Você está na página 1de 77

Toxicologic Pathology, 38: 5S-81S, 2010

Copyright # 2010 by The Author(s)


ISSN: 0192-6233 print / 1533-1601 online
DOI: 10.1177/0192623310386499

Proliferative and Nonproliferative Lesions of the


Rat and Mouse Hepatobiliary System
BOB THOOLEN1, ROBERT R. MARONPOT2*, TAKANORI HARADA3, ABRAHAM NYSKA4, COLIN ROUSSEAUX5, THOMAS NOLTE6,
DAVID E. MALARKEY7, WOLFGANG KAUFMANN8, KARIN KÜTTLER9, ULRICH DESCHL10, DAI NAKAE11, RICHARD GREGSON12,
MICHAEL P. VINLOVE13, AMY E. BRIX14, BHANU SINGH15, FIORELLA BELPOGGI16, AND JERROLD M. WARD17
1
Global Pathology Support, The Hague, The Netherlands
2
Maronpot Consulting LLC, Raleigh, North Carolina, USA
3
The Institute of Environmental Toxicology, Joso-shi, Ibaraki, Japan
4
Haharuv 18, Timrat, Israel
5
Wakefield QC, Canada
6
Boehringer Ingelheim Pharma GmbH & Co., Biberach an der Riss, Germany
7
National Toxicology Program, Cellular and Molecular Pathology Branch, Research Triangle Park, North Carolina, USA
8
Merck KGaA, Darmstadt, Germany
9
BASF Aktiengesellschaft, Ludwigshafen, Germany
10
Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
11
Tokyo Metropolitan Institute of Public Health, Shinjuku, Tokyo, Japan
12
Charles River Laboratories, Pathology Department, Senneville, QC, Canada
13
Pathology Associates, Charles River, Frederick, Maryland, USA
14
Experimental Pathology Laboratories Inc., Research Triangle Park, North Carolina, USA
15
DuPont Haskell Global Centers for Health and Environmental Science, Newark, Delaware, USA
16
Ramazzini Institute, Bentivoglio (BO), Italy
17
Global VetPathology, Montgomery Village, Maryland, USA
*Chairman of the Liver INHAND Committee
ABSTRACT

The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of
the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP) and North America (STP) to develop an interna-
tionally-accepted nomenclature for proliferative and non-proliferative lesions in laboratory animals. The purpose of this publication is to provide a
standardized nomenclature and differential diagnosis for classifying microscopic lesions observed in the hepatobiliary system of laboratory rats and
mice, with color microphotographs illustrating examples of some lesions. The standardized nomenclature presented in this document is also available
for society members electronically on the internet (http://goreni.org). Sources of material included histopathology databases from government,
academia, and industrial laboratories throughout the world. Content includes spontaneous and aging lesions as well as lesions induced by exposure
to test materials. A widely accepted and utilized international harmonization of nomenclature for lesions of the hepatobiliary system in laboratory
animals will decrease confusion among regulatory and scientific research organizations in different countries and provide a common language to
increase and enrich international exchanges of information among toxicologists and pathologists.

Keywords: diagnostic pathology; hepatobiliary system; histopathology; liver; nomenclature; rodent pathology.

Address correspondence to: Bob Thoolen, Global Pathology Support, Benoordenhoutseweg 23, The Hague 2596 BA, Netherlands; e-mail: bob.thoolen@
gpstoxpath.com.
Financial Disclosure: No money was paid for the preparation of this manuscript. During the construction of this manuscript salaries of contributors were paid by
their respective companies. None of the content of the manuscript contains any information that could be patentable or claimed as intellectual property of the
contributors or their respective companies.
Abbreviations: AE1/AE3, Two clones of anti-cytokeratin monoclonal antibodies; a.k.a., Also known as; AS, Anterior Segment; a-SMA, a-smooth muscle actin;
Bcl-2, B-cel lymphoma 2 - apoptosis regulator protein; BSTP, British Society of Toxicological Pathologists; CD (31, 34, 68), Cluster differentiation (31, 34, 68);
CEA, Carcinoembryonic antigen; CK, Cytokeratin; ED1, Rat homologue of human CD68; EM, Electron microscopy; ESTP, European Society of Toxicologic
Pathology; Factor VIII, Blood clotting factor/anti-hemophilic factor; F4/80, Rat anti-mouse macrophage monoclonal antibody; H&E, Hematoxylin and Eosin;
IHC, Immunohistochemistry; JSTP, The Japanese Society of Toxicologic Pathology; Ki-67, Nuclear protein associated with proliferation; LAMP, Lysosome-
associated protein; LLL, Lef lateral lobe; LML, Left medial lobe; MIB-1, Monoclonal antibody that detects K-67 antigen on formalin fixed paraffin embedded
sections; MS, Middle Segment; NTP, National Toxicology Program; NLDC-145, Rat anti-mouse dendritic cell monoclonal antibody; NOS, Not otherwise
specified; OX-6, MHC Class II Ia antibody; PAS, Periodic acid-Schiff; PC, Caudate Process; PCNA, Proliferator Cell Nuclear Antigen; PCR, Polymerase
Chain Reaction; PP, Papillary Process; PPA, Processus papillaris anterior; PPAR, Peroxisome Proliferator-Activated Receptor; PS, Posterior Segment; RER,
Rough Endoplasmic Reticulum; RLL, Right lateral lobe; RML, Right medial lobe; SRA-E5, Mouse monoclonal anti-macrophage antibody for Scavenger
Receptor A; SOPs, Standard Operating Procedures; STP, Society of Toxicologic Pathology.

5S
6S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

TABLE 1.—Species differences in liver lobes.

Human (4 / 8)a Monkey (4 / 8) Dog (6 / 7) Rat (4 / 7) Mouse (4 / 7) Cat (6 / 7)


Left liver Left Lobe (2 segments) Left (lateral) Lobe Left Lobe Left Lobe Left Lobe Left Lobe LLL
AS þ PS LLL þ LML LLL þ LML LLL þ LML (largest) þ LML
Right liver Right Lobe (2 segments) Right (lateral) Lobe Right Lobe RLL Right Lobe Right Lobe Right Lobe
AS þ MS þ PS (impression) þ RML RLL þ RML RLL þ RML RLL þ RML
Intermediate liver Quadrate Lobe Median Lobe (largest) Quadrate Lobe Quadrate Lobe Quadrate Lobe
(small)
Caudate lobe Caudate Lobe Caudate Lobe Caudate Lobe Caudate Lobe Caudate Lobe Caudate Lobe
PC þ PP PP þ PC PP þ PPA þ PC PP þ PC PP þ PC

LLL ¼ Left lateral lobe; RLL ¼ Right lateral lobe; PC ¼ Caudate Process; LML ¼ Left medial lobe; RML ¼ Right medial lobe; PP ¼ Papillary Process; PPA ¼ Processus papillaris
anterior; AS ¼ Anterior Segment; MS ¼ Middle Segment; PS ¼ Posterior Segment.
Gray, Williams, and Bannister (1995); Browning, Schroeder, and Berringer (1974); König, Sautet, and Liebich (2004); Rajtová, Horák, and Popesko (2002); Vons et al., 2009.
a
(Number of lobes / Number of lobes including segments).

I. GENERAL INTRODUCTION frequently observed in pathological evaluation of toxicity


studies.
The liver is a major target organ in safety assessment of
preclinical toxicity and oncogenicity studies with rodents;
hence, hepatic pathology is central to many toxicological II. ANATOMY
pathology studies. As toxicologic pathologists sometimes The liver occupies the cranial third of the abdominal
experience difficulties in distinguishing the wide variety of cavity and is comprised of multiple lobes; however, the
liver lesions in the rodents for safety evaluation purposes, this nomenclature for the liver lobes varies among authors. There
document is a consensus of senior toxicologic pathologists are basically left, middle, right, and caudate lobes (Harada
regarding suggested nomenclature that should be used for et al. 1999; Eustis et al. 1990). A thin connective tissue cap-
specific lesions. sule that is externally lined by peritoneal mesothelial cells
Standardized diagnostic criteria and nomenclature are covers the parietal and visceral surfaces of the liver. The
essential to harmonize the classification and reporting of hepa- middle lobe has an incomplete fissure where the falciform
tic nonproliferative as well as proliferative lesions. This ligament attaches. In mice the gallbladder is located in the
INHAND document serves as a framework that can be used for middle lobe fissure, whereas the rat does not have a gallblad-
the harmonization of diagnostic criteria of hepatic lesions in der. The right lobe has an anterior and posterior component
laboratory rats and mice. These recommendations for diagnos- and the small caudate lobe consists of two or more disc-
tic criteria and preferred terminology should not be considered like sublobes (See Figure 1).
mandatory; proper diagnoses are ultimately based on the dis- Nomenclature for liver lobes varies among species and
cretion of the toxicologic study pathologist. sometimes among authors. A table showing differences in liver
The INHAND (International Harmonization of Nomenclature lobes between species is included based on current anatomic
and Diagnostic Criteria for Lesions in Rats and Mice) initiative features (Table 1).
creates a framework for the harmonization of diagnostic nomen-
clature (classification of lesions using the same terminology) in
III. HISTOMORPHOLOGY
different rodent organ systems. It is a joint initiative between
Societies from the United States (STP), Great Britain (BSTP), The two-dimensional microarchitecture of the liver has
Japan (JSTP), and European countries (ESTP). been categorized in at least three perspectives (Figure 2). The
This document is organized to provide introductory material anatomic model is the classical lobule, a hexagonal structure
that reviews comparative interspecies differences in anatomy divided into concentric centrilobular, midzonal, and peripor-
and liver function, followed by a listing of liver lesions in a tal segments. The triangular portal lobule is based on bile
standardized format. The liver lesions descriptions include dif- flow and is centered on the portal triad (portal canal). The
ferential diagnoses to aid in distinguishing primary diagnoses elliptical or diamond shaped liver acinus is a functional sub-
from similar appearing lesions. Throughout the document, unit of the liver. It incorporates blood flow and metabolic
comparisons are made with respect to similar liver lesions that functions and is divided in zone 1 (periportal), zone 2 (transi-
may occur in humans. It should be noted that the preferred diag- tional; midzonal), and zone 3 (centrilobular). Functionally,
nostic terminology for some lesions in this document might zone 1 hepatocytes are specialized for oxidative liver func-
represent departures from traditional nomenclature schemes tions such as gluconeogenesis, b-oxidation of fatty acids, and
found in standard textbooks. Furthermore, illustrative photomi- cholesterol synthesis, while zone 3 cells are more important
crographs for a given diagnostic entity may occasionally depict for glycolysis, lipogenesis, and cytochrome P-450–based
additional tissue changes as this reflects actual situations drug detoxification.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 7S

1. Blood Supply and Bile Flow lymphocytes that have natural killer activity and are primarily
located in periportal areas (Wright and Stacey 1991).
The liver has a dual blood supply, the hepatic portal vein and
the hepatic artery. The hepatic artery supplies oxygenated
3. Immunohistochemistry
blood. Approximately 75% of the blood is delivered to the liver
via the hepatic portal vein that drains the spleen, stomach, Immunohistochemistry (IHC), utilizing fluorescent or chro-
intestines, and pancreas. Branches of the hepatic artery and mogen tagged antibodies, is a useful adjunct for identification
portal vein are seen in the portal triads along with bile ducts and of different cell types in the liver. Selected examples are pro-
are separated from the hepatic cords by a ‘‘limiting plate’’ of vided in Table 2.
hepatocytes. The bile ducts join to form the hepatic duct lead- Use of IHC can be helpful for diagnostic purposes and is
ing to the small intestine in rats and to the gallbladder in mice. common in human pathology where panels of immunohisto-
Blood flows from the portal areas to the central vein in the cen- chemical stains are used for supporting diagnoses. Not all com-
ter of each lobule while bile flows from the center of the hepa- mercially available preparations of a given antibody will react
tic lobule to the portal areas and on to the hepatic duct. the same way between different laboratories and between
different species. Furthermore, expertise is required for
2. Histology tissue handling to unmask cellular antigens that may be
cross-linked during tissue fixation. Diagnostic evaluation of
The two most commonly used descriptions for the structural
immunostains typically requires inclusion of both positive and
and functional units of the liver are the hepatic lobule (Kiernan
negative controls. The interpretations of IHC results are usually
1883) and the acinus (Rappaport et al., 1954) (Figure 2). The
performed in conjunction with histopathological findings and
structural unit, the hepatic lobule, is modeled on the blood flow
sometimes also with consideration of gross findings and/or
within the liver and is commonly used for descriptive pathol-
clinical pathology or other relevant study results.
ogy and morphological diagnoses. The functional unit, the
hepatic acinus, is modeled on blood flow and metabolism
IV. PHYSIOLOGY
within the liver. More recently a parenchymal unit in the liver
has been described as a cone-shaped three-dimensional struc- The liver is responsible for maintenance of many homeo-
ture comprised of approximately fourteen hepatic lobules sup- static and physiological functions. Liver size is governed both
plied and drained by common vascular tributaries (Malarkey by genetic factors and by the rate of biochemical activity to
et al. 2005; Teutsch, Schuerfeld, and Groezinger 1999; Teutsch maintain optimal functional mass. It is an organ system capable
2005). This parenchymal unit more closely explains the ran- of rapid responses to a variety of noxious stimuli. Following
dom size and shape distribution of the more classical hepatic loss of hepatocytes from stimuli such as transient toxic insult,
lobule as seen in a conventional two-dimensional histology infection, or partial hepatectomy, the liver is rapidly restored
slide. It also provides a basis for understanding the heteroge- to its optimal mass to maintain normal function.
neous response of various hepatic lobules to chemical insult. Liver functions are complex and diverse including endo-
In addition to hepatocytes, the liver is comprised of a variety crine and exocrine activity, metabolism, conjugation, detoxifi-
of cell types, including biliary cells, endothelial cells, Kupffer cation, and hematopoiesis in early embryonic and fetal
cells, Ito cells (stellate cells), fat-storing cells, and pit cells in development (Harada et al. 1999). The liver is continuously
addition to hematopoietic cells in the sinusoids and blood ves- exposed to all ingested substances absorbed through the intest-
sels. Polyhedral hepatocytes comprise approximately 60% of inal tract via the portal vein and systemically via the arterial
the liver arranged in plates or cords that radiate from the central blood supply. A pivotal hepatic function in toxicologic pathol-
vein to the portal areas. In two-dimensional sections they are ogy is xenobiotic biotransformation that leads to detoxification
typically one cell layer thick and form anastomoses (Miyai of materials absorbed in the intestinal tract. Xenobiotic
1991). On one surface they are separated from the sinusoidal metabolism by hepatocytes can occur by phase I (often the
wall by a peri-sinusoidal space, the space of Disse, where cytochrome oxidase series) and phase II reactions (often the
they are exposed to tissue fluids. On the opposite side of formation of the water soluble glucuronide) (Graham and Lake
the hepatocyte bile canaliculi are formed with hepatocytes in 2008; Martignoni, Groothuis, and de Kanter 2006). Hepatic
an adjacent hepatic cord. Desmosomes, gap junctions, and metabolic processes may also cause indirect toxicity by gener-
stud-like protrusions connect contiguous hepatocytes within a ating electrophilic species capable of reacting with proteins,
cord. Biliary cells form bile ducts in the portal areas and nucleic acids, and other cytoplasmic organelles (Xu, Li, and
constitute the portal triad with a hepatic artery and a portal Kong 2005). Intrinsic and induced enzymes responsible for
vein. Fenestrated endothelial cells line the sinusoids and hepatic function may be unevenly distributed throughout the
synthesize prostaglandins. Kupffer cells are a self-renewing hepatic lobule and between the different lobes (Greaves 2007).
fixed macrophage comprising approximately 10% of all liver The presence of background changes and undercurrent dis-
cells (Eustis et al. 1990). Kupffer cells are phagocytic, secrete ease states affects the hepatic and biliary morphology, for
mediators of inflammation, and catabolize lipids and proteins. Ito example, caloric restriction diminishes hepatocellular size and
cells (stellate cells) are peri-sinusoidal cells that store vitamin A can make interpretation of test-article–related changes more
and are also a major source of collagen in the liver. Pit cells are challenging. Other factors that influence the liver morphology
8S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

TABLE 2.—Selected immunohistochemical stains that have been Most toxicologic pathologists use a common grading scale
used to identify different cell types in liver sections. such as marginal or minimal, slight, moderate, marked, and
severe for inflammatory, necrotizing, or other degenerative and
Immunohistochemical stains of liver cells responsive lesions. Tissue-specific locators are often used, such
Cell type Antibody as portal, periportal, midzonal, centrilobular, hilar, ductal, peri-
ductal, peri-canalicular, or subcapsular to indicate the lesion
Hepatocytes CK8, CK18
distribution within the liver. Focal, multifocal, and diffuse are
Bile canaliculi Polyclonal CEA
Bile duct epithelium CK7, CK19, AE1/AE3 commonly used modifiers in the morphological diagnosis for
Endothelial cell Factor VIII, CD31, CD34 distribution parameters. Based on the formal definition, a focal
Exudate macrophages (monocytes) ED1 lesion refers to one specific area, or focus, whereas multifocal
Kupffer cells CD68, F4/80, ED2, SRA-E5 refers to more than one focus (foci). However, some patholo-
Hepatic stellate cells (activated), a-SMA
gists use focal for both focal and multifocal, referring to the
myofibroblasts and smooth muscle cells
Dendritic cells NLDC-145, OX-6 nature of the lesion rather than its actual distribution and using
Oval cells a-fetoprotein (AFP), CK20 grading to reflect the extent of the multifocality. Schemes for
Apoptosis Bcl-2, Caspase 3 and 7 scoring lesion severity vary widely and no single system is
Proliferation markers Ki67/MIB-1, PCNA likely to be accepted by all pathologists. While a sample grad-
ing scheme for focal and multifocal liver lesions is provided in
Geller, Dahll, and Alsabeh (2008); Malhotra, Sakhuja, and Gondal (2004); Hurlimann
and Gardiol (1991); Davenport et al. (2001); Kashiwagi, Kaidoh, and Inoué (2001); Faa Table 3, this should not be regarded as a universal or specific
et al. (1998). INHAND-recommended grading scheme.

are: body weight loss, blood flow, food intake, vascular and TABLE 3.—A sample grading scheme for focal and multifocal
hemodynamic changes, timing and duration of exposure, with- liver lesions (modified from Hardisty and Eustis 1990; World
drawal effects, and functional heterogeneity. Functional hetero- Health Organization 1978; Derelanko 2000).
geneity expresses itself via differences in metabolism, oxygen
supply, b-oxidation, amino acid metabolism, gluconeogenesis, Proportion of Quantifiable
Severity liver affected Grade finding
glycolysis, ureagenesis, liponeogenesis, and bile acid and biliru-
bin secretion. These factors can affect occurrence of nonproli- Marginal or minimal Very small amount 1 1-2 foci
ferative as well as proliferative liver lesions in rodents. Slight or few Small amount 2 3-6 foci
Moderate or several Medium amount 3 7-12 foci
Marked or many Large amount 4 >12 foci
V. LIVER NECROPSY AND TRIMMING PROTOCOL Severe Very large amount 5 Diffuse
At necropsy, rat and mouse liver may be weighed and
individual liver lobes examined carefully for gross lesions. In VII. NOMENCLATURE, DIAGNOSTIC CRITERIA, AND DIFFERENTIAL
conventional preclinical rodent studies, gross lesions must be DIAGNOSIS
correlated with the histopathological findings. Liver-specific
A. Congenital Lesions
trimming protocols (see Figure 1) according to standard oper-
ating procedures (SOPs) are used (e.g., see Ruehl-Fehlert Introduction
et al. 2003). Dissected lobes and trimmed liver pieces can be
Developmental anomalies occasionally occur in the liver of
fixed in 10% neutral buffered formalin (no more than 1 cm
rodents. These malformations might be expressed in different
thick in 1:10 tissue: formalin).
forms and be of different origin. They mostly occur as isolated
effects and are considered by the pathologist in distinguishing
VI. GRADING OF LIVER LESIONS
background hepatic lesions versus xenobiotic-induced lesions
Interpretation of hepatic lesions in safety assessment studies that occur in rodent preclinical toxicity studies.
requires consideration of gross and microscopic findings,
hematology, clinical chemistry, and liver weights in the con- Hepatodiaphragmatic Nodule (Figures 3 and 4)
current control groups of animals and should take into account
Pathogenesis: Developmental alteration.
species and strain, age, caging, diet, and tissue sampling.
Many pathologists use a grading system to document lesion Diagnostic features:
severity. In toxicological pathology, the generation of ordinal
data using a scoring system allows statistical analysis for  Visible grossly and tinctorially similar to normal
effects and trends (Gad and Rousseaux 2002). However, not all hepatic parenchyma.
grading systems are the same and may differ in how they incor-  Rounded extensions usually of the medial lobe(s).
porate distribution, stage, and extent of lesions. The problem of  Increased mitoses, cytological alterations, and
harmonization as it relates to lesion severity has been recog- nuclear alterations may be present.
nized and discussed in some detail (Hardisty and Eustis  Linear chromatin structures with small lateral projec-
1990; World Health Organization 1978). tions are pathognostic.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 9S

Differential diagnosis: adaptive changes usually do not result in illness or death of


rodents. Often these processes are dose and chemical related.
 Hepatocellular focus of cellular alteration—tinctorial
variation from normal parenchyma and does not pro-
trude into the diaphragm. Fatty Change
 Hepatocellular neoplasia—when visible grossly does Synonyms/subtypes: Lipidosis, vacuolation, lipid, macrovesi-
not protrude into the thoracic cavity. cular and/or microvesicular steatosis, phospholipidosis.1
 Regenerative hyperplastic nodule (nodular
hyperplasia)—typically involves multiple nodules of
Pathogenesis: Perturbations in lipid metabolism and disposition.
hyperplasia separated by proliferative bands of oval
cells or connective tissue. Diagnostic features:
Macrovesicular fatty change (Figures 5 and 6).
Comment: Hepatodiaphragmatic nodules can be seen in rats at
any age and their occurrence in fetuses is considered presump-  Hepatocytes contain a large well-defined single
tive evidence of a congenital origin. While they appear to be rounded vacuole within each cell.
protruding through the diaphragm and extending into the thor-  Nucleus and cytoplasm displaced to the periphery.
acic cavity, they actually are attached to and covered by a thin  A few hepatocytes may contain one or more smaller
fibrous portion of the diaphragm (Eustis et al. 1990). vacuoles.
An incidence ranging from 1% to 11% has been reported for
Microvesicular fatty change (Figure 7).
hepatodiaphragmatic nodules in Fischer 344 rats (Eustis et al.
1990), with few cases reported in other rat stocks and strains.  Hepatocytes partially or completely filled with
Mice do not develop such nodules but may have focal lesions numerous small lipid vacuoles.
similar to those in rat hepatodiaphragmatic nodules and with  Affected hepatocytes may have a ‘‘foamy’’ appearance.
large nuclei with large central nucleoli-like basophilic bodies.  Small vacuoles do not normally displace the nucleus to
the periphery in contrast to macrovesicular steatosis.

B. Hepatocellular Responses, Cellular Degeneration, Differential diagnosis:


Injury, and Death
 Hydropic degeneration—clear cytoplasm without
Introduction nuclear displacement.
The function and structure of most liver cells are  Glycogen accumulation—irregular and poorly defined
relatively constrained by their genetic programs of metabolism, lacy clear spaces in the cytoplasm (rarefaction) usually
differentiation, and specialization. While the cells of the hepatic with centrally located nuclei.
parenchyma have the flexibility to adapt to changing physiologi-
cal demands with reversible functional and morphological altera- Comment: There is a difference in preferred nomenclature
tions, sufficient stress, or noxious stimuli may lead to inability to among pathologists for this change. Based strictly on an
maintain homeostasis and adverse cellular adaptations. The mor- H&E-stained section, a diagnosis of cytoplasmic vacuolation
phological response to injurious stimuli depends on the nature of of hepatocytes is a universally acceptable descriptive diagno-
the injury and its severity and duration. Often at high doses, tar- sis. Based on the experience of the observer, the specific mor-
geted cells go through a sequence of cellular degeneration fol- phological features of the cytoplasmic vacuolation may be
lowed by cell death, but at lower doses degenerative changes sufficiently consistent with intracytoplasmic lipid accumula-
do not necessarily lead to cell death. Consequentially, cellular tion to warrant a presumptive diagnosis of fatty change. The
changes that do not lead to cell death or death of the animal may unequivocal demonstration of intracytoplasmic fat, however,
be called ‘‘adaptive’’ changes that can be considered either requires a special stain.
adverse or not adverse reactions, depending on the nature of the Fatty change can be induced by a number of different agents
change. There are cellular adaptations involving metabolic or and is usually divided into two main types, namely, microvesi-
functional alterations that lead to increases in cellular organelles cular and macrovesicular, although mixed forms can frequently
and intracellular accumulations of a variety of endogenous and be observed (Greaves 2007; Gopinath, Prentice, and Lewis
exogenous substances but allow the cell and animal to survive 1987; Goodman and Ishak 2006; Kanel and Korula 2005).
and often live normally. Similar changes may occur in human Macrovesicular lipidosis is a reaction to a wide variety of
liver, such as cholestasis, a common lesion in human liver injuries and can also be regarded as a physiological adapta-
after long-term drug therapy. However, in animals, when the tion demonstrated as an imbalance between uptake of lipids
limits of adaptive responses are exceeded or do not occur in from blood and secretion of lipoproteins by the hepatocyte
response to chemical exposure, irreversible cellular injury and (Goodman and Ishak 2006). Microvesicular lipidosis is usually
cellular death occurs, with possible subsequent illness and
1
death. Adaptive changes or doses of chemicals that induce Electron microscopy or special staining needed for a definitive diagnosis.
10S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

indicative of more serious hepatic dysfunction but can also Differential diagnosis:
result from nutritional disturbances (Greaves 2007).
Specific xenobiotics can induce either macrovesicular or  Fatty change—round clear vacuoles tend to be single
microvesicular lipidosis in humans (Kanel and Korula 2005). or multiple and discrete.
In animal studies, it is common to see a mixture of macrovesi-  Glycogen accumulation—irregular and poorly
cular and microvesicular lipidosis. In those situations one can defined clear spaces in the cytoplasm (rarefaction)
either diagnose the most prevalent form or record the findings usually with centrally located nuclei; positive stained
as mixed. Commentary in the pathology narrative report might with periodic acid-Schiff staining.
be appropriate, especially if recording the most prevalent form
of lipidosis. Liver with admixed presence of glycogen and fatty Comment: Definitive diagnosis of phospholipidosis is not pos-
change can be observed (Figures 8 and 9). sible based strictly on H&E-stained liver sections. A diagnosis
Fatty change and necrosis may appear together although of cytoplasmic vacuolation of hepatocytes will typically be an
they may differ in proportion. A number of causes other than acceptable descriptive diagnosis. Since the cytoplasmic
xenobiotic exposure, such as chronic hepatic injury, diet, vacuolation may mimic microvesicular fatty change, a descrip-
metabolic and hormonal status, debilitation of animals, and tive diagnosis of cytoplasmic vacuolation is recommended in
fasting before necropsy, should be taken into consideration the absence of electron microscopy or special immunostaining.
in reviewing these changes (Vollmar et al. 1999; Katoh and Phospholipidosis can be induced by xenobiotics with a
Sugimoto 1982; Nagano et al. 2007; Denda et al. 2002). The cationic amphophilic structure (Halliwell 1997; Anderson and
distribution can be either diffuse (e.g., ethionine) or zonal Borlak 2006; Reasor, Hastings, and Ulrich 2006; Chatman
(e.g., centrilobular in CCl4; periportal in phosphorus toxicity; et al. 2009) (Figures 14 and 15). It is a lipid storage disorder
midzonal in choline deficiency). Inadequate fixation proce- seen when complexes between xenobiotics and phospholipids
dures may sometimes give rise to artifacts with microvesicu- accumulate within lysosomes. Phospholipidosis refers to a spe-
lar vacuolation, although mostly with less clear cytoplasm cific form of hepatic vacuolation with the occurrence of con-
(Li et al. 2003). centric membrane bound lysosomal myeloid bodies/lamellar
Focal fatty change can sometimes be seen spontaneously and bodies that can be confirmed by specific staining and electron
is usually described as such. A specific variation occurs near the microscopy (Hruban, Slesers, and Hopkins 1972; Obert et al.
attachment of the falciform ligament and gallbladder in mice and 2007) (Figure 16). Definitive diagnosis requires electron
is referred to as ‘‘tension lipidosis’’ (Harada et al. 1999) (Figures microscopy or positive immunostaining. Immunohistochem-
10 and 11). Spontaneous fatty change can differ between strains ical staining for a lysosomal-associated protein and adipophilin
and is a normal finding in BALB mice. Livers of these mice are may be used to differentiate phospholipidosis from conven-
typically paler than in other strains. Focal fatty change in the tional fatty change (Obert et al. 2007). Both preexisting neutral
liver of rodents has previously been categorized as vacuolated fat and phospholipids can be observed in combination. The
altered hepatic foci (Eustis et al. 1990), but current practice is macrovesicular and the microvesicular fatty change (vacuola-
to diagnose this change as focal fatty change rather than as a tion) generally located at the cell periphery stains positively for
focus of hepatic alteration (Figures 12 and 13). Oil Red-O and the membranes surrounding these lipid
Fatty change can also be observed in combination with other vacuoles stain positively for adipophilin (a protein that forms
hepatotoxic injuries (e.g., chronic liver toxicity, degeneration, the membrane around non-lysosomal lipid droplets) but neg-
inflammation, and necrosis) or nutritional disturbance (e.g., ative for LAMP-2 (a lysosome-associated protein) by immu-
diet, vitamin A excess) in both animals and man. Special stains nohistochemical techniques (Obert et al. 2007). This indicates
on cryostat sections can demonstrate fat (e.g., Oil red O or that this vacuolation was due to accumulation of non-
Sudan Black) (Jones 2002). lysosomal neutral lipid. Cytoplasmic microvesiculation
located centrally in hepatocytes that exhibit positive immuno-
histochemical staining for LAMP-2 (Figure 17) but is nega-
Phospholipidosis2 tive for Oil-Red-O and adipophilin is indicative of
phospholipid accumulation (Obert et al. 2007).
Synonym: Cytoplasmic vacuolation, foam cells.
Amyloidosis (Figures 18 and 19)
Pathogenesis: Induced by xenobiotics with a cationic ampho-
philic structure. Pathogenesis: Cellular process related to misfolding of protein.
Diagnostic features: Diagnostic features:

 Multiple irregular to round clear membrane-bound  Deposition of pale, homogeneous, amorphous eosi-
vacuoles. nophilic material.
 Tends to be a diffuse change affecting hepatocytes.  Deposition often peri-sinusoidal, periportal, or
involving blood vessel walls.
2
Electron microscopy or special staining needed for a definitive diagnosis.  Localization is extracellular.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 11S

Comment: This is a rare condition in rats but is a more com- Spencer et al. 1997; Yasui, Yase, and Ota 1991; DePass
mon age-related phenomenon in hamsters and mice et al. 1986). Mineralization can sometimes be observed in
(Greaves 2007; BSTP 2007). The basis of the pathological combination with inflammation or neoplasia (Harada et al.
change is the cell’s inability to prevent protein misfolding, 1999; Kanel and Karuda 2005). Mineral deposits can be
to revert misfolded proteins to normal, or to eliminate mis- demonstrated by using additional stains (Alizarin Red, von
folded proteins by degradation. This can result in deposition Kossa) (Churukian 2002).
of potentially cytotoxic protein aggregates of amyloid as in
other protein aggregation diseases (Aigelsreiter et al. 2007).
The amyloid is predominantly composed of protein in a
Pigmentation (Pigment Deposition) (Figures 21–25)
beta-pleated sheet conformation.
The incidence of spontaneous amyloidosis usually Pathogenesis: Incidental occurrence and secondary to cellular
increases with age and is common in CD-1 mice (Harada and erythryoid breakdown products; lipid peroxidation of cel-
et al. 1996). Amyloid observed in the liver often is referred lular membranes; altered heme metabolism.
to as secondary amyloidosis (serum amyloid A protein) and
is seen in the sinusoids and within the portal vessel walls. Diagnostic features:
Hepatocytes adjacent to sinusoidal amyloid deposits are often
atrophic. A number of factors (e.g., species, age, strain, gen- Lipofuscin:
der, endocrine status, diet, stress, and parasitism) can influ-
ence the occurrence of amyloidosis (Beregi et al. 1987; Coe  Pigment can be seen in hepatocytes as well as in
and Ross 1990; Lipman et al. 1993; Harada et al. 1996; Liu Kupffer cells.
et al. 2007). Other organs are often involved in the deposition  May vary from pale yellow to deep granular
of amyloid (e.g., kidney, nasal submucosa, lamina propria brown.
intestines, heart, salivary gland, thyroid, adrenal cortex, lung,  May be sudanophilic with autofluorescence under
tongue, testis, ovary, and aorta). ultraviolet light.
Amyloidosis can be confirmed with additional histo-  Often located adjacent to bile canaliculi.
chemical staining (Congo red) where it shows pink-red stain-
ing and apple green birefringence under polarized light
(Vowles and Francis 2002; Kanel and Korula 2005) and by Iron/hemosiderin:
immunohistochemistry.
 Can be yellow to brown.
 May be finely granular.
Mineralization (Figure 20)  Usually appears intracellularly in Kupffer cells and
hepatocytes.
Pathogenesis: Hypercalcemia secondary to diet or abnormal
calcium metabolism; hepatocellular necrosis (dystrophic
mineralization). Porphyrin:

Diagnostic features:  Pigment is dense dark brown to red-brown and when


viewed with polarization is bright red with a centrally
 Intra- or extracellular basophilic deposits, sometimes located dark ‘‘Maltese cross.’’
with calcification.  Brilliant red fluorescence when viewed in fresh
frozen sections; fades with exposure to ultravio-
Differential diagnosis: let light.
 Most often located in bile ductules and bile
 Artifact—hematoxylin stain deposits in clear canaliculi.
spaces.
 Pigment deposits—may be tinctorially different from Bile (cholestasis) (Figures 23–25):
mineralization and often seen within macrophages.
 Intrabiliary accumulation of test compound or  Appears as elongated pale green-brown plugs within
metabolite. bile caniculi.
 May be associated with necrosis, inflammation, or  Will appear in Kupffer cells following rupture of
neoplasia. caniculi.
 Can appear as finely granular pigment within in hepa-
Comment: Mineralization is rarely seen in the liver and gall- tocytes, which is common in human liver but much less
bladder in rodents. Dietary factors (mineral content) and dis- common in rodents.
turbance of calcium metabolism commonly influence the  Not a common xenobiotic response in rodents; more
process of hepatic mineralization (Harada et al. 1999; common in humans and monkeys.
12S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Differential diagnosis: (Ungar, Sullman, and Zuckerman 1976). Endogenous iron


deposition can be found following breakdown of blood cells
 Artifact—hematoxylin stain deposits in clear spaces. (hemolytic event). Iron pigment can be found in Kupffer
 Formalin precipitated pigment—extracellular granu- cells, macrophages, and hepatocytes. In hepatocytes, the
lar yellow-brown deposits often associated with iron is stored in the form of ferritin (ferric iron bound to
erythrocytes. protein apoferritin) (Popp and Cattley 1991). A spontaneous
 Test compound/metabolite—may be distinctive for inherited predisposition for hepatic iron pigmentation has
the specific compound. been reported in Sprague-Dawley rats (Masson and Roome
 Mineralization—basophilic deposits; may be associ- 1997), and iron deposition can be found in the aging mouse
ated with calcification. liver (Harada et al. 1996). Iron can be demonstrated using
Perls’ Prussian blue stain in which iron stains blue.
Comment: A number of different pigments may be seen as an Hemosiderin slowly dissolves in acids, especially oxalic
incidental finding within hepatocytes and Kupffer cells in acid. Non-aldehyde fixatives can remove hemosiderin or
rodents. Some of them may increase and/or accumulate after alter it in such a way that reactions for iron are (false) neg-
treatment. Definitive diagnosis of a specific pigment typically ative (Churukian 2002). Malarial pigment is seen in hepato-
requires special stains. cytes and Kupffer cells of Plasmodium sp experimentally
Lipofuscin or ceroid is sometimes referred to as ‘‘wear and infected mice. It is the pigment from the organism and not
tear’’ or ‘‘aging’’ pigment and therefore is often observed in hemosiderin.
older animals. It is considered to represent a breakdown of cell Porphyrin pigment normally occurs in tissues only in
membranes. Lipofuscin accumulates in postmitotic and aging small amounts and is a precursor of the heme portion of
cells. It has been shown to be a mixture of oxidized proteins and hemoglobin (Churukian 2002). Porphyrin deposition in the
lipids, carbohydrates, and trace amount of metals (Seehafer and liver of rodents is found after administration of a number
Pearce 2006). A variety of stimuli can accelerate the of compounds including griseofulvin where it can be seen
accumulation of this pigment, such as drug and chemical expo- in association with hepatocellular neoplasia (Stejskal et al.
sure, trauma and circulatory factors, and diet (Greaves 2007). 1975; Zatloukal et al. 2000; Knasmuller et al. 1997;
Lipofuscin accumulation in the liver may be augmented by cer- Tschudy 1962). Griseofulvin administration in mice may
tain chemicals (Kim and Kaminsky 1988; Marsman, 1995). result in inhibition of the mitochondrial enzyme ferrochela-
Treatment of rats with PPAR alpha agonists such as fenofibrate tase and (compensatory) induction of ALA synthetase.
and associated increased lipid peroxidation seen in rodents Griseofulvin-induced accumulation of porphyrins in mouse
treated with hypolipidemic agents can induce lipofuscin accu- liver is followed by cell damage and necrotic and inflamma-
mulation in liver after prolonged treatment (Nishimura et al. tory processes (Knasmuller et al. 1997). Proto-porphyrin
2007; Goel, Lalwani, and Reddy 1986; Reddy et al. 1982). pigment in liver of rats and mice is mainly found in the bile
Increased lipofuscin accumulation has also been observed in par- ducts and leads to bile duct proliferation and portal inflam-
tially hepatectomized liver of rats (Sigal et al. 1999). Lipofuscin mation, but can also occur in hepatocytes, Kupffer cells, and
is insoluble in alcohols and xylene and other solvents normally portal macrophages (Hurst and Paget 1963). The birefrin-
used in the preparation of slides. Special stains such as Smorl’s gence of porphyrin appears to be associated with bilamellar
can be used to demonstrate the pigment. Storage granules appear components within the pigment (Stejskal et al. 1975). This
gray with Sudan Black B, may be PAS-positive, and may stain pigment is also seen in combination with liver fibrosis and
with Luxol fast blue and Ziehl-Neelsen (Jones 2002). cirrhosis, bile duct proliferation, periportal inflammation,
Porphyrin pigment, a precursor of heme protein, is seen with and hepatocarcinogenesis (Kanel and Korula 2005; Hurst
treatment of some xenobiotics. Bile pigment is a common find- and Paget 1963; Greaves 2007; Rank, Straka, and Bloomer
ing when there is cholestasis secondary to obstruction of bile 1990).
flow or when there is perturbation in bile metabolism. Bile pig-
ment stains green with Hall’s method.
Hemosiderin pigment represents precipitated iron that is Crystals (Figures 26–28)
most frequently generated as a breakdown product of ery-
throcytes and is derived from hemoglobin and accumulates Pathogenesis: Hyperlipidemia (cholesterol crystals), Chi313
in the liver following local or systemic excess of iron. (Ym1) protein (eosinophilic biliary crystals).
Deposition or iron may occur following excess dietary Diagnostic features:
intake or treatment by xenobiotics (Popp and Cattley
1991; Greaves 2007; Travlos et al. 1996). Excess of iron  Rhomboid or needle-like structures often birefringent
following injection may be stored as hemosiderin and under polarized light.
deposited in the reticuloendothelial component of the liver  Needle-like crystals in the mouse can be intracellular
(and other organs such as spleen and bone marrow) or extracellular and may be associated with intense
(Bruguera 1999; Pitt et al. 1979). Intraperitoneal injection eosinophilic epithelial cytoplasm and extracellular
of aflatoxin B1 can also induce hemosiderosis in hamsters crystals of various sizes.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 13S

Differential diagnosis: Comment: Both intranuclear and intracytoplasmic inclu-


sions are common findings in the aging mouse liver and
 Artifact—wispy blue hematoxylin deposits in clear may be seen in normal as well as neoplastic hepatocytes
spaces. (Percy and Barthold 2001; Frith and Ward 1988; Irisarri
and Hollander 1994). When the intranuclear inclusions
represent invaginations of the cytoplasm into the nucleus,
Comment: In hyperlipidemia, cholesterol crystals can deposit they may contain cytoplasmic organelles in electron micro-
in the liver with or without granulomatous inflammation graphs (van Zwieten and Hollander 1997). Ultrastructu-
(Greaves 2007; Graewin et al. 2004; Handley, Chien, and rally, three types of cytoplasmic inclusions have been
Arbeeny 1983). During gall stone formation, in addition to described: dense reticulated substance in the dilated cister-
classical rhomboid-shape monohydrate crystals, cholesterol nae of rough endoplasmic reticulum, fine granular sub-
can also crystallize transiently as needle-, spiral-, and tubule- stance in rough endoplasmic reticulum, and non–membrane
shaped crystals of anhydrous cholesterol (Dowling 2000). Eosi- bound dense granulofibrillar in the cytoplasm (Helyer and
nophilic crystals have been described in intrahepatic bile ducts Petrelli 1978).
and gallbladder of different laboratory mice strains, and some Kakizoe, Goldfarb, and Pugh (1989) have correlated the
of these crystals have been shown to contain chitinase-like pro- incidences of cytoplasmic inclusion with hepatocellular
teins confirmed by immunohistochemistry for Ym1 protein tumors in different mice strains. C57BL/6 mice are rela-
(now Chi313) (Ward et al. 2001; Harbord et al. 2002). tively more resistant to hepatocarcinogens than C3H and
Crystal formation may be associated with inflammatory C57BL/6 x C3H F1 mice. The tumors in the C57BL/6 mice
and/or proliferative bile duct changes and fibrosis in mice and were unique in their early focal development of cells con-
may also occur spontaneously (Lewis 1984; Rabstein, Peters, taining inclusions. The authors suggested that the higher
and Spahn 1973; Enomoto et al. 1974). Numerous crystals can incidence of inclusions in liver might be related to slowing
be demonstrated using a simple system of polarizing micro- of the tumor growth leading to lower incidence of hepato-
scopy. Crystals are capable of producing plane-polarized light, cellular tumors in C57BL/6 mice. Other types of intracyto-
thus showing birefringence. plasmic inclusions such as Mallory bodies, lamellated, and
crystalloid inclusions have been described in mice treated
with different chemicals and in lysosomal storage diseases
Inclusions, Intranuclear, and Cytoplasmic (Figures 29–32)
(Gebbia et al. 1985; Meierhenry et al. 1983; Rijhsinghani
Synonyms: Inclusion bodies, intranuclear cytoplasmic invagi- et al. 1980; Shio et al. 1982).
nation, acidophilic inclusions, globular bodies. Cytoplasmic vacuoles can occur in hepatocytes and
endothelial cells in a postmortem time-dependent manner in
fasted and non-fasted rats (Li et al. 2003). This artifact is espe-
Pathogenesis: Protrusion of cytoplasm into an invagination of
cially common in rats that are not exsanguinated at necropsy
the hepatocyte nuclear membrane without the actual protrusion
and the cytoplasmic vacuoles represent plasma influx into to
necessarily being present in the plane of section. Seen in spe-
affected cells (Figure 33). This artifact is more common in
cific viral infections. Deposition of protein material within
males than in females.
hepatocyte cytoplasm.
Diagnostic features:
Hypertrophy, Hepatocellular (Figures 34–41)
 Intranuclear inclusions are round, distinct, usually Synonyms: Hepatocytomegaly.
eccentrically located, and may partially or almost
completely fill the nucleus.
Pathogenesis: Metabolic enzyme induction causing increase in
 Contents of intranuclear inclusion bodies are often
endoplasmic reticulum; increase in peroxisomes; increase in
eosinophilic and may be granular or flocculent.
mitochondria.
 Intracytoplasmic inclusions are round to oval, homo-
genous, eosinophilic, and occur as single or multiple Diagnostic features:
structures in the cytoplasm.
 Enlarged hepatocytes may be tinctorially distinct.
Differential diagnosis:  Cytoplasm may be homogeneous or granular.
 Zonal pattern of distribution (centrilobular, peripor-
 Enlarged nucleolus—one or more deeply basophilic tal, midzonal) may be present.
structures in normal size nuclei.  Involving most or all lobules.
 Viral inclusion bodies (cytomegalic virus, experi-  Loss of hepatocellular plate architecture is possible.
mental viral infections).  Sinusoidal compression.
 Cytoplasmic vacuole artifact—postmortem plasma  Concurrent degeneration and/or single cell necrosis is
influx (Li et al. 2003). possible.
14S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Differential diagnosis: Diagnostic features:

 Hepatocellular neoplasia—expansile mass with  Decreased size of hepatocytes.


altered growth pattern; loss of lobular structure.  Small liver trabeculae with decreased cytoplasmic
 Regenerative hyperplastic nodules—altered growth volume, close proximity of hepatocyte nuclei, close
pattern; distorted lobular pattern. proximity of portal tracts, and increased basophilia.
 Foci of cellular alteration—usually a discrete  Hepatocyte nuclei may be smaller than normal.
collection of cells within the hepatic parenchyma.  May have a zonal distribution.
 Hepatocellular degeneration—affected cells  May be associated with hepatocellular degeneration
have increased cytoplasmic granularity and and/or single cell necrosis.
eosinophilia.  May be associated with increased sinusoidal size.
 Hepatocellular storage disease.  Depletion of cytoplasmic glycogen.
 Polyploidy—enlarged nuclei and/or binuclear
hepatocytes are often associated with increased cyto- Differential diagnosis:
plasmic volume.
 Shrinkage artifact—retraction of softer tissue from
Comment: Hepatocellular hypertrophy, secondary to increase firmer tissue.
in microsomal enzymes often occurs with a zonal or specific  Artifact of fixation or processing—poorly stained tis-
lobular pattern and commonly occurs following exposure to sue with loss of normal structure.
enzyme inducing xenobiotics. There is enlargement of the
hepatocyte cytoplasm secondary to increase in the cytosolic Comment: Hepatocellular atrophy can be caused by a number of
protein or number of organelles (e.g., smooth endoplasmic reti- factors such as inanition, starvation, hemodynamic changes, or
culum, peroxisomes, mitochondria). Classically hepatocyte pressure atrophy from neoplasia (Yu et al. 1994; Gruttadauria
hypertrophy occurs without increase in hepatocyte numbers et al. 2007; Belloni et al. 1988). Hepatocyte atrophy may be
or DNA (i.e., hyperplasia or polyploidy), however, combina- associated with decrease in absolute liver weights in rats
tions with increased mitoses do occur (e.g., PPAR-alpha (Belloni et al. 1988). Ultrastructurally atrophic hepatocytes
agonists). have reduced amounts of glycogen and decreased numbers of
Hepatocellular hypertrophy following enzyme induction mitochondria.
is considered an adaptive response to chemical stress. Strain
differences in responsiveness occur. While typically an
adaptive response, excessive hypertrophy from enzyme Degeneration
induction of hypertrophy can lead to hepatocellular degen- Introduction: In the diagnostic lexicon, degeneration is a non-
eration and necrosis. Hepatocellular hypertrophy may be asso- specific diagnosis that provides limited useful information
ciated with increase in absolute liver weights. Enzyme unless qualified to reflect the dominant morphological features.
induction leading to hepatocellular hypertrophy may be It is often at the borderline between adaptation with resolution
accompanied by some evidence of transient mitogenesis. back to normal structure and function and inability to adapt
Hypertrophy that is panlobular may be difficult to appreciate leading to cell death. In human clinical medicine degenerative
histologically because the contrast provided by a sublobular disease most often refers to chronic debility involving organs
pattern is not evident. In some cases, hepatocyte hypertrophy and tissues that slowly accumulate damage over time. In rodent
related to metabolic enzyme induction may not be evident to studies, degeneration may also be applicable to chronic
the pathologist when liver weight increase is small for a group, debility, but more often it is used to reflect acute or chronic
for example, less than 20%. cytological alterations with characteristic morphological fea-
Hepatocellular enlargement or swelling may occur from tures. Combinations of different degenerative features may
accumulation of glycogen, fat, or other substances and may occur with or without inflammation and/or necrosis.
also be a feature of degeneration and some forms of hepa- Based on H&E-stained sections, distinction between
tocellular necrosis. To avoid confusion with the more com- different forms of degeneration, hepatocellular hypertrophy
mon usage of hepatocyte hypertrophy for physiological secondary to enzyme induction, other forms of hepatocellular
enzyme induction, it is recommended that alternative forms enlargement such as glycogen accumulation/retention, and
of hepatocyte enlargement not be diagnosed as hepatocellu- even early necrosis (a.k.a. onconosis) may be difficult. In some
lar hypertrophy. cases special stains may be required to more clearly delineate
the nature of the cytologic alteration. Based strictly on H&E
staining, a descriptive diagnosis of cytoplasmic alteration is
Hepatocellular Atrophy (Figures 42 and 43)
recommended in lieu of interpretative diagnosis such as gran-
Pathogenesis: Inanition, starvation, hemodynamic changes, or ular degeneration and hyaline degeneration. However, there are
pressure atrophy from neoplasia. some degenerative lesions, such as hydropic degeneration and
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 15S

cystic degeneration, that are more clearly established in tradi- formation (Gonzalez-Quintela et al. 2000; NTP Toxicology and
tional pathology literature. The preferred diagnosis will be Caracinogenesis Studies Ethylene Glycol 1993; Peters et al.
influenced by morphological features, conventional pathology 1983; Bruni 1960; Shea 1958; Omar, Elmesallamy, and Eassa
practice, and the experience of the pathologist. 2005; Lin et al. 1996), but is rarely used as a separate description
Glycogen accumulation in hepatocytes is a type of cytoplas- since a combination of findings is often present. Cytoplasmic
mic alteration manifested on H&E-stained paraffin sections as alteration reflecting plasma influx is an artifact seen in non-
clear spaces in the cytoplasm and a centrally located nucleus. exsanguinated rats in a postmortem time-dependent manner
Intracellular accumulation of glycogen is a normal physiological (Li et al. 2003) (see Figure 33).
response following food ingestion. Since rodents eat primarily in
the evening hours, the largest amount of glycogen will be present Degeneration, Hydropic (Figure 45)
during early morning hours. Intrahepatocyte glycogen is mobi-
Synonyms: Cytoplasmic alteration, cytoplasmic change, hydro-
lized throughout the day, initially being removed from centrilob-
pic change, cloudy swelling.
ular hepatocytes. Consequently the amount present varies
depending upon whether the animals were fasted and on the time
Pathogenesis: Intracytoplasmic fluid accumulation secondary
of necropsy during the day. Failure to accumulate glycogen
to disturbance of cell membrane integrity.
because of inanition or abnormal glycogen retention may result
from treatment-induced metabolic perturbations. Diagnostic features:

Cytoplasmic Alteration (Figure 44)  Cytoplasmic vacuolation and ‘‘ballooning’’ with a


centrally located nucleus.
Synonyms: Cytoplasmic alteration, cytoplasmic change, granu-
 Lobular location may be centrilobular or periportal
lar change, granular degeneration, hyaline degeneration, glyco-
with increased clear cell change and cell swelling.
gen accumulation; ground glass change.
Differential diagnosis:
Pathogenesis: Often xenobiotic-induced and may be associated
with other forms of liver damage.
 Cytoplasmic vacuole artifact—postmortem plasma
Diagnostic features: influx.
 Glycogen accumulation—hepatocytes not markedly
 Affected cells may show increased cytoplasmic gran- enlarged; cytoplasmic clear areas are irregular.
ularity, cell swelling, and eosinophilia.
Comment: Because of disturbance of the cell membrane integ-
Differential diagnosis: rity, accumulation of intracytoplasmic fluid may occur. This
causes vacuolation and ‘‘ballooning’’ of cells. This change can
 Artifact of fixation or processing—poorly stained be caused by a number of xenobiotics with differing lobular
tissue with loss of normal structure. localization and may be a precursor to hepatocyte necrosis
 Hepatocyte hypertrophy—cytoplasmic volume (Gkretsi et al. 2007; Wang et al. 2007; Peichoto et al. 2006;
increased with uniform finely granular texture; usu- Matsumoto et al. 2006; Chengelis 1988).
ally associated with microsomal enzyme induction
or peroxisome proliferation. Degeneration, Cystic (Figure 46 and 47)
 Cytoplasmic vacuole artifact—postmortem plasma
Synonyms: Spongiosis hepatis (traditional diagnostic term pre-
influx.
ferred by many pathologists).
 Coagulation necrosis—loss of cytoplasmic and
nuclear detail.
Pathogenesis: Cystic enlargement of perisinusoidal stellate
cells (Ito cells) particularly observed in aging rats.
Comment: What has been described as granular degeneration
can be seen in combination with other forms of liver damage Diagnostic features:
(e.g., necrosis, hydropic degeneration, inflammation) (Huang
et al. 2007; Gokalp et al. 2003; Datta et al. 1998; Xu et al.  Multi-loculated cyst(s) lined by fine septa containing
1992; Aydin et al. 2003). Hepatocellular granularity may be fine flocculent eosinophilic material (PAS-positive).
due to swelling of cell organelles or increase in the numbers  The cysts are not lined by endothelial cells and do not
of cell organelles including peroxisomes, mitochondria, and compress the surrounding liver parenchyma.
smooth endoplasmic reticulum. Some pathologists do not con-  May be accompanied by occasional erythrocytes or
sider granular degeneration to be a distinct entity and do not leukocytes.
include it in their diagnostic lexicon.  May be observed within altered hepatic foci and liver
Hyaline degeneration has been described by a number of tumors.
authors, sometimes in combination with Mallory body  Affected cells may be markedly enlarged.
16S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Differential diagnosis: Single Cell Necrosis (Apoptosis) (Figures 48–50)


Diagnostic features:
 Angiectasis (Peliosis hepatis; vascular ectasia)—
dilated endothelial lined vascular spaces that often
 Affected hepatocytes may have condensed hyper-
contain blood cells.
eosinophilic cytoplasm and a somewhat angular outline.
 Sinusoidal dilatation—sinusoidal structure evident
 Not associated with an inflammatory response unless
and spaces lined by endothelial cells.
there is simultaneous necrosis.
 Necrosis—loss of nuclear and cytoplasmic detail and
 May occur spontaneously with one or two affected
loss of stain affinity.
hepatocytes present in an occasional hepatic lobule.
 Hemangioma—expansile structure lined by flattened
 May be exacerbated by treatment.
endothelial cells; may be associated with parenchy-
 In standard H&E-stained sections, apoptotic hepato-
mal compression.
cytes (apoptotic bodies) are usually rounded with
 Hemangiosarcoma—expansile mass lined by plump
condensed cytoplasm.
endothelial cells and/or layers of endothelial cells
 Rounded apoptotic bodies are typically surrounded
and associated with destruction of hepatic
by a clear halo.
parenchyma.
 Fragments of nuclear material may be present within
affected cells.
Comment: Spontaneous and xenobiotic-induced cystic degen-  Apoptotic bodies are frequently phagocytosized by
eration/spongiosis hepatis may occur in rats (Karbe and Kerlin adjacent normal cells including hepatocytes and
2002; Bannasch 2003; Babich et al. 2004; Newton et al. 2001). macrophages.
It is more common in aging rats with some male predilection. It
is less common in mice. This lesion may be seen in or associ- Differential diagnosis:
ated with other hepatic lesions (necrosis, regeneration; foci of
cellular alteration; hepatocellular neoplasms). The pathogen-  Small foci of necrosis—typically cells are swollen
esis is not fully understood (Bannasch, Block, and Zerban and there is loss of membrane integrity; usually not
1981; Karbe and Kerlin 2002). rounded; less intensely stained than apoptotic bodies;
may be accompanied by inflammatory cells.

Cell Death (Necrosis, Apoptosis) Comment: Apoptosis is a form of genetically controlled


Introduction: In the fully developed organism, cell death is the ‘‘programmed cell death.’’ Microscopically in H&E-
ultimate result of irreversible cellular injury. Cellular death in stained tissue sections, apoptosis appears as dense eosino-
the liver is manifested by a spectrum of morphological pat- philic shrunken cell bodies with maintenance of membrane
terns that can occur alone or in combinations. However, there integrity, nuclear fragmentation and cytoplasmic budding,
are two primary manifestations of cell death: necrosis and and without an inflammatory response. Definitive diagnosis
apoptosis. For decades a form of necrosis involving individual of apoptosis can be made by histological findings and
isolated hepatocytes has been diagnosed as ‘‘single cell necro- confirmed by distinctive electron microscopic features. Con-
sis.’’ This particular change is now regarded as ‘‘apoptosis’’ sequently, use of a diagnosis of ‘‘single cell necrosis’’ is
by most pathologists (Levin 1999; Levin et al. 1999; Elmore appropriate based strictly on H&E staining. The use of
2007) when the majority of injured cells have the typical TUNEL kits or caspase immunostaining may assist in diag-
apoptotic morphology. Provided that there is no accompany- nosing apoptosis and enumerating affected cells, but necro-
ing inflammatory reaction, the two terms are synonymous. sis may also be immunopositive. Inhibition of apoptosis
However, since a diagnosis of ‘‘apoptosis’’ implies a specific also plays a key role in the process of carcinogenesis (Fos-
sequence of biochemical and morphological events and ter 2000). Although apoptosis can be observed sponta-
should ideally be supported by electron microscopy, it may neously in the liver, certain chemicals may be able to
be more prudent to diagnose single cell necrosis unless there trigger direct stimulation of pro-apoptotic pathways in hepa-
is definitive proof of apoptosis provided by electron micro- tocytes (Feldmann 1997; Reed 1998). Apoptosis can also
scopy (Levin et al. 1999). It can be mentioned in the pathol- accompany treatment-related zonal necrosis in the liver,
ogy narrative that the observed ‘‘single cell necrosis’’ is especially in situations where there may be a xenobiotic-
morphologically consistent with ‘‘apoptosis.’’ induced effect (Cullen 2005; Greaves et al. 2001).
While apoptosis represents a specific genetically pro-
grammed form of cell death unaccompanied by an inflamma-
Pathogenesis: Direct or indirect cellular damage, including tory response, there are situations where small numbers of
anoxia. Apoptosis (a.k.a., single cell necrosis) can occur spon- cells and even occasional single cells characterized by cell
taneously in liver and may also be exacerbated or induced by swelling can undergo necrosis without an inflammatory
treatment. response. This represents an early stage of conventional
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 17S

necrosis, may occur within hours after exposure to a xenobio- seen after anoxia, or exposure to tannic acid, chloroform, or
tic, and should not be diagnosed as single cell necrosis (apop- other hepatotoxic agents (Gopinath, Prentice, and Lewis
tosis). A more appropriate diagnosis for this situation is focal 1987). This zone (Rappaport zone 3) is particularly vulnerable
necrosis (see the following). to ischemic damage because of its low oxygen gradient and
generation of toxic metabolites due to high content of xenobio-
Necrosis, Focal/Multifocal (Figures 51–53) tic metabolizing enzymes (Comporti 1985; Walker, Racz, and
McElligott 1985).
Diagnostic features:
Diagnostic features:
 Single or multiple foci of a few pale staining
hepatocytes.  Early necrotic hepatocytes are swollen.
 Usually retain normal morphological outline.  Cytoplasm has increased eosinophilia.
 May be associated with inflammation.  Nucleus undergoing lysis, not pyknosis.
 May have an irregular distribution but can also occur in  May have a minimal associated inflammatory
the subcapsular areas with minimal or no inflammation. reaction.
 Early lesions typically consist of three or four hepa-  Can be accompanied by glycogen depletion, hydro-
tocytes, but as the lesions progress more hepatocytes pic degeneration, fatty change, hemorrhage, and
may be involved. ‘‘ballooning’’ of hepatocytes.
 Subcapsular necrosis may sometimes be observed in
combination with hypertrophy. Midzonal (Figures 60–61)

Differential diagnosis: This necrosis is the least common form of zonal necrosis and
is mediated by specific toxicants (e.g., furan, concavalin-A,
beryllium) (Wilson et al. 1992; Boyd 1981; Seawright 1972;
 Foci of extramedullary hematopoiesis—mature and/
or immature erythroid and myeloid cell aggregates Satoh et al. 1996; Cheng 1956). The location is considered
without accompanying hepatocyte necrosis. specific and has a metabolic basis.
 Foci of inflammatory cell infiltrate—aggregates of Diagnostic features:
cells, usually mononuclear cells, in absence of obvi-
ous hepatocellular necrosis.  Seen as a band of swollen and eosinophilic cells
 Infectious disease (MHV, Ectromelia, Clostridium intermediate to the central vein (zone III) and the por-
pilliforme, Helicobacter hepaticus, Parvo virus, tal triad (zone I).
Noro virus)—a spectrum of acute to chronic active  Nucleus undergoing lysis.
inflammation, degenerative, and proliferative changes  Two to three cells in thickness in the middle of the
specific for the infectious disease entity. lobule.

Comment: Some pathologists use focal for both focal and mul- Periportal (Figure 62)
tifocal, referring to the nature of the lesion rather than its actual
distribution. A severity grade can be used to reflect the multi- Hepatic necrosis in the periportal zone is observed follow-
focal nature of the lesion. Focal, multifocal, and subcapsular ing a variety of agents (e.g., phosphorus, ferrous sulphate,
necrosis is occasionally seen in untreated rodents and may be allyl alcohol) (Kanel and Korula 2005; Atzori and Congiu
a terminal event potentially due to hypoxic change secondary 1996; Sasse and Maly 1991). Affected cells may encircle the
to impaired blood flow. Subcapsular necrosis has also been portal tract (Popp and Cattley 1991) and may be associated
reported from direct pressure secondary to gastric distention with inflammatory and other changes (Ward, Anver, et al.
and from some types of restraint (Parker and Gibson 1995; 1994; Ward, Fox, et al. 1994; NTP Technical Report on the
Nyska et al. 1992) Toxicology and Carcinogenesis Studies of a Binary Mixture
2006; NTP Technical Report on the Toxicology and Carcino-
Necrosis, Zonal (Centrilobular, Midzonal, Periportal, genesis Studies of 2, 3, 7, 8-tetracholorodibenzo-p-dioxin
Diffuse) 2006).

Pathogenesis: Secondary to direct or indirect damage from Diagnostic features:


xenobiotic exposure; tissue anoxia.
 Swollen and/or eosinophilic hepatocytes may com-
pletely encircle the portal tract.
Centrilobular (Figures 54–59)
 Nucleus undergoing lysis.
Sometimes referred to as periacinar necrosis, it consists of  May be accompanied by periportal inflammation, fibro-
irreversible cell death of centrilobular hepatocytes and is often sis, bile duct proliferation, and oval cell hyperplasia.
18S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Diffuse (Figures 63 and 64) Pathogenesis: Duplication of nuclear material in absence of


cytokinesis. Variations in nuclear size and ploidy (karyome-
Synonym: Massive necrosis, panlobular necrosis.
galy and/or anisokaryosis) are common in aging rodents.
Diagnostic features:
Diagnostic features:
 Necrosis involving a large portion of a liver lobe.
 Hepatocytes with either two or more nuclei or with a
 May be associated with torsion of a liver lobe.
single enlarged nucleus which may be tetraploid or
 May be randomly distributed throughout the liver
octaploid.
without a specific lobular localization.
 Polyploid hepatocytes are frequently larger than adja-
cent diploid hepatocytes.
Differential diagnosis:
 Anisokaryosis is randomly distributed in the hepatic
lobule with more affected hepatocytes in the centri-
 Autolysis—loss of microscopic tissue structure and
lobular region.
stain affinity; pale eosinophilic staining and absence
of nuclear detail.
Differential diagnosis:
 Torsion of a liver lobe—Affects an entire liver lobe,
loss of microscopic structure.
 Hepatocellular neoplasia—mass or expansile prolif-
 Infarction—usually an angularly shaped wedge or
eration of hepatocytes with distortion or loss of lobu-
area of tissue necrosis; may be associated with a
lar architecture.
nearby thrombus.
 Hepatocellular hypertrophy (enzyme induction)—
increase in cytoplasmic volume not typically associ-
Comment: Zonal necrosis is typically associated with expo-
ated with increased nuclear size or number.
sure to xenobiotics that either directly damage hepatocytes
or cause damage following metabolic activation by endo-
genous or induced enzymes. There is often a concentration Comment: Karyocytomegaly is a reflection of hepatocyte
gradient within the hepatic lobule with more extensive lob- polyploidy that occurs when there is duplication of nuclear
ular involvement associated with higher doses of the toxic material in the absence of cytokinesis. The result is an increase
agent. in the number of diploid nuclei per hepatocyte or an increase in
Hepatocellular necrosis can occur spontaneously in rodents the ploidy level of a single hepatocyte nucleus. Polyploidy
or be induced by xenobiotics, toxins, or following treatment at increases with age in some strains of mice as well as following
high dosages with associated tissue anoxia, circulatory some treatment regimens resulting in hepatocytomegaly as
derangements, and biliary stasis. Necrosis (centrilobular, mid- well as karyomegaly (Harada et al. 1996). Variations in cell
zonal, periportal) might be accompanied by other histological size as well as in nuclei and polyploidy are also common in
changes (fatty change, congestion, hemorrhage, inflammation, aging rats of different strains. Karyomegaly and anisokaryosis
bile stasis, fibrosis, etc.) to form a myriad of pathological are normal incidental findings, especially in older mice (Percy
changes. The distribution might also cross certain zones and and Barthold 2001). Increase in cell size (cytomegaly) may
may manifest as ‘‘bridging necrosis’’ showing confluence of accompany the increase in hepatocyte ploidy. Anisokaryosis
the lesions (e.g., central to central veins, portal tract to portal (inequality in size of nuclei) is more common and dramatic
tract, or portal tract to central zones). Bridging necrosis may in mice than in rats.
ultimately give rise to bridging fibrosis. The development of polyploidy and its pattern vary among
A specific form of necrosis, ‘‘piecemeal necrosis,’’ is char- strains. C3H and DBA mice more commonly have octaploid
acterized by necrosis of the limiting plate of the portal tract at cells with two tetraploid nuclei in adult liver while NZB and the
the interface of hepatocytes and connective tissue of the portal out-bred strain NMRI at the corresponding age show a higher
tract, usually accompanied by inflammation, can be immune- proportion of diploid cells with strikingly low proportions of
mediated, and is seen in mice with resemblance to chronic tetraploid cells. Polyploidy has been observed in the early life
active hepatitis in man (Kitamura et al. 1992; Nonomura (three weeks) in Ercc1 null mice. This premature polyploidy in
et al. 1991; Kuriki et al. 1983). Ercc1-deficient liver is most likely caused by increased levels
of p21 in response to accumulating DNA damage (Chipchase,
O’Neill, and Melton 2003). Toxic injury caused by chemicals
Karyocytomegaly and/or Multinucleated Hepatocytes
such as phenobarbitone (Martin et al. 2001) and partial hepa-
(Figures 65 and 66)
tectomy also induce an increase in ploidy, usually associated
Synonyms: Karyocytomegaly, multinucleated hepato- with extensive but transient hepatocyte proliferation (Gerlyng
cytes, binucleated hepatocytes, karyomegaly, nuclear et al. 1993).
hypertrophy, hepatocytomegaly, polyploidy, anisonucleosis, Although anisonucleosis (polyploidy) is known to occur as
anisokaryosis. an age-related phenomenon, the nuclear and cellular changes
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 19S

can also be induced by xenobiotics (Schoental and Magee are often referred to as simple or multiloculated biliary cysts
1959; Jones and Butler 1975; Singh et al. 2007; Nyska et al. (Goodman et al. 1994). Polycystic liver can be observed in
2002; Guzman and Solter 2002; Lalwani et al. 1997; Travlos the rat (Muff et al. 2006; Sato et al. 2006) and hamster (Percy
et al. 1996; Kari et al. 1995; Herman et al. 2002). In addition, and Barthold 2001), resembling Caroli’s disease in humans
multinucleated cells (formed by cell fusion rather than divi- (Clemens et al. 1980; Numan et al. 1986; Serra, Recalde, and
sion) can be formed in rats after administration of 2, 3, 7, 8- Martellotto 1987). The cysts seen in polycystic disease are mul-
tetrachloro-dibenzo-p-dioxin (Gopinath, Prentice, and Lewis tiple and seen diffusely throughout the liver and are of variable
1987; Jones and Butler 1975). Eosinophilic cytoplasmic inclu- size but generally large compared to the smaller biliary cysts.
sions may be seen in affected hepatocyte nuclei because of cell
membrane invaginations.
C. Inflammatory Cell Infiltrates and Hepatic Inflammation
Cysts, Biliary (Hepatic Cysts) (Figures 67–69) (Hepatitis)
Pathogenesis: More common in aging animals occurs as a Introduction
dilation of biliary structures.
A variety of focal, multifocal, and more generalized infiltra-
Diagnostic features: tions of inflammatory cells are frequently present in liver tis-
sue. Changes range from acute inflammatory cell infiltrate(s)
 Range in size from small to very large. or occasional aggregates of lymphocytes/lymphohistiocytic
 Single or multiple cysts. cells/foci of mononuclear cells without associated alterations
 Macroscopically may contain clear to pale yellow of adjacent hepatocytes, to large panlobular patches of distinct
fluid. hepatocyte necrosis accompanied by polymorphonuclear and
 May occur anywhere in the liver and may be unilocu- mononuclear (lymphocytes, plasma cells, macrophages) cellu-
lar or multilocular. lar infiltrates. ‘‘Mononuclear cell’’ can be used when there is a
 Multilocular cysts are divided into variably sized mixture of cell types (lymphocytes; less often macrophages and
compartments by partial or complete connective plasma cells) or the cell type is mononuclear but cannot be
tissue septa. unequivocally identified in H&E stain. If a cell type predomi-
 Cyst walls are characteristically lined by flattened to nates, then the infiltrate should be classified as lymphocytic,
cuboidal epithelium. plasmacytic, or histiocytic. While etiological agents (e.g., bac-
 May be mild compression of adjacent hepatic teria, virus, parasite) may be present, in most safety assessment
parenchyma. studies the causes of significant inflammation are either cryptic
or are attributed to a specific treatment regimen. Inflammatory
Differential diagnosis: reactions in the liver may be accompanied by oval cell and
fibroblast proliferation and the propensity for hepatocellular
 Cystic degeneration—consists of markedly enlarged proliferative responses to replace lost parenchyma.
cells with finely flocculent pale eosinophilic cytoplasm. It is recommended that use of the diagnostic term
 Angiectasis (Peliosis hepatis)—dilated vascular spaces ‘‘inflammation’’ for the liver should be used sparingly. Liver
lined by endothelial cells; may contain blood cells. inflammation (hepatitis) is operationally defined as a constella-
 Bile duct dilation—dilated bile ducts lined by cuboi- tion of changes that represent a severe and generalized liver
dal epithelium; not multiloculated. reaction and would require multiple diagnostic terms to ade-
 Parasitic cyst—may have thickened wall and contain quately characterize (Figure 70). This type of reaction is not
parasite tissue. typically encountered in conventional rodent toxicity studies.
 Cholangioma—may cause compression of adjacent Traditionally, hepatic inflammatory responses have been
parenchyma and spaces lined by more endothelial classified as acute, subacute, chronic, granulomatous, and so
cells than in biliary cysts. on. These terms are somewhat interpretative, lack precise def-
initions, vary depending upon study duration, usually do not
Comment: Biliary cysts are commonly seen in older rats (Burek consist of a singular cell type, and do not have exclusive
1978; Greaves 2007; Harada et al. 1999). Solitary cysts can be pathognomic features. A more descriptive approach is recom-
observed without major adjacent morphology changes of the sur- mended and can be qualified by lesion distribution or use of
rounding tissue. However, depending on the location, adjacent subclassification and discretionary qualifiers (Figure 71).
parenchyma may contain pressure atrophy of the hepatic cords
of the liver, fibrosis, hemosiderin deposition, proliferation of bile
Infiltration, Inflammatory Cell
ducts, or periportal lymphocytic infiltration. Single cysts are
often caused by cystic dilatation of the intrahepatic bile ducts Pathogenesis: Infiltration of different inflammatory cells is
(Sato et al. 2005). Multiple cysts are observed also in hepatic typically a response to parenchymal cell death with causes
polycystic disease, where they occur alone or in combination ranging from infectious agents, exposure to toxicants, genera-
with polycystic kidney disease (Masyuk et al. 2004, 2007). They tion of toxic metabolites, and tissue anoxia.
20S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Infiltration, Neutrophil (Figure 72) Infiltration, Mononuclear (Figures 73–79)


Synonyms: Inflammation, acute; acute inflammatory cell infil- Synonyms: Inflammation, chronic; mononuclear cell aggregates;
trate(s), focus/foci of acute inflammatory cells; aggregate(s) of inflammation, granulomatous, focus/foci of mononuclear cells.
acute inflammatory cells.
Pathogenesis: Persistent noxious stimuli associated with infec-
Diagnostic features:
tion, toxic xenobiotics, continued low level parenchymal cell
death, and immune mediated effects.
 Predominantly neutrophilic (and in some specific
Specific subtypes may include: infiltration, lymphocytes;
types of experiments, eosinophilic) cells present as
infiltration, histiocytes (monocytes); infiltration, plasma cells.
focal aggregates often associated with dying hepato-
cytes or at the periphery of large areas of hepatocyte Diagnostic features:
necrosis. A few lymphocytes and occasional macro-
phages may be present.  Infiltrating cells include lymphocytes, plasma cells,
 Infiltrating cells are usually focal or multifocal. macrophages, and occasionally multinucleated giant
 Necrosis of scattered individual cells or small clus- cells.
ters of cells without associated infiltrating neutro-  Infiltrating cells may be focal, multifocal, or diffuse
phils may also be present in some areas. in distribution.
 Can be associated with whole regions of contiguous  Randomly distributed aggregates of mononuclear
affected lobules and confluent hepatic necrosis cells (primarily lymphocytes) without any accompa-
extending between adjacent lobules (bridging hepatic nying hepatocyte degeneration or necrosis may be
necrosis). common in older animals.
 Hemorrhage may be associated with larger lesions.  Periportal aggregates of predominantly lymphoid
 Oval cell proliferation may be present. cells in portal areas may be present.
 Evidence of liver cell necrosis may be minimal to
Differential diagnosis: mild with degeneration of scattered cells.
 Mononuclear cells with disruption of the limiting
 Foci of extramedullary hematopoiesis—mature and/ plate often infiltrate portal tracts.
or immature erythroid and myeloid cell aggregates  Bile duct hyperplasia may be present in some portal
without accompanying hepatocyte necrosis. areas.
 Granulocytic leukemia—hepatic parenchyma infil-  An increase in mononuclear cells within sinusoids
trated and replaced by a monomorphic population may be seen.
of neutrophils or neutrophil precursors.  Some capsular fibrosis and periportal fibrosis may be
 Chronic inflammation—inflammatory cells include present.
primarily mononuclear cell (largely lymphocytes,  There may be evidence of hepatocellular regeneration.
macrophages) and may include fibrosis and oval cell
hyperplasia. Differential diagnosis:

 Histiocytic sarcoma—hepatic parenchyma infiltrated


Comment: While neutrophilic infiltration in the liver is and replaced by collections of histiocytic cells that
primarily a response to liver cell injury and necrosis, a few can be pleomorphic.
lymphocytes or macrophages may also be present. In addi-  Lymphoma—hepatic parenchyma infiltrated and
tion, foci of neutrophils without apparent hepatocyte necrosis replaced by a monomorphic population of lymphoid
may be present, especially in situations of a transient effect cells.
on the liver. In florid reactions, areas of necrosis include  Early lymphoma may mimic age-associated focal
degenerating neutrophils admixed with the necrotic hepatic aggregates of lymphoid cells.
parenchymal cells.  Foci of extramedullary hematopoiesis—mature and/or
The extent of parenchymal cell death eliciting inflamma- immature erythroid and myeloid cell aggregates with-
tory cell infiltration varies from minimal microfocal lesions out accompanying hepatocyte necrosis.
to large patches of coagulation necrosis encompassing multi-  Viral hepatitis—may be difficult to distinguish from
ple contiguous lobules. For xenobiotic-induced cell death and chronic (active) inflammation; evidence of a viral
inflammation, the severity of the lesions is often a function of etiology can be supportive.
the dose of the hepatotoxicant. Apoptotic cell death may
occur along with conventional necrosis. Depending upon Comment: Mononuclear cell infiltration spans a wide spec-
etiology, acute inflammation can have a specific lobular dis- trum of morphological features and severities depending upon
tribution with the possibility of portal or centilobular bridging the extent and duration of liver damage and any ongoing
between adjacent lobules. regenerative response. In distinction from acute inflammation,
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 21S

there are typically fewer neutrophils seen in chronic lesions and system is active. Others may consider chronic active inflamma-
there is more involvement of portal areas with infiltrating mono- tion simply as a form of chronic inflammation with areas of
nuclear cells and disruption of limiting plates. Granulomatous neutrophilic infiltration in the inflammatory process and
inflammation with characteristic focal or multifocal nodular col- prefer to address that in their accompanying pathology narra-
lections of mononuclear epitheloid cells and occasional giant tive. A combined neutrophilic and mononuclear infiltrate
cells associated with areas of hepatocyte necrosis can be consid- (chronic active inflammation) is a common response found in
ered a specific subtype of mononuclear cells infiltration and part mice chronically infected with Helicobacter hepaticus.
of the spectrum comprising chronic inflammation of the liver
(Figures 74–77). Some pathologists consider granulomatous Infiltration, Peribiliary (Intrahepatic) (Figure 81)
inflammation of the liver to be a form of chronic inflammation Pathogenesis: Age associated change that may be exacerbated
when there is a prolonged duration with predominant presence of by treatment.
epitheloid cells/macrophages and prefer to address the fact that
there may be areas containing mononuclear epitheloid cells and Diagnostic features:
multinucleated giant cells in the chronic inflammatory process in
an accompanying narrative. Fatty material and cholesterol  Peribiliary aggregates of inflammatory cells affecting
deposits may accumulate in some granulomatous inflammatory a few to many portal areas.
reactions. Such localized accumulations of abundant cholesterol  Sometimes accompanied by fibrosis.
have been referred to as cholesterol granulomas (Figures 78 and  May be associated with some degree of bile duct
79). hyperplasia.
When only few isolated collections of mononuclear cells are
Differential diagnosis:
present, some pathologists may diagnose them as focal mononuc-
lear cell aggregates. These focal accumulations are considered
 Cholangiofibrosis—proliferative and metaplastic
by some to be a background lesion, and for these aggregates
biliary response plus fibrosis extending into the hepa-
using the cell type in the diagnosis, instead of inflammation or
tic parenchyma; may be subcapsular.
inflammatory cell infiltrate, may be preferable and less mislead-
ing. The frequency of these mononuclear cell aggregates may
Comment: A minimal to moderate peribiliary inflammatory
be exacerbated by treatment. Helicobacter sp. and murine noro-
cell infiltration consisting primarily of mononuclear cells can
virus infections in mice may cause these incidental lesions.
occur commonly in the livers of rats and mice and increases
in incidence with animal age. Persistent obstruction to biliary
Infiltration, Mixed (Figure 80)
flow may also lead to bile duct inflammation in hepatic portal
Synonym: Infiltration, purulent, and mononuclear; chronic areas. Although this background lesion may be considered a
active inflammation; mixed inflammatory cell focus/foci. subtype of mononuclear cell infiltration (see previous), it is fre-
Diagnostic features: quently diagnosed separately when exacerbated by treatment.

Fibrosis (Figures 82 and 83)


 Infiltration of mononuclear cells along with
neutrophils. Pathogenesis: A reaction to acute or prolonged hepatotoxicity.
 Areas of active hepatocellular degeneration and/or Diagnostic features:
necrosis often present.
 Evidence of hepatocellular regeneration may be present.  The presence of connective tissue in the liver above
 Intralobular distribution is often random. the normal low rate seen in portal areas.
 May have diagnostic features common for both neutro-  Peribiliary fibrosis particularly in aged rats.
philic and mononuclear infiltration (see previous).  Three patterns of fibrosis are seen in the rodent liver:
Differential diagnosis: pericellular, peribiliary, and postnecrotic. The peri-
cellular pattern is most common in mice.
 Histiocytic sarcoma—hepatic parenchyma infiltrated  Normal hepatic architecture maintained.
and replaced by collections of histiocytic cells that  Increased prominence of fibrosis, which may sur-
can be pleomorphic. round hepatic lobules and bridge between adjacent
 Lymphoma—hepatic parenchyma infiltrated and portal areas.
replaced by a monomorphic population of lymphoid  Hyperplastic nodules of hepatocytes separated by
cells. septae of connective tissue can be seen in rats but is
less common in mice.
Comment: Some pathologists consider a combined neutrophilic  Oval cell proliferation often spreading from peripor-
and mononuclear inflammatory cell infiltration as chronic tal areas.
active inflammation. This type of response is suggestive that  Masson trichrome, van Gieson, or Silver stains can be
the adverse stimulus is still present and/or that the immune used to delineate fibrosis.
22S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Differential diagnosis: Diagnostic features:

 Cholangiofibrosis—marked pericholangial fibrosis  Focal to multifocal areas of necrosis with or without


with proliferation of biliary epithelium and metaplas- inflammation.
tic changes of glandular epithelium (e.g., goblet cells,  Silver stain positive helical to rod-shaped bacteria in
Paneth cells). or adjacent to the lesions, often can be seen between
 Regenerative hyperplasia—nodular growth pattern; hepatocytes (because organisms are in the bile
distorted lobular pattern. caniculi).
 Multiple foci of cellular alteration—discrete collec-  Chronic lesions—focal, multifocal, to diffuse may
tions of cells within the hepatic parenchyma. include inflammatory cell foci, hepatocytomegaly,
 Multiple hepatocellular adenomas. oval cell hyperplasia, and cholangitis.
 Scirrhous carcinoma—neoplastic epithelial cells will  Oval cell hyperplasia, focal to diffuse, minimal to
be embedded in a collagenous connective tissue pro- marked.
liferative response.
Differential diagnosis:
Comment: The pattern of fibrotic response to chronic
injury varies among the species. When hepatic fibrosis is  Inflammation, focal or multifocal, acute or chronic,
accompanied by a nodular or non-nodular regenerative unknown etiology.
response in the liver, it may be considered by some to repre-  Murine norovirus infection.
sent cirrhosis. Classical cirrhosis is rare in rodents in contrast  Tyzzer’s disease.
to dogs and humans and cannot reliably or consistently sep-
arate from a robust fibrotic reaction with associated nodular
regeneration, oval cell proliferation, and bile duct hyperpla- Comment: A number of different Helicobacter spp. have been
sia. Such a robust fibrosis can be induced in rodents with pro- identified that can affect the rodent liver spontaneously
longed or repeated exposure to certain chemicals (carbon (Ward, Anver, et al. 1994; Ward, Fox, et al. 1994; Goto
tetrachloride, alcohol, tetrachlorovinphos, diallylphthalate), et al. 2000; Zenner 1999). Helicobacter spp. can cause an
dietary lipotrope deficiency, or chronic hepatitis secondary increase in hepatocellular tumors in certain strains of infected
to persistent infection (Ward 1997). We see no advantage mice, but are also known to generate liver lesions (Ward, Fox,
in calling severe hepatic reaction cirrhosis in contrast to a et al. 1994; Tian et al. 2005; Rogers and Fox 2004) and can
diagnosis of hepatic fibrosis with an appropriate severity promote experimental carcinogenesis of the liver (NTP Tox-
grade. The specific morphological features of this response icology and Carcinogenesis Studies of Theophylline 1998;
can easily be addressed in the pathology narrative. It should Zenner 1999; Diwan et al. 1997) in rodents. The pathogenicity
be noted that hepatocellular neoplasms might arise in severe of the bacteria can vary with strain of bacteria and mouse
hepatic fibrosis. strain, stock or line.
Helicobacter hepaticus may cause acute to subchronic
minimal to severe lesions in livers of susceptible mice such
D. Infectious Diseases
as A strain, C3H, and BALB/c (Ward, Anver, et al. 1994).
Introduction Many mouse strains are resistant to liver infection but A
strain mice are the most susceptible (Ward, Anver, et al.
Infectious diseases of the mouse liver are an important
1994; Ward, Fox, et al. 1994). Incidental findings of focal
group of conditions that may interfere with toxicology and
or multifocal necrosis in the liver with or without inflamma-
carcinogenesis studies. The histological changes associated
tory cells such as macrophages, lymphocytes, and neutro-
with the diseases described in the following may be recorded
phils can be seen. In severely affected livers, more severe
using the nomenclature described previously under inflamma-
chronic lesions can be observed. H. hepaticus hepatic
tion and inflammatory cell infiltrates but they are also pre-
lesions are more common in males than females and inci-
sented here as a separate category of disease diagnoses to
dence is increased in mice six months of age or older (Percy
help pathologists diagnose the infections, which can be con-
and Barthold 2001). H. bilis may also cause mild hepatic
firmed by PCR, immunohistochemistry, and other diagnostic
lesions.
studies. Some major infectious diseases are mentioned in the
It is rare for H. hepaticus to cause liver lesions unless
following to separate these background lesions from xenobio-
the facility and animal room are known to be infected. If
tic induced lesions.
susceptible mouse strains are used in research, the more
severe diffuse lesions may occur. Mouse infection in two-
Helicobacter sp. Hepatitis (Figure 84; See Figure 87)
year carcinogenesis bioassays has complicated interpreta-
Pathogenesis: Infection by a number of different Helico- tion of carcinogenesis studies (Hailey et al. 1998; Stout
bacter spp. et al. 2008).
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 23S

Murine Norovirus Hepatitis (See Figure 73) Comment: Mouse hepatitis virus (MHV), a coronavirus, infects
hepatocytes, endothelium, and macrophages. In mouse liver,
Pathogenesis: Infection by murine norovirus.
virus strains may have different pathogenicities in the
Diagnostic features: various mouse strains and in mice of different ages (Percy
and Barthold 2007). Focal and multifocal necrosis is seen
 Focal, multifocal to diffuse areas of inflammation with multinucleated (syncytial) hepatocytes, endothelium, and
sometimes with vasculitis in immunodeficient mice. macrophages. Immunodeficient mice may develop a chronic
 Focal to multifocal small areas of inflammation in persistent infection with chronic lesions in liver (Ward, Collins,
most lines of mice. and Parker 1977).
 Inflammatory foci contain macrophages and Clinical MHV infection may be most commonly seen in
lymphocytes. young mice. Adult mice may have serum antibodies but no
 Immunohistochemistry shows that inflammatory clinical signs and few, if any, histopathologic lesions. Some
cells, especially macrophages in lesions and Kupffer cases show lesions in liver only. MHV is one of the most
cells, can be positive for murine norovirus antigens. prevalent murine viruses in the United States and Europe
(Homberger 1996) but appears less common today.
Differential diagnosis:
Tyzzer’s Disease (Clostridium piliforme Infection)
 Helicobacter hepatitis can appear similar but may
Pathogenesis: Infection by Clostridium piliforme (Bacillus
contain silver positive bacteria.
piliforme).
 Helicobacter hepatitis is more common in A, BALB/
c, and C3H strains that are not susceptible to MNV Diagnostic features:
infection.
 Inflammation, focal or multifocal, acute or chronic,  Focal or multifocal necrosis, coagulation to caseous
unknown etiology. with neutrophilic infiltration.
 Intrahepatocyte bundles of long basophilic bacilli.
Comment: Murine norovirus (MNV) may cause severe hepa-  Bacteria seen in Warthin-Starry, Giemsa stains, or by
titis in some lines of immunodeficient mice but only minimal the PAS method.
hepatitis or no lesions in most lines of infected immunocom-
petent mice (Ward et al. 2006). MNV infection is the most Differential diagnosis:
common viral infection in mouse colonies today. The impli-
cations for interfering in experimental results are not known.  Necrosis, of other known causes or unknown causes.
It can be assumed that chemicals or infectious agents involv-
ing the immune system or liver may be influenced by MNV Comment: Named after Ernest Tyzzer who first described it
infection. in a colony of Japanese walzing mice (Fox et al. 2002). Clos-
tridium piliforme (Bacillus piliformis) is a long, thin, spore-
forming (intracellular) bacterium. Rare lesion with sudden
Mouse Hepatitis Virus Hepatitis (See Figures 63, 64, and 70) death, with or without diarrhea. Often infection of the colon
with dissemination to the liver (focal hepatitis) and occasion-
Pathogenesis: Infection by mouse hepatitis virus affecting
ally heart (myocarditis). Special stains (e.g., Giemsa or
hepatocytes, endothelium, and macrophages (Kupffer cells).
Warthin-Starry Silver stain) can reveal intracytoplasmic fila-
Diagnostic features: mentous bacteria.
Tyzzer’s disease is rare in rodents. It is usually sporadic.
 Focal or multifocal hepatic necrosis. The gerbil is known to be extremely susceptible to infection
 Multinucleated cells (syncytia) from hepatocytes, (Fox et al. 2002).
endothelium, and macrophages. Gross lesions can include hepatomegaly, focal necrosis, sin-
 MHV lesions in other tissues, including peritoneum, gle or multifocal, small or large lesions sometimes associated
CNS, blood vessels. with lesions in other tissues, especially intestines (Percy and
 Chronic hepatitis and postnecrotic cirrhosis in immu- Barthold 2007). Multiple foci of necrosis (coagulative necro-
nodeficient mice. sis) and/or multifocal necrotizing hepatitis can be observed
microscopically.
Differential diagnosis:
E. Vascular Lesions
 Necrosis caused by toxins.
Introduction
 Necrosis caused by murine norovirus.
 Necrosis caused by Helicobacter sp. The liver has a dual blood supply consisting of a relatively
 Necrosis caused by C. piliforme. major (about 75%) venous (portal) supply via the hepatic portal
24S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

vein, which carries venous blood that is largely depleted of Angiectasis (Figures 86 and 87)
oxygen, and relatively minor (about 25%) arterial blood sup-
Synonyms: Peliosis hepatis, telangiectasis, sinusoidal dilation.
ply, via the hepatic artery. The portal blood contains toxic
materials absorbed in the intestine, and therefore the liver is the Pathogenesis: Perturbations in blood flow and/or drainage;
first tissue to be exposed to toxic substances that have been weakening of sinusoidal walls.
absorbed through the gastro-intestinal tract.
Diagnostic features:
Within the hepatic parenchyma, the hepatocytes are in
intimate contact with the sinusoidal capillaries, which are car-
 Macroscopically—seen on the surface as blood-
rying the mixture of blood originating from ramifications of
filled, thin walled cavities projecting above the sur-
the portal vein and hepatic artery to the central vein. The
face (Bannasch et al. 1997).
sinusoids are lined by modified endothelial cells containing
 Microscopically—There are two morphological
fenestrations, which allow passage of lipoproteins and other
types, as follows:
large molecules but provide a barrier to blood cells. Kupffer
1. Cystic (‘‘Phlebectatic’’)—Focal dilation (disten-
cells reside in the lumen of the sinusoids and are anchored
sion) of endothelial lined channels (photographic
to their wall.
presentation contributed by Hardisty et al. 2007).
The morphological aspect of the hepatic pathology during
Can be an isolated lesion or have a multicentric
circulatory disorders depends on the location of the vascular
form. The lacunae are densely packed with blood
structure being affected (i.e., lobular sinusoids, the outflow
and are coated by a single layer of endothelium
hepatic vein, or the inflow portal vein).
and separated from one another by cords of liver
parenchyma (Bannasch, Wayss, and Zerban
1997). The endothelial cells appear to be unal-
Congestion (Figure 85)
tered, and there is no increase of mitotic figures.
Synonym: Chronic passive congestion. The tissue adjacent to the dilated sinusoids is
well preserved and free of necrotic cells.
Pathogenesis: Circulatory failure, typically right-sided heart 2. Cavernous (‘‘Parenchymal’’)—The peliotic cysts
failure. are not, or are only partially, lined by endothelium.
Thus, the cysts involve not only the sinusoidal
Diagnostic features:
lumen, but also the space of Disse, and the blood
comes in direct contact with the neighboring par-
 Increase prominence (number) of erythrocytes in the
enchymal cells. The parenchyma undergoes focal
capillary bed or larger vessels of an organ.
necrosis without a zonal distribution. This lesion is
 No appreciable distention (angiectasis) of the vessel
much less likely to be preneoplastic and a few tox-
wall.
ins have been shown to induce the lesion. When
 Diagnosis often correlates with gross observation
endothelial lining is absent, the term Peliosis
(e.g., reddened, darkened focus).
Hepatis instead of Angiectasis is indicated.
 May be associated with centrilobular necrosis.
Differential diagnosis:
Differential diagnosis:
 Hemangioma—expansile structure lined by flattened
 Angiectasis—dilated vascular spaces lined by endothelial cells; may be associated with parenchy-
endothelial cells; dilated vascular channels and mal compression.
spaces frequently contain erythrocytes.  Cystic degeneration (Spongiosis hepatis)—cavities
 Massive necrosis. are not filled with blood but with a finely flocculent
 Hemorrhage-irregular patchy lakes of blood not con- acidophilic material.
tained within defined vascular channels.
 Autolysis-altered cellular texture and loss of staining Comment: Angiectasis is a cystic or cavernous widening of the
intensity. liver sinusoids that can occur in a variety of pathological
insults. In human, sinusoidal dilatation has been reported fol-
Comment: Congestion may result from circulatory disturbance lowing hypoxia or hyperperfusion as a result of right-sided
such as right-sided heart failure and is usually associated with heart failure, thrombosis of hepatic veins, amyloid deposition,
necrosis of the centrilobular areas (Burt, Portmann, and granulomatous, or neoplastic disease (Greaves and Faccini
MacSween 2002). Presence of blood in hepatic sinusoids seen 1992; Bruguera et al. 1978). Although these lesions can also
in animals that die or in situations where there is incomplete occur spontaneously in different rodents with different diseases
exsanguination should not be diagnosed as congestion. If it is or with certain neoplasms causing hemodynamic changes, it
absolutely necessary to record such findings, it is important can also be induced by different compounds. Focal sinusoidal
to qualify the blood stasis in the liver as passive congestion. dilatation and peliosis hepatis have been observed in the rodent
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 25S

liver after treatment with nitrosamines, pyrrolizidine alkaloids Comment: There are several potential mechanisms leading to
or glucocorticoids (Greaves 2007; Ruebner, Watanabe, and liver thrombosis. Activation of the coagulation system associ-
Wand 1970; Ungar 1986; Wolstenholme and Gardner 1950). ated with fibrin deposits and hypoxia located in the centrilob-
Altered hemodynamic and changes in the hepatic microcircula- ular sinusoids was reported to occur in the livers of rats
tion have been proposed to be of importance in the pathogen- exposed to monocrotaline (MCT) (Copple et al. 2002). It was
esis of sinusoidal dilatation (Slehria et al. 2002). Subcapsular suggested that the fibrin thrombi were formed following
sinusoidal dilatation can also be found a postmortem finding chemical-induced hepatic endothelial cell damage. In studying
in rats (Kimura and Abe 1994). an endotoxin-exposure model, it was suggested that the noted
Angiectasis is defined by multiple blood-filled cystic spaces focal and random hepatocellular necrosis was caused by circu-
of different size and shape occurs after suspected loss or weaken- latory disturbances due to fibrin thrombi in clusters of adjacent
ing of sinusoidal walls and/or supporting tissue. The two subtypes sinusoids. Using a rat model of 2-butoxyethanol induced hemo-
may occur in combination. The cystic spaces are devoid of lytic anemia associated with systemic thrombosis, fibrin
endothelial lining in the cavernous subtype (‘‘parenchymal’’) thrombi were noted in the central vein and sinusoids of the
although controversy still exists (Greaves 2007; Edwards, liver, in addition to the presence of thrombi seen in several
Colombo, and Greaves 2002). Angiectasis can be found in aged other organs (Ramot et al. 2007).
rats (Lee 1983). However, these lesions can also be induced in
both rats and mice after viral infection (Bergs and Scotti 1967) Infarction (Figures 89 and 90)
or exposure to certain drugs or chemicals (Mendenhall and Pathogenesis: Interruption of blood flow in a major vessel.
Chedid 1980; Husztik, Lazar, and Szabo 1984). Torsion of hepatic lobe.
Angiectasis has also been reported in animals or humans
Diagnostic features:
infected with Bartonella spp. (Wong et al. 2001; Breitschwerdt
and Kordick 2000) and it has been associated with a number of
 Extensive area of necrosis may be associated with
diseases in humans as well as administration of anabolic steroids
inflammation.
and oral contraceptives (Naeim, Cooper, and Semion 1973;
 The necrosis does not have acinar pattern.
Zimmerman 1998; Tsokos and Erbersdobler 2005).
Angiectasis may refer to a vascular lesion formed by dilatation Differential diagnosis:
of a group of small blood vessels. It can be observed in transgenic
mouse models (Srinivasan et al. 2003; Bourdeau et al. 2001) or  Hepatocellular necrosis resulting from direct toxicity
related to xenobiotic administration in rats and mice (Robison may have a diffuse or lobular distribution but also may
et al. 1984; Kim et al. 2004). It can be found as an incidental find- be accompanied by infarction; not mutually exclusive.
ing in aging mice and is sometimes associated with hepatocellu-
lar neoplasms (Harada et al. 1996, 1999). Angiectasis can be Comment: Aside from torsion of liver lobes, which can occur
chemically induced (Bannasch, Wayss, and Zerban 1997) and spontaneously in rodents, infarction is a very rare lesion that
has been suggested to be preneoplastic in some animal models. can be induced only under very specific experimental condi-
tions. The combined injection in mice of NG-monomethyl-L-
Thrombosis (Figure 88) arginine and aspirin after lipopolysaccharide exposure resulted
in significant hepatocellular enzyme release, characterized his-
Pathogenesis: Activation of the coagulation system associated tologically by intravascular thrombosis with diffuse infarction
with arteritis or phlebitis or secondary to atrial thrombosis. and necrosis (Harbrecht et al. 1994). Intraperitoneal injections
Diagnostic features: in rats of vasoconstrictor xenobiotics such as phenylephrine
produced infarcts of the spleen regularly and infarcts of the
 It is characterized by the formation of a thrombus liver occasionally (Levine and Sowinski 1985). Isolated perfu-
within the lumen of a blood vessel, like sinusoids and sion with 1.0 g/kg of the cytotoxic xenobiotic 5-FU or
central veins. hyperthermia of 41 degrees C  10 min resulted in 90% to
 Amorphous mass attached to the endothelium or free 100% mortality in rats, with extensive, patchy necrosis, and
within the blood vessel lumen (due to plane of section). infarction on histologic examination (Miyazaki et al. 1983).
 Contains fibrin, platelets, and entrapped blood cells.
 Damaged endothelium can be seen. Endothelial Cell Hypertrophy/Karyomegaly3 (Figure 91)
 May occur with histiocytic sarcoma in rats or mice. Synonyms: Endothelial cell enlargement, cytomegaly.

Differential diagnosis: Introduction: This is a relatively new diagnostic entity.


Because of the difficulty of identifying specific sinusoidal cell
 Postmortem clot.
 Necrotic area of tissue—loss of microscopic tissue 3
The examples provided were confirmed with special stains (not shown).
structure and stain affinity; pale eosinophilic stain- However, based solely on H&E staining, a diagnosis of ‘‘sinusoidal cell
ing, and absence of nuclear detail. hypertrophy/karyomegaly’’ is appropriate.
26S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

types from H&E-stained sections, definitive diagnosis of Hyperplasia, Endothelial


endothelial cell hypertrophy/karyomegaly requires confirma-
Synonyms: Endothelial cell hyperplasia, angiomatous
tion using special stains.
hyperplasia.

Pathogenesis: Continued DNA synthesis and cell cycle arrest Pathogenesis: Proliferation of normally present sinusoidal lin-
following exposure to certain xenobiotics. ing endothelial cells without sinusoidal dilation.

Diagnostic features: Diagnostic features:

 Irregularly shaped karyomegalic nuclei.  Focal, located to the portal triad, consisting of
 Nuclei and cells appear larger than normal. increased number of capillaries.
 Secondary changes related to the endothelial  Blood may not be present within the capillaries.
cell damage and/or obstruction of the sinusoids lead-  Absence of supporting tissue.
ing to local ischemia may be seen. Such changes  May have mitotic figures and nuclear atypia.
include sinusoidal congestion, hemorrhage, thrombi
formation, hepatocytic fatty change/degeneration, Differential diagnosis:
and necrosis (Lailach et al. 1977; Nyska et al. 2002).
 Hemangioma—expansile structure lined by flattened
Differential diagnosis: endothelial cells; may be associated with parenchy-
mal compression.
 Endothelial cell hyperplasia characterized by  Angiectasis—dilated vascular spaces lined by
increased number of endothelial cells. endothelial cells; dilated vascular channels and
 Hepatocytic cytomegaly and karyomegaly. spaces frequently contain erythrocytes.
 Anisokaryosis—variation in the size of nuclei and/or
binuclearity admixed with diploid appearing hepato- Comment: Endothelial derivation and proliferation can be
cyte nuclei. confirmed by dual immunostaining for CD31/KI-67 (Ohnishi
 Reactive Kupffer cells—enlarged histiocytic cells et al. 2007). Comparative endothelial cell kinetic studies in
with visible cytoplasm lining sinusoids. human, mice, and rats indicated that the labeling index (LI)
 Histiocytic sarcoma—hepatic parenchyma infiltrated in the male and female B6C3F1 mouse liver was significantly
and replaced by collections of histiocytic cells that higher (p < .01) compared to the LI in male and female rat
can be pleomorphic. and human liver, and the LI in the male and female rat liver
was significantly greater (p < .05) than the LI in human liver.
It was suggested that the increased rate of spontaneous
Comment: In a study of monocrotaline (Wilson et al. 2000) it hemangiosarcoma formation in mice may be related to the
was suggested that the endothelial karyomegaly was the result increased proliferation rate of endothelial cells normally
of continued DNA synthesis and concentration-dependent cell- present in the B6C3F1 strain of mice compared to rats and
cycle arrest. The exposed cells undergo a process of multiplica- humans (Ohnishi et al. 2007).
tion of chromosomal copies, defined as endopolyploidy, with
nuclear and cytoplasmic gigantism. A direct correlation
between cytoplasmic volume and nuclear DNA content was F. Non-Neoplastic Proliferative Lesions
suggested.
Introduction
A similar pathogenesis was suggested in the case of
ridelliinne-induced endothelial cytomegaly and karyomegaly A variety of non-neoplastic proliferative lesions of different
(Nyska et al. 2002). Administration of riddelliine, a naturally origin(s) occurs spontaneously in the liver of rodents and may
occurring pyrrolizidine alkaloid, to rats results in cytomegaly and also be induced by treatment with chemicals. Incidences and
karyomegaly of hepatic endothelial cells as one of its pleotrophic morphological characteristics vary considerably by animal spe-
responses to cell-specific cytotoxicity. A metabolite of this cies, strain, and sex. Some of these lesions might be regarded as
pyrrolizidine alkaloid is believed to directly interact with pre-neoplastic alterations.
endothelial cell DNA. As a non-neoplastic proliferative response, increased
Sometimes this change is interpreted as prominent hepatocyte mitoses (Figure 92) above normal background
Kupffer cells or Kupffer cell hypertrophy on H&E sections levels or increased above what is seen in control animals can
and warrants further characterization for confirmation of occur in rodent livers. Causes vary from physiological
the endothelial cell origin. Immunohistochemical stains responses such as during early growth, during pregnancy, and
and electron microscopy can be used to identify the cell type following partial hepatectomy to post-necrotic repair. This
involved. change may be diagnosed as cytologic alteration or mitotic
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 27S

alteration with a description in the pathology narrative.  Portal areas and central veins not present in small foci
In some laboratories diagnosis of ‘‘increased mitoses’’ is but can be seen in larger foci.
used.  Sinusoids within focus may be compressed, so
typical parenchymal plates are difficult to detect.
 Tortuous hepatic cords may occur due to increased
Focus of Cellular Alteration number of cells.
 Size of cells and cytoplasmic tinctorial variation
Introduction: Foci of cellular alteration are common in rodent
depend on type of focus.
studies greater than duration of twelve months and may be
 Normally absence of cellular atypia.
seen in short duration toxicity studies following exposure
 Cystic degeneration (spongiosis hepatis) and
to certain xenobiotics. Foci of cellular alteration can be iden-
angiectasis (peliosis hepatis) may occur within foci
tified by special stains. In H&E-stained slides they may be
of altered cells.
subclassified based on the predominant cell type present.
 Cytoplasmic lipid may be present.
Diagnosis of the mixed cell subtype of altered hepatic focus
 Intracytoplasmic inclusions of various types may be
varies among different laboratories. One viewpoint defines a
present.
mixed focus of cellular alteration as consisting of a combina-
tion of basophilic, vacuolated, eosinophilic, and/or clear cell
hepatocytes without a predominant cell type. An alternative
viewpoint defines a mixed cell focus as containing any two Basophilic (Figures 93–97)
phenotypes of cells in approximately a 50%/50% proportion.
Basophilic, diffuse (Figures 93 and 94).
Others regard a ‘‘true’’ mixed focus as one that contains
clearly identified basophilic and eosinophilic cells regardless  Hepatocytes of normal size or slightly enlarged with
of the proportions of each. Because of this diverse set of homogeneously staining cytoplasmic basophilia due
diagnostic opinions regarding focus subtypes, the pathologist to abundant free ribosomes.
is encouraged to describe the morphological features  Cells may be pleomorphic with enlarged vesiculated
of documented foci in detail, especially if they are altered nuclei and prominent nucleoli.
by treatment.  Dissociation of cells may occur.
 Mitotic figures may occur.
Synonyms: Areas of cellular alteration; focus of altered
Basophilic, tigroid (Figure 95).
hepatocytes; hyperplastic focus; preneoplastic focus; enzyme
altered focus; phenotypically altered focus.  Cells are usually smaller; enlarged cells may
occur.
Pathogenesis: A localized proliferation of hepatocytes pheno-  Cells are often arranged as tortuous cords.
typically different from surrounding hepatocyte parenchyma.  Cells display large abundant basophilic bodies often
arranged in clumps or long bands with striped pattern
Diagnostic features: in paranuclear or peripheral regions of cytoplasm
(due to increased rough endoplasmic reticulum).
 May occasionally be observed grossly as small white
 Mitotic rate may be increased.
foci on the liver surface, but not round nodules.
 Size may range from less than one lobule to multiple Basophilic, NOS (Figure 96).
lobules in diameter.
 Foci that are not clearly tigroid or diffusely
 Circular or ovoid shape; irregular formed foci may
basophilic.
occur.
 Peliosis or spongiosis may occur within these foci.
 Distinguished into types of foci by virtue of tinctorial
variation, size of hepatocytes, and textural appear- Basophilic (no further classification in mice) (Figure 97).
ances from surrounding parenchyma.
 May be subclassified based on predominant cell type.  Consist of cells larger or smaller than normal hepato-
The fact that 80% of the focus is composed of one cytes, in general they are smaller.
morphologic cell type (basophilic, eosinophilic, etc.)  Cytoplasm exhibits distinct basophilia due to free
or a mixed cell type. ribosomes or rough endoplasic reticulum.
 Normally no or only minimal compression of the sur-  Often cells contain obvious glycogen.
rounding liver tissue.  Intracytoplasmic basophilic clumps with relatively
 Liver plates merge imperceptible with surrounding clear intervening cytoplasm or the cytoplasmic baso-
hepatic parenchyma; nevertheless foci are sharply philia may be distributed homogeneously.
demarcated from the adjacent normal hepatocytes  Vascular pseudo-invasion may be present.
by the appearance and staining reaction of its cells.  Eosinophilic cytoplasmic inclusions may be found
 Lobular architecture preserved. occasionally within hepatocytes.
28S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Eosinophilic (Figures 98–100) the surrounding parenchyma. Distinct compression


of adjacent normal hepatocytes is present.
Synonyms: Acidophilic, ground glass.
 Carcinoma, hepatocellular—distinct trabecular,
adenoid or poorly differentiated growth is present.
 Composed of usually enlarged, polygonal hepato-
Lobular and plate architecture is not maintained.
cytes with acidophilic staining cytoplasm.
Cellular atypia and invasive growth may be present.
 Some clear cells may be present.
 Cytoplasm is distinctly granular and pale pink, inten-
Comments: Foci of cellular alteration represent a localized pro-
sely eosinophilic, or shows ground-glass appearance.
liferation of hepatocytes that are phenotypically different from
 Nuclei often enlarged; nucleoli may be prominent
surrounding hepatocyte parenchyma. They are subclassified
and centrally located.
based upon phenotypic and tinctorial features. Foci of cellular
 Eosinophilia may result from an increase in smooth
alteration can occur spontaneously in aged rats and other
endoplasmic reticulum, peroxisome, or mitochondria
rodents and can be induced by chemical treatment. The inci-
(rats, mice).
dence, size, and/or multiplicity of foci are usually increased
 Store glycogen in excess.
and time to development usually decreased after administration
of hepatocarcinogens (Hanigan, Winkler, and Drinkwater
Mixed Cell (Figures 101–104)
1993; Frith, Ward, and Turusov 1994; Bannasch and Zerban
Synonyms: Basophilic/eosinophilic mixed. 1990; Moore, Thamavit, and Bannasch 1996). Foci of cellular
Heterogeneous focus consisting of a combination of cell alteration are not necessarily preneoplastic. Foci of cellular
types (see Introduction above). alteration may have prominent fat deposition and characteristic
features of cystic degeneration and angiectasis (See B-focus
Clear Cell (Figures 105–107) with spongiosis; Figure 96).
Foci can be subclassified based on the predominant cell
 Composed of normal-sized or enlarged cells with dis-
type. If no single cell type comprises at least 80% of a given
tinct cytoplasmic clear spaces.
focus, it should be classified as mixed. Mostly these mixed foci
 Some eosinophilic or basophilic cells may be present.
consist of both basophilic and eosinophilic/clear type cells. In
 Nuclei are often small and dense, prominent and
these foci, it is not clear what is the predominant phenotype and
centrally located, sometimes exhibiting increased
are therefore indicated as ‘‘mixed.’’
volume.
Species and strain differences occur in the prevalence of
 Store glycogen in excess.
these foci. It is not uncommon for a focus predominantly
 Cell membranes may appear prominent.
of one cell type, however, to have a small number of a
different type.
Amphophilic (in Rats and Mice) (Figure 108)
Mixture of eosinophilic and clear cells can be classified into
 Hepatocyte cytoplasm having an affinity both for either eosinophilic or clear cell focus in accordance with pro-
acid and for basic dyes. portion of clear cells. Mixture of amphophilic and other pheno-
 Large cells with homogeneous, granular intensely types has never been observed in rodents.
eosinophilic cytoplasm, and randomly scattered faint Usually, amphophilic foci are less frequently observed in
basophilia. rats and mice compared to other foci.
 Nuclei slightly enlarged. A number of models have linked specific types of foci of
 Poor in or free of glycogen. cellular alteration with carcinogenesis (Mahon 1989). The
 Amphophilic (basophilic and eosinophilic) cytoplasm nitrosomorpholine model is linked with eosinophilic and clear
due to a proliferation of both mitochondria and rough cell foci as precursors. Aflatoxin is linked with basophilic foci
endoplasmic reticulum (mitochondrial-RER complex). as a tumor precursor (Bannasch, Zerban, and Hacker 1985).
 Usually less frequently observed in rats and mice It was also reported that hepatocarcinogenesis was associated
compared to other foci. with increase of basophilic or amphophilic foci (Goodman
et al. 1994). Although age-related eosinophilic or tigroid
Differential diagnosis:
basophilic foci were not associated with exposure to hepatocar-
 Hyperplasia, hepatocellular, regenerative—evidence cinogens, in hamsters, treatment with nitrosamines or other
of prior or ongoing hepatocellular damage. carcinogens caused a variety of foci including basophilic foci
 Hyperplasia, hepatocellular, non-regenerative— (Frith, Ward, and Turusov 1994; Moore, Thamavit, and
usually seen as a single large nodule of hyperplasia Bannasch 1996). As some foci may be potential precursors of
without concomitant evidence of hepatocellular dam- neoplastic formation, careful identification of altered foci is war-
age or phenotypical alteration. Rare in rats and mice. ranted (Maronpot et al. 1989). Although induced by carcinogens,
 Adenoma, hepatocellular—loss of normal lobular foci of cellular alteration can be found as non-neoplastic end
architecture with irregular growth pattern. Liver stage lesions and not all foci can be related to carcinogens (Per-
plates often impinge perpendicular or obliquely on aino et al. 1984; Harada, Maronpot, Morris, Stitzel, et al. 1989;
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 29S

Harada, Maronpot, Morris, and Boorman, 1989; Squire 1989; treatment-associated and consists of a proliferative collection
Schulte-Hermann et al. 1989). Most importantly, types of of hepatocytes spanning several lobules and without evidence
foci in controls should be compared to those found in treated ani- of prior hepatic damage. This lesion is not associated with any
mals. Some pathologists regard vacuolated foci as focal fatty evidence of existing or prior hepatocellular injury. Diagnostic
change and do not consider them a subtype of focus of cellular difficulties occur when preneoplastic foci and hepatocellular
alteration. adenomas occur in the same liver sections of older rodents.
This lesion may be similar to that of hepatic nodular hyperpla-
Hyperplasia, Hepatocellular, Non-Regenerative (Figures sia in dogs.
109 and 110) There are basically two variations of non-regenerative
hepatocellular hyperplasia. One is relatively smaller and is
Synonyms: Hyperplasia, hepatocellular, focal hepatocellular accompanied by angiectasis and/or spongiosis hepatis and the
hyperplasia. other tends to be larger than several lobules. The former
occurs in both sexes and the latter predominantly in untreated
Pathogenesis: A spontaneous or treatment-associated prolif- female control F344 rats but occasionally reported in treated
erative collection of hepatocytes spanning several lobules and rats (Tasaki et al. 2008; Hailey et al. 2005; Bach et al.
without evidence of prior hepatic damage. 2010). When present near the capsular surface, this type of
Diagnostic features: nodular hyperplasia may be evident grossly as a raised area.
The proliferating cell nuclear antigen (PCNA) labeling index
 A relatively large lesion that is often greater than sev- in these nodules is increased in comparison with surrounding
eral adjacent lobules and is occasionally accompa- parenchyma and the lesion is glutathione S-transferase pla-
nied by angiectasis and/or spongiosis hepatis (cystic cental form (GSTP) immunonegative.
degeneration). Very early non-regenerative hyperplasia may be the size of
 Comprised of slightly enlarged hepatocytes. small foci of cellular alteration and are identified by their
 Hepatocytes are tinctorially similar to surrounding altered growth pattern and tinctorial similarity to surrounding
parenchyma. parenchyma (see Figure 109).
 The liver plates in the lesion tend to merge with the
adjacent hepatic parenchyma.
 May be minimal to mild compression of adjacent Hyperplasia, Hepatocellular, Regenerative (Figures 111–113)
hepatic parenchyma. Synonyms: Hyperplasia, hepatocellular; hyperplasia, regenera-
 Lobular architecture is maintained. tive; hyperplasia, nodular; regeneration, nodular.
 Portal triads and central veins are present.
 When accompanied by angiectasis/spongiosis hepa-
Pathogenesis: A nodular regenerative response to prior or con-
tis, hepatic cords may be distorted.
tinuous hepatocellular damage.
Differential diagnosis: Diagnostic features:

 Focus of cellular alteration—phenotypical or tinctor-  Focal or multifocal (nodular) appearance.


ial variation is present. Generally not associated with  Lesion may reach several millimeters in diameter.
chronic liver damage, though foci of cellular  Spherical proliferation may be accompanied by slight
alteration may occur in damaged liver. encapsulation.
 Adenoma, hepatocellular—loss of normal lobular  Compression of surrounding liver parenchyma often
architecture with irregular growth pattern. Liver plates occurs.
often impinge perpendicular or obliquely on the sur-  Normal lobular architecture usually present but may
rounding parenchyma. Distinct compression is present. be distorted.
 Carcinoma, hepatocellular—distinct trabecular, ade-  Portal triads and central veins may be present.
noid, or poorly differentiated growth is present.  Bile duct and oval cell proliferation may be
Lobular and plate architecture is not maintained. present.
Cellular atypia and invasive growth may be present.  Hepatocytes appear slightly altered, but may have
 Hyperplasia, hepatocellular (regenerative)—evidence slightly basophilic cytoplasm or prominent nucleoli.
of ongoing or prior hepatocellular damage, fibrosis,  Increased mitotic index may be observed.
or a history of exposure to a known hepatotoxicant.  Evidence of prior or ongoing hepatocellular damage,
Lobular architecture usually distorted. such as apoptosis/necrosis, chronic inflammation,
chronic congestion, fibrosis, cirrhosis, or a known
Comment: Non-regenerative hepatocellular hyperplasia cause of toxicity.
is rare in rodents. It may occur spontaneously or be  Lesions in rats tend to be more nodular than in mice.
30S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Differential diagnosis: Differential diagnosis:

 Focus of cellular alteration—tinctorial variation is  Infiltration, mononuclear—well demarcated; usually


present. Generally not associated with chronic liver solitary; associated with cell necrosis.
damage, though foci of cellular alteration may occur  Histiocytic sarcoma—diffuse and irregular prolifera-
in damaged liver. tion of histiocytic cells throughout the liver. Usually
 Adenoma, hepatocellular—loss of normal lobular associated with destruction of hepatic parenchyma.
architecture with irregular growth pattern. Liver Occasional multinucleated giant cells may be
plates often impinge perpendicular or obliquely on present.
the surrounding parenchyma. Distinct compression  Hyperplasia, oval cell—consists of a single or double
is present. row of oval cells sometimes forming incomplete
 Carcinoma, hepatocellular—distinct trabecular, (pseudo-) duct-like structure. Cells are usually uni-
adenoid, or poorly differentiated growth is present. form in size and shape and have scant pale basophilic
Lobular and plate architecture is not maintained. Cel- cytoplasm and round to oval nuclei.
lular atypia and invasive growth may be present.
 Hyperplasia, hepatocellular (non-regenerative)—no Comment: Rare as a spontaneous finding, hyperplasia of
history or evidence of hepatocellular damage. Kupffer cells may be seen following phagocytosis of foreign
material and as a consequence of estrogen treatment and in
Comment: These lesions are considered to represent a regen- inflammatory conditions. It can be induced by cytokines.
erative response to prior or continuous hepatocellular dam- Hypertrophy and hyperplasia often accompany each other. The
age. A history of exposure to a hepatotoxicant and the hypertrophy of normal Kupffer cells gives the impression of
presence of multiple nodules of regeneration that maintain a presence of more Kupffer cells since they are difficult to visua-
lobular but usually distorted architecture makes diagnosis lize in normal livers.
more convincing. Diagnostic difficulties occur when preneo-
plastic foci and hepatocellular adenomas occur in the same
liver sections of older rodents or in livers with many induced Hyperplasia, Ito Cell (Figures 116–118)
foci and tumors.
Synonyms: Stellate cell; perisinusoidal cell; fat-storing perisi-
In livers with partial hepatectomy (PH), the pattern of
nusoidal cell.
hyperplasia at twenty-four to seventy-two hours post surgery
is diffuse hepatocyte hyperplasia with many mitotic figures and
Pathogenesis: Proliferation of fat-storing perisinusoidal cells.
no evidence of liver toxicity. Although this response is also
hyperplasia (hepatocellular, regenerative), it is not to be con- Diagnostic features:
fused with the nodular hepatic response to toxic damage to
hepatocytes. After ninety-six hours, the liver may be almost  Focal or diffuse proliferation of Ito cells.
normal histologically. In mice however, chronic biliary lesions  May grow in sheets, clusters, or along cords of
may be seen in the liver after PH. hepatocytes.
 Cells vary in size and shape and are vacuolated.
 Multiple cytoplasmic fat droplets of different size
Hypertrophy/Hyperplasia, Kupffer Cell (Figures 114 and 115) occur.
 The nuclei are ovoid or round and may be indented by
Synonyms: Kupffer cell proliferation; histiocytosis, focal or
cytoplasmic lipid droplets.
diffuse.
 Modest amount of collagenous matrix may be present.
Pathogenesis: Following phagocytosis of foreign material,
Differential diagnosis:
estrogen treatment, inflammatory conditions, and response to
cytokines. A rare spontaneous finding.
 Fatty change/Lipidosis—the cytoplasm of fat cells
Diagnostic features: may be clearer than that of Ito cells.
 Ito cell tumor—larger and more extensive than
 Diffuse to multifocal proliferation of oval to spinde- hyperplasia. Partially distinct compression of adja-
loid cells lining sinusoids. cent hepatic parenchyma.
 Cells resemble histiocytes and often contain phago-
cytic material. Comment: Ito cell hyperplasia is extremely rare and occurs
 May form as sheets or nodules. predominantly in mice. It arises from fat-storing perisinusoi-
 Hypertrophy can occur without hyperplasia and vice dal cells, better known as Ito cells (Dixon et al. 1994; Enzan
versa. 1985; Tillmann et al. 1997). The biological behavior of the
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 31S

lesion is not well established. There appears to be a continuum Diagnostic features:


with Ito cell tumor (see the following), which may be just an
exaggerated and sometimes more localized form of Ito cell  Consists of dilated to cystic bile ducts filled
hyperplasia. with mucus and cellular debris and surrounded
by inflammatory cell infiltrates and connective
Bile Duct Hyperplasia (Figures 119–122) tissue.
 Glandular epithelium is typically a single layer and
Pathogenesis: A spontaneous change in portal areas of older varies from flattened to tall columnar hyperbasophi-
animals; may be induced or exacerbated by treatment. lic and pleomorphic cells along with goblet cells and
Diagnostic feature: occasional Paneth cells.
 Glandular epithelium, particularly in cystic glands,
 Increased number of small bile ducts arising in portal may be partially lost through degeneration resulting
region. in crescent shaped structures.
 May not involve all portal areas.  Central portions of large lesions may become sclero-
 May be associated with periductular fibrosis and tic with only remnants of biliary epithelium suggest-
periductular cell infiltration. ing regression.
 Biliary epithelium is well differentiated, forming  Lesions may be limited to small foci but may occupy
normal ducts. large interconnecting areas of a lobe without mark-
 Biliary epithelium may show degenerative or edly disturbing the lobe outline.
atrophic changes.  Lesion growth typically involves contraction with
 May be associated with oval cell hyperplasia. retraction of surrounding parenchyma. Markedly
 May contain mucous metaplasia or hyalinosis in dilated or cystic mucus-filled glands along the liver
specific situations in mice. capsule may protrude above the lobe outline.
 Cystic form.  Older lesions may be shrunken from the liver surface
 Focal bile duct proliferation with cystic dilation of and appear as scars.
ducts may occur.  Regenerative hepatocellular hyperplasia may
 Usually acini lined by flattened epithelium. be present when there is extensive parenchymal
involvement.
Differential diagnosis:
Differential diagnosis:
 Cholangioma—well demarcated; usually solitary;
acini lined by cuboidal epithelium.  Cholangiocarcinoma—solid sheets, trabeculae, or
 Cholangiofibrosis—central portions become nests of closely packed biliary cells largely displacing
atrophic, collagenized, and avascular whereas the hepatic parenchyma. Intestinal metaplasia is not a
active proliferating portion is at the periphery; mucus prominent feature. Glandular dilation is absent or
production is common. minimal.
 Cholangiocarcinoma—invasive growth into sur-  Hyperplasia, bile duct—multiple small bile ducts
rounding hepatic tissue or vessels; cell are pleo- arising in portal region. Biliary epithelium is well
morphic; mucus production. differentiated, forming more normal appearing
 Oval cell hyperplasia—consists of a single or double ducts, with minimal glandular dilation in some
row of oval cells forming incomplete duct-like struc- cases.
tures. Cells usually uniform with scant basophilic
cytoplasm and round or oval nuclei. Comment: This lesion is an inflammatory, proliferative, and
metaplastic reaction involving bile duct epithelium and is
Comment: Often associated with evidence of hepatic injury and seen with hepatocellular toxicity caused by xenobiotics such
repair and obstruction of bile flow. Dilation of intrahepatic bile as dioxins, furans, and related chemicals in rats (Bannasch
ducts is a spontaneous, age-associated lesion that is more com- and Zerban 1990; Deschl et al. 1997; Eustis et al. 1990;
mon in rats than in mice. Kimbrough et al. 1973; Kimbrough and Linder 1974; Sirica
1992; Hailey et al. 2005). The initial reaction following pro-
nounced hepatic parenchymal necrosis is oval cell hyperplasia
Cholangiofibrosis (Figures 123–126)
(Engelhardt 1997).
Synonyms: Bile duct adenomatosis; intestinal cell metaplasia; Cholangiofibrosis is a controversial lesion that has been
adenofibrosis. diagnosed as cholangiocarcinoma especially when there is
extensive involvement of the liver. Cholangiofibrosis is not
Pathogenesis: Originates from an initial oval cell hyperplasia seen as a spontaneous lesion but occurs primarily in rats treated
in response to pronounced hepatic parenchymal necrosis. with a variety of xenobiotic agents that are hepatotoxic at high
32S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

dose. The intestinal cell metaplasia in cholangiofibrosis hepatocellular neoplasms and may play an important role in
includes goblet and Paneth cells and columnar cells similar hepatocarcinogenesis. Some authors support the concept that
to chief cells identified in H&E-stained paraffin sections and oval cells may participate in the lineage of hepatocellular and
enterochromaffin cells identified by special staining. The cholangiocellular carcinomas and may serve as hepatic stem
metaplastic cells have been confirmed by electron micro- cells. Oval cell hyperplasia diagnosis including grade is rec-
scopy in some cases. While cholangiofibrosis has been ommended, even when it is part of a complex set of hepatic
reported to progress to cholangiocarcinoma with malignant changes.
features (Bannasch and Zerban, 1990; Sirica 1992), unequivo-
cal metastases have not been confirmed in most cases. This
G. Neoplasms
lesion is not observed in humans.
Introduction
Oval Cell Hyperplasia (Figures 127–129)
The rodent liver is the most common target site of chemical
Synonyms: Oval cell proliferation; bile ductule cell hyperplasia. carcinogens (Maronpot et al. 1986; Evans and Lake 1998), per-
haps due to its major function as a metabolizing and detoxifying
Pathogenesis: Arises from terminal ductule epithelial cells organ for xenobiotics. Rodent hepatocarcinogens are usually
(canal of Hering cells) spontaneously, following liver infec- hepatotoxins. The chronic toxicity of these toxins may
tions, and secondary to hepatotoxic injury. contribute to hepatocarcinogenesis although genotoxic liver
Diagnostic features: carcinogens are often also hepatotoxins. There is over a thirty-
year history of experimental induction (Frith and Wiley 1982;
 Generally originates from portal areas and is often Malarkey et al. 1995; Evans et al. 1992; Ward et al. 1983,
multifocal. 1986; Ward, Lynch, and Riggs 1988; Popp 1984) and classifica-
 Consists of a single or double row of oval to round tion of preneoplastic and neoplastic lesions of the rat and mouse
cells along sinusoids in linear arrays. liver in book chapters (Bannasch and Zerban 1990; Brooks
 May form a few or many small ductules with stream- and Roe 1985; Greaves and Faccini 1984; Jones and Butler
ing into the hepatic parenchyma. 1978; Ward 1981; Harada et al. 1999; Eustis et al. 1990) and
 Formation of incomplete duct-like structures may by committee (ILAR 1980) or toxicologic pathology
be present. societies (Standardized System of Nomenclature and Diagnos-
 Cells are usually uniform in size and shape and may tic Criteria [SSNDC] Guides, http://www.toxpath.org/
be fusiform. ssndc.asp). Terminology has evolved to the present nomencla-
 Cells have scant pale basophilic cytoplasm and round ture that is also based on many publications on liver
or oval nuclei. carcinogenesis.
 Oval cells express keratin. There is evidence from experimental studies documenting
the regression of proliferative hepatocellular lesions including
Differential diagnosis: foci of cellular alteration, hepatocellular adenomas, and hepato-
cellular carcinomas following cessation of treatment (Maronpot
 Hyperplasia, bile duct—several small bile ducts 2009). A dramatic example was reported in mice following
arising in portal region. Biliary epithelium is well cessation of chronic chlordane exposure (Malarkey et al.
differentiated, forming normal ducts. 1995). Similar experience has been reported in rats and mice
 Early or mild fibrosis—collagen matrix is evident. in other studies (Lipsky et al. 1984; Greaves, Irisarri, and
 Inflammation—presence of mononuclear, polymor- Monro 1986; Marsman and Popp 1994) as well as in humans
phonuclear, or connective tissue cells will be present (Frémond et al. 1987; McCaughan, Bilous, and Gallagher
in inflammation. 1985; Emerson et al. 1980; Steinbrecher et al. 1981). Agents that
require continual administration for the stable presence and
Comment: Oval cell proliferation is considered to arise from
growth of preneoplastic and neoplastic rodent liver lesions can
terminal ductule epithelial cells (canal of Hering cells). It is
be categorized as conditional hepatocarcinogens (Maronpot
a rare spontaneous lesion in rats. Oval cell hyperplasia can
2009).
be observed following severe hepatotoxic injury and treat-
ment with hepatocarcinogens. There is often a close relation
Hepatocellular Adenoma (Figures 130-134)
to the portal tract, although more scattered groups of prolif-
erating oval cells can be seen diffusely throughout the liver Synonyms: Adenoma, hepatic; adenoma, liver parenchymal
following xenobiotic-induced hepatic injury (Engelhardt cell; hepatoma, benign; tumor, liver cell, benign; hepatoma,
1997). benign; type A nodule.
In mice oval cell hyperplasia is a feature of chronic active
hepatitis caused by H. hepaticus and H. bilis and is seen fol- Pathogenesis: Spontaneous and following treatment with
lowing treatment with various hepatocarcinogens. Hyperplas- hepatotoxins that are carcinogenic xenobiotics; with gene
tic oval cells occur in association with a high incidence of alterations in genetically engineered mice.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 33S

Diagnostic features: and/or occurrence of cellular atypia; adenoid, trabe-


cular, or poorly differentiated growth.
 Are often but not always grossly visible as small
(1 mm) to large uniformly round nodular lesions. Comment: Hepatocellular adenomas occur spontaneously with
 Histologically lesions are nodular and compressing increased incidence in older rodents and following treatment
of adjacent normal hepatocytes. with hepatotoxins that are carcinogenic xenobiotics (Harada
 Sharply demarcated from surrounding liver et al. 1999; Eustis et al. 1990; Stinson, Hoover, and Ward
parenchyma that is often compressed. The compres- 1981). In mice, hepatocellular carcinomas can be seen histologi-
sion of adjacent normal hepatocytes should be at least cally arising within adenomas of both spontaneous and induced
on two quadrants of the adenoma. adenomas (Jang et al. 1992). This process is less common in rats.
 Loss of the normal lobular architecture with irregular Occasionally large proliferative hepatocellular lesions are
growth pattern. observed that are difficult to diagnose. These lesions can com-
 Liver plates often impinge obliquely on surrounding press adjacent parenchyma and/or bulge from the surface.
liver parenchyma. They also have, at least in some areas, normal to slightly dis-
 Hepatocytes with varying size and tinctorial staining torted lobular architecture with central veins and portal triads.
pattern occur. The biologic nature of these lesions is unknown but because of
 Adenomas may also be classified morphology by their size and distinct compression, they are sometimes
tinctorial and other cytoplasmic characteristics as included in the category of hepatocellular adenoma. It is rec-
described for foci of cellular alteration. ommended that large lesions of this type with some evidence
 Staining of tumor cells may resemble that of the sur- of lobular architecture and the presence of central veins and
rounding liver parenchyma and be classified as portal areas be diagnosed as non-regenerative hepatocellular
amphophilic. hyperplasia (described previously in this document).
 Usually single nodules but multiple adenomas may
be present.
 Fibrous encapsulation may occur. Hepatocellular Carcinoma (Figures 135–140)
 An enveloped portal triad may occasionally be present.
Synonyms: Adenocarcinoma, liver cell; carcinoma, hepatic
 Mitotic index may be increased.
cell; carcinoma, hepatocellular; carcinoma, liver cell; hepa-
 Areas of cellular atypia may be present (pleomorphic
toma, malignant; hepatocarcinoma; nodule, type B.
nuclei, coarsely clumped chromatin, large nucleoli,
increased nucleus to cytoplasm ratio, cytoplasmic
Pathogenesis: Spontaneous and following treatment with hepa-
basophilia, focal attempts at trabeculae formation as
totoxins that are carcinogenic xenobiotics; with gene altera-
in carcinomas).
tions in genetically engineered mice.
 Cells within adenomas may reveal signs of degenera-
tive processes such as intracytoplasmic-inclusions, Diagnostic features:
hyaline bodies, or vacuoles.
 Sinusoids may be compressed or ectatic.  Local infiltrating growth and/or lack of distinct
 Necrosis is usually absent. demarcation.
 Pseudoinvasion of blood vessels may be present.  Marked cellular pleomorphism may occur.
 Lesions may be grossly visible and/or bulge from nat-  Alterations of tinctorial staining patterns may occur.
ural surfaces.  Loss of normal lobular architecture.
 May occur in livers with foci of cellular alteration  Vascular invasion or metastases may be observed.
and hepatocellular carcinomas.  Increased mitotic index possible.
 Bile ducts may be present.
Differential diagnosis:  Hemorrhage, necrosis, and extramedullary hemato-
poietic foci may be present.
 Focus of cellular alteration—liver plates merge  May occur as a single morphologic type or a combi-
imperceptibly with surrounding hepatic parenchyma; nation of them as in the following.
normal lobular architecture present.  May metastasize to lung.
 Regenerative hyperplasia (in damaged liver)—evi-
dence of prior or ongoing hepatocellular damage; nor- Trabecular:
mal lobular architecture is present, albeit distorted.
 Non-regenerative hyperplasia—some evidence of  Composed of well-differentiated hepatocytes form-
lobular pattern and presence of central veins and por- ing trabeculae of multiple cell layers.
tal triads.  The plates alternate with sinusoids.
 Hepatocellular carcinoma—invasive growth; no  Sometimes the sinusoids may be dilated forming
clear demarcation; loss of lobular plates, architecture, blood lakes.
34S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Acinar (synonym: glandular): The trabecular type of hepatocellular carcinoma is the most
common form in rats and to a lesser extent in mice.
 Neoplastic hepatocytes form a generally single layer Hepatocellular carcinomas may appear to arise within preexist-
around a central clear space. ing adenomas, especially in mice (Figures 139 and 140). While
 The acinar structure may vary from tiny to huge cysts. some authors refer to these lesions as focus of carcinoma in
 Glandular pattern rarely involves more than 50% of adenoma, such lesions should be diagnosed as hepatocellular
the neoplasm. carcinoma. These carcinomas exhibit the same morphological
 Acinar portion is interspersed with areas that have appearance as the carcinomas described earlier and hint toward
either trabecular or solid architecture. a progression of adenoma into carcinoma. Attention should be
paid to hemorrhagic areas, where proliferation of endothelial
Solid: cells or formation of large sinusoidal-like spaces may lead to
the erroneous diagnosis of a vascular tumor.

 Cells tend to be poorly differentiated, small, hyper-


chromatic, and pleomorphic. Hepatoblastoma (Figures 141–144)
 Sometimes predominant cells are spindle shaped.
Synonyms: Tumor, mixed, poorly differentiated.
 Single nuclei or multinucleated giant forms.
 Mitotic figures may be numerous and bizarre.
 The stroma is generally inconspicuous. Pathogenesis: Unknown but origin from liver blastema cells,
 The vascular structures are immature and often neoplastic hepatocytes, oval cells, and biliary epithelial cells
thrombosed. proposed.

Adenoid (mouse): Diagnostic features:

 Well-circumscribed nodule.
 A layer of neoplastic cells surrounds luminal structures.
 Distinct encapsulation with variable structure may be
 Lining cells usually consist of strongly basophilic
present.
cuboidal cells.
 Organoid structures are lined by vascular cavities
filled with blood.
Differential diagnosis:
 The channels are surrounded by several layers of
tumor cells.
 Focus of cellular alteration—liver plates merge
 The cells are arranged either radially or concentrically
imperceptibly with surrounding hepatic parenchyma;
forming rosettes, trabeculae, or pseudo-glandular
normal lobular architecture present.
structures.
 Regenerative hyperplasia—evidence of prior or
 The center of the rosettes sometimes contains ampho-
ongoing hepatocyte damage; normal lobular architec-
philic material or small, endothelium-lined vessel.
ture present, albeit distorted; demarcated from sur-
 Cells are small, strongly basophilic, and elongated.
rounding liver tissue.
Sometimes they may be more or less eosinophilic and
 Non-regenerative hyperplasia—some evidence of
have smaller, rounder, and less hyperchromatic nuclei.
lobular pattern and presence of central veins and por-
 Numerous mitotic figures are present.
tal triads.
 Areas of large hemorrhage, pigmentation, fibrosis, or
 Hepatocellular adenoma—no local invasiveness, less
necrosis are present.
cellular atypia, sharply demarcated from surrounding
 Osteoid, bone, squamous differentiation, and extra-
liver parenchyma. No formation of a trabecular pattern.
medullary hematopoietic foci may be present.
 Cholangiocarcinoma—mucus may be present within
the adenoid structures. If there is loss of mucus it is
difficult to distinguish. Differential diagnosis:
 Hemangiosarcoma—proliferation of atypical
endothelial cells forming blood spaces.  Cholangiocarcinoma (poorly differentiated)—gland-
ular structures with mucus production are present.
Comment: Occur spontaneously with increased incidence in Often evidence is present of extensive fibrosis, but
older rodents and following treatment with carcinogenic and not of osteoid, or bone formation.
hepatotoxic xenobiotics (Bannasch and Zerban 1990; Brooks and  Sarcoma, NOS—only mesenchymal structures are
Roe 1985; Greaves and Faccini 1984; Jones and Butler 1978; present.
Ward 1981; Harada et al. 1999; Eustis et al. 1990; Popp 1984,  Carcinoma, hepatocellular-only sparse mesenchymal
1985; Vesselinovitch, Mihailovich, and Rao 1978). Diagnosis structures are present. Hepatic cell differentiation is
may be modified based on growth pattern (Frith and Wiley 1982). obvious.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 35S

Comment: Hepatoblastomas consist of organoid structures  Hyperplasia, bile duct—multifocal, usually widespread.
often oriented around vascular spaces (Harada et al. 1999;  Cholangiofibrosis—central portions become
Nonoyama et al. 1986, 1988; Turusov, Day, et al. 1973; atrophic, collagenized, and avascular whereas the
Turusov, Deringer, et al. 1973). The cells are primitive in actively proliferating portion is at the periphery;
appearance of scant, pale basophilic cytoplasm and ovoid mucus production is common.
hyperchromatic nuclei. They are usually seen with other types  Cholangiocarcinoma—proliferation of atypical cells,
of hepatocellular tumors, especially within hepatocellular invasive growth.
adenomas. Rare reports of this lesion in rats appear in litera-
ture. They are seen in certain strains of mice and have also Comment: Cholangioma is rare in control and treated
been induced by certain hepatocarcinogens (Diwan, Ward, rodents (Bannasch and Zerban 1990; Brooks and Roe 1985;
and Rice 1989). Frith and Ward 1979; Greaves and Faccini 1984; Harada
Hepatoblastoma is generally seen within or adjacent to et al. 1999; Jones and Butler 1978; Lewis 1984; Maronpot
hepatocellular neoplasms. In these cases, some preference has et al. 1986).
been expressed for a single diagnosis of hepatoblastoma, rather
than two diagnoses (the hepatoblastoma and the hepatocellular Cholangiocarcinoma (Figures 147 and 148)
neoplasm). If this preferred convention of using the single
diagnosis of hepatoblastoma is not used, then the alternative Synonyms: Adenocarcinoma, bile duct; adenocarcinoma,
convention will need to be defined by the pathologist. Hepato- cholangiocellular; carcinoma, bile duct; carcinoma, cholangio-
blastomas can have high rates of lung metastases in some cellular; cholangioma, malignant.
experiments.
Pathogenesis: Arise from proliferating cholangial cells.

Cholangioma (Figures 145 and 146) Diagnostic features:

Synonyms: Adenoma, bile duct; adenoma, biliary; adenoma,  Biliary structures are usually glandular but may have
cholangiocellular; cholangioma, benign. solid or papillary areas, may be poor in or free of
mucus, and mostly have a minimal connective tissue
Pathogenesis: Proliferation of biliary cells. component.
Diagnostic features:  Glandular lining cells are hyperbasophilic and have
large nuclei and nucleoli but occasionally have clear
 Glandular acini may vary in size and shape. cytoplasm due to accumulated glycogen.
 Expansively growing with compression.  Glands lined by single or multilayered cuboidal or
 Nucleus is round or oval occasionally vesicular with cylindrical cells.
one or two conspicuous nucleoli.  Clear-cut invasion into vascular and lymphatic struc-
 Cytoplasm is somewhat basophilic. tures and surrounding parenchyma may be present.
 Two types can be distinguished:  May appear as a large solid mass.
 Evidence of metastasis or likelihood of metastasis is
Simple: expected.

 Generally uniform well-circumscribed neoplasm. Differential diagnosis:


 Acini are lined by a single layer of cuboidal cells
varying in size.  Cholangiofibrosis—central portions become
 Occasionally the cells are multilayered. atrophic, collagenized, and avascular whereas the
 Sparse vascular stroma may occur. actively proliferating portion is at the periphery;
dilated glands with mucus production and accompa-
Cystic: nying inflammation is common. Connective tissue
response maybe pronounced.
 Characteristically composed of dilated glandular acini.  Cholangioma—no invasion, sparse stroma, more uni-
 The acini are lined by cuboidal epithelium. form cells, single layer of cuboidal cells, may have
 Papillary structures are occasionally observed, pro- cystic glands.
jecting into the lumen of the cysts.  Carcinoma, hepatocellular (acinar)—no mucus pro-
 Clumps of liver cells may be seen between the cysts. duction; contains obvious neoplastic hepatocytes.

Differential diagnosis: Comment: Cholangiomas and cholangiocarcinomas are rarely


seen as spontaneous neoplasms in rats and mice but may occur
 Bile duct cyst—lined by flattened epithelium; no expan- following exposure to hepatotoxic xenobiotics (Eustis et al.
sive growth; not forming acini or papillary structures. 1990; Frith and Ward 1979; Harada et al. 1999; Jones and
36S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Butler 1978; Lewis 1984; Narama et al. 2003). A specific  Carcinoma, hepatocholangiocellular—features of
phenotype of cholangiocarcinoma with features of hepatocellular carcinoma and cholangiocarcinoma
cholangiofibrosis consisting of dilated biliary glands, mucus are present. Hepatocytes arranged in nests, solid, tra-
production, intestinal metaplasia, inflammatory cell infil- becular, or glandular patterns. Stratification and aty-
trates, and fibrosis has been diagnosed in rats treated with a pia of biliary epithelial component is present.
variety of hepatotoxic xenobiotics (Bannasch and Zerban
1990; Bannasch, Brenner, and Zerban 1985; Sirica 1992; Comment: Rare spontaneous lesion. A proliferative mixture of
Kimbrough and Linder 1974; Maronpot et al. 1986). The hepatocytes and intrahepatic bile duct epithelium with neither
distinction between cholangiofibrosis and this phenotype of component being malignant comprise to this tumor type
cholangiocarcinoma is difficult and controversial and is pri- (Deschl et al. 2001; Frith and Ward 1979; Harada et al.
marily based on extent of liver involvement since unequivocal 1999; Narama et al. 2003).
metastasis is rare.
These rare lesions of cholangiocarcinoma can contain para- Carcinoma, Hepatocholangiocellular (Figures 151 and 152)
meters of cholangiofibrosis but show less inflammation and
less mucus cyst(s) but with atypical ductular structures and can Pathogenesis: Proliferation of admixture of hepatocytes and
metastasize. It has been proposed to diagnose this specific form intrahepatic bile duct epithelium. Stem cell origin speculated.
of cholangiocarcinoma as ‘‘cholangiocarcinoma, intestinal Diagnostic features:
type’’ (Greaves 2007), but that nomenclature as a separate
(sub-) diagnosis was not favorably encouraged at a recent  Features of hepatocellular carcinoma and cholangio-
scientific workshop (NTP Satellite Symposium 2010). carcinoma are present.
 The hepatocytic component may be arranged in tra-
becular, glandular, or solid patterns.
Adenoma, Hepatocholangiocellular (Figures 149 and 150)  The biliary component may form acini or small nests
Pathogenesis: Proliferation of admixture of hepatocytes and without lumen. Stratification or cellular atypia may
intrahepatic bile duct epithelium. Stem cell origin speculated. occur.
 Occasionally ducts lined by both cell types, hepato-
Diagnostic features: cytic and biliary epithelial cells, may be present.
 Areas of hemorrhage and necrosis may be present.
 Features of hepatocellular adenoma and cholangioma Mitotic figures may be numerous.
are present.
 Areas composed of cords of neoplastic hepatocellular Differential diagnosis:
cells merge with areas composed of ducts lined by
neoplastic epithelium that resembles bile duct epithe-  Carcinoma, hepatocellular—composed of malignant
lium. Hepatocytes may be seen forming ductules or hepatocytes.
ducts.  Cholangiocarcinoma—composed of malignant duct
 The neoplastic biliary epithelium forms slightly epithelium.
dilated acini lined by cuboidal cells.  Adenoma, hepatocholangiocellular—features of
 Stratification and atypia of the biliary epithelium is hepatocellular adenoma and cholangioma are pres-
minimal or absent. ent. Hepatocytic component has typical cord-like
 Stromal component may be absent. arrangement of cells. Evidence of stratification and
 Hepatocytes may have some alteration in staining atypia of biliary epithelial component is minimal or
(eosinophilic, basophilic, or clear cell) compared to absent.
surrounding parenchyma.
 Mitotic figures are rare. Comment: Rare spontaneous lesion. This neoplasm contains
neoplastic elements of both hepatocytes and bile duct epithe-
Differential diagnosis: lium (Deschl et al. 2001; Frith and Ward 1979; Harada et al.
1999; Narama et al. 2003; Teredesai, Wohrmann, and Schlage
 Adenoma, hepatocellular—composed of neoplastic 2002). A diagnosis of malignancy may be based on just one of
hepatocytes. Loss of normal lobular architecture with the components being malignant.
irregular growth pattern. Liver plates often impinge
perpendicular or obliquely on the surrounding par-
Tumor, Ito Cell, Benign (Figures 153 and 154)
enchyma. Distinct compression is present.
 Cholangioma—composed of neoplastic duct Synonyms: Fat-storing cell tumor; stellate cell tumor, lipoma.
epithelium. Well demarcated; usually solitary. The
acini are lined by cuboidal uniform and well- Pathogenesis: Arises from fat-storing perisinusoidal cells,
differentiated epithelium. so-called Ito cells.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 37S

Diagnostic features:  Atypical cells are sparse, pleomorphism is usually


absent, and mitotic figures may be numerous.
 Unicentric or multicentric unencapsulated mass.  The tumors grow along sinusoids and vessels and
 Focal or diffuse accumulation of tumor cells. frequently involve other organs such as the lung and
 Partially distinct compression of adjacent hepatic spleen.
parenchyma.  Metastases and spread on serosal surfaces and in the
 May grow in sheets, clusters, or along cords of vascular spaces are common.
hepatocytes.  Minimal fibrosis may be present.
 Cells vary in size and shape and are vacuolated.
 Multiple cytoplasmic fat droplets of different size Differential diagnosis:
occur.
 The nuclei are ovoid or round and may be indented by  Malignant fibrous histiocytoma—the tumor has a
cytoplasmic lipid droplets. mixed cell population of histiocyte-like cells,
 Frequently evidence of atrophy of adjacent liver tis- bizarre tumor giant cell, fibroblasts, and undifferen-
sue is present. tiated cells. The fibrous component is always
 Modest amount of collagenous matrix may be prominent.
present.  Malignant lymphoma—no giant cells are seen and
 At the margin of the lesion, spindle-shaped cells are lymph nodes and spleen are frequently involved. His-
frequently observed. tiocytic sarcoma and lymphoma may occur together
 Unencapsulated. in the liver.

Differential diagnosis: Comment: Histiocytic sarcomas occur at a low frequency in


rats and mice (Harada et al. 1999; Eustis et al. 1990). The tumor
 Ito cell hyperplasia—multicentric lipomatous can be part of a systemic lesion involving various tissues
lesion or a singular small lesion that does not (spleen, lung, and uterus); when involving only the liver it is
show distinct compression of surrounding liver sometimes referred to as Kupffer cell sarcoma (Deschl et al.
parenchyma. 2001; Carlton et al. 1992).
 Liposarcoma—various types of fat cells, foam cells,
giant cells, myxoid cells, or fibroblast-like cells may
be present. Hemangioma (Figures 156 and 157)
Synonym: Hemangioendothelioma, benign.
Comment: Ito cell neoplasms are extremely rare. As a conse-
quence, the histogenesis and the biological behavior of these Pathogenesis: Arises from endothelial cells lining vascular
tumors are not well established (Dixon et al. 1994; Enzan spaces, most commonly of the hepatic sinusoids.
1985; Tillmann et al. 1997).
Diagnostic features:

Histiocytic Sarcoma (Figure 155)  A moderate compression of the surrounding tissues is


usually seen.
Synonym: Kupffer cell sarcoma.  The tumor is rarely encapsulated.
 Blood-filled spaces lined with a single layer of
Pathogenesis: May arise from fixed macrophages (Kupffer prominent uniform endothelial cells without
cells) attached to the sinusoidal endothelial cells or from circu- atypia.
lating macrophages, unless metastasized from other organs  Mitotic figures are rarely present.
(e.g., skin, uterus).  Solid cellular areas of uniform cells without atypia
Diagnostic features: may occur.
 Often multifocal in the liver or in livers with angiec-
 Characteristically form nodules within the liver tasis of mice.
that may contain a central area of necrosis sur-
rounded by palisaded tumor cells (multicentric Capillary type:
origin).
 Uniform population of rounded or oval cells with  Closely packed capillary structures.
foamy, eosinophilic cytoplasm, indistinct cell bound-  Minimal stroma between the vascular spaces.
aries, and elongated or folded nuclei.  Cavernous type:
 Sometimes are present multinucleated giant cells of  Large vascular channels.
foreign body scattered throughout the tumor.  Abundant connective tissue between the larger channels.
38S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Differential diagnosis: collagen bundles, and surfaces with no cytological


and histological features of malignancy.
 Angiectasis—dilated vessels or sinusoids are not  Hemangioma—no cytological and histological fea-
increased in number and have normal structure and tures of malignancy such as cellular pleomorphism,
well-differentiated endothelial cells. increased mitotic activity, tissue invasion, or metas-
 Hyperplasia, angiomatous—the hyperplastic vessels tases are present.
are lined by a normal endothelium and cause no or  Hemangiopericytoma, malignant—the tumor
only minimal compression of the surrounding consists of tightly packed spindle-shaped tumor
tissues. cells that have cytological features of malignancy
 Lymphangioma—the vascular spaces are devoid of and encircle thin-walled vascular channels
erythrocytes. (‘‘fingerprints’’).
 Hemangiosarcoma—cytological and histological  Fibrosarcoma—the tumor lacks a distinct vascular
features of malignancy are present, such as cellular pattern with prominent endothelial cells.
pleomorphism, increased mitotic activity, tissue
invasion, or metastases.
Comment: As in the case of hemangiomas, the occurrence of
 Hemangiopericytoma, benign—the tumor consists of
hemangiosarcomas in rodents has been well described in the
tightly packed spindle-shaped tumor cells encircling
published literature (Binhazim, Coghlan, and Walker 1994;
thin-walled vascular channels (‘‘fingerprints’’). In
Booth and Sundberg 1996; Faccini, Abbott, and Paulus
reticulin-stained sections, a dense reticulin meshwork 1990; Frith and Ward 1988; Frith and Wiley 1982; Giddens
surrounds individual tumor cells.
and Renne 1985; Greaves and Barsoum 1990; Greaves and
Faccini 1984; Heider and Eustis 1994; Jones and Butler
Comment: The occurrence of hemangiomas in rodents has
1975; Maita et al. 1988; Mitsumori 1990; Morgan et al.
been well documented (Booth and Sundberg 1996; Carter
1984; Peckham and Heider 1999; Popper, Maltoni, and
1973; Faccini, Abbott, and Paulus 1990; Frith and Ward
Selikoff 1981; Pozharisski and Turusov 1991; Sakamoto,
1988; Frith and Wiley 1982; Heider and Eustis 1994; Jones
Takayama, and Hosoda 1989; Solleveld et al. 1988; Stewart
and Butler 1975; Maita et al. 1988; Greaves and Barsoum
1979; Yamate et al. 1988).
1990; Greaves and Faccini 1984; Mitsumori 1990; Peckham Endothelial cell-derived hemangiosarcomas can be
and Heider 1999; Stewart 1979; Stewart et al. 1980; Squire
induced in rats and mice by a wide range of industrial, nat-
and Levitt 1975; Ward et al. 1979; Yamate et al. 1988;
ural, and pharmaceutical compounds. There are numerous
Zwicker et al. 1995). The cavernous type of hemangioma is
examples documenting the progress that is being made in
considered by some authors to be a congenital malformation
recent years in suggesting the genesis and potential rele-
rather than a neoplasm.
vance for human risk assessment of these tumors (Klaunig
and Kamendulis 2005; Laifenfeld et al. 2010; Ohnishi
Hemangiosarcoma (Figures 158-160)
et al. 2007).
Synonym: Hemangioendothelioma, malignant.

Pathogenesis: Arises from pluripotential mesenchymal stem


H. Other Liver Lesions
cells; endothelial cells of blood vessels or hepatic sinusoids.
Extramedullary Hematopoiesis (Figures 161 and 162)
Diagnostic features:
Synonym: Hematopoietic cell proliferation; myelopoiesis;
 The endothelial lining cells have a moderate erythropoiesis.
pleomorphism.
 Endothelial cells may be multilayered and/or
Pathogenesis: In adult rodent, a response to increased hemato-
clustered.
poietic demand.
 Various vascular patterns may be present, but vessels
are not well formed. Diagnostic features:
 Undifferentiated or fibrosarcomatous areas may also
be seen.  Small aggregates of hematopoietic cells are randomly
 Mitotic figures are often present. distributed in the hepatic sinusoids as well as around
 Local invasion and metastases are often present. central veins and in portal areas.
 Both erythyroid and granulocytic cells may be pres-
Differential diagnosis: ent in the aggregates; rarely megakaryocytes may
be present.
 Granulation tissue—the newly formed blood vessels  Typically not associated with hepatocellular necrosis
are typically arranged perpendicular to fibroblasts, or degeneration.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 39S

Differential diagnosis: Pancreatic Acinar Metaplasia (Figures 164 and 165)


Pathogenesis: Islands of pancreatic tissue localized within the
 Mononuclear cell aggregates—lymphocytes and his-
hepatic parenchyma are rare spontaneous occurrences in rats
tiocytic cells present alone or in addition to mature
but have been reported following prolonged exposures to poly-
myeloid cells.
chlorinated biphenyls (Kimbrough 1973; Eustis et al. 1990;
 Focal inflammation—mixed mature leukocytes,
Greaves 2007).
often associated with or a response to cellular
necrosis. Diagnostic features:

 The islands of pancreatic tissue resemble normal aci-


Comment: Extramedullary hematopoiesis (EMH) can be
nar pancreas with zymogen granules.
observed in rodent liver occasionally in response to an
 An integrated component of the hepatic tissue.
increased hematopoietic demand. Hematopoiesis is normally
found in the embryonic liver where embryonic hematopoiesis
Differential diagnosis:
dramatically expands at mid-gestation but decreases after birth.
Hepatic EMH is seen more common in rodents than in man,
 Artifact during tissue processing—tissue ‘‘floater.’’
more common in mice than rats, and more often observed in
 Metastatic pancreatic acinar neoplasia—locally inva-
females as compared to males as a general rule (Eustis et al.
sive with tissue destruction; presence of a primary
1990; Harada et al. 1996, 1999). Precipitating factors for the
pancreatic acinar neoplasm in the pancreas.
occurrence are: anemia, stress, xenobiotic toxicity, infection,
 Ectopic pancreatic tissue—a collection of pancreatic
neoplasia (e.g., histiocytic sarcoma), and pregnancy. When the
tissue adjacent to but not integrated within hepatic
erythroid precursors predominate, often the term extramedul-
parenchyma.
lary erythropoiesis is used.
Comment: In spontaneous cases, a distinction between meta-
plasia and ectopic pancreas may not be possible. Since both
Intrahepatocellular Erythrocytes (Figure 163)
pancreas and liver are embryologically related, there is a defi-
Synonyms: Emperipolesis, cytoplasmic inclusions; hepatic nite potential for metaplasia.
erythrophagocytosis.
Hepatocytes, Glandular Metaplasia (Figures 166 and 167)
Pathogenesis: Unknown. Pathogenesis: Proliferation of hepatocytes to form glandular
structures.
Diagnostic features:
Diagnostic features:
 Individual or small clusters of enlarged hepatocytes
containing intact erythrocytes.  A few to numerous glandular structures diffusely
 Affected hepatocytes are markedly enlarged. scattered in the hepatic parenchyma.
 Marginated hepatocyte nucleus.  May also be present in hyperplastic nodules and
hepatocellular adenomas.
Differential diagnosis:  Vary in size from one to ten times the diameter of a
portal bile duct.
 Angiectasis—dilated vascular spaces lined by  Lining cells resemble hepatocytes but smaller more
endothelial cells; dilated vascular channels and cuboidal glandular cells resemble biliary epithelium.
spaces frequently contain erythrocytes.  Glandular lumen may contain granular eosinophilic
 Cystic degeneration—consists of enlarged stellate material and sometime free blood cells.
cells with flocculent eosinophilic cytoplasm.
Differential diagnosis:
Comment: The intracytoplasmic inclusion of large numbers of
erythrocytes in hepatocytes has been seen exclusively in mice  Cholangioma; glandular acini may vary in size and
(Harada et al. 1999). It has occurred in at least nine separate shape, expansively growing with compression.
cancer bioassays and one fourteen-day study in B6C3F1 mice.  Hepatocholangioma; features of hepatocellular ade-
In two of these studies it appears to have been exacerbated or noma and cholangioma are present.
possibly caused by treatment. Attempts to demonstrate active
erythrophagocytosis by electron microscopy have been unsuc- Comment: Partial replacement of hepatic parenchyma by
cessful. A potential mechanism is emperiopolesis. Internaliza- glandular structures with features resembling hepatocytes has
tion of erythrocytes in hepatocytes has been reported in been observed in chronic studies of 3, 3’, 4, 4’, 5-
hibernating frogs (Barni and Bernocchi 1991). pentachlorobiphenyl and 2, 3’, 4, 4’, 5-pentachlorobiphenyl
40S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

(NTP Toxicology and Carcinogenesis Studies 2006). It is Diagnostic features:


speculated that the glandular structures represent abnormal dif-
ferentiation of hepatic precursor cells (NTP Toxicology and  Mature pancreatic acinar tissue located in wall of the
Carcinogenesis Studies 2006). gallbladder (Harada et al. 1999).

Intravascular Hepatocytes (Figure 168) Differential diagnosis:

Synonyms: Vascular pseudoinvasion; vascular infiltration of  Metastatic pancreatic acinar carcinoma—locally


hepatocytes. invasive neoplasm with distortion and/or destruction
of gallbaldder tissue.
Pathogenesis: Unknown. Sporadic occurrence.  Pancreatic metaplasia—smoothly integrated normal
Diagnostic features: appearing pancreatic tissue within the gallbladder
mucosa.
 Protrusion of normal appearing hepatocytes into
hepatic veins and within the contour of the vessel. Degenerative Lesions
 Usually involves medium to large size hepatic veins.
 Infiltrating hepatocytes are covered by an endothelial Hyalinosis, Gallbladder (Figures 171–174)
cell lining. Synonyms: Hyalinosis, cytoplasmic inclusions, crystals.

Differential diagnosis: Pathogenesis: A change in the gallbladder epithelium that can


be induced by inflammation and unknown factors.
 Extension of perivascular focus of cellular alteration,
usually basophilic. Diagnostic features:
 Metastatic hepatocellular carcinoma.
 The gallbladder epithelial cells contain a hyaline
Comment: Intravascular infiltration of hepatocytes is rarely cytoplasm, which is uniformly eosinophilic.
seen in control and treated mice. A similar change was  The protein is usually immunoreactive for Ym1/
reported in diethylnitrosamine treated mice as part of a baso- Ym2.
philic focus of cellular alteration response (Goldfarb et al.  Epithelial cells may contain eosinophilic needle-like
1983; Koen, Pugh, and Goldfarb 1983). The significance of crystals and the same and larger crystals may be seen
this change is unknown. extracellularly.
 Maybe associated with hyalinosis in other tissues,
such as bile duct epithelium, stomach, and lung.
I. Gallbladder Lesions
Differential diagnosis:
Congenital Lesions
Heterotopic Hepatocytes  Other degenerative cellular changes without hyali-
nized eosinophilic cytoplasm.
Pathogenesis: Developmental anomaly; postnatal
 Amyloidosis—pale eosinophilic extracellular deposits.
transdifferentiation.
Diagnostic features: Comment: The hyaline protein in the cells has been shown to
be Ym1/Ym2 (now Chi313), a chitinase-like protein, with
 Cells morphologically identical to mature hepato- unknown functions. In sickle cell mice, it is associated with
cytes are present in the submucosa of the gallbladder gallstones. Hyalinosis is rare in most lines of mice (Harada
(Harada et al. 1999). et al. 1999; Hsu et al. 2006; Yang and Campbell 1964) but may
occur in high incidence in 129 and B6;129 mice (Ward et al.
Differential diagnosis: 2001) and in some genetically engineered mouse lines. Hyali-
nosis is reported to occur in increased incidence in B6C3F1
 Metastatic hepatocellular neoplasm—locally inva- female mice exposed to penicillin.
sive expansile mass of atypical hepatocytes.
 Artifact—tissue ‘‘floater.’’ Glandular Metaplasia
Synonym: Adenomatoid change.
Heterotopic Acinar Pancreas (Figures 169 and 170)
Pathogenesis: Developmental abnormality; postnatal Pathogenesis: Spontaneous and associated with inflammatory
transdifferentiation. and proliferative changes in gallbladder.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 41S

Diagnostic features: Comment: Gallstones are rare spontaneously in mice but


may be experimentally induced by various methods (Chang,
 Thickened mucosa with diffuse or focal proliferation Suh, and Kwon 1999; Hsu et al. 2006; Ichikawa et al. 2009; Lee
of tall columnar cells forming numerous glands in the and Scott 1982; Lewis 1984; Rege and Prystowsky 1998;
lamina propria. Tepperman, Caldwell, and Tepperman 1964; Trotman et al.
 The gallbladder epithelial cells may show increased 1983; Xie et al. 2009).
cell proliferation.
 Hypertrophic columnar cells with uniform, homoge-
Inflammatory Lesions
neous, bright eosinophilic cytoplasm forming gland-
ular structures with lumen containing eosinophilic Cholecystitis (Figures 177 and 178)
crystals.
Synonym: Inflammation, gallbladder.
 Chronic inflammation of the gallbladder may be
present.
Pathogenesis: Toxicant exposure and bacterial and viral infec-
tions (Greaves 2007; Harada et al. 1999).
Comment: Glandular metaplasia has been observed at low pre-
Diagnostic features:
valence in the gall bladder and intra-hepatic bile ducts (all asso-
ciated with cholelithiasis), cholecystitis, cholangitis,
 May be accompanied by ulcers or erosions of the
papillomatous hyperplasia, papilloma, intra-mural cysts, and
mucosal lining cells.
focal epithelial ulceration of aged mice from life span carcino-
 Types of inflammation range from acute to chronic,
genicity studies. The lesions are found predominantly in female
including a granulomatous reaction.
mice (Lewis 1984) and can occur spontaneously in some strains
 Lumen may contain necrotic cellular debris.
of mice. Other metaplastic changes have also been described in
 Mucosal hyperplasia and mucinous metaplasia may
the human gallbladder, including goblet cells, paneth cells,
be present in some cases of inflammation.
and/or enterochromaffin cells in the mucosa (Hruban, Argani,
 Initial change may be submucosal edema.
and Ali 2006).

Proliferative Lesions
Gallbladder, Calculi (Figures 175 and 176)
Hyperplasia, Gallbladder (Figures 179 and 180)
Synonyms: Stones, gallstones, choleliths.
Pathogenesis: Irritation of gallbaldder mucosa and after xeno-
biotic exposure.
Pathogenesis: Excess dietary factors and altered metabolism.
Diagnostic features:
Diagnostic features:
 Lesion is often small.
 Grossly visible concretion(s) in the gallbladder of  Lesion varies from a few cells on papillary folds to
mice. small papillary projections.
 Grossly, may be solid or soft, single or multiple, and  Epithelium is usually single layered.
of various colors including white and pigmented (yel-  Cells are well differentiated.
low, grey).  Minimal atypia may be present.
 Depending on the etiology, the stone may be com-
posed of a mixture of cholesterol, calcium salts, Differential diagnosis:
hemoglobin, and occasionally as a pure stone com-
posed of just one of these substances.  Adenoma—growth pattern is disordered. May exhi-
 Often associated with inflammatory lesions of the bit some atypia
gallbladder.  Adenocarcinoma—increased cellular atypia. Disor-
dered growth pattern. Invasive growth.
Differential diagnosis:  Adenomatoid change/glandular metaplasia—focal or
diffuse proliferation of epithelial cells forming gland-
 Mineralization—associated with necrosis, necrotic ular structures. Cells are well differentiated, usually
benign or malignant tumor, and inflammatory columnar, and eosinophilic, with little or no cellular
conditions. atypia. Distinct eosinophilic crystals are present in
 Neoplasia (Gross) —histologically, it is neoplastic. cytoplasm, or in lumen of glands.
 Inflammation—inflammatory exudates may be seen
without gallstone formation. Comment: Details related to gallbladder hyperplasia can be
 Parasites—can be seen histologically as parasites. found in several references (Deschl et al. 2001; Harada et al.
42S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

1996, 1999; Yoshitomi, Alison, and Boorman 1986; Yoshitomi et al. 2001; Harada et al. 1996, 1999; Lewis 1984; Yoshitomi,
and Boorman 1994). Alison, and Boorman 1986; Yoshitomi and Boorman 1994).

Adenoma, Gallbladder (Figures 181–183) Adenocarcinoma, Gallbladder


Synonym: Adenoma, papillary. Pathogenesis: Arises from epithelium of the gallbladder.
Diagnostic features:
Pathogenesis: Arises from epithelium of gallbladder.
Diagnostic features:  May be a sessile broad-based mass or characterized
by diffusely thickening of mucosa.
 In general well differentiated and solitary.  Growth pattern is disordered.
 Growth is disordered, predominantly papillary- or  Cellular atypia is present.
cauliflower-like.  Cytoplasm is scant and basophilic.
 Epithelium is single layered, but occasionally may be  Nuclei are enlarged.
multilayered.  Mitotic figures are common.
 Amount of fibrovascular stroma is variable.  Invasion of the wall of the gallbladder or of adjacent
 Cells may exhibit some atypia, with enlarged nuclei tissue.
or even giant nuclei with one or two nucleoli.
 Mitotic figures may be present. Differential diagnosis:
 Often inflammatory cellular infiltration and focal
mineralization of the stroma may be present.  Hyperplasia—lesion is small. Exhibits only little evi-
dence of atypia.
Differential diagnosis:  Adenoma—no evidence of invasive growth. Less cel-
lular atypia.
 Hyperplasia (focal)—small lesion. Exhibits only lit-  Adenomatoid change (glandular metaplasia)—
tle evidence of atypia. focal or diffuse proliferation of epithelial cells
 Adenocarcinoma—increased cellular atypia. Disor- forming glandular structures. Cells are well differ-
dered growth pattern. Invasive growth. entiated, usually columnar, and eosinophilic, with
 Adenomatoid change/glandular metaplasia—focal or little or no cellular atypia. Distinct eosinophilic
diffuse proliferation of epithelial cells forming gland- crystals are present in cytoplasm, or in lumen of
ular structures. Cells are well differentiated, usually glands.
columnar, and eosinophilic, with little or no cellular
atypia. Distinct eosinophilic crystals are present in Comment: Benign and malignant epithelial neoplasms of the
cytoplasm, or in lumen of glands. gallbladder are described in several published references
(Deschl et al. 2001; Harada et al. 1996, 1999; Lewis 1984;
Comment: Benign and malignant epithelial neoplasms of the Yoshitomi, Alison, and Boorman 1986; Yoshitomi and
gallbladder are described in several published references (Deschl Boorman 1994).
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 43S

FIGURE 1.—Gross appearance and tissue trimming recommendations for a normal rodent liver. Ref. to http://reni.item.fraunhofer.de/reni/trimming/
index.php. FIGURE 2.— Two-dimensional microarchitecture of the liver. FIGURE 3.—Rat liver. Hepatodiaphragmatic nodule. FIGURE 4.—Rat liver.
Hepatodiaphragmatic nodule with intranuclear inclusions (chromatin). Higher magnification of Figure 3. FIGURE 5.—Rat liver. Macrovesicular fatty
change. FIGURE 6.—Rat liver. Macrovesicular fatty change. Higher magnification of Figure 5.
44S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 7.—Mouse liver. Fatty change, microvesicular. FIGURE 8.—Mouse liver. Mixture of fatty change and cytoplasmic glycogen. FIGURE 9.—
Mouse liver. Mixture of fatty change and cytoplasmic glycogen. Higher magnification of Figure 8. FIGURE 10.—Mouse liver. Tension lipidosis.
FIGURE 11.—Mouse liver. Tension lipidosis. Higher magnification of Figure 10. FIGURE 12.—Rat liver. Focal fatty change.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 45S

FIGURE 13.—Rat liver. Focal fatty change. Higher magnification of Figure 12. FIGURE 14.—Rat liver. Phospholipidosis. FIGURE 15.—Rat liver.
Phospholipidosis. Higher magnification of Figure 14. FIGURE 16.—Rat liver. Phospholipidosis. EM concentric membrane bound lysosomal myeloid
bodies/lamellar bodies. FIGURE 17.—Rat liver. Phospholipidosis. Central microvesiculation; positive LAMP-2 staining. FIGURE 18.—Mouse liver.
Amyloidosis.
46S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 19.—Mouse liver. Amyloidosis. Higher magnification of Figure 18. FIGURE 20.—Mouse liver. Focal mineralization associated with centri-
lobular necrosis. FIGURE 21.—Mouse liver. Pigment deposition in a focus of histiocytes. FIGURE 22.—Rat liver. Pigmentation. FIGURE 23.—Mouse
liver. Centrilobular hypertrophy with cholestasis. FIGURE 24—Mouse liver. Cholestasis.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 47S

FIGURE 25.—Rat liver. Pigment in hypertrophic hepatocytes consistent with bile. Cholestasis. FIGURE 26.—Mouse liver. Hyalinosis of hyper-
plastic bile ducts with crystal formation and peribiliary inflammatory cell infiltrate. FIGURE 27.—Mouse liver. Hyalinosis of hyperplastic bile
ducts with crystal formation and peribiliary inflammatory cell infiltrate. Higher magnification of Figure 26. FIGURE 28.—Mouse liver.
Hyalinosis of hyperplastic bile ducts with crystal formation. FIGURE 29.—Mouse liver. Numerous intracytoplasmic hyaline bodies in an hepa-
tocellular adenoma. FIGURE 30.—Mouse liver. Intranuclear inclusion body, cytoplasmic invagination.
48S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 31.—Mouse liver. Intranuclear inclusion bodies. FIGURE 32.—Mouse liver. Cytoplasmic inclusions in an hepatocellular adenoma. FIGURE
33.—Mouse liver. Plasma influx. FIGURE 34.—Mouse liver. Centrilobular hepatocellular hypertrophy. FIGURE 35.—Mouse liver. Higher magni-
fication of centrilobular hepatocellular hypertrophy. FIGURE 36.—Mouse liver. Hepatocellular hypertrophy.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 49S

FIGURE 37.—Mouse liver. Hepatocellular hypertrophy. FIGURE 38.—Mouse liver. Centrilobular hepatocellular hypertrophy. FIGURE 39.—Rat
liver. Hepatocellular hypertrophy with eosinophilic granular cytoplasm following treatment with a peroxisome proliferating xenobiotic.
FIGURE 40.—Rat liver. Hepatocellular hypertrophy following treatment with a peroxisome proliferating xenobiotic. FIGURE 41.—Rat liver.
Hepatocyte enlargement confirmed as mitochondrial hypertrophy. FIGURE 42.—Rat liver. Atrophy.
50S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 43.—Mouse liver. Hepatic atrophy. FIGURE 44.—Mouse liver. Cytoplasmic alteration. FIGURE 45.—Mouse liver. Hydropic degeneration.
FIGURE 46.—Rat liver. Cystic degeneration. FIGURE 47.—Rat liver. Cystic degeneration. FIGURE 48.—Mouse liver. Apoptosis.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 51S

FIGURE 49.—Mouse liver. Apoptosis. FIGURE 50.Mouse liver. Apoptosis. FIGURE 51 and 53 Mouse liver. Necrosis, focal. FIGURE 52. Mouse liver.
Necrosis, multifocal. FIGURE 54. Rat liver. Bridging centrilobular necrosis.
52S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 55.—Rat liver. Bridging centrilobular necrosis. Higher magnification of Figure 54. FIGURE 56.—Rat liver. Centrilobular necrosis with early
bridging. FIGURE 57.—Rat liver. Centrilobular necrosis with early bridging. Higher magnification of Figure 56. FIGURE 58.—Rat liver. Centrilob-
ular bridging necrosis with mineralization. FIGURE 59.—Rat liver. Centrilobular bridging necrosis with mineralization. Higher magnification of
Figure 58. FIGURE 60.—Rat liver. Midzonal necrosis.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 53S

FIGURE 61.—Rat liver. Midzonal necrosis. Higher magnification of Figure 60. FIGURE 62.—Mouse liver. Periportal necrosis. FIGURE 63.—Mouse
liver. Diffuse necrosis with inflammation and bile duct hyperplasia in lower left of figure. FIGURE 64.—Mouse liver. Diffuse necrosis.
Multinucleated giant cells in MHV infection. Higher magnification of Figure 63. FIGURE 65.—Mouse liver. Karyocytomegaly. FIGURE 66.—Mouse
liver. Multinucleated hepatocytes. Karyocytomegaly.
54S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 67.—Rat liver. Biliary cysts. FIGURE 68.—Rat liver. Biliary cysts. FIGURE 69.—Rat liver. Biliary cysts. Higher magnification of Figure 68.
FIGURE 70.—Mouse liver. Generalized inflammation, postnecrotic mild fibrosis. Mouse hepatitis virus infection. FIGURE 71.—A descriptive
approach for classifying inflammatory responses in the liver. FIGURE 72.—Mouse liver. Focal neutrophil infiltrate associated with hepatocyte
necrosis.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 55S

FIGURE 73.—Mouse liver. Multifocal mononuclear cell infiltrates. Norovirus infection. FIGURE 74.—Rat liver. Infiltration, mononuclear. Granu-
lomatous inflammation with pigment-laden histiocytes and multinucleated giant cells. FIGURE 75.—Rat liver. Mixed inflammatory cell infiltrate
and granulomatous inflammation. FIGURE 76.—Infiltration, mononuclear (microgranulomas). FIGURE 77.—Rat liver. Infiltration, mononuclear.
FIGURE 78.—Rat liver. Infiltration, mononuclear. Granulomatous inflammation (cholesterol granuloma).
56S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 79.—Rat liver. Infiltration, mononuclear. Granulomatous inflammation (cholesterol granuloma). Higher magnification of Figure 78.
FIGURE 80.—Mouse liver. Focal neutrophil infiltrate. FIGURE 81.—Mouse liver. Periportal inflammatory cell infiltrate. FIGURE 82.—Rat liver.
Fibrosis. Early bridging between lobules. FIGURE 83.—Rat liver. Fibrosis. Higher magnification of Figure 82. FIGURE 84.—Mouse liver. Silver
stain demonstrating Helicobacter sp.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 57S

FIGURE 85.—Rat liver. Chronic passive congestion. FIGURE 86.—Rat liver. Angiectasis. FIGURE 87.—Rat liver. Angiectasis. FIGURE 88.—Rat liver.
Thrombosis with associated area of necrosis. FIGURE 89.—Mouse liver. Infarcted lobe. FIGURE 90.—Mouse liver. Infarct.
58S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 91.—Rat liver. Endothelial karyomegaly (sinusoidal cell karyomegaly). FIGURE 92.—Mouse liver. Increased mitoses. FIGURE 93.—Rat
liver. Diffuse basophilic focus. FIGURE 94.—Rat liver. Diffuse basophilic focus. Higher magnification of Figure 93. FIGURE 95.—Rat liver. Baso-
philic (tigroid) focus of cellular alteration. FIGURE 96.—Rat liver. Basophilic focus of cellular alteration with cystic degeneration.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 59S

FIGURE 97.—Rat liver. Basophilic focus of cellular alteration. FIGURE 98.—Rat liver. Eosinophilic focus of cellular alteration. FIGURE 99.—Rat
liver. Eosinophilic focus of cellular alteration. Higher magnification of Figure 98. FIGURE 100.—Rat liver. Eosinophilic focus of cellular alteration
with pale pink cytoplasm. FIGURE 101.—Mouse liver. Large basophilic focus of cellular alteration with glycogen present in centrally located hepa-
tocytes. FIGURE 102.—Mouse liver. Large basophilic focus of cellular alteration with glycogen present in centrally located hepatocytes. Higher
magnification of the edge of Figure 102.
60S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 103.—Rat liver. Mixed cell foci. FIGURE 104.—Rat liver. Mixed cell focus. Higher magnification of Figure 103. FIGURE 105.—Mouse liver.
Clear cell focus. FIGURE 106.—Mouse liver. Clear cell focus. Higher magnification of Figure 105. FIGURE 107.—Rat liver. Clear cell focus of
cellular alteration. FIGURE 108.—Rat liver. Amphophilic focus.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 61S

FIGURE 109.—Rat liver. Early non-regenerative hyperplasia. FIGURE 110.—Rat liver. Non-regenerative hyperplasia. FIGURE 111.—Rat liver.
Regenerative hyperplasia. FIGURE 112.—Rat liver. Regenerative hyperplasia. Higher magnification of Figure 111. FIGURE 113.—Rat liver. Regen-
erative hyperplasia. Higher magnification of Figure 111. FIGURE 114.—Mouse liver. Kupffer cell hyperplasia.
62S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 115.—Mouse liver. Kupffer cell hyperplasia. FIGURE 116.—Mouse liver. Ito cell hyperplasia. FIGURE 117.—Mouse liver. Ito cell hyper-
plasia. Higher magnification of Figure 116. FIGURE 118.—Mouse liver. Ito cell hyperplasia, multifocal. FIGURE 119.—Rat liver. Bile duct hyper-
plasia. FIGURE 120.—Rat liver. Bile duct hyperplasia with associated mononuclear inflammatory cell infiltrate.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 63S

FIGURE 121.—Rat liver. Bile duct hyperplasia with mononuclear inflammatory cell infiltration and early fibrosis. FIGURE 122.—Rat liver. Bile
duct hyperplasia. FIGURE 123.—Rat liver. Cholangiofibrosis. FIGURE 124.—Rat liver. Cholangiofibrosis. FIGURE 125.—Rat liver. Cholangiofibro-
sis. FIGURE 126.—Rat liver. Cholangiofibrosis.
64S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 127.—Rat liver. Oval cell hyperplasia. FIGURE 128.—Mouse liver. Oval cell hyperplasia. FIGURE 129.—Mouse liver. Oval cell hyperplasia.
FIGURE 130.—Mouse liver. Large hepatocellular adenoma. FIGURE 131.—Mouse liver. Hepatocellular adenoma, eosinophilic. FIGURE 132.—
Mouse liver. Hepatocellular adenoma, eosinophilic. Higher magnification of Figure 131.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 65S

FIGURE 133.—Mouse liver. Hepatocellular adenoma. FIGURE 134.—Mouse liver. Hepatocellular adenoma. High magnification of Figure 133.
FIGURE 135.—Mouse liver. Hepatocellular carcinoma. FIGURE 136.—Mouse liver. Hepatocellular carcinoma. Prominent glandular formation.
Higher mignification of Figure 135. FIGURE 137.—Mouse liver. Hepatocellular carcinoma. Prominent glandular and trabecular formation. Higher
mignification of Figure 135. FIGURE 138.—Mouse liver. Hepatocellular carcinoma with trabecular and glandular formation.
66S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 139.—Mouse liver. Hepatocellular carcinoma arising in an hepatocellular adenoma. FIGURE 140.—Mouse liver. Hepatocellular carcinoma
arising in a hepatocellular adenoma. Higher magnification of Figure 139. FIGURE 141.—Mouse liver. Hepatoblastoma. FIGURE 142.—Mouse liver.
Hepatoblastoma. Higher magnification of Figure 141. FIGURE 143.—Mouse liver. Hepatoblastoma. Osseous metaplasia is present. FIGURE 144.—
Mouse liver. Hepatoblastoma. High magnification of Figure 143.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 67S

FIGURE 145.—Mouse liver. Cholangioma. FIGURE 146.—Mouse liver. Cholangioma. Higher magnification of Figure 145. FIGURE 147.—Mouse liver.
Cholangiocarcinoma. FIGURE 148.—Mouse liver. Cholangiocarcinoma. Higher magnification of Figure 147. FIGURE 149.—Rat liver. Hepatocholan-
gioma (adenoma, hepatocholangial). FIGURE 150.—Rat liver. Hepatocholangioma (adenoma, hepatocholangial). Higher magnification of Figure 149.
68S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 151.—Rat liver. Hepatocholangiocarcinoma (Carcinoma, hepatocholangial). FIGURE 152.—Rat liver. Hepatocholangiocarcinoma
(Carcinoma, hepatocholagial). High magnification of Figure 151. FIGURE 153.—Mouse liver. Ito cell tumor. FIGURE 154.—Mouse liver. Ito cell tumor.
Higher magnification of Figure 153. FIGURE 155.—Mouse liver. Histiocytic sarcoma. Perivascular infiltration. FIGURE 156.—Mouse liver.
Hemangioma.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 69S

FIGURE 157.—Mouse liver. Hemangioma. High magnification of Figure 156. FIGURE 158.—Mouse liver. Hemangiosarcoma. FIGURE 159.—Mouse
liver. Hemangiosarcoma. Higher magnification of Figure 158. FIGURE 160.—Mouse liver. Hemangiosarcoma. Higher magnification of Figure 159.
FIGURE 161.—Mouse liver. Extramedullary hematopoiesis. FIGURE 162.—Mouse liver. Extramedullary hematopoiesis. Myelopoiesis.
70S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 163.—Mouse liver. Intrahepatic erythrocytes. FIGURE 164.—Rat liver. Pancreatic acinar metaplasia. FIGURE 165.—Rat liver. Ectopic
acinar pancreas. FIGURE 166.—Rat liver. Hepatocyte glandular metaplasia. FIGURE 167.—Rat liver. Hepatocyte glandular metaplasia. FIGURE
168.—Mouse liver. Subintimal intravascular hepatocyte proliferation.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 71S

FIGURE 169.—Mouse gallbladder. Heterotopic acinar pancreas. FIGURE 170.—Mouse gallbladder. Heterotopic acinar pancreas. Higher magni-
fication of Figure 169. FIGURE 171.—Mouse gallbladder. Hyalinosis. Part of a biliary calculus is present. FIGURE 172.—Mouse gallbladder.
Hyalinosis with crystal formation. FIGURE 173.—Mouse gallbladder. Hyalinosis and mucosal hyperplasia. Higher magnification of Figure 172.
FIGURE 174.—Mouse gallbladder. Hyalinosis with crystal formation. Higher magnification of Figure 172.
72S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

FIGURE 175.—Mouse gallbladder. Biliary calculus (biliary stone). FIGURE 176.—Mouse gallbladder. Hyalinosis and biliary calculus.
FIGURE 177. Mouse gallbladder. Cholecystitis. FIGURE 178.—Mouse gallbladder. Submucosal edema. FIGURE 179.—Mouse gallbladder. Mucosal
hyperplasia. FIGURE 180.—Mouse gallbladder. Mucosal hyperplasia. Higher magnification of Figure 179.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 73S

FIGURE 181.—Mouse gallbladder. Papillary adenoma. FIGURE 182.—Mouse gallbladder. Papillary adenoma. FIGURE 183.—Mouse gallbladder.
Papillary adenoma. Higher magnification of Figure 182.
74S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

ACKNOWLEDGEMENTS Bergs, V. V., and Scotti, T. M. (1967). Virus-induced peliosis hepatitis in rats.
Science 158, 377–78.
The authors wish to express their thanks for the excellent Binhazim, A. A., Coghlan, L. G., and Walker, C. (1994). Spontaneous heman-
editorial support and suggestions provided by John Vahle, Bob giosarcoma in the tail of a Long-Evans rat carrying the Eker mutation.
Greenhill, John Foster, Chirukandath Gopinath, Rupert Lab Anim Sci 44, 191–94.
Booth, C. J., and Sundberg, J. P. (1996). Hemangiomas and hemangiosarco-
Kellner, Beth Mahler, Stuart Levin, Ken Schafer, Gordon Hard, mas. In Pathobiology of the Aging Mouse. Vol 1. Cardiovascular
Ian Pyrah, Charlotte Keenan, Maria-Luisa Phan Lung Whu, System (U. Mohr, D. L. Dungworth, C. C. Capen, W. W. Carlton, J.
and Suzy Tirtodikromo. Special thanks to Beth Mahler and P. Sundberg, and J. M. Ward, eds.), pp 393–401. ILSI Press,
Emily Singletary for photography support. The majority of the Washington, DC.
photomicrographs used in this document were provided cour- Bourdeau, A., Faughnan, M. E., McDonald, M. L., Paterson, A. D., Wanless, I.
R., and Letarte, M. (2001). Potential role of modifier genes influencing
tesy of the National Toxicology Program Archives, National transforming growth factor-beta1 levels in the development of vascular
Institute of Environmental Health Sciences, Research Triangle defects in endoglin heterozygous mice with hereditary hemorrhagic tel-
Park, North Carolina. angiectasia. Am J Pathol 158, 2011–020.
Boyd, M. R. (1981). Toxicity mediated by reactive metabolites of furans. Adv
Exp Med Biol 136, B865–79.
Breitschwerdt, E. B., and Kordick, D. L. (2000). Bartonella infection in ani-
REFERENCES mals: Carriership, reservoir potential, pathogenicity, and zoonotic poten-
Aigelsreiter, A., Janig, E., Stumptner, C., Fuchsbichler, A., Zatloukal, K., and tial for human infection. Clin Microbiol Rev 13, 428–38.
Denk, H. (2007). How a cell deals with abnormal proteins. Pathogenetic Brooks, P. N., and Roe, F. J. C. (1985). Hepatocellular adenoma, liver, rat. In
mechanisms in protein aggregation diseases. Pathobiology 74, 145–58. Monographs on Pathology of Laboratory Animals. Digestive System
Anderson, N., and Borlak, J. (2006). Drug-induced phospholipidosis. FEBS (T. C. Jones, U. Mohr, and R. D. Hunt, eds.), pp. 47–52. Springer,
Lett 580, 5533–540. New York.
Atzori, L., and Congiu, L. (1996). Effect of verapamil on allyl alcohol hepato- Browning, F. M., Schroeder, C. R., and Berringer, O. M. (1974). An Atlas and
toxicity. Drug Metabol Drug Interact 13, 87–98. Dissection Manual of Rhesus Monkey Anatomy. 2nd printing. Rose Print-
Aydin, G., Ozcelik, N., Cicek, E., and Soyoz, M. (2003). Histopathologic ing Company Inc., Tallahassee, Florida.
changes in liver and renal tissues induced by Ochratoxin A and melatonin Bruguera, M. (1999). Biopsia hepática en las enfermedades hepáticas por
in rats. Hum Exp Toxicol 22, 383–91. depósito. Gastroenterologı´a Y Hepatologı´a 22, 20–4.
Babich, M. A., Chen, S. B., Greene, M. A., Kiss, C. T., Porter, W. K., Smith, T. Bruguera, M., Aranguibel, F., Ros, E., and Rodes, J. (1978). Incidence and clin-
P., Wind, M. L., and Zamula, W. W. (2004). Risk assessment of oral ical significance of sinusoidal dilatation in liver biopsies. Gastroenterol-
exposure to diisononyl phthalate from children’s products. Regul Toxicol ogy 75, 474–78.
Pharmacol 40, 151–67. Bruni, C. (1960). Hyaline degeneration of rat liver cells studied with the elec-
Bach, U., Hailey, J. R., Hill, G. D., Kaufmann, W., Latimer, K. S., Malarkey, tron microscope. Lab Invest 9, 209–15.
D. E., Maronpot, R. M., and Elmore, S. A. (2010). Proceedings of the BSTP. (2007). Modular Education Programme in Toxicological Pathology.
2009 National Toxicology Program Satellite Symposium. Toxicol Pathol Module 8: Liver. Cambridge University, Department of Pathology, Cam-
38, 9–36. bridge, UK.
Bannasch, P. (2003). Comments on R. Karbe and R. L. Kerlin (2002). Cystic Burek, J. D. (1978). Pathology of Aging Rats. CRC Press, West Palm Beach,
degeneration/spongiosis hepatis (Toxicol Pathol 30, 216-227). Toxicol Florida.
Pathol 5, 566–70. Burt, A. D., Portmann, B. C., and MacSween, R. N. M. (2002). Liver pathology
Bannasch, P., Bloch, M., and Zerban, H. (1981). Spongiosis hepatis. Specific associated with diseases of other organs or systems. In Pathology of the
changes of the perisinusoidal liver cells induced in rats by N- Liver (R. N. M. MacSween, ed.). 4th edition, pp. 827–83. Churchill
nitrosomorpholine. Lab Invest 44, 252–64. Livingstone, London.
Bannasch, P., Brenner, U., and Zerban, H. (1985). Cholangiofibroma and Carlton, W. W., Ernst, H., Faccini, J. M., Greaves, P., Krinke, G. J., Long,
cholangiocarcinoma, liver, rat. In Monographs on Pathology of Labora- P. H., Maekawa, A., Newsholme, S. J., and Weisse, G. (1992). Soft tissue
tory Animals. Digestive System (T. C. Jones, U. Mohr, and R. D. Hunt, and musculoskeletal system 2. In International Classification of Rodent
eds.), pp. 52–65. Springer, New York. Tumours—Part 1—The Rat (U. Mohr, ed.), p. 31. WHO IARC Scientific
Bannasch, P., Wayss, K., and Zerban, H. (1997). Peliosis hepatis, rodents. Publications, Lyon, France.
In Digestive System (T. C. Jones, U. Mohr, and R. D. Hunt, eds.), Carter, R. L. (1973). Tumours of the soft tissues. In Pathology of Tumours in
pp. 154–60. Springer-Verlag, Berlin, New York. Laboratory Animals. Vol I. Tumours of the Rat, Part 1 (V. S. Turusov,
Bannasch, P., and Zerban, H. (1990). Tumours of the liver. In Pathology of ed.), pp 151–68. IARC Scientific Publications, Lyon, France.
Tumours in Laboratory Animals. Vol. I: Tumours of the Rat (V. S. Chang, H. J., Suh J. I., and Kwon, S. Y. (1999). Gallstone formation and gall-
Turusov and U. Mohr, eds.), pp. 199–240, 2nd edition. IARC Scientific bladder mucosal changes in mice fed a lithogenic diet. J Korean Med Sci
Publications No. 99, Lyon, France. 14, 286–92.
Bannasch, P., Zerban, H., and Hacker, H. J. (1985). Foci of altered hepatocytes, Chatman, L. A., Morton, D., Johnson, T. O., and Anway, S. D. (2009). A strat-
rat. In Monographs on Pathology of Laboratory Animals. Digestive Sys- egy for risk management of drug-induced phospholipidosis. Toxicol
tem (T. C. Jones, U. Mohr, and R. D. Hunt, eds.), pp 10–30. Springer, Pathol 37, 997–1005.
New York. Cheng, K. K. (1956). Experimental studies on the mechanism of the zonal dis-
Barni, S., and Bernocchi, G. (1991). Internalization of erythrocytes into liver tribution of beryllium liver necrosis. J Pathol Bacteriol 71, 265–76.
parenchymal cells in naturally hibernating frogs (Rana esculenta L.). Chengelis, C. P. (1988). Changes in hepatic glutathione concentrations during
J Exp Zoology 258, 143–50. carbon disulfide induced hepatotoxicity in the rat. Res Commun Chem
Belloni, A. S. Rebuffat, P., Gottardo, G., Meneghelli, V., Coi, A., Mazzocchi, Pathol Pharmacol 61, 97–109.
G., and Nussdorfer, G. G. (1988). A morphometric study of the effects of Chipchase, M. D., O’Neill, M., & Melton, D. W. (2003). Liver biology
short-term starvation on rat hepatocytes. J Submicrosc Cytol Pathol 20, and pathobiology—Characterization of premature liver polyploidy
751–57. in DNA repair (Ercc1)-deficient mice. Hepatology. Official Journal
Beregi, E., Penzes, L., Regius, O., and Izsak, J. (1987). Biological changes and of the American Association for the Study of Liver Diseases 38,
diseases in aged CBA/Ca mice. Compr Gerontol [A] 1, 72–4. 958–66.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 75S

Churukian, C. J. (2002). Pigments and minerals In Theory and Practice of His- Enomoto, M., Naoe, S., Harada, M., Miyata, K., Saito, M., and Noguchi, Y.
tological Techniques (J. D. Bancroft and M. Gamble, eds.), pp. 243–67. (1974). Carcinogenesis in extrahepatic bile duct and gallbladder—carci-
Churchill Livingstone, Edinburgh, Scotland. nogenic effect of N-hydroxy-2-acetamidofluorene in mice fed a ‘‘gall-
Clemens, M., Jost, J. O., Kautz, G., Schwering, H., Kessler, B., and Galansky, stone-inducing’’ diet. Jpn J Exp Med 44, 37–54.
M. (1980). Das Caroli-Syndrom. Der Chirurg; Zeitschrift Fur Alle Enzan, H. (1985). Proliferation of Ito cells (fat-storing cells) in acute carbon
Gebiete Der Operativen Medizen 51, 219–22. tetrachloride liver injury. A light and electron microscopic autoradio-
Coe, J. E., and Ross, M. J. (1990). Amyloidosis and female protein in the Syr- graphic study. Acta Pathol Jpn 35, 1301–308.
ian hamster. Concurrent regulation by sex hormones. J Exp Med 171, Eustis, S. L., Boorman, G. A., Harada, T., and Popp, J. A. (1990). Pathology of
1257–267. the Fischer Rat. Reference and Atlas (G. A. Boorman, S. L. Eustis, M. R.
Comporti, M. (1985). Lipid peroxidation and cellular damage in toxic liver Elwell, C. A. Montgomery, Jr, and W. F. MacKenzie, eds.), pp. 71–92.
injury. Lab Inves 53, 599–623. Academic Press, San Diego, California.
Copple, B. L., Banes, A., Ganey, P. E., and Roth, R. A. (2002). Endothelial cell Evans, J. G., Collins, M. A., Lake, B. G., and Butler, W. H. (1992). The histol-
injury and fibrin deposition in rat liver after monocrotaline exposure. ogy and development of hepatic nodules and carcinoma in C3H/He and
Toxicol Sci 65, 309–18. C57BL/6 mice following chronic phenobarbitone administration. Toxicol
Cullen, J. M. (2005). Mechanistic classification of liver injury. Toxicol Pathol Pathol 20, 585–94.
33, 6–8. Evans, J. G., and Lake, B. G. (1998). The digestive system II: The hepatobiliary
Datta, K., Chin, A., Ahmed, T., Qing, W. G., Powell, K. L., Simhambhatla, P., system In Target Organ Pathology—A Basic Text (J. Turton and J. Hoo-
MacLeod, M. C., Stoica, G., and Kehrer, J. P. (1998). Mixed effects of 2, son, eds.), pp. 61–97. Taylor & Francis Ltd., London.
6-dithiopurine against cyclophosphamide mediated bladder and lung Faa, G., Van, E. P., Roskams, T., Miyazaki, H., Serreli, S., Ambu, R., & Des-
toxicity in mice. Toxicology 125, 1–11. met, V. J. (1998). Expression of cytokeratin 20 in developing rat liver and
Davenport, M., Gonde, C., Redkar, R., Koukoulis, G., Tredger, M., Mieli- in experimental models of ductular and oval cell proliferation. J Hepatol
Vergani, G., Portmann, B., and Howard, E. (2001). Immunohistochemis- 29, 628–33.
try of the liver and biliary tree in extrahepatic biliary atresia. J Ped Surg Faccini, J. M., Abbott, D. P., and Paulus, G. J .J. (1990). Mouse Histopathol-
36, 1017–025. ogy. A Glossary for Use in Toxicity and Carcinogenicity Studies. Else-
Denda, A., Kitayama, W., Kishida, H., Murata, N., Tsutsumi, M., Tsujiuchi, T., vier, Oxford, UK.
Nakae, D., and Konishi, Y. (2002). Development of hepatocellular ade- Feldmann, G. (1997). Liver apoptosis. J Hepatol 26 Suppl 2, 1–11.
nomas and carcinomas associated with fibrosis in C57BL/6J male mice Foster, J. R. (2000). Cell death and cell proliferation in the control of normal
given a choline-deficient, L-amino acid-defined diet. Jpn J Cancer Res and neoplastic tissue growth. Toxicol Pathol 28, 441–46.
93, 125–32. Fox, J. G., Anderson, L. C., Loew, F. M., and Quimby, F. W. (2002).
DePass, L. R., Garman, R. H., Woodside, M. D., Giddens, W. E., Maronpot, R. R., Laboratory Animal Science. 2nd edition. Academic Press, San Diego,
and Weil, C. S. (1986). Chronic toxicity and oncogenicity studies of ethylene California.
glycol in rats and mice. Fundam Appl Toxicol 7, 547–65. Frémond, B., Jouan, H., Sameh, A. H., LeGall, E., Bergerson, C., Manac’h, A.,
Derelanko, M. J. (2000). Acute/chronic toxicology. In Toxicologist’s Pocket Gruel, Y., and Babut, J. M. (1987). Tumeurs du foie secondaires á
Handbook, p. 13. CRC Press, Boca Raton, Florida. l’androgénothérapie. A propos de 2 cas chez l’enfant. Chirurgie
Deschl, U., Cattley, R., Harada, T., Küttler, K., Hailey, J. R., Hartig, F., Pe´diatrique 28, 97–101.
Leblanc, B., Marsman, D. S., and Shirai, T. (2001). Liver, gallblad- Frith, C. H., and Ward, J. M. (1979). A morphologic classification of prolifera-
der, and exocrine pancreas, In International Classification of tive and neoplastic hepatic lesions in mice. J Environ Pathol Toxicol 3,
Rodent Tumours: The Mouse (U. Mohr, ed.), pp. 59–86. Springer, 329–51.
New York. Frith, C. H., and Ward, J. M. (1988). Color Atlas of Neoplastic and Non-
Deschl, U., Ernst, H., Frantz, J. D., Goodman, D. G., Hartig, F., Konishi, Y., Neoplastic Lesions in Aging Mice. Elsevier, Oxford, UK.
Küttler, K., Popp, J. A., Püschner, H., Squire, R. A., Tatematsu, M., Frith, C. H., Ward, J. M., and Turusov, V. S. (1994). Tumours of the Liver.
Tuch, K., Ward, J. M., and Whiteley L. O. (1997). Digestive system. Pathology of Tumours in Laboratory Animals. Vol 2. Tumours of the
In International Classification of Rodent Tumours Part 1—The Rat Mouse, 2nd edition. IARC Scientific Publications, Lyon, France.
(U. Mohr, ed.), pp. 65–98. WHO IARC Scientific Publications, Lyon, Frith, C. H., and Wiley, L. (1982). Spontaneous hepatocellular neoplasms and
France. hepatic hemangiosarcomas in several strains of mice. Lab Anim Sci 32,
Diwan, B. A., Ward, J. M., Ramljak, D., and Anderson, L. M. (1997). 157–62.
Promotion by Helicobacter hepaticus-induced hepatitis of hepatic tumors Gad, S. C., and Rousseaux, C. G. (2002). Use and misuse of statistics as an aid
initiated by N-nitrosodimethylamine in male A/JCr mice. Toxicol Pathol in study interpretation. In Handbook of Toxicologic Pathology (W. M.
25, 597–605. Haschek, C. G. Rousseaux, and M. A. Wallig, eds.), Vol. 1, pp. 327–
Diwan, B. A., Ward, J. M., and Rice, J. M. (1989). Promotion of malignant 418. Academic Press, San Diego, California.
‘‘embryonal’’ liver tumors by phenobarbital: Increased incidence and Gebbia, N., Leto, G., Gagliano, M., Tumminello, F. M., and Rausa, L. (1985).
shortened latency of hepatoblastomas in (DBA/2 x C57BL/6)F1 mice ini- Lysosomal alterations in heart and liver of mice treated with doxorubicin.
tiated with N-nitrosodiethylamine. Carcinogenesis 10, 1345–348. Cancer Chemotherapy and Pharmacology 15, 26–30.
Dixon, D., Yoshitomi, K., Boorman, G. A., and Maronpot, R. R. (1994). ‘‘Lipo- Geller, S. A., Dahll, D., and Alsabeh, R. (2008). Application of immunohisto-
matous’’ lesions of unknown cellular origin in the liver of B6C3F1 mice. chemistry to liver and gastrointestinal neoplasms: Lysosomal alterations
Vet Pathol 31, 173–82. in heart and liver of mice treated with doxorubicin. Arch Pathol Lab Med
Dowling, R. H. (2000). Review: Pathogenesis of gallstones. Aliment Pharma- 132, 490–99.
col Ther 14, Suppl 2, 39–47. Gerlyng, P., Abyholm, A., Grotmol, T., Erikstein, B., Huitfeldt, H. S., Stokke,
Edwards, R., Colombo, T., and Greaves, P. (2002). ‘‘Have you seen this?’’ T., and Seglen, P. O. (1993). Binucleation and polyploidization patterns
peliosis hepatis. Toxicol Pathol 30, 521–23. in developmental and regenerative rat liver growth. Cell Proliferation 26,
Elmore, S. (2007). Apoptosis: A review of programmed cell death. Toxicol 557–65.
Pathol 35, 495–516. Giddens, W. E., and Renne, R. A. (1985). Haemangiosarcoma, nasal cavity,
Emerson, Q. B., Nachtnebel, K. L., Penkava, R. R., and Rothenberg, J. (1980). mouse. In Monographs on Pathology of Laboratory Animals. Respiratory
Oral-contraceptive-associated liver tumors. Lancet 1, 1251. Systems (T. C. Jones, U. Mohr, and R. D. Hunt, eds.), pp 72–4. Springer,
Engelhardt, N. V. (1997). Oval cells in rodent liver, mouse, rat. In Digestive New York.
System (T. C. Jones, J. A. Popp, and U. Mohr, eds.), pp. 162–66. ILSI Gkretsi, V., Mars, W. M., Bowen, W. C., Barua, L., Yang, Y., Guo, L.,
Monograph, ILSI Press, Washington, DC. St-Arnaud, R., Dedhar, S., Wu, C., and Michalopoulos, G. K.
76S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

(2007). Loss of integrin linked kinase from mouse hepatocytes in Hailey, J. R., Walker, N. J., Sells, D. M., Brix, A. E., Jokinen, M. P., and Nyska,
vitro and in vivo results in apoptosis and hepatitis. Hepatology 45, A. (2005). Classification of proliferative Hepatocellular lesions in Harlan
1025–034. Sprague-Dawley rats chronically exposed to dioxin-like compounds.
Goel, S. K., Lalwani, N. D., and Reddy, J. K. (1986). Peroxisome proliferation Toxicol Pathol 33, 165–74.
and lipid peroxidation in rat liver. Cancer Res 46, 1324–330. Halliwell, W. H. (1997). Cationic amphiphilic drug-induced phospholipidosis.
Gokalp, O., Gulle, K., Sulak, O., Cicek, E., and Altuntas, I. (2003). The effects Toxicol Pathol 25, 53–60.
of methidathion on liver: Role of vitamins E and C. Toxicol Ind Health Handley, D. A., Chien, S., and Arbeeny, C. M. (1983). Ultrastructure of hepatic
19, 63–7. cholesterol crystals in the hypercholesterolemic-diabetic rat. Pathol Res
Goldfarb, S., Pugh, T. D., Koen, H., and He, Y. Z. (1983). Preneoplastic and Pract 177, 13–21.
neoplastic progression during hepatocarcinogenesis in mice injected Hanigan, M. H., Winkler, M. L., and Drinkwater, N. R. (1993). Induction of
with diethylnitrosamine in infancy. Environ Health Perspect 50, three histochemically distinct populations of hepatic foci in C57BL/6J
149–61. mice. Carcinogenesis 14, 1035–040.
Gonzalez-Quintela, A., Mella, C., Perez, L. F., Abdulkader, I., Caparrini, A. Harada, T., Maronpot, R. R., Enomoto, A., Tamano, S., and Ward, J. M.
M., and Lojo, S. (2000). Increased serum tissue polypeptide specific anti- (1996). Changes in the liver and gallbladder. In Pathobiology of the
gen (TPS) in alcoholics: a possible marker of alcoholic hepatitis. Alcohol Aging Mouse. Vol. 2 (U. Mohr, D. L. Dungworth, C. C. Capen, W. W.
Clin Exp Res 24, 1222–226. Carlton, J. P. Sundberg, and J. M. Ward, eds.), pp 207–41. ILSI Press,
Goodman, D. G., Maronpot, R. R., Newberne, P. M., Popp, J. A., and Squire, R. Washington, DC.
A. (1994). Proliferative and selected other lesions in the liver of rats. In Harada, T., Maronpot, R. R., Morris, R. W., and Boorman, G. A. (1989). Obser-
Guides for Toxicologic Pathology (STP/ARP/AFIP), pp 1–24. Society of vation on altered hepatocellular foci in National Toxicology Program 2-
Toxologic Pathology, Washington, DC. year carcinogenicity studies in rats. Toxicol Pathol 17, 690–708.
Goodman, Z. D., and Ishak, K. G. (2006). Hepatobiliary system and pancreas. Harada, T., Maronpot, R. R., Morris, R. W., Stitzel, K. A., and Boorman, G. A.
In Surgical Pathology and Cytopathology (S. G. Silverberg, ed.), Vol. 2, (1989). Morphological and sterological characterization of hepatic foci of
4th ed., pp. 1465–526. Elsevier, Oxford, UK. cellular alteration in control Fischer 344 rats. Toxicol Pathol 17, 579–93.
Gopinath, C., Prentice, D. E., and Lewis, D. J. (1987). The liver. In Atlas of Harada, T., Enomoto, A., Boorman, G. A., and Maronpot, R. R. (1999). Liver
Experimental Toxicological Pathology (G. A. Grasham, ed.), Vol. 13, and gallbladder, In Pathology of the Mouse (R. R. Maronpot, ed.), pp.
pp. 43–60. MTP Press Limited, Lancaster, England. 119–83. Cache River Press, Vienna, IL.
Goto, K., Ohashi, H., Takakura, A., and Itoh, T. (2000). Current status of Heli- Harbord, M., Novelli, M., Canas, B., Power, D., Davis, C., Godovac-
cobacter contamination of laboratory mice, rats, gerbils, and house musk Zimmermann, J., Roes, J., and Segal, A.W. (2002). Ym1 is a neutrophil
shrews in Japan. Curr Microbiol 41, 161–66. granule protein that crystallizes in p47phox-deficient mice. The J Biol
Graewin, S. J., Lee, K. H., Tran, K. Q., Goldblatt, M. I., Svatek, C. L., Nakeeb, Chem 277, 5468–475.
A., and Pitt, H. A. (2004). Leptin-resistant obese mice do not form biliary Harbrecht, B. G., Stadler, J., Demetris, A. J., Simmons, R. L., and Billiar, T. R.
crystals on a high cholesterol diet. J Surg Res 122, 145–49. (1994). Nitric oxide and prostaglandins interact to prevent hepatic dam-
Graham, M. J., and Lake, B. G. (2008). Induction of drug metabolism: Species age during murine endotoxemia. Am J Physiol 266, G1004–010.
differences and toxicological relevance. Toxicology 254, 184–91. Hardisty, J. F., Elwell, M. R., Ernst, H., Greaves, P., Kolenda-Roberts, H.,
Gray, H., Williams, P. L., and Bannister, L. H. (1995). Gray’s Anatomy: The Malarkey, D. E., Mann, P. C., and Tellier, P. A. (2007). Histopathology
Anatomical Basis of Medicine and Surgery (L. H. Bannister, M. M. of hemangiosarcomas in mice and hamsters and liposarcomas/fibrosarco-
Berry, P. Collins, M. Dyson, J. E. Dusch, and M. W. J. Ferguson, eds.), mas in rats associated with PPAR agonists. Toxicol Pathol 35, 928–41.
pp 1795–802. Churchill Livingstone, New York. Hardisty, J. F., and Eustis, S. L. (1990). Toxicologic pathology: A critical stage
Greaves, P. (2007). Liver and pancreas. In Histopathology of Preclinical Toxi- in study interpretation. In Progress in Predictive Toxicology (D. B. Clay-
city Studies. 3rd edition. pp 457–514. Elsevier, Oxford, UK. son, ed.). CRC Press, Boca Raton, Florida.
Greaves, P., and Barsoum, N. (1990). Tumours of soft tissues. In Pathology of Helyer, B. J., and Petrelli, M. (1978). Cytoplasmic inclusions in spontaneous
Tumours in Laboratory Animals. Vol. I. Tumours of the Rat, 2nd edition hepatomas of CBA/H-T6T6 mice. Histochemistry and electron micro-
(V. S. Turusov and U. Mohr, eds), pp 597–23. IARC Scientific Publica- scopy. Journal of the National Cancer Institute 60, 861–69.
tions, Lyon, France. Heider, K., and Eustis, S. L. (1994). Tumours of the soft tissues. In Pathology
Greaves, P., Edwards, R., Cohen, G. M., and MacFarlane, M. (2001). ‘‘Have of Tumours in Laboratory Animals. Vol. 2. Tumours of the Mouse, 2nd
you seen this?’’ Diffuse hepatic apoptosis. Toxicol Pathol 29, 398–400. edition (V. Turusov and U. Mohr, eds.), pp. 611–49. IARC Scientific
Greaves, P., and Faccini, J. M. (1984). Rat Histopathology. A Glossary for Use Publications, Lyon, France.
in Toxicity and Carcinogenicity Studies, pp 88–97. Elsevier, Oxford, UK. Homberger, F. R. (1996). Mouse hepatitis virus. Schweiz Arch Tierheilkd 138,
Greaves, P., and Faccini, J. M. (1992). Digestive system. In Rat Histopathol- 183–88.
ogy: A Glossary for Use in Toxicity and Carcinogenicity Studies, pp. Herman, J. R., Dethloff, L. A., McGuire, E. J., Parker, R. F., Walsh, K. M.,
105–48. Elsevier, Oxford, UK. Gough, A. W., Masuda, H., and de la Iglesia, F. A. (2002). Rodent carci-
Greaves, P., Irisarri, E., and Monro A. M. (1986). Hepatic foci of cellular and nogenicity with the thiazolidinedione antidiabetic agent troglitazone.
enzymatic alteration and nodules in rats treated with clofibrate or diethyl- Toxicol Sci 68, 226–36.
nitrosamine followed by phenobarbital: Their rate of onset and their Hruban, R. H., Argani, P., and Ali, S. Z. (2006). The pancreas and extrahepatic
reversibility. J Natl Cancer Inst 76, 475–84. biliary system. In Silverberg’s Principles and Practice of Surgical Pathol-
Gruttadauria, S., Mandala, L., Miraglia, R., Caruso, S., Minervini, M. I., ogy and Cytopathology (G. G. Silverberg, ed.), pp. 1576–593. Churcill
Biondo, D., Volpes, R., Vizzini, G., Marsh, J. W., Luca, A., Marcos, Livingstone, Oxford, UK.
A., and Gridelli, B. (2007). Successful treatment of small-for-size syn- Hruban, Z., Slesers, A., and Hopkins, E. (1972). Drug-induced and naturally
drome in adult-to-adult living-related liver transplantation: single center occurring myeloid bodies. Lab Invest 27, 62–70.
series. Clin Transplant 21, 761–66. Hsu, L., Diwan, B., Ward, J. M., and Noguchi, C. T. (2006). Pathology of ‘‘Ber-
Guzman, R. E., and Solter, P. F. (2002). Characterization of sublethal keley’’ sickle-cell mice includes gallstones and priapism. Blood 107,
microcystin-LR exposure in mice. Vet Pathol 39, 17–26. 3414–415.
Hailey, J. R., Haseman, J. K., Bucher, J. R., Radovsky, A. E., Malarkey, D. E., Huang, J., Ren, R. N., Chen, X. M., and Ye, L. Y. (2007). An experimental
Miller R. T., Nyska, A., and Maronpot, R. R. (1998). Impact of Helico- study on hepatotoxicity of topiramate in young rats. Zhongguo Dang Dai
bacter hepaticus infection in B6C3F1 mice from twelve National Toxi- Er Ke Za Zhi 9, 54–8.
cology Program two-year carcinogenesis studies. Toxicol Pathol 26, Hurlimann, J., and Gardiol, D. (1991). Immunohistochemistry in the differen-
602–11. tial diagnosis of liver carcinomas. Am J Surg Pathol 15, 280–88.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 77S

Hurst, E. W., and Paget, G. E. (1963). Protoporphyrin, cirrhosis and hepatoma Knasmuller, S., Parzefall, W., Helma, C., Kassie, F., Ecker, S., and Schulte-
in livers of mice given griseofulvin. Br J Dermatol 75, 105–12. Hermann, R. (1997). Toxic effects of griseofulvin: Disease models,
Husztik, E., Lazar, G., and Szabo, E. (1984). Immunologically induced peliosis mechanisms, and risk assessment. Crit Rev Toxicol 27, 495–537.
hepatis in rats. Br J Exp Pathol 65, 313–18. Koen, H., Pugh, T. D., and Goldfarb, S. (1983). Centrilobular distribution of
Ichikawa, H., Imano, M., Takeyama, Y., Shiozaki, H., and Ohyanagi, H. diethylnitrosamine-induced hepatocellular foci in the mouse. Lab Invest
(2009). Involvement of osteopontin as a core protein in cholesterol gall- 49, 78–81.
stone formation. J Hepatobiliary Pancreat Surg 16, 197–203. König, H. E., Sautet, J., and Liebich, H. G. (2004). 7. Digestive system (appa-
Institute of Laboratory Animal Resources, National Research Council, ratus digetorius). In Veterinary Anatomy of Domestic Mammals: Text-
National Academy of Sciences. (1980). Histologic typing of liver tumors book and Colour Atlas (H. E. König, H.-G. Liebich, and H. Bragulla,
of the rat. J Natl Cancer Inst 64, 177–206. eds.), pp. 332–37. Schattauer, Stuttgart.
Irisarri, E., and Hollander, C. F. (1994). Digestive system—Aging of the liver Kuriki, J., Murakami, H., Kakumu, S., Sakamoto, N., Yokochi, T., Nakashima,
In Pathobiology of the Aging Rat—Vol. 2. (U. Mohr, D. L. Dungworth, I., and Kato, N. (1983). Experimental autoimmune hepatitis in mice after
and C. C. Capen, eds.), pp. 341–49. ILSI Press, Washington, DC. immunization with syngeneic liver proteins together with the polysac-
Jang, J. J., Weghorst, C. M., Henneman, J. R., Devor, D. E., and Ward, J. M. charide of Klebsiella pneumoniae. Gastroenterology 84, 596–603.
(1992). Progressive atypia in spontaneous and N-nitrosodiethylamine- Laifenfeld, D., Gilchrist, A., Drubin, D., Jorge, M., Eddy, S. F., Frushour, B. P.,
induced hepatocellular adenomas of C3H/HeNCr mice. Carcinogenesis Ladd, B. Obert, L. A., Gosink, M. M., Cook, J. C., Criswell, K., Somps,
13, 1541–547. C. J., Koza-Taylor, P., Elliston, K. L., and Lawton, M. P. (2010). The role
Jones, G., and Butler, W. H. (1975). Morphology and spontaneous neoplasia. In of Hypoxia in 2-butoxyethanol–induced hemangiosarcoma. Toxicol Sci
Mouse Hepatic Neoplasia (W. M. Butler and P. M. Newberne, eds.), pp 113, 254–66.
21–59. Elsevier, Oxford, UK. Lailach, J. J., Johnson, W. D., Raczniak, T. J., and Shumaker, R. C. (1977).
Jones, G., and Butler, W. H. (1978). Light microscopy of rat hepatic neoplasia. Fibrin thrombosis in monocrotaline pyrrole-induced cor pulminale in
In Rat Hepatic Neoplasia (P. M. Newberne and W. H. Butler, eds.), pp rats. Arch Pathol Lab Med 101, 69–73.
114–35. MIT Press, Cambridge, Massachusetts. Lalwani, N. D., Dethloff, L. A., Haskins, J. R., Robertson, D. G., and de La,
Jones, M. L. (2002). Lipids In Theory and Practice of Histological Techniques Iglesia. (1997). Increased nuclear ploidy, not cell proliferation, is
(J. D. Bancroft and M. Gamble, eds.), pp. 201–30. Churchill Livingstone, sustained in the peroxisome proliferator-treated rat liver. Toxicol Pathol
Edinburgh, Scotland. 25, 165–76.
Kakizoe, S., Goldfarb, S., and Pugh, T. D. (1989). Focal impairment of growth Lee, K. P. (1983). Peliosis hepatis-like lesion in aging rats. Vet Pathol 20, 410–23.
in hepatocellular neoplasms of C57BL/6 mice: A possible explanation Lee, S. P., and Scott, A. J. (1982). The evolution of morphologic changes in the
for the strain’s resistance to hepatocarcinogenesis. Cancer Res 49, gallbladder before stone formation in mice fed a cholesterol-cholic acid
3985–989. diet. Am J Pathol 108, 1–8.
Kanel, G. C., and Korula, J. (2005). Atlas of Liver Pathology, 2nd edition. Levin, S. (1999). Commentary: Implementation of the STP recommendations
Elservier Saunders, Los Angeles, California. on the nomenclature of cell death. Society of Toxicologic Pathologists.
Karbe, E., and Kerlin, R. L. (2002). Cystic degeneration/Spongiosis hepatis in Toxicol Pathol 27, 491.
rats. Toxicol Pathol 30, 216–27. Levin, S., Bucci, T. J., Cohen, S. M., Fix, A. S., Hardisty, J. F., LeGrand, E. K.,
Kari, F., Bucher, J., Haseman, J., Eusti, S., and Huff, J. (1995). Long-term Maronpot, R. R., and Trump, B. F. (1999). The nomenclature of cell
exposure to the anti-inflammatory agent phenylbutazone induces kidney death: Recommendations of an ad hoc Committee of the Society of Tox-
tumors in rats and liver tumors in mice. Jpn J of Cancer Res 86, 252–63. icologic Pathologists. Toxicol Pathol 27, 484–90.
Kashiwagi, R., Kaidoh, T., and Inoué, T. (2001). Immunohistochemical study Levine, S., and Sowinski, R. (1985). Splenic infarcts produced in rats by vaso-
of derndritic cells and kupffer cells in griseofulvin-induced protoporhyric constrictor drugs. Exp Pathol 28, 13–9.
mice. Yonago Act Med 44, 7–16. Lewis, D. J. (1984). Spontaneous lesions of the mouse biliary tract. J Comp
Katoh, M., and Sugimoto, T. (1982). Effect of malotilate (diisopropyl 1, 3- Pathol 94, 263–71.
dithiol-2-ylidenemalonate) on chronic liver injury caused by carbon tet- Li, X., Elwell, M. R., Ryan, A. M., and Ochoa, R. (2003). Morphogenesis of
rachloride. Nippon Yakurigaku Zasshi 80, 83–91. post mortem hepatocyte vacuolation and liver weight increases in
Kiernan, F. (1883). The anatomy and physiology of the liver. Philosophical Trans- Sprague-Dawley rats. Toxicol Pathol 31, 682–88.
actions of the Royal Society of London Series B, Biol Sci 123, 711–70. Lin, C. C., Chang, C. H., Yang, J. J., Namba, T., and Hattori, M. (1996). Hepa-
Kim, J. C., and Kaminsky, L. S. (1988). 2,2,2-Trifluoroethanol toxicity in aged toprotective effects of emodin from Ventilago leiocarpa. J Ethnopharma-
rats. Toxicol Pathol 16, 35–45. col 52, 107–11.
Kim, J. C., Shin, D. H., Kim, S. H., Kim, J. K., Park, S. C., Son, W. C., Lee, H. S., Lipman, R. D., Gaillard, E. T., Harrison, D. E., and Bronson, R. T. (1993). Hus-
Suh, J. E., Kim, C. Y., Ha, C. S., and Chung, M. K. (2004). Subacute toxicity bandry factors and the prevalence of age-related amyloidosis in mice. Lab
evaluation of a new camptothecin anticancer agent CKD-602 administered Anim Sci 43, 439–44.
by intravenous injection to rats. Regul Toxicol Pharmacol 40, 356–69. Lipsky, M. M., Tanner, D. C., Hinton, D. E., Trump, B. F. (1984). Reversibility,
Kimbrough, R. D. (1973). Pancreatic-type tissue in livers of rats fed polychlori- persistence, and progression of safrole-induced mouse liver lesions follow-
nated biphenyls. J Natl Cancer Inst 51, 679–81. ing cessation of exposure. In Mouse Liver Neoplasia (J. A. Popp, ed.), pp
Kimbrough, R. D., and Linder, R. E. (1974). Induction of adenofibrosis and 161–77. Hemisphere Publishing Corporation, Washington, DC.
hepatomas of the liver in BALB-cJ mice by polychlorinated biphenyls Liu, Y., Cui, D., Hoshii, Y., Kawano, H., Une, Y., Gondo, T., and Ishihara, T.
(Aroclor 1254). J Natl Cancer Inst 53, 547–52. (2007). Induction of murine AA amyloidosis by various homogeneous
Kimbrough, R. D., Linder, R. E., Burse, V. W., and Jennings, R. W. (1973). amyloid fibrils and amyloid-like synthetic peptides. Scand J Immunol
Adenofibrosis in the rat liver, with persistence of polychlorinated biphe- 66, 495–500.
nyls in adipose tissue. Arch Environ Health 27, 390–95. Mahon, D. C. (1989). Altered hepatic foci in rat liver as weight of evidence of
Kimura, M., and Abe, M. (1994). Histology of postmortem changes in rat livers carcinogenicity: The Canadian perspective. Toxicol Pathol 17, 709–15.
to ascertain hour of death. Int J Tissue React 16, 139–50. Maita, K., Hirano, M., Harada, T., Mitsumori, K., Yoshida, A., Takahashi, K.,
Kitamura, K., Iwasaki, H. O., Yasoshima, A., Yoshikawa, H., Yoshikawa, T., Nakashima, N., Kitazawa, T., Enomoto, A., Inui, K., and Shirasu,Y.
and Okaniwa, A. (1992). Pathology of chemically induced chronic active (1988). Mortality, major cause of moribundity, and spontaneous tumors
hepatitis in mice. Exp Mol Pathol 57, 153–66. in CD-1 mice. Toxicol Pathol 16, 340–49.
Klaunig, J. E., and Kamendulis, L. M. (2005). Mode of action of Malarkey, D. E., Devereux, T. R., Dinse, G. E., Mann, P. C., and Maronpot,
butoxyethanol-induced mouse liver hemangiosarcomas and hepatocellu- R. R. (1995). Hepatocarcinogenicity of chlordane in B6C3F1 and
lar carcinomas. Toxicol Lett 156, 107–15. B6D2F1 male mice: Evidence for regression in B6C3F1 mice and
78S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

carcinogenesis independent of ras proto-oncogene activation. Carcino- Morgan, K. T., Frith, C. H., Swenberg, J. A., McGrath J. T., Zulch K. J., and
genesis 16, 2617–625. Crowder D. M. (1984). A morphologic classification of brain tumors
Malarkey, D., Johnson, K., Ryan, L., Boorman, G., and Maronpot, R. (2005). found in several strains of mice. J Natl Cancer Inst 72, 151–60.
New insights into functional aspects of liver morphology. Toxicol Pathol Muff, M. A., Masyuk, T. V., Stroope, A. J., Huang, B. Q., Splinter, P. L., Lee,
33, 27–34. S. O., and Larusso, N. F. (2006). Development and characterization of a
Malhotra, V., Sakhuja, P., and Gondal, R. (2004). Immunohistochemistry in cholangiocyte cell line from the PCK rat, an animal model of Autosomal
liver diseases. J Gastroenterol Hepatol 19, S364–69. Recessive Polycystic Kidney Disease. Lab Invest 86, 940–50.
Maronpot, R. R. (2009). Biological basis of differential susceptiblity to hepa- Naeim, F., Copper, P. H., and Semion, A. A. (1973). Peliosis hepatis. Possible
tocarcinogenesis amoung mouse strains. J Toxicol Pathol 22, 11–33. etiologic role of anabolic steroids. Arch Pathol 95, 284–85.
Maronpot, R. R., Harada, T., Murthy, A. S. K., and Boorman, G. A. (1989). Nagano, K., Sasaki, T., Umeda, Y., Nishizawa, T., Ikawa, N., Ohbayashi, H.,
Documenting foci of hepatocellular alteration in two-year carcinogeni- Arito, H., Yamamoto, S., and Fukushima, S. (2007). Inhalation carcino-
city studies: Current practices of the national toxicology program. Toxi- genicity and chronic toxicity of carbon tetrachloride in rats and mice.
col Pathol 17, 675–84. Inhal Toxicol 19, 1089–103.
Maronpot, R. R., Montgomery, C. A., Boorman G. A., and McConnell, E. E. Narama, I., Imaida, K., Iwata, H., Nakae, D., Nishikawa, A., and Harada, T.
(1986). National Toxicology Program nomenclature for hepatoprolifera- (2003) A review of nomenclature and diagnostic criteria for proliferative
tive lesions of rats. Toxicol Pathol 14, 263–73. lesions in the liver of rats by a working group of the Japanese Society of
Marsman, D. S. (1995). NTP Technical Report on Toxicity Studies of Dibutyl Toxicologic Pathology. J Toxicol Pathol 16, 1–17.
Phthalate (CAS No. 84-74-2) Administered in Feed to F344/N Rats and Newton, P. E., Wooding, W. L., Bolte, H. F., Derelanko, M. J., Hardisty, J. F., and
B6C3F? Mice. Toxicity Report Series, No. 30. U.S. Department of Health Rinehart, W. E. (2001). A chronic inhalation toxicity/oncogenicity study of
and Human Services, Public Health Service, National Institutes of methylethylketoxime in rats and mice. Inhal Toxicol 13, 1093–116.
Health, Research Triangle Park, North Carolina. Nishimura, J., Dewa, Y., Muguruma, M., Kuroiwa, Y., Yasuno, H., Shima, T.,
Marsman, D. S., and Popp, J. A. (1994). Biological potential of basophilic Jin, M., Takahashi, M., Umemura, T., and Mitsumori, K. (2007). Effect
hepatocellular foci and hepatocellular adenoma induced by the peroxi- of fenofibrate on oxidative DNA damage and on gene expression related
some proliferator, Wy-14,643. Carcinogenesis 15, 111–17. to cell proliferation and apoptosis in rats. Toxicol Sci 97, 44–54.
Martignoni, M., Groothuis, G. M., and de Kanter, R. (2006). Species differ- Nonomura, A., Kono, N., Yoshida, K., Mizukami, Y., Nakanuma, Y., and
ences between mouse, rat, dog, monkey and human CYP-mediated drug Matsubara, F. (1991). Histological changes of the liver in experimental
metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol graft-versus-host disease across minor histocompatibility barriers. V. A
2, 875–94. light and electron microscopic study of the intralobular changes. Liver
Martin, N. C., McGregor, A. H., Sansom, N., Gould, S., and Harrison, D. J. 11, 149–57.
(2001). Phenobarbitone-induced ploidy changes in liver occur indepen- Nonoyama, T., Fullerton, F., Reznik, G., Bucci, T. J., and Ward, J. M. (1988).
dently of p53. Toxicology Letters 119, 109–15. Mouse hepatoblastomas: A histologic, ultrastructural, and immunohisto-
Masson, R., and Roome, N. O. (1997). Spontaneous iron overload in Sprague- chemical study. Vet Pathol 25, 286–96.
Dawley rats. Toxicol Pathol 25, 308–16. Nonoyama, T., Reznik, G., Bucci, T. J., and Fullerton, F. (1986). Hepatoblas-
Masyuk, T. V., Huang, B. Q., Masyuk, A. I., Ritman, E. L., Torres, V. E., toma with squamous differentiation in a B6C3F1 mouse. Vet Pathol 23,
Wang, X., Harris, P. C., and La Russo, N. F. (2004). Biliary Dysgenesis 619–22.
in the PCK Rat, an Orthologous Model of Autosomal Recessive Polycys- NTP Satellite Symposium. (2010). Pathology Potpourri. Chicago Marriott
tic Kidney Disease. The Am J of Pathol 165, 1719–730. Downtown, Chicago, Illinois.
Masyuk, T., Masyuk, A., Torres, V., Harris, P., and Larusso, N. (2007). Octreo- NTP technical report on the toxicology and carcinogenesis studies of 2, 3, 7, 8-
tide inhibits hepatic cystogenesis in a rodent model of polycystic liver tetrachlorodibenzo-p-dioxin (TCDD) (CAS No. 1746-01-6) in female
disease by reducing cholangiocyte adenosine 3’,5’-cyclic monopho- Harlan Sprague-Dawley rats (Gavage Studies) (2006). Natl Toxicol Pro-
sphate. Gastroenterology 132, 1104–116. gram Tech Rep Ser 521, 4–232.
Matsumoto, M., Aiso, S., Umeda, Y., Arito, H., Nagano, K., Yamamoto, S., NTP technical report on the toxicology and carcinogenesis studies of a binary
and Matsushima, T. (2006). Thirteen-week oral toxicity of para- and mixture of 3,3’,4,4’,5-pentachlorobiphenyl (PCB 126) (CAS no. 57465-
ortho- chloronitrobenzene in rats and mice. J Toxicol Sci 31, 9–22. 28-8) and 2,3’,4,4’,5-pentachlorobiphenyl (PCB 118) (CAS no. 31508-
McCaughan, G. W., Bilous, M. J., and Gallagher, N. D. (1985). Long-term sur- 00-6) in female Harlan Sprague-Dawley rats (Gavage studies)(2006).
vival with tumor regression and androgen-induced liver tumors. Cancer NIH Publication, No. 07-4467, Natl Toxicol Program Tech Rep Ser 531.
56, 2622–626. NTP toxicology and carcinogenesis studies of ethylene glycol (CAS No. 107-
Meierhenry, E. F., Ruebner, B. H., Gershwin, M. E., Hsieh, L. S., and French, 21-1) in B6C3F1 mice (feed studies). (1993). NIH Publication, No. 93-
S. W. (1983). Dieldrin-induced mallory bodies in hepatic tumors of mice 3144. Natl Toxicol. Program Tech Rep Ser 413.
of different strains. Hepatology 3, 90–5. NTP toxicology and carcinogenesis studies of 3, 30 , 4, 40 ,
Mendenhall, C. L., and Chedid, A. (1980). Peliosis hepatis. Its relationship to 5-pentachlorobiphenyl (PCB 126) (CAS No. 57465-28-8) in female Har-
chronic alcoholism, aflatoxin B1, and carcinogenesis in male Holtzman lan Sprague-Dawley rats (Gavage Studies). (2006). Natl Toxicol Program
rats. Digestive Diseases and Sciences 25, 587–92. Tech Rep Ser 520, 4-246.
Mitsumori, K. (1990). Blood and lymphatic vessels. In Pathology of the Fischer NTP toxicology and carcinogenesis studies of theophylline (CAS No. 58-55-9)
Rat. Reference and Atlas (G. A. Boorman, S. L. Eustis, M. R. Elwell, C. in F344/N Rats and B6C3F1 mice (Feed and Gavage Studies) (1998).
A. Montgomery, Jr., and W. F. MacKenzie, eds.), pp. 473–84. Academic NIH Publication, No. 98-3963, Natl Toxicol Program Tech Rep Ser 473.
Press, San Diego, California. Numan, F., Cokyuksel, O., Camuscu, S., Demir, K., and Duren, M. (1986).
Miyai, K. (1991). The discovery of new diseases by utilizing laboratory Caroli syndrome. Rontgenblatter 39, 191–92.
data for clinical research. Perspectives in Biology and Medicine 34, Nyska, A., Moomaw, C. R., Foley, J. F., Maronpot, R. R., Malarkey, D. E.,
542–48. Cummings, C. A., Peddada, S., Moyer, C. F., Allen, D. G., Travlos, G.,
Miyazaki, M., Makowka, L., Falk, R. F., Falk, J. A., McDonell, M., and and Chan, P. C. (2002). The hepatic endothelial carcinogen riddelliine
Venturi, D. (1983). Protection of thermochemotherapeutic-induced induces endothelial apoptosis, mitosis, S phase, and p53 and hepatocytic
lethal acute hepatic necrosis in the rat by 16,16-dimethyl prostaglandin vascular endothelial growth factor expression after short-term exposure.
E2. J Surg Res 34, 415–26. Toxicol Appl Pharmacol 184, 153–64.
Moore, M. A., Thamavit, W., and Bannasch, P. (1996). Tumours of the liver. In Nyska, A., Waner, T., Wormser, U., Gur, E., Kuttin, E., and Dayan, D. (1992).
Pathology of Tumours in Laboratory Animals—Vol. 3 (V. Turusov and Possible pitfalls in rat extended dermal toxicity testing: an hepatic-ocular
U. Mohr, eds.), pp. 79–125. IARC, Lyon, France. syndrome. Arch Toxicol 66, 339–46.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 79S

Obert, L. A., Sobocinski, G. P., Bobrowski, W. F., Metz, A. L., Rolsma, M. D., Reddy, J. K., Lalwani, N. D., Reddy, M. K., and Qureshi, S. A. (1982). Exces-
Altrogge, D. M., and Dunstan, R. W. (2007). An immunohistochemical sive accumulation of autofluorescent lipofuscin in the liver during hepa-
approach to differentiate hepatic lipidosis from hepatic phospholipidosis tocarcinogenesis by methyl clofenapate and other hypolipidemic
in rats. Toxicol Pathol 35, 728–34. peroxisome proliferators. Cancer Res 42, 259–66.
Ohnishi, T., Arnold, L. L., Clark, N. M., Wisecarver, J. L., and Cohen, S. M. Reed, D. J. (1998). Evaluation of chemical-inducedoxidative stress as a mechan-
(2007). Comparison of endothelial cell proliferation in normal liver and ism of hepatocyte death In Toxicology of the Liver (G. L. Plaa and W. R.
adipose tissue in B6C3F1 mice, F344 rats, and humans. Toxicol Pathol Hewitt, eds.), pp. 187–220. Taylor & Francis, Washington, DC.
35, 904–09. Rege, R. V., and Prystowsky J. B. (1998). Inflammation and a thickened mucus
Omar, A., Elmesallamy, G.-S., and Eassa, S. (2005). Comparative study of the layer in mice with cholesterol gallstones. J Surg Res 74, 81–5.
hepatotoxic, genotoxic and carcinogenic effects of praziquantel distocide Rijhsinghani, K., Krakower, C., Swerdlow, M., Abrahams, C., and Ghose, T.
& the natural myrrh extract Mirazid on adult male albino rats. J Egypt Soc (1980). Alpha-1-antitrypsin in intracellular inclusions of diethylnitrosa-
Parasitol 35, 313–29. mine induced hepatomas of C57BLxC3H F1 mice. Carcinogenesis 1,
Parker, G. A., and Gibson, W. B. (1995). Liver lesions in rats associated with 473–79.
wrapping of the torso. Toxicol Pathol 23, 507–12. Robison, R. L., Van Ryzin, R. J., Stoll, R. E., Jensen, R. D., and Bagdon, R. E.
Peckham, J. C., and Heider, K. (1999). Skin and subcutis. In Pathology of the (1984). Chronic toxicity/carcinogenesis study of temazepam in mice and
Mouse. Reference and Atlas (R. R. Maronpot, G. A. Boorman, and B. W. rats. Fundam Appl Toxicol 4, 394–405.
Gaul, eds.), pp. 555–12. Cache River Press, Vienna. Rogers, A. B., and Fox, J. G. (2004). Inflammation and cancer. I. Rodent mod-
Peichoto, M. E., Teibler, P., Ruiz, R., Leiva, L., and Acosta, O. (2006). Sys- els of infectious gastrointestinal and liver cancer. Am J Physiol Gastroint-
temic pathological alterations caused by Philodryas patagoniensis colu- est Liver Physiol 286, G361–66.
brid snake venom in rats. Toxicon 48, 520–28. Ruebner, B. H., Watanabe, K., and Wand, J. S. (1970). Lytic necrosis resem-
Peraino, C., Staffeldt, E. F., Carnes, B. A., Ludeman, V. A., Blomquist, J. A., bling peliosis hepatis produced by lasiocarpine in the mouse liver. A light
and Vesselinovitch, S. D. (1984). Characterization of histochemically and electron microscopic study. Am J Pathol 60, 247–71.
detectable altered hepatocyte foci and their relationship to hepatic tumor- Ruehl-Fehlert, C., Kittel, B., Morawietz, G., Deslex, P., Keenan, C., Mahrt, C.
igenesis in rats treated once with diethylnitrosamine or benzo(a)pyrene R., Nolte, T., Robinson, M., Stuart, B. P., and Deschl, U. (2003). Revised
within one day after birth. Cancer Res 44, 3340–347. guides for organ sampling and trimming in rats and mice—Part 1. Exp
Percy, D. H., and Barthold, S. W. (2001). Hamster. In Pathology of Laboratory and Toxicol Pathol 55, 91–106.
Rodents and Rabbits, pp. 194–95. Blackwell Publishing, Ames, Iowa. Sakamoto, M., Takayama, S., and Hosoda, Y. (1989). Hemangioendothelial
Percy, D. H., and Barthold, S. W. (2007). Pathology of Laboratory Rodents and sarcoma in brown adipose tissue of mouse induced by carcinogenic het-
Rabbits, 3rd edition, p. 356. Blackwell Publishing, Ames, Iowa. erocyclic amine, Glu-P-1. Toxicol Pathol 17, 754–58.
Peters, M., Liebman, H. A., Tong, M. J., and Tinberg, H. M. (1983). Alcoholic Sasse, D., and Maly, I. P. (1991). Studies on the periportal hepatotoxicity of
hepatitis: Granulocyte chemotactic factor from Mallory body-stimulated allyl alcohol. Prog Histochem Cytochem 23, 146–49.
human peripheral blood mononuclear cells. Clin Immunol Immunopathol Sato, Y., Harada, K., Furubo, S., Kizawa, K., Sanzen, T., Yasoshima, M.,
28, 418–30. Ozaki, S., Isse, K., Sasaki, M., and Nakanuma, Y. (2006). Inhibition of
Pitt, C. G., Gupta, G., Estes, W. E., Rosenkrantz, H., Metterville, J. J., Crum- intrahepatic bile duct dilation of the polycystic kidney rat with a novel
bliss, A. L., Palmer, R. A., and Sciortino, C. V. (1979). The selection and tyrosine kinase inhibitor gefitinib. Am J Pathol 169, 1238–250.
evaluation of new chelating agents for the treatment of iron overload. Sato, Y., Harada, K., Kizawa, K., Sanzen, T., Furubo, S., Yasoshima, M.,
J Pharmacol Exp Therap 208, 12–8. Ozaki, S., Ishibashi, M., and Nakanuma, Y. (2005). Activation of the
Popp, J. A. (1984). Mouse Liver Neoplasia. Current Perspectives. Hemisphere MEK5/ERK5 cascade is responsible for biliary dysgenesis in a rat model
Publishing Co., New York. of Caroli’s disease. Am J Pathol 166, 49–60.
Popp, J. A. (1985). Hepatocellular carcinoma, liver, rat. In Monographs on Satoh, M., Kobayashi, K., Ishii, M., Igarashi, T., and Toyota, T. (1996). Mid-
Pathology of Laboratory Animals. Digestive System (T. C. Jones, U. zonal necrosis of the liver after concanavalin A-injection. The Tohoku J
Mohr, and R. D. Hunt, eds.), pp. 39–46. Springer, New York. Exp Med 180, 139–52.
Popp, J. A., and Cattley, R. C. (1991). Hepatobiliary system In Handbook of Schoental, R., and Magee, P. N. (1959). Further observations on the subacute
Toxicologic Pathology (W. M. Haschek and C. G. Rousseaux, eds.), and chronic liver changes in rats after a single dose of various pyrrolizi-
pp. 279–314. Academic Press, San Diego, California. dine (Senecio) alkaloids. J Pathol Bacteriol 78, 471–82.
Popper, H., Maltoni, C., and Selikoff, J. (1981). Vinyl chloride-induced hepatic Schulte-Hermann, R., Kraupp-Grasl, B., Bursch, W., Gerbracht, U., and
lesions in man and rodents. A comparison. Liver 1, 7–20. Timmermann-Trosiener, I. (1989). Effects of non-genotoxic hepatocarci-
Pozharisski, K. M., and Turusov, V. S. (1991). Angiosarcoma of the renal cap- nogens phenobarbital and nafenopin on phenotype and growth of differ-
sule, mouse. In Monographs on Pathology of Laboratory Animals. Cardi- ent populations of altered foci in rat liver. Toxicol Pathol 17, 642–50.
ovascular and Musculoskeletal Systems (T. C. Jones, U. Mohr, and R. D. Seawright, A. A. (1972). A metabolic basis for midzonal necrosis of the liver.
Hunt, eds.), pp. 91–7. Springer, New York. J Pathol 107, Pxv.
Rabstein, L. S., Peters, R. L., and Spahn, G. J. (1973). Spontaneous tumors and Seehafer, S. S., and Pearce, D. A. (2006). You say lipofuscin, we say ceroid:
pathologic lesions in SWR-J mice. J Natl Cancer Inst 50, 751–58. Defining autofluorescent storage material. Neurobiology of Aging 27,
Rajtová, V., Horák, J., and Popesko, P. (2002). A Colour Atlas of the Anatomy 576–88.
of Small Laboratory Animals. Saunders, London. Serra, C. A., Recalde, E. B., and Martellotto, G. I. (1987). Caroli’s disease:
Ramot, Y., Lewis, D., Ortel, T., Streicker, M., Moser, G., Elmore, S., Ward, S., Congenital cystic and segmentary dilatation of the intrahepatic bile ducts.
Peddada S., and Nyska, A. (2007). Age and dose related sensitivity in 2- Rev Fac Cien Med Univ Nac Cordoba 45, 28–30.
butoxyethanol f344 rat model of hemolytic anemia and disseminated Shea, S. M. (1958). The method of paired comparisons in histopathological
thrombosis. Exp Toxicol Pathol 58, 311–22. ranking; hyaline degeneration of liver cells. A M A Arch Pathol 65,
Rank, J. M., Straka, J. G., and Bloomer, J. R. (1990). Liver in disorders of por- 77–80.
phyrin metabolism. J Gastroenterol Hepatol 5, 573–85. Shio, H., Fowler, S., Bhuvaneswaran, C., and Morris, M. D. (1982). Lysosome
Rappaport, A. M., Borowy, Z. J., Lougheed, W. M., and Lotto, W. N. (1954). lipid storage disorder in NCTR-BALB/c mice. II. Morphologic and cyto-
Subdivision of hexagonal liver lobules into a structural and functional chemical studies. The Am J of Pathol 108, 150–59.
unit; role in hepatic physiology and pathology. Anat Rec 119, 11–33. Sigal, S. H., Rajvanshi, P., Gorla, G. R., Sokhi, R. P., Saxena, R., Gebhard, D.
Reasor, M. J., Hastings, K. L., and Ulrich, R. G. (2006). Drug-induced phos- R. Jr., Reid, L. M., and Gupta, S. (1999). Partial hepatectomy-induced
pholipidosis: Issues and future directions. Expert Opin Drug Saf 5, polyploidy attenuates hepatocyte replication and activates cell aging
567–83. events. The Am J of Physiol 276, 1260–272.
80S THOOLEN ET AL. TOXICOLOGIC PATHOLOGY

Singh, N. D., Sharma, A. K., Dwivedi, P., Patil, R. D., and Kumar, M. (2007). Trotman, B. W., Bernstein, S. E., Balistreri, W. F., and Martin, R. A. (1983).
Citrinin and endosulfan induced maternal toxicity in pregnant Wistar Interaction of hemolytic anemia and genotype on hemolysis-induced
rats: Pathomorphological study. J Appl Toxicol 27, 589–601. gallstone formation in mice. Gastroenterology 84,719–24.
Sirica, A. E. (1992). The Role of Cell Types in Hepatocarcinogenesis. CRC Tschudy, D. P. (1962). Biochemical studies of experimental porphyria. Meta-
Press, Boca Raton, Florida. bolism 11, 1287–301.
Slehria, S., Rajvanshi, P., Ito, Y., Sokhi, R. P., Bhargava, K. K., Palestro, C. J., Tsokos, M., and Erbersdobler, A. (2005). Pathology of peliosis. Forensic Sci
McCuskey, R. S., and Gupta, S. (2002). Hepatic sinusoidal vasodilators Int 149, 25–33.
improve transplanted cell engraftment and ameliorate microcirculatory Turusov, V. S., Day, N. E., Tomatis, L., Gati, E., and Charles, R. T. (1973).
perturbations in the liver. Hepatology 35, 1320–328. Tumors in CF-1 mice exposed for six consecutive generations to DDT.
Solleveld, H. A., Miller, R. A., Banas, D. A., and Boorman, G. A. (1988). Pri- J Natl Cancer Inst 51, 983–97.
mary cardiac hemangiosarcomas induced by 1,3-butadiene in B6C3F1 Turusov, V. S., Deringer M. K., Dunn T. B., and Stewart H. L. (1973). Malig-
hybrid mice. Toxicol Pathol 16, 46–52. nant mouse-liver tumors resembling human hepatoblastomas. J Natl Can-
Spencer, A. J., Wilson, S. A., Batchelor, J., Reid, A., Rees, J., and Harpur, E. cer Inst 51, 1689–695.
(1997). Gadolinium chloride toxicity inthe rat. Toxicol Pathol 25, 245–55. Ungar, H. (1986). Venoocclusive disease of the liver and phlebectatic peliosis
Squire, R. A. (1989). Evaluation and grading of rat liver foci in carcinogenicity in the golden hamster exposed to dimethylnitrosamine. Pathol Res Pract
tests. Toxicol Pathol 17, 685–89. 181, 180–87.
Squire, R. A., and Levitt, M. H. (1975). Report of a workshop on classifi- Ungar, H., Sullman, S. F., and Zuckerman A. J. (1976). Acute and
cation of specific hepatocellular lesions in rats. Cancer Res 35, protracted changes in the liver of Syrian hamsters induced by a
3214–223. single dose of aflatoxin B1. Observations on pathological effects
Srinivasan, S., Hanes, M. A., Dickens, T., Porteous, M. E., Oh, S. P., Hale, L. of the solvent (dimethylformamide). Brit J of Exp Pathol 57,
P., and Marchuk, D. A. (2003). A mouse model for hereditary hemorrha- 157–64.
gic telangiectasia (HHT) type 2. Hum Mol Genet 12, 473–82. van Zwieten, M. J., and Hollander, C. F. (1997). Intranuclear and intracytoplas-
Steinbrecher, U. P., Lisbona, R., Huang, S. N., and Mishkin, S. (1981). Com- mic inclusions, liver, rat. In Monographs on Pathology of Laboratory
plete regression of hepatocellular adenoma after withdrawal of oral con- Animals: Digestive System (T. C. Jones, J. A. Popp, and U. Mohr eds.),
traceptives. Dig Dis Sci 26, 1045–050. pp. 133–39. Springer, New York.
Stejskal, R., Itabashi, M., Stanek, J., and Hruban, Z. (1975). Experimental por- Vesselinovitch, S. D., Mihailovich, N., and Rao, K. V. N. (1978). Morphology
phyria induced by 3-(2,4,6-trimethylphenyl)-thioethyl)-4 methylsyd- and metastatic nature of induced hepatic nodular lesions in C57BL x C3H
none. Virchows Arch B Cell Pathol 18, 83–100. F1 mice. Cancer Res 38, 2003–010.
Stewart, H. L. (1979). Tumours of the soft tissues. In Pathology of Tumours in Vollmar, B., Siegmund, S., Richter, S., and Menger, M. D. (1999). Microvas-
Laboratory Animals. Vol. II. Tumours of the Mouse (V. S. Turusov, ed.), cular consequences of Kupffer cell modulation in rat liver fibrogenesis.
pp. 487–526. IARC Scientific Publications, Lyon, France. J Pathol 189, 85–91.
Stewart, H. L., Williams, G., Keysser, C. H., Lombard, L. S., and Montali, R. J. Vons, C., Beaudoin, S., Helmy, N., Dagher, I., Weber, A., and Franco, D.
(1980). Histologic typing of liver tumors of the rat. J Natl Cancer Inst 64, (2009). First description of the surgical anatomy of the cynomolgus mon-
179–206. key liver. Am J of Primatol 71, 400–08.
Stinson, S.F., Hoover, K. L., and Ward, J. M. (1981). Quantitation of differ- Vowles, G. H., and Francis, R. J. (2002). Amyloid. In Theory and Practice of
ences between spontaneous and induced liver tumors in mice with an Histological Techniques (J. D. Bancroft and M. Gamble, eds.), pp. 303–
automated image analyzer. Cancer Lett 14, 143–50. 24. Churchill Livingstone, Edinburgh, Scotland.
Stout, M. D., Kissling, G. E., Suárez, F. A., Malarkey, D. E., Herbert, R. A., and Walker, R. M., Racz, W. J., and McElligott, T. F. (1985). Acetaminophen-
Bucher, J. R. (2008). Influence of Helicobacter hepaticus infection on the induced hepatotoxic congestion in mice. Hepatology 5, 233–40.
chronic toxicity and carcinogenicity of triethanolamine in B6C3F1 mice. Wang, J., Zhou, G., Chen, C., Yu, H., Wang, T., Ma, Y., Jia, G., Gao, Y., Li, B.,
Toxicol Pathol 36, 783–94. Sun, J., Li, Y., Jiao, F., Zhao, Y., and Chai, Z. (2007). Acute toxicity and
Tasaki, M., Umemura, T., Inoue, T., Okamura, T., Kuroiwa, Y., Ishii, Y., biodistribution of different sized titanium dioxide particles in mice after
Maeda, M., Hirose, M., and Nishikawa, A. (2008). Induction of char- oral administration. Toxicol Lett 168, 176–85.
acteristic hepatocyte proliferative lesion with dietary exposure of Ward, J. M. (1981). Morphology of foci of altered hepatocytes and naturally
Wistar Hannover rats to tocotrienol for 1 year. Toxicology 250, occurring hepatocellular tumors in F344 rats. Virchows Arch A 390, 339–45.
143–50. Ward, J. M. (1997). Cirrhosis, mouse. In Monographs on Pathology of Labora-
Tepperman, J., Caldwell, F. T., and Tepperman, H. M. (1964). Induction of tory Animals: Digestive System (T. C. Jones, J. A. Popp, and U. Mohr,
gallstones in mice by feeding a cholesterol-cholic acid containing diet. eds.), pp. 151–54. 2nd Edition. Springer, New York.
Am J Physiol 206, 628–34. Ward, J. M., Anver, M. R., Haines, D. C., and Benveniste, R. E. (1994).
Teredesai, A., Wohrmann, T., and Schlage, W. (2002). Hepatocholangiocellu- Chronic active hepatitis in mice caused by Helicobacter hepaticus. Am
lar carcinoma in a rat—Case report. J Vet Med A Physiol Pathol Clin Med J Pathol 145, 959–68.
49, 541–44. Ward, J. M., Collins, M. J., Jr., and Parker, J. C. (1977). Naturally occuring
Teutsch, H. F. (2005). The modular microarchitecture of human liver. Hepatol- mouse hepatitis virus infection in nude mouse. Lab Anim Sci 27, 372–76.
ogy 42, 317–25. Ward, J. M., Diwan, B. A., Ohshima, M., Hu, H., Schuller, H. M., and Rice, J.
Teutsch, H. F., Schuerfeld, D., and Groezinger, E. (1999). Three-dimensional M. (1986). Tumor-initiating and promoting activities of di(2-ethylhexyl)
reconstruction of parenchymal units in the liver of the rat. Hepatology 29, phthalate in vivo and in vitro. Environ Health Perspect 65, 279–91.
494–505. Ward, J. M., Fox, J. G., Anver, M. R., Haines, D. C., George, C. V., Collins, M.
Tian, X. F., Fan, X. G., Fu, C. Y., Huang, Y., and Zhu, C. (2005). Experimental J., Jr., Gorelick, P. L., Nagashima, K., Gonda, M. A., Gilden, R. V. et al.
study on the pathological effect of Helicobacter pylori on liver tissues. (1994b). Chronic active hepatitis and associated liver tumors in mice
Zhonghua Gan Zang Bing Za Zhi 13, 780–83. caused by a persistent bacterial infection with a novel Helicobacter spe-
Tillmann, T., Kamino, K., Dasenbrock, C., Ernst, H., Kohler, M., Morawietz, cies. J Natl Cancer Inst 86, 1222–227.
G., Campo, E., Cardesa, A., Tomatis, L., and Mohr, U. (1997). Subcuta- Ward, J. M., Goodman, D. G., Squire, R. A., Chu, K. C., and Linhart, M. S.
neous soft tissue tumours at the site of implanted microchips in mice. Exp (1979). Neoplastic and nonneoplastic lesions in aging (C57BL/6N x
Toxic Pathol 49, 197–200. C3H/HeN)F1 (B6C3F1) mice. J Natl Cancer Inst 63, 849–54.
Travlos, G. S., Mahler, J., Ragan, H. A., Chou, B. J., and Bucher, J. R. (1996). Ward, J. M., Lynch, P., and Riggs, C. (1988). Rapid development of hepatocel-
Thirteen-week inhalation toxicity of 2- and 4-chloronitrobenzene in lular neoplasms in aging male C3H/HeNCr mice given phenobarbital.
F344/N rats and B6C3F1 mice. Fund and Appl Toxicol 30, 75–92. Cancer Lett 39, 9–18.
Vol. 38, No. 7S, 2010 LESIONS OF THE HEPATOBILIARY SYSTEM 81S

Ward, J. M., Rice, J. M., Creasia, D., Lynch, P., and Riggs, C. (1983). Dissim- Xu, C., Li, C. Y., and Kong, A. N. (2005). Induction of phase I, II and III drug
ilar patterns of promotion by di(2-ethylhexyl)phthalate and phenobarbital metabolism/transport by xenobiotics. Arch Pharm Res 28, 249–68.
of hepatocellular neoplasia initiated by diethylnitrosamine in B6C3F1 Xu, S. X., Zheng, D., Sun, Y. M., Wang, S. H., Shao, L. Q., Huang, Q. Y., and
mice. Carcinogenesis 4, 1021–029. Xiang, D. L. (1992). Subchronic toxicity studies of fenbendazole in rats.
Ward, J. M., Wobus, C. E., Thackray, L. B., Erexson, C. R., Faucette, L. J., Vet Hum Toxicol 34, 411–13.
Belliot, G., Copeland, M. K., Mandel, M., Barron, E. L., Sosnovtsev S. Yamate, J., Tajima, M., Ihara, M., Shibuya, K., and Kudow, S. (1988). Spon-
V., and Green K. Y.(2006). Pathology of immunodeficient mice taneous vascular endothelial cell tumors in aged B6C3F1 mice. Jpn J Vet
with naturally-occurring murine norovirus infection. Toxicol Path 34, Sci 50, 453–61.
708–15. Yang, Y. H., and Campbell, J. S. (1964). Crystalline excrements in bronchitis
Ward, J. M., Yoon, M., Anver, M. R., Haines, D. C., Kudo, G., Gonzalez, F. J., and cholescystitis of mice. Am J Pathol 45, 337–45.
and Kimura, S. (2001). Hyalinosis and Ym1/Ym2 gene expression in the Yasui, M., Yase, Y., and Ota, K. (1991). Distribution of calcium in central ner-
stomach and respiratory tract of 129S4/SvJae and wild-type and vous system tissues and bones of rats maintained on calcium-deficient
CYP1A2-null B6, 129 mice. Am J Pathol 158, 323–32. diets. J Neurol Sci 105, 206–10.
Wilson, D. M., Goldsworthy, T. L., Popp, J. A., and Butterworth, B. E. Yoshitomi, K., Alison, R. H., and Boorman, G. A. (1986). Adenoma and ade-
(1992). Evaluation of genotoxicity, pathological lesions, and cell nocarcinoma of the gallbladder in aged laboratory mice. Vet Pathol 23,
proliferation in livers of rats and mice treated with furan. Environ Mol 523–27.
Mutagen 19, 209–22. Yoshitomi, K., and Boorman, G. A. (1994). Tumours of the gallbladder. In
Wilson, D., Lame, M., Dunston, S., and Segall, H. (2000). DNA damage cell Pathology of Tumours in Laboratory Animals. Vol. 2. Tumours of the
checkpoint activities are altered in monocrotaline pyrrole-induced cell Mouse (V. Turusov and U. Mohr, eds.), pp. 271–79, 2nd edition. IARC
cycle arrest in human pulmonary artery endothelial cells. Toxicol Appl Scientific Publications, Lyon, France.
Pharmacol 166, 69–80. Yu, W., Wan, X., Wright, J. R., Jr., Coddington, D., and Bitter-Suermann, H.
Wolstenholme, J. T., and Gardner, W. U. (1950). Sinusoidal dilatation occur- (1994). Heterotopic liver transplantation in rats: Effect of intrahepatic
ring in livers of mice with a transplanted testicular tumor. Proc Soc Exp islet isografts and split portal blood flow on liver integrity after auxiliary
Biol Med 74, 659–61. liver isotransplantation. Surgery 115, 108–17.
Wong, A. K., Alfert, M., Castrillon, D. H., Shen, Q., Holash, J., Yancopoulos, Zatloukal, K., Stumptner, C., Lehner, M., Denk, H., Baribault, H., Eshkind, L.
G. D., and Chin, L. (2001). Excessive tumor-elaborated VEGF and its G., and Franke, W. W. (2000). Cytokeratin 8 protects from hepatotoxi-
neutralization define a lethal paraneoplastic syndrome. Proc Natl Acad city, and its ratio to cytokeratin 18 determines the ability of hepatocytes
Sci U.S.A 98, 7481–486. to form Mallory bodies. Am J Pathol. 156, 1263–274.
World Health Organization. (1978). Principles and Methods for Evaluating the Zenner, L. (1999). Pathology, diagnosis and epidemiology of the rodent Heli-
Toxicity of Chemicals. Part I. World Health Organization, Geneva. cobacter infection. Comp Immunol Microbiol Infect Dis 22, 41–61.
Wright, P. F., and Stacey, N. H. (1991). A species/strain comparison of hepatic Zimmerman, H. J. (1998). Drug-induced hepatic disease. In Toxicology of the
natural lymphocytotoxic activities in rats and mice. Carcinogenesis 12, Liver (G. L. Plaa, and W. R. Hewitt, eds.), pp. 3–60. Taylor & Francis,
1365–370. Washington, DC.
Xie, Y., Newberry, E. P., Kennedy, S. M., Luo, J., and Davidson, N. O. (2009). Zwicker, G. M., Eyster, R. C., Sells, D. M., and Gass, J. H. (1995). Spontaneous
Increased susceptibility to diet-induced gallstones in liver fatty acid bind- vascular neoplasms in aged Sprague-Dawley rats. Toxicol Pathol 23,
ing protein (L-Fabp) knockout mice. J Lipid Res 50, 977–87. 518–26.

For reprints and permissions queries, please visit SAGE’s Web site at http://www.sagepub.com/journalsPermissions.nav.

Você também pode gostar