Functional Role of Cellular Senescence in Biliary Injury

8
REVIEW Functional Role of Cellular Senescence in Biliary Injury Q22 Luke Meng,* yz Morgan Quezada, y Phillip Levine, yx Yuyan Han,* y Kelly McDaniel,* yx Tianhao Zhou,* y Emily Lin,* y Shannon Glaser,* y Fanyin Meng,* yx Heather Francis,* yx and Gianfranco Alpini* y Q1 Q2 Q3 From Research, Central Texas Veterans Health Care System,* Temple; the Department of Medicine, y Digestive Disease Research Center, Scott & White Healthcare, Texas A&M Health Science Center, College of Medicine, Baylor Scott & White Health, Temple; the University of Texas Southwestern Medical Center, z Dallas; and Academic Operations, x Scott & White Memorial Hospital, Temple, Texas Accepted for publication October 28, 2014. Address correspondence to Gianfranco Alpini, Ph.D., Scott & White Digestive Disease Research Center, Central Texas Veterans University Health Care System, Texas A&M Health Science Center, Olin E. Teague Medical Center, 1901 S First St, Bldg 205, 1R60, Temple, TX 76504. E-mail: [email protected]. Cellular senescence is a state of irreversible cell cycle arrest that has been involved in many gastrointestinal diseases, including human cholestatic liver disorders. Senescence may play a role in biliary atresia, primary sclerosing cholangitis, cellular rejection, and primary biliary cirrhosis, four liver diseases affecting cholangiocytes and the biliary system. In this review, we examine proposed mechanisms of senescence-related biliary diseases, including hypotheses associated with the senescence-associated phenotype, induction of senescence in nearby cells, and the depletion of stem cell subpopulations. Current evidence for the molecular mechanisms of senescence in the previously mentioned diseases is discussed in detail, with attention to recent advances on the role of pathways associated with senescence-associated phenotype, stress-induced senescence, telo- mere dysfunction, and autophagy. (Am J Pathol 2015, -:1e8; http://dx.doi.org/10.1016/ j.ajpath.2014.10.027) Cellular Q6 senescence is a state of irreversible growth arrest in the G 1 phase of the cell cycle. 1,2 Telomere shortening, double-stranded DNA damage, inamma- tion, and other forms of cell stress all function as stimuli for cell senescence. Although traditionally cellular senescence has been viewed as a protective response to cellular injury or disruption, the study of the senescence-associated secretory phenotype (SASP) has become increasingly implicated in the pathogenesis of hepatobiliary disease. Specically, senescence has been hypothesized to play a prominent role in chol- angiopathies, including primary biliary sclerosis [or primary sclerosing cholangitis (PSC)], primary biliary cholangitis (PBC), cellular rejection (CR), and biliary atresia (BA). Over time, the SASP may provide proinammatory or other adverse stimulus to hep- atobiliary stem/progenitor cells, leading to the observed disease phenotypes. 3e5 This review summarizes current evidence for the molecular mechanisms of cellular senescence in the pathogenesis of select biliary diseases. Cellular Senescence First observed in 1965, human broblasts were noted to have a limited ability to replicate in culture. 6 Senescent cells are metabolically active cells that often remain in situ but no longer maintain proliferative activity. This devel- opment is associated with morphological changes causing the cells to appear large and at with characteristic nuclear changes, including vacuolation. 7e10 Two main tumor- suppressor pathways, p53 and p16 INK4a /pRB, have been shown to regulate senescence responses. 11 The expression of associated cell cycle inhibitors, including p15 INK4B , p16 INK4a , p21 WAF1/Cip1 , p53, and increased senescence- associated b-galactosidase (SA-b-GAL) activity, is Supported in part by NIH R01 grants DK062975, DK054811, and DK07698 (G.A., S.G., and F.M Q4 .), the Department of Veterans Affairs Merit Review Awards (G.A., S.G., and F.M.), the VA Q5 CD-2 Award (H.F.), and the PSC Foundation grant (H.F.). L.M., M.Q., and P.L. contributed equally to this work. Disclosures: None declared. Copyright ª 2014 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2014.10.027 ajp.amjpathol.org The American Journal of Pathology, Vol. -, No. -, - 2015 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 REV 5.2.0 DTD AJPA1934_proof 22 January 2015 6:08 pm EO: AJP14_0219

Transcript of Functional Role of Cellular Senescence in Biliary Injury

REVIEWFunctional Role of Cellular Senescence in BiliaryInjury

Q22 Luke Meng,*yz Morgan Quezada,y Phillip Levine,yx Yuyan Han,*y Kelly McDaniel,*yx Tianhao Zhou,*y Emily Lin,*y

Shannon Glaser,*y Fanyin Meng,*yx Heather Francis,*yx and Gianfranco Alpini*y

Q1Q2Q3

From Research, Central Texas Veterans Health Care System,* Temple; the Department of Medicine,y Digestive Disease Research Center, Scott & WhiteHealthcare, Texas A&M Health Science Center, College of Medicine, Baylor Scott & White Health, Temple; the University of Texas Southwestern MedicalCenter,z Dallas; and Academic Operations,x Scott & White Memorial Hospital, Temple, Texas

Accepted for publicationOctober 28, 2014.

Address correspondence toGianfranco Alpini, Ph.D., Scott& White Digestive DiseaseResearch Center, Central TexasVeterans University HealthCare System, Texas A&MHealth Science Center, Olin E.Teague Medical Center, 1901 SFirst St, Bldg 205, 1R60,Temple, TX 76504. E-mail:[email protected].

Cellular senescence is a state of irreversible cell cycle arrest that has been involved in manygastrointestinal diseases, including human cholestatic liver disorders. Senescence may play a rolein biliary atresia, primary sclerosing cholangitis, cellular rejection, and primary biliary cirrhosis,four liver diseases affecting cholangiocytes and the biliary system. In this review, we examineproposed mechanisms of senescence-related biliary diseases, including hypotheses associated withthe senescence-associated phenotype, induction of senescence in nearby cells, and the depletion ofstem cell subpopulations. Current evidence for the molecular mechanisms of senescence in thepreviously mentioned diseases is discussed in detail, with attention to recent advances on the roleof pathways associated with senescence-associated phenotype, stress-induced senescence, telo-mere dysfunction, and autophagy. (Am J Pathol 2015, -: 1e8; http://dx.doi.org/10.1016/

j.ajpath.2014.10.027)

CellularQ6 senescence is a state of irreversible growtharrest in the G1 phase of the cell cycle.1,2 Telomereshortening, double-stranded DNA damage, inflamma-tion, and other forms of cell stress all function asstimuli for cell senescence. Although traditionallycellular senescence has been viewed as a protectiveresponse to cellular injury or disruption, the study ofthe senescence-associated secretory phenotype (SASP)has become increasingly implicated in the pathogenesisof hepatobiliary disease. Specifically, senescence hasbeen hypothesized to play a prominent role in chol-angiopathies, including primary biliary sclerosis [orprimary sclerosing cholangitis (PSC)], primary biliarycholangitis (PBC), cellular rejection (CR), and biliaryatresia (BA). Over time, the SASP may provideproinflammatory or other adverse stimulus to hep-atobiliary stem/progenitor cells, leading to the observeddisease phenotypes.3e5 This review summarizes currentevidence for the molecular mechanisms of cellularsenescence in the pathogenesis of select biliarydiseases.

Cellular Senescence

First observed in 1965, human fibroblasts were noted tohave a limited ability to replicate in culture.6 Senescentcells are metabolically active cells that often remain in situbut no longer maintain proliferative activity. This devel-opment is associated with morphological changes causingthe cells to appear large and flat with characteristic nuclearchanges, including vacuolation.7e10 Two main tumor-suppressor pathways, p53 and p16INK4a/pRB, have beenshown to regulate senescence responses.11 The expressionof associated cell cycle inhibitors, including p15INK4B,p16INK4a, p21WAF1/Cip1, p53, and increased senescence-associated b-galactosidase (SA-b-GAL) activity, is

Supported in part by NIH R01 grants DK062975, DK054811, andDK07698 (G.A., S.G., and F.M Q4.), the Department of Veteran’s AffairsMerit Review Awards (G.A., S.G., and F.M.), the VA Q5CD-2 Award (H.F.),and the PSC Foundation grant (H.F.).L.M., M.Q., and P.L. contributed equally to this work.Disclosures: None declared.

Copyright ª 2014 American Society for Investigative Pathology.Published by Elsevier Inc. All rights reserved.http://dx.doi.org/10.1016/j.ajpath.2014.10.027

ajp.amjpathol.org

The American Journal of Pathology, Vol. -, No. -, - 2015

1234567891011121314151617181920212223242526272829303132333435363738394041424344454647484950515253545556575859606162

63646566676869707172737475767778798081828384858687888990919293949596979899100101102103104105106107108109110111112113114115116117118119120121122123124

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

commonly observed. In addition, the production ofsenescence-associated heterochromatin foci and DNA damageresponse proteins have all been used to aid in the identificationof senescent cells in association with the absence of replicativemarkers (Ki-67 or thymidine analogue bromodeoxyuridinein vitro).12 Overall, senescence has been shown to causedifferent effects in different tissues and cell types (Table 1½T1"½T1" ).Correlating expression of these markers with disease states hasdemonstrated the widespread prevalence of the senescentstates in hepatobiliary diseases.

Replicative Senescence

Multiple effectors have been identified leading to the devel-opment of senescence. Classically, replicative senescence de-velops from telomere erosion associated with the accumulationof replication cycles (aging) in the absence of corrective telo-merase (Figure 1½F1"½F1" ). The process of telomere erosion elicits aDNA damage response, which acts to induce the expression ofg-H2Ax (a phosphorylated histone variant of H2Ax) and theDNA damage response proteins p53-binding protein 1, nibrin,

and mediator of DNA damage checkpoint protein 1. DNAdamage kinases ataxia telangiectasia mutated and ataxia tel-angiectasia and Rad3-related protein are subsequently acti-vated and, in turn, activate checkpoint kinases 1 and 2.20,21 Theresultant signal cascade leads to differential expression of p53isoforms, and has been linked directly to senescent pheno-types, although the process, including breakpoints betweenapoptosis and senescence, remains incompletely understood,with some role of NF-kB in its regulation.22,23 In cholestaticliver disease, the role of age-related telomere erosion does notseem prominent. In a study of telomeres using quantitativefluorescent in situ hybridization (Q-FISH) and confirmed bySouthern blot analysis from 73 normal liver samples, chol-angiocytes and hepatocytes were shown to maintain theirtelomere lengths independent of age with only Kupffer andstellate cells, demonstrating age-related attrition.24

Premature Senescence

Numerous other mechanisms independent of replicativesenescence have been identified as pathways of so-called

Table 1 Summary of Effects of the Consequences on Various Cell Types

Cell type Consequences of senescence*

Hepatocytes Unclear, but senescence correlated with fibrosis stage

13

Cholangiocytes Unclear, but senescence correlated with Banff grade in acute rejection

14e16

Hepatic stellate cells Ameliorate fibrosis during acute liver damage via secretion of metalloproteinases

17

Retinal pigment epithelial cells Disruption of division and migration and atrophy of retina in adult macular degeneration

18

Fibroblasts Disrupted tissue structure locally via secretion of VEGF

1

Melanocytes Reinforcing senescence via secretion of IL-6, IL-8, and PAI-1

19

*Senescence causes different effects in different tissues and cell types.PAI, plasminogen activator inhibitor; VEGF, vascular endothelial growth factor.

SASP

Tumor SuppressionSenescence

IL-6, IL-8,MMPs

Tissue Repair /Tumor Progression

Immune Clearance

IL-1a

Tissue Aging

Self-Sustaining Feedback Loop

Growth Arrest

SenescenceSmuli

Figure 1 The senescence-associated secretoryphenotype (SASP) during aging, cancer develop-ment, and progression. Cellular senescence hasdual effects on human health during the pro-gression of aging and various human disorders.The beneficial effects include tumor suppressionand tissue repair after injury; the deleterious ef-fects include tumor and aging promotion thatinvolve several key SASP molecules, includingIL-6, IL-8, matrix metalloproteinases (MMPs), andspecific miRNAs.

print&web4C=F

PO

Meng et al

2 ajp.amjpathol.org - The American Journal of Pathology

125126127128129130131132133134135136137138139140141142143144145146147148149150151152153154155156157158159160161162163164165166167168169170171172173174175176177178179180181182183184185186

187188189190191192193194195196197198199200201202203204205206207208209210211212213214215216217218219220221222223224225226227228229230231232233234235236237238239240241242243244245246247248

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

premature senescence, including chromatin instability, DNAdamage, dysfunctional telomeres, oncogenic mutations,overexpression of cell cycle inhibitors, and stress signals(Figure 2½F2"½F2" ). Oncogenic-induced senescence has beencommonly characterized in vitro using mutant H-RasV12, anoncogene, to induce cell cycle arrest signals by activatingp53 and p16INK4A/pRB pathways.25 p53 contributes tocellular senescence by transactivating genes that blockcellular proliferation, including the p21/Cip1/WAF1 cyclin-dependent kinase inhibitor and miR-34 class ofQ7 miRNAs.26

Similarly, loss of tumor suppressors (PTEN and NF1Q8 ) hasbeen shown to induce premature senescent pathways.27,28

Although our understanding of DNA damageerelatedsenescence provides new insights into these molecularprocesses, stress-induced senescence is currently more afocal point for research on the pathobiology of chol-angiopathies. In vitro, stress-induced senescence has beenmodeled with alterations in culture medium, growth factors,and oxidative stress in various human cell lines.29e32 Inmouse cholangiocytes, proinflammatory cytokines, such astumor necrosis factor a, interferons b and g, have been usedto generate reactive oxygen species and were shown toinduce senescence via the p53/p21WAF1/Cip1 pathway.33 Theevidence linking autoinflammatory cholangiopathies to thispathway will be discussed later.

Potential Mechanisms of Senescence-RelatedHuman Diseases

Senescence-Associated Secretory Phenotype

Senescent cells exhibit altered gene expression and promi-nently secrete cytokines (eg, IL-1Q9 and IL-6), chemokines

(eg, IL-8, CXCL-1, and CXCL-2), insulin-like growthfactorebinding proteins (IGFBPs), and several other factors,including matrix metalloproteinases, serine proteases, andproinflammatory colony-stimulating factors. These solublesignaling factors are referred to as the SASP.34 Acting viaparacrine and autocrine mechanisms, these proteins alter themicroenvironment of the cells, which may lead to adisruption of tissue structure and function (Figure 3 ½F3"½F3").Although SASP has attracted interest for its potentialpathological role in many human diseases, the biologicalfunction of SASP as a regulatory mechanism in tumorsuppression and other areas remains incompletely under-stood. Evidence has emerged related to the antifibroticeffects of SASP in the setting of acute liver injury. Ashepatic stellate cells proliferate and secrete extracellularmatrix (ECM) components and undergo senescence todevelop SASP (Figure 4 ½F4"½F4"), they secrete metalloproteinasesand other proinflammatory molecules that digest the ECMproteins counterbalancing the initial fibrotic response.35

Because senescence has also been reported in chronic bileduct inflammation in the setting of primary biliary cirrhosisand primary sclerosing cholangitis,36 and cholangiocytes inPSC liver exhibit increased expression of SASP compo-nents,15 the senescence program may also limit the fibro-genic response to cholestatic liver injury through the similarHSC Q10-related mechanisms.

Induction of Senescence in Nearby Cells

The ability of SASP to propagate senescence through pos-itive feedback has been hypothesized to potentially be apathological mechanism in cholangiopathies. To date, theevidence for this effect remains limited. The observation of

Division Competent Cell

Oxida!ve Stress

Senescent Cell

SA-β-GALp16INK4a

p21WAF1/cipl

TelomereDysfunc!on

DNA Damage

Oncogenic Ac!va!on

Figure 2 Overview of senescence. Variousfactors, such as telomeric dysfunction, DNA dam-age, oncogenic activation, and oxidative stress,induce senescence in division competent cells. Onbecoming senescent, the cell and its nucleusbecome enlarged and begin to express p16INK4a,p21WAF1/Cip1, and senescence-associated b-galac-tosidase (SA-b-GAL). Senescent cells present alarge flattened morphology and build up a SA-b-GAL activity that distinguishes them from mostquiescent cells.

print&web4C=F

PO

Senescence in Biliary Injury

The American Journal of Pathology - ajp.amjpathol.org 3

249250251252253254255256257258259260261262263264265266267268269270271272273274275276277278279280281282283284285286287288289290291292293294295296297298299300301302303304305306307308309310

311312313314315316317318319320321322323324325326327328329330331332333334335336337338339340341342343344345346347348349350351352353354355356357358359360361362363364365366367368369370371372

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

clustered expression of p16INK4a in nevi has led to thehypothesis that senescent cells secrete senescence-inducingagents that act on other cells locally.37 Subsequently, inhuman melanoma cell lines, high levels of IGFBP P-rP1(IGFBP-rP1, previously known as IGFBP7 and mac25) inhibitBRAF-MEK-ERKQ11 oncogene signaling and activate senes-cence or apoptotic pathways.38 As discussed later, over-expression of IGFBP-rP1 has also been observed in BA.However, its exact function in the setting of the diseaseremains unclear. IGFBP-5 has also been implicated as a pro-tein, which may act locally to induce senescence. Humanumbilical endothelial cells treated with exogenous rh-IGFBP-5Q12

were found to increase expression of p53 and p21 anddecrease proliferation compared to untreated controls.39

Depletion of Progenitor Cell Populations

The potential pathological link between senescence and thedepletion of progenitor cells is perhaps theoretically moreobvious. The antiproliferative properties of senescence canbe detrimental to essential functions of progenitor cell pop-ulations, which are crucial to tissue repair and regeneration innumerous organs, particularly the liver. It is possible that asinjury or other stimuli deplete these populations, it interfereswith proper tissue function. Some of the most direct evidencefor this pathological mechanism comes from research oncardiac anthracycline toxicity using a rat model. Treatment ofmice with doxorubicin led to expansion of a p16INK4a-positive cardiac stem cell population exhibiting irreversiblegrowth arrest, and this population was significantly associ-ated with left ventricular dysfunction. By using intra-myocardial injections of syngeneic cardiac stem cells,researchers were able to rescue the cardiac function of the

treated animals, which led to significantly improved survivalcompared to untreated controls.40 The functional role of astrong cellular senescence inducer, the homeobox transcrip-tion factor prospero homeobox protein 1 (Prox1), has beenrecently clarified in liver progenitors.41 Prox1 depletion inbipotent hepatoblasts significantly decreased the expressionof multiple hepatocyte genes and led to defective hepatocytemorphogenesis. Consequently, abnormal epithelial structuresexpressing hepatocyte and cholangiocyte markers or resem-bling ectopic bile ducts developed in the Prox1-deficient liverparenchyma. Nevertheless, excessive commitment of hep-atoblasts into cholangiocytes, premature intrahepatic bileduct morphogenesis, and biliary hyperplasia occurred inperiportal areas of Prox1-deficient livers.41 Certainly, thecrucial role of liver progenitor cells could suggest that theremay be some contribution of this model in hepatobiliarydisease as well.

Cholangiopathies

Biliary Atresia

BA is a progressive, fibro-obliterative disease of the extra-hepatic biliary tree that represents the most common causeof pediatric liver transplantation. Even with early Kasaihepatoportoenterostomy, liver transplantation remainsnecessary for 60% to 80% of patients, and without trans-plant, the 5-year survival is an unfortunate 60%.42e44 Thepathophysiology of this disease remains unknown, althougha combination of viral, toxic, genetic, and immunologicaletiologies have been generally considered.Interestingly, senescence markers have been observed in

affected pediatric patients. In two patients with cirrhosis

DNA Damage Foci(CDNA-SCARS/TIF)

Heterochroma!n Foci(SAHF)

-

Growth

SASP

Arrest

p16INK4a

Proinflammatory State

SenescenceSmuli

Nonsenescent Cells(Cholangiocytes)

SA-β-GAL

Figure 3 Formation of senescence-associatedsecretory phenotype (SASP). Senescent cells/cholangiocytes display different phenotypes fromquiescent or terminally differentiated cells (nondi-viding), whereas the definite feature of the senescentphenotype is notably defined. The significant markersof senescent cells/cholangiocytes include an essen-tially irreversible growth arrest; expression ofsenescence-associated b-galactosidase (SA-b-GAL)and p16INK4a; nuclear foci containing DNA damageresponse proteins (DNA-SCARS/TIF Q20) or senescence-associated heterochromatin foci (SAHF); and robustsecretion of various growth factors, cytokines(proinflammatory), proteases, and other proteins,such as SASP. Proinflammatory SASP mediators(C-X-C chemokine receptor 2, IL-6, and IL-6 receptor)can reinforce cellular senescence in an autocrine orparacrine manner. The secretion of SASP is the mostsignificant of these effects because it turns senescentcells/cholangiocytes into a proinflammatory statethat has the ability to promote disease progression.

print&

web4C=F

PO

Meng et al

4 ajp.amjpathol.org - The American Journal of Pathology

373374375376377378379380381382383384385386387388389390391392393394395396397398399400401402403404405406407408409410411412413414415416417418419420421422423424425426427428429430431432433434

435436437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468469470471472473474475476477478479480481482483484485486487488489490491492493494495496

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

secondary to BA, p53 and SA-b-GAL were seen expressed innodular hepatocytes, canals of Hering, and cholangioles withan absent expression of p16 or p21.45 These observationscorrelate with the results of a genome-wide gene expressionanalysis of normal, diseased control, and end-stage BA livers,which found strong expression of IGFBP-rP1 compared to thenormal and non-BA diseased controls. Previously, IGFBP-rP1has been shown to be up-regulated in various cell lines withsenescent phenotypes and has shown to be a p53-responsivegene.46,47 Although the data are limited, taken together,these findings may suggest a possible mechanism for senes-cence of cholangiocytes in BA.

Although previously there have been little data involvingtelomere dysfunction as a mechanism leading to BA, a studyby Sanada et al48 measured hepatocyte telomeres usingQ-FISH and showed preserved telomere length, but normal-ized telomere/centromere ratios, being markedly reducedcompared to healthy controls. Whether these findings repre-sent a true cause or consequence of the BA remains unclear.Previously, estimations of hepatocyte telomere lengths usingQ-FISH in patients with cirrhosis from chronic viral hepatitis,autoimmune hepatitis, primary sclerosing cholangitis, primarybiliary cirrhosis, and alcoholic liver disease have found thattelomere length correlates to Child-Pugh score independent ofassociated disease.49 Although the role of telomere dysfunc-tion in BA remains unclear, these studies suggest that hepa-tocyte telomere length may have some potential role as anadjunct biomarker to clinical scoring models of cirrhosis, suchas pediatric end-stage liver disease, model for end-stage liverdisease, and Child-Pugh in certain diseases.

Primary Sclerosing Cholangitis

PSC is an idiopathic, autoinflammatory disorder characterizedby fibrosis, stricturing, and obliteration of medium and largeducts throughout the biliary epithelium. This incurable diseasehas a prominent associationwith inflammatory bowel disease in70% of patients, particularly ulcerative colitis, and its

progressive nature leads to complications, including cholestasis,hepatic failure, and cholangiocarcinoma.50 Median survival inthe absence of liver transplant is between 10 and 12 years.51,52

The role of cell senescence in PSC remains an emergingfield of research. Cholangiocytes in PSC have been shown tohave increased expression of SA-b-GAL, p16INK4a, andp21WAF1.53 We have demonstrated that two cellular senes-cence markers, plasminogen activator inhibitor-1 and p57,are significantly increased in isolated cholangiocytes fromMDR2 knockout mice, which develop periportal fibrosissimilar to human PSC (unpublished data). Plasminogenactivator inhibitor-1 is an essential mediator of replicativesenescence in vitro and is one of the biochemical fingerprintsof senescence in vivo.54 The p57 overexpression inducedcellular senescence through cell cycle arrest in the G1

phase.55 It has been demonstrated that p57 is involved inhepatocyte growth arrest at two distinct points during liverdevelopment: the perinatal period and the postnatal transitionto a quiescent adult hepatocyte phenotype.56 Recently,Tabibian et al15 Q13demonstrated increased expression ofg-H2Ax, a marker of DNA damage and up-regulation ofSASP markers IL-6, IL-8, chemokine ligand 2, and plas-minogen activator inhibitor-1 in diseased samples. Corollaryto these findings, by using the lipopolysaccharide inflam-matory stress model, they reproduced cholangiocytesenescence-inducing expression of SASP markers andbystander cholangiocyte senescence. Furthermore, theirstudy showed significantly increased expression of N-Rasprotein colocalization with activated RAS in PSC, which wasabsent in PBC, hepatitis C, or control samples. The data buildon their previous work suggesting that N-Ras protein medi-ates lipopolysaccharide-induced inflammation, further sup-porting a potential role for N-Ras signaling in thepathogenesis of PSC, adding a molecular mechanism to ourunderstanding of PSC as an inflammatory disease.57

One recent study has characterized and identifiedphenotypic and signaling features of isolated PSC patient-derived cholangiocytes.58 The cholangiocytes from stage

Figure 4 Recovery effect of hepatic stellatecells drives the senescence-associated secretoryphenotype (SASP) during liver fibrosis. Thesecretion of SASP by activated stellate cells canprevent further proliferation of extracellular ma-trix (ECM)-producing cells, promote ECM degra-dation, and accelerate clearance of activatedhepatic stellate cells from the site of liver injury/fibrosis. COL1A1, type I collagen; COL1A2,collagen 1 chain type II; CTGF, connective tissuegrowth factor; MMP, matrix Q21metalloproteinases;PDF, ---; ROS, reactive oxygen species; TGF,transforming growth factor; TIMP, tissue inhibitorof metalloproteinases.

print&

web4C=F

PO

Senescence in Biliary Injury

The American Journal of Pathology - ajp.amjpathol.org 5

497498499500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554555556557558

559560561562563564565566567568569570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604605606607608609610611612613614615616617618619620

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

4 PSC patient liver explants were isolated by dissection,differential filtration, and immune-magnetic bead separa-tion. The proportion of cholangiocytes staining positivefor senescence-associated b-galactosidase was found to bemuch higher in PSC cholangiocytes compared withcultured normal human cholangiocytes (48% versus 5%;P < 0.01).58 Interestingly, by using Q-FISH, telomerelength in the cholangiocytes of nine PSC samples wasfound to be maintained compared to normal controls.15

Adjacent hepatocyte telomere length was not reported.If the data from Wiemann et al49 on shortened hepatocytetelomeres in PSCs are substantiated, a complex interac-tion of senescent pathways could be involved in thepathogenesis of this disease. Certainly, differences instages of liver disease may play a role, but further clari-fication of the data on the telomere length of hepatocytesand cholangiocytes in PSCs may be helpful.

Cellular Rejection

Acute CR has historically been associated with a progressive,obliterative cholangiopathy. Histopathology shows a non-suppurative cholangitis of the interlobular bile ducts withcholangiocytes exhibiting cellular and nuclear enlargement,multinucleation with uneven nuclear spacing, and thickeningof the basement membrane.59 In recent years with moreaggressive immunosuppression, the rate of CR has decreasedfrom 60% to 70% to 15% to 20% in graft recipients, withductopenia becoming even rarer.60 Despite therapeutic ad-vances, CR remains a significant cause of morbidity and costin liver transplant.

Even in early stages of CR, senescence has beenobserved with absent proliferation of cholangiocytesdespite their stereotypical proliferative response toinjury.61 The number of p21WAF1/Cip1-positive chol-angiocytes has been shown to correlate with the Banffgrade of acute rejection and declines with treatment.62,63

Previously, it has been observed that patients with failedallografts who had received cyclosporine had more duc-topenia than patients who had received tacrolimus.64

Interestingly in other epithelial tissues, cyclosporine,but not tacrolimus, has been shown to augment trans-forming growth factor-b expression, which is known toinduce p21WAF1/Cip1 expression.65 Building on thesefindings, Brain et al14 presented observational data onS100A4 expression in CR and in an in vitro model thatdescribed dedifferentiation driven by transforming growthfactor-b2. Given the diverse expression of S100A4, theprecise implications of this work are uncertain, yetintriguing.

Primary Biliary Cirrhosis

Primary biliary cirrhosisQ14 (PBC) is a progressive, autoim-mune cholangiopathy involving the small intrahepatic bileducts. The disease tends to affect women >40 years who

may present with symptoms of cholestasis, fibrotic biliarylesions, and hepatomegaly.66,67 Over time if untreated, thesustained loss of bile duct epithelial cells in PBC leads tocirrhosis and liver failure. Serum antimitochondrial anti-bodies can be detected in the 90% of cases. These anti-bodies target the E2 and E3BP subunits of the pyruvatedehydrogenase complex (PDC-E2 and PDC-E3BP).68,69

However, their involvement in the pathogenesis of PBChas been uncertain.Sasaki et al53 Q15have led research on the role of cellular

senescence in pathogenesis of PBC. The authors haveshown that PBC cholangiocytes exhibit senescence markersSA-b-GAL, p16INK4, and p21WAF1/Cip far more frequentlythan normal and viral hepatitis controls and observed theincreased presence of infiltrating myeloperoxidase-positiveinflammatory cells in PBC.53 The role of the inflammatorycells remains unclear, but it suggests that oxidativestresseinduced senescence may contribute to the patho-genesis of PBC. Examining telomere length using Q-FISH,they demonstrated that diseased small bile ducts and duct-ules in PBC compared with normal-appearing bile ducts inPBC, chronic viral hepatitis, and normal livers had signifi-cantly shorter telomeres, although the role of telomerelength in PBC is unknown.More interestingly, autophagy, the lysosomal pathway

involving the catabolism of cellular components, has beenidentified as a necessary component to the activation of cellsenescence.70 Sasaki et al Q16showed that senescent PBCcholangiocytes accumulated markers of autophagy,including microtubule-associated proteins-light chain 3b,cathepsin D, and lysosome-associated membrane protein-1.They further showed aggregation of p62 sequestosome-1, aspecific marker for autophagy, in diseased small bile ducts,suggesting some impairment of autophagy in PBC.2,70e72

Subsequently, the group has shown colocalization of mito-chondrial antigen PDC-E2 with autophagy marker lightchain 3b and increased in vitro cell surface expression ofPDC-E2 in cultured BECs Q17on exposure to various stresses.73

Taken together, these findings provide insight into therelationship between autophagy, the expression of mito-chondrial antigens, and autoimmunity in PBC. The exactmechanisms behind this pathological interaction remainunclear, but certainly this work suggests further under-standing of the pathways related to senescence, particularlyautophagy, may yield new insights in the development ofbiliary disease.

Conclusion and Perspectives

Cellular senescence is linked to the development of varioushuman diseases, and its role in biliary disease remains to becompletely elucidated. Even as recent data suggest newquestions related to the mechanisms of senescence in thedevelopment of cholangiopathies, the role cellular senes-cence plays in the regulation of biliary growth, injury, and

Meng et al

6 ajp.amjpathol.org - The American Journal of Pathology

621622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653654655656657658659660661662663664665666667668669670671672673674675676677678679680681682

683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739740741742743744

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

response to injury continues to expand our understanding ofbiliary disease and hopefully, in the future, yield new op-portunities for targeted therapies.

References

1. Rodier F, Campisi J: Four faces of cellular senescence. J Cell Biol2011, 192:547e556

2. Sasaki M, Nakanuma Y: Novel approach to bile duct damage inprimary biliary cirrhosis: participation of cellular senescence andautophagy. Int J Hepatol 2012, 2012:452143

3. Yildiz G, Arslan-Ergul A, Bagislar S, Konu O, Yuzugullu H, Gursoy-Yuzugullu O, Ozturk N, Ozen C, Ozdag H, Erdal E, Karademir S,Sagol O, Mizrak D, Bozkaya H, Ilk HG, Ilk O, Bilen B, Cetin-Atalay R,Akar N, Ozturk M: Genome-wide transcriptional reorganization asso-ciated with senescence-to-immortality switch during human hepatocel-lular carcinogenesis. PLoS One 2013, 8:e64016

4. Krizhanovsky V, Xue W, Zender L, Yon M, Hernando E, Lowe SW:Implications of cellular senescence in tissue damage response, tumorsuppression, and stem cell biology. Cold Spring Harb Symp QuantBiol 2008, 73:513e522

5. Ozturk M, Arslan-Ergul A, Bagislar S, Senturk S, Yuzugullu H:Senescence and immortality in hepatocellular carcinoma. Cancer Lett2009, 286:103e113

6. Hayflick L: The limited in vitro lifetime of human diploid cell strains.Exp Cell Res 1965, 37:614e636

7. Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW: Onco-genic ras provokes premature cell senescence associated with accu-mulation of p53 and p16INK4a. Cell 1997, 88:593e602

8. Chen Q, Ames BN: Senescence-like growth arrest induced byhydrogen peroxide in human diploid fibroblast F65 cells. Proc NatlAcad Sci U S A 1994, 91:4130e4134

9. Parrinello S, Samper E, Krtolica A, Goldstein J, Melov S, Campisi J:Oxygen sensitivity severely limits the replicative lifespan of murinefibroblasts. Nat Cell Biol 2003, 5:741e747

10. Aravinthan A, Verma S, Coleman N, Davies S, Allison M,Alexander G: Vacuolation in hepatocyte nuclei is a marker ofsenescence. J Clin Pathol 2012, 65:557e560

11. Lowe SW, Cepero E, Evan G: Intrinsic tumour suppression. Nature2004, 432:307e315

12. Collado M, Gil J, Efeyan A, Guerra C, Schuhmacher AJ, Barradas M,Benguría A, Zaballos A, Flores JM, Barbacid M: Tumour biology:senescence in premalignant tumours. Nature 2005, 436:642

13. Hoare M, Das T, Alexander G: Ageing, telomeres, senescence, andliver injury. J Hepatol 2010, 53:950e961

14. Brain JG, Robertson H, Thompson E, Humphreys EH, Gardner A,Booth TA, Jones DE, Afford SC, von Zglinicki T, Burt AD,Kirby JA: Biliary epithelial senescence and plasticity in acute cellularrejection. Am J Transplant 2013, 13:1688e1702

15. Tabibian JH, O’Hara SP, Splinter PL, Trussoni CE, LaRusso NF:Cholangiocyte senescence by way of N-ras activation is a charac-teristic of primary sclerosing cholangitis. Hepatology 2014, 59:2263e2275

16. Taguchi K, Hirano I, Itoh T, Tanaka M, Miyajima A, Suzuki A,Motohashi H, Yamamoto M: Nrf2 enhances cholangiocyte expansionin Pten-deficient livers. Mol Cell Biol 2014, 34:900e913

17. Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA: Liverfibrosis and repair: immune regulation of wound healing in a solidorgan. Nat Rev Immunol 2014, 14:181e194

18. Ambati J, Atkinson JP, Gelfand BD: Immunology of age-relatedmacular degeneration. Nat Rev Immunol 2013, 13:438e451

19. Chinta SJ, Lieu CA, Demaria M, Laberge RM, Campisi J,Andersen JK: Environmental stress, ageing and glial cell senescence:a novel mechanistic link to Parkinson’s disease? J Intern Med 2013,273:429e436

20. di Fagagna FdA, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, vonZglinicki T, Saretzki G, Carter NP, Jackson SP: A DNA damagecheckpoint response in telomere-initiated senescence. Nature 2003,426:194e198

21. Ward IM, Chen J: Histone H2AX is phosphorylated in an ATR-dependent manner in response to replicational stress. J Biol Chem2001, 276:47759e47762

22. Fujita K, Mondal AM, Horikawa I, Nguyen GH, Kumamoto K,Sohn JJ, Bowman ED, Mathe EA, Schetter AJ, Pine SR: p53 isoformsD133p53 and p53b are endogenous regulators of replicative cellularsenescence. Nat Cell Biol 2009, 11:1135e1142

23. Crescenzi E, Pacifico F, Lavorgna A, De Palma R, D’Aiuto E,Palumbo G, Formisano S, Leonardi A: NF-kB-dependent cytokinesecretion controls Fas expression on chemotherapy-induced prema-ture senescent tumor cells. Oncogene 2011, 30:2707e2717

24. Verma S, Tachtatzis P, Penrhyn-Lowe S, Scarpini C, Jurk D, VonZglinicki T, Coleman N, Alexander GJ: Sustained telomere length inhepatocytes and cholangiocytes with increasing age in normal liver.Hepatology 2012, 56:1510e1520

25. Ben-Porath I, Weinberg RA: The signals and pathways activatingcellular senescence. Int J Biochem Cell Biol 2005, 37:961e976

26. He L, He X, Lowe SW, Hannon GJ: microRNAs join the p53 net-workeanother piece in the tumour-suppression puzzle. Nat RevCancer 2007, 7:819e822

27. Chen Z, Trotman LC, Shaffer D, Lin H-K, Dotan ZA, Niki M,Koutcher JA, Scher HI, Ludwig T, Gerald W: Crucial role of p53-dependent cellular senescence in suppression of Pten-deficienttumorigenesis. Nature 2005, 436:725e730

28. Courtois-Cox S, Genther Williams SM, Reczek EE, Johnson BW,McGillicuddy LT, Johannessen CM, Hollstein PE, MacCollin M,Cichowski K: A negative feedback signaling network underliesoncogene-induced senescence. Cancer Cell 2006, 10:459e472

29. Chen Q, Fischer A, Reagan JD, Yan L-J, Ames BN: Oxidative DNAdamage and senescence of human diploid fibroblast cells. Proc NatlAcad Sci U S A 1995, 92:4337e4341

30. Yuan H, Kaneko T, Matsuo M: Relevance of oxidative stress to thelimited replicative capacity of cultured human diploid cells: the limitof cumulative population doublings increases under low concentra-tions of oxygen and decreases in response to aminotriazole. MechAgeing Dev 1995, 81:159e168

31. Bennett DC, Medrano EE: Molecular regulation of melanocytesenescence. Pigment Cell Res 2002, 15:242e250

32. Wright WE, Shay JW: Historical claims and current interpretations ofreplicative aging. Nat Biotechnol 2002, 20:682e688

33. Sasaki M, Ikeda H, Sato Y, Nakanuma Y: Proinflammatory cytokine-induced cellular senescence of biliary epithelial cells is mediated viaoxidative stress and activation of ATM pathway: a culture study. FreeRadic Res 2008, 42:625e632

34. Coppé J-P, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J,Nelson PS, Desprez P-Y, Campisi J: Senescence-associated secretoryphenotypes reveal cell-nonautonomous functions of oncogenic RASand the p53 tumor suppressor. PLoS Biol 2008, 6:e301

35. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C,Yee H, Zender L, Lowe SW: Senescence of activated stellate cellslimits liver fibrosis. Cell 2008, 134:657e667

36. Sasaki M, Ikeda H, Yamaguchi J, Nakada S, Nakanuma Y: Telomereshortening in the damaged small bile ducts in primary biliary cirrhosisreflects ongoing cellular senescence. Hepatology 2008, 48:186e195

37. Gray-Schopfer V, Wellbrock C, Marais R: Melanoma biology andnew targeted therapy. Nature 2007, 445:851e857

38. Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR: Onco-genic BRAF induces senescence and apoptosis through pathwaysmediated by the secreted protein IGFBP7. Cell 2008, 132:363e374

39. Kim KS, Seu YB, Baek S-H, Kim MJ, Kim KJ, Kim JH, Kim J-R:Induction of cellular senescence by insulin-like growth factor bindingprotein-5 through a p53-dependent mechanism. Mol Biol Cell 2007,18:4543e4552

Senescence in Biliary Injury

The American Journal of Pathology - ajp.amjpathol.org 7

745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774775776777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806

807808809810811812813814815816817818819820821822823824825826827828829830831832833834835836837838839840841842843844845846847848849850851852853854855856857858859860861862863864865866867868

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219

40. De Angelis A, Piegari E, Cappetta D, Marino L, Filippelli A,Berrino L, Ferreira-Martins J, Zheng H, Hosoda T, Rota M:Anthracycline cardiomyopathy is mediated by depletion of the car-diac stem cell pool and is rescued by restoration of progenitor cellfunction. Circulation 2010, 121:276e292

41. Seth A, Ye J, Yu N, Guez F, Bedford DC, Neale GA, Cordi S,Brindle PK, Lemaigre FP, Kaestner KH, Sosa-Pineda B: Prox1ablation in hepatic progenitors causes defective hepatocyte specifi-cation and increases biliary cell commitment. Development 2014,141:538e547

42. Chardot C, Carton M, Spire-Bendelac N, Le Pommelet C,Golmard JL, Auvert B: Prognosis of biliary atresia in the era of livertransplantation: French national study from 1986 to 1996. Hepatology1999, 30:606e611

43. Serinet M-O, Wildhaber BE, Broué P, Lachaux A, Sarles J,Jacquemin E, Gauthier F, Chardot C: Impact of age at Kasai operationon its results in late childhood and adolescence: a rational basis forbiliary atresia screening. Pediatrics 2009, 123:1280e1286

44. Had�zic N, Davenport M, Tizzard S, Singer J, Howard ER, Mieli-Vergani G: Long-term survival following Kasai portoenterostomy: ischronic liver disease inevitable? J Pediatr Gastroenterol Nutr 2003,37:430e433

45. Gutierrez-Reyes G, de Leon MdCG, Varela-Fascinetto G, Valencia P,Tamayo RP, Rosado CG, Labonne BF, Rochilin NM, Garcia RM,Valadez JA: Cellular senescence in livers from children with endstage liver disease. PLoS One 2010, 5:e10231

46. Swisshelm K, Ryan K, Tsuchiya K, Sager R: Enhanced expression ofan insulin growth factor-like binding protein (mac25) in senescenthuman mammary epithelial cells and induced expression with retinoicacid. Proc Natl Acad Sci U S A 1995, 92:4472e4476

47. Suzuki H, Igarashi S, Nojima M, Maruyama R, Yamamoto E, Kai M,Akashi H, Watanabe Y, Yamamoto H, Sasaki Y: IGFBP7 is a p53-responsive gene specifically silenced in colorectal cancer with CpGisland methylator phenotype. Carcinogenesis 2010, 31:342e349

48. Sanada Y, Kawano Y, Miki A, Aida J, Nakamura K, Shimomura N,Ishikawa N, Arai T, Hirata Y, Yamada N, Okada N, Wakiya T,Ihara Y, Urahashi T, Yasuda Y, Takubo K, Mizuta K: Maternal graftsprotect daughter recipients from acute cellular rejection after pediatricliving donor liver transplantation for biliary atresia. Transpl Int 2014,27:383e390

49. Wiemann SU, Satyanarayana A, Tsahuridu M, Tillman HL, Zender L,Klempnaur J, Flemming P, Franco S, Blasco MA, Manns MP: He-patocyte telomere shortening and senescence are general markers ofhuman liver cirrhosis. FASEB J 2002, 16:935e942

50. Boonstra K, Beuers U, Ponsioen CY: Epidemiology of primarysclerosing cholangitis and primary biliary cirrhosis: a systematic re-view. J Hepatol 2012, 56:1181e1188

51. Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, Maccarty RL,Hunter EB, Fleming TR, Fisher LD, Beaver SJ, Larusso NF: Primarysclerosing cholangitis: natural history, prognostic factors and survivalanalysis. Hepatology 1989, 10:430e436

52. Broome U, Olsson R, Lööf L, Bodemar G, Hultcrantz R,Danielsson A, Prytz H, Sandberg-Gertzen H, Wallerstedt S,Lindberg G: Natural history and prognostic factors in 305 Swedishpatients with primary sclerosing cholangitis. Gut 1996, 38:610e615

53. Sasaki M, Ikeda H, Haga H, Manabe T, Nakanuma Y: Frequentcellular senescence in small bile ducts in primary biliary cirrhosis: apossible role in bile duct loss. J Pathol 2005, 205:451e459

54. Eren M, Boe AE, Murphy SB, Place AT, Nagpal V, Morales-Nebreda L, Urich D, Quaggin SE, Budinger GR, Mutlu GM,Miyata T, Vaughan DE: PAI-1-regulated extracellular proteolysisgoverns senescence and survival in Klotho mice. Proc Natl Acad SciU S A 2014, 111:7090e7095

55. Tsugu A, Sakai K, Dirks PB, Jung S, Weksberg R, Fei YL, Mondal S,Ivanchuk S, Ackerley C, Hamel PA, Rutka JT: Expression of

p57(KIP2) potently blocks the growth of human astrocytomas andinduces cell senescence. Am J Pathol 2000, 157:919e932

56. Awad MM, Sanders JA, Gruppuso PA: A potential role forp15(Ink4b) and p57(Kip2) in liver development. FEBS Lett 2000,483:160e164

57. O’Hara SP, Splinter PL, Trussoni CE, Gajdos GB, Lineswala PN,LaRusso NF: Cholangiocyte N-Ras protein mediates lipopolysaccharide-induced interleukin 6 secretion and proliferation. J Biol Chem 2011, 286:30352e30360 Q18

58. Tabibian JH, Trussoni CE, O’Hara SP, Splinter PL, Heimbach JK,LaRusso NF: Characterization of cultured cholangiocytes isolatedfrom livers of patients with primary sclerosing cholangitis. Lab Invest2014, 94:1126e1133

59. Vierling JM, Fennell RH: Histopathology of early and late humanhepatic allograft rejection: evidence of progressive destruction ofinterlobular bile ducts. Hepatology 1985, 5:1076e1082

60. Maluf DG, Stravitz RT, Cotterell AH, Posner MP, Nakatsuka M,Sterling RK, Luketic VA, Shiffman ML, Ham JM, Marcos A: Adultliving donor versus deceased donor liver transplantation: a 6-yearsingle center experience. Am J Transplant 2005, 5:149e156

61. Koukoulis GK, Shen J, Karademir S, Jensen D, Williams J: Chol-angiocytic apoptosis in chronic ductopenic rejection. Hum Pathol2001, 32:823e827

62. Ray MB, Schroeder T, Michaels SE, Hanto DW: Increased expres-sion of proliferating cell nuclear antigen in liver allograft rejection.Liver Transpl Surg 1996, 2:337e342

63. Lunz JG III, Contrucci S, Ruppert K, Murase N, Fung JJ, Starzl TE,Demetris AJ: Replicative senescence of biliary epithelial cells pre-cedes bile duct loss in chronic liver allograft rejection: increasedexpression of p21WAF1/Cip1 as a disease marker and the influenceof immunosuppressive drugs. Am J Pathol 2001, 158:1379e1390

64. Blakolmer K, Seaberg EC, Batts K, Ferrell L, Markin R, Wiesner R,Detre K, Demetris A: Analysis of the reversibility of chronic liverallograft rejection implications for a staging schema. Am J SurgPathol 1999, 23:1328e1339

65. Zhang JG, Walmsley M, Moy J, Cunningham A, Talbot D, Dark J,Kirby J: Differential effects of cyclosporin A and tacrolimus on theproduction of TGF-b: implications for the development of obliterativebronchiolitis after lung transplantation. Transpl Int 1998, 11:S325eS327

66. Jones DE: Pathogenesis of primary biliary cirrhosis. Clin Liver Dis2008, 12:305e321; viii

67. Invernizzi P, Gershwin ME: Primary biliary cirrhosis: bad genes, badluck. Dig Dis Sci 2012, 57:599e601

68. Fussey SP, Guest JR, James OF, Bassendine MF, Yeaman SJ:Identification and analysis of the major M2 autoantigens in primarybiliary cirrhosis. Proc Natl Acad Sci U S A 1988, 85:8654e8658

69. Shimoda S, Van de Water J, Ansari A, Nakamura M, Ishibashi H,Coppel RL, Lake J, Keeffe EB, Roche TE, Gershwin ME: Identifi-cation and precursor frequency analysis of a common T cell epitopemotif in mitochondrial autoantigens in primary biliary cirrhosis. JClin Invest 1998, 102:1831e1840 Q19

70. Young AR, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JF,Tavaré S, Arakawa S, Shimizu S, Watt FM: Autophagy mediates themitotic senescence transition. Genes Dev 2009, 23:798e803

71. Sasaki M, Miyakoshi M, Sato Y, Nakanuma Y: Autophagy mediatesthe process of cellular senescence characterizing bile duct damages inprimary biliary cirrhosis. Lab Invest 2010, 90:835e843

72. Sasaki M, Miyakoshi M, Sato Y, Nakanuma Y: A possibleinvolvement of p62/sequestosome-1 in the process of biliary epithe-lial autophagy and senescence in primary biliary cirrhosis. Liver Int2012, 32:487e499

73. Sasaki M, Miyakoshi M, Sato Y, Nakanuma Y: Increased expressionof mitochondrial proteins associated with autophagy in biliaryepithelial lesions in primary biliary cirrhosis. Liver Int 2013, 33:312e320

Meng et al

8 ajp.amjpathol.org - The American Journal of Pathology

869870871872873874875876877878879880881882883884885886887888889890891892893894895896897898899900901902903904905906907908909910911912913914915916917918919920921922923924925926927928929930931932933

934935936937938939940941942943944945946947948949950951952953954955956957958959960961962963964965966967968969970971972973974975976977978979980981982983984985986987988989990991992993994995996997998

REV 5.2.0 DTD ! AJPA1934_proof ! 22 January 2015 ! 6:08 pm ! EO: AJP14_0219