<<

58 3

t b doan and others Nuclear receptors in 58: 3 R169–R190 Review

Emerging functional roles of nuclear receptors in breast cancer

Tram B Doan, J Dinny Graham and Christine L Clarke Correspondence should be addressed Westmead Institute for Medical Research, Sydney Medical School – Westmead, University of Sydney, to T B Doan Sydney, New South Wales, Australia Email [email protected]

Abstract

Nuclear receptors (NRs) have been targets of intensive drug development for decades Key Words due to their roles as key regulators of multiple developmental, physiological and disease ff nuclear receptors processes. In breast cancer, expression of the and remains ff breast cancer clinically important in predicting prognosis and determining therapeutic strategies. ff circadian More recently, there is growing evidence supporting the involvement of multiple nuclear ff metabolism receptors other than the estrogen and progesterone receptors, in the regulation of ff migration and metastasis various processes important to the initiation and progression of breast cancer. We review new insights into the mechanisms of action of NRs made possible by recent advances in genomic technologies and focus on the emerging functional roles of NRs in breast cancer biology, including their involvement in circadian regulation, metabolic reprogramming Journal of Molecular and breast cancer migration and metastasis. Endocrinology (2017) 58, R169–R190 Journal of Molecular Endocrinology Introduction

Breast cancer is the most common cancer diagnosed nearly all physiological aspects of life and the availability in women worldwide (Ferlay et al. 2015). Over the past of drugs targeting NRs that resulted from intensive drug decades, substantial progress toward treatment of primary development targeting NRs for a range of pathological -positive (ER+) breast cancer has been conditions. The human NR superfamily consists of 48 made with the development of endocrine therapies highly evolutionarily conserved factors with targeting the estrogen biosynthesis and signaling the ability to act as molecular sensors of physiological pathways. Despite the success of endocrine therapies, and environmental stimuli (Mangelsdorf et al. 1995). there remain subgroups of women for whom available Development of technologies that allow genome-wide, treatment offers little help. These include patients with unbiased profiling of genomic binding sites and ER+ breast tumors who develop endocrine treatment expression has allowed deeper insights into the molecular resistance (Dixon 2014), patients with estrogen receptor- mechanisms of NR actions. These studies highlight the negative (ER−) breast cancers (Hudis & Gianni 2011) complexity of gene regulatory networks governed by and cancers that recur in all age groups (EBCTCG 2005). NRs, the extensive of NR crosstalk and the role Women in these subgroups currently face the challenge of coregulators and the chromatin landscape of the cell of living with advanced disease; therefore, there exists as important modulators of NR cell type-specific and the need to develop rational treatments targeting these context-dependent function. subgroups of breast cancer. In the context of breast cancer, it is recognized that Nuclear receptors (NRs) are potential promising targets estrogen receptor (ESR1) and (PGR) due to the importance of NRs as master regulators of play critical roles in the development and progression

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access

10.1530/JME-16-0082 Journal of Molecular Endocrinology (AF1); theDNA-bindingdomain (DBD)consistingof The N-terminalA/Bdomain withactivationfunction1 Mangelsdorf 2014),whichconsistsoffourmajor domains: structuralorganization (Evans & evolutionarily conserved of theNRsuperfamilyareidentified throughtheirhighly acid andoxysterols(Mangelsdorf vitamins and cholesterol metabolites including retinoic for hormones, thyroid hormones, lipophilic members. Thesetranscriptionfactorsincludethereceptors The humannuclearreceptorsuperfamilyconsistsof48 The nuclearreceptor superfamily genomic technologies. of actionNRsmadepossiblethroughadvances in as wellnewinsightsintothemolecularmechanisms these emergingfunctionalrolesofNRsinbreastcancer, and metastasis.Inthisreview, wewilldiscussevidencefor regulation ofthecircadian clock,metabolism,migration of breastcancertumorigenesisandprogressionincluding and that NRs are important regulators of several aspects involvement inregulatingproliferationandapoptosis involvement ofNRsinbreastcancerextendsbeyondtheir unexplored. Recentstudiesareestablishingthatthe the normalbreastandcancerhavelargelybeen of theremainingmembershumanNRfamilyin 2014). However, thefunctionalrolesofmajority 2002, al. et (RARB) inbreastcancer(Yang effect of the receptor beta the anti-migratory al.2009, et signaling (Hua the retinoicacidreceptoralpha(RARA)andestrogen considerable attention,particularlythecrosstalkbetween of theretinoidreceptorsubfamilyhavealsoreceived and anti-apoptoticeffectsinbreastcancer. Members have alsobeeninvestigatedfortheiranti-proliferative al.2011, et (GR) (Vilasco 2010, including thevitaminD3receptor(VDR)(Krishnan et al. 2015).Anumberofothernuclearreceptors, Barton et al. as apotentialtreatmenttarget(Cordera&Jordan2006, 2015), has emerged both as a marker of outcome and al.2010, et breasttumors(Park in primary (AR),whichisfrequentlyexpressed disease (Cordera&Jordan2006).Morerecently, the the estrogensignalingaxisarestandardofcareforER+ clinical managementprogramsandtreatmentstargeting and disease outcome.Consequently, measurementofESR1 breast tumorremainsthesinglemostrobustpredictorof of breastcancer, andmoreover, their expression ina DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review PGR expressioninbreastcancersisamainstayof 2013)andtheglucocorticoid receptor Mehta et al. t al.2010),and et Ross-Innes t al.2015b) et Abduljabbar t

b t al.1995).Members et

© 2017SocietyforEndocrinology doan andothers Printed inGreatBritain t al. et Flamini t al. et Grogg wide varietyofbiologicalprocesses innormalphysiology been showntoaffectthecell typespecificityofNRs. coregulators andthechromatin landscapeofthecellhave al. 2013).Boththeinteractionwith et (Millard factors, the chromatin landscape as well as non-coding target genesthroughinteractionswithothertranscription complexes thataffectNRtranscriptionalregulation of majority of coregulators function as members of large and includebothcoactivatorscorepressors.The to themechanismsbywhichNRsexerttheirfunctions conformation of the AF2 region. Coregulators are integral interaction ofNRswithcoregulatorsbyinfluencingthe Gronemeyer 2002, et al.2004). selective recognition by different NR subclasses (Laudet & this canonicalhormoneresponseelementallowfor repeats separatedbyaspacerwithvariablelength)of and duplications(organizedasdirect,invertedoreverted canonical hexamericsequenceRGGTCA.Modifications Nuclear receptorresponseelementsarederivativesofthe to increasebindingaffinity, specificityanddiversity. orphan receptors.Dimerizationisageneralmechanism heterodimerization partnersforVDR,THR,RARand heterodimers. RXRsinparticularcanactaspromiscuous THR, RARandRXRcanformbothhomodimers receptors generallybindashomodimers,whereasVDR, another familymember. Forexample,steroidhormone asmonomers,homodimersorheterodimerswith regionsoftarget response elementswithintheregulatory activation, NRsregulategenetranscriptionbybinding in circadian andmetabolic pathways(reviewedhere). inhuman,especiallyforNRsthatareinvolved observed ), whichpartiallyreflectthefunctionalgroupings (Fig. 1B of NRsintointegratedphysiologicfunctionalgroups NRs. In the mouse, this revealed a hierarchical organization feasible, basedonprofilingtheanatomicalexpressionof Organization ofNRsintofunctionalgroupingsisalso al.1999, et construction (Auwerx based onsequencealignmentandphylogenetictree groups 48 humanNRsareorganizedintosixevolutionary ligands fortheseNRshavenotbeenfound(Table 1). The designated asorphansbecauseendogenousphysiological 1A).ApproximatelyhalfoftheNRsare 1995) (Fig. 1986, (Giguere et al. have anAF2regionthatmediatescoactivatorinteraction -binding domain(LBD),whichmayornot specificity andthehingeregionlinkingDBDto two zincfingermotifswhichconfersresponseelement breast cancer Nuclear receptorsin Published byBioscientifica Ltd. NRs regulatethetranscription ofgenesthatcontrola Ligand bindingalsoallostericallycontrolsthe In theclassicalmodelofNRaction,uponligand

1987, Kumar et al. Downloaded fromBioscientifica.com at09/27/202109:55:09AM t al.2006). et Germain 58 Mangelsdorf : 3 170 170 et al. via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 171 breast cancer

Figure 1 (A) Schematic diagram of the structural and functional organization of nuclear receptors (reprinted by permission from Macmillan Publishers Ltd: Nature Reviews Drug Discovery (Gronemeyer et al. 2004), copyright 2004). The evolutionary conserved regions C and E are indicated as boxes (green and orange, respectively), and a black bar represent the divergent regions A/B, D and F. Domain functions are depicted above and below the scheme (AD, activation domain; AF1, activation function 1; NLS, nuclear localization signal). (B) Hierarchical clustering of NR tissue expression profile (reprinted from Cell, volume 126, Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM & Mangelsdorf DJ, Anatomical profiling of expression reveals a hierarchical transcriptional network, pages 789–799, copyright (2006), with permission from Elsevier). The relationship between receptor expression, function and is depicted as a circular dendrogram using the hierarchical, unsupervised clustering of NR tissue expression distribution profiles in mouse. The analysis reveals the existence of a higher order network tying nuclear receptor function to , development, central and basal metabolic functions, dietary-lipid metabolism and energy homeostasis. The NRs in red boxes are discussed in this review with a focus on their functional roles in circadian regulation, metabolism and breast cancer migration and metastasis.

including embryonic development, reproduction, also observed by Lin and coworkers (Lin et al. 2015). metabolism, homeostasis and cell proliferation. NRs In addition to ESR1, PGR and AR, expression levels of other are also known to play a role in various pathological NRs were shown to be associated with histological grade Journal of Molecular Endocrinology processes including cancer, inflammation and metabolic (THRB, NUR77, RORC, COUP-TF2 and LRH-1), to classify disorders (McKenna & O’Malley 2010) and as such have breast tissues (THRA, ESR1, NURR77, EAR2 and RARG) and been targets of intensive drug development for decades to predict metastasis-free survival in -treated (Ottow & Weinmann 2008). patients (THRB, COUP-TF2, MR and PPARG). Thirty-three of the 48 human NRs were also observed to be expressed NR expression in the normal breast and in stromal cells and four of these (RORA, THRB, VDR and breast cancer PPARG) were shown to have differential expression profiles in cancer-associated fibroblasts compared to normal Recent studies have begun to provide a global view of the breast adipose fibroblasts (Knower et al. 2013). Together, functional roles and expression of the NR superfamily these studies highlight that multiple members of the NR in primary normal breast (Muscat et al. 2013) and family are likely to play important roles in breast cancer breast cancer tissues (Muscat et al. 2013, Lin et al. 2015). growth and development and to have discriminant and Muscat and coworkers found that the majority of NRs prognostic value in breast cancer. are expressed in the breast (41 of 48). Many NRs showed differential expression in breast cancer compared to the NR expression in breast cancer subtypes normal breast indicative of potential involvement in breast cancer biology (Muscat et al. 2013). In particular, Breast cancer is a heterogeneous disease encompassing breast cancer is associated with the overexpression of the multiple subtypes with distinct molecular profiles, NR4A subgroup as well as EAR2, and pan-repression of therapeutic response and clinical outcomes (Prat et al. the majority of NRs relative to the normal breast. High 2015). Several NRs have been shown to either act expression of the NR4A subgroup in breast cancer was co-operatively with ESR1 and/or have differential

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology

Steroid hormonereceptors

Orphan nuclearreceptors

Non-steroid hormonereceptors Name Table 1

Adopted orphans Estrogen receptor Tailess-related receptor Testicular orphanreceptors Hepatocyte nuclearfactor4 Short heterodimericpartner DSS-AHC criticalregiononthe Steroidogenic factor-like Xreceptor Vitamin Dreceptor-like Xreceptor-like Rev-erbs RAR-related orphanreceptor Thyroid hormonereceptor 3-Ketosteroid receptor Chicken ovalbuminupstream Photoreceptor cell-specific Peroxisome proliferator- Vitamin Dreceptor Retinoic acidreceptor DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review X ,gene1 promoter- receptor activated receptor Human nuclearreceptors. Nomenclature NR1A2 NR3A1 NR2E1 NR2C2 NR2C1 NR2A2 NR2A1 NR0B2 NR0B1 NR5A2 NR5A1 NR2B3 NR2B2 NR2B1 NR1I3 NR1I2 NR1H4 NR1H3 NR1H2 NR1D2 NR1D1 NR1C3 NR2F2 NR1C2 NR1F2 NR1F1 NR1B3 NR1B2 NR1A1 NR3C4 NR3C3 NR3C2 NR3C1 NR2F1 NR2E3 NR1C1 NR1I1 NR1F3 NR1B1 NR3A2 t

b

© 2017SocietyforEndocrinology doan andothers Printed inGreatBritain Symbol THRB ESR1 TLX TR4 TR2 HNF4g HNF4a SHP DAX-1 LRH-1 SF-1 RXRG RXRB RXRA CAR PXR FXRa LXRa LXRb REV-ERBb REV-ERBa PPARG PPARD RORB RORA RARG RARB THRA AR PGR MR GR COUP-TF1 PNR PPARA VDR RORC RARA ESR2 COUP-TF2 ID 10062 10002 7068 2099 7101 7182 7181 3174 3172 8431 2494 2516 6258 6257 6256 9970 8856 9971 7376 9975 9572 5468 5467 6096 6095 5916 5915 7067 5241 4306 2908 7025 5465 7421 6097 5914 2100 7026 367

breast cancer Nuclear receptorsin

Published byBioscientifica Ltd. Ligands Thyroxine (T Estogens Not known All-trans retinoicacid All-trans retinoicacid Fatty acids Fatty acids CD437 Not known Phospholipids Phospholipids 9-cis-retinoic acid 9-cis-retinoic acid 9-cis-retinoic acid Androstanol, Bile acids Bile acids Oxysterols Retinoic acid Heme Heme Fatty acids Fatty acids Cholesterol, cholesteryl Oxysterols All-trans and9-cis All-trans and9-cis Thyroxine (T , Progesterone Cortisols Not known Benzimidazoles Fatty acids Calcitriol (1′,25 Retinoic acid All-trans and9-cis Estradiols /ATRA androstenol sulphate retinoic acid retinoic acid dihydrotestosterone (hydrocortisone) dihydroxy vitaminD3) retinoic acid triiodothyronine (T triiodothyronine (T 4 4 ), ),

3 3 ) ) Downloaded fromBioscientifica.com at09/27/202109:55:09AM vs normal Expression inBC Decreased Decreased Increased Not expressed Decreased Decreased Decreased Not expressed Not expressed Not expressed Decreased Not expressed Decreased Decreased Decreased Not expressed Decreased Not expressed Decreased Decreased Decreased Decreased Decreased Decreased Decreased Decreased Decreased Decreased Increased Expressed inBC Decreased Increased Decreased Decreased Decreased Decreased Decreased Decreased Increased Decreased 58 : 3 ESR1 expression Association with Yes Yes Yes

Yes Yes

Yes Yes Yes

Yes Yes Yes Yes

a,b a,b a,b a,b a,b a,b b b b b (Continued) 172 172 via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 173 breast cancer

Table 1 Continued.

Expression in BC Association with Name Nomenclature Symbol Entrez ID Ligands vs normal ESR1 expression ErbA2-related gene 2 NR2F6 EAR2 2063 Not known Increased Estrogen receptor-related NR3B1 ESRRA 2101 Isoflavones, Decreased receptor , chlordane NR3B2 ESRRB 2103 Isoflavones, Decreased diethylstilbestrol, 4-hydroxy tamoxifen NR3B3 ESRRG 2104 Isoflavones, Decreased Yesb diethylstilbestrol, 4-hydroxy tamoxifen Nerve IB-like NR4A1 NUR77 3164 Cyclosporone-B, Increased diindolylmethane analogs NR4A2 NURR1 4929 6-Mercaptopurine, Increased benzimidazoles, C-DMIs NR4A3 NOR1 8013 6-Mercaptopurine, Increased prostaglandin A2 nuclear factor NR6A1 GCNF 2649 Not known Decreased

aMuscat et al. (2013); bLin et al. (2015).

actions in ER+ vs ER− breast cancers. For instance, AR Similarly, the expression of GR has been shown has different roles in ER+ vs ER− breast cancers. AR has to be differentially associated with survival in been investigated extensively as a therapeutic target in ER+ vs ER− breast cancers. In ER+ breast cancers, ER− breast cancer. It was reported that in ER−/HER2+ high GR expression is associated with better survival breast tumors, AR stimulates oncogenic Wnt and HER2 (Pan et al. 2011, Abduljabbar et al. 2015b), whereas in signaling pathways thereby stimulating cancer cell ER− breast cancers, high GR expression is correlated with growth (Ni et al. 2011). However, the role of androgen poorer breast cancer-specific survival (Pan et al. 2011, signaling in ER+ breast cancer is less clear. Expression of Abduljabbar et al. 2015b). AR is associated with good prognosis in ER+ breast cancer Recent studies profiling the expression of NRs in Journal of Molecular Endocrinology (Hu et al. 2011, Hilborn et al. 2016), with anti-estrogenic ER+ and ER− cancers have also identified other NRs and anti-proliferative effects in ER+ breast cancer being whose expression levels differ significantly between ER+ reported (Peters et al. 2009, D’Amato et al. 2015). The and ER− breast cancers (Table 1). In particular, AR, PGR, anti-proliferative effect of AR is partly attributed to its PNR, RARA and THRB were reported to be significantly inhibitory effect on ESR1 signaling in ER+ breast cancer differentially expressed between ER+ and ER− breast cells (Peters et al. 2009, Fioretti et al. 2014). However, AR cancers (Muscat et al. 2013) and significantly associated has also been reported to contribute to therapy resistance with ESR1 expression (Lin et al. 2015). The expression of in ER+ breast cancer cells overexpressing AR, with increase DAX-1, LRH-1, PPARA, RORB, RORC and TLX were also in AR-to-ESR1 ratio associated with worse outcome for reported to be significantly associated with ESR1 expression tamoxifen-treated patients (Cochrane et al. 2014) and in multiple microarray datasets (Lin et al. 2015). increased activity of Tam in AR overexpressing cells. In this context, AR reportedly increased tamoxifen Mechanisms of NR action agonist activity via the activation of EGFR in ER+ breast cancer, and this was blocked by dual treatment with the Alterations in normal transcriptional programs are anti-androgen enzalutamide and EGFR inhibitor gefitinib a fundamental feature of cancer pathogenesis and (Ciupek et al. 2015). In addition, AR appears to promote progression. Nuclear receptors are important regulators proliferation of molecular apocrine breast cancer; a that directly couple small-molecule signaling with subset of TNBC expressing AR, through activation of transcriptional regulation. An understanding of their genes that are normally regulated by ESR1 in ER+ cancer mechanisms of action at the genomic and network level, (Robinson et al. 2011). and how these processes are altered in breast cancer, is of

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology genomic action in which NRs frequently act on long-range genomic actioninwhichNRs frequentlyactonlong-range these studiessuggestamore complexmodelofNR chromatin interactions (Liu & Cheung 2014 ). Together, by studiesontheestrogen receptor-mediated long-range regulation thatinvolveschromatin loopingasillustrated NRs arecapableofmediatinglong-rangetranscriptional 2011).Furthermore, intestinal epithelialcells(Surjit et al. confers transactivationtoligandedGRinepidermisand canonical glucocorticoidresponseelements(GRE)that (nGRE) thatconferstransrepressioninadditiontothe (GR) iscapableofbindinganegativeresponseelement responses ofNR.Forexample,theglucocorticoidreceptor elements can be a mechanism to fine tune transcriptional ( in ameta-analysisofNRcistromesdifferentcelltypes canonical motifandhalf-siterecognitionisalsoconfirmed but alsotodistalregionsandtheprevalenceofnon- that NRslocalizenotonlytoproximalpromoterregions al.2013).Theobservation et (Kittler NRs beingobserved localization, withbothproximal-biasedanddistal-biased and foundthatNRvariedsignificantlyintheirgenomic distribution ofthebindingsites24NRsinMCF-7cells 2006). Kittlerandcoworkersexaminedthegenomic not contain the canonical ESR1-binding motif (Carroll et al. distal regionsofthegenomeandthatmanyESR1sitesdid cancer celllineshowedthat most ESR1binding occurs at mapping ofESR1genomiclocalizationintheMCF-7breast or ChIP-Seq. In the context of breast cancer, genome-wide nuclear receptors and transcription factors using ChIP-chip that profiled the genome-widebinding sites ofseveral classical modelshavebeenchallengedbyrecentstudies orientation (Mangelsdorf recognition ofstringentpairedhalf-siteswithhighlyspecific bind proximallytothepromotersoftargetgenes,through hormone-responsive genesgaverisetothemodelthatNRs Traditionally, studiesonnuclearreceptoractionat Shifts from the‘classical’modelofNRaction in breastcancermadepossiblebythesegenomic methods. section, wereviewnewinsightsofNRmechanismsaction networksinbreastcancer.receptor regulatory Inthissub- nuclear receptorsandhighlightthecomplexityof revealed newinsightsintothemechanismsofaction wide manner. Studiesemployingthesemethodshave genomic locationsofNRbindinginanunbiased,genome- chip, ChIP-SeqandGRO-Seqhasenabledprofilingofthe cancer development. fundamental importancetoourunderstandingofbreast ). NR binding at non-canonical response 2011).NRbindingatnon-canonicalresponse Tang et al. DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review The developmentoftechnologiessuchasChIP- t al.1995).However, these et t

b

© 2017SocietyforEndocrinology doan andothers Printed inGreatBritain

with PGR-bindingsitesthat showstrongnucleosome occurs, and progesterone-responsive genesare associated nucleosomes containingfunctional PGR-bindingsites occupancy. Upon hormonetreatment,remodelingof with DHSregionsthatstill exhibithighnucleosome that PGRbindinguponhormonestimulationcorresponds (MNase) followedbynext-generationsequencingshowed Profiling ofthePGRbyDNaseandMicrococcalnuclease al. 2013). et affinity estrogenresponseelements(Gertz characterized byinaccessiblechromatincontaininghigh- ESR1-binding sitessharedbymultiplecelltypesare correspond tocelltype-specificgeneregulation,whereas co-occurrence ofothertranscriptionfactorbinding,and binding sitescoincidewithpre-existingopenchromatin, ESR1 revealedasimilarpattern.ThemajorityofESR1- al. ). Profilingof 2015 et range interactions(Kuznetsova the formationofactiveenhancersandincreasedlong- subset of sites containing the consensus GRE results in occurs atpre-accessiblechromatin.BindingofGRtoa that, similartobreastcells,themajorityofGRbinding and afterdexamethasoneinductioninHeLacellsfound 2011). Arecentstudyoflong-rangeinteractionsbefore al. et hormone-induced remodeledchromatin(John DHS sites,whereasonly~20%ofGRbindingoccursat majority (>70%)ofGRbindingoccursatpre-induction hypersensitive sites(DHS)inbreastcellsrevealedthatthe of NRaction.Genome-widescreeningDNAseI induction vspost-inductionchromatinasadeterminant several groupshaveinvestigatedthequestionofpre- conformational changes induced uponligand binding, widely accepted model of NR binding to DNA only after cell type-specifictranscriptionalregulation.Duetothe NR–chromatin interactionisamajordeterminantofNR regulates aswellisregulatedby, NRactions,andthat existing inadynamiccontinuumofcondensationstates, landscape ofthecell.Itisbecomingclearthatchromatin, and complexinteractionsbetweenNRthechromatin through advancesingenomictechnologiesisthedynamic section), anothermajoraspectofNRactionilluminated transcriptional responseofNRs(discussedinthenext of coregulatorsindeterminingthecelltype-specific Clarke &Graham2012).Inadditiontotheinfluence 2008, of thesameNRindifferentcelltypes(Krum et al. remarkable differenceinthegenome-widebindingprofiles Studies investigatingNRcistromeshighlightthe Cell typespecificityandthechromatin landscape of differentresponseelements. enhancers whoseactionscanbefine-tunedbyrecognition breast cancer Nuclear receptorsin Published byBioscientifica Ltd.

Downloaded fromBioscientifica.com at09/27/202109:55:09AM 58 : 3 174 174 via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 175 breast cancer

remodeling upon hormone induction (Ballare et al. 2013). SRC to E2-stimulated genes while the absence of SRC The chromatin landscape of the cell therefore plays an was observed at E2-repressed genes (Kininis et al. 2007). important role in determining the genomic binding In a subsequent study, Zwart and coworkers mapped the profiles of specific NRs. Although some NRs such as genome-wide binding sites of several ESR1 coregulators ESR1and GR rely mostly on pre-programmed sites made (SRC1, SRC2, SRC3, p300 and CBP) in MCF-7 cells and accessible by other cell- and tissue-specific pioneer DNA- showed a complex network of ESR1-coregulator binding, binding factors, other NRs such as PGR require ligand- with preferential binding sites for each coregulator. dependent active . They identified a subset of ESR1-regulated genes that are co-occupied by SRC3, but not SRC1 or SRC2, that predicts poor or good survival outcome depending on whether Pioneer factors and coregulators as modulators of the genes were upregulated or downregulated (Zwart et al. NR function 2011). Lupien and coworkers also identified association Pioneer factors are transcription factors that can bind between a subset of E2-induced ESR1 binding sites condensed chromatin and influence transcription by co-occupied by CARM1 and gene repression, independent facilitating subsequent recruitment of other transcriptional of the presence of FOXA1 binding. They also identified regulators. Recent studies have highlighted the importance another subset of E2-induced, FOXA1-independent, ESR1 of pioneer factors in facilitating chromatin accessibility and and CARM1 co-occupied sites that result primarily in also in determining the cell type-specific response of NRs. repression of expression (Lupien et al. 2009). These studies This is exemplified by the dependence of ESR1 on FOXA1 demonstrate that selective interaction with different in breast cancer. FOXA1 expression is luminal restricted coregulators at specific subsets of target genes may be an (Perou et al. 2000) and is thought to function as a pioneer important mechanism through which the specificity of factor (Cirillo et al. 2002). FOXA1 has been observed to NR action is modulated. influence genome-wide chromatin accessibility and is Due to their central role in regulating NR-mediated a key determinant of cell type-specific ESR1 genomic transcription, many coregulators are known to be localization (Lupien et al. 2008, Hurtado et al. 2011), involved in human diseases (Lonard et al. 2007, Lonard reflecting the importance of interaction with other factors & O’Malley 2012). In the breast, changes in expression on control of cell specificity of ESR1. Similarly, AP-1 has levels of coregulators have been implicated in the tissue- been shown to recruit GR to regions of the genome that specific response to tamoxifen (Keeton & Brown 2003). are basally accessible prior to hormonal treatment. AP-1 The majority of coregulators show differential expression

Journal of Molecular Endocrinology is thought to maintain chromatin accessibility for GR between breast cancer and normal breast tissues, as well as binding as well as recruiting GR indirectly in the absence between ER+ and ER− breast cancers. This is accompanied of a canonical GR motif (Biddie et al. 2011). by changes in the expression correlations between subsets In addition to pioneer factors, coregulators interact of coregulators and specific NRs, whose expression showed with NRs and other transcription factors to facilitate prognostic value in breast cancer (Doan et al. 2014). transcription of target genes. According to the Nuclear Coregulators are therefore critical determinants of Receptor Signaling Atlas (NURSA: www.nursa.org), there NR-mediated transcriptional regulation in the breast and are 320 NR coregulators identified to date. However, this disruptions in the normal NR–coregulator interaction number is likely to be a gross underestimation according network are potentially an important aspect of breast to a recent proteomic study (Malovannaya et al. 2011). cancer biology. Coregulators can assist in transcriptional regulation of NRs by facilitating chromatin accessibility, stabilizing Combinatorial control of by NRs in NR–DNA interaction or facilitating indirect NR–DNA breast cancer interaction through tethering (McKenna & O’Malley 2002, Millard et al. 2013). Recent large-scale integrative analyses of transcription Selective recruitment of coregulators to subsets of factor genomic localization data from the ENCODE NR-binding sites can also be a mechanism through which project highlighted the extraordinary complexity of transcriptional regulation of subsets of NR target genes are transcriptional regulation and the extensive functional fine-tuned. An early study of ESR1 and steroid receptor crosstalk between transcription factors (ENCODE Project (SRC) binding using promoter tiling Consortium 2012, Gerstein et al. 2012, Xie et al. 2013). arrays showed E2-dependent recruitment of ESR1 and This observation also holds for NR in breast cancer.

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology ESR1-binding events,resulting inageneexpressionprofile shown to be recruited to the ESR1 complex and to redirect presence ofbothestrogen and progesterone,PGRwas share genomicbindingsites withPGRandLRH-1.Inthe between theseNRs. time, targetandcell-specificmodesofco-operation co-operative andantagonistic,perhapsreflectingdifferent GR, thenatureoftheirinteractionisreportedtobeboth ESR1 co-occupies many target genes with RARA as well as 2013).Therefore,although target genes(Karmakar et al. estrogen receptor-mediatedtranscriptionalactivation of with displacementofESR1leadingtotherepression of dexamethasone, GRrecruitmenttothesesitesisassociated several ESR1-bindingsitesincellstreatedwithbothE2and MCF-7 cellsrevealedthat,althoughGRco-occupies study profilingESR1andGRgenomiclocalizationin 2013). Another to DNAfortheotherNR(Miranda et al. facilitates chromatinremodelingtherebyenablingaccess assisted loadingmechanism,inwhichbindingofoneNR significant co-operationofthesetwoNRs,throughan epithelialcelllinesrevealed and GRinmousemammary Innes or co-operativeisstillcontroversial(Hua profiles, althoughwhethertheinteractionisantagonistic similarbinding showed thatESR1andRARAsharevery (Fontana et al. 1992).StudiesofESR1andRARAcistromes and antagonizethegrowthstimulationeffectofestrogen been knowntoinhibitproliferationinbreastcancercells action in breast cancer cells. For example, retinoic acid has including otherNRs,areimportantdeterminantsofESR1 not act on its own and that other transcription factors, tumors. Mounting evidence indicates that ESR1 does to drive growth and proliferation in the majority of breast comparing theoverlapoftheircistromes.ESR1isknown various other NRs in breast cancer cells by individually convergence ingenomicbindingbetweenESR1and regionsinbreastcancercells. active regulatory relevant genesandarehypothesizedtobeimportant These regions are also enriched with breast cancer- chromatin marksandincreasedaccessibility. elements,includingactive with activeregulatory These HOTregionsareenrichedwithfeaturesassociated coordinately occupiedbymultipleNRs(HOTregions). connected andtherearemanyregionsinthegenome They showedthattheresultingnetworkishighlyinter- associated coregulators in MCF-7 breast cancer cells. map fromcistromicdataof24NRsand14breastcancer- Kittler and coworkers (Kittler DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review In additiontoRARAandGR, ESR1wasreportedto On asmallerscale,otherstudieshavealsorevealed t al.2010).Likewise,cistromicprofilingofESR1 et et al. 2013)builtaregulatory t

b

© 2017SocietyforEndocrinology doan andothers et al. 2009, Printed inGreatBritain Ross- change inESR1actionwas showntobeinfluencedby in sampleswithdifferent outcomes.Thisdynamic process, withdistinctESR1-binding regionsobserved they showedthatESR1binding isahighlydynamic insampleswithgoodprognosis.Further,was observed metastatic samples,whereaslowerESR1signalintensity highsignalintensityatESR1-binding sitesin observed al.2012).They et and cancerprogression(Ross-Innes to directlyexplorethecorrelationbetweenESR1binding seq ofESR1inclinicalsampleswithdifferentprognoses outcomes. Ross-InnesandcoworkersperformedChIP- with breastcancerdiseaseprogressionandclinical in NRtranscriptionalprogramsandtheirassociation Recent studieshavehighlightedtheeffectofchanges type-specific changesinPGRtranscriptionalregulation. Graham 2012),reflectingcancer-associatedandcell breast cellsdisplayremarkablylowoverlap(Clarke& breast cancer cells and AB-32 immortalized normal profiles. ThetranscriptionaltargetsofPGRin T-47D disease-associated shiftsintheirgenomicbinding factors displayingtemporal,celltypespecificand regulation isahighlydynamicprocesswithtranscription It isbecomingincreasinglyevidentthattranscriptional Reprogramming ofNRbinding anddiseaseprogression have thepotentialtoleadnoveltherapeuticstrategies. the functionalinteractionsbetweenNRsinbreastcancer is stilltobedetermined;nevertheless,theseinsightsinto supporting thisasamechanismcommontootherNRs particularly a feature of ER or whether data will emerge studies sofarreflecttranscriptionalplasticitythatis analyses in unraveling this complexity. Whether the networks andtheutilityofintegrated,genome-wide particularly betweenERandotherNRs,ingeneregulatory responsive genesinMCF-7cells(Lai et al.2013). of ESR1andsynergisticallyregulateasubsetestrogen- substantial portion(~35%)ofitsbindingsiteswiththat are yettobeestablished.LRH-1wasreportedsharea line models,andtheimplicationsforbreastcancerinvivo Furthermore, themajorityofdataarederivedfromcell PGR andtheircrosstalkarehighlycontextdependent. (Daniel et al. 2015).ItislikelythattheactionofESR1and in amoreaggressiveproliferativeresponsetoestrogen transcriptional complexwithESR1andPELP1,resulting by actingasamolecularscaffoldfortheformationof un-liganded PGRBactivatedasubsetofERtargetgenes 2015). However, inthepresenceofestrogenalone, associated withbetterclinicaloutcome(Mohammed et al. breast cancer Nuclear receptorsin Published byBioscientifica Ltd. These studies highlight the complexityof cross talk,

Downloaded fromBioscientifica.com at09/27/202109:55:09AM 58 : 3 176 176 via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 177 breast cancer

the action of other NRs, such as PGR (Mohammed et al. Emerging functional roles of NR in BC 2015), as well as the signaling context leading to It is now clear that a number of NRs are implicated in ESR1 activation. For example, ESR1 displayed distinct breast cancer growth and development (Conzen 2008). It genomic binding profiles depending on whether ESR1 is emerging that, in addition to influencing was activated by estrogen or through the epidermal and proliferation, NRs also play important roles in other growth factor (EGF) pathway (Lupien et al. 2010). The aspects of breast cancer biology. Table 2 summarizes EGF-induced ESR1 cistrome specifically regulates genes reported involvement of NRs in selected aspects of breast that are overexpressed in ERBB2-positive breast cancers cancer biology. This section outlines some of the emerging and associated with poor clinical outcomes. The EGF functional roles of NRs in breast cancer. pathway can therefore be an alternative for ESR1 signaling in breast cancer and provides a molecular explanation for the endocrine therapy resistance often Nuclear receptors as key components of the seen in ER+ERBB2+ breast cancers. Analysis of ESR1 and ESRRA cistromes in tamoxifen-sensitive vs -resistant breast cancer cells showed that despite regulating A is any biological process that displays distinct transcriptional networks, their cistromes are an endogenous, entrainable oscillation of roughly reprogramed in tamoxifen-resistant breast cancer cells 24 h, driven by a self-sustained timekeeping mechanism, toward the regulation of genes functionally relevant the circadian clock. The circadian clock governs many to resistance (Thewes et al. 2015). These studies show aspects of human physiology, including body temperature, that NR binding profiles can be shifted in a context- sleep–wake cycle, blood pressure, hormone secretion and dependent manner, resulting in altered transcriptional metabolism. In line with this, at least 10% of all genes networks that affect disease progression and outcome. are under the control of the circadian system and display

Table 2 NR function in breast cancer.a

Pro-oxidative Circadian Anti-proli­ Pro- Anti- phosphoryla- Pro- Anti- Pro- Pro- involve- Nomenclature Symbol ferative apoptotic migratory tion proliferative apoptotic migratory glycolytic ment NR3A1 ESR1 Y Y Y NR3A2 ESR2 Y Y Y NR3C1 GR Y Y Journal of Molecular Endocrinology NR3C3 PGR Y Y Y Y NR3C4 AR Y Y NR1B1 RARA Y Y NR1B2 RARB Y Y Y NR1B3 RARG Y Y NR1F1 RORA Y NR1F2 RORB Y NR1F3 RORC Y NR1I1 VDR Y Y NR1C1 PPARA Y NR1C2 PPARD Y NR1C3 PPARG Y Y Y Y NR1D1 REV-ERBa Y Y Y NR1D2 REV-ERBb Y Y Y NR2B1 RXRA Y NR2B2 RXRB Y NR2B3 RXRG Y NR2F1 COUP-TF1 Y NR2F2 COUP-TF2 Y NR3B1 ESRRA Y Y NR3B3 ESRRG Y Y NR4A1 NUR77 Y

aNR effect on cancer-related biological processes based on studies in breast primary tissue or cell lines. The influence of NR on breast cancer proliferation, apoptosis, migration, metabolism as well as involvement in circadian regulation/response is tabulated. Y, indicates reported involvement.

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology cortosol, estrogen),nutrientsignals (e.g. derivativesoffattyacidsandretinoids)cellularredoxstatus(NADH/NAD+ ratio). feedback ontotheclockgenesthemselves. NRregulationofclockgenesalsorenderstheresponsiveto numerouscirculatinghormones(e.g. NRs (showningreen)interactclosely withthecircadianfeedbackloopsandareresponsivetoclock(exhibit rhythmicexpression)andableto most notableinterlockingloopisthat involvingthenuclearhormonereceptors(NRs),REV-ERB andROR.Inaddition toREV-ERB andROR,severalother and physiology. Actingontheprimaryfeedback loopareauxiliaryloops,whichappeartoincreasethestability androbustnessoftheoscillations.The timing anddrivestherhythmicexpression ofseveralclock-controlledandclock-modulatedgenes,whichin turn mediatecircadianrhythmsinbehavior the nucleusandrepresstheirowngenetranscriptionthroughdirectinteraction withtheCLOCK/BMAL1complex.Thisfeedbackcycleprovidesnear24-h CRYPTOCHROME (CRY1 andCRY2) astranscriptionalrepressors.AslevelsofcytosolicPERandCRY proteinsrise,theyassociate,translocateto model involvesaprimaryloopwithCLOCK(orhomologueNPAS2) andBMAL1astranscriptionalactivators,(PER1,PER2PER3) consists ofacomplexcircuitrytranscriptional/translationalregulatory feedbackloops(clockcomponentsshowningray).Inmammals,thecurrent dysfunction indisease,pages191–198,copyright(2010),withpermission fromElsevier).Themolecularmachinerythatprovidescircadiantimekeeping The molecularclockmachinery(reprintedfromTrends inPharmacologicalSciences , volume31BechtoldDA,GibbsJE&LoudonAS,Circadian Figure 2 with permissionfromElsevier).RORsrecognizethesame al.(2010), et 2,reusedfromBechtold and RORs(Fig. that involvemultipleNRs,mostprominentlyREV-ERBs that the circadian loops clock contains other auxiliary 2).Itisnowemerging co-repressors, PERandCRY (Fig. transcription factorsCLOCKandBMALtheir relatively simplefeedbackloopconsistingofthekey and breastcancer. growing bodyofevidencelinkingcircadian disruption between NRs and circadian regulation, as well as the including cancer. Here,wediscusstheemerginglink disruption tovariousclinicalandpathologicalconditions 2005). Thereismountingevidencelinkingcircadian 2002, circadian oscillation(Akhtar et al. DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review Initially, thecircadian clockwasdefinedtobea t

b

© 2017SocietyforEndocrinology doan andothers 2003, Oishi et al. Printed inGreatBritain (Zhao et al. 2014). display circadian expression in various metabolic tissues circadian clockisextensive,withatleast35NRsknownto The listofotherNRspotentiallyundercontrolthe al. 2011). et al.2010)andESRRA(Dufour et Campbell by thecircadian clock,mostnotablyGR(Conway- have beenshowntoregulateaswellbeingregulated (Preitner et al.2002). machinery the pivotalroleofREV-ERBs intheuniversal circadian deletion studies and cistrome analyses have confirmed of clockgeneexpression(Akashi&Takumi 2005).Gene have demonstratedtheirimportanceintheregulation elementsasREV-ERBs,regulatory andarangeofstudies breast cancer Nuclear receptorsin Published byBioscientifica Ltd. In additiontoREV-ERBs andRORs,severalotherNRs

Downloaded fromBioscientifica.com at09/27/202109:55:09AM 58 : 3 178 178 via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 179 breast cancer

To date, epidemiological studies reported mixed ESR1 has been shown to be an important regulator findings on the association between increased risk of linking the circadian system and breast cancer breast cancer and circadian disruption in the form tumorigenesis, acting as a regulator of (as well as being of night shift work. Although a few studies reported regulated by) the core clock genes. ESR1 appears to no significant association between breast cancer and participate in a feedback loop involving the clock protein night work (O’Leary et al. 2006, Pronk et al. 2010), PER2, which was shown to be a tumor suppressor in the majority of recent studies reported significant or luminal breast cancer. Although PER2 is E2 inducible, borderline associations, with the strongest association PER2 itself is thought to mediate ESR1 degradation found with long-term exposure to night work (Hansen through the proteasome pathway. Suppression of PER2 2001, Schernhammer et al. 2001, 2006, Hansen & Lassen leads to ESR1 stabilization (Gery et al. 2007). In addition, 2012, Hansen & Stevens 2012, Knutsson et al. 2013, ESR1 was shown to bind estrogen response elements in the Menegaux et al. 2013, Rabstein et al. 2013, Akerstedt et al. promoter region of CLOCK leading to the upregulation 2015, Papantoniou et al. 2015). Meta-analysis studies have of CLOCK in breast cancer cells. Knockdown of CLOCK also reported a positive association between night work attenuated proliferation in breast cancer cells (Xiao et al. and breast cancer risk (Jia et al. 2013) and that a positive 2014). Therefore, CLOCK appears to be an important dose–response gradient is observed for breast cancer with mediator of the proliferative effect of E2 in breast cancer. increased years of night shift and cumulative night work In summary, NRs play important regulatory roles in (Wang et al. 2013). both the universal and tissue-specific circadian systems. Although circadian disruption is prominently linked In particular, REV-ERBs and RORs are integral regulators to breast cancer tumorigenesis through epidemiological of the universal circadian clock, and various other NRs studies, the molecular mechanisms and NRs involved exhibit circadian expression. Although epidemiological are still largely unknown, with most published studies studies have linked circadian disruption and breast cancer focusing on the ESR1 as a link between the circadian system tumorigenesis, a lot remains to be learned about the and breast cancer development. Many studies suggest that underlying molecular mechanism. Lowered melatonin lowered melatonin level due to exposure to night light due to night light exposure is thought to play a role and contributes toward breast cancer development and drug there is evidence supporting the involvement of ESR1 resistance. Melatonin is thought to exert its oncostatic as an integral regulator linking circadian regulation and effect through multiple mechanisms including anti- breast cancer development. , anti-angiogenesis, anti-oxidation and regulation

Journal of Molecular Endocrinology of immune response (Viswanathan & Schernhammer Regulation of energy metabolism 2009). The oncostatic effect of melatonin in breast cancer is thought be mediated through its anti-estrogenic actions As the initial observation by Warburg that proliferating as well as its role in regulating the activity of aromatases, tumor cells converted the majority of their glucose to the enzymes responsible for the local biosynthesis of lactate even in oxygen-rich condition and his hypothesis from androgens (Cos et al. 2006). Recent studies that altered metabolism is a characteristic of cancer using MCF-7 breast cancer xenografts in mice showed cells, metabolic reprogramming has now attained cancer that suppression of melatonin production by dim light hallmark status (Ward & Thompson 2012). Specifically, exposure at night leads to both tamoxifen (Dauchy et al. the Warburg effect describes a shift toward aerobic 2014) and doxorubicin resistance (Xiang et al. 2015). glycolysis in preference to oxidative phosphorylation as In addition to the effect of melatonin, there is the major means of ATP generation in cancer cells, even evidence linking the core clock genes as well as the REV- in oxygen-rich conditions. Although aerobic glycolysis ERBs with breast cancer growth and metastasis. The is not as efficient as oxidative phosphorylation (yielding rhythmic expression of the PER genes are reported to be only 2 mol of ATP/mole of glucose instead of 36 mol ATP/ altered in primary breast tumors (Chen et al. 2005) as well mole of glucose), it has certain benefits for tumor cells. as in breast cancer cell lines (Xiang et al. 2012). A recent Glycolysis results in more rapid production of ATP, fatty study demonstrated the association of the core clock gene acids and nucleotides that are needed by proliferating BMAL2 in ER breast cancer metastasis (Ha et al. 2016). tumor cells that need to double their biomass to divide. Furthermore, activation of REV-ERBA and REV-ERBB by a Furthermore, the generation of lactic acid promotes synthetic ligand is anti-proliferative in breast cancer cells. a tumor microenvironment that is protective against

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology related receptors (ESRRA/NR3B1, ESRRB/NR3B2 and Estrogen-related receptors (ERRs) level ofeachisoform. in aparticularcelltypedepends ontherelativeexpression transcriptional regulation,andfunctionalpredominance REV-ERB isoforms probably have redundant functions in 2010). These studies suggest that the two (Kourtidis et al. function ofREV-ERBA inERBB2-positive breastcancercells co-expression ofREV-ERBA andERBB2pro-survival negative cells,consistentwithapreviousstudyreporting have higherexpressionofREV-ERBA comparedtoERBB2- However, ERBB2-positive breastcancercellsappearto cells whileknockdownofREV-ERBA hadnoeffect. circadian andmetabolictargetgenesinbreastcancer REV-ERBB resultedintheenhancedexpressionofboth innormalcells.Knockdownof the oppositeisobserved abundant incancercellscomparedtoREV-ERBA, whereas cancer (DeMei et al. 2015).REV-ERBB issignificantlymore and metabolicpathwaysincancercells,includingbreast the dominantisoformcontrollingbothcircadian rhythm a dominantroleinnormalcells,REV-ERBB seemstobe recent publicationsuggeststhatalthoughREV-ERBA has al. 2015). A et type-specific transcription factors (Zhang the recruitmentofHDAC3,whichistetheredbycell REV-ERBA toitscognatesite,metaboliccontrolinvolves circadian regulationrequires directDNAbindingof to circadian controland metaboliccontrol.Although of generegulationemployedbyREV-ERBA withregard ERBA invarioustissuesreporteddifferentmechanisms A recentstudylookingatgenome-widelocationofREV- 2012, pathways (Cho et al. as integratorsofcircadian regulationandmetabolic roles incircadian regulation, REV-ERBs arerecognized REV-ERBs of therolesNRsinbreastcancercellularmetabolism. tumors cells.Here,wereviewourcurrentunderstanding intimately linkedtothemetabolicreprogrammingof Their alteredexpressioninbreastcancersispotentially downstream pathwaysinvolvedinglucosemetabolism. from regulatingglucosetransporterstocontrollingthe many ofthepathwayscontrollingenergymetabolism, act asatriggerfortheWarburg effect. 2010);hence,glucosefluxcanpotentially (Vazquez et al. glycosis inpreferencetooxidativephosphorylation accumulation ofglucoseactuallypromotesaerobic been suggestedthroughglucosefluxmodelingthat al.2012).Ithasalso et immune attack(Calcinotto DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review It hasemergedthatNRsarecentrallyplacedin In addition and related to their prominent Gerhart-Hines &Lazar2015). t

b

© 2017SocietyforEndocrinology doan andothers Printed inGreatBritain The estrogen- All three ESRR isoforms were identified ascoactivating and themajorityofgenes intheglycolyticpathways. (Cai a complexwithMYCto co-regulate glycolyticgenes regulated byMYC.ESRRA was foundtoparticipatein and modulators ofmetabolicpathwayssuchasMYC, genes (Dufour et al.2007). regions of many shared target and bind to regulatory and thatESRRAESRRGreadilyformheterodimers of enzymes in the glycolytic pathways (Cai overexpression ofESRRGalsoleadstoincreasedexpression probably involvesmuchgreatercomplexity, giventhat metabolic reprogrammingbyERRsinbreastcancers the tricarboxylicacidcycle(Eichner et al. 2010).However, through regulation of genes that control phosphorylation al.2013),ESRRG is thought to sustain oxidative et (Cai of enzymesthatregulatetheglycolysispathway promotes aerobic glycolysis through the upregulation cancer cells (Deblois& Giguere 2013).Although ESRRA in the regulation of metabolic reprogramming of breast suggested thatESRRAandESRRGhaveoppositeroles of favorableoutcome(Ariazi is correlatedwithESR1andERBB4,whicharemarkers poor outcome(Thewes ESRRA isassociatedwithincreasedrecurrenceand 2002).Although outcomes(Ariazi et al. distinct survival Giguere 2011). of mitochondrialbiogenesisandfunction(Eichner& regionsof705genesinvolvedinallaspects regulatory Together, thethreeERRisoformsrecognizeproximal and metabolicgenenetworks(Dufour co-operate withPROX1andBMAL1toregulatecircadian 2004).Intheliver, ESRRAwasreportedto (Mootha et al. genesinmousemyoblastcells oxidative phosphorylation positive feedbacklooptoregulatetheexpressionofmany gamma coactivatoralphagene(PGC-1α)inadouble- with theperoxisomeproliferator-activatedreceptor- al.2007).ESRRAwasreportedtoco-operate et Dufour across themitochondrialmembrane(Alaynick et al. 2007, of energysubstrates,productionandtransportATP ESRRG co-operatetocontrolgenesinvolvedinuptake of metabolicgenenetworks.Intheheart,ESRRAand maintaining energyhomeostasisthroughtheregulation 2013). BothESRRAandESRRGhavedominantrolesin high energydemand(Giguere2008, regulators ofenergymetabolisminmultipletissueswith ESRRG/NR3B3) havebecomerecognizedasthemaster breast cancer Nuclear receptorsin Published byBioscientifica Ltd. ESRRs alsointeractwithoncogenesthatareknown In thebreast,ESRRAandESRRGareassociatedwith et al. 2013).HIFdirectlyactivatesglucose transporters ERBB2. Manygenesintheglycolyticpathwaysare

Downloaded fromBioscientifica.com at09/27/202109:55:09AM t al.2015), et t al.2002).Ithasbeen et 58 Deblois &Giguere ESRRG expression : 3 t al.2011). et t al. 2013) et 180 180 HIF via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 181 breast cancer

factors of HIF and to enhance HIF-induced glycolytic and on the cell compartment in which PPARG is activated angiogenic gene expression in hypoxic condition (Ao et al. though, with activation in cancer cells resulting in growth 2008). ESRRA also regulates the expression of ERRB2 inhibition while activation in stromal cells results in (Deblois et al. 2010), which was reported to translocate growth enhancement of co-injected breast cancer cells to the mitochondria of tumor cells and regulate energy (Avena et al. 2013). Recent studies have also suggested an metabolism. Overexpression of mitochondrial ERBB2 involvement of PPARG and energy metabolism in breast decreases mitochondrial electron transport chain activity cancer. Firstly, several genes controlling the glycolytic and enhances cellular glycolysis (Ding et al. 2012). In pathways are found to have peroxisome proliferator addition, ERBB2 influences the glycolytic response element (PPRE) suggesting their regulation by through upregulation of lactate dehydrogenase (Zhao et al. PPARs (Sakharkar et al. 2013). Secondly, overexpression of 2009) as well as REV-ERBA, an NR that was reported to be PPARG in fibroblasts increased the production of l-lactate involved in glycolysis and fatty acid synthesis in breast and mitochondrial dysfunction. In addition, PPARG cancer cells (Kourtidis et al. 2010). induces the activation of HIF1A, a transcription factor In summary, the ERRs are prominent regulators that promotes glycolysis (Avena et al. 2013). of gene networks controlling glycolysis and oxidative phosphorylation in metabolic tissues. In the breast, Control of breast cancer migration and metastasis ESRRA expression is associated with poor outcome and appears to be involved in reprogramming the cell to Breast cancer metastasis accounts for the majority of favor aerobic glycolysis, a metabolic state characteristic of deaths from breast cancer. Early detection and a deeper cancer cells. ESRRG, on the other hand, is associated with understanding of the metastatic process are critical to better outcome and is involved in maintaining oxidative develop therapeutic interventions. Metastasis involves a phosphorylation, a metabolic state characteristic of complex cascade of steps starting with invasion by the normal cells. However, given that ESRRA and ESRRG can primary tumor of the surrounding host tissues, followed by form heterodimers and readily bind to many shared target intravasation and dissemination of the tumors cells via the genes, the complexity of the mechanisms for controlling blood or lymphatic system, infiltration and colonization their specificity of action is still to be deciphered. at the distant site. Several factors and pathways are known to influence cancer metastasis including disintegration Peroxisome proliferator-activated receptors of cell-to-cell adhesion, proteolytic remodeling of the (PPARs) PPARs function as heterodimers with the extracellular matrix, cell motility, evasion of immune and Journal of Molecular Endocrinology retinoid receptors (RXR) and regulate various genes that apoptotic signals and angiogenesis. Although substantial are involved in lipid metabolism and energy homeostasis. information is known about the process of metastasis, the Through these pathways, PPARs influence various molecular basis of breast cancer metastasis remains poorly cancer-related cellular processes such as proliferation, understood. NRs, being master regulators of almost every differentiation and survival (Michalik et al. 2004). Three physiological aspect of life, are likely to be intimately PPAR isotypes (PPARA/NR1C1, PPARD/NR1C2 and involved in the metastasis process. Here, we summarize PPARG/NR1C3) are known, each with unique expression the current understanding of the roles of various NRs in profile and functional roles. The PPARs showed decreased biological pathways affecting breast cancer metastasis. expression in breast cancer compared to normal breast (Muscat et al. 2013). The role of PPARD in breast cancer Estrogen receptors The estrogen receptors alpha tumorigenesis is unclear. Although activation of PPARD (ESR1) and beta (ESR2) appear to have distinct roles in has been shown to inhibit breast cancer cell line breast cancer progression and metastasis, with reports of tumorigenicity (Yao et al. 2014), inhibition of PPARD ESR1 signaling enhancing breast cancer cell migration and by inverse was reported to inhibit breast cancer invasion, whereas ESR2 signaling has the opposite effects. cell invasion (Adhikary et al. 2013). PPARG is the best Emerging evidence suggests that ’s positive studied of the 3 PPAR subtypes and has been reported effect on cell migration and invasion appears to be to have an anti-proliferative effect on breast cancer cells mediated through ESR1’s extranuclear signaling. ESR1 (Pon et al. 2015) and is associated with better outcomes extranuclear signaling is mediated through the PI3K, based on survival analysis of PPARG protein expression MAPK and c-Jun N-terminal kinase pathways (Li et al. level in BC tissue microarrays (Abduljabbar et al. 2015a). 2010, Zheng et al. 2011) with the involvement of PELP1 The downstream effect of PPARG seems to be dependent (Chakravarty et al. 2010), focal adhesion kinase (FAK)

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology signaling involvingESR1 (an ESR1variant),itwassuggestedthatnon-genomic unclear mechanisms. while ESR2exertstheopposite effectthroughasyet to regulatecelladhesionand cytoskeletalremodeling and pro-invasiveeffectsthrough extranuclearsignaling emerging pictureisthatofESR1exertingpro-migratory divergent effectsofsubtypevariantsandcrosstalk.The having opposingroles,whichisfurthercomplicatedby cancer cellmetastasisiscomplexwithdifferentisoforms the effectsofestrogenreceptorsignalingonbreast 2014).Insummary, lymph nodemetastasis(Rosin et al. tumors, wasreportedtocorrelatewithhigherrisk of expression ofanESR2variant,ESR2cxinprimary al.2012).Furthermore, et pro-invasive effect(Ohshiro andactivation of p-ERK1/2 to have pro-migratory 2000, al. et less invasiveandproliferativetumors(Jarvinen cancer cells(Chakravarty et al.2010). resulted inenhancedmotilityandmigrationofbreast formation ofrufflesandfilopodia-likestructures,which cells suchasdynamicactincytoskeletonremodelingand E2-mediated signalingpathwaysinducefeaturesofmotile signaling influencesthecytoskeletalorganization.These reported aspossiblemechanismsthroughwhichESR1 of theactin-bindingproteinezrin(Zheng protein moesin (Giretti 2008, al. et ROCK-2 cascadehasalsobeenreported(Giretti for activation.DownstreamactivationoftheRhoA/ al.2010),c-Src andpaxillin(Li et (Sanchez breast cancercells,whichexpressbothESR2andESR1 2011).Ininflammatory in ER+MCF-7cells(Chen et al. to co-activateIL-8resultinginincreasedinvasiveness different ESR2variants.andPEA3werereported to crosstalkwithESR1aswelldifferentialeffectof effects, especiallyinspecificbreastcancersubtypes,due of ESR2havingbothpro-invasiveandpro-migratory reportsand anti-proliferativeeffect,withcontradictory more complexthansimplyhavingananti-migratory and invasion. strengthens celladhesionandreducesmigration actin filaments(Lindberg et al. 2010),whichconsequently formation of vinculin-containing focal complexes and 2015), integrinalpha1,beta1andenhancethe ESR2 wasreportedtoupregulate E-cadherin (Zhou et al. 2012)and to reducedcellgrowthandmobility(Li et al. DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review ESR2, ontheotherhand,hasbeenassociatedwith However, ESR2’s influenceoncellmigrationispossibly Zheng ). 2001 Lazennec et al. Overexpression of ESR2 leads et al. 2011).Recruitmentoftheactin-binding ) and phosphorylation 2008) and phosphorylation et al. 36 , ESR2andGPR30through t

b

© 2017SocietyforEndocrinology doan andothers et al. 2011)were Printed inGreatBritain t al.2010) et 36

adhesion kinase(FAK) (Graham offocal progestins alsoenhancethephosphorylation is increasedbyprogestins.Inaddition,progesterone/ 2013) suggestingamechanismthroughwhichmigration ( matrix metalloproteases(MMPs),β plasminogen activator (uPA), uPA receptor(uPAR), modulated theexpressionofgenessuchasurokinase activation ofMAPK,PI3K/AktandGTPase/RhoA that contributedtobreastcancermetastasis.Progestin that PGRactivatedcytoplasmicsignalingcascades Carnevale andcoworkers(Carnevale and metastasis.UsingaDNA-bindingmutantPGR, pathways isimplicatedinbreastcancermigration PGRA-predominant breast cancers. potentially contributingtothepooreroutcomesseenin migration andincreasedinvasion(McGowan al.2005),enhanced et adhesion signaling(Graham altered progestin response, including increased focal 2004). However, predominanceofPGRAresultsinan isoform levels,isinhibitedbyprogestins(McGowan of T-47D breastcancercells,expressingequivalentPGR and decreasedmigration(Lin and focalcontacts,consistentwithincreasedadhesion PGR, progestintreatmentincreasesactinstressfibers 2004, al. 2000, 2001, et invasiveness (Lin acting through nuclear PGR inhibit cell migration and PGR isoformandcontextdependent.Ingeneral,progestins and invasivepotential.However, theseeffectsareligand, progestin effectsonbreastcancercelladhesion,migration (Mote et al. 2015).Anumberofstudieshavedemonstrated with apoorerresponsetotheanti-estrogentamoxifen 2002)andisassociated in diseaseprogression(Mote et al. 1995).Thisdisruptionisseenearly PGRA) (Graham et al. cancer resulting in a predominance of one isoform (usually commonly theirexpressionbecomesderegulatedinbreast are equivalentlyexpressedinmostnormalcells,but al. 1990). PGRA and PGRB et a single gene (Kastner PGRB) arisingfromalternatepromoteractivitiesdriving expressed astwofunctionallydifferentforms(PGRAand Progesterone receptors ( and influencingbreastcancer progressionandmetastasis cytoskeletal reorganization involved incellularmotility formation andturnoverof focaladhesionpointsand predominance (Graham Bellance breast cancer Nuclear receptorsin Carnevale Luo &Guan2010). Published byBioscientifica Ltd. Crosstalk betweenPGRandcytoplasmicsignaling t al.2007).Incellstransfectedwith et Carnevale t al.2013),andthisisenhancedwithPGRA et t al.2007, et

Downloaded fromBioscientifica.com at09/27/202109:55:09AM t al.2008, et Fu t al.2005),promotingthe et 2000, et al. PGR inthehumanis et al. 2005, 1-integrin andPAI-1 et al. 2007)reported 58 t al. et McGowan : 3 2001). Invasion t al. et Bellance Fu 2004), et al. et al. 2008, 182 182 et al. via freeaccess

PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 183 breast cancer

PGR has also been reported to exert ligand- findings highlight that RARB is necessary for mediating the independent effects on breast cancer cell morphology anti-migratory effect of retinoic acid through modulation and migration (Jacobsen et al. 2005, Carnevale et al. 2007, of expression of genes involved in cell migration. Bellance et al. 2013). Un-liganded PGRA was shown to differentially regulate genes involved in cell adhesion Estrogen-related receptors Aside from their and increase adhesiveness and migration in inducible prominent role as master regulators of energy metabolism, cells expressing just one isoform (Jacobsen et al. 2005). the ERRs have been reported to influence the migratory However, in a recent study using bi-inducible MDA-MB-231 potential of breast cancer cells. Knockdown of ESRRA cells, un-liganded PGRB but not PGRA increased cell dramatically decreased the migratory potential of breast migration via differential effects on focal adhesion cancer cells (Stein et al. 2008), later determined to be signaling (Bellance et al. 2013). Furthermore, it has been mediated through VEGF (Stein et al. 2009) and WNT11 demonstrated that extensive ligand-independent action (Dwyer et al. 2010) signaling pathways, which are known of PGRB are dependent on specific post-translational regulators of angiogenesis. Inactivation of ESRRA also modifications, particularly phosphorylation, and resulted in impaired directional migration of breast cancer associated with pro-survival pathways (Daniel et al. 2007, cells, mediated through enhanced stability of RHOA, a Faivre et al. 2008, Knutson et al. 2012). These findings in protein involved in controlling oriented cellular migration the absence of ligand suggest that disrupted PGR isoform (Sailland et al. 2014). ESRRA itself was found to be a target expression may have particular importance in the context of miR-137 (Zhao et al. 2012). Therefore, consistent with of post-menopausal breast cancer, where circulating reports of its association with poor prognosis, activation progesterone is low or absent. of ESRRA has a pro-migratory effect on breast cancer cells. In summary, the emerging picture is that the effects By contrast, and consistent with its association with better of PGR and progesterone on breast cancer metastasis, breast cancer outcome, ESRRG was reported to promote adhesion and invasion are context dependent and mesenchymal-to-epithelial transition and decrease influenced by the relative ratio of PGR isoforms being breast cancer cell invasiveness through activation of expressed, the presence of ligands and whether PGR E-cadherin (Tiraby et al. 2011). Therefore, similar to their functions as a transcription factor or activator of signaling opposing roles in metabolic reprogramming, ESSRA and cascades, but that PGR contributes to increased invasive ESSRG display opposing influences on breast cancer cell and metastatic behavior in a number of contexts. migration and invasiveness. Journal of Molecular Endocrinology Retinoic acid receptors Several studies demonstrated NUR77 NUR77 (NR4A1) expression is elevated in both the anti-proliferative and anti-migratory effect of retinoids ER+ and ER− breast tumors (Muscat et al. 2013), and on breast cancer cells. Specifically, all-trans retinoid acid NUR77 expression is correlated with decreased relapse- (ATRA) was reported to modulate several migration-related free survival in breast cancer (Alexopoulou et al. 2010), proteins such as downregulation of MMP-1, MMP-2, consistent with reports of its pro-oncogenic role in breast cancer (Hedrick et al. 2015). Contradictory to a MMP-9, FAK, NF-κB and p-ERK and upregulation of E-cadherin in breast cancer cell lines (Liu et al. 2003, previous report of NUR77 having anti-migratory effects in Dutta et al. 2009, 2010). The anti-migratory effect of MCF-10A breast cancer cells, recently, NUR77 expression retinoids on breast cancer cells were shown to be was shown to have pro-migratory effects through activation mediated through RARB with treatment of RA or RARB of TGF-beta signaling (Zhou et al. 2014), which is known agonist resulting in significantly reduced cell migration to have an important role in breast cancer metastasis while treatment with RARA or RARG agonists did not (Moore et al. 2008, Kohn et al. 2012, Luwor et al. 2015). (Flamini et al. 2014). RARB is the only member of the RARs showing decreased expression in breast cancers COUP-TF1 and COUP-TF2 COUP-TFs are orphan (Muscat et al. 2013). RARB showed progressively decreased nuclear receptors primarily known for their important expression during breast , with only roles in organogenesis including angiogenesis, cellular around 50% of invasive breast carcinoma expressing growth, differentiation and migration (Boudot et al. RARB (Xu et al. 1997). Transduction of RARB2 resulted 2011). Their roles in breast cancer growth and progression in significant reduction in metastasis rate in a mouse have recently been highlighted. Both COUP-TF1 and xenograft model (Treuting et al. 2002). Together, these COUP-TF2 expression are lower in breast tumors

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology supported thiswork. Foundation AustraliaandaUPA Scholarship fromtheUniversityofSydney A CollaborativeProgramGrant from theNationalBreastCancer Funding perceived asprejudicingtheimpartialityofresearchreported. The authorsdeclarethatthereisnoconflictofinterestcould be Declaration ofinterest reprogramming andmigrationmetastasis. processes includingthecircadian clock,metabolic of adiverserangeothercancer-associatedbiological proliferation andapoptosis,NRsarecriticalregulators state. Finally, itisevidentthatinaddition toregulating programsofNRareassociatedwiththetumor regulatory expression oftargetgenes,andhowchangesinthegene the chromatinlandscapetofinetunecell-specific work togetheraswellwith other coregulatorsand NR transcriptionalregulation,howmultipleNRsoften Cistromic profilingstudieshighlightthecomplexityof expressed and are potentially functional in breast cancer. shown thatmanyNRs,inadditiontoESR1andPGR,are functional interactions.Expressionprofilingstudieshave mechanisms of action, as well as the complexity of their cancer biology: their expression, genomic distribution, integrated insightsintotherolesofNRsinbreast Advances ingenomictechnologieshaveenabledmore Concluding remarks cancer cells. to-mesenchymal transitionandmigrationofbreast a potentialinvolvementofCOUP-TFsinepithelial- 2004).Together thesefindingssuggest cells (Navab et al. resulted in increasedmigrationand invasion ofthese report that transfection of COUP-TF2 into MDA-MB-231 a roleinbreastcancercellmigrationandinvasionwith The otherCOUP-TFmember, COUP-TF2,mayalsohave reported todrivebreastcancermetastasis(Ray et al. 2015). tumorshasrecentlybeen CXCL12 expressioninprimary breast cancercells(LeDily leads to enhancedmotilityandinvasiveness of MCF-7 in MCF-7 cellsresultsinadecreaseCXCL12andincrease ( low expressionof E-cadherin and expression ofvimentin dedifferentiated breast cancer cells and correlated with COUP-TF1 expressionwasreportedtobehigherin compared tonormalbreasttissue(Muscat in al.2008).OverexpressionofCOUP-TF1in et Le Dily DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review CXCR4 expression,mediatedbyEGFsignaling,and et al. 2008, t

b

© 2017SocietyforEndocrinology doan Boudot et al. 2014). andothers Printed inGreatBritain ). al.2013). et Barton VN,D’AmatoNC,GordonMA, ChristensonJL,EliasA& Ballare C,CastellanoG,GavegliaL,AlthammerS,Gonzalez-Vallinas J, Avena P, AnselmoW, Whitaker-MenezesD,Wang C,PestellRG, Auwerx J,BaulieuE,BeatoM,Becker-AndreBurbachPH, Ariazi EA,ClarkGM&MertzJE2002Estrogen-relatedreceptoralpha Ao A,Wang H,KamarajugaddaS&LuJ2008Involvement ofestrogen- Alexopoulou AN,LeaoM,CaballeroOL,DaSilvaL,ReidLakhaniSR, Alaynick WA, KondoRP, XieW, HeW, DufourCR,DownesM, Akhtar RA,ReddyAB,MaywoodES,ClaytonJD,KingVM,SmithAG, Akerstedt T, KnutssonA,NarusyteJ,SvedbergP, KecklundG& T,Adhikary BrandtDT, KaddatzK,Stockert J,NaruhnS,MeissnerW, Abduljabbar R,NegmOH,LaiCF, JerjeesDA,Al-KaabiM,HamedMR, Abduljabbar R,Al-KaabiMM,NegmOH,JerjeesD,MuftahAA, References Akashi M&Takumi T2005TheorphannuclearreceptorRORalpha breast cancer Nuclear receptorsin Published byBioscientifica Ltd. Hormones andCancer Hormones cancer: acaseforclassificationas AR+ orquadruplenegativedisease. 2015Androgenreceptor biologyintriplenegativebreast Richer JK Molecular Cell Nucleosome-driven transcriptionfactor bindingandgeneregulation. Eyras E,LeDilyF, ZaurinR,SoronellasD,Vicent GP, Cell Cycle cancer tumorgrowth,viametabolicreprogrammingandsymbiosis. 2013 Compartment-specificactivationofPPARgamma governsbreast HulitJ,CasaburiI,AndoS,Martinez-OutschoornUE, Lamb RS, Cell A unifiednomenclaturesystemforthenuclearreceptorsuperfamily. ChambonP,Camerino G, CooneyA,DejeanDreyerC, Research favorable biomarkers,respectively, inhumanbreastcancer. Cancer and estrogen-relatedreceptorgammaassociatewithunfavorable pnas.0711677105) of solidtumors.PNAS related receptorsintranscriptionalresponsetohypoxiaandgrowth Research the migrationofnormalandbreastcancercelllines.Breast Cancer transcriptional networksunderlyingbreastcancer:NR4A1reduces Simpson AJ,MarshallJF, NevilleAM&JatPS2010Dissectingthe cmet.2007.06.007) postnatal .CellMetabolism directs andmaintainsthetransitiontooxidativemetabolismin Jonker JW, GilesW, Naviaux RK,GiguereV, (doi:10.1016/S0960-9822(02)00759-5) by thesuprachiasmaticnucleus.Current Biology mouse livertranscriptome,asrevealedbycDNAmicroarray, isdriven Gant TW, HastingsMH&KyriacouCP2002Circadian cyclingofthe bmjopen-2015-008127) Swedish cohortstudy. BMJOpen Alexanderson K2015Nightworkandbreastcancerinwomen:a (doi:10.1038/nsmb925) 5241–5252. ANGPTL4 geneandinhibitcancercellinvasion.Oncogene PPARbeta/delta agonistssuppressoncogenicsignalingtothe Finkernagel F, ObertJ,LieberS,ScharfeM, (doi:10.1007/s10549-015-3335-1) in breastcancer. Breast CancerResearch andTreatment and biologicalsignificanceofglucocorticoidreceptor(GR)expression Tighe PJ,BuluwelaL,MukherjeeA,GreenAR, 3348-9) Research andTreatment activated receptor-gammainluminalbreastcancer. Breast Cancer Prognostic andbiologicalsignificanceofperoxisomeproliferator- Mukherjee A,LaiCF, BuluwelaL,AliS,Tighe PJ, Bmal1. regulates circadian transcriptionofthemammaliancore-clock 97 161–163.(doi:10.1016/S0092-8674(00)80726-6) Nature Structural andMolecularBiology Nature Structural 62 6510–6518. 12 R51.(doi:10.1186/bcr2610) 12 1360–1370.(doi:10.4161/cc.24289) (doi:10.1038/onc.2012.549) 49 67–79.(doi:10.1016/j.molcel.2012.10.019)

6 206–213.(doi:10.1007/s12672-015-0232-3) 105 7821–7826.(doi:10.1073/ 150 511–522.(doi:10.1007/s10549-015- Downloaded fromBioscientifica.com at09/27/202109:55:09AM 6 13–24.(doi:10.1016/j. 5 e008127.(doi:10.1136/ 2013Inverse et al. 2007ERRgamma et al. 58 12 441–448. 2015bClinical et al. : 12 540–550. 3 2015a et al. 150 335–346. 2013 et al. 1999 et al. 32

184 184 et al. via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 185 breast cancer

Bechtold DA, Gibbs JE & Loudon AS 2010 Circadian dysfunction in ERalpha-positive breast cancer. Breast Cancer Research and Treatment disease. Trends in Pharmacological Sciences 31 191–198. (doi:10.1016/j. 154 225–237. (doi:10.1007/s10549-015-3609-7) tips.2010.01.002) Clarke CL & Graham JD 2012 Non-overlapping progesterone receptor Bellance C, Khan JA, Meduri G, Guiochon-Mantel A, Lombes M & cistromes contribute to cell-specific transcriptional outcomes.PLoS Loosfelt H 2013 Progesterone receptor isoforms PRA and PRB ONE 7 e35859. (doi:10.1371/journal.pone.0035859) differentially contribute to breast cancer cell migration through Cochrane DR, Bernales S, Jacobsen BM, Cittelly DM, Howe EN, interaction with focal adhesion kinase complexes. Molecular Biology D’Amato NC, Spoelstra NS, Edgerton SM, Jean A, Guerrero J, et al. 2014 of the Cell 24 1363–1374. (doi:10.1091/mbc.E12-11-0807) Role of the androgen receptor in breast cancer and preclinical analysis Biddie SC, John S, Sabo PJ, Thurman RE, Johnson TA, Schiltz RL, of enzalutamide. Breast Cancer Research 16 R7. (doi:10.1186/bcr3599) Miranda TB, Sung MH, Trump S, Lightman SL, et al. 2011 Conway-Campbell BL, Sarabdjitsingh RA, McKenna MA, Pooley JR, Transcription factor AP1 potentiates chromatin accessibility and Kershaw YM, Meijer OC, De Kloet ER & Lightman SL 2010 binding. Molecular Cell 43 145–155. Glucocorticoid ultradian rhythmicity directs cyclical gene pulsing of (doi:10.1016/j.molcel.2011.06.016) the clock gene period 1 in rat hippocampus. Journal of Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM & Mangelsdorf DJ Neuroendocrinology 22 1093–1100. 2006 Anatomical profiling of nuclear receptor expression reveals a (doi:10.1111/j.1365-2826.2010.02051.x) hierarchical transcriptional network. Cell 126 789–799. Conzen SD 2008 Nuclear receptors and breast cancer. Molecular (doi:10.1016/j.cell.2006.06.049) Endocrinology 22 2215–2228. (doi:10.1210/me.2007-0421) Boudot A, Le Dily F & Pakdel F 2011 Involvement of COUP-TFs in Cordera F & Jordan VC 2006 Steroid receptors and their role in the cancer progression. Cancers 3 700–715. (doi:10.3390/cancers3010700) biology and control of breast cancer growth. Seminars in Oncology 33 Boudot A, Kerdivel G, Lecomte S, Flouriot G, Desille M, Godey F, 631–641. Leveque J, Tas P, Le Drean Y & Pakdel F 2014 COUP-TFI modifies Cos S, Gonzalez A, Martinez-Campa C, Mediavilla MD, Alonso- CXCL12 and CXCR4 expression by activating EGF signaling and Gonzalez C & Sanchez-Barcelo EJ 2006 Estrogen-signaling pathway: a stimulates breast cancer cell migration. BMC Cancer 14 407. link between breast cancer and melatonin oncostatic actions. Cancer (doi:10.1186/1471-2407-14-407) Detection and Prevention 30 118–128. (doi:10.1016/j.cdp.2006.03.002) Cai Q, Lin T, Kamarajugadda S & Lu J 2013 Regulation of glycolysis and D’Amato NC, Jacobsen BM, Cochrane DR, Spoelstra NS, Babbs BL, Elias A the Warburg effect by estrogen-related receptors. Oncogene 32 & Richer JK 2015 Inhibiting androgen receptor nuclear localization 2079–2086. (doi:10.1038/onc.2012.221) decreases estrogen receptor (ER) activity and tumor growth in ER plus Calcinotto A, Filipazzi P, Grioni M, Iero M, De Milito A, Ricupito A, breast cancer. Cancer Research 75 (9 Suppl) abstract P3-04-06. Cova A, Canese R, Jachetti E, Rossetti M, et al. 2012 Modulation of (doi:10.1016/j.cdp.2006.03.002) microenvironment acidity reverses anergy in human and murine Daniel AR, Faivre EJ & Lange CA 2007 Phosphorylation-dependent tumor-infiltrating T lymphocytes.Cancer Research 72 2746–2756. antagonism of sumoylation derepresses progesterone receptor action (doi:10.1158/0008-5472.CAN-11-1272) in breast cancer cells. Molecular Endocrinology 21 2890–2906. Carnevale RP, Proietti CJ, Salatino M, Urtreger A, Peluffo G, Edwards DP, (doi:10.1210/me.2007-0248) Boonyaratanakornkit V, Charreau EH, Bal de Kier Joffe E, Schillaci Daniel AR, Gaviglio AL, Knutson TP, Ostrander JH, D’Assoro AB, R, et al. 2007 Progestin effects on breast cancer cell proliferation, Ravindranathan P, Peng Y, Raj GV, Yee D & Lange CA 2015 proteases activation, and in vivo development of metastatic Progesterone receptor-B enhances estrogen responsiveness of breast phenotype all depend on progesterone receptor capacity to activate cancer cells via scaffolding PELP1- and estrogen receptor-containing cytoplasmic signaling pathways. Molecular Endocrinology 21 transcription complexes. Oncogene 34 506–515. (doi:10.1038/ 1335–1358. (doi:10.1210/me.2006-0304) onc.2013.579) Journal of Molecular Endocrinology Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, Dauchy RT, Xiang S, Mao L, Brimer S, Wren MA, Yuan L, Anbalagan M, Brodsky AS, Keeton EK, Fertuck KC, Hall GF, et al. 2006 Genome- Hauch A, Frasch T, Rowan BG, et al. 2014 Circadian and melatonin wide analysis of estrogen receptor binding sites. Nature Genetics 38 disruption by exposure to light at night drives intrinsic resistance to 1289–1297. (doi:10.1038/ng1901) tamoxifen therapy in breast cancer. Cancer Research 74 4099–4110. Chakravarty D, Nair SS, Santhamma B, Nair BC, Wang L, Bandyopadhyay (doi:10.1158/0008-5472.CAN-13-3156) A, Agyin JK, Brann D, Sun LZ, Yeh IT, et al. 2010 Extranuclear functions De Mei C, Ercolani L, Parodi C, Veronesi M, Lo Vecchio C, Bottegoni G, of ER impact invasive migration and metastasis by breast cancer cells. Torrente E, Scarpelli R, Marotta R, Ruffili R, et al. 2015 Dual Cancer Research 70 4092–4101. (doi:10.1158/0008-5472.CAN-09-3834) inhibition of REV-ERBbeta and autophagy as a novel Chen ST, Choo KB, Hou MF, Yeh KT, Kuo SJ & Chang JG 2005 pharmacological approach to induce cytotoxicity in cancer cells. Deregulated expression of the PER1, PER2 and PER3 genes in breast Oncogene 34 2597–2608. (doi:10.1038/onc.2014.203) cancers. Carcinogenesis 26 1241–1246. (doi:10.1093/carcin/bgi075) Deblois G & Giguere V 2013 Oestrogen-related receptors in breast Chen Y, Chen L, Li JY, Mukaida N, Wang Q, Yang C, Yin WJ, Zeng XH, cancer: control of cellular metabolism and beyond. Nature Reviews Jin W & Shao ZM 2011 ERbeta and PEA3 co-activate IL-8 expression Cancer 13 27–36. (doi:10.1038/nrc3396) and promote the invasion of breast cancer cells. Cancer Biology and Deblois G, Chahrour G, Perry MC, Sylvain-Drolet G, Muller WJ & Therapy 11 497–511. (doi:10.4161/cbt.11.5.14667) Giguere V 2010 Transcriptional control of the ERBB2 amplicon by Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong LW, ERRalpha and PGC-1beta promotes tumorigenesis. DiTacchio L, Atkins AR, Glass CK, et al. 2012 Regulation of circadian Cancer Research 70 10277–10287. (doi:10.1158/0008-5472.CAN-10- behaviour and metabolism by REV-ERB-alpha and REV-ERB-beta. 2840) Nature 485 123–127. (doi:10.1038/nature11048) Ding Y, Liu Z, Desai S, Zhao Y, Liu H, Pannell LK, Yi H, Wright ER, Owen Cirillo LA, Lin FR, Cuesta I, Friedman D, Jarnik M & Zaret KS 2002 LB, Dean-Colomb W, et al. 2012 Receptor tyrosine kinase ErbB2 Opening of compacted chromatin by early developmental translocates into mitochondria and regulates cellular metabolism. transcription factors HNF3 (FoxA) and GATA-4. Molecular Cell 9 Nature Communications 3 1271. (doi:10.1038/ncomms2236) 279–289. (doi:10.1016/S1097-2765(02)00459-8) Dixon JM 2014 Endocrine resistance in breast cancer. New Journal of Ciupek A, Rechoum Y, Gu G, Gelsomino L, Beyer AR, Brusco L, Science 2014 1–27. Covington KR, Tsimelzon A & Fuqua SA 2015 Androgen receptor Doan TB, Eriksson NA, Graham D, Funder JW, Simpson ER, Kuczek ES, promotes tamoxifen agonist activity by activation of EGFR in Clyne C, Leedman PJ, Tilley WD, Fuller PJ, et al. 2014 Breast cancer

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology Fu X-D,GirettiMS,BaldacciC,Garibaldi S,FlaminiM,SanchezAM, C,ShaoZM&JettenAM1992Retinoid antagonism Fontana JA,Nervi Flamini MI,GaunaGV, SottileML,NadinBS,SanchezAM&Vargas- Fioretti FM,Sita-LumsdenA,BevanCL&BrookeGN2014Revisingthe Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Ferlay J,SoerjomataramI,DikshitR,EserS,MathersC,RebeloM, Faivre EJ,DanielAR,HillardCJ&LangeCA2008Progesteronereceptor Evans RM&MangelsdorfDJ2014Nuclearreceptors,RXR,andthebig ENCODE ProjectConsortium2012AnintegratedencyclopediaofDNA MC,DufourCR,BertosN,ParkM,St-PierreJ& Eichner LJ,Perry Eichner LJ&GiguereV2011Estrogenrelatedreceptors(ERRs):anew Dutta A,SenT, BanerjiA,DasS&ChatterjeeA2009Studieson Dufour CR,LevasseurMP, PhamNH,EichnerLJ,Wilson BJ,Charest- Dufour CR,Wilson BJ,HussJM,KellyDP, AlaynickWA, DownesM, Early BreastCancerTrialists’ CollaborativeGroup(EBCTCG)2005Effects Dwyer MA,JosephJD,Wade HE,EatonML,KunderRS,KazminD, Dutta A,SenT&ChatterjeeA2010All-transretinoicacid(ATRA) DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review (doi:10.1371/journal.pone.0002790) and invasionthroughtheactincytoskeleton. PLoSONE signaling ofprogesteronereceptorto breastcancercellmovement Gadducci A,GenazzaniAR&Simoncini T2008Extra-nuclear 3938–3945. gene expressioninbreastcarcinoma cells.CancerResearch of estrogen-responsivetransforminggrowthfactoralphaandpS2 Molecular Medicine ofCellularand cancer cells:roleofretinoicacidreceptorbeta.Journal 2014Retinoicacidreducesmigrationofhumanbreast Roig LM Endocrinology role oftheandrogenreceptorinbreastcancer. ofMolecular Journal ijc.29210) 2012. worldwide: sources, methods andmajorpatternsinGLOBOCAN FormanD&BrayF2015Cancerincidenceandmortality Parkin DM, 22 823–837.(doi:10.1210/me.2007-0437) to specificityprotein1transcriptionfactors.MolecularEndocrinology andtethering rapid signalingmediatesserine345phosphorylation bang. nature11247) elements inthehumangenome.Nature Metabolism cells viathePGC-1beta/ERRgammatranscriptionalpathway. Cell Giguere V2010miR-378(*)mediatesmetabolicshiftinbreastcancer Mitochondrion dawn intranscriptionalcontrolofmitochondrialgenenetworks. Journal ofOncology breast cancercells(MCF-7).Journal moleculesinhuman metalloproteinase-2 (MMP-2)anditsregulatory multifunctional effectofall-transretinoicacid(ATRA) onmatrix (doi:10.1371/journal.pgen.1002143) in thecontrolofmetabolicclockoutputs.PLoSGenetics 2011 GenomicconvergenceamongERRalpha,PROX1,andBMAL1 Marcotte A,DuguayD,Poirier-HeonJF, CermakianN&GiguereV cmet.2007.03.007) ERRalpha andgamma.CellMetabolism orchestration ofcardiacfunctionsbytheorphannuclearreceptors Evans RM,BlanchetteM&GiguereV2007Genome-wide Molecular Oncology prognosis predictedbynuclearreceptor-coregulatornetworks. trials. oftherandomised anoverview recurrence and15-yearsurvival: of chemotherapyandhormonaltherapyforearlybreastcanceron 70 9298–9308.(doi:10.1158/0008-5472.CAN-10-0226) autocrine mannertoincreasecancercellmigration.CancerResearch estrogen-related receptoralphaandbeta-cateninactsinan Chang CY&McDonnellDP2010WNT11expressionisinducedby Adhesion andMigration molecules.Cell downregulates MMP-9bymodulatingitsregulatory International Journal ofCancer Journal International Cell Lancet 157 255–266.(doi:10.1016/j.cell.2014.03.012) 12 352–361.(doi:10.1016/j.cmet.2010.09.002) 365 1687–1717.(doi:10.1016/s0140-6736(05)66544-0) 52 R257–R265.(doi:10.1530/JME-14-0030) 11 544–552.(doi:10.1016/j.mito.2011.03.121) 18 1113–1123.(doi:10.1111/jcmm.12256) 8 998–1013.(doi:10.1016/j.molonc.2014.03.017) 4 409–418.(doi:10.4161/cam.4.3.11682) 136 E359–E386.(doi:10.1002/ t 5 345–356.(doi:10.1016/j.

b 489 57–74.(doi:10.1038/

© 2017SocietyforEndocrinology doan andothers 2009 627840. Printed inGreatBritain 7 e1002143. 3 e2790. 52

Gery S,VirkGery RK,ChumakovK,Yu A&KoefflerHP2007Theclockgene Gertz J,SavicD,Varley KE,PartridgeEC,SafiA,Jain P, CooperGM, Gerstein MB,KundajeA,HariharanM,LandtSG,Yan KK,ChengC, Germain P, StaelsB,DacquetC,SpeddingM&LaudetV2006Overview Gerhart-Hines Z&LazarMA2015Rev-erbandthecircadian Graham JD,Yeates SS,MillikenJS,BilousAM& C,BalleineRL,Harvey Giretti MS,FuX-D,DeRosaG,SarottoI,BaldacciC,GaribaldiS, Giguere V, HollenbergSM,RosenfeldMG&EvansRM1986Functional Giguere V2008Transcriptional controlofenergyhomeostasisbythe Graham JD,Yager ML,HillHD,BythK,O’NeillGM&ClarkeCL2005 Grogg A,Trippel M,PfaltzK,LadrachC,DroeserRA,CihoricN, Gronemeyer H,GustafssonJA&LaudetV2004Principlesfor Hedrick E,LeeSO,DoddapaneniR,SinghM&SafeS2015Nuclear Hansen J&StevensRG2012Case-controlstudyofshift-workandbreast Hansen J&LassenCF2012Nestedcase-controlstudyofnightshift Hansen J2001Increasedbreastcancerriskamongwomenwhowork Ha NH,LongJ,CaiQ,ShuXO&HunterKW2016Thecircadian Hu R,DawoodS,HolmesMD,Collins LC,SchnittSJ,ColeK, Hilborn E,GacicJ,FornanderT, NordenskjoldB,StalO&JanssonA breast cancer Nuclear receptorsin Published byBioscientifica Ltd. 7916–7920. Per2 linksthecircadian systemtotheestrogenreceptor. Oncogene Cell type-specific andsharedtranscriptionfactorbindingsites.Molecular Reddy TE,CrawfordGE&MyersRM2013Distinctpropertiesofcell- Nature networkderivedfromENCODEdata. of thehumanregulatory KhuranaE,RozowskyJ,AlexanderR, Mu XJ, 685–704. Reviews of nomenclaturenuclearreceptors.Pharmacological Metabolism transcriptional regulationofmetabolism.DiabetesObesityand expression inhumanbreastcancer. CancerResearch Clarke CL1995CharacterizationofprogesteronereceptorAandB (doi:10.1371/journal.pone.0002238) remodelling, migrationandinvasion.PLoSONE nuclear signallingofestrogenreceptortobreastcancercytoskeletal Mannella P, BigliaN,SismondiP, GenazzaniAR, (doi:10.1016/0092-8674(86)90339-9) domains ofthehumanglucocorticoidreceptor. Cell (doi:10.1210/er.2008-0017) estrogen-related receptors.EndocrineReviews 2713–2735. responsiveness ofbreastcancercells.MolecularEndocrinology Altered progesteronereceptorisoformexpressionremodelsprogestin Cancer duringtumorprogressionofbreastcancer.highly conserved BMC ZweifelM&TapiaSalhia B, C2015Androgenreceptorstatusis Discovery modulation ofthenuclearreceptorsuperfamily. Nature ReviewsDrug Endocrine-Related Cancer receptor 4A1asadrugtargetforbreastcancerchemotherapy. ofCancer Journal cancer riskinDanishnurses:impactofshiftsystems.European oemed-2011-100240) Occupational andEnvironmental Medicine work andbreastcancerriskamongwomenintheDanishmilitary. (doi:10.1097/00001648-200101000-00013) predominantly atnight.Epidemiology (doi:10.1371/journal.pgen.1006267) receptor-negative breastcancer. PLoSGenetics rhythm geneArntl2isametastasissusceptibilityforestrogen Marotti JD, HankinsonSE,Colditz GA&TamimiMarotti JD, RM2011 Androgen ofCancer Journal response inoestrogenreceptor-alpha-negative breastcancer. British 2016 Androgenreceptorexpression predictsbeneficialtamoxifen 52 25–36.(doi:10.1016/j.molcel.2013.08.037) 489 91–100.(doi:10.1038/nature11245) 15 872.(doi:10.1186/s12885-015-1897-2) 3 950–964.(doi:10.1038/nrd1551) (doi:10.1124/pr.58.4.2) 17 12–16.(doi:10.1111/dom.12510) (doi:10.1038/sj.onc.1210585) (doi:10.1210/me.2005-0126) 48 1722–1729.(doi:10.1016/j.ejca.2011.07.005) 114 248–255.(doi:10.1038/bjc.2015.464)

22 831–840.(doi:10.1530/ERC-15-0063) Downloaded fromBioscientifica.com at09/27/202109:55:09AM 12 74–77. 69 551–556.(doi:10.1136/ 29 677–696. 58 2012Architecture et al. 12 e1006267. : 3 3 e2238. 2008Extra- et al. 55 5063–5068. 46 645–652. 19 58 186 186 26 via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 187 breast cancer

receptor expression and breast cancer survival in postmenopausal Kohn EA, Yang YA, Du Z, Nagano Y, Van Schyndle CM, Herrmann MA, women. Clinical Cancer Research 17 1867–1874. (doi:10.1158/1078- Heldman M, Chen JQ, Stuelten CH, Flanders KC, et al. 2012 0432.CCR-10-2021) Biological responses to TGF-beta in the mammary epithelium show a Hua S, Kittler R & White KP 2009 Genomic antagonism between complex dependency on Smad3 gene dosage with important retinoic acid and estrogen signaling in breast cancer. Cell 137 implications for tumor progression. Molecular Cancer Research 10 1259–1271. (doi:10.1016/j.cell.2009.04.043) 1389–1399. (doi:10.1158/1541-7786.MCR-12-0136-T) Hudis CA & Gianni L 2011 Triple-negative breast cancer: an unmet Kourtidis A, Jain R, Carkner RD, Eifert C, Brosnan MJ & Conklin DS medical need. Oncologist 16 (Supplement 1) 1–11. (doi:10.1634/ 2010 An RNA interference screen identifies metabolic regulators theoncologist.2011-S1-01) NR1D1 and PBP as novel survival factors for breast cancer cells with Hurtado A, Holmes KA, Ross-Innes CS, Schmidt D & Carroll JS 2011 the ERBB2 signature. Cancer Research 70 1783–1792. FOXA1 is a key determinant of estrogen receptor function and (doi:10.1158/0008-5472.CAN-09-1550) endocrine response. Nature Genetics 43 27–33. (doi:10.1038/ng.730) Krishnan AV, Swami S & Feldman D 2010 Vitamin D and breast cancer: Jacobsen BM, Schittone SA, Richer JK & Horwitz KB 2005 Progesterone- inhibition of estrogen synthesis and signaling. Journal of Steroid independent effects of human progesterone receptors (PRs) in Biochemistry and Molecular Biology 121 343–348. (doi:10.1016/j. estrogen receptor-positive breast cancer: PR isoform-specific gene jsbmb.2010.02.009) regulation and tumor biology. Molecular Endocrinology 19 574–587. Krum SA, Miranda-Carboni GA, Lupien M, Eeckhoute J, Carroll JS & (doi:10.1210/me.2004-0287) Brown M 2008 Unique ER alpha cistromes control cell type-specific Jarvinen TA, Pelto-Huikko M, Holli K & Isola J 2000 Estrogen receptor gene regulation. Molecular Endocrinology 22 2393–2406. (doi:10.1210/ beta is coexpressed with ERalpha and PR and associated with nodal me.2008-0100) status, grade, and proliferation rate in breast cancer. American Journal Kumar V, Green S, Stack G, Berry M, Jin JR & Chambon P 1987 of Pathology 156 29–35. (doi:10.1016/S0002-9440(10)64702-5) Functional domains of the human estrogen receptor. Cell 51 Jia Y, Lu Y, Wu K, Lin Q, Shen W, Zhu M, Huang S & Chen J 2013 Does 941–951. (doi:10.1016/0092-8674(87)90581-2) night work increase the risk of breast cancer? A systematic review Kuznetsova T, Wang SY, Rao NA, Mandoli A, Martens JH, Rother N, and meta-analysis of epidemiological studies. Cancer Epidemiology 37 Aartse A, Groh L, Janssen-Megens EM, Li G, et al. 2015 197–206. (doi:10.1016/j.canep.2013.01.005) Glucocorticoid receptor and nuclear factor kappa-b affect three- John S, Sabo PJ, Thurman RE, Sung MH, Biddie SC, Johnson TA, dimensional chromatin organization. Genome Biology 16 264. Hager GL & Stamatoyannopoulos JA 2011 Chromatin accessibility (doi:10.1186/s13059-015-0832-9) pre-determines glucocorticoid receptor binding patterns. Nature Lai C-F, Flach KD, Alexi X, Fox SP, Ottaviani S, Thiruchelvam PTR, Kyle Genetics 43 264–268. (doi:10.1038/ng.759) FJ, Thomas RS, Launchbury R, Hua H, et al. 2013 Co-regulated gene Karmakar S, Jin Y & Nagaich AK 2013 Interaction of glucocorticoid expression by oestrogen receptor alpha and liver receptor homolog-1 receptor (GR) with estrogen receptor (ER) alpha and activator protein is a feature of the oestrogen response in breast cancer cells. Nucleic 1 (AP1) in dexamethasone-mediated interference of ERalpha activity. Acids Research 41 10228–10240. (doi:10.1093/nar/gkt827) Journal of Biological Chemistry 288 24020–24034. (doi:10.1074/jbc. Laudet V & Gronemeyer H 2002 2 – DNA recognition by nuclear M113.473819) receptors. In The Nuclear Receptor FactsBook, pp 22–36. Eds V Laudet Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H & & H Gronemeyer. London, UK: Academic Press. Chambon P 1990 Two distinct estrogen-regulated promoters Lazennec G, Bresson D, Lucas A, Chauveau C & Vignon F 2001 ER beta generate transcripts encoding the two functionally different human inhibits proliferation and invasion of breast cancer cells. progesterone receptor forms A and B. EMBO Journal 9 1603–1614. Endocrinology 142 4120–4130. (doi:10.1210/en.142.9.4120) Keeton EK & Brown M 2003 Coregulator expression and breast cancer: Le Dily F, Metivier R, Gueguen MM, Le Peron C, Flouriot G, Tas P & Journal of Molecular Endocrinology improving the predictive power of . Clinical Pakdel F 2008 COUP-TFI modulates estrogen signaling and Cancer Research 9 1229–1230. influences proliferation, survival and migration of breast cancer cells. Kininis M, Chen BS, Diehl AG, Isaacs GD, Zhang T, Siepel AC, Clark AG Breast Cancer Research and Treatment 110 69–83. (doi:10.1007/s10549- & Kraus WL 2007 Genomic analyses of transcription factor binding, 007-9693-6) histone acetylation, and gene expression reveal mechanistically Li Y, Wang JP, Santen RJ, Kim TH, Park H, Fan P & Yue W 2010 Estrogen distinct classes of estrogen-regulated promoters. Molecular and stimulation of cell migration involves multiple signaling pathway Cellular Biology 27 5090–5104. (doi:10.1128/MCB.00083-07) interactions. Endocrinology 151 5146–5156. (doi:10.1210/en.2009- Kittler R, Zhou J, Hua S, Ma L, Liu Y, Pendleton E, Cheng C, Gerstein M 1506) & White KP 2013 A comprehensive nuclear receptor network for Li H, Tu Z, An L, Qian Z, Achilefu S & Gu Y 2012 Inhibitory effects of breast cancer cells. Cell Reports 3 538–551. (doi:10.1016/j. ERbeta on proliferation, invasion, and tumor formation of MCF-7 celrep.2013.01.004) breast cancer cells–prognostication for the use of ERbeta-selective Knower KC, Chand AL, Eriksson N, Takagi K, Miki Y, Sasano H, Visvader therapy. Pharmaceutical Biology 50 839–849. (doi:10.3109/13880209.2 JE, Lindeman GJ, Funder JW, Fuller PJ, et al. 2013 Distinct nuclear 011.637506) receptor expression in stroma adjacent to breast tumors. Breast Lin VC, Ng EH, Aw SE, Tan MG, Ng EH & Bay BH 2000 Progesterone Cancer Research and Treatment 142 211–223. (doi:10.1007/s10549- induces focal adhesion in breast cancer cells MDA-MB-231 013-2716-6) transfected with progesterone receptor complementary DNA. Knutson TP, Daniel AR, Fan D, Silverstein KA, Covington KR, Fuqua SA Molecular Endocrinology 14 348–358. (doi:10.1210/mend.14.3.0426) & Lange CA 2012 Phosphorylated and sumoylation-deficient Lin VC, Eng AS, Hen NE, Ng EH & Chowdhury SH 2001 Effect of progesterone receptors drive proliferative gene signatures during progesterone on the invasive properties and tumor growth of breast cancer progression. Breast Cancer Research 14 R95. progesterone receptor-transfected breast cancer cells MDA-MB-231. (doi:10.1186/bcr3211) Clinical Cancer Research 7 2880–2886. Knutsson A, Alfredsson L, Karlsson B, Akerstedt T, Fransson EI, Lin M-L, Patel H, Remenyi J, Banerji CRS, Lai C-F, Periyasamy M, Westerholm P & Westerlund H 2013 Breast cancer among shift Lombardo Y, Busonero C, Ottaviani S, Passey A, et al. 2015 workers: results of the WOLF longitudinal cohort study. Scandinavian Expression profiling of nuclear receptors in breast cancer identifies Journal of Work, Environment and Health 39 170–177. (doi:10.5271/ TLX as a mediator of growth and invasion in triple-negative breast sjweh.3323) cancer. Oncotarget 6 21685–21703. (doi:10.18632/oncotarget.3942)

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology Menegaux F, Truong T, AngerA,Cordina-DuvergerE,Lamkarkach F, Mehta RG,PengX,AlimirahF, MurilloG&MehtaR2013Vitamin D McKenna NJ&O’MalleyBW2010SnapShot:nuclearreceptorsI.Cell McKenna NJ&O’MalleyBW2002Combinatorialcontrolofgene McGowan EM,SaadS,BendallLJ,BradstockKF&ClarkeCL2004Effect Mangelsdorf DJ,ThummelC,BeatoM,HerrlichP, SchutzG, Malovannaya A,LanzRB,JungSY, BulynkoY, LeNT, ChanDW, DingC, Luwor RB,HakmanaD,IariaJ,NheuTV, Simpson RJ&ZhuHJ2015 Lupien M,MeyerCA,BaileyST, EeckhouteJ,CookWesterling T, Lupien M,EeckhouteJ,MeyerCA,KrumSA,RhodesDR,LiuXS& Lupien M,EeckhouteJ,MeyerCA,Wang Q,ZhangY, LiW, CarrollJS, Luo M&GuanJL2010Focaladhesionkinase:aprominentdeterminant Lonard DM,LanzRB&O’MalleyBW2007Nuclearreceptorcoregulators Lonard DM&O’MalleyBW2012Nuclearreceptorcoregulators: Liu H,ZangC,FennerMH,PossingerK&ElstnerE2003PPARgamma Liu MH&CheungE2014Estrogenreceptor-mediatedlong-range Lindberg K,StromA,LockJG,GustafssonJA,HaldosenLA& DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review (doi:10.1002/ijc.27669) ofCancer Journal CECILE study).International breast cancer:apopulation-basedcase-control studyinFrance(the P,Arveux KerbratP, FevotteJ&GuenelP2013Nightworkand (doi:10.1016/j.canlet.2012.10.034) and breastcancer:emergingconcepts.CancerLetters 142 822–822.e821.(doi:10.1016/j.cell.2010.08.026) (doi:10.1016/S0092-8674(02)00641-4) expression bynuclearreceptorsandcoregulators.Cell Treatment migration intobonemarrowfibroblasts.Breast CancerResearch and of progesteronereceptorapredominanceonbreastcancercell 839. The nuclearreceptorsuperfamily:theseconddecade.Cell BlumbergB,KastnerP,Umesono K, MarkM,ChambonP, (doi:10.1016/j.cell.2011.05.006) endogenous coregulatorcomplexome.Cell Shi Y, Yucer N,KrenciuteG, (doi:10.1186/s12943-015-0309-1) dynamic TGF-beta-Smad3activity. MolecularCancer motility andmigrationisdrivenbysub-populationsofcellswith Single livecellTGF-betasignallingimaging:breastcancer (doi:10.1101/gad.1944810) cancer endocrineresistance.GenesandDevelopment stimulation inducesadistinctER(alpha)cistromeunderlyingbreast Zhang X,CarrollJS,RhodesDR,LiuXS, (doi:10.1128/MCB.00020-09) receptor alphacistrome.MolecularandCellularBiology Brown M2009Coactivatorfunctiondefinestheactiveestrogen (doi:10.1016/j.cell.2008.01.018) enhancer-driven lineage-specifictranscription.Cell Liu XS&BrownM2008FoxA1translatesepigeneticsignaturesinto 289 127–139.(doi:10.1016/j.canlet.2009.07.005) in breastcancerinitiation,progressionandmetastasis.CancerLetters er.2007-0012) and humandisease.EndocrineReviews Endocrinology modulators ofpathologyandtherapeutictargets.Nature Reviews 3/A:1023366117157) in vitro.Breast CancerResearch andTreatment ligands andATRA inhibittheinvasionofhumanbreastcancercells mce.2013.09.019) and CellularEndocrinology chromatin interactionsandtranscriptioninbreastcancer. Molecular (doi:10.1002/jcp.21932) ofCellularPhysiology breast cancercells.Journal integrin alpha1andbeta1levelsenhancesadhesionof 2010Expressionofestrogenreceptorbetaincreases Helguero LA (doi:10.1016/0092-8674(95)90199-X) 83 211–220.(doi:10.1023/B:BREA.0000014041.58977.80) 8 598–604.(doi:10.1038/nrendo.2012.100) 382 624–632.(doi:10.1016/j. 2011Analysisofthehuman et al. t 28 575–587.(doi:10.1210/

b 2010Growthfactor et al.

© 2017SocietyforEndocrinology doan 145 787–799. 79 63–74.(doi:10.102 132 924–931. andothers 222 156–167. Printed inGreatBritain 132 958–970. 24 2219–2227. 14 50. 334 95–100. 108 465–474. 29 3413–3423. 83 835– 1995 et al. Oishi K,AmagaiN,ShiraiH,Kadota K,OhkuraN&Ishida2005 Oishi K,MiyazakiKadotaKikunoR,NagaseT, AtsumiG, Ohshiro K,SchwartzAM,LevinePH&KumarR2012Alternateestrogen ES,SchoenfeldER,StevensRG,KabatGC,Henderson K, O’Leary Ni M,ChenY, LimE,Wimberly H,BaileyST, ImaiY, RimmDL,LiuXS Navab R,Gonzalez-SantosJM,JohnstonMR,LiuJ,BrodtP, Tsao MS& Muscat GEO,ErikssonNA,BythK,LoiS,GrahamD,JindalDavisMJ, Mote PA, GompelA,HoweC,HiltonHN,SestakI,CuzickJ,DowsettM, Michalik L,DesvergneB&Wahli W2004Peroxisome-proliferator- Mohammed H,RussellIA,StarkR,RuedaOM,HickeyTE,Tarulli GA, Miranda TB,Voss TC,SungM-H,BaekS,JohnHawkinsM,Grontved Millard CJ,Watson PJ,FairallL&SchwabeJWR2013Anevolving Mootha VK,HandschinC,ArlowD,XieX,StPierreJ,SihagS,Yang W, Moore LD,IsayevaT, SiegalGP&PonnazhaganS2008Silencingof Mote PA, BartowS,Tran N&ClarkeCL2002Lossofco-ordinate breast cancer Nuclear receptorsin Published byBioscientifica Ltd. 191–202. dependent circadian genesinthemouseliver. DNAResearch Genome-wide expressionanalysisreveals 100adrenalgland- (doi:10.1074/jbc.M304564200) ofBiologicalChemistry output genes.Journal expression analysisofmouseliverrevealsCLOCK-regulatedcircadian AzamaT,Ohkura N, MesakiM,Yukimasa S, pone.0030725) non-genomic signaling.PLoSONE breastcancer cellsvia receptors promoteinvasionofinflammatory of Epidemiology night, andbreastcanceronLongIsland,NewYork. AmericanJournal Grimson R,GammonMD&LeskeMC2006Shiftwork,lightat ccr.2011.05.026) negative breastcancer. CancerCell & BrownM2011Targeting androgenreceptorinestrogenreceptor- 5472.CAN-03-1185) carcinoma cells.CancerResearch transcription factorIIenhancesinvasivenessofhumanlung Hu J2004Expressionofchickenovalbuminupstreampromoter- (doi:10.1210/me.2012-1265) value inbreastcancer. MolecularEndocrinology nuclear receptorsastranscriptome:discriminantandprognostic Clyne C,FunderJW, SimpsonER, (doi:10.1007/s10549-015-3397-0) in theATAC trial.Breast CancerResearch andTreatment predominance isadiscriminatorofbenefitfromendocrinetherapy Hugol D,ForgezP, BythK, 163–172. Cancer activated receptorsandcancers:complexstories.Nature Reviews receptor modulatesERalphaactioninbreastcancer. Nature Serandour AA,BirrellSN,BrunaA,SaadiA, 5472.CAN-13-0742) the genomiclevel.CancerResearch landscape: interplayoftheestrogenandglucocorticoidreceptorsat L, SchiltzRL&HagerGL2013Reprogrammingthechromatin Molecular Endocrinology of understanding ofnuclearreceptorcoregulatorproteins.Journal 6570–6575. gene expressionthatisalteredindiabeticmuscle.PNAS Gabpa/b specifyPGC-1alpha-dependentoxidativephosphorylation P,Altshuler D,Puigserver PattersonN, (doi:10.1158/1078-0432.CCR-07-4604) and metastasisinvivo.ClinicalCancerResearch breast cancer:implicationsforproliferationandmigrationinvitro transforming growthfactor-beta1insitubyRNAinterferencefor 313–317. breast carcinogenesis. Breast CancerResearch andTreatment expression ofprogesteronereceptorsAandBisanearlyeventin 4 61–70.(doi:10.1038/nrc1254) (doi:10.1093/dnares/dsi003) (doi:10.1023/A:1014820500738) (doi:10.1038/nature14583) (doi:10.1073/pnas.0401401101) 164 358–366.(doi:10.1093/aje/kwj211)

51 T23–T36.(doi:10.1530/JME-13-0227) 2015ProgesteronereceptorA et al. Downloaded fromBioscientifica.com at09/27/202109:55:09AM 64 5097–5105.(doi:10.1158/0008- 2013Research resource: et al. 7 e30725.(doi:10.1371/journal. 20 119–131.(doi:10.1016/j. 73 5130–5139.(doi:10.1158/0008- 2004Erralphaand et al. 2015Progesterone et al. 278 41519–41527. 2003Genome-wide et al. 58 27 350–365. 14 4961–4970. : 3 151 309–318. 101 72 523 12

188 188

via freeaccess PROOF ONLY

Review t b doan and others Nuclear receptors in 58:3 189 breast cancer

Ottow E & Weinmann H 2008 Nuclear receptors as drug targets: a Sailland J, Tribollet V, Forcet C, Billon C, Barenton B, Carnesecchi J, historical perspective of modern drug discovery. In Nuclear Receptors Bachmann A, Gauthier KC, Yu S, Giguere V, et al. 2014 Estrogen- as Drug Targets, pp 1–23. Weinheim, Germany: Wiley-VCH Verlag related receptor a decreases RHOA stability to induce orientated cell GmbH & Co. KGaA. (doi:10.1002/9783527623297.ch1) migration. PNAS 111 15108–15113. (doi:10.1073/pnas.1402094111) Pan D, Kocherginsky M & Conzen SD 2011 Activation of the Sakharkar MK, Shashni B, Sharma K, Dhillon SK, Ranjekar PR & glucocorticoid receptor is associated with poor prognosis in estrogen Sakharkar KR 2013 Therapeutic implications of targeting energy receptor-negative breast cancer. Cancer Research 71 6360–6370. metabolism in breast cancer. PPAR Research 2013 109285. (doi:10.1158/0008-5472.CAN-11-0362) (doi:10.1155/2013/109285) Papantoniou K, Castano-Vinyals G, Espinosa A, Aragones N, Perez- Sanchez AM, Flamini MI, Baldacci C, Goglia L, Genazzani AR & Gomez B, Ardanaz E, Altzibar JM, Sanchez VM, Gomez-Acebo I, Simoncini T 2010 Estrogen receptor-alpha promotes breast cancer Llorca J, et al. 2015 Breast cancer risk and night shift work in a case- cell motility and invasion via focal adhesion kinase and N-WASP. control study in a Spanish population. European Journal of Molecular Endocrinology 24 2114–2125. (doi:10.1210/me.2010-0252) Epidemiology 31 867–878. (doi:10.1007/s10654-015-0073-y) Schernhammer ES, Laden F, Speizer FE, Willett WC, Hunter DJ, Kawachi Park S, Koo J, Park HS, Kim JH, Choi SY, Lee JH, Park BW & Lee KS 2010 I & Colditz GA 2001 Rotating night shifts and risk of breast cancer Expression of androgen receptors in primary breast cancer. Annals of in women participating in the nurses’ health study. Journal of the Oncology 21 488–492. (doi:10.1093/annonc/mdp510) National Cancer Institute 93 1563–1568. (doi:10.1093/jnci/93.20.1563) Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack Schernhammer ES, Kroenke CH, Laden F & Hankinson SE 2006 Night JR, Ross DT, Johnsen H, Akslen LA, et al. 2000 Molecular portraits of work and risk of breast cancer. Epidemiology 17 108–111. human breast tumours. Nature 406 747–752. (doi:10.1038/35021093) (doi:10.1097/01.ede.0000190539.03500.c1) Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Centenera MM, Stein RA, Chang CY, Kazmin DA, Way J, Schroeder T, Wergin M, Harris JM, Jindal S, Segara D, Jia L, Moore NL, et al. 2009 Androgen Dewhirst MW & McDonnell DP 2008 Estrogen-related receptor receptor inhibits estrogen receptor-alpha activity and is prognostic alpha is critical for the growth of estrogen receptor-negative breast in breast cancer. Cancer Research 69 6131–6140. (doi:10.1158/0008- cancer. Cancer Research 68 8805–8812. (doi:10.1158/0008-5472.CAN- 5472.CAN-09-0452) 08-1594) Pon CK, Firth SM & Baxter RC 2015 Involvement of insulin-like growth Stein RA, Gaillard S & McDonnell DP 2009 Estrogen-related receptor factor binding protein-3 in peroxisome proliferator-activated receptor alpha induces the expression of vascular endothelial growth factor gamma-mediated inhibition of breast cancer cell growth. Molecular and in breast cancer cells. Journal of Steroid Biochemistry and Molecular Cellular Endocrinology 399 354–361. (doi:10.1016/j.mce.2014.10.023) Biology 114 106–112. (doi:10.1016/j.jsbmb.2009.02.010) Prat A, Pineda E, Adamo B, Galvan P, Fernandez A, Gaba L, Diez M, Surjit M, Ganti KP, Mukherji A, Ye T, Hua G, Metzger D, Li M & Viladot M, Arance A & Munoz M 2015 Clinical implications of the Chambon P 2011 Widespread negative response elements mediate intrinsic molecular subtypes of breast cancer. Breast 24 (Supplement direct repression by agonist-liganded glucocorticoid receptor. Cell 2) S26–S35. (doi:10.1016/j.breast.2015.07.008) 145 224–241. (doi:10.1016/j.cell.2011.03.027) Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U Tang Q, Chen Y, Meyer C, Geistlinger T, Lupien M, Wang Q, Liu T, & Schibler U 2002 The orphan nuclear receptor REV-ERBalpha Zhang Y, Brown M & Liu XS 2011 A comprehensive view of nuclear controls circadian transcription within the positive limb of the receptor cancer cistromes. Cancer Research 71 6940–6947. mammalian circadian oscillator. Cell 110 251–260. (doi:10.1016/ (doi:10.1158/0008-5472.CAN-11-2091) S0092-8674(02)00825-5) Thewes V, Simon R, Schroeter P, Schlotter M, Anzeneder T, Buettner R, Pronk A, Ji BT, Shu XO, Xue S, Yang G, Li HL, Rothman N, Gao YT, Benes V, Sauter G, Burwinkel B, Nicholson RI, et al. 2015 Zheng W & Chow WH 2010 Night-shift work and breast cancer risk Reprogramming of the ERR alpha and ER alpha target gene Journal of Molecular Endocrinology in a cohort of Chinese women. American Journal of Epidemiology 171 landscape triggers tamoxifen resistance in breast cancer. Cancer 953–959. (doi:10.1093/aje/kwq029) Research 75 720–731. (doi:10.1158/0008-5472.CAN-14-0652) Rabstein S, Harth V, Pesch B, Pallapies D, Lotz A, Justenhoven C, Baisch Tiraby C, Hazen BC, Gantner ML & Kralli A 2011 Estrogen-related C, Schiffermann M, Haas S, Fischer HP, et al. 2013 Night work and receptor gamma promotes mesenchymal-to-epithelial transition and breast cancer estrogen receptor status–results from the German suppresses breast tumor growth. Cancer Research 71 2518–2528. GENICA study. Scandinavian Journal of Work, Environment and Health (doi:10.1158/0008-5472.CAN-10-1315) 39 448–455. (doi:10.5271/sjweh.3360) Treuting PM, Chen LI, Buetow BS, Zeng W, Birkebak TA, Seewaldt VL, Ray P, Stacer AC, Fenner J, Cavnar SP, Meguiar K, Brown M, Luker KE & Sommer KM, Emond M, Maggio-Price L & Swisshelm K 2002 Luker GD 2015 CXCL12-gamma in primary tumors drives breast beta2 inhibition of metastasis in mouse cancer metastasis. Oncogene 34 2043–2051. (doi:10.1038/onc.2014.157) mammary gland xenografts. Breast Cancer Research and Treatment 72 Robinson JL, Macarthur S, Ross-Innes CS, Tilley WD, Neal DE, Mills IG 79–88. (doi:10.1023/A:1014906529407) & Carroll JS 2011 Androgen receptor driven transcription in Vazquez A, Liu J, Zhou Y & Oltvai ZN 2010 Catabolic efficiency of molecular apocrine breast cancer is mediated by FoxA1. EMBO aerobic glycolysis: the Warburg effect revisited. BMC Systems Biology Journal 30 3019–3027. (doi:10.1038/emboj.2011.216) 4 58. (doi:10.1186/1752-0509-4-58) Rosin G, de Boniface J, Karthik GM, Frisell J, Bergh J & Hartman J 2014 Vilasco M, Communal L, Mourra N, Courtin A, Forgez P & Gompel A Oestrogen receptors beta1 and betacx have divergent roles in breast 2011 Glucocorticoid receptor and breast cancer. Breast Cancer cancer survival and lymph node metastasis. British Journal of Cancer Research and Treatment 130 1–10. (doi:10.1007/s10549-011-1689-6) 111 918–926. (doi:10.1038/bjc.2014.398) Viswanathan AN & Schernhammer ES 2009 Circulating melatonin and Ross-Innes CS, Stark R, Holmes KA, Schmidt D, Spyrou C, Russell R, the risk of breast and endometrial cancer in women. Cancer Letters Massie CE, Vowler SL, Eldridge M & Carroll JS 2010 Cooperative 281 1–7. (doi:10.1016/j.canlet.2008.11.002) interaction between retinoic acid receptor-alpha and estrogen Wang F, Yeung KL, Chan WC, Kwok CC, Leung SL, Wu C, Chan EY, Yu receptor in breast cancer. Genes and Development 24 171–182. IT, Yang XR & Tse LA 2013 A meta-analysis on dose-response (doi:10.1101/gad.552910) relationship between night shift work and the risk of breast cancer. Ross-Innes CS, Stark R, Teschendorff AE, Holmes KA, Ali HR, Dunning Annals of Oncology 24 2724–2732. (doi:10.1093/annonc/mdt283) MJ, Brown GD, Gojis O, Ellis IO, Green AR, et al. 2012 Differential Ward PS & Thompson CB 2012 Metabolic reprogramming: a cancer oestrogen receptor binding is associated with clinical outcome in hallmark even warburg did not anticipate. Cancer Cell 21 297–308. breast cancer. Nature 481 389–393. (doi:10.1038/nature10730) (doi:10.1016/j.ccr.2012.02.014)

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0082 Printed in Great Britain Downloaded from Bioscientifica.com at 09/27/2021 09:55:09AM via free access Journal of Molecular Endocrinology Zhang YX,FangB,EmmettMJ,DamleM,SunZ,FengD,ArmourSM, Yao P-L,MoralesJL,ZhuB,KangB-H,GonzalezFJ&PetersJM2014 Yang QF, SakuraiT&KakudoK2002Retinoid,retinoicacidreceptor Xu XC,SneigeN,LiuX,NandagiriR,LeeJJ,LukmanjiF, HortobagyiG, Xie D,BoyleAlanP, Wu L,ZhaiJ,KawliT&SnyderM2013Dynamic Xiao L,ChangAK,ZangMX,BiH,LiS,Wang M,XingX&Wu H2014 Xiang SL,DauchyRT, HauchA,MaoLL,Yuan L,Wren MA,BelancioVP, Xiang S,MaoL,DuplessisT, Yuan L,DauchyR,E,BlaskDE, DOI: 10.1530/JME-16-0082 http://jme.endocrinology-journals.org Review Review Remsberg JR,JagerJ,SoccioRE, (doi:10.1158/1535-7163.MCT-13-0836) tumorigenicity. (ppar-beta/delta) inhibitshumanbreastcancercellline Activation ofperoxisomeproliferator-activatedreceptor-beta/delta 167–173. beta andbreastcancer. Breast CancerResearch andTreatment breast carcinogenesis. CancerResearch nuclear retinoicacidreceptorbetamessengerRNAlevelduring Lippman SM,DhingraK&LotanR1997Progressivedecreasein (doi:10.1016/j.cell.2013.09.043) trans-acting factorcolocalizationinhumancells.Cell journal.pone.0095878) proliferation ofbreastcancercells.PLoSONE Induction oftheCLOCKgenebyE2-ERalphasignalingpromotes (doi:10.1111/jpi.12239) circadian ofPinealResearch melatoninsignal.Journal breast cancerisdrivenbylightatnight-induceddisruptionofthe Mondal D,FraschT, BlaskDE, cancer cells:regulationbymelatonin.Breast Cancer genes inducedbyserumshockinhumanbreastepithelialand Frasch T&HillSM2012Oscillationofclockandcontrolled (doi:10.1023/A:1020576606004) Molecular CancerTherapeutics 2015Doxorubicinresistancein et al. 2015Discretefunctionsof et al. 57 4992–4996. t

b

© 2017SocietyforEndocrinology doan 13 1008–1017. 9 e95878.(doi:10.1371/ andothers Printed inGreatBritain 6 137–150. 59 60–69. 155 713–724. 76

Zwart W, TheodorouV, KokM,CanisiusS,LinnS&CarrollJS2011 Zhou Y, MingJ,XuY, Zhang Y&JiangJ2015ERbeta1inhibitsthe Zhou F, DrabschY, DekkerTJA,deVinuesa AG,LiY, HawinkelsLJAC, Zheng S,HuangJ,ZhouK,ZhangC,XiangQ,Tan Z,Wang T&FuX Zhao X,ChoH,Yu RT, AtkinsAR,DownesM&EvansRM2014Nuclear Zhao Y, LiY, LouG,ZhaoL,XuZ,ZhangY&HeF2012MiR-137 Zhao YH,ZhouM,LiuH,DingY, KhongHT, Yu D,FodstadO&Tan M Accepted Preprint published online 13 January 2017 Accepted Preprintpublishedonline13January 2017 Accepted 12January Received infinalform13December2016 breast cancer Nuclear receptorsin Published byBioscientifica Ltd. 4764–4776. transcriptional regulationofbreastcancer. EMBOJournal Oestrogen receptor-co-factor-chromatinspecificityinthe Research Communications E-cadherin inaId1-dependentmanner. BiochemicalandBiophysical migration andinvasionofbreastcancercellsthroughupregulation 5 3388.(doi:10.1038/ncomms5573) metastasis byactivatingTGF-betasignalling.Nature Communications Nuclear receptorNR4A1promotesbreastcancerinvasionand Sheppard K-A,GoumansM-J,LuworRB,deVries CJ, PLoS ONE invasion viaextra-nuclearactivationofactin-bindingproteinezrin. 2011 17beta-Estradiolenhancesbreastcancercellmotilityand (doi:10.1002/embr.201338271) receptors rockaroundtheclock.EMBOReports (doi:10.1371/journal.pone.0039102) capacityofbreastcancercells.PLoSONE and migratory targets estrogen-relatedreceptoralphaandimpairstheproliferative growth. heat shockfactor1promotesbreastcancercellglycolysisand 2009 UpregulationoflactatedehydrogenaseAbyErbB2through Science nuclear receptorRev-erbalphacouplemetabolismtotheclock. 348 1488–1492.(doi:10.1126/science.aab3021) Oncogene 6 e22439.(doi:10.1371/journal.pone.0022439) (doi:10.1038/emboj.2011.368) 28 3689–3701.(doi:10.1038/onc.2009.229)

457 141–147.(doi:10.1016/j.bbrc.2014.12.038) Downloaded fromBioscientifica.com at09/27/202109:55:09AM 58 15 518–528. : 3 . 2014 et al 7 e39102. 30

190 190 via freeaccess