hepatocellular carcinoma associated with liver-gender ... · hepatocellular carcinoma (hcc) is a...

12
Hepatocellular Carcinoma Associated with Liver-Gender Disruption in Male Mice Arlin B. Rogers, 1,2 Elizabeth J. Theve, 1 Yan Feng, 1 Rebecca C. Fry, 2 Koli Taghizadeh, 2 Kristen M. Clapp, 1 Chakib Boussahmain, 1 Kathleen S. Cormier, 1 and James G. Fox 1,2 1 Division of Comparative Medicine and 2 Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts Abstract Hepatocellular carcinoma (HCC) is a male-predominant cancer associated with chronic hepatitis. Like human viral hepatitis, murine Helicobacter hepaticus infection produces inflammation and HCC with a masculine bias. We used this model to identify potential mechanisms of male HCC predisposition. Male weanling A/JCr mice (n = 67) were gavaged with H. hepaticus or vehicle. At 1 year, mice were distributed into four groups: surgical castration, chemical castration, castration followed by dihydrotestosterone sup- plementation, or sexually intact controls. Responses to infection were compared with IFN-; challenge alone. At 21 months, there was no significant difference in hepatitis between groups. Neither castration nor androgen receptor agonism altered tumor incidence. Infected mice with severe, but not mild, disease exhibited a mosaic of alterations to sexually dimorphic genes and microsomal long-chain fatty acids. By microarray, tumorigenic hepatitis was strongly associated with liver-gender disruption, defined as the loss of a gender-identifying hepatic molecular signature. IFN-; alone produced similar changes, demonstrating a role for proinflammatory cytokines in this process. In conclusion, hepatocarcinogenesis in male mice with chronic hepatitis is maturationally imprinted and androgen-independent. Proinflammatory cytokines may promote HCC in a male- predominant fashion due to high sensitivity of the masculin- ized liver to loss of sex-specific transcriptional balance. Liver-gender disruption has pleiotropic implications for hepatic enzyme activity, lipid processing, nuclear receptor activation, apoptosis, and proliferation. We propose a multistep model linking chronic hepatitis to liver cancer through cytokine-mediated derangement of gender-specific cellular metabolism. This model introduces a novel mecha- nism of inflammation-associated carcinogenesis consistent with male-predominant HCC risk. [Cancer Res 2007;67(24): 11536–46] Introduction Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing condition, but other significant risks include alcoholism, aflatoxin B1 ingestion, fatty liver disease, and inborn errors of metabolism (2). Cirrhosis often, but not always, precedes cancer. Even after controlling for known risks, the incidence of HCC in men is more than twice that of women (1). To date, no cause for this gender discrepancy in humans has been identified. The liver is a sexually dimorphic organ. Gender differences exist with regard to gene expression, mitochondrial function, microsomal enzyme activity, membrane lipid composition, and immune responses (3–6). Liver-gender differentiation commences early in development, but the greatest divergence occurs at puberty (7). In males, testosterone is aromatized within the central nervous system to initiate an endocrine cascade resulting in periodic release of growth hormone from the anterior pituitary (8). Hepatocyte growth hormone receptors transduce pulsed hormone through a tyrosine phosphorylation cascade involving Jak2 and Stat5b. Activated Stat5b migrates to the nucleus and, in conjunction with recognized and unrecognized cofactors, enhances transcription of masculine genes while repressing feminine genes (9). In females, growth hormone is secreted at lower but more continuous levels from the pituitary, Stat5b remains unphosphorylated in the hepatocyte cytoplasm, and the default program of feminine transcription is maintained (7). Pathways other than growth hormone/Stat5b also contribute to liver-gender differentiation, although these are not well defined (10). Sexual dimorphism of the liver has significant metabolic consequences. Xenobiotic clearance of a wide variety of drugs and toxins differs between women and men due, in part, to gender-dimorphic expression of enzymes, such as cytochrome P450 3A4 (3, 11, 12). Rodents also exhibit pronounced liver-gender dimorphism (13, 14). In spite of the strong influence of gender on liver cancer risk, therapeutic trials targeting sex hormone pathways have proved disappointing. Preliminary clinical results demonstrating a modest benefit with tamoxifen and cyproterone acetate were not replicated in expanded multicenter trials (15). Furthermore, no large placebo-controlled study has shown an improvement in tumor size or patient survival using the competitive androgen receptor antagonist flutamide, leuteinizing hormone–releasing hormone antagonist leuprorelin, or progestin megestrol acetate (16–18). Notably, the above cited trials enrolled only patients with inoperable tumors. Antiandrogen therapy as a chemopro- phylactic strategy to delay or prevent HCC in high-risk patients has not been reported. Like human viral hepatitis, Helicobacter hepaticus infection of A/JCr mice produces chronic inflammation and HCC with a male bias (19, 20). Tumors typically arise after 18 months. At 1 year of age, susceptible males exhibit chronic hepatitis, but not HCC (21). This time point is analogous to the chronic but premalignant stage of human viral hepatitis, when infection frequently is first recognized. We undertook the present study to Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Requests for reprints: Arlin Rogers, Division of Comparative Medicine 16-849, Massachusetts Institute of Technology, Cambridge, MA 02139. Phone: 617-253-9442; Fax: 617-252-1882; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-07-1479 Cancer Res 2007; 67: (24). December 15, 2007 11536 www.aacrjournals.org Research Article Research. on January 26, 2021. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 26-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

Hepatocellular Carcinoma Associated with Liver-Gender

Disruption in Male Mice

Arlin B. Rogers,1,2Elizabeth J. Theve,

1Yan Feng,

1Rebecca C. Fry,

2Koli Taghizadeh,

2

Kristen M. Clapp,1Chakib Boussahmain,

1Kathleen S. Cormier,

1and James G. Fox

1,2

1Division of Comparative Medicine and 2Center for Environmental Health Sciences, Massachusetts Institute of Technology,Cambridge, Massachusetts

Abstract

Hepatocellular carcinoma (HCC) is a male-predominantcancer associated with chronic hepatitis. Like human viralhepatitis, murine Helicobacter hepaticus infection producesinflammation and HCC with a masculine bias. We used thismodel to identify potential mechanisms of male HCCpredisposition. Male weanling A/JCr mice (n = 67) weregavaged with H. hepaticus or vehicle. At 1 year, mice weredistributed into four groups: surgical castration, chemicalcastration, castration followed by dihydrotestosterone sup-plementation, or sexually intact controls. Responses toinfection were compared with IFN-; challenge alone. At21 months, there was no significant difference in hepatitisbetween groups. Neither castration nor androgen receptoragonism altered tumor incidence. Infected mice with severe,but not mild, disease exhibited a mosaic of alterations tosexually dimorphic genes and microsomal long-chain fattyacids. By microarray, tumorigenic hepatitis was stronglyassociated with liver-gender disruption, defined as the lossof a gender-identifying hepatic molecular signature. IFN-;alone produced similar changes, demonstrating a role forproinflammatory cytokines in this process. In conclusion,hepatocarcinogenesis in male mice with chronic hepatitisis maturationally imprinted and androgen-independent.Proinflammatory cytokines may promote HCC in a male-predominant fashion due to high sensitivity of the masculin-ized liver to loss of sex-specific transcriptional balance.Liver-gender disruption has pleiotropic implications forhepatic enzyme activity, lipid processing, nuclear receptoractivation, apoptosis, and proliferation. We propose amultistep model linking chronic hepatitis to liver cancerthrough cytokine-mediated derangement of gender-specificcellular metabolism. This model introduces a novel mecha-nism of inflammation-associated carcinogenesis consistentwith male-predominant HCC risk. [Cancer Res 2007;67(24):11536–46]

Introduction

Hepatocellular carcinoma (HCC) is a male-predominant cancer(1). Chronic viral hepatitis is the most common predisposingcondition, but other significant risks include alcoholism, aflatoxin

B1 ingestion, fatty liver disease, and inborn errors of metabolism(2). Cirrhosis often, but not always, precedes cancer. Even aftercontrolling for known risks, the incidence of HCC in men is morethan twice that of women (1). To date, no cause for this genderdiscrepancy in humans has been identified.

The liver is a sexually dimorphic organ. Gender differencesexist with regard to gene expression, mitochondrial function,microsomal enzyme activity, membrane lipid composition, andimmune responses (3–6). Liver-gender differentiation commencesearly in development, but the greatest divergence occurs atpuberty (7). In males, testosterone is aromatized within thecentral nervous system to initiate an endocrine cascade resultingin periodic release of growth hormone from the anteriorpituitary (8). Hepatocyte growth hormone receptors transducepulsed hormone through a tyrosine phosphorylation cascadeinvolving Jak2 and Stat5b. Activated Stat5b migrates to thenucleus and, in conjunction with recognized and unrecognizedcofactors, enhances transcription of masculine genes whilerepressing feminine genes (9). In females, growth hormone issecreted at lower but more continuous levels from the pituitary,Stat5b remains unphosphorylated in the hepatocyte cytoplasm,and the default program of feminine transcription is maintained(7). Pathways other than growth hormone/Stat5b also contributeto liver-gender differentiation, although these are not welldefined (10). Sexual dimorphism of the liver has significantmetabolic consequences. Xenobiotic clearance of a wide varietyof drugs and toxins differs between women and men due, inpart, to gender-dimorphic expression of enzymes, such ascytochrome P450 3A4 (3, 11, 12). Rodents also exhibit pronouncedliver-gender dimorphism (13, 14).

In spite of the strong influence of gender on liver cancer risk,therapeutic trials targeting sex hormone pathways have proveddisappointing. Preliminary clinical results demonstrating amodest benefit with tamoxifen and cyproterone acetate werenot replicated in expanded multicenter trials (15). Furthermore,no large placebo-controlled study has shown an improvement intumor size or patient survival using the competitive androgenreceptor antagonist flutamide, leuteinizing hormone–releasinghormone antagonist leuprorelin, or progestin megestrol acetate(16–18). Notably, the above cited trials enrolled only patientswith inoperable tumors. Antiandrogen therapy as a chemopro-phylactic strategy to delay or prevent HCC in high-risk patientshas not been reported.

Like human viral hepatitis, Helicobacter hepaticus infection ofA/JCr mice produces chronic inflammation and HCC with amale bias (19, 20). Tumors typically arise after 18 months. At 1year of age, susceptible males exhibit chronic hepatitis, but notHCC (21). This time point is analogous to the chronic butpremalignant stage of human viral hepatitis, when infectionfrequently is first recognized. We undertook the present study to

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).

Requests for reprints: Arlin Rogers, Division of Comparative Medicine 16-849,Massachusetts Institute of Technology, Cambridge, MA 02139. Phone: 617-253-9442;Fax: 617-252-1882; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-07-1479

Cancer Res 2007; 67: (24). December 15, 2007 11536 www.aacrjournals.org

Research Article

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

test the hypothesis that castration would slow progression toHCC in mature males with chronic hepatitis and, conversely,that androgen receptor agonism would accelerate tumorigenesis.Unexpectedly, we found that androgens played no direct role intumor promotion. Instead, we identified a strong associationbetween tumorigenic hepatitis, proinflammatory cytokines, andliver-gender disruption, defined as the loss of a gender-identifying hepatic molecular signature. These observations formthe basis of new model of inflammation-associated carcinogen-esis that is androgen-independent yet consistent with male-predominant expression of HCC.

Materials and Methods

Study design. Sixty-seven Helicobacter -free male A/JCr weanling mice

were purchased from the National Cancer Institute. Mice were gavaged with

H. hepaticus 3B1 (ATCC 51449; n = 43) or broth only (n = 24), and infectionwas confirmed by fecal PCR as described previously (21–23). At 1 year, livers

from five infected intact mice were evaluated by histopathology. The

remaining 62 were distributed into four groups: (a) surgical castration

(n = 14), (b) ‘‘chemical castration’’ with the gonadotropin-releasinghormone antagonist abarelix depot (Plenaxis, 50 Ag/g body weight, i.p.

once monthly; n = 14), (c) surgical castration followed by dihydrotestoster-

one supplementation (5 mg of 90-day-release hormone pellets, InnovativeResearch of America; implanted s.c. over alternating shoulders every

3 months; n = 13), or (d) sexually intact controls (n = 21). In a separate

experiment, Helicobacter -free male and female A/J mice (n = 10 each) were

implanted s.c. with a 7-day Alzet osmotic minipump (Durect Corporation)containing 20 Ag recombinant murine IFN-g (R&D Systems) or vehicle as

described elsewhere (24). Mouse husbandry conditions have been described

(21). Protocols were compliant with the USPHS Policy on Humane Care and

Use of Laboratory Animals and approved by the Massachusetts Institute ofTechnology Committee on Animal Care.

Histopathology. To minimize the effect of circadian variation, mouse

necropsies were performed between 9:00 and 11:30 am. After euthanasia by

CO2 inhalation, mouse serum was processed and tissues were collected and

evaluated by histopathology as described (21). Liver sections were scored by

a comparative pathologist blinded to sample identity. A hepatitis index was

generated by combining scores for lobular, portal, and interface hepatitis

along with the number of lobes (of four) containing five or more

inflammatory lesions (Table 1). Mice with a histologic hepatitis index of

z4 were defined as having hepatitis. Preneoplastic and neoplastic lesion

criteria were adapted from the National Toxicology Program (25).

Intralobular foci of cellular alteration were assigned a score of 1, and

translobular foci were assigned a score of 2. Foci of cellular alteration are

useful as precancerous disease markers (26). Adenomas received a score

of 3, and HCC a score of 4. Mice meeting histologic criteria of hepatitis

(inflammation index, z4) and neoplasia (grade, z3) were defined as having

tumorigenic hepatitis. Photomicrographs were obtained using a BH-2

microscope (Olympus America) and DXM1200 camera (Nikon Instruments).

White color balance and size bars were applied using Photoshop CS2

software (Adobe Systems). Kruskall-Wallis one-way ANOVA and Mann-

Whitney U nonparametric statistics were performed using Prism software

(GraphPad) with significance set at P < 0.05 as described previously (21).Quantitative reverse transcription–PCR. Liver RNA was isolated and

assessed for quality as described previously (27). Quantitative reversetranscription–PCR (qRT-PCR) was performed with Taqman assay-on-demand or SYBR green reagents (Applied Biosystems). Samples were runin duplicate, and dissociation curves were performed for SYBR green assaysto confirm specificity. Genes were selected based on previous diseaseassociations in intact male mice documented by us and others (21, 28).Original primer sequences were designed using MacVector 7.2.3 software(MacVector; Supplementary Table S1). To determine the effect of castrationF dihydrotestosterone supplementation on basal gene expression, fold-change values from uninfected androgen-interrupted groups were com-pared against uninfected sexually intact controls. Parametric ANOVA andStudent’s t test were performed similar to the nonparametric assays in theprevious section. For the IFN-g experiment, differential gene expression wasdetermined for gender by comparing vehicle-treated male versus female

mice and for cytokine effect by comparing IFN-g–supplemented versusvehicle-only treated mice of the same sex.

Table 1. Histopathologic scoring criteria used to determine hepatitis index

Criterion 1 2 3 4

Lobular hepatitis Small, randomly scatteredlobular inflammatory

foci

Moderate multifocalinflammation

with areas of

coagulative

hepatocellularnecrosis

Moderate-severe coalescingfoci of inflammation and

coagulative necrosis

Translobular inflammationand coagulative necrosis

with significant hepatocyte

loss

Portal hepatitis Small foci of leukocytes

(lymphocyte-predominant)

in scattered portal triads

Moderately large,

portal-restricted,

well-circumscribedmononuclear

cell-rich aggregates

Large lymphocyte-predominant

aggregates forming follicle-like

structures

Portal-portal bridging,

well-developed follicles

Interfacehepatitis

Small foci of indistinctlybounded portal/periportal

inflammation extending

through limiting plate

Multifocal inflammatoryfoci extending through

limiting plate and beyond

first two rows of periportal

hepatocytes

Moderate-severe inflammatoryfoci traversing limiting

plates; often accompanied

by oval cell hyperplasia

Severe, poorly circumscribedbridging inflammation with

hepatocyte loss and

widespread oval cell

hyperplasiaHepatitis index

(0–16)

Sum of lobular, portal, and

interface hepatitis scores

plus number of liver lobes

(of four) with five ormore inflammatory foci;

hepatitis defined by

histologic index of z4

Liver-Gender Disruption and HCC

www.aacrjournals.org 11537 Cancer Res 2007; 67: (24). December 15, 2007

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

Microarray. RNA samples from two sham-inoculated sexually intactmice, surgically castrated mice, and H. hepaticus–infected surgically

castrated mice with hepatitis and HCC were submitted for microarray

using the GeneChip Mouse Genome 430 2.0 Array (Affymetrix) as described

(27). Mice for these analyses were collected in a narrow time windowmidmorning and were of identical age (21 months) but were not litter-

matched or fasted. Mice selected for microarray had no identified

extrahepatic lesions (e.g., dermatitis). Nonexpressed transcripts on

microarray were filtered with GeneChip Operating Software algorithms(Affymetrix). Gene expression differences of z1.5-fold with P < 0.05 were

considered significant. For castration and tumor-associated directional

analysis, results were normalized by Robust Multichip Average (29). Protein-

protein interactions were mapped with Ingenuity Pathways Analysis(Ingenuity Systems). Microarray data were submitted to the Gene

Expression Omnibus Database (Series GSE6632).

Immunohistochemistry and ELISA. Immunohistochemistry was per-formed for h-catenin (1:100; Lab Vision), interleukin-6 (IL-6; 1:600; Abcam),

Ki-67 (1:50; BD Biosciences), and nuclear factor-nB (NF-nB; p65 subunit,

1:100; Invitrogen) as described (30). The ARK kit (DAKO) was used for

mouse-on-mouse applications. Species, isotype, and concentration-matchedirrelevant primary antibody controls were included in all immunohisto-

chemistry assays. Ki-67 (proliferation) labeling indices were determined

by counting the number of positive hepatocyte nuclei per 10 high-power

(40� ocular objective) fields. Serum dihydrotestosterone (Alpha DiagnosticInternational) and growth hormone (Diagnostic Systems Laboratories)

were measured by ELISA according to manufacturer protocols. Serum from

all mice was collected at necropsy between 9:00 and 11:30 am. All mice wereevaluated for serum growth hormone along with three Helicobacter-free

female control mice from an unrelated study. Intact and surgically castrated

dihydrotestosterone-supplemented mice were assayed for dihydrotestoster-

one, along with two uninfected and two infected mice from the surgicaland chemical castration groups. Parametric statistics were performed as

described above.

Microsomal long-chain fatty acid analysis. Long-chain fatty acid

(LCFA) methyl esters from enriched liver microsome fractions isolatedwith the NE-PER kit (Pierce Biotechnology) were extracted by the Bligh-

Dyer method and derivatized with 14% borontrifluoride-methanol

(Sigma) based on a published protocol (5). Specific LCFAs were identifiedand quantified using a commercial gas chromatography/mass spectrom-

etry system composed of a 6890N gas chromatograph and 5973N mass

selective detector (Agilent Technologies) operated in positive chemical

ionization mode with methane as reagent gas. Samples were derivedfrom five intact infected and five intact uninfected males along with

five uninfected adult female mice from an unrelated study. Retention

times for the six major microsomal LCFA species (see Results) were

determined with commercial standards (Sigma). Data were processedwith ChemStation software (Agilent) and analyzed statistically as

described above.

Results

Neither castration nor androgen receptor agonism alteredHCC incidence. In agreement with prior experience, H.hepaticus–infected male A/JCr mice at 1 year of age exhibitedchronic hepatitis but not HCC (21). This confirmed that the H.hepaticus inoculum was virulent and that androgen interventionswere instituted before malignancy. At 21 months, we confirmedby histopathology of androgen-sensitive salivary glands, renalglomeruli, and accessory sex glands (31) that the hormonalinterventions had the intended physiologic effect. Histopatho-logic evaluation of liver samples from sham-inoculated miceshowed no significant disease, whereas males in all infectedcohorts developed characteristic lesions of chronic activehepatitis (Fig. 1A). Most mice with hepatitis also had areas ofhepatocellular atypia ranging in progressive severity from

intralobular and translobular foci of cellular alteration toadenoma and carcinoma (Fig. 1A ; ref. 25). There was nosignificant difference in hepatitis or tumor incidence betweengroups (Fig. 1B). Among mice with hepatitis, intact malesexhibited a higher mean inflammation index than all castratedgroups, but the difference was not statistically significant (Fig.1B). Because there was no difference in HCC incidence betweengroups, we sought an androgen-independent mechanism oftumor promotion in mature male mice with preestablishedhepatitis.Castration and disease-associated gene alterations by qRT-

PCR. We previously identified genes associated with preneoplastichepatitis in H. hepaticus–infected mice (27). Here, we extendedthose evaluations to mice with HCC and determined the effect ofandrogenic intervention on gene expression. Our qRT-PCR panelincluded genes known to be expressed in a sexually dimorphicfashion, including the female-predominant genes Cyp2a4 andCyp4a14 and the male-predominant genes Cyp2d9, Cyp4a12,epidermal growth factor receptor, hydroxysteroid dehydrogenase3h-V, insulin-like growth factor binding protein-2, and trefoilfactor-3 (intestinal; ref. 14). As expected, feminine genes were up-regulated by surgical and chemical castration in uninfected mice,and masculine genes were down-regulated (Fig. 2A). Furthermore,with the exception of Cyp4a14, none of these changes wasreversed by the powerful androgen receptor agonist dihydrotes-tosterone (Fig. 2A). Liver masculinization is regulated predomi-nantly by indirect androgen effects and not by androgen receptorligand binding, as our findings confirm. H. hepaticus–infectedmice with hepatitis regardless of group showed statisticallysignificant up-regulation of H19 and trefoil factor-3 (Fig. 2B). Alldiseased mice except those supplemented with dihydrotestoster-one also exhibited significantly elevated tumor necrosis factor-a(TNF-a) and ubiquitin D (Fig. 2B). Sexually intact but notcastrated groups exhibited disease-associated down-regulation ofCyp2a4, epidermal growth factor receptor, hydroxysteroid dehy-drogenase 3h-V, insulin-like growth factor binding protein-2, andup-regulated lipocalin 2 (not shown). Cyp4a14 was down-regulated by disease in both castrated and intact mice, but onlyreached significance in the latter group. An important confound-ing variable on hepatic gene expression was ulcerative dermatitis,a common aging lesion of A/JCr and other mice (31). In ourpanel, ulcerative dermatitis independently down-regulated all fourCyp genes, as well as Igfbp2 (not shown). Therefore, mice withdermatitis were removed from comparative analyses for thosegenes. Mice with dermatitis were not selected for microarrayanalysis (below).Tumor-associated liver-gender disruption revealed by

microarray. Because no clear pattern of gene alterations emergedfrom the qRT-PCR panel, we performed microarray. First, weassessed the effect of castration alone on liver gene expression.Of 45,037 probesets representing f39,000 unique transcripts,16,074 were called present in sexually intact mice by GeneChipOperating Software (Supplementary Table S2). Therefore, f36% ofthe murine genome were expressed in the adult male liver. Inuninfected castrated mice, we identified 2,123 gene alterations(1,413 up-regulated and 710 down-regulated) compared withuninfected sexually intact controls. Thus, f13% of all liver geneswere castration sensitive. Most of these genes have been shownelsewhere to be gender dimorphic and/or associated with failedhepatic masculinization due to impaired growth hormone signaling(e.g., Ames dwarf, lit/lit, and Stat5b�/� mice; refs. 14, 32). Therefore,

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11538 www.aacrjournals.org

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

Figure 1. Histopathology and immunohistochemistry of H. hepaticus–induced hepatitis and tumorigenesis in mature male androgen-modulated mice.A, histopathologic features of H. hepaticus–induced chronic active hepatitis and HCC. a, normal liver from sham-inoculated control. b, lobular hepatitis with randommixed inflammation surrounding and traversing central focus of coagulative necrosis. c, marked portal hepatitis with lympoid follicle-like structures; note sharpdelineation along hepatic limiting plate. d, interface hepatitis featuring portocentric inflammation extending into adjacent lobule with disregard for limiting plate boundary.e, normal liver. f, clear cell focus of cellular alteration; note absence distinct border at margin (arrowheads ). g, adenoma; note distinct margin with slight compression ofoverlying hepatocytes (arrowheads ). h, mixed cell HCC including basophilic-tigroid hepatocytes with subplasmalemmal radial striations (arrowheads ). B, hepatitisincidence, inflammation severity (hepatitis index), and tumor incidence summary demonstrating no significant difference in disease outcome between surgically andchemically castrated mice with or without supplementation of the powerful androgen receptor agonist dihydrotestosterone when compared with sexually intact controls.C, immunohistochemistry. a, noKi-67+ proliferating hepatocytes in normalmouse liver. b, marked cellular proliferation (intranuclear Ki-67 expression; arrowheads ) inHCC.HCC. c, in healthy mouse liver, note presence of circulating IL-6 in blood vessels (arrow) and along sinusoidal surfaces; occasional random hepatocytes are alsopositive (arrowhead). d, increased IL-6+ hepatocytes in regions of fatty degeneration within HCC. e, intranuclear NF-nB (p65) localization in mononuclear inflammatorycells (arrow ). f, in healthy mouse liver, h-catenin localized to hepatocyte surface membranes; in contrast, biliary epithelium (arrow ) shows h-catenin along the cellsurface and diffusely throughout cytoplasm. g, no nuclear translocation of h-catenin in neoplastic hepatocytes. h, proliferating oval cells (arrows ) in tumor show thebiliary pattern of surface and cytoplasmic h-catenin staining. A, H&E; bar, 80 Amol/L (a, b, d ), 160 Amol/L (c ), and 40 Amol/L (e–h ). C, diaminobenzidine withhematoxylin counterstain; bar, 40 Amol/L (a–d, f–h ) and 10 Amol/L (e).

Liver-Gender Disruption and HCC

www.aacrjournals.org 11539 Cancer Res 2007; 67: (24). December 15, 2007

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

we referred to genes up-regulated by castration as feminine andthose down-regulated as masculine. In agreement with the knowndimorphism of the murine immune system (6), many of thecastration-sensitive genes in our panel were attributable to residentleukocyte populations, including Kuppfer cells and natural killerT cells (Supplementary Table S2).

Next, we assessed genes associated with tumorigenic hepatitisin castrated mice. Compared with uninfected surgically castratedcontrols, H. hepaticus–infected castrated males displayed alter-ations to 2,848 genes (1,856 up-regulated and 992 down-regulated). Of these, 829 were shown in the previous analysisto be castration sensitive (i.e., gender-dimorphic). Therefore, 29%of disease-associated genes were also castration sensitive (P <0.0001), demonstrating a highly significant bias for gender-dimorphic gene involvement in tumorigenic hepatitis. Many ofthe castration-sensitive genes altered in diseased mice wereassociated with inflammation, suggesting that dysregulation ofboth gender-specific immunity and hepatocellular functioncontributed to global liver-gender disruption (Table 2). Toprecisely characterize the direction change of gender-dimorphicgenes, we used conservative Robust Multichip Average normal-ization criteria (29). By this method, 379 genes met the statisticalcutoff for castration sensitivity (276 up-regulated and 103 down-regulated; Supplementary Table S3). Diseased mice exhibited 80up-regulated masculine genes, 163 up-regulated feminine genes,23 down-regulated masculine genes, and 113 down-regulatedfeminine genes (Supplementary Fig. S1). Gene ontology analysisand functional network mapping highlighted the central role ofproinflammatory cytokine pathways in the altered expression ofgender-dimorphic genes. Especially noteworthy were pathwayscentered on Stat1, TNF-a, IFN-g, and RelA (NF-nB subunit p65;Supplementary Fig. S1). Because of the insensitivity of themicroarray for detecting IFN-g (33), we performed qRT-PCR forthis gene. IFN-g was up-regulated >8-fold in diseased versusuninfected mice (P < 0.001, data not shown). Because we usedwhole liver slices, we were not able to distinguish hepatocytefrom nonhepatocyte contributions to total gene expression. Themurine immune system also exhibits sexual dimorphism (6), andit is likely that castration-sensitive inflammatory response genesinfluenced disease outcomes. Taken together, gene expressionanalyses showed that gender-dimorphic gene alterations weresignificantly overrepresented in mice with tumorigenic hepatitisand that proinflammatory cytokines were centrally involved inpathogenesis.Microsomal LCFAs reflected gender and disease status. LCFA

composition in healthy hepatocyte microsomes (endoplasmicreticulum) is gender specific. In agreement with studies in the rat(5), male mice in our study exhibited a higher proportion than age-matched females of palmitic acid (C16:0) as a fraction of the totalLCFA pool (Fig. 3A). Females had a higher proportion of stearic(C18:0) and arachidonic acid (C20:4), although only the latterachieved statistical significance. This is consistent with female-predominant expression of LCFA elongase (Elovl6), which wasincreased nearly 2-fold by castration in our study (SupplementaryTable S2). Although we had insufficient samples for statisticalanalysis, castrated male mice exhibited an intermediate phenotype(not shown). In H. hepaticus–infected males with hepatitis, therewas significantly decreased stearic (C18:0) and linoleic acid (C18:2)and increased oleic acid (C18:1). Accumulation of the monounsat-urated fatty acid oleate correlated with a 5-fold increase inexpression of stearoyl-CoA desaturase 2 (Supplementary Table S2).

It is possible that inflammatory cells contributed to altered fattyacid profiles, although we previously showed by morphometricanalysis that leukocytes account for a relatively small percentage ofliver area in mice with H. hepaticus–induced hepatitis (21). Takentogether, assessment of microsomal LCFA confirmed that genderand disease-associated differences in gene expression had func-tional metabolic consequences. Further work will be required todetermine the clinical relevance of fatty acid composition in thisand other subcellular compartments.Cell proliferation and disease markers in situ. As expected,

nuclear labeling of the cell proliferation marker Ki-67 was muchhigher in tumors than in normal liver (Fig. 1C). Mice in allandrogen-interrupted groups exhibited a modest but statisticallysignificant increase in basal hepatocyte proliferation versussexually intact uninfected controls (mean 2.1 Ki-67+ hepatocytenuclei versus 0.2 per 10 high-power fields, respectively). However,there was no difference in HCC incidence or Ki-67 labeling indexin neoplasms derived from different mouse groups (not shown),demonstrating that basal hepatocyte replication rate did notcorrelate with tumor risk. In healthy uninfected mice, IL-6 wasidentified in circulating form in blood vessels and alongsinusoidal linings and intracytoplasmically in randomly scatteredhepatocytes. In H. hepaticus–infected mice, IL-6 was expressed inleukocytes and was detected with increased frequency indysplastic and neoplastic hepatocytes in regions of fattydegeneration (Fig. 1C). As detected by immunohistochemistry,NF-nB (p65 subunit) was expressed primarily by inflammatorycells (Fig. 1C). Because microarray results implicated disease-associated genes in the Wnt/h-catenin signaling family, includingWnt-5a protein, cadherin 1 (E-cadherin), a-catenin, and cateninsrc (Supplementary Table S2), we performed h-catenin immu-nohistochemistry on representative tissues. However, unlike thenuclear translocation seen in colon and other cancers (34), inboth normal and cancerous liver, h-catenin was localizedexclusively to hepatocellular surface membranes. In contrast,bile duct epithelium and proliferating oval cells showed diffusecytoplasmic, as well surface-anchored, protein (Fig. 1C). Fromthis, we concluded that increased Wnt/h-catenin pathway geneproducts likely reflected biliary and oval cell hyperplasia.Regardless of cell type, nuclear signal was rare. Therefore,H. hepaticus–induced neoplasms would seem to fall into theh-catenin–negative category of HCC subtypes in agreement withtheir relatively slow-growing behavior (35).Growth hormone correlated poorly with masculinization in

aged males. Most liver masculinization occurs after pulsatilegrowth hormone induction at puberty (7). We sought to determinewhether the panhepatic feminization in mice castrated at 1 yearwas associated with loss of growth hormone pulsatility. Unexpect-edly, all male groups regardless of androgenic intervention showeda widely dispersed serum growth hormone distribution patternconsistent with pulsatile secretion (Fig. 3B). By microarray weobserved a 1.5-fold increase in Jak2 mRNA expression in mice withHCC but no difference in Stat5b (Supplementary Table S2).However, for these proteins it is the phosphorylation state thatgoverns in vivo activity (7). We confirmed in a separate assay thatcirculating dihydrotestosterone was increased in hormone-supple-mented mice (Fig. 3C). Because high levels of dihydrotestosteronedid not masculinize gene expression (Fig. 2A), testosterone, but notgrowth hormone or androgen receptor signaling, seemed necessaryto maintain male liver phenotype in this cohort of aged mice.Therefore, mechanisms that maintain masculine liver phenotype in

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11540 www.aacrjournals.org

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

Figure 2. Liver gene expression changes associated with castration, H. hepaticus–induced hepatitis and recombinant murine IFN-g challenge. A, surgical andchemical castration resulted in up-regulated transcription of feminine liver genes and down-regulation of masculine genes. None of these were reversed by the powerfulandrogen receptor agonist dihydrotestosterone except Cyp4a14, highlighting the absence of a significant role for androgen receptor signaling in liver masculinization.B, in our qRT-PCR panel, two genes (H19, Tff3) were significantly up-regulated in all groups of H. hepaticus–infected mice with hepatitis F tumors compared withgroup-matched uninfected controls. Two additional genes (Tnfa, Ubd) were significantly up-regulated in all groups except castrated mice supplemented withdihydrotestosterone.

Liver-Gender Disruption and HCC

www.aacrjournals.org 11541 Cancer Res 2007; 67: (24). December 15, 2007

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

mature mice may differ from those that induce gender differen-tiation at puberty. These results highlight the complexity ofendocrine regulation of liver-gender determination and show thatfactors in addition to growth hormone periodicity should be

considered when evaluating sex-specific hepatic transcriptionalregulation.IFN-; alone recapitulatedH. hepaticus–associated transcrip-

tional changes. To assess the potential role of proinflammatory

Table 2. Liver-gender disruption in mice with chronic hepatitis and HCC highlighted by mosaic of disease-associatedalterations to castration–up-regulated (feminine) and down-regulated (masculine) genes

Castration up (fem), hepatitis/HCC up DFold Castration down (masc), hepatitis/HCC up DFold

CD14 antigen 12.0 Discoidin domain receptor family, member 1 21.2

Caspase 12 10.8 Cadherin 1 16.4

IFN-induced protein 2; Ifit2 10.8 Serine protease inhibitor 12 14.7

Small inducible cytokine A5 (CCL5) 7.7 Lipoprotein lipase 9.0Cytochrome b-245, h polypeptide 6.4 Atrazine-specific IgG2b heavy chain variable 5.2

Chemokine (C-X-C motif) ligand 10 6.0 Immunoglobulin heavy chain 1 (serum IgG2a) 4.5

Calmodulin-related protein 5.5 CD8 antigen, a chain 4.3

T-cell specific GTPase 4.9 Homologue to LYVE-1 3.6C-type lectin, superfamily member 12 4.5 Coagulation factor II (thrombin) receptor 3.5

Tropomyosin 2, h 4.4 TNF-a–induced protein 2 3.3

Castration up (fem), hepatitis/HCC down DFold Castration down (masc), hepatitis/HCC down DFold

Sphingosine kinase 2 �14.5 Ribosomal protein S4, Y-linked 2 �6.3D site albumin promoter binding protein �14.1 Mouse major urinary protein 3 �5.7

Major facilitator superfamily domain containing 2 �5.9 Major urinary proteins 11 and 8 �4.2

Period homologue 2 �5.3 Major urinary protein 1 �3.0

Dopachrome tautomerase �4.6 Alanine-glyoxylate aminotransferase 2-like 1 �2.9Nocturnin �4.6 Cytochrome P450, 7b1 �2.5

Rigui �4.5 Amiloride-sensitive sodium channel �2.3

Thyroid hormone responsive SPOT14 homologue �3.8 NADPH oxidase 4 �2.0

Period 2 �3.8 Kruppel-like factor 9 �1.9Hepcidin antimicrobial peptide �3.7 Guanine nucleotide binding protein, a 14 �1.8

NOTE: Note significant representation of castration-sensitive inflammation-associated genes. Fold-changes shown are mean of H. hepaticus–infected

surgically castrated males versus sham-inoculated castrated males. The top 10 characterized genes in each category are listed here. Complete results arepresented in Supplementary Table S2.

Abbreviations: fem, feminine; masc, masculine.

Figure 2 Continued. C, concordance ofchanges in transcription of feminine,masculine, and gender-neutral liver geneseffected by chronic H. hepaticus infectionversus 7-d exposure to recombinantmurine IFN-g via osmotic minipump inuninfected mice, suggesting a role forIFN-g in chronic hepatitis and tumorigenesis.A complete comparison is presented inSupplementary Fig. S2. Sx cast, surgicalcastration; Chem cast, chemical castration(abarelix); DHT, dihydrotestosterone;Hh, H. hepaticus . Gene key: Cyp2a4,cytochrome P450 2A4; Egfr, epidermalgrowth factor receptor; Hsd3b5,hydroxysteroid dehydrogenase 3h-V;Igfbp2, insulin-like growth factor bindingprotein 2; Tff3, trefoil factor-3 (intestinal);H19, H19 fetal liver mRNA; Tnfa, TNF-a;Ubd, ubiquitin D; H2-aa, histocompatibility2 class II antigen A a; Ifit 2, IFN-inducedprotein with tetratricopeptide repeats 2;Nox4, NADPH oxidase 4; Cxcl10/IP10,chemokine (C-X-C motif) ligand 10(IFN-activated gene 10); Stat1, signaltransducer and activator of transcription 1.For all genes shown, P < 0.05, exceptwhere denoted by asterisk (*).

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11542 www.aacrjournals.org

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

cytokines in liver-gender disruption, we determined transcrip-tional responses in Helicobacter -free mice challenged withrecombinant murine IFN-g. With few exceptions (Cxcl10, Hspa1b,Lpl), genes altered by castration were confirmed as genderdimorphic in this experiment (Supplementary Fig. S2). Thisvalidated our assumption that castration sensitivity, largely, wassynonymous with sexual dimorphism. Of greater importance,almost all genes up-regulated or down-regulated in mice withtumorigenic hepatitis were altered in the same direction by IFN-g,suggesting a role for this proinflammatory cytokine in liver-gender disruption and disease pathogenesis (Fig. 2C). Concordantchanges were observed for the feminine genes Cd14, Cxcl9 (MIG),H2-aa, Ifit2, Ccrn4l, Dbp, Hamp1, and Mfsd2, masculine genes

Tff3, Cyp7b1, and Nox4, and gender-neutral genes Ccl2 (MCP-1),Cxcl10 (IP-10), Lpl, Stat1, and Tgtp (Supplementary Fig. S2).However, IFN-g did not up-regulate in males the exquisitelysensitive feminine gene sulfotransferase hydroxysteroid-preferring2 and produced a relatively modest reduction in the male-specifictranscript HSD3h-V (Supplementary Fig. S2). Therefore, IFN-gwas not responsible for the hepatic feminization associated withacute H. hepaticus infection. Rather, IFN-g had a more directeffect on a subset of gender-dimorphic genes resulting intranscriptional alterations in both the masculine and femininedirections.

Discussion

Because HCC is a male-predominant cancer, we tested thehypothesis that castration would reduce tumor formation inmice with preestablished hepatitis and, conversely, that androgenreceptor agonism would accelerate tumorigenesis. Both arms ofthis hypothesis were disproved. Neither surgical nor chemicalcastration of 1-year-old mice with chronic hepatitis reducedHCC at 21 months nor did supplementation of the powerfulandrogen receptor agonist dihydrotestosterone promote tumors.These findings contrast with the proved efficacy of peripubertalcastration for reducing chemical and transgene-promoted livercancer (36, 37). Therefore, the effectiveness of castration forreducing HCC in male mice is age sensitive. Our results confirmand extend those of Vesselinovitch, who showed that castrationat 38 weeks was significantly less effective than castration at20 weeks or earlier for reducing diethylnitrosamine-initiatedHCC (38). Taken together, murine studies show that malesusceptibility to hepatocarcinogenesis is maturationallyimprinted and that the benefit of androgen blockade againstHCC is inversely correlated with age. This agrees with theabsence of benefit of antiandrogen therapy for HCC in oldermen and argues against a direct role for male sex hormones inliver carcinogenesis (15–18).

A review of microarray data in the mouse literature showsthat the male liver responds to acute injury in part throughfeminization. We documented this in a prior H. hepaticus study,and similar responses have been reported in widely divergentmodels of acute hepatic injury (21, 39, 40). Intriguingly,hepatocyte-targeted disruption of h-catenin also results in liverfeminization, including decreased expression of the male-predominant gene Egfr (41). Thus, the male liver is predisposedto gender-dimorphic gene alterations in response to a widevariety of perturbations. One potential mechanism for feminiza-tion of the male liver after acute injury is inhibition of Stat5b bysuppressor of cytokine signaling 3 (Socs3) in response to IL-6(42). Nevertheless, in the masculinized mature liver, disease-associated gene alterations in the feminine direction do notresult in the balanced expression characteristic of the female anddo not confer protection against tumor formation.

In contrast to the simple feminization associated with acuteinjury, chronic tumorigenic hepatitis in our study was associatedwith a widespread mosaic of sex-dependent gene alterations,resulting in loss of liver-gender identity. Liver-gender disruptionwas not observed in infected mice without hepatitis, proving thatH. hepaticus infection alone was insufficient to incite thesechanges. In contrast, exogenous IFN-g did reproduce many ofthese changes, implicating a role for proinflammatory cytokines inthis process. Viewed as a whole, disease expression in this system

Figure 3. Assessment of androgenic intervention, sex, and/or H. hepaticusinfection on gender-sensitive biomarkers. A, liver microsomal LCFAs asdetermined by gas chromatography/mass spectrometry. Note gender differencein palmitic (C16:0) and arachidonic (C20:4) acids relative to total LCFA pool.Males with H. hepaticus–induced hepatitis had a significant increase in themonounsaturated oleic acid (C18:1) and decrease in saturated stearic (C18:0)and polyunsaturated linoleic acid (C18:2) relative to uninfected males, possiblyreflecting a disease-associated increase in stearoyl-CoA desaturase 2transcription. B, serum growth hormone, ELISA. Note wide distribution of serumgrowth hormone levels consistent with pulsatile secretion in all male groups (firstfour columns ) regardless of androgenic intervention. In contrast, female valuesclustered tightly as expected (fifth column ). C, serum dihydrotestosterone,ELISA. Dihydrotestosterone was significantly decreased in surgically andchemically castrated males (left two columns ) relative to castrated malesreceiving hormonal supplement (third column ) and sexually intact controls (fourthcolumn ). *, P < 0.05.

Liver-Gender Disruption and HCC

www.aacrjournals.org 11543 Cancer Res 2007; 67: (24). December 15, 2007

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

was largely a function of the host response. Because we examinedwhole liver slices, it was not possible to determine which liver cellpopulations contributed to up-regulated expression of specificgenes. We identified castration-sensitive disease-associated genesattributable both to hepatocyte and nonhepatocyte (e.g., leukocyte)cell populations (Table 2). Therefore, loss of gender specificity bothin immunity and hepatocyte function likely contributed to diseasepropagation.

Taken together, results of our study implicate cytokine-drivenliver-gender disruption and not androgen signaling as amechanism of male-predominant HCC promotion. Combiningdata presented here with results published by others, we havedeveloped a multistep working model of hepatocarcinogenesiswith a focus on the male liver (Fig. 4A). In our model,testosterone masculinizes the liver through a well-documentedseries of events (4). Acute injury invokes a feminizing responsethat includes alterations to a predictable set of genes, includingmembers of cytochrome P450, hydroxysteroid dehydrogenase,and sulfotransferase enzyme families (27). This may be mediatedin part by IL-6–induced Socs3 inhibition of Stat5b (42). A criticalrole for IL-6 in liver cancer risk was shown in a recent chemicalcarcinogenesis study wherein IL6�/� male mice displayed afemale phenotype of HCC resistance (43). In our working model,if acute hepatic injury does not resolve, the male liver continues

down one of two paths: (a) containment through appropriateand balanced physiologic responses or (b) propagation through aself-feeding cycle of inflammation and cellular distress. In thelatter case, continuous exposure to IFN-g and related proin-flammatory mediators alters the expression of cytokine-sensitivesexually dimorphic genes resulting in loss of liver-gender identity(confirmed in our study by microarray heat map comparison;Fig. 4B), metabolic derangement, and HCC. It is likely thatmolecular crosstalk exists between multiple inflammatory path-ways and sex-specific liver gene regulation. An NF-nB responseelement has been identified in the promoter region of thefemale-predominant gene CD36 ( fatty acid translocase; ref. 44).Whereas some studies have suggested an antitumorigenic rolefor IFN-g, these conclusions were based on the behavior ofcultured or transplanted tumor cell lines (45, 46). Our in vivomodel predicts a protumorigenic effect of chronic proinflamma-tory cytokine exposure. In all likelihood, the effect of IFN-g andother cytokines on liver carcinogenesis is context specific.

We postulate that deranged hepatocellular metabolism is thephysiologic connection between liver-gender disruption and HCC.For example, murine enzymes involved in cholesterol and bile acidmetabolism, Cyp7a1, Cyp7b1, and Cyp8b1, are gender dimorphic.Cyp7a1, the rate-limiting enzyme in classic bile acid synthesis, isfemale predominant, whereas Cyp7b1 and Cyp8b1 in the mouse are

Figure 4. Working model of inflammation-associatedliver-gender disruption and hepatocarcinogenesis. A, atpuberty, testosterone masculinizes liver gene transcription(light blue tiles ) through pulsatile growth hormone and Stat5b.Acute injury incites feminization (light pink tiles ) of a subsetof gender-dimorphic genes, possibly through IL-6 and/orTNF-a–induced up-regulation of Cish/Socs and inhibition ofStat5b (42). This may destabilize gender-specific metabolicbalance. In the face of subsequent chronic injury, the maleliver may respond appropriately and contain disease (bottomleft ) or inappropriately with a self-perpetuating cycle ofproinflammatory cytokines, such as IFN-g, and cell damage,resulting in global liver-gender disruption, metabolicderangement, and HCC (bottom right ). B, microarray heatmap of castration-sensitive hepatic genes demonstrating cleargender-identifying molecular signatures in healthy intact andcastrated males, but not in castrated males with hepatitis andHCC.

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11544 www.aacrjournals.org

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

masculine genes (10). Animals in our study with tumorigenichepatitis showed suppression of Cyp7b1 and Cyp8b1 but no changein Cyp7a1 mRNA levels. Elsewhere, mice with gene knockout of thebile acid sensor farnesoid X receptor were shown to developspontaneous liver neoplasms, and alterations to the above Cypgenes were associated with tumorigenesis (47). Based on the widerange of transcriptional alterations, the diseased male liver likelyaccumulates altered pools of cholesterol, bile acids, and LCFAs, asshown in our study for microsomal LCFA. This could result inshunting toward alternative pathways, oxidative damage, andactivation of proinflammatory second messengers, as well asdisrupted signaling through lipid-sensitive nuclear receptors, suchas peroxisome proliferator–activated receptors and retinoid X andliver X receptors (48).

The model we have developed regards gender-specifichepatocellular function not merely as a product of individualgenes but a balanced and interdependent system. As such,hepatic feminization associated with acute responses could leavethe male liver in precarious metabolic balance, amplifying thesubsequent effect of type I cytokines, such as IFN-g. Together,these forces would result in global liver-gender disruption,derangement of hepatocellular metabolism, and HCC with

a male bias. Nevertheless, the similar transcriptional profilesobserved in male and female mice challenged with IFN-gsuggest that proinflammatory cytokines are important intumor promotion in both sexes. Regardless of gender, tumorrisk in our model is determined primarily at the level of thehepatocyte, in agreement with the cell autonomous origin ofHCC in chimeric mouse models (49, 50). Further study will beneeded to determine at the molecular level how the loss ofgender-specific metabolic balance contributes to the oxidativedamage, epigenetic alterations, and dysregulated proliferationand apoptosis kinetics that are the hallmarks of hepatocellulartransformation.

Acknowledgments

Received 4/27/2007; revised 8/14/2007; accepted 10/8/2007.Grant support: NIH grants R01CA67529, T32RR07036, and P01CA26731 (J.G. Fox),

and pilot grant P30ES02109 (A.B. Rogers).The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Juri Ann Miyamae and Katherine Rydstrom for assistance with tissuecollection and processing, Nancy Taylor for laboratory administration, PraecisPharmaceuticals for the gift of Plenaxis, and members of the MIT Center forEnvironmental Health Sciences for helpful discussions and logistical support.

References1. El-Serag HB, Rudolph KL. Hepatocellular carcinoma:epidemiology and molecular carcinogenesis. Gastroen-terology 2007;132:2557–76.

2. Bosch FX, Ribes J, Diaz M, Cleries R. Primary livercancer: worldwide incidence and trends. Gastroenterol-ogy 2004;127:S5–16.

3. Dhir RN, Dworakowski W, Thangavel C, Shapiro BH.Sexually dimorphic regulation of hepatic isoforms ofhuman cytochrome p450 by growth hormone.J Pharmacol Exp Ther 2006;316:87–94.

4. Justo R, Boada J, Frontera M, Oliver J, Bermudez J,Gianotti M. Gender dimorphism in rat liver mitochon-drial oxidative metabolism and biogenesis. Am J PhysiolCell Physiol 2005;289:C372–8.

5. Gueraud F, Paris A. Hepatic microsomal membranelipidic composition and growth hormone effect in adultmale rat: evidence for a ‘feminization’ process of totalphospholipid fatty acid pattern. Biochim Biophys Acta1997;1329:97–110.

6. Lotter H, Jacobs T, Gaworski I, Tannich E. Sexualdimorphism in the control of amebic liver abscess ina mouse model of disease. Infect Immun 2006;74:118–24.

7. Waxman DJ, O’Connor C. Growth hormone regulationof sex-dependent liver gene expression. Mol Endocrinol2006;20:2613–29.

8. Veldhuis JD, Bowers CY. Three-peptide control ofpulsatile and entropic feedback-sensitive modes ofgrowth hormone secretion: modulation by estrogenand aromatizable androgen. J Pediatr Endocrinol Metab2003;16 Suppl 3:587–605.

9. Park SH, Wiwi CA, Waxman DJ. Signalling cross-talk between hepatocyte nuclear factor 4a andgrowth-hormone-activated STAT5b. Biochem J 2006;397:159–68.

10. Verma AS, Dhir RN, Shapiro BH. Inadequacy ofthe Janus kinase 2/signal transducer and activator oftranscription signal transduction pathway to mediateepisodic growth hormone-dependent regulation ofhepatic CYP2C11. Mol Pharmacol 2005;67:891–901.

11. Mugford CA, Kedderis GL. Sex-dependent metabo-lism of xenobiotics. Drug Metab Rev 1998;30:441–98.

12. Wolbold R, Klein K, Burk O, et al. Sex is a majordeterminant of CYP3A4 expression in human liver.Hepatology 2003;38:978–88.

13. Ahluwalia A, Clodfelter KH, Waxman DJ. Sexualdimorphism of rat liver gene expression: regulatoryrole of growth hormone revealed by deoxyribonu-cleic acid microarray analysis. Mol Endocrinol 2004;18:747–60.

14. Clodfelter KH, Holloway MG, Hodor P, Park SH, RayWJ, Waxman DJ. Sex-dependent liver gene expression isextensive and largely dependent upon signal transducerand activator of transcription 5b (STAT5b): STAT5b-dependent activation of male genes and repression offemale genes revealed by microarray analysis. MolEndocrinol 2006;20:1333–51.

15. Llovet JM. Updated treatment approach to hepato-cellular carcinoma. J Gastroenterol 2005;40:225–35.

16. GRETCH. Randomized trial of leuprorelin andflutamide in male patients with hepatocellular carci-noma treated with tamoxifen. Hepatology 2004;40:1361–9.

17. Chao Y, Chan WK, Huang YS, et al. Phase II study offlutamide in the treatment of hepatocellular carcinoma.Cancer 1996;77:635–9.

18. Colleoni M, Nelli P, Vicario G, Mastropasqua G,Manente P. Megestrol acetate in unresectable hepato-cellular carcinoma. Tumori 1995;81:351–3.

19. Fox JG, Li X, Yan L, et al. Chronic proliferativehepatitis in A/JCr mice associated with persistentHelicobacter hepaticus infection: a model of Helico-bacter -induced carcinogenesis. Infect Immun 1996;64:1548–58.

20. Rogers AB, Fox JG. Inflammation and Cancer. I.Rodent models of infectious gastrointestinal and livercancer. Am J Physiol Gastrointest Liver Physiol 2004;286:G361–6.

21. Rogers AB, Boutin SR, Whary MT, et al. Progressionof chronic hepatitis and preneoplasia in Helicobacterhepaticus -infected A/JCr mice. Toxicol Pathol 2004;32:668–77.

22. Fox JG, Dewhirst FE, Tully JG, et al. Helicobacterhepaticus sp. nov., a microaerophilic bacterium isolatedfrom livers and intestinal mucosal scrapings from mice.J Clin Microbiol 1994;32:1238–45.

23. Ge Z, White DA, Whary MT, Fox JG. FluorogenicPCR-based quantitative detection of a murine patho-gen, Helicobacter hepaticus . J Clin Microbiol 2001;39:2598–602.

24. Brooling JT, Campbell JS, Mitchell C, Yeoh GC, FaustoN. Differential regulation of rodent hepatocyte and oval

cell proliferation by interferon g. Hepatology 2005;41:906–15.

25. Harada T, Enomoto A, Boorman GA, Maronpot RR.Liver and Gallbladder. In: Maronpot RR, editor.Pathology of the Mouse. St. Louis (MO): Cache RiverPress; 1999. p. 119–84.

26. Cardiff RD, Anver MR, Boivin GP, et al. Precancer inmice: animal models used to understand, prevent, andtreat human precancers. Toxicol Pathol 2006;34:699–707.

27. Boutin SR, Rogers AB, Shen Z, et al. Hepatictemporal gene expression profiling in Helicobacterhepaticus -infected A/JCr mice. Toxicol Pathol 2004;32:678–93.

28. Croker BA, Krebs DL, Zhang JG, et al. SOCS3negatively regulates IL-6 signaling in vivo . Nat Immunol2003;4:540–5.

29. McGee M, Chen Z. Parameter estimation for theexponential-normal convolution model for backgroundcorrection of affymetrix GeneChip data. Stat Appl GenetMol Biol 2006;5:Article 24.

30. Rogers AB, Cormier KS, Fox JG. Thiol-reactivecompounds prevent nonspecific antibody binding inimmunohistochemistry. Lab Invest 2006;86:526–33.

31. Percy DH, Barthold SW. Mouse. In: Pathology ofLaboratory Rodents and Rabbits. Ames (IA): Iowa StatePress; 2001. p. 3–106.

32. Amador-Noguez D, Yagi K, Venable S, Darlington G.Gene expression profile of long-lived Ames dwarf miceand Little mice. Aging Cell 2004;3:423–41.

33. Myles MH, Livingston RS, Livingston BA, Criley JM,Franklin CL. Analysis of gene expression in ceca ofHelicobacter hepaticus -infected A/JCr mice before andafter development of typhlitis. Infect Immun 2003;71:3885–93.

34. Nambiar PR, Nakanishi M, Gupta R, et al. Geneticsignatures of high- and low-risk aberrant crypt foci in amouse model of sporadic colon cancer. Cancer Res 2004;64:6394–401.

35. Calvisi DF, Conner EA, Ladu S, Lemmer ER, FactorVM, Thorgeirsson SS. Activation of the canonical Wnt/h-catenin pathway confers growth advantages in c-Myc/E2F1 transgenic mouse model of liver cancer. J Hepatol2005;42:842–9.

36. Vesselinovitch SD, Itze L, Mihailovich N, Rao KV.Modifying role of partial hepatectomy and gonadectomyin ethylnitrosourea-induced hepatocarcinogenesis. Can-cer Res 1980;40:1538–42.

Liver-Gender Disruption and HCC

www.aacrjournals.org 11545 Cancer Res 2007; 67: (24). December 15, 2007

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

37. Matsumoto T, Takagi H, Mori M. Androgen depen-dency of hepatocarcinogenesis in TGFa transgenicmice. Liver 2000;20:228–33.

38. Vesselinovitch SD. Certain aspects of hepatocarcino-genesis in the infant mouse model. Toxicol Pathol 1987;15:221–8.

39. Deaciuc IV, Doherty DE, Burikhanov R, et al. Large-scale gene profiling of the liver in a mouse model ofchronic, intragastric ethanol infusion. J Hepatol 2004;40:219–27.

40. Weng Y, DiRusso CC, Reilly AA, Black PN, Ding X.Hepatic gene expression changes in mouse models withliver-specific deletion or global suppression of theNADPH-cytochrome P450 reductase gene. Mechanisticimplications for the regulation of microsomal cyto-chrome P450 and the fatty liver phenotype. J Biol Chem2005;280:31686–98.

41. Tan X, Behari J, Cieply B, Michalopoulos GK, MongaSP. Conditional deletion of h-catenin reveals its role in

liver growth and regeneration. Gastroenterology 2006;131:1561–72.

42. Denson LA, Held MA, Menon RK, Frank SJ, Parlow AF,Arnold DL. Interleukin-6 inhibits hepatic growth hor-mone signaling via up-regulation of Cis and Socs-3. Am JPhysiol Gastrointest Liver Physiol 2003;284:G646–54.

43. Naugler WE, Sakurai T, Kim S, et al. Gender disparityin liver cancer due to sex differences in MyD88-dependent IL-6 production. Science 2007;317:121–4.

44. Stahlberg N, Rico-Bautista E, Fisher RM, et al.Female-predominant expression of fatty acid translo-case/CD36 in rat and human liver. Endocrinology 2004;145:1972–9.

45. Okada T, Sawada T, Kubota K. Deferoxamineenhances anti-proliferative effect of interferon-gagainst hepatocellular carcinoma cells. Cancer Lett2007;248:24–31.

46. Komita H, Homma S, Saotome H, Zeniya M, Ohno T,Toda G. Interferon-g produced by interleukin-12-acti-

vated tumor infiltrating CD8+ T cells directly inducesapoptosis of mouse hepatocellular carcinoma. J Hepatol2006;45:662–72.

47. Yang F, Huang X, Yi T, Yen Y, Moore DD, Huang W.Spontaneous development of liver tumors in theabsence of the bile acid receptor farnesoid X receptor.Cancer Res 2007;67:863–7.

48. Anderson SP, Dunn C, Laughter A, et al. Overlappingtranscriptional programs regulated by the nuclearreceptors peroxisome proliferator-activated receptor a,retinoid X receptor, and liver X receptor in mouse liver.Mol Pharmacol 2004;66:1440–52.

49. Chiaverotti TA, Drinkwater NR. C57BR/cdJ hepato-carcinogen susceptibility genes act cell-autonomously inC57BR/cdJ<->C57BL/6J chimeras. Cancer Res 2003;63:4914–9.

50. Poole TM, Drinkwater NR. Strain dependent effectsof sex hormones on hepatocarcinogenesis in mice.Carcinogenesis 1996;17:191–6.

Cancer Research

Cancer Res 2007; 67: (24). December 15, 2007 11546 www.aacrjournals.org

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 12: Hepatocellular Carcinoma Associated with Liver-Gender ... · Hepatocellular carcinoma (HCC) is a male-predominant cancer (1). Chronic viral hepatitis is the most common predisposing

2007;67:11536-11546. Cancer Res   Arlin B. Rogers, Elizabeth J. Theve, Yan Feng, et al.   Disruption in Male MiceHepatocellular Carcinoma Associated with Liver-Gender

  Updated version

  http://cancerres.aacrjournals.org/content/67/24/11536

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2007/12/12/67.24.11536.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/24/11536.full#ref-list-1

This article cites 48 articles, 15 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/24/11536.full#related-urls

This article has been cited by 12 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/67/24/11536To request permission to re-use all or part of this article, use this link

Research. on January 26, 2021. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from