Submit manuscript...
Journal of
eISSN: 2471-1381

Liver Research, Disorders & Therapy

Review Article Volume 1 Issue 1

Liver injury models for induction of hepatic oval cells in rodents

Ek Khoon Tan,1,2 Maureen Shuh,1 Ari J Cohen1,3,4

1Laboratory of Transplant Research, Institute of Translational Research, USA
2Department of General Surgery, Singapore General Hospital, Singapore
3Multi-Organ Transplant Center, Ochsner Health System, USA
4Ochsner Clinical School, University of Queensland School of Medicine, USA

Correspondence: Ek Khoon Tan, Laboratory of Transplant Research, Institute of Translational Research, Ochsner Clinic Foundation, 1514 Jefferson Highway, New Orleans, LA 7012, USA, Tel 1504 8425764, Fax 1504 8425746

Received: June 24, 2015 | Published: July 24, 2015

Citation: Tan EK, Shuh M, Cohen A. Liver injury models for induction of hepatic oval cells in rodents. J Liver Res Disord Ther. 2015;1(1):15-27. DOI: 10.15406/jlrdt.2015.01.00005

Download PDF

Abstract

Hepatic oval cells are progenitor stem cells residing in the liver that play an important role in liver regeneration when hepatocyte response to the injury is inadequate. On-going studies continue to characterize these elusive cells, understand their role in carcinogenesis, and determine their potential for therapeutics. Since oval cells proliferate only upon exposure to liver injury, a multitude of injury models have been developed in the past years to investigate their activity. These injury models comprise chemical, surgical and biological components. Depending on the chemical toxicity or lethality of injury, different extents and time required to induce oval cell responses are seen. Here, we review the various strategies available that can be used to induce hepatic oval cells in rodents.

Keywords: animal models, liver, liver regeneration, oval cells, rodentia

Introduction

The liver is a remarkable organ with immense regenerative potential. Hesiod’s Theogony describes the punishment meted out to Prometheus for restoring fire to humanity–his liver would be eaten by an eagle daily only for it to regenerate overnight before the ordeal is revisited. Modern science now affords us the opportunity to investigate why and how the liver regenerates. Most humans tolerate major hepatectomies with the remnant liver undergoing hypertrophy to maintain homeostasis. Animal experiments reveal that after a partial hepatectomy (PH), rats and mice regenerate their liver mass almost completely by 7 and 14days, respectively.1‒3

Injury models to explore liver behavior with chemical induction of cirrhosis and carcinogenesis led to the work of Opie EL.4 He recognized the development of biliary ductular cells proliferating around portal tracts of rats fed butter yellow (dimethyl aminoazobenzene). These cells were further characterized and given the name “oval cells” by Farber E.5 Since then, numerous studies using a multitude of injury models in rodents have identified oval cells as hepatic progenitor or stem cells. We reviewed the MEDLINE database with a PubMed search using terms “hepatic oval cells” up till Dec 2013. Publications written in English pertaining to injury models and oval cells were reviewed in-depth. We begin with a concise summary of hepatic oval cells before reviewing the chemical and surgical strategies available to induce hepatic oval cells in rodents.

Hepatic oval cells

After massive injury, the liver regenerates by compensatory proliferation of hepatocytes.6 However, when this pathway is inhibited or inadequate such as hepatocyte replicative senescence, hepatic oval cells may proliferate.7 In such situations, oval cells constitute an available transit amplifying cell compartment to continue liver regeneration and repopulation.8,9

Oval cells appear as small cells approximately 1/3 the size of hepatocytes.10 As so named, they are oval-shaped with unclear margins and scant, slightly basophilic-like cytoplasm.5 The cell has a large round or oval-shaped blue-staining nuclei on routine hematoxylin & eosin staining, that contains a fine chromatin network and prominent nucleolus. In mice, oval cells have a diameter of 7-10µm.11,12 In rats, they measure 7-12µm.13‒17 They are even slightly larger in hamsters at 7-14µm.18 When oval cells proliferate, active DNA synthesis occurs and this can be observed using bromodeoxyuridine or 5-ethynyl-2’-deoxyuridine labeling.15 Electron microscopy indicate that oval cells initially exhibit characteristics of undifferentiated cells with a high ratio of nucleus to cytoplasm with obvious nucleoli, rare cytoplasmic organelles with only few mitochondria, endoplasmic reticulum and some villus-like apophysis the cell surface.10,19 The nuclei and cytoplasmic organelles resemble bile duct cells except that oval cells contain greater numbers of ribonucleoprotein granules.20

While the scientific literature on the origin of oval cells is still incomplete, oval cells are likely to originate from the embryonic ductal plate and reside facultatively at the canal of Hering.12,21‒23 Upon injury, they first appear near biliary ductules, spreading along portal tracts before infiltrating the rest of the liver.5,24,25 Initially, they resemble proliferating bile duct cells, save that they do not form recognizable ductular profiles.26 Cell surface markers have identified oval cells in normal rodent liver.27 Normal mice have on average 8.07±5.02 oval cells per portal tract,28 or 0.04% of total liver cells.29 In normal rats, 2.5±0.5 x105 oval cells have been isolated per 100g body weight.30

Oval cell surface markers

A major impediment in studying oval cells is the lack of consistent and specific cellular markers. Identification begins with hematoxylin and eosin staining to visualize typical histological characteristics. In the past decade, developments in immunohistochemistry and hybridoma technology have defined the oval cell population further.31Numerous cellular markers are reported to identify oval cell populations although only few have been specific to oval cells. This may be due to the progressive differentiation of oval cells in which the expression patterns of markers change. Due to the cross-reactivity of markers with other cell types, conflicting studies are often found positing for and against the same marker. CD44, for example, has been linked to oval cells,32 undifferentiated oval cell phenotypes,33 and small hepatocytes.34 Thymocyte antigen 1 (Thy-1), a hematopoietic stem cell marker, is expressed on oval cells35 although other studies report the opposite and that bone marrow cells,36 myofibroblasts and activated stellate cells are Thy-1 positive.37

Nonetheless, some markers more commonly applied by the research community are described. Markers related to immature oval cell phenotypes include c-kit, stem cell antigen-1 (Sca-1), neural cell adhesion molecule and CD3411,38‒40 while markers for mature phenotypes share similarities to adult biliary cells or hepatocytes. Murine oval cell markers include A6, Cytokeratin 19 (CK19), OV-6 and M2-isozyme of pyruvate kinase (M2PK).41‒45 For rats, markers include but are not limited to gamma-glutamyl transpeptidase (γGT), CK19, OV-6, BD-2, OC-2, Thy-1, β-catenin, M2PK, alpha-fetoprotein (AFP), delta-like 1 homolog (Dlk-1), albumin and epithelial cell adhesion molecule. 16,26,32,35,37,46‒52 Other animals in the Rodentia family share similar antigens on their oval cells. Oval cells in hamsters express CK19, AFP, albumin and OV-618,53 while the woodchuck expresses CK18.54

To isolate oval cells, they are first separated from hepatocytes through centrifugation with the oval cells partitioning into the non-parenchymal cell fraction. Thereafter, methods such as Nycodenz gradient separation, flow cytometry or immuno-magnetic sorting (using above-stated cell markers) are applied to isolate oval cells.55 These methods allowed isolation of highly-pure fractions of oval cells of more than 95% with demonstrable proliferative potential.35,56 Nevertheless to the lack of specific cell surface antigens, a combination of criteria is used to define oval cell populations during cell isolation.

Two of these criteria are described:

  1. diameter of 10-15µm, presence of CK19, presence of γGT activity, presence of albumin, absence of peroxidase activity.46
  2. diameter of 10-15µm, immunoreactivity to OV-6, expression of CK19 and placental glutathione S-transferase (πGST), presence of γGT activity, partial expression of albumin and AFP, and absence of peroxidase activity.13

Functions of hepatic oval cells and potential for clinical use

Oval cells are hepatic progenitor cells as they possess some stem cell characteristics. Stem cells are defined as undifferentiated cells capable of proliferation, self-maintenance, producing a large number of differentiated functional progeny, regenerating tissue after injury and flexibility in the use of these options.57 As mentioned earlier, hepatic oval cells proliferate to regenerate the liver after injury. Established stem cell markers such as Sca-1,11 CD34,11,40 CD35;11 and markers of hepatoblasts such as CD44,32,33 Dlk-133,52,58 have been identified on oval cells.59 Oval cells also demonstrate the side population phenotype characteristic of stem cells by their ability to efflux Hoechst 33342 dye.60 Importantly, oval cells are able to differentiate into both hepatocytes and biliary epithelial cells.16,37,61‒64 Oval cells can also differentiate into other cells. For example, intestinal metaplasia has been observed in 0.02% N-acetyl-2-aminofluorene (2-AAF)/PH model.65,66 Trans-differentiation of oval cells toward pancreatic beta-cell phenotype with production of pro-insulin67 occurs when exposed to high glucose environments.68,69

In keeping with having properties of a stem-cell, oval cells have been reported to differentiate into cells outside an endodermal lineage. Oval cells have been found to express vimentin and N-cadherin indicating their ability to acquire mesenchymal properties,32,37,40 and can differentiate into mesenchymal cells after being implanted subcutaneously into a host.70 Deng et al reported oval cells undergoing neural trans-differentiation after being injected into brain tissue, adopting the morphology and antigenic phenotype of macro- and microglial cells and assuming immune-positivity for nestin.71

Stem cells, which possess the ability for tissue repair and regeneration, may be the key to treating severe and chronic illnesses. Hence, much interest lies in the potential of oval cells as a therapeutic agent to reconstitute injured livers.72 Cultured oval cells have been propagated to 100 passages without malignant transformation, highlighting the possibility of an expandable cell-source for hepatocytes.73 When transfused into a host, oval cells demonstrate liver-tropism and can repopulate up to 90% of the liver after PH.37,74 Transplanting oval cells in rats improves liver function and survival after D-galactosamine (GaIN) induced fulminant hepatic failure75 and ameliorates ischemia/reperfusion injury.64 In rat liver transplantation, addition of oval cells prolonged median survival from 21 to 38days,14 improved liver function and reduced rejection activity.14,64 These findings suggest significant potential for oval cells as therapy for acute liver failure and modulating liver transplantation outcomes.72

Background on rodent liver injury models

The Solt-Farber protocol was the first consistent and reproducible method to induce oval cell proliferation.76 This involved an intra-peritoneal dose of di-ethylnitrosamine (DEN) 20mg per 100g body weight at day 0 followed by a recovery period of 2weeks. Thereafter, 2-AAF was added in the basal diet and PH performed one week later. Oval cells were identified 30hours after PH.

Since then, variations to streamline this induction process, new toxicology tests yielding oval cells, and gene modification have led to a myriad of injury models. We propose an adaptation of Terblanche et al.77 model for acute hepatic failure.

An ideal injury model to study oval cells would comprise:

  1. a standardized animal model,
  2. an option for gene modification to investigate activation pathways,
  3. reproducible oval cell response,
  4. minimize mortality to allow studying long-term effects of oval cell activation and carcinogenesis,
  5. minimal hazard to personnel.

We will first discuss the factors surrounding animal models followed by the injury agents.

Animal models

Species, gender and strain: Rats and mice are the two most common rodents used as they are easily standardized animal models. Few studies have used hamsters and woodchucks. Each species possesses different metabolic pathways and liver micro-environments. Between species, the same agent does not generate equivalent oval cell responses and cellular expression, possibly even of oval cell phenotype, as indicated by discrepant markers.52 Gender discrepancies have been observed in rats. In the pioneering work by Farber, oval cell responses after ethionine and 2-AAF were seen only in male Wistar rats while livers of corresponding females were normal.5 Bisgaard et al.78 also found that 2-AAF elicited smaller mitogenic responses in female Fischer-344 rats compared to males. This phenomenon may be due to female rats having a smaller enzymatic capacity to N-hydroxylate 2-AAF into N-hydroxy-2-AAF, the chemical that elicits the mitogenic effects of 2-AAF.79 Notwithstanding, oval cell proliferation in female Wistar, Sprague-Dawley and Fischer-344 rats has been reported in other studies.80‒84

Limited information is available on strain-related differences although one study by Wood et al.85 compared oval cell responses between 2 strains of age-matched rats. The oval cell response in Fischer-344 rats was extensive but oval cells remained localized to the portal areas in Copenhagen rats. Further work is required to characterize the gender discrepancies within rats, the different rat strains and how they affect interpretation of oval cell responses.

Age: Consistent with other stem cell types, oval cells are subject to age-related quiescence and decline in function.86 While experiments with mice ranging from 3 up to 12weeks have identified oval cells,43,87,88 age-related discrepancies have been noted in terms of number of cells and proliferative ability. With regard to number of oval cells, conflicting evidence has been reported. Comparing mice older and younger than 8weeks undergoing an interval-feeding injury model, no oval cells were seen in mice younger than 8 weeks. It was suggested that this may be related to how oval cells respond to cumulative proliferative demand over extended periods of time.3 On the other hand, studies exist reporting age dependent oval cell responses in the opposite direction, with younger animals demonstrating greater responses compared to older animals.89,90 Concerning the actual proliferative ability of the oval cells isolated, evidence has been consistent in finding that oval cells from older mice have lower regenerative potential.25,90 Cell lines cultured from mice 4weeks old were maintained for more than 30 passages, but those from mice 8weeks old were maintained for only 20 passages.25 Therefore, the age of the animal may influence the extent of oval cell response as well as the ability of the oval cells to effect a regenerative response.

Injury agent

Basis and methods of oval cell induction: There are two differing hypotheses on how oval cells are activated. One hypothesis is the resistant hepatocyte model that involves the use of a toxic carcinogen.76 The cytotoxic effect of the carcinogen causes hepatic cell death which selects for cells resistant to or acquire resistance to the toxicity and gain a proliferative advantage. Oval cells become selected and activated through this mechanism. Experiments by Anil kumar et al.91 instead indicate that oval cells belong to a facultative stem cell compartment. The alternative two-hit hypothesis suggests that oval cells exist facultatively and that both hepatocytes and oval cells respond to the injury, except that hepatocytes proliferate preferentially. A “first hit” exhausts or inhibits hepatocyte responses while the “second hit” initiates the oval cell response.92,93

Cellular injury does not guarantee oval cell proliferation.94 The site of injury within the liver (centri-lobular vs peri-portal) influences the time taken and magnitude of oval cell response. Chemicals such as N-acetyl-p-aminophenol (APAP) and carbon tetrachloride (CCl4) which preferentially damage centri-lobular liver have been observed to induce quicker and greater oval cell responses.28,92 This has been hypothesized to be due to oval cells residing in the peri-portal region. Hence, agents targeting centri-lobular regions spare the oval cells, allowing for more prolific responses.28 Combining agents may result in synergistic effects. For example, allyl alcohol (AA) and CCl4 were compared using dosages that resulted in equivalent levels of hepatic necrosis. When 2-AAF was added to these dosages of AA and CCl4, the 2-AAF/CCl4 model generated significantly greater and more persistent responses as compared to 2-AAF/AA.92

Classification of injury agents: Injury models can be broadly classified into chemical, surgical and biological, or a combination. The 2 most common injury methods are chemical agents (Table 1) and the combination of chemical agents with PH (Table 2). The high number of permutations of agents, dosage, delivery method, kinetics, and duration presents a challenge in providing a meaningful analysis. We have elected to discuss the published results of more commonly used agents.

Model

Species

Selected References

CDE

Mouse, Rat

[17,20,27,44,99,192]

DDC

Mouse

[11,12,52,100,101,106,193,194]

GaIN

Rat

[94,111-115]

DEN

Mouse

[117-119]

APAP

Mouse

[28,52,127]

CCl4

Mouse, Rat

[130,131]

2-AAF

Rat

[5,133]

2-AAF/CCl4

Rat

[33,92,94,136,182]

2-AAF/AA

Rat

[92,136,139]

Choline deficient diet/DEN/2-AAF

Hamster

[53]

3’-Me-DAB

Rat

[5,51,144,188]

Phenobarbital/Cocaine hydrochloride

Mouse

[145]

TCPOBOP

Mouse

[45]

DDC/TCPOBOP

Mouse

[101]

Retrorsine/GaIN

Rat

[115]

Fumosinin B1

Rat

[148,149]

Ethanol

Mouse, Rat

[7,151]

High fat diet & ethanol

Mouse

[150]

Iron overload

Rat

[152]

CDE: Choline-Deficient Diet Supplemented with Ethionine; 3’-Me-DAB: 3’-Methyl-4-Dimethyl Aminoazobenzene; DDC: 3,5-Diethoxycarbonyl-1,4-Dihydrocollidine; Gain: D-Galactosamine; DEN: Di-Ethyl Nitrosamine; APAP: N-Acetyl-P-Aminophenol; Ccl4: Carbon Tetrachloride; 2-AAF: N-Acetyl-2-Aminofluorene; AA: Allyl Alcohol; TCPOBOP: 1,4 Bis[2-(3,5-Dichloropyridyloxy)] Benzene.

Table 1 Chemical liver injury models in rodents.

Model

Species

Selected References

DEN/PH

Rat

[120]

2-AAF/PH

Mouse, Rat

[14,24,37,52,62,75,141,162,195]

DEN/2-AAF/PH

Rat

[76,85,121,122]

Choline deficient diet/DEN/2-AAF/PH

Hamster

[18]

Dipin/PH

Mouse

[42,146,147]

Retrorsine/PH

Rat

[32,196]

Ethanol/PH

Mouse

[45]

DEN: Di-Ethylnitrosamine; PH: 2/3 Partial Hepatectomy; 2-AAF: N-Acetyl-2-Aminofluorene; Dipin: 1,4-Bis [N,N’-Di(Ethylene)-Phosphamide]-Piperazine.

Table 2 Combined chemical and surgical liver injury models in rodents. 

Chemical

  1. Choline-deficient diet supplemented with ethionine (CDE): Choline, a quaternary compound (trimethyl-β-hydroxy-ethylammonium), is an essential nutrient and precursor of the acetylcholine. It is needed to form cellular membranes and is an important methyl-group donor necessary for converting homocysteine to methionine.95 Choline deficiency leads to inflammation and oxidative stress, causing steatohepatitis and liver fibrosis. Its involvement with methylation and epigenetic changes also causes genetic mutations and cancer when deficient.96
  2. Choline-deficient diets can be supplemented with ethionine in drinking water to exacerbate their toxicity.97 Ethionine is the ethyl antagonist of methionine and results in hypomethylation of hepatic DNA due to the accumulation of S- adenosyl ethionine.98 This is the most commonly applied chemical injury model on both rats and mice.20,44 Akhurst et al.99 suggested that an ideal response using CDE in mice was achieved with 50% choline-deficient diet and 0.15% DL–ethionine for 2-3weeks. Full choline-deficient diets resulted in extensive hepatic steatosis while 50% choline-deficient diet maintained the mice in a good condition with minimal steatosis. Reducing ethionine resulted in fewer oval cells seen.99 Peak oval cell response and plateauing of doubling time is seen at around 2weeks.27,99

  3. 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC): DDC is a porphyrinogenic hepatotoxin that can be reconstituted at 0.1% in the diet to generate a prolific oval cell response at 3-6weeks.52‒100 It activates constitutive androstane receptor (CAR, NR113) that induces transcription of the cytochrome p450 2b gene in hepatocytes.101 CAR also regulates endogenous energy metabolism by inhibiting key gluconeogenic genes that encode phosphoenoyl pyruvate carboxylkinase and glucose-6-phosphatase.102,103
  4. DDC causes the accumulation of N-methylprotoporphyrin IX, a potent inhibitor of the enzyme ferrochelatase which converts protoporphyrin IX into heme.104 This results in aggregation of protoporphyrin IX and porphyrin precursor δ-aminolevulinic acid.105 Porphyrin crystals form within hepatocytes in the peri-portal regions,101 causing hepatocyte, ductal and oval cell proliferation.12 In contradistinction to other models where hepatocyte proliferation is inhibited, hepatocyte proliferation continues allowing the study of interactions between hepatocytes and oval cells.106

  5. D-Galactosamine (GaIN): GaIN is a hexosamine metabolized through the galactose pathway in the liver.107 GaIN sequesters available uridine which results in depletion of intracellular uridine derivatives in hepatocytes and loss of intracellular calcium homeostasis.108 Cell membranes and organelles are disrupted, and RNA and protein synthesis are arrested.77,109 GaIN liver injury in rats causes pan-lobular, focal hepatic necrosis that is microscopically and biochemically similar to that of viral hepatitis in humans.110 GaIN is administered intra-peritoneally as a single dose to rats between 50-140mg per 100g body weight. Maximal oval cell proliferation is seen between 2-5days94,111,112 and oval cells disappeared 7-10days after the injection.113,114 GaIN can also be given 2weeks after retrorsine, a tumorigenic pyrrolizidine alkaloid, to generate greater numbers of oval cells.115
  6. Di-ethylnitrosamine (DEN): DEN belongs to the family of carcinogenic N-nitroso compounds.116 It causes alkylation of nucleic acids and proteins in hepatocytes resulting in cirrhosis and cancer.117 DEN can be given per-oral or intra-peritoneally, and/or in combination with other agents.47,76,118 Mouse models use DEN alone (intra-peritoneal)119 while rat and hamster models use DEN (either per-oral or intra-peritoneal) in combination with 2-AAF and/or PH.53,120,121 For rats, oral preparations are dosed at 1mg per 100g body weight120 while the intra-peritoneal dose is 15-20mg per 100g body weight.122,123 In the DEN/2-AAF/PH model, peak oval cell response is seen 7 and 14days after PH.85,124
  7. N-acetyl-p-aminophenol (APAP): APAP is an aminobenzene that is metabolized by cytochrome p450 into the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). NAPQI is usually conjugated by glutathione in the liver into harmless cysteine and mercapturic acid conjugates. When given in overdose quantities, glutathione stores are depleted and NAPQI covalently binds to DNA and cysteine residues on hepatic proteins leading to 3-(cysteine-S-yl) APAP adducts.125 Changes in the liver may be detected within 30minutes of injury – sinusoidal endothelial cells swell and microvascular congestion occurs in the centri-lobular region.126 Necrosis of hepatocytes develops at around 2hours.126 Oval cell responses are detected as early as 3hours after injury and it is hypothesized that this rapid response is due to a centri-lobular form of injury.28 For mice, APAP is given as a single intra-peritoneal dose between 25-100mg per 100g body weight28,52 or in multiple doses at 30mg per 100g body weight.127
  8. Carbon Tetrachloride (CCl4): CCl4 is a halogenated alkane that is metabolized in the liver into trichloromethyl radical (CCl3*) and chlorine. CCl3* binds to numerous cellular molecules, produces reactive oxygen species and forms DNA adducts.128 Similar to APAP, CCl4 preferentially damages the centri-lobular region.92 Studies in rats have found that CCl4 alone cannot generate an oval cell response94,114,129,130 although the converse has been reported.131 In mice, few A6 positive oval cells were seen after chronic CCl4 exposure.130 CCl4 is commonly combined with 2-AAF for use in rats. In 2-AAF/CCl4 models, the oral dose of CCl4 is 90mg per 100g body weight81 while the intra-peritoneal dose is 150mg per 100g body weight.36,83,132 Maximal oval cell responses after 2-AAF/CCl4 are between days 7-9 after the start of the protocol.36,83,132
  9. N-acetyl-2-aminofluorene (2-AAF): N-acetyl-2-aminofluorene (2-AAF) is an aromatic amine that becomes metabolized to N-hydroxy-2-AAF which is the potent proximate carcinogen.78 It blocks proliferation of hepatocytes94 and forms DNA adduct leading eventually to cancer.133 After 1week of 0.02% 2-AAF in the diet, at least 95% of hepatocyte replicative response is inhibited when PH is performed.134 The liver mass of 2-AAF/PH treated rats is arrested until 11days after PH (from 33 to 57%) compared to near complete regeneration in 7days after PH alone.2 Since 2-AAF needs to be metabolically activated, various oval cell responses have been seen depending on the gender, strain and species.5,78,79,135
  10. 2-AAF mixed in the diet (constituting 0.02-0.04%) generates an oval cell response after 2-4weeks.5,133 2-AAF is frequently combined with PH, CCl4 or AA in mouse and rat models.62,76,92,136 These studies delivered 2-AAF at doses of 0.4-2mg per 100g body weight daily33,52,66,137 or 1-1.5mg per day by gavage.24,138 In recent years, 2-AAF is commonly administered with a time-released pellet implanted subcutaneously or intra-peritoneally, delivering 2.5mg per day.35,139,140 In 2-AAF/PH models, 2-AAF is started for 4-6days prior to PH, omitted (if administered orally) on the day of PH, and resumed thereafter for another 4-10days.37,62,75,141 Maximal oval cell proliferation after 2-AAF/PH is reported at 6-11days after PH.15,37,52,142

  11. Others: Numerous other chemical agents are available. One example is 3’-methyl-4-dimethylaminoazobenzene (3’-Me-DAB), an azo dye with significant hepatocarcinogenic properties. Nearly 100% of rats develop hepatocellular carcinoma after 20weeks of 3’-Me-DAB diet.143 Measuring serum AFP levels while on 3’-Me-DAB demonstrates two peaks: the first corresponds with the appearance of oval cells while the second indicates the development of hyperplastic nodules and hepatocellular carcinoma.143,144 Cocaine hydrochloride is a hepatotoxin that, in contra-distinction to APAP and CCl4, causes peri-portal injury.145 TCPOBOP (1, 4 bis [2-(3,5-dichloropyridyloxy)]-benzene) activates the same CAR-ligand as DDC. Interestingly, there is preferential CYP2B10 induction in centri-lobular hepatocytes, as opposed to DDC.101 Dipin (1, 4-bis [N,N’-di(ethylene)-phosphamide]-piperazine) is an alkylating drug that causes irreversible damage to DNA, RNA and proteins.146 Dipin is combined with PH to induce oval cell proliferation although the time taken varies from 1-11weeks.42,142,146,147 Fumosinin B1, a mycotoxin produced by Fusarium moniliforme, is a non-genotoxic carcinogen that induces oval cell proliferation but allows for hepatocytes to proliferate, similar to DDC.148,149 Diet modification using high fat or alcohol diets leads to steatohepatitis and oval cells responses can be observed after chronic exposure.7,150,151 Chronic diet-induced iron overload also shows similar oval cell responses.152

Surgical

  1. Partial hepatectomy (PH): The surgical procedures that can be performed on rodent livers to induce injury are listed in Table 3. The most commonly used procedure is PH, which involves removing 50-83.4% of the liver.76,137,152 A widely described technique is the 70% or 2/3 PH model described by Higgins et al.154 The left lateral and median lobes are approximately 72% of total liver weight.155 PH alone does not always generate oval cells 114,156,157 and is used in combination with chemical agents. However, some studies have isolated oval cells in a pure PH model.153,158,159 This is a unique opportunity to study oval cell responses in livers that are otherwise healthy and naïve to toxic injury. The modified Solt-Farber protocol uses 2-AAF/PH without DEN.62,100,160 2-AAF/PH can be used on mice and rats, and peak responses are between 7 to 11days after PH.37,52,60,156,161,162
  2. Bile duct or portal vein branch ligation: Bile duct ligation causes cholestatic liver injury and biliary ductal proliferation. Oval cell proliferation is not commonly associated with this model156,157,163 although few studies have identified oval cells 7 and 10days post-ligation using M2PK and CK19 as markers, respectively.164,165 Ligating a portal vein branch causes ischemia, atrophy of the affected lobes and compensatory hypertrophy of non-affected lobes. Ligating the left portal vein branch produces a peak oval cell response after 7days.166,167 Similar to PH alone, these ligation models allows the study of oval cells induced by non-carcinogenic injury.167
  3. Other surgical adjuncts: Parasympathetic stimulation is known to modulate oval cell responses. Vagotomized rats generated a smaller response due to the loss of stimulatory effects of acetylcholine via the muscarinic acetylcholine receptor type-3.168 Inhibiting the sympathetic nervous system chemically with 6-hydroxydopamine or pharmacologically with prazosin generated heightened oval cell responses but less hepatic necrosis and steatosis.169 These findings have implications on the role of oval cells in liver transplantation since donor grafts are denervated.

Model

Species

Selected References

PH

Mouse, Rat

[153,158,159]

Bile duct ligation

Rat

[164,165]

Portal vein branch ligation

Mouse, Rat

[166,167]

PH: 2/3 Partial hepatectomy.

Table 3 Surgical liver injury models in rodents.

Biological: Biological injury models involve gene modification or the introduction of and infection or immune-mediated injury (Table 4). Gene modification generates animals which develop different forms of liver injury. Mutation of the Atp7b gene creates Long-Evans Cinnamon rats that have toxic accumulation of copper within the liver. Spontaneous hepatitis develops at 16weeks of age with concomitant oval cell proliferation.170,171 Homozygous obese (ob/ob) mice develop steatohepatitis and oval cell responses similar to alcohol-induced models.7,45 AL-uPA transgenic mice express hepatotoxic urokinase-type plasminogen activator in hepatocytes, generating a proliferative response from trans-gene deficient hepatocytes and oval cells.172 Gene-transfer models with suicide genes injure the liver biochemically. One example is recombinant adenoviral vector AdCMVtk which contains herpes simplex virus thymidine kinase (HSV-tk) gene. When ganciclovir is administered to animals treated with AdCMVtk, HSV-tk phosphorylates ganciclovir into a cytotoxic derivative that causes severe liver injury and massive oval cell responses.173 Adenoviral vectors can also introduce growth factors such as hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) to modulate oval cell behavior.155,174 In the cases of HGF and VEGF, increased oval cell responses were seen after liver injury.155,174

Model

Species

Selected References

Long-Evans Cinnamon

Rat

[170,171]

Obese mutant (ob/ob)

Mouse

[7,45]

Urokinase-type plasminogen activator transgene

Mouse

[172]

Herpes simplex virus thymidine kinase gene

Rat

[173]

Taurine transporter knockout (taut-/-)

Mouse

[175]

Helicobacterhepaticus

Mouse

[177,178]

Woodchuck hepatitis virus

Woodchuck

[54,179]

Woodchuck hepatitis virus/ Aflatoxin B1

Woodchuck

[179,197]

Fas receptor/CD95

Mouse

[25]

Table 4 Biological liver injury models in rodents.

Gene knockout models can be used to induce liver injury or investigate pathways of oval cell activation. Taurine transporter knockout (taut-/-) mice develop non-specific non-steatotic hepatitis and oval cells could be identified in these mice beyond 1year of age.175 Interleukin-6 (IL-6), WW45 and p53 knock-out models are some examples used with injury models described above to delineate oval cell signaling and carcinogenic pathways.87,145,176 Infective hepatitis models include Helicobacter hepaticus infection in mice177,178 and chronic woodchuck hepatitis virus in woodchucks.54,179 Acute and chronic hepatitis develops in these animals, eliciting oval cell responses and subsequently, hepatocellular carcinoma. Direct immune-mediated injury can be induced by stimulating the Fas receptor/CD95 causing apoptosis of hepatocytes and fulminant hepatic failure.180 Tsuchiya et al.180 injected mice intra-peritoneally with 3 doses of 0.03mg per 100g body weight anti-mouse Fas every 2days and isolated oval cells after 5days.25

Dose, duration and delivery of injury agent: The dose of injurious agent should be titrated to deliver significant liver injury and yet minimize both short and long-term mortality. In the case of chemicals, we can introduce the agent at LD50 dose.92,114 The LD50 dose by definition induces mortality in 50% of experimental animals and has served as a historical test to compare the toxicity between different agents. This may be appropriate for single dose agents such as GaIN and CCl4. Other injury models such as choline-deficient ethionine supplemented (CDE) diet and 3, 5-diethoxycarbonyl-1, 4-dihydrocollidine (DDC) require sequential dosing, and their dosages have been derived empirically.

Extent of injury can be objectively assessed by measuring the transaminitis induced145 or by quantifying the amount of hepatic necrosis on microscopy.92 It should be noted that oval cells may not respond equally even if the extent of injury is comparable biochemically. One study compared GaIN with CCl4 dose-adjusted to equivalent extents of hepatic necrosis and levels of serum aspartate aminotransferase.94 In this study, GalN induced a generous oval cell response while CCl4 did not.94

Depending on the injury agent, the first appearance of oval cells may be anytime between 1 day to 3weeks after inducing the injury51,62,80,143,181 while peak response varies between 5days to 6weeks.20,50,148,161 Cessation of the injury agent leads to termination of oval cell response182,183 although changes to the liver such as cholangiofibrosis are not reversed once formed.50 Prolonging exposure to injury may allow greater oval cell proliferation but risks increased hepatic fibrosis. If the intent is to isolate oval cells, distorting the liver architecture may impede perfusion digestion methods thereby reducing yields.46

The method of delivering the agent is an important component of the experimental design that may lead to variable results.184 Animal-dependent feeding (with the drug mixed in the diet or drinking water) is certainly influenced by the keenness of the animal to feed, but may also be influenced by the cage design leading to non-standardized oral-intake and dosing.185 In this case, it was a cage that limited access to food. Alternative methods of gavage feeding, subcutaneously implanted time-release pellets, or intra-peritoneal injections addresses this problem of consistency in dosimetry. Gavage feeding delivers the drug directly to the stomach and is associated with some animal discomfort but is otherwise safe and avoids anesthetic agents that may interfere with the metabolic profile of the animal. The disadvantage is enterally-delivered drugs are subject to a first-pass effect with its associated issues of variable absorption and metabolism. This leads to variable effects and potentially a slower onset of action. Parenteral methods (subcutaneously-implanted pellet or intra-peritoneal injection) avoid the first-pass effect but are still subject to absorption (either through the subcutaneous tissue or peritoneum). They also involve a minor surgical procedure and anesthetic and risks infective or surgical complications that may compromise the experiment.184 Another concern albeit unrelated to the animal is the hazard to personnel. Ethionine, for example, is an airborne carcinogen and can be potential risk to staff when administered in powder or pellet form. Administering ethionine in the drinking water instead will reduce this hazard for research personnel.99

Comparing oval cell responses: Last, the degree of oval cells responses can be compared between injury models. Four scoring systems were identified and listed in Table 5. Each system proposes reasonable methods of assessing oval cell response. Smaller scores indicate oval cells only around the portal tracts. Higher scores reflect greater proliferation and more extensive infiltration into liver parenchyma.119,124,149,172 Based upon the articles and reports with negative findings identified in this review, the most reliable method appears to favor the use of 2-AAF/PH liver injury model as described above. In the absence of an available animal surgeon, the reliable chemical induction methods identified were choline-deficient diet with ethionine supplementation (CDE) or 3, 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC).

Liver injury models and carcinogenesis: The process of cancer induction and the entity cancer stem cells are comprehensive topics that warrant discourses of their own. Nevertheless, we briefly introduce some relevant issues. Many injury models inducing oval cell responses also form cancer nodules as chronic injury leads to acquisition of mutations over periods of hyper-proliferation. Oval cells have been suspected to be tumor-initiating cells or cancer stem cells.186,187 Indeed, oval cell responses often correlate directly with the likelihood of tumor formation.44,188 Cancerous and oval cells often co-express markers such as OV-6, c-kit, Glypican-3, and AFP as well.39,111,114,122 Since much interest in oval cells come from the potential for therapy, this raises the concern of transplanting potentially cancerous cells.189 It was reported that oval cell lines became tumorigenic and formed carcinomas when inoculated into rats and nude mice.17,87 However, non-tumorigenic propagable oval cell lines have also been reported in literature.73,190 The significance of oval cells in carcinogenesis needs to be further explored and perhaps, may lead to developments in targeted cancer therapy.191‒197

Future direction

Oval cells may play an important role in liver regeneration. We summarized pertinent information on oval cells and reviewed various injury models used to generate them. Future work to characterize these cells will provide greater insight into the exact pathways of activation and translate toward therapeutic utility. Developing surgical methods to induce oval cells will allow research on oval cells naïve to toxic chemicals and recreate scenarios that better mirror clinical situations. Elucidation of malignant transformation pathways will determine the exact role oval cells play in carcinogenesis and may potentially contribute toward targeted therapy.

Acknowledgements

None.

Conflict of interest

Author declares that there is no conflict of interest.

References

  1. Batusic DS, von Bargen A, Blaschke S, et al. Different physiology of interferon-alpha/-gamma in models of liver regeneration in the rat. Histochem Cell Biol. 2011;136(2):131‒144.
  2. Cimica V, Batusic D, Chen Y, et al. Transcriptome analysis of rat liver regeneration in a model of oval hepatic stem cells. Genomics. 2005;86(3):352‒364.
  3. Ueberham E, Lindner R, Kamprad M, et al. Oval cell proliferation in p16INK4a expressing mouse liver is triggered by chronic growth stimuli. J Cell Mol Med. 2008;12(2):622‒638.
  4. Opie EL. The pathogenesis of tumors of the liver produced by butter yellow. J Exp Med. 1944;80(3):231‒246.
  5. Farber E. Similarities in the sequence of early histological changes induced in the liver of the rat by ethionine, 2-acetylamino-fluorene, and 3'-methyl-4-dimethylaminoazobenzene. Cancer Res. 1956;16(2):142‒148.
  6. Alison M, Golding M, Lalani el N, et al. Wound healing in the liver with particular reference to stem cells. Philos Trans R Soc Lond B Biol Sci. 1998;353(1370):877‒894 .
  7. Yang S, Koteish A, Lin H, et al. Oval cells compensate for damage and replicative senescence of mature hepatocytes in mice with fatty liver disease. Hepatology. 2004;39(2):403‒411.
  8. Assimakopoulos SF, Tsamandas AC, Alexandris IH, et al. Stimulation of oval cell and hepatocyte proliferation by exogenous bombesin and neurotensin in partially hepatectomized rats. World J Gastrointest Pathophysiol. 2011;2(6):146‒154.
  9. Sanchez A, Factor VM, Schroeder IS, et al. Activation of NF-kappaB and STAT3 in rat oval cells during 2-acetylaminofluorene/partial hepatectomy-induced liver regeneration. Hepatology. 2004;39(2):376‒385.
  10. Fang CH, Gong JQ, Zhang W. Function of oval cells in hepatocellular carcinoma in rats. World J Gastroenterol. 2004;10(17):2482‒2487.
  11. Petersen BE, Grossbard B, Hatch H, et al. Mouse A6-positive hepatic oval cells also express several hematopoietic stem cell markers. Hepatology. 2003;37(3):632‒640.
  12. Wang X, Foster M, Al-Dhalimy M, et al. The origin and liver repopulating capacity of murine oval cells. Proc Natl Acad Sci U S A. 2003;100:11881‒11888.
  13. Kaplanski C, Pauley CJ, Griffiths TG, et al. Differentiation of rat oval cells after activation of peroxisome proliferator-activated receptor alpha43. Cancer Res. 2000;60(3):580‒587.
  14. Li Z, Chen J, Li L, et al. In vitro proliferation and differentiation of hepatic oval cells and their potential capacity for intrahepatic transplantation. Braz J Med Biol Res. 2013;46(8):681‒688.
  15. Qin AL, Zhou XQ, Zhang W, et al. Characterization and enrichment of hepatic progenitor cells in adult rat liver. World J Gastroenterol. 2004;10(10):1480-1486.
  16. Wang P, Liu T, Cong M, et al. Expression of extracellular matrix genes in cultured hepatic oval cells: an origin of hepatic stellate cells through transforming growth factor beta? Liver Int. 2009;29(4):575‒584.
  17. Yoshimura H, Harris R, Yokoyama S, et al. Anaplastic carcinomas in nude mice and in original donor strain rats inoculated with cultured oval cells. Am J Pathol. 1983;110(3):322‒332.
  18. Yoon BI, Choi YK, Kim DY. Differentiation processes of oval cells into hepatocytes: proposals based on morphological and phenotypical traits in carcinogen-treated hamster liver. J Comp Pathol. 2004;131(1):1‒9.
  19. Alison MR, Poulsom R, Jeffery R, et al. Expression of hepatocyte growth factor mRNA during oval cell activation in the rat liver. The Journal of Pathology. 1993;171(4):291‒299.
  20. Grisham JW, Hartroft WS. Morphologic identification by electron microscopy of "oval" cells in experimental hepatic degeneration. Lab Invest. 1961;10:317‒332.
  21. Carpentier R, Suner RE, van Hul N, et al. Embryonic ductal plate cells give rise to cholangiocytes, periportal hepatocytes, and adult liver progenitor cells. Gastroenterology. 2011;41(4):1432‒1438 .
  22. Theise ND, Saxena R, Portmann BC, et al. The canals of Hering and hepatic stem cells in humans. Hepatology. 1999;30(6):1425‒1433.
  23. Petersen BE, Bowen WC, Patrene KD, et al. Bone marrow as a potential source of hepatic oval cells. Science. 1999;284(5417):1168‒1170.
  24. Thorgeirsson SS, Evarts RP, Bisgaard HC, et al. Hepatic stem cell compartment: activation and lineage commitment. Proc Soc Exp Biol Med. 1993;204(3):253‒260.
  25. Tsuchiya A, Heike T, Baba S, et al. Long-term culture of postnatal mouse hepatic stem/progenitor cells and their relative developmental hierarchy. Stem Cells. 2007;25(4):895‒902.
  26. Lenzi R, Liu MH, Tarsetti F, et al. Histogenesis of bile duct-like cells proliferating during ethionine hepatocarcinogenesis. Evidence for a biliary epithelial nature of oval cells. Lab Invest. 1992;66(3):390‒402.
  27. Knight B, Matthews VB, Akhurst B, et al. Liver inflammation and cytokine production, but not acute phase protein synthesis, accompany the adult liver progenitor (oval) cell response to chronic liver injury. Immunol Cell Bio. 2005;83(4):364‒374.
  28. Kofman AV, Morgan G, Kirschenbaum A, et al. Dose- and time-dependent oval cell reaction in acetaminophen-induced murine liver injury. Hepatology. 2005;41(6):1252‒1261.
  29. Qiu Q, Hernandez JC, Dean AM, et al. CD24-positive cells from normal adult mouse liver are hepatocyte progenitor cells. Stem Cells Dev. 2011;20(12):2177‒2188.
  30. Guo J, Li J, Lu Y, et al. A novel technique for hepatic progenitor cell isolation from normal adult rat livers. ASAIO J. 2012;58(1):73‒78.
  31. Yang L, Faris RA, Hixson DC. Phenotypic heterogeneity within clonogenic ductal cell populations isolated from normal adult rat liver. Proc Soc Exp Biol Med. 1993;204(3):280‒288.
  32. Yovchev MI, Grozdanov PN, Joseph B, et al. Novel hepatic progenitor cell surface markers in the adult rat liver. Hepatology. 2007;45(1):139‒149.
  33. Chiu CC, Sheu JC, Chen CH, et al. Global gene expression profiling reveals a key role of CD44 in hepatic oval-cell reaction after 2-AAF/CCl4 injury in rodents. Histochem Cell Biol. 2009;132(5):479‒489.
  34. Kon J, Ooe H, Oshima H, et al. Expression of CD44 in rat hepatic progenitor cells. J Hepatol. 2006;45(1):90‒98.
  35. Petersen BE, Goff JP, Greenberger JS, et al. Hepatic oval cells express the hematopoietic stem cell marker Thy-1 in the rat. Hepatology. 1998;27(2):433‒445.
  36. Corcelle V, Stieger B, Gjinovci A, et al. Characterization of two distinct liver progenitor cell subpopulations of hematopoietic and hepatic origins. Exp Cell Res. 2006;312(15):2826‒2836.
  37. Yovchev MI, Grozdanov PN, Zhou H, et al. Identification of adult hepatic progenitor cells capable of repopulating injured rat liver. Hepatology. 2008;47(2):636‒647.
  38. Bird TG, Lorenzini S, Forbes SJ. Activation of stem cells in hepatic diseases. Cell Tissue Res. 2008;331(1):283‒300.
  39. Fang CH, Zhang W, Zhu XY, et al. The expression of c-kit and proliferating cell nuclear antigen in oval cells of rats with hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int. 2003;2(4):537‒544.
  40. Kubota K, Soeda J, Misawa R, et al. Bone marrow-derived cells fuse with hepatic oval cells but are not involved in hepatic tumorigenesis in the choline-deficient ethionine-supplemented diet rat model. Carcinogenesis. 2008;29(2):448‒454.
  41. Dorrell C, Erker L, Lanxon-Cookson KM, et al. Surface markers for the murine oval cell response. Hepatology. 2008;48(4):1282‒1291.
  42. Engelhardt NV, Factor VM, Yasova AK, et al. Common antigens of mouse oval and biliary epithelial cells. Expression on newly formed hepatocytes. Differentiation. 1990;45(1):29‒37.
  43. Okabe M, Tsukahara Y, Tanaka M, et al. Potential hepatic stem cells reside in EpCAM+ cells of normal and injured mouse liver. Development. 2009;136(11):1951‒1960.
  44. Knight B, Yeoh GC, Husk KL, et al. Impaired preneoplastic changes and liver tumor formation in tumor necrosis factor receptor type 1 knockout mice. J Exp Med. 2000;192(12):1809‒1818.
  45. Roskams T, Yang SQ, Koteish A, et al. Oxidative stress and oval cell accumulation in mice and humans with alcoholic and nonalcoholic fatty liver disease. Am J Pathol. 2003;163(4):1301‒1311.
  46. Pack R, Heck R, Dienes HP, et al. Isolation, biochemical characterization, long-term culture, and phenotype modulation of oval cells from carcinogen-fed rats. Exp Cell Res. 1993;204(2):198‒209.
  47. Dunsford HA, Karnasuta C, Hunt JM, et al. Different lineages of chemically induced hepatocellular carcinoma in rats defined by monoclonal antibodies. Cancer Res. 1989;49(17):4894‒4900.
  48. Hixson DC, Allison JP. Monoclonal antibodies recognizing oval cells induced in the liver of rats by N-2-fluorenylacetamide or ethionine in a choline-deficient diet. Cancer Res. 1985;45(8):3750‒3760.
  49. Yang L, Jung Y, Omenetti A, et al. Fate-mapping evidence that hepatic stellate cells are epithelial progenitors in adult mouse livers. Stem Cells. 2008;26(8):2104‒2113.
  50. Steinberg P, Klingelhoffer A, Schafer A, et al. Expression of pyruvate kinase M2 in preneoplastic hepatic foci of N-nitrosomorpholine-treated rats. Virchows Arch. 1999;434(3):213‒220.
  51. Dempo K, Chisaka N, Yoshida Y, et al. Immunofluorescent study on alpha-fetoprotein-producing cells in the early stage of 3'-methyl-4-dimethylaminoazobenzene carcinogenesis. Cancer Res. 1975;35(5):1282‒1287.
  52. Jelnes P, Santoni-Rugiu E, Rasmussen M, et al. Remarkable heterogeneity displayed by oval cells in rat and mouse models of stem cell-mediated liver regeneration. Hepatology. 2007;45(6):1462‒1470.
  53. Yoon BI, Jung SY, Hur K, et al. Differentiation of hamster liver oval cell following Clonorchis sinensis infection. J Vet Med Sci. 2000;62(12):1303‒1310.
  54. Su Q, Zerban H, Otto G, et al. Cytokeratin expression is reduced in glycogenotic clear hepatocytes but increased in ground-glass cells in chronic human and woodchuck hepadnaviral infection. Hepatology. 1998;28(2):347‒359.
  55. Shupe TD, Piscaglia AC, Oh SH, et al. Isolation and characterization of hepatic stem cells, or "oval cells," from rat livers. Methods Mol Biol. 2009;482:387‒405.
  56. Xu C, Chen X, Chang C, et al. Genome-wide expression profiling of hepatic oval cells after partial hepatectomy in rats. Tissue Cell. 2011;43(5):291‒303.
  57. Potten CS, Loeffler M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development. 1990;110(4):1001‒1020.
  58. Tanimizu N, Tsujimura T, Takahide K, et al. Expression of Dlk/Pref-1 defines a subpopulation in the oval cell compartment of rat liver. Gene Expr Patterns. 2004;5(2):209‒218.
  59. Shiojiri N, Lemire JM, Fausto N. Cell lineages and oval cell progenitors in rat liver development. Cancer Res. 1991;51(10):2611‒2620.
  60. Shimano K, Satake M, Okaya A, et al. Hepatic oval cells have the side population phenotype defined by expression of ATP-binding cassette transporter ABCG2/BCRP1. Am J Pathol. 2003;163(1):3‒9.
  61. Darwiche H, Oh SH, Steiger-Luther NC, et al. Inhibition of Notch signaling affects hepatic oval cell response in rat model of 2AAF-PH. Hepat Med. 2011;3:89‒98.
  62. Evarts RP, Nagy P, Marsden E, et al. A precursor-product relationship exists between oval cells and hepatocytes in rat liver. Carcinogenesis. 1987;8(11):1737‒1740.
  63. Fougere-Deschatrette C, Imaizumi-Scherrer T, Strick-Marchand H, et al. Plasticity of hepatic cell differentiation: bipotential adult mouse liver clonal cell lines competent to differentiate in vitro and in vivo. Stem Cells. 2006;24(9):2098‒2109.
  64. Li Z, Chen J, Li L, et al. In vitro and in vivo characteristics of hepatic oval cells modified with human hepatocyte growth factor. Cell Mol Biol Lett . 2013;18(4):507‒521.
  65. Barut V, Sarraf CE. Intestinal metaplasia in liver of rats after partial hepatectomy and treatment with acetylaminofluorene. Cell Prolif. 2009;42(5):657‒660.
  66. Zhang W, Chen XP, Zhang WG, et al. Hepatic non-parenchymal cells and extracellular matrix participate in oval cell-mediated liver regeneration. World J Gastroenterol. 2009;15(5):552‒560.
  67. Vorobeychik M, Bloch K, Zemel R, et al. Immunohistochemical evaluation of hepatic oval cell activation and differentiation toward pancreatic beta-cell phenotype in streptozotocin-induced diabetic mice. J Mol Histol. 2008;39(4):463‒468.
  68. Li Y, Zhao LJ, Xia FZ, et al. Transdifferentiation of hepatic oval cells into pancreatic islet beta-cells. Front Biosci. 2012;17:2391‒2395.
  69. Yang L, Li S, Hatch H, et al. In vitro trans-differentiation of adult hepatic stem cells into pancreatic endocrine hormone-producing cells. Proc Natl Acad Sci U S A. 2002;99(12):8078‒8083.
  70. Dong HH, Xiang S, Chen XP, et al. The epithelial-mesenchymal transition promotes transdifferentiation of subcutaneously implanted hepatic oval cells into mesenchymal tumor tissue. Stem Cells Dev. 2009;18(9):1293‒1298.
  71. Deng J, Steindler DA, Laywell ED, et al. Neural trans-differentiation potential of hepatic oval cells in the neonatal mouse brain. Exp Neurol. 2003;182(2):373‒382.
  72. Forbes SJ, Newsome PN. New horizons for stem cell therapy in liver disease. J Hepatol. 2012;56(2):496‒499.
  73. Wang P, Cong M, Liu TH, et al. Primary isolated hepatic oval cells maintain progenitor cell phenotypes after two-year prolonged cultivation. J Hepatol. 2010;53(5):863‒871.
  74. Chen JZ, Hong H, Xiang J, et al. A selective tropism of transfused oval cells for liver. World J Gastroenterol. 2003;9(3):544‒546.
  75. Wu CX, Zou Q, Zhu ZY, et al. Intrahepatic transplantation of hepatic oval cells for fulminant hepatic failure in rats. World J Gastroenterol. 2009;15(12):1506‒1511.
  76. Solt DB, Medline A, Farber E. Rapid emergence of carcinogen-induced hyperplastic lesions in a new model for the sequential analysis of liver carcinogenesis. Am J Pathol. 1977;88(3):595‒618.
  77. Terblanche J, Hickman R. Animal models of fulminant hepatic failure. Dig Dis Sci. 1991;36(6):770‒774.
  78. Bisgaard HC, Nagy P, Santoni-Rugiu E, et al. Proliferation, apoptosis, and induction of hepatic transcription factors are characteristics of the early response of biliary epithelial (oval) cells to chemical carcinogens. Hepatology. 1996;23(1):62‒70.
  79. Kriek E. Fifty years of research on N-acetyl-2-aminofluorene, one of the most versatil
  80. Anilkumar T, Ryan CJ, Aslam M, et al. The anti-proliferative effect of plasma from rats with acute fulminant hepatic failure. Scand J Gastroenterol. 1997;32(11):1152‒1161.
  81. e compounds in experimental cancer research. J Cancer Res Clin Oncol. 1992;118(7):481‒489.
  82. Ito Y, Watanabe T, Nagatomo S, et al. Annexin A3-expressing cellular phenotypes emerge from necrotic lesion in the pericentral area in 2-acetylaminofluoren/carbon tetrachloride-treated rat livers. Biosci Biotechnol Biochem. 2007;71(12):3082‒3089.
  83. Piscaglia AC, Shupe TD, Oh SH, et al. Granulocyte-colony stimulating factor promotes liver repair and induces oval cell migration and proliferation in rats. Gastroenterology. 2007;133(2):619‒631 .
  84. Yin L, Lynch D, Ilic Z, et al. Proliferation and differentiation of ductular progenitor cells and littoral cells during the regeneration of the rat liver to CCl4/2-AAF injury. Histol Histopathol. 2002;17(1):65‒81.
  85. Wojcik M, Bobowiec R, Lisiecka U, et al. Proliferation, differentiation and apoptosis of choline deficient ethionine supplemented diet-rat oval cells under the influence of 2-methoxyestradiol. J Physiol Pharmacol. 2012;63(6):669‒676.
  86. Wood GA, Sarma DS, Archer MC. Resistance to the promotion of glutathione S-transferase 7-7-positive liver lesions in Copenhagen rats. Carcinogenesis. 1999;20(7):1169‒1175.
  87. Chakkalakal JV, Jones KM, Basson MA, et al. The aged niche disrupts muscle stem cell quiescence. Nature. 2012;490(7420):355‒360.
  88. Dumble ML, Croager EJ, Yeoh GC, et al. Generation and characterization of p53 null transformed hepatic progenitor cells: oval cells give rise to hepatocellular carcinoma. Carcinogenesis. 2012;23(3):435‒445.
  89. Strick-Marchand H, Masse GX, Weiss MC, et al. Lymphocytes support oval cell-dependent liver regeneration. J Immunol. 2008;181(4):2764‒2771.
  90. Guest I, Ilic Z, Sell S. Age dependence of oval cell responses and bile duct carcinomas in male fischer 344 rats fed a cyclic choline-deficient, ethionine-supplemented diet. Hepatology. 2010;52(5):1750‒1757.
  91. Tarnowski M, Koryciak-Komarska H, Czekaj P, et al. The comparison of multipotential for differentiation of progenitor mesenchymal-like stem cells obtained from livers of young and old rats. Folia Histochem Cytobiol. 2007;45(3):245‒254.
  92. Anilkumar TV, Golding M, Edwards RJ, et al. The resistant hepatocyte model of carcinogenesis in the rat: the apparent independent development of oval cell proliferation and early nodules. Carcinogenesis. 1995;16(4):845‒853.
  93. Petersen BE, Zajac VF, Michalopoulos GK. Hepatic oval cell activation in response to injury following chemically induced periportal or pericentral damage in rats. Hepatology. 1998;27(4):1030‒1038.
  94. Yoon SM, Gerasimidou D, Kuwahara R, et al. Epithelial cell adhesion molecule (EpCAM) marks hepatocytes newly derived from stem/progenitor cells in humans. Hepatology. 2011;53(3):964‒973.
  95. Schmiedeberg P, Biempica L, et al. Timing of protooncogene expression varies in toxin-induced liver regeneration. J Cell Physiol. 1993;154(2):294‒300.
  96. Al-Humadi H, Zarros A, Kyriakaki A, et al. Choline deprivation: an overview of the major hepatic metabolic response pathways. Scand J Gastroenterol. 2012;47(8‒9):874‒886.
  97. Zeisel SH. Dietary choline deficiency causes DNA strand breaks and alters epigenetic marks on DNA and histones. Mutat Res 2012;733(1‒2):34‒38.
  98. Niederau C, Luthen R, Niederau MC, et al. Acute experimental hemorrhagic-necrotizing pancreatitis induced by feeding a choline-deficient, ethionine-supplemented diet. Methodology and standards. Eur Surg Res. 1992;24:40‒54.
  99. Shivapurkar N, Wilson MJ, Poirier LA. Hypomethylation of DNA in ethionine-fed rats. Carcinogenesis. 1984;5(8):989‒992.
  100. Akhurst B, Croager EJ, Farley-Roche CA, et al. A modified choline-deficient, ethionine-supplemented diet protocol effectively induces oval cells in mouse liver. Hepatology. 2001;34(3):519‒522.
  101. Apte U, Thompson MD, Cui S, et al. Wnt/beta-catenin signaling mediates oval cell response in rodents. Hepatology. 2008;47(1):288‒295.
  102. Yamazaki Y, Moore R, Negishi M. Nuclear receptor CAR (NR1I3) is essential for DDC-induced liver injury and oval cell proliferation in mouse liver. Lab Invest. 2011;91(11):1624‒1633.
  103. Dong B, Saha PK, Huang W, et al. Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease. Proc Natl Acad Sci U S A. 2009;106(44):18831‒18836.
  104. Ueda A, Hamadeh HK, Webb HK, et al. Diverse roles of the nuclear orphan receptor CAR in regulating hepatic genes in response to phenobarbital. Mol Pharmacol. 2002;61(1):1‒6.
  105. Cole SP, Marks GS. Ferrochelatase and N-alkylated porphyrins. Mol Cell Biochem. 1984;64(2):127‒37.
  106. Fonia O, Weizman R, Coleman R, et al. PK 11195 aggravates 3,5-diethoxycarbonyl-1,4-dihydrocollidine-induced hepatic porphyria in rats. Hepatology. 1996;24(3):697‒701.
  107. Hu M, Kurobe M, Jeong YJ, et al. Wnt/beta-catenin signaling in murine hepatic transit amplifying progenitor cells. Gastroenterology. 2007;133(5):1579‒1591.
  108. Tunon MJ, Alvarez M, Culebras JM, et al. An overview of animal models for investigating the pathogenesis and therapeutic strategies in acute hepatic failure. World J Gastroenterol. 2009;15(25):3086‒3098 .
  109. Ferencikova R, Cervinkova Z, Drahota Z. Hepatotoxic effect of D-galactosamine and protective role of lipid emulsion. Physiol Res. 2003;52(1):73‒78.
  110. Shinozuka H, Farber JL, Konishi Y, et al. D-galactosamine and acute liver cell injury. Fed Proc. 1973;32(4):1516‒1526.
  111. Kucera O, Lotkova H, Kand'ar R, et al. The model of D-galactosamine-induced injury of rat hepatocytes in primary culture. Acta Medica (Hradec Kralove). 2006;49(1):59‒65.
  112. Grozdanov PN, Yovchev MI, Dabeva MD. The oncofetal protein glypican-3 is a novel marker of hepatic progenitor/oval cells. Lab Invest. 2006;86(12):1272‒1284.
  113. Holic N, Suzuki T, Corlu A, et al. Differential expression of the rat gamma-glutamyl transpeptidase gene promoters along with differentiation of hepatoblasts into biliary or hepatocytic lineage. Am J Pathol. 2000;157(2):537‒548 .
  114. Ichinohe N, Kon J, Mitaka T. Isolation of hepatic progenitor cells from the galactosamine-treated rat liver. Methods Mol Biol. 2012;826:49‒58.
  115. Kuhlmann WD, Peschke P. Hepatic progenitor cells, stem cells, and AFP expression in models of liver injury. Int J Exp Pathol. 2006;87(5):343‒359.
  116. Yu CH, Chang MH, Chien CS, et al. Hepatocyte transplantation and the differentiation fate of host oval cells in acute severe hepatic injury. Cell Transplant. 2010;19(2):231‒243.
  117. Lewis DF, Brantom PG, Ioannides C, et al. Nitrosamine carcinogenesis: rodent assays, quantitative structure-activity relationships, and human risk assessment. Drug Metab Rev. 1997;29(4):1055‒1078.
  118. Xu XM, Yuan GJ, Deng J, et al. Hepatic oval cells activated by hepatocyte apoptosis in diethylnitrosamine-induced rat liver cirrhosis. Saudi Med J. 2010;31(5):490‒494.
  119. Wu K, Ding J, Chen C, et al. Hepatic transforming growth factor beta gives rise to tumor-initiating cells and promotes liver cancer development. Hepatology. 2012;56(6):2255‒2267 .
  120. He XY, Smith GJ, Enno A, et al. Short-term diethylnitrosamine-induced oval cell responses in three strains of mice. Pathology. 1994;26(2):154‒160.
  121. Oredipe OA, Schumm DE, Mercurio F, et al. A carcinogenesis- and tumorigenesis-associated rat fetal protein: an immuno-histochemical and immuno-biochemical study utilizing a new monoclonal antibody, MOFP. Carcinogenesis. 1989;10(12):2175‒2181.
  122. Zhu HZ, Zhang XL, Chen YS. Expression of glutathione S-transferase placental mRNA in hepatic preneoplastic lesions in rats. World J Gastroenterol. 1998;4(1):38‒40.
  123. Faris RA, Monfils BA, Dunsford HA, et al. Antigenic relationship between oval cells and a subpopulation of hepatic foci, nodules, and carcinomas induced by the "resistant hepatocyte" model system. Cancer Res. 1991;51(4):1308‒1317.
  124. He RK, Gascon-Barre M. Influence of the vitamin D status on the early hepatic response to carcinogen exposure in rats. J Pharmacol Exp Ther. 1997;281(1):464‒469.
  125. Park YN, Jung WH, Park C. The effect of deferoxamine on the preneoplastic lesions in the chemically induced hepatocarcinogenesis. Yonsei Med J. 1994;35(4):388‒395.
  126. Qiu Y, Benet LZ, Burlingame AL. Identification of the hepatic protein targets of reactive metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and mass spectrometry. J Biol Chem. 1998;273(28):17940‒17953.
  127. Ito Y, Bethea NW, Abril ER, et al. Early hepatic microvascular injury in response to acetaminophen toxicity. Microcirculation. 2003;10(5):391‒400.
  128. Shen K, Chang W, Gao X, et al. Depletion of activated hepatic stellate cell correlates with severe liver damage and abnormal liver regeneration in acetaminophen-induced liver injury. Acta Biochim Biophys Sin (Shanghai). 2011;43(4):307‒315.
  129. Weber LW, Boll M, Stampfl A. Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol. 2003;33(2):105‒136.
  130. Dabeva MD, Alpini G, Hurston E, et al. Models for hepatic progenitor cell activation. Proc Soc Exp Biol Med. 1993;204(3):242‒252.
  131. Pritchard MT, Nagy LE. Hepatic fibrosis is enhanced and accompanied by robust oval cell activation after chronic carbon tetrachloride administration to Egr-1-deficient mice. Am J Pathol. 2010;176(6):2743‒2752.
  132. Jeong WI, Jeong DH, Do SH, et al. Expression of cytokeratins 8 and 18 on Mallory body and oval cell in rats during hepatic fibrosis. In Vivo. 2005;19(4):769‒775.
  133. Suh H, Song MJ, Park YN. Behavior of isolated rat oval cells in porous collagen scaffold. Tissue Eng. 2003;9(3):411‒420.
  134. Huitfeldt HS, Brandtzaeg P, Poirier MC. Carcinogen-induced alterations in rat liver DNA adduct formation determined by computerized fluorescent image analysis. Lab Invest. 1991;64(2):207‒214.
  135. Solt D, Farber E. New principle for the analysis of chemical carcinogenesis. Nature. 1976;263:701‒703.
  136. Holme JA, Trygg B, Soderlund E. Species differences in the metabolism of 2-acetylaminofluorene by hepatocytes in primary monolayer culture. Cancer Res. 1986;46(4):1627‒1632.
  137. Hatch HM, Zheng D, Jorgensen ML, et al. SDF-1alpha/CXCR4: a mechanism for hepatic oval cell activation and bone marrow stem cell recruitment to the injured liver of rats. Cloning Stem Cells. 2002;4(4):339‒351.
  138. Li Z, Chen J, Li L, et al. Human hepatocyte growth factor (hHGF)-modified hepatic oval cells improve liver transplant survival. PLoS One . 2012;7(9):e44805..
  139. Evarts RP, Nakatsukasa H, Marsden ER, et al. Expression of transforming growth factor-alpha in regenerating liver and during hepatic differentiation. Mol Carcinog. 1992;5(1):25‒31.
  140. Leite AR, Correa-Giannella ML, Dagli ML, et al. Fibronectin and laminin induce expression of islet cell markers in hepatic oval cells in culture. Cell Tissue Res. 2007;327(3):529‒537.
  141. Yovchev MI, Dabeva MD, Oertel M. Isolation, characterization, and transplantation of adult liver progenitor cells. Methods Mol Biol. 2013;976:37‒51.
  142. Petersen BE, Zajac VF, Michalopoulos GK. Bile ductular damage induced by methylene dianiline inhibits oval cell activation. Am J Pathol. 1997;151(4):905‒909.
  143. He ZP, Tan WQ, Tang YF, et al. Activation, isolation, identification and in vitro proliferation of oval cells from adult rat livers. Cell Prolif. 2004;37(2):177‒187.
  144. Sakamoto S, Hirai H, Taga H, et al. Thymidine kinase and alpha-fetoprotein as biochemical markers of hepatocarcinogenesis induced by 3'-methyl-4-dimethylaminoazobenzene treatment in rats. Carcinogenesis. 1990;11(1):145‒150.
  145. Iwai M, Kashiwadani M, Takino T, et al. Demonstration by light and ultrastructural immunoperoxidase study of alpha-fetoprotein-positive non-hepatoma cells and hepatoma cells during 3'-methyl-4-dimethylaminoazobenzene hepatocarcinogenesis. Virchows Arch B Cell Pathol Incl Mol Pathol. 1988;55(2):117‒123.
  146. Rosenberg D, Ilic Z, Yin L, et al. Proliferation of hepatic lineage cells of normal C57BL and interleukin-6 knockout mice after cocaine-induced periportal injury. Hepatology. 2000;31(4):948‒955.
  147. Factor VM, Radaeva SA, Thorgeirsson SS. Origin and fate of oval cells in dipin-induced hepatocarcinogenesis in the mouse. Am J Pathol. 1994;145(2):409‒422.
  148. Braun KM, Sandgren EP. Cellular origin of regenerating parenchyma in a mouse model of severe hepatic injury. Am J Pathol. 2000;157(2):561‒569.
  149. Lemmer ER, de la Motte Hall P, Omori N, et al. Histopathology and gene expression changes in rat liver during feeding of fumonisin B1, a carcinogenic mycotoxin produced by Fusarium moniliforme. Carcinogenesis. 1999;20(5):817‒824.
  150. Lemmer ER, Vessey CJ, Gelderblom WC, et al. Fumonisin B1-induced hepatocellular and cholangiocellular tumors in male Fischer 344 rats: potentiating effects of 2-acetylaminofluorene on oval cell proliferation and neoplastic development in a discontinued feeding study. Carcinogenesis. 2004;25(7):1257‒1264 .
  151. Jung Y, Brown KD, Witek RP, et al. Accumulation of hedgehog-responsive progenitors parallels alcoholic liver disease severity in mice and humans. Gastroenterology. 2008;134(5):1532‒1543.
  152. Smith PG, Tee LB, Yeoh GC. Appearance of oval cells in the liver of rats after long-term exposure to ethanol. Hepatology. 1996;23(1):145‒154.
  153. Smith PG, Yeoh GC. Chronic iron overload in rats induces oval cells in the liver. Am J Pathol. 1996;149(2):389‒398.
  154. Gong P, Wang Y, Zhang J, et al. Differential hepatic stem cell proliferation and differentiation after partial hepatectomy in rats. Mol Med Rep. 2013;8(4):1005‒1010.
  155. Higgins GM, Anderson RM. Experimental pathology of the liver - restoration of the liver of the white rat following partial surgical removal. Arch Pathol. 1931;12:186‒202.
  156. Oe H, Kaido T, Mori A, et al. Hepatocyte growth factor as well as vascular endothelial growth factor gene induction effectively promotes liver regeneration after hepatectomy in Solt-Farber rats. Hepatogastroenterology. 2005;52(65):1393‒1397.
  157. Bisgaard HC, Holmskov U, Santoni-Rugiu E, et al. Heterogeneity of ductular reactions in adult rat and human liver revealed by novel expression of deleted in malignant brain tumor 1. Am J Pathol. 2002;161(4):1187‒1198.
  158. Jensen CH, Jauho EI, Santoni-Rugiu E, et al. Transit-amplifying ductular (oval) cells and their hepatocytic progeny are characterized by a novel and distinctive expression of delta-like protein/preadipocyte factor 1/fetal antigen 1. Am J Pathol. 2004;164(4):1347‒1359.
  159. Chang C, Xu C. Transcriptome atlas of glutamine family amino acid metabolism-related genes in eight regenerating liver cell types. Cell Biol Int. 2010;34(12):1189‒1198.
  160. Wang J, Clark JB, Rhee GS, et al. Proliferation and hepatic differentiation of adult-derived progenitor cells. Cells Tissues Organs. 2003;173(4):193‒203.
  161. Dezso K, Papp V, Bugyik E, et al. Structural analysis of oval-cell-mediated liver regeneration in rats. Hepatology. 2012;56(4):1457‒1467.
  162. Hu Z, Evarts RP, Fujio K, et al. Expression of hepatocyte growth factor and c-met genes during hepatic differentiation and liver development in the rat. Am J Pathol. 1993;142(6):1823‒1830.
  163. Arai M, Yokosuka O, Fukai K, et al. Gene expression profiles in liver regeneration with oval cell induction. Biochem Biophys Res Commun. 2004;317(2):370‒376.
  164. Paku S, Schnur J, Nagy P, et al. Origin and structural evolution of the early proliferating oval cells in rat liver. Am J Pathol. 2001;158(4):1313‒1323.
  165. Assimakopoulos SF, Tsamandas AC, Georgiou CD, et al. Bombesin and neurotensin exert antiproliferative effects on oval cells and augment the regenerative response of the cholestatic rat liver. Peptides. 2010;31(12):2294‒2303.
  166. Olynyk JK, Yeoh GC, Ramm GA, et al. Gadolinium chloride suppresses hepatic oval cell proliferation in rats with biliary obstruction. Am J Pathol. 1998;152(2):347‒352.
  167. Sakai H, Tagawa Y, Tamai M, et al. Isolation and characterization of portal branch ligation-stimulated Hmga2-positive bipotent hepatic progenitor cells. Biochem Biophys Res Commun. 2010;403(3‒4):298‒304.
  168. Ise N, Sato T, Yasui O, et al. Enhanced proliferation of hepatic progenitor cells in rats after portal branch occlusion. Liver Transpl. 2004;10(6):748‒754.
  169. Cassiman D, Libbrecht L, Sinelli N, et al. The vagal nerve stimulates activation of the hepatic progenitor cell compartment via muscarinic acetylcholine receptor type 3. Am J Pathol. 2002;161(2):521‒530.
  170. Oben JA, Roskams T, Yang S, et al. Sympathetic nervous system inhibition increases hepatic progenitors and reduces liver injury. Hepatology. 2003;38(3):664‒673.
  171. Fujimoto Y, Takahashi H, Dempo K, et al. Hereditary hepatitis in LEC rats: accumulation of abnormally high ploid nuclei. Cancer Detect Prev. 1989;14(2):235‒237.
  172. Irani AN, Malhi H, Slehria S, et al. Correction of liver disease following transplantation of normal rat hepatocytes into Long-Evans Cinnamon rats modeling Wilson's disease. Mol Ther. 2001;3(3):302‒309.
  173. Braun KM, Thompson AW, Sandgren EP. Hepatic microenvironment affects oval cell localization in albumin-urokinase-type plasminogen activator transgenic mice. Am J Pathol. 2003;162(1):195‒202.
  174. Bustos M, Sangro B, Alzuguren P, et al. Liver damage using suicide genes. A model for oval cell activation. Am J Pathol. 2000;157(2):549‒559.
  175. Shiota G, Kunisada T, Oyama K, et al. In vivo transfer of hepatocyte growth factor gene accelerates proliferation of hepatic oval cells in a 2-acetylaminofluorene/partial hepatectomy model in rats. FEBS Lett. 2000;470(3):325‒330.
  176. Warskulat U, Borsch E, Reinehr R, et al. Chronic liver disease is triggered by taurine transporter knockout in the mouse. FASEB J. 2006;20(3):574‒576.
  177. Lee KP, Lee JH, Kim TS, et al. The Hippo-Salvador pathway restrains hepatic oval cell proliferation, liver size, and liver tumorigenesis. Proc Natl Acad Sci U S A. 2010;107(18):8248‒8253.
  178. Diwan BA, Ward JM, Ramljak D, et al. Promotion by Helicobacter hepaticus-induced hepatitis of hepatic tumors initiated by N-nitrosodimethylamine in male A/JCr mice. Toxicol Pathol. 1997;25(6):597‒605.
  179. Ihrig M, Schrenzel MD, Fox JG. Differential susceptibility to hepatic inflammation and proliferation in AXB recombinant inbred mice chronically infected with Helicobacter hepaticus. Am J Pathol. 1999;155(2):571‒582.
  180. Bannasch P, Khoshkhou NI, Hacker HJ, et al. Synergistic hepatocarcinogenic effect of hepadnaviral infection and dietary aflatoxin B1 in woodchucks. Cancer Res. 1995;55(15):3318‒3330.
  181. Eichhorst ST, Krueger A, Muerkoster S, et al. Suramin inhibits death receptor-induced apoptosis in vitro and fulminant apoptotic liver damage in mice. Nat Med. 2004;10(6):602‒609.
  182. Yin L, Lynch D, Sell S. Participation of different cell types in the restitutive response of the rat liver to periportal injury induced by allyl alcohol. J Hepatol. 1999;31(3):497‒507.
  183. Ghoshal AK, Mullen B, Medline A, et al. Sequential analysis of hepatic carcinogenesis. Regeneration of liver after carbon tetrachloride-induced liver necrosis when hepatocyte proliferation is inhibited by 2-acetylaminofluorene. Lab Invest. 1983;48(2):224‒230.
  184. Lipsky MM, Hinton DE, Klaunig JE, et al. Biology of hepatocellular neoplasia in the mouse. I. Histogenesis of safrole-induced hepatocellular carcinoma. J Natl Cancer Inst. 1981;67(2):365‒376.
  185. Turner PV, Brabb T, Pekow C, et al. Administration of substances to laboratory animals: routes of administration and factors to consider. J Am Assoc Lab Anim Sci. 2011;50(5):600‒613.
  186. Digernes V, Iversen OH. Flow cytometry of nuclear DNA content in liver cirrhosis and liver tumours in rats exposed to acetylaminofluorene. Virchows Arch B Cell Pathol Incl Mol Pathol. 1984;47(2):139‒146.
  187. Fausto N. Oval cells and liver carcinogenesis: an analysis of cell lineages in hepatic tumors using oncogene transfection techniques. Prog Clin Biol Res. 1990;331:325‒334.
  188. Wu XZ, Yu XH. Bone marrow cells: the source of hepatocellular carcinoma? Med Hypotheses. 2007;69(1):36‒42.
  189. Sano T, Kagawa M, Okuno M, et al. Prevention of rat hepatocarcinogenesis by acyclic retinoid is accompanied by reduction in emergence of both TGF-alpha-expressing oval-like cells and activated hepatic stellate cells. Nutr Cancer. 2005;51(2):197‒206.
  190. Sell S, Leffert HL. Liver cancer stem cells. J Clin Oncol. s2008;26(17):2800‒2805.
  191. Tirnitz-Parker JE, Tonkin JN, Knight B, et al. Isolation, culture and immortalisation of hepatic oval cells from adult mice fed a choline-deficient, ethionine-supplemented diet. Int J Biochem Cell Biol. 2007;39(12):2226‒2239.
  192. Alison MR. Liver stem cells: implications for hepatocarcinogenesis. Stem Cell Rev. 2005;1(3):253‒260.
  193. Sanders JA, Brilliant KE, Clift D, et al. The inhibitory effect of rapamycin on the oval cell response and development of preneoplastic foci in the rat. Exp Mol Pathol. 2012;93(1):40‒49.
  194. Fan L, Xu C, Wang C, et al. Bmi1 is required for hepatic progenitor cell expansion and liver tumor development. PLoS One. 2012;7(9):e46472.
  195. Weng HL, Feng DC, Radaeva S, et al. IFN-gamma inhibits liver progenitor cell proliferation in HBV-infected patients and in 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet-fed mice. J Hepatol. 2013;59(4):738‒745.
  196. Bisgaard HC, Nagy P, Ton PT, et al. Modulation of keratin 14 and alpha-fetoprotein expression during hepatic oval cell proliferation and liver regeneration. J Cell Physiol. 1994;59(3):475‒484.
  197. Pichard V, Aubert D, Ferry N. Direct in vivo cell lineage analysis in the retrorsine and 2AAF models of liver injury after genetic labeling in adult and newborn rats. PLoS One . 2009;4(9):e7267.
  198. Radaeva S, Bannasch P. Changes in catalase and glucose-6-phosphatase distribution patterns within oval cell compartment as possible differentiation markers during viral hepatocarcinogenesis in woodchucks. Differentiation. 1996;60(3):169‒178.
Creative Commons Attribution License

©2015 Tan, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.