDEC cytotoxicity discussed here are priorstudies investigating carcinogenesis in medaka (Okihiro andHinton, 1999), which revealed that larval medaka exposed todiethyl nitrosamine (DEN) exhibited a higher prevalence of bil-iary tumors (46.4% of all tumors in larval-exposed medaka werebiliary versus 8.1% in adult-exposed fish), as opposed to hepa-tocellular carcinomas, which were the prevalent neoplasm whenmedaka were exposed as adults (100% of hepatocellular tumors

Page 14: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 33

in adult-exposed medaka were malignant, while only 51.5% oflarval hepatocellular tumors were malignant). These findings,describing a differential response to DEN, depending on thelife stage at which exposure occurred, may reflect differences incell populations during development (e.g. presence and preva-lence of BPDECs), and perhaps the role of these cell types ongrowth and differentiation of tumors in embryonic and adultlivers. Given: (1) the findings discussed here, (2) that previ-ous findings show the livers of early life stage medaka, at leastup to 40 days post fertilization, to be replete with BPDECs(Hardman et al., 2007b; Okihiro and Hinton, 2000), and (3)that these cell types are putative bipotent/pluripotent stem-likecells, akin to mammalian oval cells (Farber, 1956; Fausto andCampbell, 2003; Golding et al., 1996; Okihiro and Hinton,1999; Theise et al., 1999); it is possible that BPDEC toxicitymay be responsible for the age dependent differences in thetypes of liver tumors observed by Okihiro and Hinton (1999)(e.g. stem cell hypothesis of carcinogenesis). Although anatom-ical variations between mammalian and medaka liver exist (e.g.while BPDECs are located throughout the hepatic parenchymaof medaka, mammalian oval cells are localized to the peri-portalcanals of Hering) (Hardman et al., 2007b; Hinton et al., 2007),response of BPDECs to ANIT is intriguing in the presence ofour current understanding of ANIT induced changes in progen-itor cells of the mammalian liver (Alpini et al., 1992; Faa et al.,1998; Roskams et al., 1998, 2003). It follows that BPDECs mayplay important roles in further elucidating comparative struc-ture/function relationships, and toxic response, in piscine andmammalian livers.

4.3. Hydropic vacuolation

Hydropic vacuolation (HV), the result of the intracellu-lar accumulation of water and symptomatic of cell swelling,has been employed as a biomarker of exposure to assess theresponse of wild fishes to environmental contaminants, particu-larly polynuclear aromatic hydrocarbons, halogenated aromatichydrocarbons, and pesticides (Bodammer and Murchelano,1990; Gardner and Pruell, 1988; Moore et al., 1996, 2005;Murchelano and Wolke, 1985; Myers et al., 1998a,b). By exam-ple, winter flounder (Pleuronectes americanus) were annuallysurveyed for trends in hepatotoxicity and chemical body bur-den to evaluate water quality and effluent treatment efficacyin Boston Harbor, with HV employed as a key indicator ofhepatic injury (Moore et al., 2005). HV as a biomarker ofenvironmental stress has also been employed on the U.S. Westcoast to monitor contaminated coastal environments (Bodammerand Murchelano, 1990; Gardner and Pruell, 1988; Moore etal., 1996, 2005), and was most commonly observed in biliaryepithelial cells and hepatocytes in starry flounder (Platichthysstellatus), white croaker (Genyonemus lineatus) and rock sole(Lepidopsetta bilineata).Of interest, Nayak et al. (1996) sug-gest HV may reflect an adaptive, as opposed to toxic, cellularresponse. Observations in animal and human livers suggestvacuolated hepatocytes observed during liver injury are cellsadaptively altered to resist further insult, as opposed to cellsundergoing hydropic degeneration. By example, morphologi-

cal and biochemical investigations have shown that cytoplasmicvacuolation of hepatocytes following low doses of CCL4 wasdue to excess accumulation of glycogen, predominantly of themonoparticulate form. Low dose CCL4 exposed cells lackedfeatures of degeneration or regeneration, and were much less sus-ceptible to injury by larger subsequent CCl4 doses, as assessedby structural and serum enzyme analyses (Nayak et al., 1996).

The occurrence of HV in medaka was found to be con-centration dependent, with increasing prevalence up to 6 �MANIT (LC50 = 5 �M), beyond which HV prevalence declined.While electron dense particulates, which may represent glyco-gen deposits, where observed sporadically in HVs, we did notattempt to further characterize this response, and cannot elabo-rate on whether HV was an adaptive versus toxic response. Thein vivo findings presented do however yield a clearer under-standing of the morphology of this response at the cellular andsystem level of organization.

4.4. Biliary epithelial cell proliferation and biliary treearborization

While several of the structural and functional changesobserved in ANIT treated medaka are consistent with acholestatic response, relatively well described in rodents andhumans, such as a mosaic of either dilated or collapsed canali-culi and BEC toxicity (Anwer, 2004; Muller and Jansen, 1998;Trauner et al., 2005), arborization of the biliary tree (hyperpla-sia of biliary epithelia of bile ductules and ducts), a commonresponse observed during chronic cholestasis in the mammalianliver (Alpini et al., 1989; Lesage et al., 2001; Masyuk et al.,2003), was not observed in STII medaka in response to ANIT.While arborization appeared to be absent, examination of theliver hilus revealed BEC proliferation, suggesting a biliary tree“arborization-like” response in medaka liver, as compared withits mammalian counterparts. Lack of biliary tree arborization (ascompared to rodent) may be due to at least two important fac-tors: (1) biliary tree arborization described in mammalian liversis a function of proliferation of biliary epithelia of bile ductulesand ducts, which are largely localized to portal tracts, and (2)because larger bile ducts in medaka are found predominantly inthe hilar/peri-hilar region of liver, arborization of the biliary treein medaka would be localized primarily to liver hilus. These twopoints are discussed in the following.

First, in vivo and ex vivo (histological, immunohisto-chemical, TEM) analyses presented show ANIT inducesspecific changes in the medaka hepatobiliary system involv-ing hepatocytes (vacuolation, cytomegaly), biliary epithelialcells (hyperplasia, vacuolation, cytomegaly) and BPDECs(cytomegaly, vacuolation). AE1/AE3 staining suggests responseBECs of the intrahepatic biliary passageways, and PCNA analy-ses suggest BEC proliferation in the hilar and peri-hilar region ofthe liver, in response to chronic ANIT exposure (Figs. 4–6 Figs.4C, F, 5D, E and 6). These findings, particularly BEC hyperpla-sia in the hilar/peri-hilar region of the liver, are consistent withprior observations correlating medaka and mammalian hepato-biliary structure/function relationships (Hardman et al., 2007b),and suggest the hepatobiliary system of medaka responds to

Page 15: Non invasive high resolution

Author's personal copy

34 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

ANIT in a manner consistent with that observed in the mam-malian liver. Previously investigations suggested that the liver ofmedaka, in total, can be considered the structural and functionalanalogue of an individual mammalian lobule, with both livers(medaka and mammalian) sharing a common functional unit.However, in medaka, “arborization” of the biliary tree wouldbe localized to the liver hilus, the sole region of medaka liverdistinctly akin to the mammalian portal tract. Because the mam-malian liver is comprised of numerous lobules, with portal tractscontaining bile ductules and small ducts at the lobule periphery(which, structurally, yields a biliary tree), arborization occursthroughout the mammalian liver. In contrast, bile ductules andducts of the medaka liver are largely found near or within theliver hilus (single lobule hypothesis). Hence, while arborizationof the biliary tree occurs in medaka and mammals, this response,as suggested earlier (Hardman et al., 2007b), and reconfirmedhererin, will be different in pattern, site and amount.

Importantly, because of the comparative anatomical arrange-ment of mammalian and medaka biliary systems, there will bedifferences as to the interpretation of responses of these liversto insult, though the response may, fundamentally, be similar.Lack of distinct “biliary tree arborization” throughout the livercorpus of medaka, and localization of BEC proliferation to thehilar region of the liver, is illustrative of this concept and animportant comparative finding given arborization of portal tracts(BECs) in rodents is a hallmark response to reduced bile flowand BEC injury. By example, studies by Masyuk et al. (2003)found total biliary tree volume (one measure of arborization) inANIT treated rats to increase 18 times above controls. In con-junction, hepatic artery volume and portal vein volume increased4 times and 3 times that of control animals, respectively, whilebile duct diameter between ANIT treated and control rodentsremained unchanged. Note: Arborization of the biliary tree hasalso recently been described in three dimensions in the humanliver (Ludwig et al., 1998).

It follows from the above discussion, and previous findings(Hardman et al., 2007b), that arborization of the biliary “tree” inmedaka, in response to proliferative agents, would likely be lesspronounced than that observed in the mammalian liver, givenmedaka can be considered to possess a single portal tract (theliver hilus), as opposed to mammals, which possess myriad por-tal tracts throughout the liver corpus. It should be noted thefindings presented here were be no means all inclusive in termsof investigation of BEC proliferation in response to ANIT, andit is interesting to consider that, upon chronic ANIT exposureextending for months, one may see BEC proliferation infiltratemore distal regions of the liver (relative to hilus), with an increasein intrahepatic ductules and ducts.

4.5. Hepatobiliary transport

No impairment of blood to bile transport of the fluorophoresFITC, Bodipy HPC and Bodipy C5 Ceramide was observed atANIT concentrations <6 �M. The only observed impairment ofhepatobiliary transport function in vivo was at aqueous ANITconcentrations >6 �M, which approached the LC100 (10 �M,48 h), which cannot be necessarily attributed to altered hepa-

tocellular transport mechanisms (e.g. ATP binding cassette oftransmembrane transporters). At 8 �M ANIT STII medaka werenon-motile by 6 h of exposure and exhibited passive hepatic con-gestion in tandem with decreased heart rate (determined by rateof ventricular contraction). Reduced heart rate was likely asso-ciated with reduced cardiac output. Given the apparent systemictoxicity at 6 �M ANIT and above, the observed decrease influorophore transport could have resulted from: (1) decreasedrespiratory (branchial) uptake of the fluorophores (which invivo observations suggest is via gill uptake mechanisms), (2)a decrease in intrahepatic circulation (reduced heart rate, sinu-soidal flow rates, reduced fluorophore availability over time)and/or (3) from general systemic toxicity not characterized inthese studies. Because impaired transport function was asso-ciated with cardiovascular changes and overall morbidity, itis likely that the loss of transport function observed (in vivo)at ANIT concentrations >6 �M was symptomatic of generalsystemic toxicity, rather than ANIT mediated disruption of hepa-tocellular and biliary epithelial cell transmembrane transporters,which play key roles in bile transport and cholestasis.

In contrast to in vivo transport studies with fluorescent probes,volumetric analyses of 3D reconstructions, while performed inonly two 2 ANIT treated medaka and three control livers, sug-gest modest cholestatic and choleretic responses. The findingare noteworthy given; (1) 3D analyses from 3 control livers at8, 12, and 30 dpf yielded average intrahepatic biliary (canalic-ular, bile preductular) volumes of 0.97% (±0.09) relative tothe volume of liver examined, and (2) in hepatobiliary met-rics were consistent across 2D and 3D in vivo analyses, andex vivo evaluations, revealing accuracy of in vivo and ex vivoquantitative assessments. Where the volumes of sinusoidal, hep-atocellular, and parenchymal space, relative to total liver volumeexamined, remained unchanged in ANIT treated medaka, bilecanalicular volume was found to be diminished (cholestasis?) atlow ANIT concentrations, and increased (choleresis?) at higherconcentrations (Table 2). Assuming 3D reconstructions fromin vivo imaging accurately represent physiological change, theresults are of interest given prior studies describing an adaptivecholeretic response reported at non-toxic doses of ANIT (Alpiniet al., 1999), and altered intrahepatic biliary volume (Masyuket al., 2003), in rodent livers. While statistical analyses are notpossible, the changes in biliary volume suggested in volumetricanalyses are perhaps representative of real physiological changein response to ANIT, and may illustrate the ability to performvolumetric analyses in vivo in STII medaka.

What can explain the discrepancy between volumetric andin vivo fluorophore transport studies? Foremost, bile trans-port in mammals is a multi-phasic process (bi-directionalapical and basal membrane transport) governed in part by asuite of transmembrane transporters such as; the basolateralNa+/taurocholate cotransporter (NTCP), organic anion trans-porting proteins (OATPs), apical bile salt export pump (BSEP),and the multidrug resistant family of transporters (MDR/MRP)(see reviews by (Boyer, 1996a,b; Trauner and Boyer, 2003;Trauner et al., 2000)). Because fluorophore physico-chemicalproperties determine transporter substrate specificity, it is pos-sible that the fluorophores employed were not substrates for

Page 16: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 35

affected transporters (if affected by ANIT exposure). By exam-ple, FITC, an organic anion, is a potential candidate for OATPtransport on the basal membrane, and MRP2 on the apicalmembrane, neither of which may be affected by ANIT (noinformation on the effects of ANIT on transporter proteins infish exists). Hence, while no loss of fluorophore transport wasobserved in vivo, it is important to recognize that: (1) being thefirst in vivo transport studies of their kind, these studies wereof a screening nature, and (2) we did not attempt to elucidatewhich transporters the employed fluorophores were substratesfor, nor did we evaluate other fluorophores that may have provedbetter substrates for the variety of transporters present (as under-stood in the mammalian liver). Given these factors discrepanciesbetween volumetric and in vivo fluorophore transport studies arenot altogether surprising, and more refined study designs willbe required to elucidate the mechanisms of altered fluorophoretransport in vivo.

4.6. Cholestasis

While we have been reluctant to use the term cholesta-sis here, we feel discussion of this response important, givenits lack of description in piscine species. Cholestasis, a hall-mark response of the mammalian liver to injury and insult,is a complex response that involves not only the hepatobil-iary system but many other organ systems (e.g. gastrointestinaltract, kidney, cardiovascular system, endocrine system) as well.Because a variety of pharmaceuticals and environmental con-taminants have been shown to alter bile transport in mammals(Chang and Schiano, 2007; Mohi-ud-din and Lewis, 2004;Sakurai et al., 2007), in vivo models and methodologies, asdescribed here, may prove valuable for investigation of alteredbile transport in piscine species. Evaluation of this response inthe piscine liver is important given the relevance of this typeof hepatic injury/adaptation to ecotoxicological considerations,for instance; the effects of antibacterial agents, pesticides, andhormones employed in aquaculture, and other environmentalcontaminants, on fish reproduction, fitness and population health(Alderman and Hastings, 1998; Cabello, 2006; Goldburg andNaylor, 2005; Rhodes et al., 2000). In short, understanding acholestatic type response in fish is imperative to more fullyelucidating the effects of environmental contaminants and pro-phylactic substances on hepatobiliary metabolic and transportfunction, and health of the individual.

5. Conclusions

While the responses described are largely morphological innature, these findings reveal the intrahepatic biliary system ofmedaka to be targeted by the reference hepatotoxicant, ANIT.The cytological changes (e.g. BEC hyperplasia, cytomegaly,hydropic vacuolation, attenuated/dilated canaliculi), and puta-tive changes in bile volume and transport in ANIT exposedmedaka, are consistent with those observed in rodent ANIT stud-ies. Biliary tree “arborization”, or rather, the interpretation of thisresponse, differs, since the vast majority of the liver corpus ofmedaka is comprised of a canaliculo-bile preductular network,

while BECs, and their associated bile ductules and ducts, arelargely localized to the liver hilus.

These findings, which describe similarities and differencesbetween mammalian and medaka hepatobiliary systems inresponse to a reference hepatotoxicant (ANIT), in conjunctionwith prior in vivo work characterizing normalcy (Hampton etal., 1988, 1989; Hardman et al., 2007b; Hinton et al., 2004,2007; and others), illustrate the importance of our compara-tive understanding of the vertebrate liver, and the significanceof this understanding on the interpretation and communicationof xenobiotic induced injury in piscine livers. From these andprevious findings it is apparent that appreciating the spectrumof responses of the piscine liver to xenobiotics that target thisorgan system, particularly in a comparative sense, requires moreattention to bile preductular epithelial cells, bile preductules,and their relationship to the interconnected intrahepatic biliarynetwork. This is becoming increasingly important given thattoxicity screening in embryos and eleutheroembryos is a keyfactor in the regulatory evaluation of chemicals of environmentalconcern (e.g. REACh protocol; regulatory framework for Regis-tration, Evaluation, Authorization and Restriction of Chemicals)(ECHA, 2007), and that the liver is a key target organ of toxicity.

The findings presented have also shown for the first timein vivo evaluation of toxicity in the STII medaka hepatobiliarysystem, and demonstrate the ability to study and image, with highresolution, normalcy and toxicity in living medaka; a valuablediagnostic and investigatory tool. Given the described couplingof in vivo and ex vivo investigations, this suggests the futureability to integrate molecular mechanisms of disease and toxicitywith system level phenotypes, a current research aim in thislaboratory.

Acknowledgments

Thanks to Dr. David Miller, Laboratory of Pharmacologyand Chemistry, National Institute of Environmental Health Sci-ences, Research Triangle Park, for providing access to their laserscanning confocal microscopy facility, and to the Duke Uni-versity Integrated Toxicology Program. This publication wasmade possible by Grant Number 1 RO1 RR018583-02 from theNational Center for Research Resources (NCRR), a componentof the National Institutes of Health (NIH), and Grant NumberR21CA106084-01A1 from the National Cancer Institute (NCI),also a component of NIH. Its contents are solely the responsi-bility of the authors and do not necessarily represent the officialviews of NCRR or NIH.

References

Alderman, D.J., Hastings, T.S., 1998. Antibiotic use in aquaculture: developmentof antibiotic resistance – potential for consumer health risks. Int. J. Food Sci.Technol. 33 (2), 139–155.

Al-Gubory, K.H., 2005. Fibered confocal fluorescence microscopy for imagingapoptotic DNA fragmentation at the single-cell level in vivo. Exp. Cell. Res.310 (2), 474–481.

Alpini, G., Lenzi, R., Zhai, W.R., Slott, P.A., Liu, M.H., Sarkozi, L., Tavoloni,N., 1989. Bile secretory function of intrahepatic biliary epithelium in the rat.Am. J. Physiol. 257 (1 Pt 1), G124–G133.

Page 17: Non invasive high resolution

Author's personal copy

36 R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37

Alpini, G., Aragona, E., Dabeva, M., Salvi, R., Shafritz, D.A., Tavoloni, N.,1992. Distribution of albumin and alpha-fetoprotein mRNAs in normal,hyperplastic, and preneoplastic rat liver. Am. J. Pathol. 141 (3), 623–632.

Alpini, G., Glaser, S.S., Ueno, Y., Rodgers, R., Phinizy, J.L., Francis, H., Baioc-chi, L., Holcomb, L.A., Caligiuri, A., LeSage, G.D., 1999. Bile acid feedinginduces cholangiocyte proliferation and secretion: evidence for bile acid-regulated ductal secretion. Gastroenterology 116 (1), 179–186.

Alpini, G., Ueno, Y., Glaser, S.S., Marzioni, M., Phinizy, J.L., Francis, H.,Lesage, G., 2001. Bile acid feeding increased proliferative activity and apicalbile acid transporter expression in both small and large rat cholangiocytes.Hepatology 34 (5), 868–876.

Alpini, G., Glaser, S., Alvaro, D., Ueno, Y., Marzioni, M., Francis, H., Baioc-chi, L., Stati, T., Barbaro, B., Phinizy, J.L., 2002a. Bile acid depletion andrepletion regulate cholangiocyte growth and secretion by a phosphatidyli-nositol 3-kinase-dependent pathway in rats. Gastroenterology 123 (4),1226–1237.

Alpini, G., McGill, J.M., Larusso, N.F., 2002b. The pathobiology of biliaryepithelia. Hepatology 35 (5), 1256–1268.

Anwer, M.S., 2004. Cellular regulation of hepatic bile acid transport in healthand cholestasis. Hepatology 39 (3), 581–590.

Arias, I.M., 1988. In: Irwin, M., Arias, W.B.J., Hans, P., David, S., David, A.S.(Eds.), The Liver: Biology and Pathobiology. Raven Press, New York.

Arrese, M., Ananthananarayanan, M., Suchy, F.J., 1998. Hepatobiliary transport:molecular mechanisms of development and cholestasis. Pediatr. Res. 44 (2),141–147.

Ballatori, N., Hager, D.N., Nundy, S., Miller, D.S., Boyer, J.L., 1999. Carrier-mediated uptake of lucifer yellow in skate and rat hepatocytes: a fluid-phasemarker revisited. Am. J. Physiol. 277 (4 Pt 1), G896–G904.

Ballatori, N., Rebbeor, J.F., Connolly, G.C., Seward, D.J., Lenth, B.E., Henson,J.H., Sundaram, P., Boyer, J.L., 2000. Bile salt excretion in skate liver ismediated by a functional analog of Bsep/Spgp, the bile salt export pump.Am. J. Physiol. Gastrointest. Liver Physiol. 278 (1), G57–G63.

Bodammer, J.E., Murchelano, R.A., 1990. Cytological study of vacuolated cellsand other aberrant hepatocytes in winter flounder from Boston Harbor. Can-cer Res. 50 (20), 6744–6756.

Bove, K.E., Daugherty, C.C., Tyson, W., Mierau, G., Heubi, J.E., Balistreri, W.F.,Setchell, K.D., 2000. Bile acid synthetic defects and liver disease. Pediatr.Dev. Pathol. 3 (1), 1–16.

Boyer, J.L., 1996a. Bile duct epithelium: frontiers in transport physiology. Am.J. Physiol. 270 (1 Pt 1), G1–G5.

Boyer, J.L., 1996b. Bile secretion—models, mechanisms, and malfunctions. Aperspective on the development of modern cellular and molecular conceptsof bile secretion and cholestasis. J. Gastroenterol. 31 (3), 475–481.

Boyer, J.L., Schwarz, J., Smith, N., 1976a. Biliary secretion in elasmobranches.I. Bile collection and composition. Am. J. Physiol. 230 (4), 970–973.

Boyer, J.L., Schwarz, J., Smith, N., 1976b. Selective hepatic uptake and biliaryexcretion of 35S-sulfobromophthalein in marine elasmobranchs. Gastroen-terology 70 (2), 254–256.

Brown-Peterson, N.J., Krol, R.M., Zhu, Y., Hawkins, W.E., 1999. N-nitrosodiethylamine initiation of carcinogenesis in Japanese medaka(Oryzias latipes): hepatocellular proliferation, toxicity, and neoplasticlesions resulting from short term, low level exposure. Toxicol. Sci. 50 (2),186–194.

Cabello, F.C., 2006. Heavy use of prophylactic antibiotics in aquaculture: agrowing problem for human and animal health and for the environment.Environ. Microbiol. 8 (7), 1137–1144.

Carpenter-Deyo, L., Marchand, D.H., Jean, P.A., Roth, R.A., Reed, D.J.,1991. Involvement of glutathione in 1-naphthylisothiocyanate (ANIT)metabolism and toxicity to isolated hepatocytes. Biochem. Pharmacol. 42(11), 2171–2180.

Chang, C.Y., Schiano, T.D., 2007. Review article: drug hepatotoxicity. AlimentPharmacol. Ther. 25 (10), 1135–1151.

Chignard, N., Mergey, M., Veissiere, D., Parc, R., Capeau, J., Poupon, R., Paul,A., Housset, C., 2001. Bile acid transport and regulating functions in thehuman biliary epithelium. Hepatology 33 (3), 496–503.

Connolly, A., Price, S., Connelly, J., Hinton, R., 1988. Early changesin bile duct lining cells and hepatocytes in rats treated with alpha-naphthylisothiocyanate. Toxicol. Appl. Pharmacol. 93 (2), 208–219.

ECHA, European Chemicals Agency, June 2007. Internet Website:http://ec.europa.eu/echa/reach en.html.

Faa, G., Van Eyken, P., Roskams, T., Miyazaki, H., Serreli, S., Ambu, R., Desmet,V.J., 1998. Expression of cytokeratin 20 in developing rat liver and in exper-imental models of ductular and oval cell proliferation. J. Hepatol. 29 (4),628.

Farber, E., 1956. Similarities in the sequence of early histological changesinduced in the liver of the rat by ethionine, 2-acetylamino-fluorene, and3′-methyl-4-dimethylaminoazobenzene. Cancer Res. 16 (2), 142–148.

Fausto, N., 2000. Liver regeneration. J. Hepatol. 32 (1 Suppl.), 19–31.Fausto, N., Campbell, J.S., 2003. The role of hepatocytes and oval cells in liver

regeneration and repopulation. Mech. Dev. 120 (1), 117–130.Furuta, T., Takeuchi, H., Isozaki, M., Takahashi, Y., Kanehara, M., Sugi-

moto, M., Watanabe, T., Noguchi, K., Dore, T.M., Kurahashi, T., 2004.Bhc-cNMPs as either water-soluble or membrane-permeant photoreleasablecyclic nucleotides for both one- and two-photon excitation. Chembiochem5 (8), 1119–1128, and others.

Gardner, G.R., Pruell, R.J., 1988. A histopathological and chemical assess-ment of winter flounder, lobster and soft-shelled clam indigenous to QuincyBay Boston Harbor and an in-situ evaluation of oysters including sedi-ment (surface and cores) chemistry. U.S. Environmental Protection Agency,Environmental Research Laboratory, Narragansett, RI.

Goldburg, R., Naylor, R., 2005. Future seascapes, fishing, and fish farming.Front. Ecol. Environ. 3 (1), 21–28.

Golding, M., Sarraf, C., Lalani, E.N., Alison, M.R., 1996. Reactive biliaryepithelium: the product of a pluripotential stem cell compartment? Hum.Pathol. 27 (9), 872–884.

Groothuis, G.M., Meijer, D.K., 1996. Drug traffic in the hepatobiliary system.J. Hepatol. 24 (Suppl. 1), 3–28.

Hampton, J., Lantz, R., Goldblatt, P., Lauren, D., Hinton, D., 1988. Functionalunits in rainbow trout (Salmo gairdneri, Richardson) liver. II. The biliarysystem. Anat. Rec. 221 (2), 619–634.

Hampton, J.A., Lantz, R.C., Hinton, D.E., 1989. Functional units in rainbowtrout (Salmo gairdneri, Richardson) liver. III. Morphometric analysis ofparenchyma, stroma, and component cell types. Am. J. Anat. 185 (1), 58–73.

Hardman, R., Kullman, S., Hinton, D., 2007a. Non invasive in vivo investigationof hepatobiliary structure and function in STII medaka (Oryzias latipes):methodology and applications. Compar. Hepatol., in press.

Hardman, R., Volz, D., Kullman, S., Hinton, D.E., 2007b. An in vivo look atvertebrate liver architecture: 3-dimensional reconstructions from medaka(Oryzias latipes). Anat. Rec. 290 (7), 770–782.

Hill, D.A., Roth, R.A., 1998. Alpha-naphthylisothiocyanate causes neutrophilsto release factors that are cytotoxic to hepatocytes. Toxicol. Appl. Pharmacol.148 (1), 169–175.

Hill, D.A., Jean, P.A., Roth, R.A., 1999. Bile duct epithelial cells exposedto alpha-naphthylisothiocyanate produce a factor that causes neutrophil-dependent hepatocellular injury in vitro. Toxicol. Sci. 47 (1), 118–125.

Hinton, D., Lantz, R., Hampton, J., McCuskey, P., McCuskey, R., 1987. Nor-mal versus abnormal structure: considerations in morphologic responses ofteleosts to pollutants. Review. Environ. Health Perspect. 71, 139–146.

Hinton, D., Segenr, H., Braunbeck, T., 2001. Toxic responses of the liver. In:Schlenk, D., Benson, W. (Eds.), New Perspectives: Toxicology and the Envi-ronment Target Organ Toxicity in Marine and Freshwater Teleosts. Taylorand Francis, New York, pp. 225–268.

Hinton, D.E., Wakamatsu, Y., Ozato, K., Kashiwada, S., 2004. Imaging liverdevelopment/remodeling in the see-through medaka fish. Comp. Hepatol. 3(Suppl. 1), S30.

Hinton, D., Segner, H., Braunbeck, T., Kullman, S., Hardman, R., 2007. Chapter7: Liver Toxicity. In: DiGiulo, R., Hinton, D.E. (Eds.), The Toxicology ofFishes: Unit 2 Key Target Systems. Taylor and Francis, Boca Raton, FL.

Inudo, M., Ishibashi, H., Matsumura, N., Matsuoka, M., Mori, T., Taniyama,S., Kadokami, K., Koga, M., Shinohara, R., Hutchinson, T.H., 2004. Effectof estrogenic activity, and phytoestrogen and organochlorine pesticide con-tents in an experimental fish diet on reproduction and hepatic vitellogeninproduction in medaka (Oryzias latipes). Comp. Med. 54 (6), 673–680, andothers.

Kan, K.S., Coleman, R., 1986. 1-Naphthylisothiocyanate-induced permeabilityof hepatic tight junctions to proteins. Biochem. J. 238 (2), 323–328.

Page 18: Non invasive high resolution

Author's personal copy

R. Hardman et al. / Aquatic Toxicology 86 (2008) 20–37 37

Kossor, D.C., Handler, J.A., Dulik, D.M., Meunier, P.C., Leonard, T.B., Gold-stein, R.S., 1993. Cholestatic potentials of alpha-naphthylisothiocyanate(ANIT) and beta-naphthylisothiocyanate (BNIT) in the isolated perfusedrat liver. Biochem. Pharmacol. 46 (11), 2061–2066.

Kossor, D.C., Goldstein, R.S., Ngo, W., DeNicola, D.B., Leonard, T.B., Dulik,D.M., Meunier, P.C., 1995. Biliary epithelial cell proliferation followingalpha-naphthylisothiocyanate (ANIT) treatment: relationship to bile ductobstruction. Fundam. Appl. Toxicol. 26 (1), 51–62.

Leonard, T.B., Popp, J.A., Graichen, M.E., Dent, J.G., 1981. alpha-Naphthylisothiocyanate induced alterations in hepatic drug metabolizingenzymes and liver morphology: implications concerning anticarcinogenesis.Carcinogenesis 2 (6), 473–482.

Lesage, G., Glaser, S., Ueno, Y., Alvaro, D., Baiocchi, L., Kanno, N., Phinizy,J.L., Francis, H., Alpini, G., 2001. Regression of cholangiocyte proliferationafter cessation of ANIT feeding is coupled with increased apoptosis. Am. J.Physiol. Gastrointest. Liver Physiol. 281 (1), G182–G190.

Lowe, P.J., Kan, K.S., Barnwell, S.G., Sharma, R.K., Coleman, R., 1985.Transcytosis and paracellular movements of horseradish peroxidaseacross liver parenchymal tissue from blood to bile: effects of alpha-naphthylisothiocyanate and colchicines. Biochem. J. 229 (2), 529–537.

Ludwig, J., Ritman, E.L., LaRusso, N.F., Sheedy, P.F., Zumpe, G., 1998.Anatomy of the human biliary system studied by quantitative computer-aidedthree-dimensional imaging techniques. Hepatology 27 (4), 893–899.

Masyuk, T.V., Ritman, E.L., LaRusso, N.F., 2003. Hepatic artery and portalvein remodeling in rat liver: vascular response to selective cholangiocyteproliferation. Am. J. Pathol. 162 (4), 1175–1182.

McLean, M.R., Rees, K.R., 1958. Hyperplasia of bile ducts induced by alpha-naphthylisothiocyanate: experimental biliary cirrhosis free from biliaryobstruction. Vet. Immunol. Immunopathol. 76, 175–188.

Mohi-ud-din, R., Lewis, J.H., 2004. Drug- and chemical-induced cholestasis.Clin. Liver Dis. 8 (1), 95–132, vii.

Moore, M.J., Shea, D., Hillman, R.E., Stegeman, J.J., 1996. Trends in hepatictumours and hydropic vacuolation, fin erosion, organic chemicals and stableisotope ratios in winter flounder from Massachusetts, USA. Mar. Pollut.Bull. 32 (6), 458.

Moore, M., Lefkovitz, L., Hall, M., Hillman, R., Mitchell, D., Burnett, J., 2005.Reduction in organic contaminant exposure and resultant hepatic hydropicvacuolation in winter flounder (Pseudopleuronectes americanus) followingimproved effluent quality and relocation of the Boston sewage outfall intoMassachusetts Bay, USA: 1987–2003. Mar. Pollut. Bull. 50 (2), 156.

Motta, P.G.F., 1975. Structure of rat bile canaliculi as revealed by scanningelectron microscopy. Anat. Rec. 182, 499–513.

Muller, M., Jansen, P.L., 1998. The secretory function of the liver: new aspectsof hepatobiliary transport. J. Hepatol. 28 (2), 344–354.

Murchelano, R.A., Wolke, R.E., 1985. Epizootic Carcinoma in the Winter Floun-der, Pseudopleuronectes-Americanus. Science 228 (4699), 587–589.

Myers, M.S., Johnson, L.L., Hom, T., Collier, T.K., Stein, J.E., Varanasi, U.,1998a. Toxicopathic hepatic lesions in subadult English sole (Pleuronectesvetulus) from Puget Sound, Washington USA: relationships with otherbiomarkers of contaminant exposure. Mar. Environ. Res. 45 (1), 47–67.

Myers, M.S., Johnson, L.L., Olson, O.P., Stehr, C.M., Horness, B.H., Collier,T.K., McCain, B.B., 1998b. Toxicopathic hepatic lesions as biomarkers ofchemical contaminant exposure and effects in marine bottomfish speciesfrom the Northeast and Pacific Coasts, USA. Mar. Pollut. Bull. 37 (1–2),92–113.

Nayak, N.C., Sathar, S.A., Mughal, S., Duttagupta, S., Mathur, M., Chopra,P., 1996. The nature and significance of liver cell vacuolation followinghepatocellular injury—an analysis based on observations on rats renderedtolerant to hepatotoxic damage. Virchows Arch. 428 (6), 353–365.

Okihiro, M.S., Hinton, D.E., 1999. Progression of hepatic neoplasia in medaka(Oryzias latipes) exposed to diethylnitrosamine. Carcinogenesis 20 (6),933–940.

Okihiro, M.S., Hinton, D.E., 2000. Partial hepatectomy and bile duct ligation inrainbow trout (Oncorhynchus mykiss): histologic, immunohistochemical andenzyme histochemical characterization of hepatic regeneration and biliaryhyperplasia. Toxicol. Pathol. 28 (2), 342–356.

Orsler, D.J., Ahmed-Choudhury, J., Chipman, J.K., Hammond, T., Coleman, R.,1999. ANIT-induced disruption of biliary function in rat hepatocyte couplets.Toxicol. Sci. 47 (2), 203–210.

Rhodes, G., Huys, G., Swings, J., McGann, P., Hiney, M., Smith, P., Pickup,R.W., 2000. Distribution of oxytetracycline resistance plasmids betweenaeromonads in hospital and aquaculture environments: implication ofTn1721 in dissemination of the tetracycline resistance determinant Tet A.Appl. Environ. Microbiol. 66 (9), 3883–3890.

Rocha, E., Monteiro, R.A., Pereira, C.A., 1997. Liver of the brown trout, Salmotrutta (Teleostei Salmonidae): a stereological study at light and electronmicroscopic levels. Anat. Rec. 247 (3), 317–328.

Rocha, E., Monteiro, R.A., Oliveira, M.H., Silva, M.W., 2001. The hepatocytesof the brown trout (Salmo trutta f. fario): a quantitative study using design-based stereology. Histol. Histopathol. 16 (2), 423–437.

Roskams, T., De Vos, R., Van Eyken, P., Myazaki, H., Van Damme, B., Desmet,V., 1998. Hepatic OV-6 expression in human liver disease and rat exper-iments: evidence for hepatic progenitor cells in man. J. Hepatol. 29 (3),455.

Roskams, T.A., Libbrecht, L., Desmet, V.J., 2003. Progenitor cells in diseasedhuman liver. Semin. Liver Dis. 23 (4), 385–396.

Sakurai, A., Kurata, A., Onishi, Y., Hirano, H., Ishikawa, T., 2007. Prediction ofdrug-induced intrahepatic cholestasis: in vitro screening and QSAR analysisof drugs inhibiting the human bile salt export pump. Expert Opin. Drug. Saf.6 (1), 71–86.

Santos, F., MacDonald, G., Rubel, E.W., Raible, D.W., 2006. Lateral line haircell maturation is a determinant of aminoglycoside susceptibility in zebrafish(Danio rerio). Hear Res. 213 (1–2), 25–33.

Steiner, J., Carruthers, J., 1961. Studies on the fine structure of the terminalbranches of the biliary tree. Am. J. Pathol. 38, 639–661.

Theise, N.D., Saxena, R., Portmann, B.C., Thung, S.N., Yee, H., Chiriboga, L.,Kumar, A., Crawford, J.M., 1999. The canals of Hering and hepatic stemcells in humans. Hepatology 30 (6), 1425–1433.

Thung, S.N., Gerber, M.A., 1992. The liver. In: Sternber, S. (Ed.), Histology forPathologists. Raven Press, New York, pp. 625–638.

Trauner, M., Boyer, J.L., 2003. Bile salt transporters: molecular characterization,function, and regulation. Physiol. Rev. 83 (2), 633–671.

Trauner, M., Meier, P.J., Boyer, J.L., 1998. Molecular pathogenesis of cholesta-sis. N. Engl. J. Med. 339 (17), 1217–1227.

Trauner, M., Fickert, P., Stauber, R.E., 2000. Hepatocellular bile salt trans-port: lessons from cholestasis. Can. J. Gastroenterol. 14 (Suppl. D),99D–104D.

Trauner, M., Wagner, M., Fickert, P., Zollner, G., 2005. Molecular regulationof hepatobiliary transport systems: clinical implications for understand-ing and treating cholestasis. J. Clin. Gastroenterol. 39 (4 Suppl. 2),S111–S124.

Wakamatsu, Y., Pristyazhnyuk, S., Kinoshita, M., Tanaka, M., Ozato, K., 2001.The see-through medaka: a fish model that is transparent throughout life.Proc. Natl. Acad. Sci. U.S.A. 98 (18), 10046–10050.

Waters, N.J., Holmes, E., Williams, A., Waterfield, C.J., Farrant, R.D., Nichol-son, J.K., 2001. NMR and pattern recognition studies on the time-relatedmetabolic effects of alpha-naphthylisothiocyanate on liver, urine, and plasmain the rat: an integrative metabonomic approach. Chem. Res. Toxicol. 14(10), 1401–1412.

Waters, N.J., Holmes, E., Waterfield, C.J., Farrant, R.D., Nicholson, J.K., 2002.NMR and pattern recognition studies on liver extracts and intact livers fromrats treated with alpha-naphthylisothiocyanate. Biochem. Pharmacol. 64 (1),67–77.

Wlodkowic, D., Skommer, J., Pelkonen, J., 2007. Towards an understanding ofapoptosis detection by SYTO dyes. Cytom. A 71 (2), 61–72.

Wolkoff, A.W., Cohen, D.E., 2003. Bile acid regulation of hepatic physiology.I. Hepatocyte transport of bile acids. Am. J. Physiol. Gastrointest. LiverPhysiol. 284 (2), G175–G179.

Woolley, J., Mullock, B., Hinton, R., 1979. Reflux of billiary componentsinto blood in experimental intrahepatic cholestasis induced in rats bytreatment with alpha-naphthylisothiocyanate. Clin. Chim. Acta 92 (3),381–386.