Sei sulla pagina 1di 9

l ab orator y

Special Article
a n imals
l i m i t e d

Laboratory Animals
2015, Vol. 49(S1) 21–29
! The Author(s) 2015
Standard Operating Procedures in Reprints and permissions:
sagepub.co.uk/
Experimental Liver Research: journalsPermissions.nav
DOI: 10.1177/0023677215573040
Thioacetamide model in mice and rats la.sagepub.com

MC Wallace1, K Hamesch2, M Lunova2, Y Kim5, R Weiskirchen3,


P Strnad2,4 and SL Friedman1

Abstract
In addition to carbon tetrachloride (CCl4), thioacetamide (TAA) represents a second widely used model for the
induction of experimental liver fibrosis, but can also be employed for the development of acute liver failure
and liver tumours. While TAA itself is not hepatotoxic, its reactive metabolites covalently bind to proteins and
lipids thereby causing oxidative stress and centrilobular necrosis. Compared with CCl4, TAA leads to more
periportal infiltrates and more pronounced ductal proliferation. While TAA has been shown to induce liver
fibrosis development in several different mouse strains, wide variations in the administration routes, doses
and treatment durations have been reported. Therefore, an adoption of a universal standard operating pro-
cedure for the administration of TAA is urgently needed. For that purpose, we are presenting here two TAA
models (intraperitoneal administration of 150 mg/kg of TAA three times per week for 11 weeks in rats, and
TAA administration in drinking water at 300 mg/L for 2–4 months in mice) with which we have had success in
reliably and reproducibly developing chronic liver injury and fibrosis.

Keywords
hepatotoxin, CYP2E1, liver fibrosis, cirrhosis, animal models

Historical background and current usage


been shown to produce cirrhosis more reliably than
The organosulfur compound thioacetamide (TAA; subcutaneous carbon tetrachloride (CCl4); however,
molecular formula C2H5NS), that was formerly used this route of administration of CCl4 is uncommon
as a fungicide and was occasionally found as a contam- and thus the results must be interpreted with caution.8
inant in orange juice,1 was first identified as being hep- A variable, weight-based oral regimen for TAA
atotoxic to rats in 1948. After two years of chronic
exposure in drinking water, experimental animals 1
Division of Liver Diseases, Icahn School of Medicine at Mount
developed advanced fibrosis, cirrhosis and liver Sinai, New York, New York, USA
tumours in a dose-dependent fashion.2 It was not 2
Department of Internal Medicine III, RWTH University Hospital
until almost 40 years later that chronic TAA intoxica- Aachen, Aachen, Germany
3
tion was established as a reliable and reproducible Institute of Molecular Pathobiochemistry, Experimental Gene
Therapy and Clinical Chemistry, RWTH University Hospital
experimental model of fibrosis and cirrhosis in rodents
Aachen, Aachen, Germany
by either the oral or intraperitoneal (IP) routes.3–5 In 4
Interdisciplinary Center for Clinical Research (IZKF), RWTH
addition, acute exposure to TAA at higher doses is rec- University Aachen, Aachen, Germany
5
ommended by the International Society for Hepatic Kyung Hee Medical Center, Seoul, Korea
Encephalopathy and Nitrogen Metabolism (ISHEN)
as a well-characterized model of acute liver failure, Corresponding author:
Scott L Friedman, MD, Division of Liver Diseases, Box 1123, Mount
encephalopathy and other events (Table 1) but these Sinai School of Medicine, 1425 Madison Ave, Room 11-70C,
will not be discussed in detail here.6,7 In a cirrhosis New York, NY 10029, USA.
imaging study, chronic oral TAA exposure in rats has Email: scott.friedman@mssm.edu
22 Laboratory Animals 49(S1)

Table 1. Usage of thioacetamide-induced liver damage in rodents.

Disease model Application Advantages Disadvantages References

Liver fibrosis IP commonly used Most closely resembles Long latency of fibrogenic 18, 30, 33, 35, 36
IP: 100–200 mg/kg alcoholic fibrogenesis. effects.
body weight, Reproducible results, Heterogenous results due
PO: 200 mg/L drink- low mortality, to missing
ing water less toxic than other standardization
chemicals
Acute hepatic IP, single dose 200 to Produces hepatic enceph- Monitoring and supportive 7, 37, 38
failure 1200 mg/kg body alopathy, care due to hypother-
weight large time windows mia, hypglycaemia and
before death others
Hepatocellular car- IP commonly used, Reproducible results Better models available 39, 40
cinoma or cholan- IP: 100–200 mg/kg (e.g. diethyl-
gio-carcinoma body weight, nitrosamine)
PO: 200 mg/L drink
ing water
Iron overload IP: 100 mg/kg body First non-transgenic 41
weight model of chronic liver-
injury-related iron
overload
Overview of frequently used rodent disease models employing thioacetamide application. IP ¼ intraperitoneal; PO ¼ per oral (drinking water).

administration in rats has been developed9 and remainder (6) used TAA in conjunction with other
refined10 and has been proposed to produce cirrhosis models such as CCl4 or bile duct ligation, for example.
and portal hypertension more reliably and with less Unfortunately, neither the proportion of experimental
toxicity than a non-variable dosing regimen. Less com- animals which developed advanced fibrosis or cirrhosis,
monly, TAA may be used in conjunction with another nor the mortality rates were reported in these studies. It
hepatocarcinogen such as N-diethylnitrosamine (DEN) is therefore difficult to make recommendations regard-
in a two-stage model to induce hepatocellular carcin- ing the most appropriate regimen based on recently
oma (HCC).11 Another variation is the combination of published studies. Local expertise and personal experi-
TAA with alcohol to produce stronger fibrosis.12 ence with the model is likely to be an important factor
The use of chronic TAA varies widely, particularly in when planning an experiment with TAA. Here we pre-
the route of administration; dosage, timing and length of sent our experience with the TAA model, which
treatment; and animal used (Table 2). In the first half of includes chronic oral exposure in C57BL/6 mice at a
2014, there have been 18 papers published in peer- fixed dose (300 mg/L in drinking water) for 2–4
reviewed journals that utilized chronic TAA to produce months,13 IP injection of TAA in Sprague–Dawley
fibrosis or cirrhosis experimentally (references available rats for 11 weeks with or without an experimental
upon request). Thirteen studies used rats of various gen- drug,14 and a time-course pilot study of TAA by IP
etic backgrounds (Sprague–Dawley, Wistar, F344, and injection in Sprague–Dawley rats for 12 weeks
albino) and five used mice of either C57BL/6 or (Figure 1). We report the rate and degree of fibrosis
Kunming backgrounds. The majority delivered TAA and cirrhosis development, changes in portal pressures
by the IP route (12); the remainder (5) used the oral and liver collagen content, as well as the mortality rate
route, three of which used a fixed dose (300 mg/L in in the experimental animals.
drinking water), the others (2) a variable, weight-based
dose. One paper did not report the route of administra-
tion. The oral and IP routes were used in both mice and
Mechanisms and pattern of liver injury
rats. Dosages for the IP route ranged from 10 mg/kg to Despite the hepatotoxic properties of TAA being well
400 mg/kg, two to three times per week from between 5 known since 1948, the mechanisms by which it causes
and 20 weeks while recently most researches have used liver injury are complex and remain poorly understood.
dosages of between 100 mg/kg and 200 mg/kg, three TAA requires oxidative bioactivation of its toxic
times per week for at least six weeks. Most used TAA metabolites to cause liver injury that affects both hep-
as the only model of fibrosis and cirrhosis (12); the atocytes and cholangiocytes.15,16 With regard to the
Wallace et al. 23

Table 2. Representative examples of thioacetamide usage in mouse models of experimental liver fibrosis.

Application Mice References

IP: 100 mg/kg body weight C57BL/6 J and FVB/N background, 18, 41
3/wk, 8 wk old, mainly female
6 wk course
IP: 150 mg/kg body weight C57BL/6 J background, 36, 42
3/wk, 6–8 wk old, male
6 wk course
IP: 200 mg/kg body weight C57BL/6 background, 35
3/wk, 6 wk old, male
4–8 wk course
IP: 200 mg/kg body weight BALB/c background, 32
3/wk, 8–10 wk old, male
4 wk course
PO: 200 mg/L drinking water C57BL/6 J background, 33
18 wk course 6 wk old, unknown gender
PO: 200 mg/L drinking water C3H/He background, 30
6–16 wk course 10 wk old, unknown gender
The main application routes for thioacetamide treatment are intraperitoneal injection or administration in drinking water.
IP ¼ intraperitoneal; PO ¼ per oral (drinking water), wk ¼ week.

latter, an enlargement of portal tracts without signs of increased length of intoxication, rodent liver microscop-
cholestasis is frequently observed.17–19 TAA is first oxi- ically demonstrates more clearly a mixed inflammatory
dized to TAA-S-oxide and then to TAA-S,S-oxide infiltrate of the portal tract with bile duct proliferation
(Figure 2); this process can be carried out by hepatic and some hepatocyte necrosis, than a typical pattern of
cytochrome P450 enzymes or FAD-containing mono- fibrosis with portal–portal and portal–central septa pro-
oxygenases.20 The lethal dose (LD50) in rats and mice gressing to eventual features of macronodular cirrhosis
after both oral and IP administration was determined with bridging fibrosis and regenerative nodules.
to be approximately 300 mg/kg body weight.21 Even at Although both CCl4 and TAA result in centrilobular
high doses, TAA is not toxic to cultured hepatocytes liver damage, several differences between the models
while TAA-S-oxide and TAA-S,S-oxide cause toxicity should be noted.18,28 TAA displays a longer latency
as demonstrated by increases in cytosolic lactate between the exposure to the drug and the development
dehydrogenase (LDH) release.20 The oxidation of TAA of liver injury.3,29 TAA also leads to more periportal
appears to be mediated primarily through CYP2E1, infiltrates and more pronounced ductal proliferation,
although under varying physiological conditions, the appearance of acidophilic bodies, and vacuolated
FAD-containing monooxygenases may play a more nuclei with prominent nucleoli, as well as more pro-
important role.20,22,23 When Cyp2e1-null mice were nounced hepatocyte vacuolization.18,19 Bile duct ligation
acutely exposed to TAA, they were protected from liver and repeated application of CCl4 have traditionally been
injury, elevation in markers of inflammation, and oxida- employed as the most widely used experimental model of
tive stress. By contrast, wild-type mice showed markedly fibrosis. Common to all these three models is the devel-
increased mRNA levels of tumour necrosis factor opment of hepatic fibrosis that can be most easily visua-
(TNF)-a, interleukin (IL)-6 and glutathione peroxidase lized with Sirius Red staining (Figure 3).
plus developed centrilobular necrosis and inflamma- With prolonged treatment using TAA (18 weeks and
tion.22 In addition to causing oxidative stress, glutathi- longer), cellular atypia may become evident and mitotic
one depletion and thus cellular injury, the reactive figures may be observed.10 As demonstrated in the
metabolites of TAA covalently bind to cellular proteins original TAA publication,2 chronic intoxication fre-
and lipids. Moreover, this results in alterations in cell quently results in malignancy, although TAA is cur-
cycle-related proteins, protein folding, enzyme inhibition rently rarely used in isolation as a model of HCC or
and mitochondrial/chaperone function, all of which cholangiocarcinoma. In an oral, weight-based TAA
leads to significant cytotoxicity.24–26 model, rats developed portal hypertension and
The administration of TAA causes a predictable and splenomegaly after 12 weeks of exposure, indicating a
reliable dose- and time-dependent pattern of liver injury strong functional relationship to human cirrhosis.10
that resembles human fibrosis and cirrhosis.10,27 With an Additionally, and perhaps most importantly, rodent
24 Laboratory Animals 49(S1)

Portal Pressure
(a) AST (c)
20 Control
1250
TAA
Control
1000 15
TAA

mm H2O
750
IU/L 10
500
5
250

0 0
2 4 6 8 10 12 2 4 6 8 10 12
Week Week

(d) Collagen Content


ALT
20 Control
750
TAA
Control
15
TAA

% Collagen
500
IU/L

10

250
5

0 0
2 4 6 8 10 12 2 4 6 8 10 12
Week Week

(b)
mRNA
Fold change relative to GAPDH

10.0

Col1A1
7.5
Asma
Pdgrfb
5.0 Mmp2
Timp2
2.5

0.0
0 2 4 6 8 10 12
Week

Figure 1. Time course experiment of thioacetamide (TAA) by intraperitoneal (IP) injection and phosphate-buffered saline
(PBS) controls in rats. (a) Aspartate aminotransferase (AST) (upper panel) and alanine aminotransferase (ALT) (lower
panel) levels spiked initially and then returned to normal at week 4. (b) Several fibrosis related genes demonstrated
consistent elevation throughout the course of the experiment. (c, d) Portal pressures (c) and collagen content in liver
tissue (d) plateaued at week 8. Col1A1: collagen type 1a1; Asma: a-smooth muscle actin; Pdgrfb: Platelet-derived growth
factor receptor type b; Mmp2: matrix metalloproteinase-2; Timp2: tissue inhibitor of matrix metalloproteinases 2; GAPDH:
glyceraldehyde phosphate dehydrogenase.

hepatic stellate cell activation in TAA intoxication is [PBS]-injected animal) was sacrificed every 12 weeks
central to the observed fibrosis and cirrhosis and assessment was performed of various fibrosis asso-
development.30 ciated genes by quantitative real-time reverse transcrip-
To validate the model, we performed a time-course tion-polymerase chain reaction (qRT-PCR), portal
experiment in which Sprague–Dawley rats were pressures by portal vein cannulation, biochemical ana-
exposed to TAA by IP injection (150 mg/kg, three lysis of liver injury by serum alanine aminotransferase
times/week) for 12 weeks. One group (2 experimental (ALT) and aspartate aminotransferase (AST) measure-
animals and one control phosphate-buffered saline ments, and quantification of collagen content by the
Wallace et al. 25

Figure 2. Simplified model for the formation of noxious thioacetamide (TAA) metabolites and their effects in hepatocytes.
In a first biotransformation step, TAA is reversibly metabolized to TAASO. In a second oxidation step, the highly reactive
species TAASO2 is formed. Subsequently, TAASO2 can directly react with amine groups (R-NH2) on cellular proteins or
lipids resulting in dysfunction. In addition, TAA gives rise to glutathione depletion and elevated levels of reactive oxygen
species (ROS), lipid peroxides, cytotoxicity and mitochondrial injury. Together the events may lead to apoptosis, necrosis
and the formation of hepatocellular carcinoma and cholangiocarcinoma. Details of the sequence of chemical reactions
induced by TAA can be found elsewhere.20,34

Figure 3. Histological characterization of the most widely used experimental liver fibrosis models. To visualize the most
widely used experimental models, liver sections were prepared from untreated animals, mice subjected to bile duct
ligation (BDL) for three weeks or administered carbon tetrachloride (CCl4) (2/week, 0.2 mL/kg for 12 weeks) and
thioacetamide (TAA) (3/week 100 mg/kg for 15 weeks), respectively. Haematoxylin and eosin (H&E) staining was
employed for the overall histological evaluation whereas Sirius Red staining was carried out to evaluate the extent of liver
fibrosis. Scale bar: 100 mm (top), 200 mm (bottom).
26 Laboratory Animals 49(S1)

BIO-QUANT Life Science morphometry systemÕ their effectiveness in modulating different phases of
(BIOQUANT Image Analysis Corp, Nashville, TN, fibrosis. The TAA model may also be used to study
USA). The results are presented in Figure 1. After an functional consequences of cirrhosis such as portal
initial spike in inflammation and liver injury at two hypertension, and as previously mentioned, acute
weeks as assessed by serum ALT and AST, these TAA exposure in high doses can be used to study
values returned to normal at week 4. Sustained acute liver failure and encephalopathy. Finally, TAA
mRNA elevation of fibrosis associated genes including administration may also be used to study the biology
collagen 1A1 (Col1A1), a-smooth muscle actin (Asma), of fibrosis, as it provides a reliable stimulus for stel-
platelet-derived growth factor receptor b (Pdgfrb), late cell activation and fibrosis development. Genetic
matrix metalloproteinase 2 (Mmp2) and tissue inhibitor knockdown and overexpression techniques are best
of metalloproteinase 2 (Timp2) were demonstrated. utilized in this way. The remainder of this SOP will
Both portal pressures and collagen content of liver focus on the use of TAA to produce fibrosis and
tissue reached maximal levels in TAA rats at week 8 cirrhosis.
and then plateaued. These results reinforce the efficacy . Analysis of treatment and control cohorts: Selection of
of the TAA IP injection strategy for the production of readouts will depend on the aims of the project. In a
significant fibrosis between 8 and 12 weeks. study in which the degree of fibrosis development is
the major outcome being assessed, a thorough ana-
lysis of liver histology plus a fibrosis quantification
General considerations technique should be used. Techniques used will
depend upon local expertise, but important consider-
. Safety concerns: Based on the high toxicity of TAA, ations are the use of an experienced liver histopath-
personal protective equipment including eyeglasses, ologist to assess random sections in a blinded fashion.
protective gloves, appropriate clothing and respira- We suggest formalin-fixed, paraffin embedded liver
tory mask are necessary whenever handling this hep- sections should be stained with haematoxylin and
atotoxin. To avoid chemical decomposition, TAA eosin for histological assessment of fibrosis/cirrhosis
should be stored in a cool, dry and safe place to grade plus assessment of any ductular reaction, cellu-
which only authorized staff have access (e.g. poison lar atypia or steatosis, for example. For fibrosis quan-
cabinet). tification, four sections should be stained with Sirius
. Genetic background: While the susceptibility to liver Red with nine random images taken per slide for a
fibrosis development in different experimental total of 36 images that can be analysed by the
models has been shown to display significant strain BIO-QUANT Life Science morphometry systemÕ .
differences,31 no systematic analyses on strain-speci- Alternatively, hydroxyproline quantification can be
fic susceptibility to TAA have been reported. In fact, performed. Col1A1 mRNA can be assessed by qRT-
the successful induction of TAA-mediated liver PCR and protein by Western blot. Portal pressures
fibrosis has been described in a variety of genetic can be assessed at the time of sacrifice by inserting a
backgrounds of both mice and rats.18,32 16 G angiocatheter into the portal vein and measur-
. Route, dosage, timing and duration of treatment: As ing the height of a water column.
discussed above in ‘Historic background and current . Biometric assessment In a typical experiment an
usage’, there is wide variation in currently used TAA experimental drug is concomitantly administered
models and it is beyond the scope of this standard along with TAA. A possible readout system to
operating procedure (SOP) to discuss the differences. assess the efficacy of the drug is the mRNA expres-
As a guide to planning a TAA experiment, we pre- sion of Col1A1 which may change at the end of the
sent here two models with which we have both per- experiment by a factor of 8 (with an inter-individual
sonal experience and confidence in their safety and variance of 25%) (cf. Figure 1d). The control group
efficacy for fibrosis and cirrhosis induction. will receive only TAA. It can be estimated that the
. Experimental uses and design: A typical TAA experi- drug may reduce the expression of collagen by a
ment involves the comparison of the rate or degree factor of 2. In addition, experience in our labora-
of fibrosis or cirrhosis development between a con- tories has shown that a difference of less than 30%
trol group and a treatment or intervention group. In in collagen mRNA expression does not necessarily
this era of rapidly increasing interest in anti-fibrotic impact intrahepatic collagen deposition and scar-
therapies, it is a convenient and reliable tool for the ring. For such an experiment, in our view it is advis-
relatively rapid in vivo screening of potential thera- able to use nine animals per group. If the mortality
pies, as it allows for the generation of results within rate of TAA when applied for eight weeks is further
4–5 months. The timing of the introduction of the assumed to be approximately 10%, this experiment
experimental therapies can be modified to assess should be planned with 10 animals in each group.
Wallace et al. 27

For details on how to calculate biometric param- volume isoflurane plus analgesia and laparotomy per-
eters, please refer to Scholten et al.’s article ‘The formed. Portal pressures can be measured as described
carbon tetrachloride model in mice’ published in prior to the removal of organs.
this special issue of Laboratory Animals. Side-effects: TAA administration can lead to cirrhosis
and its associated complications; however, these are
rarely the source of any discomfort to the animals.
Mild body weight loss is likely to be observed.
Practical implementation Mortality of up to 15% can be expected in the treat-
We provide here two TAA protocols, by IP injection in ment groups. This should be reported in the Methods
Sprague–Dawley rats and oral administration in section of any publications.
C57BL/6 mice. We have found these approaches to Efficacy of treatment: After 11 weeks of treatment at the
produce reliable and reproducible fibrosis and cirrhosis dose of 150 mg/kg three times per week, virtually all of
that allows for an easy distinction between control and the animals will have developed the histological fea-
treatment groups. tures of cirrhosis.

TAA via IP injection


Our experience with IP injection of rats with TAA was
TAA via oral administration
recently published14 and is summarized below as a Our experience with orally administered TAA was
standard approach for reference: recently published13 and is summarized below as a
standard for reference:
Animals: Sprague–Dawley rats between 280–300 g pur-
chased from The Jackson Laboratory (Bar Harbor, 1. Animals: 8–10-week-old C57BL/6 mice purchased
ME, USA) are kept in a 12 h light–dark cycle with from The Jackson Laboratory are kept in a 12 h
free access to water. It should be noted that recent light–dark cycle with free access to water. It should
guidelines from the US National Institutes of Health be noted that recent guidelines from the US NIH
(NIH) suggest equal numbers of male and female ani- suggest equal numbers of male and female animals be
mals be used in all future experiments. used in all future experiments. Of note, male
TAA preparation and administration: TAA purchased mice have been reported to be more susceptible to
from Sigma Chemical Co (St Louis, MO, USA) is development of TAA-induced liver fibrosis,33 which
dissolved in 0.9% normal saline to a total volume of is in line with similar findings in other fibrosis models.
1–1.5 mL per rat approximately one hour before injec- 2. TAA preparation and administration: TAA pur-
tion. Treatment groups receive 150 mg/kg of TAA chased from Sigma Chemical Co (St Louis, MO,
three times per week for a total of 11 weeks while USA) is dissolved in drinking water at 300 mg/L
control groups receive the same volume of 0.9% and administered for 2–4 months. Control animals
normal saline. are given normal water with the same volume. In
General handling: Rats are examined regularly for order to reduce the risk of infection, drinking
signs of distress or ill health and weighed prior to each water should be filtered and acidified (pH around
injection to accurately calculate the dosage of TAA. 4.0). The water volume should be measured on a
Injection technique: Rats are handled at all times with daily basis and changed twice weekly.
care, as the experience can be distressing. We recom- 3. General handling: Mice are examined daily for signs
mend that lab personnel with specific training in animal of distress or ill health. The body weight is deter-
handling be responsible for the IP injections. Rats are mined twice a week starting at the beginning of the
scuffed behind the neck between the middle and index experiment.
fingers with the tail hooked between the little and ring 4. Animal sacrifice: At the end of the study period ani-
fingers to expose the abdomen. A sterile, disposable mals are placed under inhalation anaesthesia with
syringe (25–27 G) is used to inject the TAA into the 1–5% by volume isoflurane plus analgesia and lapar-
lower side of the abdomen with care taken to avoid otomy is performed. Portal pressures can be measured
injection into the bladder or abdominal organs. The as described prior to removal of organs.
solution should be at room temperature at time of 5. Side-effects: TAA administration can lead to cirrho-
injection. The routine administration of anaesthesia is sis and its associated complications; however, these
generally not recommended as it results in unnecessary are rarely the source of any discomfort to the ani-
stress to the animals. mals. Mild body weight loss is likely to be observed.
Animal sacrifice: At the end of the study period animals Low mortality can be expected in the treatment
are placed under inhalation anaesthesia with 1–5% by groups. This should be reported in the Methods
28 Laboratory Animals 49(S1)

section of any publications. In general, oral adminis- have presented two TAA models with which we have
tration of TAA leads to less extrahepatic toxicity than had success in reliably and reproducibly induce chronic
IP administration due to the first-pass effect. In con- liver injury and fibrosis in rodents.
trast to the intraperitoneal treatment, mice are not
known to experience pain and a more continuous Ethical statement
dosing is achieved. Because of these advantages, we All animal experiments were performed in accordance with
prefer oral administration to IP dosage, although the the United States Animal Welfare Act and the Guide for the
latter has been used more frequently in the literature. Care and Use of Laboratory Animals of the National
6. Efficacy of treatment: There is a progressive develop- Institutes of Health. The Institutional Animal Care and Use
Committee (IACUC) of the Icahn School of Medicine at
ment of fibrosis and cirrhosis development across the
Mount Sinai approved all the animal experiments performed.
study period. At the end of four months virtually all
of the treatment animals will have developed the
histological features of cirrhosis. Funding
Work in Dr. Friedman’s laboratory is supported by NIH
Grants RO1DK56621 and RO1AA020709.

Classification of severity of procedure Conflict of interest statement


According to Article 15 of the EU Directive 2010/63 The authors have no conflicts of interest to declare.
(http://eur-lex.europa.eu/LexUriServ/
LexUriServ.do?uri¼OJ:L:2010:276:0033:0079:en:PDF) References
the estimated degree of pain, suffering, distress or last- 1. Childs JFL and Siegler EA. Controlling orange decay.
ing harm to the animals subjected to TAA application Ind Eng Chem 1946; 38: 82–87.
should be classified according to a scoring system that 2. Fitzhugh OG and Nelson AA. Liver tumors in rats fed
consists of ‘non-recovery’, ‘mild’, ‘moderate’ or thiourea or thioacetamide. Science 1948; 108: 626–628.
‘severe’. The classification criteria that underpin this 3. Dashti H, Jeppsson B, Hägerstrand I, et al.
Thioacetamide- and carbon tetrachloride-induced liver
assessment have been established by the Expert
cirrhosis. Eur Surg Res 1989; 21: 83–91.
Working Group on severity classification of scientific
4. Müller A, Machnik F, Zimmermann T and Schubert H.
procedures performed on animals. Further details can Thioacetamide-induced cirrhosis-like liver lesions in rats
be found at: http://ec.europa.eu/environment/chem- – usefulness and reliability of this animal model. Exp
icals/lab_animals/pdf/report_ewg.pdf. Pathol 1988; 34: 229–236.
The severity of the procedure depends on the TAA 5. Muñoz Torres E, Paz Bouza JI, López Bravo A,
dose and duration of the experiment. TAA application Abad Hernández MM and Carrascal Marino E.
to induce fibrosis with no major impairment of liver Experimental thioacetamide-induced cirrhosis of the
function is classified as a moderate procedure accord- liver. Histol Histopathol 1991; 6: 95–100.
ing to Article 15 of the aforementioned EU Directive 6. Peeling J, Shoemaker L, Gauthier T, Benarroch A,
2010/63. The application of TAA to induce liver failure Sutherland GR and Minuk GY. Cerebral metabolic and
histological effects of thioacetamide-induced liver failure.
with associated complications such as cerebral oedema
Am J Physiol 1993; 265: G572–G578.
and encephalopathy, coagulopathy, renal, haemo-
7. Butterworth RF, Norenberg MD, Felipo V, Ferenci P,
dynamic and cardiorespiratory failure is classified as Albrecht J and Blei AT Members of the ISHEN
a severe procedure. According to the 3 R principle, Commission on Experimental Models of HE.
the procedure should undergo a refinement, and Experimental models of hepatic encephalopathy:
humane endpoints must be implemented with frequent ISHEN guidelines. Liver Int 2009; 29: 783–788.
observation points to restrict pain, suffering, distress or 8. Kreft B, Dombrowski F, Block W, Bachmann R, Pfeifer
lasting harm to the animals, at which point the proced- U and Schild H. Evaluation of different models of experi-
ure can be reclassified as moderate. mentally induced liver cirrhosis for MRI research with
correlation to histopathologic findings. Invest Radiol
1999; 34: 360–366.
Concluding remarks 9. Li X, Benjamin IS and Alexander B. Reproducible pro-
duction of thioacetamide-induced macronodular cirrho-
The administration of TAA via the oral or IP route is a
sis in the rat with no mortality. J Hepatol 2002; 36:
safe and effective technique for the study of fibrosis in 488–493.
rodents. There is a wide variation in the route, dose, 10. Laleman W, Vander Elst I, Zeegers M, et al. A stable
timing and animal used. The scientific community model of cirrhotic portal hypertension in the rat: thioa-
would benefit from the adoption of a universal SOP cetamide revisited. Eur J Clin Invest 2006; 36: 242–249.
for the administration of TAA with the intent of study- 11. Kimura M, Fujii Y, Yamamoto R, et al. Involvement of
ing fibrosis and its associated complications. Here we multiple cell cycle aberrations in early preneoplastic liver
Wallace et al. 29

cell lesions by tumor promotion with thioacetamide in a in relation to changes due to thioacetamide. Mol Cell
two-stage rat hepatocarcinogenesis model. Exp Toxicol Biochem 2000; 208: 1–10.
Pathol 2013; 65: 979–988. 28. Popov Y and Schuppan D. Targeting liver fibrosis: stra-
12. Kornek M, Raskopf E, Guetgemann I, et al. Combination tegies for development and validation of antifibrotic
of systemic thioacetamide (TAA) injections and ethanol therapies. Hepatology 2009; 50: 1294–1306.
feeding accelerates hepatic fibrosis in C3H/He mice and 29. Zimmermann T, Müller A, Machnik G, Franke H,
is associated with intrahepatic up regulation of MMP-2, Schubert H and Dargel R. Biochemical and morpho-
VEGF and ICAM-1. J Hepatol 2006; 45: 370–376. logical studies on production and regression of experi-
13. Wang X, Lopategi A, Ge X, et al. Osteopontin induces mental liver cirrhosis induced by thioacetamide in
ductular reaction contributing to liver fibrosis. Gut 2014; Uje:WIST rats. Z Versuchstierkd 1987; 30: 165–180.
63: 1805–1818. 30. Salguero Palacios R, Roderfeld M, et al. Activation of
14. Traber PG, Chou H, Zomer E, et al. Regression of fibro- hepatic stellate cells is associated with cytokine expression
sis and reversal of cirrhosis in rats by galectin inhibitors in thioacetamide-induced hepatic fibrosis in mice. Lab
in thioacetamide-induced liver disease. PLoS One 2013; 8: Invest 2008; 88: 1192–1203.
e75361. 31. Walkin L, Herrick SE, Summers A, et al. The role of mouse
15. Dyroff MC and Neal RA. Studies of the mechanism of strain differences in the susceptibility to fibrosis: a system-
metabolism of thioacetamide S-oxide by rat liver micro- atic review. Fibrogenesis Tissue Repair 2013; 6: 18.
somes. Mol Pharmacol 1983; 23: 219–227. 32. Yang MC, Chang CP and Lei HY. Induction of liver
16. Hunter AL, Holscher MA and Neal RA. Thioacetamide- fibrosis in a murine hepatoma model by thioacetamide
induced hepatic necrosis. I. Involvement of the mixed- is associated with enhanced tumor growth and sup-
function oxidase enzyme system. J Pharmacol Exp Ther pressed antitumor immunity. Lab Invest 2010; 90:
1977; 200: 439–448. 1782–1793.
17. Zimmermann C, Ferenci P, Pifl C, et al. Hepatic enceph- 33. Schnur J, Olah J, Szepesi A, Nagy P and Thorgeirsson S.
alopathy in thioacetamide-induced acute liver failure in Thioacetamide-induced hepatic fibrosis in transforming
rats: characterization of an improved model and study of growth factor b-1 transgenic mice. Eur J Gastroenterol
amino acid-ergic neurotransmission. Hepatology 1989; 9: Hepatol 2004; 16: 127–133.
594–601. 34. Sarma D, Hajovsky H, Koen YM, et al. Covalent modi-
18. Strnad P, Tao GZ, Zhou Q, et al. Keratin mutation pre- fication of lipids and proteins in rat hepatocytes and
disposes to mouse liver fibrosis and unmasks differential in vitro by thioacetamide metabolites. Chem Res
effects of the carbon tetrachloride and thioacetamide Toxicol 2012; 25: 1868–1877.
models. Gastroenterology 2008; 134: 1169–1179. 35. Honda H, Ikejima K, Hirose M, et al. Leptin is required
19. Jang JH, Kang KJ, Kim YH, Kang YN and Lee IS. for fibrogenic responses induced by thioacetamide in the
Reevaluation of experimental model of hepatic fibrosis murine liver. Hepatology 2002; 36: 12–21.
induced by hepatotoxic drugs: an easy, applicable, and 36. Wasmuth HE, Lammert F, Zaldivar MM, et al.
reproducible model. Transplant Proc 2008; 40: Antifibrotic effects of CXCL9 and its receptor CXCR3
2700–2703. in livers of mice and humans. Gastroenterology 2009; 137:
20. Hajovsky H, Hu G, Koen Y, et al. Metabolism and tox- 309–319.
icity of thioacetamide and thioacetamide S-oxide in rat 37. Tuñón MJ, Alvarez M, Culebras JM and
hepatocytes. Chem Res Toxicol 2012; 25: 1955–1963. González-Gallego J. An overview of animal models for
21. Lewis RJ Sr and Sax NI. Sax’s dangerous properties of investigating the pathogenesis and therapeutic strategies
industrial materials. Volumes 1–3, 9th ed. New York: Van in acute hepatic failure. World J Gastroenterol 2009; 15:
Nostrand Reinhold, 1996. 3086–3098.
22. Kang JS, Wanibuchi H, Morimura K, et al. Role of 38. Avraham Y, Grigoriadis N, Poutahidis T, et al.
CYP2E1 in thioacetamide-induced mouse hepatotoxicity. Cannabidiol improves brain and liver function in a ful-
Toxicol Appl Pharmacol 2008; 228: 295–300. minant hepatic failure-induced model of hepatic enceph-
23. Chilakapati J, Korrapati MC, Shankar K, et al. Role of alopathy in mice. Br J Pharmacol 2011; 162: 1650–1658.
CYP2E1 and saturation kinetics in the bioactivation of 39. Becker FF. Thioacetamide hepatocarcinogenesis. J Natl
thioacetamide: effects of diet restriction and phenobar- Cancer Inst 1983; 71: 553–558.
bital. Toxicol Appl Pharmacol 2007; 219: 72–84. 40. Munoz Torres E, Paz Bouza JI, Lopez Bravo A, Abad
24. Jeong DH, Jang JJ, Lee SJ, et al. Expression patterns of cell Hernandez MM and Carrascal Marino E. Experimental
cycle-related proteins in a rat cirrhotic model induced by thioacetamide-induced cirrhosis of the liver. Histol
CCl4 or thioacetamide. J Gastroenterol 2001; 36: 24–32. Histopathol 1991; 6: 95–100.
25. Staňková P, Kučera O, Lotková H, Roušar T, Endlicher R 41. Mueller K, Sunami Y, Stuetzle M, et al. CHOP-mediated
and Cervinková Z. The toxic effect of thioacetamide on hepcidin suppression modulates hepatic iron load.
rat liver in vitro. Toxicol In Vitro 2010; 24: 2097–2103. J Pathol 2013; 231: 532–542.
26. Koen YM, Sarma D, Hajovsky H, et al. Protein targets of 42. Zaldivar MM, Pauels K, von Hundelshausen P, et al.
thioacetamide metabolites in rat hepatocytes. Chem Res CXC chemokine ligand 4 (Cxcl4) is a platelet-derived
Toxicol 2013; 26: 564–574. mediator of experimental liver fibrosis. Hepatology
27. Al-Bader A, Mathew TC, Abul H, Al-Sayer H, Singal PK 2010; 51: 1345–1353.
and Dashti HM. Cholangiocarcinoma and liver cirrhosis

Potrebbero piacerti anche