19
Overexpression of transcription factor AP-2 suppresses mammary gland growth and morphogenesis J. Zhang, a,1 S. Brewer, a,2 J. Huang, b,2 and T. Williams a,b, * a Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA b Departments of Craniofacial Biology and Cellular & Structural Biology, BRB151, Campus Box C286, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Denver, CO 80262, USA Received for publication 28 March 2002, revised 11 December 2002, accepted 11 December 2002 Abstract AP-2 transcription factors are key regulators of mouse embryonic development. Aberrant expression of these genes has also been linked to the progression of human breast cancer. Here, we have investigated the role of the AP-2 gene family in the postnatal maturation of the mouse mammary gland. Analysis of AP-2 RNA and protein levels demonstrates that these genes are expressed in the mammary glands of virgin and pregnant mice. Subsequently, AP-2 expression declines during lactation and then is reactivated during involution. The AP-2 and AP-2 proteins are localized in the ductal epithelium, as well as in the terminal end buds, suggesting that they may influence growth of the ductal network. We have tested this hypothesis by targeting AP-2 expression to the mouse mammary gland using the MMTV promoter. Our studies indicate that overexpression of AP-2 inhibits mammary gland growth and morphogenesis, and this coincides with a rise in PTHrP expression. Alveolar budding is severely curtailed in transgenic virgin mice, while lobuloalveolar development and functional differentiation are inhibited during pregnancy and lactation, respectively. Our studies strongly support a role for the AP-2 proteins in regulating the proliferation and differentiation of mammary gland epithelial cells in both mouse and human. © 2003 Elsevier Science (USA). All rights reserved. Keywords: Transcription factors; AP-2; AP-2; Transgenic mice; Mammary gland; Alveolar budding; Lobuloalveolar development; Breast cancer Introduction The AP-2 family of transcription factors regulates im- portant aspects of vertebrate embryogenesis and has also been linked to the control of cell proliferation and tumori- genesis. The AP-2 proteins constitute a distinct class of transcription factor, which is defined by the presence of a “basic– helix–span– helix” DNA binding and dimerization motif (Williams and Tjian, 1991a,b). To date, four AP-2 genes have been characterized in mammalian species, AP- 2, AP-2, AP-2, and AP-2 (Bosher et al., 1996; Chazaud et al., 1996; Imagawa et al., 1987; Mitchell et al., 1987; Moser et al., 1995; Williams et al., 1988; Zhao et al., 2001). Considerable sequence identity exists between the DNA binding domains of these four AP-2 proteins, and they can all bind as homo- or heterodimers to a GC-rich element typified by the consensus sequence 5-GCCNNNGGC-3 (Bosher et al., 1996; McPherson and Weigel, 1999; Mo- hibullah et al., 1999). Many genes that mediate cell growth, cell shape, cell movement, and cell communication fre- quently possess important AP-2 binding sites as a compo- nent of their cis-regulatory sequences (Bosher et al., 1995; Gaubatz et al., 1995; Gille et al., 1997; McPherson et al., 1997; Zeng et al., 1997). Indeed, the finding that genes, including cytokeratins, E-cadherin, integrins, and matrix metalloproteinases, are regulated by AP-2 reflects an im- portant role for these transcription factors in morphogenesis (Byrne et al., 1994; De Clerk et al., 1994; Fini et al., 1994; * Corresponding author. Departments of Craniofacial Biology and Cel- lular & Structural Biology, BRB151, Campus Box C286, University of Colorado Health Sciences Center, 4200 East Ninth Avenue, Denver CO 80262, USA. Fax: 1-303-315-3013. E-mail address: [email protected] (T. Williams). 1 Present address: College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R.C. 2 These authors contributed equally to the project. R Available online at www.sciencedirect.com Developmental Biology 256 (2003) 127–145 www.elsevier.com/locate/ydbio 0012-1606/03/$ – see front matter © 2003 Elsevier Science (USA). All rights reserved. doi:10.1016/S0012-1606(02)00119-7

Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Overexpression of transcription factor AP-2� suppresses mammarygland growth and morphogenesis

J. Zhang,a,1 S. Brewer,a,2 J. Huang,b,2 and T. Williamsa,b,*a Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA

b Departments of Craniofacial Biology and Cellular & Structural Biology, BRB151, Campus Box C286, University of Colorado Health Sciences Center,4200 East Ninth Avenue, Denver, CO 80262, USA

Received for publication 28 March 2002, revised 11 December 2002, accepted 11 December 2002

Abstract

AP-2 transcription factors are key regulators of mouse embryonic development. Aberrant expression of these genes has also been linkedto the progression of human breast cancer. Here, we have investigated the role of the AP-2 gene family in the postnatal maturation of themouse mammary gland. Analysis of AP-2 RNA and protein levels demonstrates that these genes are expressed in the mammary glands ofvirgin and pregnant mice. Subsequently, AP-2 expression declines during lactation and then is reactivated during involution. The AP-2� andAP-2� proteins are localized in the ductal epithelium, as well as in the terminal end buds, suggesting that they may influence growth of theductal network. We have tested this hypothesis by targeting AP-2� expression to the mouse mammary gland using the MMTV promoter.Our studies indicate that overexpression of AP-2� inhibits mammary gland growth and morphogenesis, and this coincides with a rise inPTHrP expression. Alveolar budding is severely curtailed in transgenic virgin mice, while lobuloalveolar development and functionaldifferentiation are inhibited during pregnancy and lactation, respectively. Our studies strongly support a role for the AP-2 proteins inregulating the proliferation and differentiation of mammary gland epithelial cells in both mouse and human.© 2003 Elsevier Science (USA). All rights reserved.

Keywords: Transcription factors; AP-2�; AP-2�; Transgenic mice; Mammary gland; Alveolar budding; Lobuloalveolar development; Breast cancer

Introduction

The AP-2 family of transcription factors regulates im-portant aspects of vertebrate embryogenesis and has alsobeen linked to the control of cell proliferation and tumori-genesis. The AP-2 proteins constitute a distinct class oftranscription factor, which is defined by the presence of a“basic–helix–span–helix” DNA binding and dimerizationmotif (Williams and Tjian, 1991a,b). To date, four AP-2genes have been characterized in mammalian species, AP-

2�, AP-2�, AP-2�, and AP-2� (Bosher et al., 1996;Chazaud et al., 1996; Imagawa et al., 1987; Mitchell et al.,1987; Moser et al., 1995; Williams et al., 1988; Zhao et al.,2001). Considerable sequence identity exists between theDNA binding domains of these four AP-2 proteins, and theycan all bind as homo- or heterodimers to a GC-rich elementtypified by the consensus sequence 5�-GCCNNNGGC-3�(Bosher et al., 1996; McPherson and Weigel, 1999; Mo-hibullah et al., 1999). Many genes that mediate cell growth,cell shape, cell movement, and cell communication fre-quently possess important AP-2 binding sites as a compo-nent of their cis-regulatory sequences (Bosher et al., 1995;Gaubatz et al., 1995; Gille et al., 1997; McPherson et al.,1997; Zeng et al., 1997). Indeed, the finding that genes,including cytokeratins, E-cadherin, integrins, and matrixmetalloproteinases, are regulated by AP-2 reflects an im-portant role for these transcription factors in morphogenesis(Byrne et al., 1994; De Clerk et al., 1994; Fini et al., 1994;

* Corresponding author. Departments of Craniofacial Biology and Cel-lular & Structural Biology, BRB151, Campus Box C286, University ofColorado Health Sciences Center, 4200 East Ninth Avenue, Denver CO80262, USA. Fax: �1-303-315-3013.

E-mail address: [email protected] (T. Williams).1 Present address: College of Life Science, Hunan Normal University,

Changsha, Hunan 410081, P.R.C.2 These authors contributed equally to the project.

R

Available online at www.sciencedirect.com

Developmental Biology 256 (2003) 127–145 www.elsevier.com/locate/ydbio

0012-1606/03/$ – see front matter © 2003 Elsevier Science (USA). All rights reserved.doi:10.1016/S0012-1606(02)00119-7

Page 2: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Hennig et al., 1996; Mitchell et al., 1987; Somasundaram etal., 1996; Zutter et al., 1994). Currently, the AP-2�, AP-2�,and AP-2� genes are the best characterized. During embry-ogenesis, all three genes are expressed in tissues undergoingcomplex morphogenetic changes, principally in the neuralcrest, neural tube, kidney, eye, facial prominences, and limbbuds (Chazaud et al., 1996; Mitchell et al., 1991; Moser etal., 1997b). The expression of the individual AP-2 genesoften coincides in particular tissues, but overall, each familymember has a unique spatiotemporal pattern of expression.Mouse molecular genetic studies have demonstrated thatthere is a critical requirement for the AP-2 genes duringmammalian development. A homozygous null mutation forany of the AP-2 genes results in lethality either duringembryogenesis or shortly after birth. In particular, the dis-ruption of the AP-2� gene leads to defects in the formationof the neural tube, face, eyes, heart, and limbs during em-bryogenesis (Brewer et al., 2002; Nottoli et al., 1998;Schorle et al., 1996; West-Mays et al., 1999; Zhang et al.,1996). Moreover, these AP-2� knockout mice lack a ventralbody-wall, resulting in a severe thoracoabdominoschisis.Mice lacking the AP-2� gene die shortly after birth, due toinappropriate morphogenesis of the kidneys (Moser et al.,1997a), but do not display any other major developmentalabnormalities. AP-2�-null mice die around E7.5, shortlyafter implantation (Auman et al., 2002; Werling andSchorle, 2002). In this instance, the defects occur within theextraembryonic cell lineages and there is no requirement forAP-2� in the embryo proper (Auman et al., 2002). The factthat none of the AP-2 knockout mice survive until weaningindicates the collective importance of the AP-2 genes, but atpresent, precludes an understanding of how these genesmight regulate later developmental events, in particularmammary gland maturation.

Although much is known about the spatiotemporal reg-ulation of the AP-2 genes during embryogenesis, little in-formation exists concerning their expression in the postnatalmammary gland. Nevertheless, it is clear from studies ofhuman tissue samples that the AP-2 proteins can be ex-pressed in the adult breast (Turner et al., 1998). Moreover,several studies have indicated that the AP-2 proteins mayregulate gene expression in breast cancer (Gee et al., 1999;Turner et al., 1998). The AP-2�, AP-2�, and AP-2� pro-teins have been detected in breast cancer biopsies, and thereis a significant up-regulation of AP-2� expression in early-stage breast tumors compared with benign tissue (Turner etal., 1998). The presence of various combinations of theAP-2 proteins in tumor tissue also coincides with the ex-pression of several genes involved in cell growth and pro-liferation. Significant correlations have been reported forthe presence of the insulin-like growth factor I receptor(IGF-I-R) and AP-2� expression, the estrogen receptor (ER)and AP-2�, and p21waf1/cip1 and AP-2 family members (Geeet al., 1999; Turner et al., 1998). However, the most strikingfinding was that the majority of tumors containing signifi-cant levels of ErbB2 (HER2/Neu) protein expressed both

the AP-2� and AP-2� proteins (Turner et al., 1998). Thesignificance of these various correlations is strengthened bythe finding that AP-2 binding sites are present within thepromoters of the human IGF-I-R, ER, p21waf1/cip1, andErbB2 genes (Bosher et al., 1995; McPherson et al., 1997;Turner et al., 1998; Zeng et al., 1997). Thus, AP-2 mayregulate the transcription of these targets in human breastcancer. Evidence obtained in vitro concerning the role ofAP-2 in cellular transformation suggests two alternativehypotheses for the presence of increased expression of AP-2in breast cancer (Kannan et al., 1994; Zeng et al., 1997).One possibility is that the AP-2 proteins are proliferativesignals that activate a variety of growth factor pathways. Analternative hypothesis is that the AP-2 proteins are moreakin to tumor suppressors, and the activation of the AP-2genes would represent a failed attempt to halt cell prolifer-ation. However, it has not been possible to distinguishbetween these potential roles for AP-2 using the currentstrains of AP-2 knockout mice, due to their early lethality.Therefore, in the current analysis, we have generated andanalyzed transgenic mice in which the AP-2� gene is over-expressed in the postnatal mammary gland. We have com-bined these transgenic studies with an analysis of AP-2expression during normal mammary gland maturation.Taken together, our findings demonstrate that the AP-2proteins can regulate the growth and morphogenesis of themammary gland. In particular, the data support the hypoth-esis that the presence of exogenous AP-2� inhibits prolif-eration of the ductal epithelia, and we therefore infer that theendogenous AP-2 genes may be activated for the samepurpose in human breast cancer.

Materials and methods

Tissue collection and histological analysis

Mammary gland tissue samples were obtained by remov-ing the fourth inguinal gland from mice at various stages ofpostnatal development, i.e., from virgin, pregnant, and lac-tating mice, and from mice in which the mammary glandwas regressing. In virgin wild-type and transgenic mice,mammary glands were removed at equivalent stages of theestrous cycle (Rugh, 1968). In lactating mice, the pups wereremoved from the mothers 5–6 h before sacrifice and mam-mary gland isolation. The tissue specimens were fixed withfreshly made 10% neutral buffered formalin or 4% parafor-maldehyde, dehydrated in a graded series of ethanol andxylene, then embedded in paraffin wax. Sections of 5 �mwere cut and mounted on poly-L-lysine-coated slides. Thesesections were stained with hematoxylin and eosin or usedfor immunohistochemical analysis. Whole-mount analysisof mouse mammary tissue was performed as previouslydescribed (Wysolmerski et al., 1995). The detection of�-galactosidase activity in mouse mammary tissue was asdescribed (Brisken et al., 1998), and slides were counter-

128 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 3: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Fig. 1. Expression of the AP-2 gene family in the mouse mammary gland. (A) RNase protection analyses of AP-2� (upper panel), AP-2� (middle), and AP-2�(lower) expression at various stages of mammary gland maturation (as indicated at top: V, virgin; Preg, pregnant; Lact, lactation; Reg, regressing). Sampleswere normalized to �-actin RNA (data not shown), except during lactation, when the RNA levels of the milk protein genes skewed the total RNA population.Note that two to three mice were used for the virgin and regression stage RNA samples, while one mouse was utilized for the other stages. (B–G) Detectionof AP-2 protein expression in the mouse mammary gland. The presence of AP-2� (brown) was detected in cell nuclei by using the 3B5 monoclonal antibody

Page 4: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Fig. 2. The expression of �-galactosidase in the mammary glands of mice heterozygous for the AP-2� lacZ knock-in allele. The detection of �-galactosidase activitywas performed on whole- mount preparations (A, C, E, G), and subsequently visualized in cross-section (B, D, F, H). Mammary glands were derived from miceat 5 weeks of age (virgins; A and B); at 10 days of pregnancy (C and D); at 7 days of lactation (E and F); and during regression, 7 days after weaning (G and H).

(B–D). AP-2� protein was visualized by using the �96 polyclonal antiserum (E–G). Sections shown are ductal tissue (B, E) and terminal end bud structures(C, F) from a 7-week virgin mouse; and alveolar tissue from a mouse at 14 days of pregnancy (D, G). Body cells (arrow) and cap cells (arrowhead) of theterminal end bud are marked in (C) and (F).

Page 5: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

stained with nuclear fast red. The generation and character-ization of mice containing the AP-2� lacZ knock-in allelehave been described elsewhere (Brewer et al., 2002).

Antibodies and immunohistochemistry

Immunohistochemical analysis of endogenous AP-2 pro-tein expression was performed by using either an AP-2�-specific monoclonal antibody (3B5) or an AP-2�-specificpolyclonal antiserum (�96), as previously described (Turneret al., 1998; Zhang et al., 1996). The analysis of transgeneexpression was performed by using the polyclonal anti-serum SC-807 (Santa Cruz), which recognizes the FLAGepitope tag. Sections were incubated with a 1:100 dilutionof SC-807 overnight at 4°C and subsequently processed forreactivity by using the avidin–biotin peroxidase technique(Vectastain ABC Kit; Vector Laboratories). The assessmentof cell proliferation in mammary tissue was performed byusing the Phosphohistone H3 (Ser10) (6G3) (Cell SignalingTechnology) mouse monoclonal antibody in combinationwith the M.O.M. blocking procedure (Vector Laboratories)as recommended by the manufacturer. Subsequently, sec-tions were incubated for 30 min in a solution of 1� PBS and0.05% Triton that contained Alexa Fluor 488 goat anti-rabbit IgG and Alexa Fluor 594 goat anti-mouse IgG (Mo-lecular Probes). Images were captured on a SPOT 2 digitalcamera by using a Leica microscope with fluorescence op-tics, and were manipulated in Adobe Photoshop. Cell pro-liferation was also measured by using the “BrdU Cell Pro-liferation Assay” (Oncogene Research Products). Briefly,13.5-day or 18.5-day pregnant transgenic or wild-type micewere injected with bromodeoxyuridine solution (50 mg/kg,i.p.) and euthanized 2 h later. The fourth inguinal mammarygland was isolated and fixed in 4% paraformaldehyde. Fol-lowing immunological detection of BrdU on sectioned ma-terial, the number of BrdU-positive cells per 1000 epithelialcells was counted from 5 different randomly chosen areas inthe mammary gland tissue sections, and the data were an-alyzed by using the Student’s t test.

For apoptotic cell analysis, frozen sections were cut at 12�M and mounted on poly-L-lysine-coated slides. TheTUNEL assay was performed by using the “In Situ CellDetection Kit” (Boehringer Mannheim) or the “FrageELDNA Fragmentation Kit” (Oncogene Research Products),and slides were then counterstained with nuclear fast red.Data were quantitated as above for the BrdU-labeled tissue.

Construction of transgenes

Transgenes were constructed by using standard subclon-ing techniques. The plasmid SPRSV-AP2 (Williams andTjian, 1991a) was used as a starting vector for attachment ofa sequence encoding a FLAG epitope tag to the 3� end of thehuman AP-2� cDNA. Specifically, the sequence 5�- GGCGGC GAT TAC AAA GAC GAC GAT GAT AAA TAGGAA TTC CTC GAG -3� was used to replace the normal

TGA stop codon as well as the 3� flanking sequences upuntil the XhoI site, which is present in the adjacentpolylinker. The new plasmid, termed SP(RSV)AP-2 FLAG,encodes a version of AP-2� in which the normal 437-amino-acid protein is extended by 10 residues (gly gly asptyr lys asp asp asp asp lys Stop), of which the final 8 encodethe FLAG epitope. This tag enables expression of the trans-gene to be distinguished from the endogenous gene at thelevel of RNA and protein expression. The AP-2 FLAGfusion construct was then placed under the control of mousemammary tumor virus long terminal repeat (MMTV LTR).The SP(RSV)AP-2 FLAG plasmid possesses a secondpolylinker sequence immediately upstream of the AP-2�cDNA open reading frame. A HindIII site is present at the5� end of this polylinker, and an initiator codon occupies thelast three nucleotide positions, 5�-AAG CTT GAA TTCGGT ACC CGC CAT G-3�. The SP(RSV) AP-2 FLAGplasmid was digested with the restriction enzyme HindIIIand subsequently repaired with T4 DNA polymerase in thepresence of dNTPs. The AP-2� cDNA insert was thenremoved by digestion with XhoI and placed between theSmaI and XhoI sites of the plasmid pMSG (AmershamPharmacia) to generate the plasmid MSG AP-2 FLAG. Thefidelity of the construct was confirmed by DNA sequenceanalysis. For microinjection, MSG AP-2 FLAG was di-gested with HindIII and XbaI restriction enzymes, and thefragment corresponding to the transgene was then separatedfrom vector sequences by sucrose density gradient ultracen-trifugation. The fractions that contained the transgene frag-ment were collected and dialyzed extensively against 10mM Tris–Cl, 0.25 mM EDTA, pH 7.5, and the DNA con-centration was adjusted to 2 �g/ml prior to microinjection.

Generation of transgenic mice

Transgenic mice were generated by injecting DNA intopronuclei of fertilized eggs of inbred FVB mice (Taconic).The embryos surviving the microinjection were transferredinto the oviducts of pseudopregnant females (CD1 strain;Charles River). After birth, founders carrying the transgenewere identified by Southern blot analysis of genomic DNA.The offspring of the founders carrying the transgene wereidentified by PCR. Transgene expression was monitored byRNase protection of total RNA isolated from mammarytissue of 14-day pregnant mice.

Isolation of genomic DNA, southern blot, and PCRanalysis

Genomic DNA was isolated from tails of 3- to 4-week-old mice as described (Laird et al., 1991). A 12-�g aliquotof genomic DNA was digested with the restriction enzymeBglII, electrophoresed on a 0.8% agarose gel, and trans-ferred to nitrocellulose (Schleicher & Schuell) or Hybond Nfilters (Amersham). The filter was hybridized with a SmaI–NcoI fragment corresponding to nucleotides 557 to 1290 of

131J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 6: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

the human AP-2� cDNA (Williams et al., 1988). This DNAfragment was either radioactively labeled by random-prim-ing in the presence of 32P dATP, or labeled nonradioactivelywith the “Genius Nonradioactive Nucleic Acid Labelingand Detection Kit” (Boehringer-Mannheim). The hybrid-ized products were visualized by autoradiography or bylight-emission, respectively. PCR was performed by usingforward primer MMTV-5 (corresponding to MMTV LTRnucleotides 7591–7611 in the plasmid pMSG): 5�-TCACAA GAG CGG AAC GGA CTC-3�, and reverse primerTWY3 (corresponding to positions 128-111 of the humanAP-2� cDNA): 5�-GCT GGT GCC GTC GTC ACG-3�.PCR conditions were: 1 cycle at 94°C for 1 min 20 s; 32cycles at 94°C for 45 s, 53°C for 45 s, 72°C for 2 min; and1 cycle at 72°C for 10 min. A fragment of 320 bp wasamplified from mice containing the transgene.

Isolation of RNA, RNase protection, and in situhybridization

Mouse mammary tissue was homogenized in guanidineisothiocyanate buffer, and the total RNA was isolated asdescribed (Chomczynski and Sacchi, 1987). RNase protec-tion was performed essentially as published (Williams et al.,1988), except that 10 �g of total RNA was used for eachprotection assay. Riboprobes specific for transcripts corre-sponding to the human AP-2� transgene, or the endogenousmouse AP-2�, AP-2�, or AP-2� genes, were generatedfrom the following four plasmids. The plasmid TRIP1 con-tains the mouse AP-2� cDNA sequences corresponding toamino acids 97 to 437 inserted between the HindIII andEcoRI sites of pBS II KS� (Stratagene). The plasmidJSmu�5� contains the mouse AP-2� cDNA sequences cor-responding to amino acids 66 to 214 inserted between theHindIII and Acc65I sites of pBS II SK� (Stratagene). Theplasmid mu-�-3� contains the mouse AP-2� cDNA se-quences corresponding to amino acids 364 to 449 insertedbetween the HindIII and KpnI sites of pBS II SK� (Strat-agene). The plasmid P4 contains the human AP-2� cDNAcorresponding to amino acids 1 to 437, and the sequencesencoding the C-terminal FLAG epitope tag, inserted be-tween the EcoRI and XhoI sites of pBluescript SK� (Strat-agene). TRIP1, JSmu�5�, mu-�-3�, and P4 were linearizedwith MluI, XbaI, HindIII, and NcoI, respectively, and thecRNA probes were synthesized with T7 RNA polymerase inthe presence of �32P UTP. The protected fragments were580 nt for AP-2�, 450 nt for AP-2�, 300 nt for AP-2�, and150 nt for the human transgene. The riboprobe specific forthe transgene incorporated sequences corresponding to theFLAG epitope tag, as well as a 120-nt fragment of the 3�end of human AP-2� cDNA (see Fig. 3). A 380-bp mousePTHrP cDNA fragment, corresponding to nucleotide posi-tions 121–501 of exon 5 (from GenBank Accession No.M60057), was derived from total cellular RNA of 12- daypregnant mammary tissue by RT-PCR using the primers5�-TG GAA TTC CAA GGA CAC GTT ACA GGA TT-3�

(forward) and 5�-CAT GAA TTC ATG CAC AGA AGGAAA TCA GT-3� (reverse). Subsequently, this fragmentwas subcloned into the EcoRI site of pBS II KS (Strat-agene), and an antisense riboprobe was made by using T3RNA polymerase after linearization with BamHI. A 231-bpp21waf1/cip1 cDNA fragment, corresponding to nucleotides20–241 (from GenBank Accession No. U24173), was ob-tained similarly, using the primers 5�-ATTC GAA TTCGTC AGA GTC TAG GGG AAT TG-3� (forward) and5�-ATCC GGA TCC ACG AAG TCA AAG TTC CACCG-3� (reverse). The PCR product was digested with EcoRIand BamHI and subcloned into the corresponding sites ofpBS II KS (Stratagene). Subsequently, the plasmid waslinearized with EcoRI, and an antisense transcript was ob-tained by using T7 RNA polymerase. The mouse K18cDNA was a gift of Dr. Robert Oshima (the BurnhamInstitute), and a riboprobe was made by using SP6 RNApolymerase after the plasmid was linearized with EcoRI.The �-actin control riboprobe was obtained from Ambion,Inc (Austin, TX). The mouse EGF-R, ErbB2, ErbB3, wheyacidic protein (WAP), �- casein, and �-lactalbumin cDNAswere obtained from Drs. Frank Jones and David Stern in theDepartment of Pathology at Yale University School of Med-icine. The ErbB2 and ErbB3 plasmids were both linearizedwith BamHI, prior to transcription with T7 RNA polymer-ase, while the EGF-R plasmid was linearized with HindIIIand transcribed with T3 RNA polymerase. In situ hybrid-ization with the other plasmids was performed essentially asdescribed (Jones et al., 1999), except that probes wereradiolabeled by using �33P UTP.

Results

Expression of endogenous AP-2 genes in the mousemammary gland

The majority of mammary gland development occursduring puberty and pregnancy, in response to changes inhormone levels (Daniel and Silverstein, 1987; Hen-nighausen and Robinson, 1998). At birth, the mammarygland consists of a rudimentary network of ducts that oc-cupies a small region of the fat pad in the vicinity of thenipple. Outgrowth and bifurcation of the ducts occur inresponse to estrogen during puberty. Expansion of the lobu-loalveolar network and differentiation into a secretory organoccur in pregnancy and lactation, respectively. Weaningleads to involution, and the mammary gland is eventuallyremodeled to resemble the virgin state. To determine howthe AP-2 genes are regulated during these various stages ofpostnatal mammary gland development, we examined bothRNA and protein expression. Transcripts corresponding toall AP-2 family members, with the exception of AP-2�,were readily detected in mammary tissue (Fig. 1A, and datanot shown). RNase protection analysis indicated that theAP-2�, AP-2�, and AP-2� genes were all expressed in a

132 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 7: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Fig. 3. The MMTV-AP-2� transgene. (A) The structure of the MMTV-AP-2� construct. The locations of the PCR primers used to identify transgenic progeny(arrows), and of the cRNA probe used for the RNase protection analyses (underlined), are shown. (B) RNase protection analysis of transgene expression inthe mammary tissue of transgenic founder offspring. The numbers refer to the identity of the founder mice. (C–F) Detection of transgene protein expressionusing a polyclonal antiserum specific for the FLAG epitope tag. Mammary tissues were derived from normal mice (C, E) or from siblings that possessed theMA44 transgene insertion (D, F). Mammary tissues were obtained from 6-month-old virgin mice (C, D) and mice at 14 days of pregnancy (E, F). Thelocations of ductal and alveolar epithelial cells are indicated, respectively, by arrows and arrowheads.

133J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 8: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

similar manner at various stages in the growth of the mam-mary gland (Fig. 1A, and data not shown). Transcripts fromall three genes were readily detected in the early virginmammary tissue, and expression levels continued to in-crease as the mammary gland matured. Higher levels ofAP-2 expression were observed in pregnant mammary tis-sue. As gestation progressed, the expression of all threeAP-2 genes continued to increase and reached a peak at 15days of pregnancy, the latest pregnant mammary tissueanalyzed. The expression of these genes was sharply re-duced in lactating mammary tissue, but expression wasreactivated during the stage of involution.

We next utilized immunological reagents specific forAP-2� and AP-2� to examine the mammary gland celltypes in which these two AP-2 proteins were expressed(Turner et al., 1998; Zhang et al., 1996). Immunohistochem-ical analysis with these reagents revealed that both theAP-2� and AP-2� transcription factors were localized in thenuclei of the ductal epithelium of the virgin mouse mam-

mary gland (Fig. 1B and E). Staining for both proteins wasalso apparent in the body cells of the terminal end buds (Fig.1C and F). At later stages, the presence of AP-2� andAP-2� could also be observed in the alveolar epithelium ofpregnant mice (Fig. 1D and G). The AP-2 proteins werereduced to an undetectable level in the lactating mammarygland, but became evident again during involution (data notshown). The AP-2� protein was expressed in a similardevelopmental profile in the ductal and alveolar epithelia,although in this instance, detailed examination was notpossible with the available immunological reagents (datanot shown). Together, these data show an excellent corre-lation with the RNase protection analyses concerning thedevelopmental profile of AP-2 expression in the mousemammary gland. However, there were qualitative differ-ences between the expression of AP-2� and AP-2�. First,during the initial phase of lactation, AP-2� transcripts weremaintained at higher levels than AP-2� transcripts. Second,in both virgin and pregnant animals, the AP-2� protein was

Fig. 4. Morphology of transgenic mammary glands from virgin mice. Whole-mount preparations are shown of the fourth inguinal mammary gland of eithernormal mice (A, C, and E), or transgenic siblings (B, D, and F). Mammary glands are from 8-week-old virgin mice (A, B) or from virgin mice at 6 monthsof age (C–F). The dark oval in the center of the gland is a lymph node. (E, F) Higher magnifications of the periphery of the glands shown in (C) and (D).Arrows indicate alveolar buds.

134 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 9: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

present in the majority of ductal and alveolar epithelial cells.In contrast, AP-2� was expressed in approximately 70–80% of the virgin ductal epithelial cells and displayed var-iegated expression in the alveolar clusters during preg-nancy. Further differences between AP-2� and AP-2�expression were observed in the terminal end buds (TEBs).These structures are responsible for the outgrowth of theductal network into the surrounding fat pad. Two cell pop-ulations, termed cap cells and body cells, have been iden-tified within the TEB. Cap cells will form the myoepitheliallayer, and body cells will form ductal epithelial tissue. TheAP-2� protein was present in the majority of both the body(Fig. 1C, arrow) and cap cells (Fig. 1C, arrowhead), al-though the latter cells had lower expression levels. AP-2�was also present in both body and cap cells. However,AP-2� was present within fewer cells of the TEB, and thelevel of expression within an individual body cell (Fig. 1F,arrow) or cap cell (Fig. 1F, arrowhead) was equivalent.

We have recently generated a mouse strain in which thebacterial lacZ gene is inserted in- frame into the codingregion of the AP-2� gene locus (Brewer et al., 2002). Micethat are heterozygous for this allele are viable and fertile andprovide an additional means by which the expression profileof the AP-2� locus can be followed during postnatal devel-opment of the mammary gland. In these mice, �-galactosi-dase activity is detected in the ductal epithelium and TEBsof virgin animals, and continues to be expressed in the ductsand lobuloalveoli during pregnancy (Fig. 2A–D). Expres-sion of the AP-2� lacZ allele is undetectable during lacta-tion, but �-galactosidase activity is restored during regres-sion (Fig. 2E–H). These results provide an independentconfirmation of our previous findings concerning the spatialand temporal expression pattern of the AP-2� gene in thepostnatal mammary gland.

Generation and characterization of AP-2� transgenicmouse lines

The dynamic expression profile of the AP-2 proteins inthe mammary gland suggested that these transcription fac-tors could be involved in regulating gene expression duringmammary gland growth and morphogenesis. We utilized atransgenic approach to test this hypothesis. The humanAP-2� cDNA was placed under the control of the MMTVLTR (Fig. 3A), and injected into one- cell mouse embryos.The human and mouse AP-2� proteins are almost identicalat the protein level, differing by only 1 out of 437 aminoacids. The utilization of the human transgene ensured thatits expression would be readily distinguished from the en-dogenous mouse gene by RNase protection analysis. Wealso attached a FLAG epitope tag to the C terminus of thetransgene, and this tag enables the protein derived from thetransgene to be distinguished from the endogenous AP-2�protein. Ten transgenic founder mouse lines were generatedas assessed by Southern blot analysis. To examine transgeneexpression, RNase protection assays were performed on

offspring of the MMTV AP-2� founders by using totalRNA isolated from mammary tissue of 14-day pregnantmice. Seven of these transgenic lines expressed significantlevels of the human AP-2� transgene in the mammaryglands (Fig. 3B), and three lines, MA7, MA14, and MA44,were kept for further analysis. We also determined that, inthese three lines, lower levels of transgene expression oc-curred in the lung, salivary gland, and kidney, but no ex-pression was detected in the ovaries (data not shown). Withthe exception of the mammary gland phenotype discussedbelow, no gross anatomical differences between transgenicmice and their wild-type littermates were detected in anytissue.

We next utilized immunohistochemistry to examine therelative expression levels of the transgene in the variousmammary gland cell populations of the MA44 transgenicmice and nontransgenic siblings (Fig. 3C–F). Mammarytissues taken from both virgin and pregnant mice wereprobed with a polyclonal antiserum specific for the FLAGepitope tag. Transgene expression was clearly detected inboth ductal (arrows) and alveolar (arrowheads) epithelialcells of the MA44 mouse line (Fig. 3D and F). In contrast,transgene product was not detected in these cell types innontransgenic sibling mammary tissue (Fig. 3C and E).Note that expression of the transgene was found to benonuniform in the ductal epithelia of the mammary gland atvarious stages of development (for example, Fig. 3D). Var-iegated expression is frequently seen for mammary glandtransgenes driven by the MMTV, WAP, or �-lactoglobulinpromoters (Barash et al., 1999; Deckard-Janatpour et al.,1997; Faerman et al., 1995; Jones and Stern, 1999; Jones etal., 1999; Robinson et al., 1995). There is also evidence thatendogenous genes, such as the progesterone receptor andthose encoding milk proteins, are not uniformly expressedin all cells of the ductal or lobuloalveolar epithelium of themouse mammary gland (Robinson et al., 1995; Silbersteinet al., 1996). Therefore, the differential transgene stainingwe observe is not atypical. Nevertheless, the mosaic natureof our transgene’s expression does preclude a straightfor-ward morphometric analysis of the mammary gland as awhole.

Overexpression of AP-2� inhibits alveolar budding in thevirgin mouse mammary gland

As shown above, the analysis of the FLAG-stained tissuefrom virgin and pregnant mice indicated that we had beensuccessful in targeting the AP-2� transgene to the appro-priate mammary epithelial layers. It was also evident froman examination of the sections at lower magnification thatthere was a reduced amount of epithelial tissue in the micethat overexpressed AP-2� compared with their wild-typesiblings (data not shown, and see below). To determinefurther the effects of overexpression of AP-2�, mammaryglands from virgin, pregnant, and lactating mice were ex-amined by whole-mount analysis. All three transgenic lines

135J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 10: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

studied in our analysis (MA44, MA14, and MA7) generateda similar mammary gland phenotype (data not shown).Since the MA44 transgenic line produced the most pro-nounced alterations in morphology, mammary glands fromthese mice were studied in detail. The initial phase ofpostnatal mammary gland development, involving ductaloutgrowth through the mammary fat pad, was not affectedin the transgenic mice. Indeed, a comparison of whole-mount mammary gland tissues from a normal littermate(Fig. 4A) and a transgenic mouse (Fig. 4B) at 8 weeks ofage indicated that the ductal network had filled the entire fatpad in both instances. In older virgin mice, however, therewas a dramatic difference in the morphology of the ducts.At 6 months of age, the ductal system of a normal littermatehad a highly branched organization that was associated withmultiple alveolar bud structures (Fig. 4C and E, arrows). Incontrast, mammary glands from the transgenic mice dis-played a simpler, sparser, ductal network. While there wasno alteration in the number of major side-branches, wenoted a dramatic reduction in the frequency of the alveolarbuds (Fig. 4D and F, arrows).

Overexpression of AP-2� causes defects in lobuloalveolardevelopment during pregnancy and lactation

Subsequent examination of the transgenic mice duringpregnancy demonstrated significant underdevelopment ofthe lobuloalveolar structures when compared with their non-transgenic littermates. By day 15 of pregnancy, the normalmammary gland had developed well-formed alveoli (Fig.5A and C, arrows). In contrast, the transgenic gland had farfewer alveoli, and these were smaller and more compact(Fig. 5B and D, arrows). Strikingly, ducts were clearlyvisible in whole-mount preparations of transgenic mam-mary glands (Fig. 5D, arrowheads), whereas these structureswere obscured by lobuloalveolar expansion in normal con-trol mice. Histological examination confirmed that trans-genic mice had fewer clusters of lobuloalveoli, and showedthat each individual alveolus was smaller and more con-densed as compared with the normal littermate (Fig. 5E andF). Moreover, in contrast to the abundant presence of cel-lular and luminal lipids in normal littermates at this stage(Fig. 5E, red arrows), we did not observe the accumulationof such lipids in the lobuloalveoli of transgenic mice (Fig.5F).

We next examined whether the decreased quantity oflobuloalveolar tissue in the transgenic mice resulted fromalterations in cell death and/or cell proliferation. TheTUNEL assay was used to examine the incidence of celldeath in the mammary glands of wild type and transgenicmice at 13.5 and 18.5 days of pregnancy (Table 1, and datanot shown). Few apoptotic cells were observed in the wildtype mammary epithelial tissue at either time point (0.38and 0.16% cells, respectively), but there was a significantincrease in apoptosis at both stages of mammary glanddevelopment in the transgenic mice (0.96 and 0.5% cells,

respectively). In the transgenic mammary glands, apoptoticcells were often found in regions that contained the charac-teristic compact alveolar tissue described above (data notshown). We next determined the distribution of proliferat-ing cells after 2 h of BrdU labeling in wild type andtransgenic mammary glands at 13.5 and 18.5 days of preg-nancy (Table 2). In both strains of mice, the greatest num-bers of labeled cells were observed at the 13.5-day timepoint, when the alveolar network is rapidly increasing insize. However, at this time point, nontransgenic mammaryglands displayed two- to threefold greater levels of prolif-erating cells than their transgenic counterparts. Similar dif-ferences in proliferation between the two strains of micewere also observed at 18.5 days of pregnancy. We alsoexamined cell proliferation in the wild type and transgenicanimals using immunohistochemistry for the phosphohis-tone H3 cell proliferation marker. Again, we found thatthere were more labeled cells in wild-type than transgenicmammary glands at these two time points and the quiescentstate tended to coincide with regions of high transgeneexpression (data not shown). Note that the differences seenin proliferation and apoptosis between wild type transgenicmice probably underestimate the influence of ectopic AP-2�expression on these cellular processes since the MA44mammary glands are mosaic, not uniform, with respect totransgene expression. Taken together, these findings indi-cate that the reduction in lobuloalveolar tissue in the trans-genic mice results from a combination of decreased prolif-eration and increased cell death.

Morphological differences between the mammary glandsof transgenic and control mice were also apparent duringlactation (Fig. 6). Whole-mount staining performed at 1 daypostpartum revealed an extensive secretory network in nor-mal mice, in which lobuloalveoli were expanded due to thepresence of lactation products (Fig. 6A). In contrast, thelobuloalveoli of transgenic mice were sparser and remainedmore condensed (Fig. 6B). One consequence of the under-developed lobuloalveolar structures is that the ducts werestill clearly visible in the transgenic mice compared withtheir normal siblings (Fig. 6B, blue arrows). The ductsthemselves had a similar distended appearance in both wild-type and transgenic animals, consistent with milk secretion(see below). Histological analysis at 1 day postpartum re-vealed further differences between wild-type and transgenicmammary glands. First, more lobuloalveolar clusters werepresent in the normal mammary glands (Fig. 6C) than insibling transgenic mice (Fig. 6D). Second, alterations werealso apparent with respect to the overall architecture of themammary gland and lobuloalveolar morphology. In controlanimals, the lobuloalveolar lumens were engorged withlactation products, and the alveolar epithelial cells had aflattened morphology, indicating that these cells had under-gone secretory differentiation (Fig. 6C). Moreover, al-though there was a limited degree of heterogeneity in thecontrol animals, the lobuloalveolar structures from thesemice were generally uniform in appearance throughout the

136 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 11: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

gland. In contrast, the mammary glands from transgenicmice displayed considerable heterogeneity in the morphol-ogy of the lobuloalveoli (Fig. 6D). A subset of the glandcontained alveolar epithelial cells typical of this maturationstage (Fig. 6D, arrows), although sometimes the lumens inthese regions could be more distended, a phenotype also

noted for mice carrying a dominant negative FGFR2 trans-gene (Jackson et al., 1997). However, other regions of thegland appeared to be at an earlier stage in the maturationprocess, since alveolar cells were more cuboidal in shape,and the lumens contained lower amounts of lactation prod-ucts (Fig. 6D, arrowheads).

Fig. 5. Morphology of transgenic mammary glands at 15 days of pregnancy. (A–D) Whole-mount preparations are shown of the fourth inguinal mammarygland from either a normal mouse (A, C) or a transgenic sibling (B, D). (C, D) Higher magnifications of the periphery of the glands shown in (A) and (B).Arrows indicate alveoli, and arrowheads show ducts. (E, F) Histology of mammary tissue taken from a normal mouse (E) or from a transgenic sibling (F).Red arrows indicate intracellular lipids; red arrowheads indicate condensed lobuloalveoli.

137J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 12: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

By 7 days postpartum, lobuloalveolar lumens wereprominent throughout the whole mammary pad of nontrans-genic mice (Fig. 6E). Histological sections of these normalmice indicated that stromal fat cells were rarely observed

and that the lobuloalveolar lumens were engorged withsecretory proteinacious materials (Fig. 6E, red arrows). Incontrast, fat cells were still readily apparent in the trans-genic mammary glands at 7 days postpartum (Fig. 6F,

Fig. 6. Morphology of postpartum transgenic mammary glands. (A, B) Whole-mount preparations are shown of the fourth inguinal mammary gland of anormal mouse (A), or a transgenic sibling (B), at 1 day postpartum. Blue arrows indicate ducts. (C, D) Histology of the mammary tissue shown in (A) and(B), respectively. Arrows show tissue that has a normal appearance for this stage of mammary gland maturation, and arrowheads indicate tissue that is lesswell-developed. (E, F) Histology of mammary tissues taken from 7 days postpartum normal (E), or transgenic (F), sibling mice. Red arrows indicate secretoryproteinacious material; red arrowheads show lipids within the immature epithelial cells; and the asterisk identifies fat cells. Pups were removed from themothers 5–6 h before sacrifice and mammary gland isolation.

138 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 13: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

asterisk). Furthermore, some alveolar epithelial cells fromthe transgenic mice still retained a cuboidal shape, and largeintracellular fat droplets were still visible (Fig. 6F, redarrowheads), features which are more typical of late preg-nancy (Hollmann, 1969). Despite the variegated phenotype,the AP-2� transgenic mothers were able to feed litters ofnormal size, indicating that sufficient milk production isoccurring.

Analysis of gene expression during pregnancy andlactation

Interestingly, the mammary glands of transgenic micethat overexpressed dominant negative versions of ErbB2 orErbB4 also contained regions in which alveolar clustersmore typical of late pregnancy were present during lactation(Jones and Stern, 1999; Jones et al., 1999). However, theinfluence of these two transgenes on lactogenesis could bedistinguished by their influence on milk protein gene ex-pression. Therefore, we further investigated the effect ofexogenous AP-2� on mammary gland development by ex-amining the expression of three molecular markers of lac-tation: whey acidic protein, �-lactalbumin, and �-casein. Insitu hybridization studies were performed on serial sectionsof wild-type and transgenic mammary tissue at 1 day post-partum, and the results were compared with the presence ofthe FLAG epitope on adjacent sections. FLAG epitopestaining revealed that exogenous AP-2� protein was presentthroughout the transgenic gland, in both the cuboidal epi-thelial cells and the more flattened epithelial cells (Fig. 7,and data not shown). However, the level of transgene ex-pression was variable within an individual gland, with atendency for higher levels of FLAG reactivity to occur inthe epithelial cells with the less mature, cuboidal, morphol-ogy. In situ hybridization studies were performed by usingsense and anti-sense probes specific for the three milk pro-tein genes. No significant hybridization was observed withthe sense probes (data not shown). Data obtained using theanti-sense probes indicated that these three milk protein

genes were expressed almost uniformly in both transgenicmice (Fig. 7) and wild-type mice (data not shown). Notably,in the transgenic mice, uniform expression of these genesoccurred despite the distinct lobuloalveolar morphologiespresent in different regions of the gland. Taken together,these findings indicated that the overexpression of AP-2�could alter lobuloalveolar morphology but did not preventtranscription of the major milk protein genes.

Next, we investigated the mechanism by which the over-expression of AP-2� might influence the postnatal devel-opment of the mammary gland. The expression of severalgenes known to regulate the growth and morphogenesis ofthe mammary gland and/or postulated to be AP-2 targetswere examined in 12-day pregnant wild-type and transgenicmice, using a combination of RNase protection and semi-quantitative RT-PCR analyses (Fig. 8, and data not shown).These studies revealed an �3-fold increase in the RNAlevels for the parathyroid hormone-related protein (PTHrP)in the mammary glands of transgenic mice compared withtheir wild-type littermates (Fig. 8). The increase in PTHrPexpression was also observed by using semiquantitativeRT-PCR (J.Z., data not shown). Previous studies haveshown that the PTHrP promoter is directly activated byAP-2 in vitro (Prager et al., 1994). Moreover, mice thatexpress a PTHrP transgene in the mammary gland alsodisplay an inhibition of development (Wysolmerski et al.,1995). Therefore, the increase in PTHrP expression seen inthe AP-2� transgenic mice represents a plausible mecha-nism by which mammary gland morphology is altered inthese animals. In contrast to the changes in PTHrP expres-sion, we did not detect any differences in transcript levelsbetween transgenic mice and wild-type littermates for threefurther potential AP-2 target genes, p21waf1/cip1, ErbB2, andErbB3 (Fig. 8). We also found no significant alteration inthe expression of several additional genes which encodegrowth factor signaling molecules associated with mam-mary gland morphogenesis, including wnt4, the progester-one receptor, the prolactin receptor, EGF-R, and ErbB4(Fig. 8, and data not shown).

Table 1Comparison of apoptosis levels in mammary epithelial cells betweentransgenic and wild type mice

No. apoptotic cells per 1000

at 13.5 days of pregnancy at 18.5 days of pregnancy

MA44 transgenic Nontransgenic MA44 transgenic Nontransgenic

8 4 6 210 2 4 011 6 8 47 3 3 1

12 4 4 1Average 9.6 Average 3.8 Average 5 Average 1.60.96% 0.38% 0.5% 0.16%

Note. Student’s t test probability at 13.5 days: P � 0.001; at 18.5 days:P � 0.0004.

Table 2Comparison of proliferation levels in mammary epithelial cells betweentransgenic and wild type mice

No. proliferating cells per 1000

at 13.5 days of pregnancy at 18.5 days of pregnancy

MA44 transgenic Nontransgenic MA44 transgenic Nontransgenic

21 42 4 1130 36 8 1717 48 2 2023 56 5 1419 47 6 16Average 22 Average 45.8 Average 5 Average 15.62.2% 4.6% 0.5% 1.6%

Note. Student’s t test probability at 13.5 days: P � 0.0003; at 18.5 days:P � 0.016.

139J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 14: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Discussion

Previous studies have demonstrated that the AP-2 familyof transcription factors regulates multiple aspects of mam-malian development. Formation of the neural tube, face,eyes, limbs, heart, ventral body-wall, and kidneys all relyupon the appropriate expression of the AP-2 genes duringmouse embryogenesis (Brewer et al., 2002; Moser et al.,1997a; Nottoli et al., 1998; Schorle et al., 1996; West-Mayset al., 1999; Zhang et al., 1996). The observation that thehuman AP-2 genes are also expressed in both benign mam-mary epithelial tissue and early stage breast cancer indicatedthat these transcription factors might also be important forregulating the growth and morphogenesis of the postnatalmammary gland (Gee et al., 1999; Turner et al., 1998). Ourcurrent studies, in which we have first examined the expres-sion profile of the AP-2�, AP-2�, and AP-2� genes in thepostnatal mouse mammary gland, and subsequently manip-ulated the levels of the AP-2� protein in this organ, stronglysupport this hypothesis.

The spatiotemporal expression pattern of the AP-2 genefamily throughout mouse mammary gland maturation andremodeling was determined by using a combination ofRNase protection, immunohistochemistry, and lacZ geneknock-in technology. These complimentary techniques in-dicated that all three genes were present in the ductal epi-thelia of the postnatal mouse mammary gland, but showedthat they were not expressed in the surrounding myoepithe-lium. These findings support our previous contention thatthe AP-2 proteins are a regular component of the genenetwork expressed in normal human breast epithelial tissue(Turner et al., 1998). However, in our previous studies usinghuman tissue samples, it was not possible to assess theexpression profile of the AP-2 genes at specific stages ofdevelopment. Using the mouse model system, we now showthat expression of all three AP-2 genes occurs in the virginmammary gland. AP-2 expression increases in the virgingland during the period of ductal outgrowth and continuesto rise during pregnancy as lobuloalveolar developmentproceeds. Comparison with cytokeratin expression indi-cated that the general rise in AP-2 expression reflected thelarge increase in mammary ductal epithelial tissue duringthese growth periods and was not caused by increased AP-2levels in individual cells (J.Z., unpublished observations). Incontrast, it is clear that AP-2 expression drops off dramat-ically in the mammary epithelial cells during lactation,before it is reactivated with the remodeling of the mammarygland that occurs during involution. Detailed examinationindicated that the AP-2� and AP-2� proteins were not onlypresent in the ductal epithelial tissue, but were also foundwithin the TEBs. Significant AP-2� expression occurred inthe majority of the TEB body cells, while much lower levelswere apparent in cap cells. In contrast, AP-2� expressionwas apparent in a subpopulation of both the body cells andcap cells. Differences in the distribution of AP-2� andAP-2� proteins within these TEB regions indicate that they

may perform distinct functions in the growth and morpho-genesis of the ductal network. One possibility is that highlevels of AP-2� are consistent with a body cell and ductalepithelial cell lineage, while the down-regulation and even-tual loss of AP-2� would mark cap and myoepithelial cells.Similarly, the differential expression of AP-2� in the TEBmight indicate cells that have different fates with respect togene expression, proliferation, or apoptosis (Humphreys,1999; Seagroves et al., 2000).

The expression profile of the AP-2 genes is similar tothat of several key regulators of mammary gland develop-ment and maturation. In other development processes, theAP-2 transcription factors, and in particular AP-2�, controlmultiple aspects of growth and morphogenesis. We there-fore wished to determine whether this key regulatory mol-ecule could also influence the morphology of the mammaryductal network. Moreover, since the levels of AP-2 areincreased in breast cancer, we wished to ascertain whetherincreased levels of AP-2� might lead to mammary tumorformation. For these purposes, we generated transgenicmice that overexpressed the AP-2� gene under the controlof the MMTV LTR. Our findings indicate that the overex-pression of AP-2� causes a unique spatiotemporal sequenceof changes in mammary gland morphology but does notresult in increased tumorigenesis. In some instances, trans-gene expression in the endocrine organs, especially theovaries, can affect mammary gland morphology by chang-ing circulating hormone levels. However, two lines of evi-dence indicate that AP-2� is acting autonomously withinthe mammary glands of our transgenic mice. First, we didnot detect transgene expression within the ovaries. Second,the mammary glands examined in our studies displayedmorphological heterogeneity that coincides with transgeneexpression. Indeed, the presence of the epitope tag served asa powerful means by which transgene expression could becorrelated with morphological differences in the gland.

In the transgenic mice, there were no obvious defects inthe outgrowth of the ducts, nor in the bifurcation of theindividual ducts, two processes that occur during puberty inresponse to estrogen. However, there was a significant im-pairment of alveolar bud formation in the mammary glandsof virgin mice. Furthermore, during pregnancy, transgenicmice had a less extensive ductal network than was observedin control mice, and the lobuloalveolar cells also remainedcondensed and failed to accumulate lipids at the appropriatetime points. One mechanism by which the overexpressionof AP-2� might directly inhibit ductal proliferation isthrough increased expression of the p21waf1/cip1 cell cycleinhibitor, since the human version of this gene is regulatedby AP-2 in vitro (Zeng et al., 1997). However, we wereunable to detect significant changes in the levels of mousep21waf1/cip1 in vivo between wild-type and transgenic mam-mary tissue. Nevertheless, we did note that there was asignificant decrease in cell proliferation in the transgenicepithelial tissue as measured by BrdU incorporation and theanalysis of markers of cell proliferation, including phospho-

140 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 15: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

histone H3. These findings indicate that a reduction inactively cycling cells in the mammary gland indeed accom-panied the overexpression of AP-2�, and this may in partaccount for the phenotype of the transgenic tissue in thevirgin and pregnant animals. Cell death, which is increased

in the presence of the transgene, may act as a secondmechanism by which the lobuloalveolar tissue is reduced inthe transgenic mice. A link between altered levels of AP-2expression and an increase in cell death has also beenpreviously noted in both AP-2� and AP-2� knockout mice

Fig. 7. Milk protein gene expression in transgenic mouse mammary glands at 1 day postpartum. (A, C, and E) Immunohistochemical detection of the FLAGepitope tag. Darkfield images of in situ hybridizations performed with antisense probes corresponding to WAP (B), �-casein (D), and �-lactalbumin (F) arepresented for adjacent sections.

141J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 16: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

(Moser et al., 1997a; Schorle et al., 1996; Zhang et al.,1996). In these latter instances, it is possible that the in-crease in cell death results from either inappropriate cellcycle progression or defective cell:cell signaling. Similarly,the alterations in cell death and cell proliferation we notedabove may be the direct result of AP-2� overexpression, ormay occur indirectly if AP-2� instead influences the perti-nent signal transduction pathways for mammary gland de-velopment. Note that we have recently obtained furthersupport for the hypothesis that AP-2� can control prolifer-ation of mammary epithelia from transplantation studies inwhich mammary buds derived from AP-2�-null embryos orfrom heterozygous siblings have been transplanted into thecleared fat pads of immunodeficient host mice. Preliminaryresults indicate an increase in lobuloalveolar structures dur-ing pregnancy in transplants derived from AP-2�-null em-bryos compared with those obtained from heterozygouscontrols (S.B., J. Wysolmerski, and T.W., unpublished ob-servations). Together, these findings support a model inwhich the levels of AP-2� can modulate alveolar density—the loss of AP-2� expression causes an increase in alveolardevelopment, whereas excess AP-2� is inhibitory.

Normal side-branching and lobuloalveolar developmentrely on the presence of intact progesterone and prolactinsignaling pathways in the ductal epithelia (Robinson et al.,2000). The misexpression of several additional key regula-tory molecules, some of which act as targets for thesehormone signaling cascades, can also result in an underde-veloped ductal network. In particular, such phenotypes canbe caused by a mutation of either cyclin D1, Stat5a, wnt4,inhibin�B, C/EBP�, or A-myb (Fantl et al., 1999; Robinsonand Hennighausen, 1997; Robinson et al., 2000; Teglund etal., 1998; Toscani et al., 1997), or by overexpression ofcomponents of the TGF-�, FGF, and parathyroid hormone-related protein (PTHrP) signaling pathways (Jackson et al.,1997; Jhappan et al., 1993; Pierce et al., 1993; Wysolmerskiet al., 1995). We have examined several of these genes todetermine whether their transcript levels were altered inresponse to the AP-2� transgene. We did not detect anydifferences between transgenic mice and their wild-typelittermates with respect to the progesterone receptor, theprolactin receptor, or wnt4. However, we did observe a

consistent �3-fold increase in transcripts corresponding tothe signaling molecule parathyroid hormone-related protein(PTHrP) in the transgenic mice. Intriguingly, previous stud-ies have shown that the overexpression of PTHrP in themyoepithelial compartment of the mammary gland inhibitsboth side-branching in virgin mice and lobuloalveolar de-velopment during pregnancy (Wysolmerski et al., 1995).Therefore, it is possible that altered AP-2� levels causechanges in mammary gland development in part by affect-ing PTHrP expression, especially since the PTHrP promotercan be activated by AP-2 in vitro (Prager et al., 1994). In thefuture, it will be of interest to determine whether compo-nents of the PTHrP pathway are indeed downstream targetsfor the AP-2 transcription factors. The finding that trans-genic mice show no change in the expression of otherpostulated AP-2 gene targets, such as p21waf1/cip1, couldresult from a number of factors. First, the results may reflectdifferences in promoter activity measured here in vivo,versus in vitro, where many of these human targets werecharacterized. Alternatively, these findings may indicatethat there are sequence differences between the promotersof the human and mouse target genes that may alter theirability to bind and respond to AP-2 (Bates and Hurst, 1997).Finally, it is also possible that a different AP-2 protein isrequired for expression of these target genes or that AP-2�cannot act alone but needs to function in concert with otherAP-2 proteins or other transcription factors that are limitingin vivo (Turner et al., 1998).

Following parturition, the sparser appearance of the duc-tal tree in AP-2� transgenic mice also persisted into lacta-tion. Even at 7 days of lactation, some areas of the glandwere still occupied by lobuloalveolar structures that weremore appropriate for an earlier stage of the differentiationprocess, namely late pregnancy or parturition. Note that wedid not detect transgene expression at 7 days of lactation byeither immunohistochemistry or RNase protection in thesealtered regions (J.Z., data not shown), a result that has alsobeen observed for at least one other MMTV transgeneconstruct (Jones and Stern, 1999). Therefore, we postulatethat the earlier presence of high levels of AP-2� during latepregnancy has prevented maturation in these areas. We alsohypothesize that regions having lower levels of transgeneexpression during pregnancy would instead escape from theinfluence of AP-2� and would proliferate and differentiateappropriately. Different levels of transgene expressionwould therefore lead to the observed variegation in themorphology of the mammary glands. This hypothesis issupported by the observation that, at 1 day postpartum, thereis a higher incidence of transgene expression in regions ofthe mammary gland that are delayed in differentiation com-pared with areas that are more mature.

We further addressed the differentiation state of themammary epithelia by studying the expression of threemolecular markers of lactation: whey acidic protein (WAP),�-casein, and �-lactalbumin. Using in situ hybridization, wefound that all three genes were expressed in the transgenic

Fig. 8. Gene expression in normal and transgenic mammary glands at 12days of pregnancy. The top panel shows RNase protection data for trans-genic (�) and wild-type (�) littermates for the genes indicated above.Reactions were normalized by simultaneously assaying for the expressionof the mouse K18 gene (bottom panel), a cytokeratin that is expressed inthe mammary epithelium. Each RNA sample was derived from two miceof the respective genotype.

142 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 17: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

mammary gland at 1 day of lactation. Moreover, transcriptswere present not only in the flattened epithelial cells, char-acteristic of this stage of lactation, but also in the atypicaland morphologically distinct cuboidal epithelial cells. Thus,our data suggest that, although the MMTV AP-2� transgenemay alter the morphology of the mammary ductal epithe-lium, it does not inhibit the expression of the major lactationproteins. A similar relationship between abnormal morphol-ogy and normal transcript levels of these milk protein genesis typical of inhibin�B knockout mice as well as miceexpressing the dominant negative ErbB2 transgene (Jonesand Stern, 1999; Robinson and Hennighausen, 1997). Inother instances, notably dominant negative ErbB4 trans-genic mice and mice lacking the STAT5a gene, alteredmorphology is accompanied by a decrease in expression ofone or more of the milk protein genes (Jones et al., 1999;Liu et al., 1997; Teglund et al., 1998). When taken together,the AP-2� transgenic phenotype has a distinctive combina-tion of changes in morphology and in gene expression thatdistinguish it from other mutations affecting mammarygland development.

Despite the link between AP-2 protein expression andhuman breast cancer, to date we have not detected anymammary tumors in the MMTV- AP-2� transgenic mice,many of which have been kept for greater than 18 months.Indeed, the overexpression of AP-2� suppressed the growthand development of the ductal network. We therefore pos-tulate that, in human breast cancer, the rise in AP-2� ex-pression is not responsible for the inappropriate cell cycleprogression. Instead, we suggest that the activation of theAP-2 genes in breast cancer represents a failed attempt tohalt cell proliferation. The conclusion that the AP-2 genesmay have an inhibitory role in tumor progression is sup-ported by recent in vitro and in vivo studies. First, it hasbeen shown that transfection of AP-2� into HepG2 hepa-tocarcinoma cells or SW480 adenocarcinoma cells can in-hibit their growth and tumorigenicity, possibly by activatingthe transcription of p21waf1/cip1 (Zeng et al., 1997). Second,there is a tendency for AP-2 gene expression to be lostduring the process of metastasis in both breast cancer andmelanoma (Bar-Eli, 1999; Gee et al., 1999), while the ec-topic expression of AP-2� in melanoma cell lines can re-duce the tumorigenicity of these cells when they are trans-planted into nude mice (Bar-Eli, 1999).

Considerable amino acid identity exists between the AP-2�, AP-2�, and AP-2� proteins, and indeed these transcrip-tion factors can all bind to essentially the same recognitionsite (Bosher et al., 1996). Nevertheless, human breast cancerstudies indicate that these transcription factors may regulatedifferent target genes (Gee et al., 1999; Turner et al., 1998).Subtle differences in the spatiotemporal patterns of AP-2expression also occur within the mammary epithelium.Therefore, while the overexpression of AP-2� may beequivalent to expressing any or all of the AP-2 genes, it ispossible that each gene could perform a unique functionwithin the mammary gland—a function that is subverted by

the overexpression of one particular family member. Con-tinued analysis of how these AP-2 genes might act alone orin concert within the mammary gland is clearly warranted.In the context of the whole organism, the demonstration thatthe AP-2� protein can profoundly influence postnatal mam-mary gland morphology again illustrates the importance ofthis gene family for regulating growth and morphogenesis.Furthermore, the ability of AP-2� to inhibit lobuloalveolarproliferation strongly suggests that this gene family canmodulate cell growth in human breast cancer.

Acknowledgments

We thank F. Jones, J. Wysolmerski, D. Stern, HelenHurst, and the Section of Critical Technologies of the YaleUniversity School of Medicine for their considerable assis-tance. We thank Mei Han, Yu Ji, and Linda B. Lewis fortechnical help, and Margaret Neville and Michael Rudolphfor discussion of their gene microarray data prior to publi-cation. We are also grateful to the Williams Laboratory, inparticular H. Auman, for critical reading of the manuscript.J.Z. was the recipient of a breast cancer research fellowshipfrom the Department of Defense (DAMD17-96-6094). Thisresearch was supported by National Institutes of HealthGrant CA77833 (to T.W.).

References

Auman, H.J., Nottoli, T., Lakiza, O., Winger, Q., Donaldson, S., Williams,T., 2002. Transcription factor AP-2� is essential in the extraembryoniclineages for early postimplantation development. Development 119,2733–2747.

Bar-Eli, M., 1999. Role of AP-2 in tumor growth and metastasis of humanmelanoma. Cancer Metastasis Rev. 18, 377–385.

Barash, I., Faerman, A., Richenstein, M., Kari, R., Damary, G.M., Shani,M., Bissell, M.J., 1999. In vivo and in vitro expression of human serumalbumin genomic sequences in mammary epithelial cells with beta-lactoglobulin and whey acidic protein promoters. Mol. Reprod. Dev.52, 241–252.

Bates, N.P., Hurst, H.C., 1997. Transcriptional regulation of type I receptortyrosine kinases in the mammary gland. J. Mammary Gland Biol.Neoplasia 2, 153–163.

Bosher, J.M., Totty, N.F., Hsuan, J.J., Williams, T., Hurst, H.C., 1996. Afamily of AP-2 proteins regulates c-erbB-2 expression in mammarycarcinoma. Oncogene 13, 1701–1707.

Bosher, J.M., Williams, T., Hurst, H.C., 1995. The developmentally reg-ulated transcription factor AP-2 is involved in c-erbB-2 overexpressionin human mammary carcinoma. Proc. Natl. Acad. Sci. USA 92, 744–747.

Brewer, S., Jiang, X., Donaldson, S., Williams, T., Sucov, H.M., 2002.Requirement for AP-2� in cardiac outflow tract morphogenesis. Mech.Dev. 110, 139–149.

Brisken, C., Park, S., Vass, T., Lydon, J.P., O’Malley, B.W., Weinberg,R.A., 1998. A paracrine role for the epithelial progesterone receptor inmammary gland development. Proc. Natl. Acad. Sci. USA 95, 5076–5081.

Byrne, C., Tainsky, M., Fuchs, E., 1994. Programming gene expression indeveloping epidermis. Development 120, 2369–2383.

143J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 18: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Chazaud, C., Oulad-Abdelghani, M., Bouillet, P., Decimo, D., Chambon,P., Dolle, P., 1996. AP-2.2, a novel gene related to AP-2, is expressedin the forebrain, limbs and face during mouse embryogenesis. Mech.Dev. 54, 83–94.

Chomczynski, P., Sacchi, N., 1987. Single-step method of RNA isolationby acid guanidinium thiocyanate-phenol-chloroform extraction. Anal.Biochem. 162, 156–159.

Daniel, C.W., Silverstein, G.B., 1987. Postnatal development of rodentmammary gland, in: Neville, M.C., Daniel, C.W. (Eds.), The MammaryGland: Development, Regulation and Function, Plenum Press, NewYork, pp. 3–36.

De Clerk, Y.A., Darville, M.A., Eeckhout, Y., Rousseau, G.G., 1994.Characterization of the promoter of the gene encoding human tissueinhibitor of metalloproteinase-2 (TIMP-2). Gene 139, 185–191.

Deckard-Janatpour, K., Muller, W.J., Chodosh, L.A., Gardner, H.P., Mar-quis, S.T., Coffey, R., Cardiff, R.D., 1997. Differential expression ofthe neu transgene in murine mammary tissues. Int. J. Oncol. 11,235–241.

Faerman, A., Barash, I., Puzis, R., Nathan, M., Hurwitz, D.R., Shani, M.,1995. Dramatic heterogeneity of transgene expression in the mammarygland of lactating mice: a model system to study the synthetic activityof mammary epithelial cells. J. Histochem. Cytochem. 43, 461–470.

Fantl, V., Edwards, P.A., Steel, J.H., Vonderhaar, B.K., Dickson, C., 1999.Impaired mammary gland development in Cyl-1(�/�) mice duringpregnancy and lactation is epithelial cell autonomous. Dev. Biol. 212,1–11.

Fini, M.E., Bartlett, J.D., Matsubara, M., Rinehart, W.B., Mody, M.K.,Girard, M.T., Rainville, M., 1994. The rabbit gene for 92-kDa matrixmetalloproteinase. Role of AP-1 and AP-2 in cell type-specific tran-scription. J. Biol. Chem. 269, 28620–28628.

Gaubatz, S., Imhof, A., Dosch, R., Werner, O., Mitchell, P., Buettner, R.,Eilers, M., 1995. Transcriptional activation by Myc is under negativecontrol by the transcription factor AP-2. EMBO J. 14, 1508–1519.

Gee, J.M., Robertson, J.F., Ellis, I.O., Nicholson, R.I., Hurst, H.C., 1999.Immunohistochemical analysis reveals a tumour suppressor-like rolefor the transcription factor AP-2 in invasive breast cancer. J. Pathol.189, 514–520.

Gille, J., Swerlick, R.A., Caughman, S.W., 1997. Transforming growthfactor alpha induced transcriptional activation of the vascular perme-ability factor (VPF/VEGF) gene requires AP-2-dependent DNA bind-ing and transcriptional activation. EMBO J. 16, 750–759.

Hennig, G., Lowrick, O., Birchmeier, W., Behrens, J., 1996. Mechanismsidentified in the transcriptional control of epithelial gene expression.J. Biol. Chem. 271, 595–602.

Hennighausen, L., Robinson, G.W., 1998. Think globally, act locally: themaking of a mouse mammary gland. Genes Dev. 12, 449–455.

Hollmann, K.H., 1969. Quantitative electron microscopy of sub-cellularorganization in mammary gland cells before and after parturition, in:Reynolds, M., Folley, S.J. (Eds.), Lactogenesis: The Initiation of MilkSecretion at Parturition, University of Pennsylvania Press, Philadel-phia, pp. 37–41.

Humphreys, R.C., 1999. Programmed cell death in the terminal endbud. J.Mammary Gland Biol. Neoplasia 4, 213–220.

Imagawa, M., Chiu, R., Karin, M., 1987. Transcription factor AP-2 medi-ates induction by two different signal-transduction pathways: proteinkinase C and cAMP. Cell 51, 251–260.

Jackson, D., Bresnick, J., Rosewell, I., Crafton, T., Poulsom, R., Stamp, G.,Dickson, C., 1997. Fibroblast growth factor receptor signalling has arole in lobuloalveolar development of the mammary gland. J. Cell Sci.110, 1261–1268.

Jhappan, C., Geiser, A.G., Kordon, E.C., Bagheri, D., Hennighausen, L.,Roberts, A.B., Smith, G.H., Merlino, G., 1993. Targeting expression ofa transforming growth factor beta 1 transgene to the pregnant mam-mary gland inhibits alveolar development and lactation. EMBO J. 12,1835–1845.

Jones, F.E., Stern, D.F., 1999. Expression of dominant-negative ErbB2 inthe mammary gland of transgenic mice reveals a role in lobuloalveolardevelopment and lactation. Oncogene 18, 3481–3490.

Jones, F.E., Welte, T., Fu, X.Y., Stern, D.F., 1999. ErbB4 signaling in themammary gland is required for lobuloalveolar development and Stat5activation during lactation. J. Cell Biol. 147, 77–88.

Kannan, P., Buettner, R., Chiao, P.J., Yim, S.O., Sarkiss, M., Tainsky,M.A., 1994. N-ras oncogene causes AP-2 transcriptional self-interfer-ence, which leads to transformation. Genes Dev. 8, 1258–1269.

Laird, P.W., Zijderveld, A., Linders, K., Rudnicki, M.A., Jaenisch, R.,Berns, A., 1991. Simplified mammalian DNA isolation procedure.Nucleic Acids Res. 19, 4293.

Liu, X., Robinson, G.W., Wagner, K.U., Garrett, L., Wynshaw-Boris, A.,Hennighausen, L., 1997. Stat5a is mandatory for adult mammary glanddevelopment and lactogenesis. Genes Dev. 11, 179–186.

McPherson, L.A., Baichwal, V.R., Weigel, R.J., 1997. Identification ofERF-1 as a member of the AP-2 transcription factor family. Proc. Natl.Acad. Sci. USA 94, 4342–4347.

McPherson, L.A., Weigel, R.J., 1999. AP2alpha and AP2gamma: a com-parison of binding site specificity and trans-activation of the estrogenreceptor promoter and single site promoter constructs. Nucleic AcidsRes. 27, 4040–4049.

Mitchell, P.J., Timmons, P.M., Hebert, J.M., Rigby, P.W.J., Tjian, R.,1991. Transcription factor AP-2 is expressed in neural crest cell lin-eages during mouse embryogenesis. Genes Dev. 5, 105–119.

Mitchell, P.J., Wang, C., Tjian, R., 1987. Positive and negative regulationof transcription in vitro: enhancer-binding protein AP-2 is inhibited bySV40 T antigen. Cell 50, 847–861.

Mohibullah, N., Donner, A., Ippolito, J.A., Williams, T., 1999. SELEX andmissing phosphate contact analyses reveal flexibility within the AP-2[alpha] protein: DNA binding complex. Nucleic Acids Res. 27, 2760–2769.

Moser, M., Imhof, A., Pscherer, A., Bauer, R., Amselgruber, W., Sinowatz,F., Hofstadter, F., Schule, R., Buettner, R., 1995. Cloning and charac-terization of a second AP-2 transcription factor: AP-2�. Development121, 2779–2788.

Moser, M., Pscherer, A., Roth, C., Becker, J., Mucher, G., Zerres, K.,Dixkens, C., Weis, J., Guay-Woodford, L., Buettner, R., Fassler, R.,1997a. Enhanced apoptotic cell death of renal epithelial cells in micelacking transcription factor AP-2�. Genes Dev. 11, 1938–1948.

Moser, M., Ruschoff, J., Buettner, R., 1997b. Comparative analysis ofAP-2 alpha and AP-2 beta gene expression during mouse embryogen-esis. Dev. Dyn. 208, 115–124.

Nottoli, T., Hagopian-Donaldson, S., Zhang, J., Perkins, A., Williams, T.,1998. AP-2-null cells disrupt morphogenesis of the eye, face, and limbsin chimeric mice. Proc. Natl. Acad. Sci. USA 95, 13714–13719.

Pierce Jr., D.F., Johnson, M.D., Matsui, Y., Robinson, S.D., Gold, L.I.,Purchio, A.F., Daniel, C.W., Hogan, B.L., Moses, H.L., 1993. Inhibi-tion of mammary duct development but not alveolar outgrowth duringpregnancy in transgenic mice expressing active TGF-beta 1. GenesDev. 7, 2308–2317.

Prager, D., Rosenblatt, J.D., Ejima, E., 1994. Hypercalcemia, parathyroidhormone-related protein expression and human T-cell leukemia virusinfection. Leuk. Lymphoma 14, 395–400.

Robinson, G.W., Hennighausen, L., 1997. Inhibins and activins regulatemammary epithelial cell differentiation through mesenchymal–epithe-lial interactions. Development 124, 2701–2708.

Robinson, G.W., Hennighausen, L., Johnson, P.F., 2000. Side-branching inthe mammary gland: the progesterone-Wnt connection [In ProcessCitation]. Genes Dev. 14, 889–894.

Robinson, G.W., McKnight, R.A., Smith, G.H., Hennighausen, L., 1995.Mammary epithelial cells undergo secretory differentiation in cyclingvirgins but require pregnancy for the establishment of terminal differ-entiation. Dev. Suppl. 121, 2079–2090.

Rugh, R., 1968. The Mouse: Its Reproduction and Development. BurgesPublishing, Minneapolis, MN.

144 J. Zhang et al. / Developmental Biology 256 (2003) 127–145

Page 19: Overexpression of transcription factor AP-2 suppresses ...analyzed transgenic mice in which the AP-2 gene is over-expressed in the postnatal mammary gland. We have com-bined these

Schorle, H., Meier, P., Buchert, M., Jaenisch, R., Mitchell, P.J., 1996.Transcription factor AP-2 essential for cranial closure and craniofacialdevelopment. Nature 381, 235–238.

Seagroves, T.N., Lydon, J.P., Hovey, R.C., Vonderhaar, B.K., Rosen, J.M.,2000. C/EBPbeta (CCAAT/enhancer binding protein) controls cell fatedetermination during mammary gland development. Mol. Endocrinol.14, 359–368.

Silberstein, G.B., Van Horn, K., Shyamala, G., Daniel, C.W., 1996. Pro-gesterone receptors in the mouse mammary duct: distribution anddevelopmental regulation. Cell Growth Differ. 7, 945–952.

Somasundaram, K., Jayaraman, G., Williams, T., Moran, E., Frisch, S.,Thimmapaya, B., 1996. Repression of a matrix metalloprotease gene byE1A correlates with its ability to bind to cell type-specific transcriptionfactor AP-2. Proc. Natl. Acad. Sci. USA 93, 3088–3093.

Teglund, S., McKay, C., Schuetz, E., van Deursen, J.M., Stravopodis, D.,Wang, D., Brown, M., Bodner, S., Grosveld, G., Ihle, J.N., 1998. Stat5aand Stat5b proteins have essential and nonessential, or redundant, rolesin cytokine responses. Cell 93, 841–850.

Toscani, A., Mettus, R.V., Coupland, R., Simpkins, H., Litvin, J., Orth, J.,Hatton, K.S., Reddy, E.P., 1997. Arrest of spermatogenesis and defec-tive breast development in mice lacking A-myb. Nature 386, 713–717.

Turner, B.C., Zhang, J., Gumbs, A.A., Maher, M.G., Kaplan, L., Carter, D.,Glazer, P.M., Hurst, H.C., Haffty, B.G., Williams, T., 1998. Expressionof AP-2 transcription factors in human breast cancer correlates with theregulation of multiple growth factor signalling pathways. Cancer Res.58, 5466–5472.

Werling, U., Schorle, H., 2002. Transcription factor gene AP-2 gammaessential for early murine development. Mol. Cell. Biol. 22, 3149–3156.

West-Mays, J.A., Zhang, J., Nottoli, T., Hagopian-Donaldson, S., Libby,D., Strissel, K.J., Williams, T., 1999. AP-2alpha transcription factor is

required for early morphogenesis of the lens vesicle. Dev. Biol. 206,46–62.

Williams, T., Admon, A., Luscher, B., Tjian, R., 1988. Cloning andexpression of AP-2, a cell-type-specific transcription factor that acti-vates inducible enhancer elements. Genes Dev. 2, 1557–1569.

Williams, T., Tjian, R., 1991a. Analysis of the DNA binding and activationproperties of the human transcription factor AP-2. Genes Dev. 5,670–682.

Williams, T., Tjian, R., 1991b. A novel dimerization motif in transcriptionfactor AP-2 can be utilized by other DNA binding proteins. Science251, 1067–1071.

Wysolmerski, J.J., McCaughern-Carucci, J.F., Daifotis, A.G., Broadus,A.E., Philbrick, W.M., 1995. Overexpression of parathyroid hormone-related protein or parathyroid hormone in transgenic mice impairsbranching morphogenesis during mammary gland development. Devel-opment 121, 3539–3547.

Zeng, Y.-X., Somasundaram, K., El-Deiry, W.F., 1997. AP2 inhibits can-cer cell growth and activates p21waf1/cip1 expression. Nat. Genet. 15,78–82.

Zhang, J., Hagopian-Donaldson, S., Serbedzija, G., Elsemore, J., Plehn-Dujowich, D., McMahon, A.P., Flavell, R.A., Williams, T., 1996.Neural tube, skeletal and body wall defects in mice lacking transcrip-tion factor AP-2. Nature 381, 238–241.

Zhao, F., Satoda, M., Licht, J.D., Hayashizaki, Y., Gelb, B.D., 2001.Cloning and characterization of a novel mouse AP-2 transcriptionfactor, AP-2�, with unique DNA binding and transactivation properties.J. Biol. Chem. 276, 40755–40760.

Zutter, M.M., Santoro, S.A., Painter, A.S., Tsung, Y.L., Gafford, A., 1994.The human alpha 2 integrin gene promoter. Identification of positiveand negative regulatory elements important for cell-type and develop-mentally restricted gene expression. J. Biol. Chem. 269, 463–469.

145J. Zhang et al. / Developmental Biology 256 (2003) 127–145