14
Genomes & Developmental Control Preferential reduction of β cells derived from Pax6MafB pathway in MafB deficient mice Wataru Nishimura a,b , Sheldon Rowan b,c , Therese Salameh a , Richard L. Maas b,c , Susan Bonner-Weir a,b , Susan M. Sell d , Arun Sharma a,b, a Section of Islet Transplantation and Cell Biology, Joslin Diabetes Center, USA b Department of Medicine, Harvard Medical School, Boston, MA 02215, USA c Division of Genetics, Department of Medicine, Brigham and Womens Hospital, Boston, MA 02215, USA d Bioinformatics Research Center and Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223, USA Received for publication 30 August 2007; revised 6 December 2007; accepted 10 December 2007 Available online 23 December 2007 Abstract During pancreatic development insulin + cells co-express the transcription factors MafB and Pax6, and transition from a MafA to MafA + state. To examine the role of Pax6 and MafB in the development of β-cells, we analyzed embryonic pancreata from Pax6- and MafB-deficient mice. Pax6 deficiency, as manifest in the Pax6 SeyNeu allele, reduced not only the number of cells expressing insulin or glucagon, but also the number of MafB, PDX-1 and MafA expressing cells. We show that MafB can directly activate expression of insulin and glucagon, and a MafB protein engineered to contain N248S mutation in the MafB (kr ENU ) results in significantly reduced activation. Furthermore, pancreata from MafB deficient (kr ENU /kr ENU ) mice exhibited reduced number of cells expressing insulin, glucagon, PDX-1 and MafA, with only a minor reduction in MafB expressing cells. MafB deficiency does not affect endocrine specification but does affect the lineage commitment of the endocrine cells and their maturation. Similar to Pax6 deficient mice, MafB deficient mice showed reductions both in insulin and glucagon expressing cells and in the ability of MafB and PDX-1 expressing cells to activate expression of these hormones. However, MafB deficient mice exhibited no effect on Pax6 expression. These results suggest that MafB may function as a downstream mediator of Pax6 in regulating the specification of insulin and glucagon expressing cells. Interestingly, the remaining insulin + cells in these knockouts preferentially express Hb9, suggesting the existence of an alternate pathway for the generation of insulin expressing cells, even in the absence of Pax6 and MafB function. Thus, Pax6 acts upstream of MafB, which in turn may trigger the expression of insulin and regulate the PDX-1 and MafA expression required for β-cell maturation. © 2007 Elsevier Inc. All rights reserved. Keywords: MafB; MafA; Pax6; Sey; Kreisler; Insulin gene transcription factor; Pancreatic development; Endocrine differentiation; Pancreatic islets Introduction To develop reliable sources of glucose-responsive β-cells for the treatment of diabetes, it is essential to understand how β- cells form during development. The pancreas develops as a dorsal and ventral evagination from the developing foregut that expresses the homeodomain transcription factor PDX-1 (Edlund, 2002; Leonard et al., 1993; Miller et al., 1994; Ohlsson et al., 1993). PDX-1 is essential for the normal proliferation and differentiation of embryonic pancreatic precursors (Edlund, 2002; Jonsson et al., 1994; Offield et al., 1996). Another transcription factor, Ngn3, regulates the differentiation of endocrine cells (Gradwohl et al., 2000). In addition, several other transcription factors play critical roles in both pancreatic development and endocrine differentiation (Collombat et al., 2006; Grapin-Botton and Melton, 2000; Jensen, 2004; Kim and MacDonald, 2002; Murtaugh, 2007). For example, transcription factors Arx and Pax4 play critical roles in regulating specifica- tion of endocrine progenitors towards α- or β- and δ-cell fates (Collombat et al., 2005, 2006, 2007). Pax6 is required for maintaining these hormone-expressing cells and for regulating specification of ghrelin-expressing ε-cells (Ashery-Padan et al., 2004; Heller et al., 2005; Wang et al., 2004). In addition, Available online at www.sciencedirect.com Developmental Biology 314 (2008) 443 456 www.elsevier.com/developmentalbiology Corresponding author. Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA. Fax: +1 617 732 2589. E-mail address: [email protected] (A. Sharma). 0012-1606/$ - see front matter © 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.ydbio.2007.12.009

Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Embed Size (px)

Citation preview

Page 1: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Available online at www.sciencedirect.com

Developmental Biology 314 (2008) 443–456www.elsevier.com/developmentalbiology

Genomes & Developmental Control

Preferential reduction of β cells derived from Pax6–MafB pathway inMafB deficient mice

Wataru Nishimura a,b, Sheldon Rowan b,c, Therese Salameh a, Richard L. Maas b,c,Susan Bonner-Weir a,b, Susan M. Sell d, Arun Sharma a,b,⁎

a Section of Islet Transplantation and Cell Biology, Joslin Diabetes Center, USAb Department of Medicine, Harvard Medical School, Boston, MA 02215, USA

c Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USAd Bioinformatics Research Center and Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223, USA

Received for publication 30 August 2007; revised 6 December 2007; accepted 10 December 2007Available online 23 December 2007

Abstract

During pancreatic development insulin+ cells co-express the transcription factors MafB and Pax6, and transition from a MafA− to MafA+ state.To examine the role of Pax6 and MafB in the development of β-cells, we analyzed embryonic pancreata from Pax6- and MafB-deficient mice.Pax6 deficiency, as manifest in the Pax6Sey–Neu allele, reduced not only the number of cells expressing insulin or glucagon, but also the number ofMafB, PDX-1 and MafA expressing cells. We show that MafB can directly activate expression of insulin and glucagon, and a MafB proteinengineered to contain N248S mutation in the MafB (krENU) results in significantly reduced activation. Furthermore, pancreata from MafB deficient(krENU/krENU) mice exhibited reduced number of cells expressing insulin, glucagon, PDX-1 and MafA, with only a minor reduction in MafBexpressing cells. MafB deficiency does not affect endocrine specification but does affect the lineage commitment of the endocrine cells and theirmaturation. Similar to Pax6 deficient mice, MafB deficient mice showed reductions both in insulin and glucagon expressing cells and in the abilityof MafB and PDX-1 expressing cells to activate expression of these hormones. However, MafB deficient mice exhibited no effect on Pax6expression. These results suggest that MafB may function as a downstream mediator of Pax6 in regulating the specification of insulin andglucagon expressing cells. Interestingly, the remaining insulin+ cells in these knockouts preferentially express Hb9, suggesting the existence of analternate pathway for the generation of insulin expressing cells, even in the absence of Pax6 and MafB function. Thus, Pax6 acts upstream ofMafB, which in turn may trigger the expression of insulin and regulate the PDX-1 and MafA expression required for β-cell maturation.© 2007 Elsevier Inc. All rights reserved.

Keywords: MafB; MafA; Pax6; Sey; Kreisler; Insulin gene transcription factor; Pancreatic development; Endocrine differentiation; Pancreatic islets

Introduction

To develop reliable sources of glucose-responsive β-cells forthe treatment of diabetes, it is essential to understand how β-cells form during development. The pancreas develops as adorsal and ventral evagination from the developing foregut thatexpresses the homeodomain transcription factor PDX-1(Edlund, 2002; Leonard et al., 1993; Miller et al., 1994; Ohlssonet al., 1993). PDX-1 is essential for the normal proliferation and

⁎ Corresponding author. Research Division, Joslin Diabetes Center, OneJoslin Place, Boston, MA 02215, USA. Fax: +1 617 732 2589.

E-mail address: [email protected] (A. Sharma).

0012-1606/$ - see front matter © 2007 Elsevier Inc. All rights reserved.doi:10.1016/j.ydbio.2007.12.009

differentiation of embryonic pancreatic precursors (Edlund,2002; Jonsson et al., 1994; Offield et al., 1996). Anothertranscription factor, Ngn3, regulates the differentiation ofendocrine cells (Gradwohl et al., 2000). In addition, severalother transcription factors play critical roles in both pancreaticdevelopment and endocrine differentiation (Collombat et al.,2006; Grapin-Botton and Melton, 2000; Jensen, 2004; Kim andMacDonald, 2002; Murtaugh, 2007). For example, transcriptionfactors Arx and Pax4 play critical roles in regulating specifica-tion of endocrine progenitors towards α- or β- and δ-cell fates(Collombat et al., 2005, 2006, 2007). Pax6 is required formaintaining these hormone-expressing cells and for regulatingspecification of ghrelin-expressing ε-cells (Ashery-Padan et al.,2004; Heller et al., 2005; Wang et al., 2004). In addition,

Page 2: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

444 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

members of the Nkx and Maf families regulate the differentia-tion of hormone positive cells.

The identification of MafA as a β-cell specific basic leucinezipper (bZIP) transcription factor has initiated the systematiccharacterization of Maf factors in pancreatic development andin β-cell function (Olbrot et al., 2002; Kataoka et al., 2002;Matsuoka et al., 2003). MafA (formerly known as RIPE3b1) is aglucose-responsive insulin gene transcription factor that bindsto the insulin Maf Response Element (MARE) and activatesinsulin gene expression (Sharma and Stein, 1994; Olbrot et al.,2002; Kataoka et al., 2002; Matsuoka et al., 2003). Another Maffactor, MafB [also known as Kreisler (Krml1/MafB)], isexpressed in pancreatic endocrine cells and regulates differ-entiation of several cell types (Eichmann et al., 1997;Manzanares et al., 1997; Moriguchi et al., 2006; Sieweke etal., 1996). The gene was first identified as the locus mutated inthe Kreisler (kr) mouse, and a second ethylnitrosourea (ENU)induced mutation [krENU (also known asMafBENU)] in this genehas been described (Cordes and Barsh, 1994). MafB, expressedonly in α-cells in adults, is expressed in the insulin positive cellsduring pancreatic development (Artner et al., 2006; Nishimuraet al., 2006). We proposed that endocrine progenitors aftercommitting to the insulin positive lineage continue a furtherdifferentiation/maturation process. Early insulin+ cells expressMafB, and following the induction of PDX-1, mature intoinsulin+ and MafA+ cells (Nishimura et al., 2006). MafAdeficient Mice have normal pancreatic islets at birth, but theratio of β to α cells gradually reduce after birth, resulting inglucose intolerance by 8–12 weeks (Zhang et al., 2005). Thisphenotype suggests a critical role of MafA in the maturationstep required for the function and survival of β-cells. Theimportance of this maturation step is highlighted by a recentpublication on the differentiation of human ES cells intoinsulin+ cells (D'Amour et al., 2006). Although these insulin+

cells expressed MafB, they did not maintain the expression ofPDX-1 and did not secrete insulin in response to glucose(D'Amour et al., 2006). We hypothesize that these cells may nothave switched from MafB+ MafA− to MafB− MafA+ state. It islikely that the lack of MafA expression prevents early insulin+

cells from expressing the downstream targets of MafA that arerequired for the insulin synthesis and secretion (Kato et al.,2006; Wang et al., 2007). Thus, elucidation of the mechanismsthat regulate the conversion of insulin+ cells into maturefunctional β-cells should facilitate our ability to generateglucose-responsive, insulin-producing cells for cell-baseddiabetes therapy.

In the present study, we characterized the roles of Pax6 andMafB in the differentiation of insulin expressing cells. Loss ofeither Pax6 or MafB function results in reduced numbers ofcells that express insulin, glucagon, PDX-1 or MafA, anddecreases the ability of MafB+ and PDX-1+ cells to expressinsulin. Our results suggest that MafB may function as adownstream mediator of Pax6 in regulating the formation ofinsulin and glucagon positive cells. We present data supportingthe initiation of insulin expression in the endocrine progenitorsby at least two pathways: the formation of insulin-expressingcells from one pathway requires the function of Pax6 and

MafB, while specification of the remaining insulin-expressingcells may occurs via a Pax4–Hb9 dependent pathway. Theseresults suggest that MafB can regulate the initiation of insulinexpression in some, but not all, endocrine progenitors destinedto express insulin.

Materials and methods

Construction of expression vectors

Various expression vectors were constructed by conventional molecularbiology techniques. Insulin promoter luciferase reporter constructs −238 WTLUC and its derivative mutant 121–22m LUC have been described previously(Harrington and Sharma, 2001). The glucagon promoter luciferase reporter GLULUC was kindly provided by Dr. Dan Drucker (Toronto, Canada); the PDX-1promoter luciferase reporter (Eto et al., 2007), by Dr. Melissa Thomas(Massachusetts General Hospital, Boston, MA). The expression vectors pcDNAMafA, pCMVSport6 MafB and pcDNA cMaf have been described previously(Nishimura et al., 2006). pCMVSport6 krENU expression vector was generatedby PCR amplification of homozygous krENU mouse genomic DNA witholigonucleotide primers (5′-GACCCGCCAGGACTCACAGAAA-3′and 5′-CCGCGCAACAGCTACCCACTA-3′) for 35 cycles of 30 sec at 94.0C,40 sec at 62.3C and 1 min at 72C. PCR product was digested with AflII andPvuII and was used to replace the corresponding fragment frompCMVSport6 MafB to give full-length krENU cDNA containing a pointmutation at base pair 743 in the DNA binding region of MafB. Constructs wereconfirmed by sequencing.

Luciferase assays

HeLa cells were transfected with 1 μg of reporter constructs of −238 WTLUC, 121–22 m LUC, GLU LUC or PDX-1 promoter, 1 μg of pSVβ-galplasmid (Promega, Madison, WI) as an internal control and 1 μg of indicatedexpression vector. Whole cell extracts were prepared and luciferase activity wasmeasured as previously described (Nishimura et al., 2005).

Animals

Mice with mutation in kreisler/MafB generated by ethylnitrosourea (ENU)induced chemical mutagenesis have been reported earlier (Cordes and Barsh,1994). Mutagenesis results in a point mutation in the DNA-binding region ofMafB that changes amino acid 248 from asparagine to serine (krENU orMafBENU)leading to compromised DNA binding affinity of krENU to Maf ResponseElement (MARE)(Sadl et al., 2002). Heterozygous mice with the ENU-inducedkrENU allele were received from Dr. Greg Barsh (Stanford, CA), were maintainedon C57Bl/6J background for more than 10 generation and used in this study.Small eye mutant (Pax6Sey–Neu) mice (Glaser et al., 1994; Hill et al., 1991; Sanderet al., 1997), also generated by chemical mutagenesis, have a point mutation in asplice donor site of 3′ end of the homeobox that results in incorrect splicing andtruncated Pax6 protein lacking the carboxy-terminal 115 amino acidscorresponding to the transcriptional activation domain. The day of vaginal-plug discovery was designated as embryonic day E 0.5.

Immunohistochemistry and quantification

Immunohistochemical and immunofluorescent analyses were performed asdescribed previously (Nishimura et al., 2006). The primary antibodies usedwere: guinea pig anti-insulin (Linco, Billerica, MA); guinea pig anti-glucagon(Linco); rabbit anti-somatostatin (Santa Cruz Biotechnology, CA); rabbit anti-pancreatic polypeptide (Linco); rabbit anti-Ghrelin (Phoenix Pharmaceuticals,Belmont, CA); rabbit anti-Hb9 (provided by Dr. S. Pfaff); rabbit anti-MafB(Bethyl Laboratories, Montgomery, TX); mouse anti-Nkx2.2 (DevelopmentalStudies Hybridoma Bank, Iowa City, IA); rabbit anti-Nkx6.1 (provided by Dr. P.Serup); mouse anti-Neurogenin3 (provided by Drs. Serup and Madsen, BetaCell Biology Consortium); goat anti-PDX-1 (provided by Dr. C. Wright); rabbitanti-Pax6 (Covance, Princeton, NJ). Rabbit anti-MafA antibody was described

Page 3: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

445W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

previously (Nishimura et al., 2006). For amplification, biotinylated anti-rabbit,anti-mouse or anti-goat antibodies (Jackson ImmunoResearch, West Grove, PA)were used at a 1:400 dilution followed by streptavidin-conjugated Texas red(1:400) or streptavidin-conjugated Alexa fluor 488 (1:400) (Molecular Probes,Eugene, OR). The secondary antibodies were: FITC- or Texas red-conjugatedanti-rabbit, anti-mouse or anti-guinea pig (Jackson ImmunoResearch). Nuclearcounterstaining was performed by DAPI mounting medium (Vector, Burlin-game, CA). For the double staining using rabbit anti-MafB and rabbit anti-Ghrelin antibodies, pancreatic sections were first stained with rabbit anti-MafBantibody using secondary biotinylated anti-rabbit antibody followed bystreptavidin-conjugated Alexa fluor 488, and images of areas expressingMafB were taken. The same imaged sections were next stained with rabbit anti-Ghrelin antibody and Texas red conjugated anti-rabbit secondary antibody andimages of the same areas were taken again to examine co-expression of MafBand Ghrelin. The results of immunohistochemistry and quantification werederived from several replicates of at least three embryonic pancreases for eachgenotype. Every 10th section of embryonic pancreas was analyzed forimmunofluorescent hormone-positive areas as a proportion of epithelial area.For total number of transcription factor+ cells (for Fig. 2A), every 10th sectionwas stained with DAB and counted. For number of transcription factor+ insulin+

cells per total number of indicated transcription factor+ cells or total number ofinsulin+ cells, cells were counted from at least three different pancreatic blocks.Immunofluorescence data were collected as confocal images on Zeiss LSM410(Zeiss, Thornwood, NY); all DAB-stained images were analyzed on anOlympus BH2 bright field microscope. Images were quantitated using NIHImage J software.

Western blot analysis

Cell lysates used in luciferase assays were subjected to 10% SDS-PAGE andgels were transferred to polyvinyldenedifluoride membrane filters (BioRadLaboratories, Hercules, CA). Western bolt analyses were performed using MafBand β-actin antibody (Santa Cruz Biotechnology, CA) and detected withhorseradish peroxidase using ECL reagents (Amersham).

Results

Pax6 deficiency results in reduced numbers of MafA, MafB andPDX-1 expressing cells

Previously, we showed that Pax6+ insulin+ cells transitionfrom a MafB+ MafA− stage to a mature MafA+ MafB−

phenotype (Nishimura et al., 2006). An association betweenPax6 and Maf factors has been reported in different systems(Reza et al., 2002; Reza and Yasuda, 2004). Pax6 deficiencyresults in the inability of endocrine cells to express markers ofterminal differentiation and maturation (Ashery-Padan et al.,2004). Hence, we examined the role of Pax6 in regulating theexpression and function of Maf factors during pancreaticdevelopment. E15.5 Pax6Sey–Neu/Sey–Neu (subsequently referredas SeyNeu/SeyNeu) pancreata completely lack Pax6 expressingcells (antibody recognizes C-terminal domain of Pax6), whilewild type littermates have significant numbers of Pax6+ cells,some of which co-expressed insulin (Supplementary Fig. 1). Asreported earlier (Sander et al., 1997; Wang et al., 2004), loss ofPax6 results in a significant reduction in insulin and glucagon-expressing cells but does not affect the pancreatic expression ofNkx transcription factors at E15.5. Nkx6.1 expression in thepancreas marks the area of endocrine differentiation and, asshown in Supplementary Fig. 1, was unaffected by loss of Pax6function. Similarly, expression of the pro-endocrine gene Ngn3was not affected in Pax6 deficient mice (Supplementary Fig. 1),

consistent with the earlier observation (Ashery-Padan et al.,2004; Wang et al., 2004) that Pax6 is not required for theinitiation of endocrine differentiation.

We next examined whether the loss of Pax6 affected theexpression of Maf factors. The number of cells in Pax6-deficient (SeyNeu/SeyNeu) E15.5 pancreas expressing MafB,PDX-1 (induced PDX-1 expression during secondary transi-tion or PDX-1high) and MafA (Figs. 1B–D, F–H) werereduced. Some cells still expressed these transcription factorsdemonstrating the presence of a Pax6 independent mechanismfor their expression. The number of insulin+ cells expressingthese three transcription factors was significantly reduced inSeyNeu/SeyNeu mice (Figs. 1B–D, F–H). Thus, the loss ofPax6 function not only reduces the expression of terminalhormone markers but also reduces the expression of thetranscription factors implicated in the terminal differentiationprocess.

We next examined whether expression of MafB, PDX-1high

and MafA were maintained in insulin+ cells in the absence ofPax6 function (Figs. 1J–L, N–P). As reported previously(Sander et al., 1997), at E17.5 the number of insulin+ cellsremained low in the SeyNeu/SeyNeu embryos. Additionally,MafA and PDX-1 expression was restricted to insulin+ cells,and the number of cells expressing these transcription factorswas reduced (Figs. 1K, L, O, P); most Nkx6.1+ cells expressedinsulin, with only occasional Nkx6.1+ insulin− cells (Figs. 1I,M). We also observed a greater reduction in insulin+ and MafB+

cells in the SeyNeu/SeyNeu embryos at E17.5 (Fig. 1N) than atE15.5 (Fig. 1F).

Pax6-deficiency reduces the proportion of MafB+ andPDX-1high cells expressing insulin

Our observation that MafB+ pancreatic cells are present inthe SeyNeu/SeyNeu embryos (Figs. 1F, N) is consistent with theconclusion of Artner and colleagues (Artner et al., 2006) thatPax6 and Pax4 are not essential for MafB expression. Todetermine whether Pax6 deficiency reduces only the number ofcells expressing MafB, PDX-1 and MafA or has additionaleffects on their function, we quantified stained E15.5 pancreaticsections (Fig. 2). The number of MafB+ cells was significantlyreduced in SeyNeu/SeyNeu pancreas (Figs. 1F and 2A). While thenumber of Nkx6.1+ cells did not change, those of insulin+,MafB+ and MafA+ cells were reduced by about 75% (Fig. 2A),clearly demonstrating that loss of Pax6 function significantlyaffects MafB+ and MafA+ cells.

Since Pax6 is not required for the specification of endocrinecells but it regulates their ability to acquire final differentiationmarkers (Supplementary Fig. 1, Ashery-Padan et al., 2004;Heller et al., 2005; Wang et al., 2004), we analyzed in E15.5pancreas whether the loss of Pax6 expression affected 1) theability of MafB+, PDX-1high and MafA+ cells to express insulinand 2) the ability of remaining insulin+ cells to express normallevels of these factors. MafA was expressed only in insulin+

cells, while several MafB+ and PDX-1high cells did not expressinsulin. Similarly, several insulin+ cells did not express thesetranscription factors (Fig. 1). Quantification was performed to

Page 4: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 1. SeyNeu/SeyNeu embryos have reduced expression MafB, PDX-1 and MafA. Adjacent sections from E15.5 wild type (A–D) and SeyNeu/SeyNeu (E–H) embryonicpancreata (n=6 each) were stained to detect the expression of Nkx6.1, MafB, PDX-1 and MafA in green and insulin in red. In wild type pancreas adjacent sectionsshow cells expressing Nkx6.1, MafB, PDX-1 andMafA, but in SeyNeu/SeyNeu embryos MafB, PDX-1 andMafA expression was significantly reduced. E17.5 pancreatafrom wild type (I–L) and SeyNeu/SeyNeu (M–P) embryos (n=3 each) were stained for Nkx6.1, MafB, PDX-1 and MafA in green and insulin in red. At this stage loss ofPax6 function results in significant reduction in the number of insulin+ cells, and the majority of Nkx6.1+, MafB+, PDX-1high and MafA+ cells are restricted to theseinsulin+ cells. Bars: 20 μm.

446 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

determine the proportion of transcription factor+ cells expres-sing insulin (Fig. 2B) and the proportion of insulin+ cellsexpressing different transcription factors (Fig. 2C), as depictedin the Venn diagrams. Nearly 65–70% of MafB+ cells(390 MafB+, 273 Ins+ and 259 MafB+Ins+ cells; 259 out of390), 75–80% of PDX-1high cells (346 PDX-1high, 324 Ins+ and276 PDX1highIns+ cells) and all MafA+ cells (179 MafA+, 268Ins+ and 179 MafA+Ins+ cells) expressed insulin in wild typepancreas whereas loss of Pax6 function significantly reducedthe ability of MafB+ (∼35%)(125 MafB+, 58 Ins+ and49 MafB+Ins+ cells; 49 out of 125) and PDX-1high (∼45%)(82 PDX-1high, 58 Ins+ and 49 PDX1highIns+ cells) cells to co-express insulin. Importantly, while the number of insulin+ cellswas reduced in SeyNeu/SeyNeu pancreas, there was no change in

the proportion of MafA+ cells expressing insulin (100%)(50 MafA+, 81 Ins+ and 50 MafA+Ins+ cells), which suggeststhat MafA is expressed only after insulin+ cells have reached alatter maturation stage. Next, we quantified the proportion ofinsulin+ cells expressing these transcription factors (Fig. 2C).Unlike the reduction in the proportion of MafB+ and PDX-1high

cells expressing insulin in the absence of Pax6, the proportionof insulin+ cells expressing these different transcription factorsin wild type and SeyNeu/SeyNeu mice remained unchanged.Nearly 95%, 75% and 65% of insulin+ cells were MafB+, PDX-1high and MafA+, respectively, in both wild type and Pax6deficient mice (Fig. 2C). This observation suggests that theremaining insulin+ cells derived from a Pax6 independentpathway appear to undergo normal maturation.

Page 5: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 2. Pax6-deficiency reduces the proportion of MafB+ and PDX-1+ cells that express insulin. Quantification of immunostained E15.5 embryonic pancreatic sectionsfrom wild type and SeyNeu/SeyNeu embryos (n=6 each). (A) Total number of Nkx6.1+, MafB+ andMafA+ cells in wild type and SeyNeu/SeyNeu embryonic pancreas weredetermined along with the number of insulin+ cells in the same sections. In SeyNeu/SeyNeu embryonic pancreas, the numbers of MafB+, MafA+ and insulin+ cells weresignificantly reduced (p=0.03, 0.01 and 0.01 respectively), while the number of Nkx6.1+ cells remained unchanged (p=0.46). (B) The ability of transcription factorexpressing cells to express insulin was determined by quantifying the proportion of transcription factor+ insulin+ cells per total number of transcription factor+ cells, asrepresented by cells in green circle in adjacent Venn diagram. Proportions of MafB+Ins+ cells/total MafB+ cells and PDX-1+Ins+ cells/total PDX-1+ cells in SeyNeu/SeyNeu pancreas were significantly less than the wild-type (p=0.005 and 0.01 respectively), while all of the MafA+ cells express insulin. (C) To determine the ability ofinsulin+ cells to undergo further maturation in the absence of Pax6 function, the proportion of insulin+ cells expressing these transcription factors was quantified, asrepresented by cells in red circle in adjacent Venn diagram. The proportion of MafB+Ins+, PDX-1+Ins+ and MafA+Ins+ cells to the total Ins+ cells was not significantlydifferent between wild type and SeyNeu/SeyNeu mice.

447W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

Pax6 deficiency reduces the proportion of MafB+ cellsexpressing glucagon

Since Pax6 deficiency reduced the number of MafB+ cellsthat co-express insulin, we also used SeyNeu/SeyNeu mice toexamine whether the MafB+ cells that did not express insulin

instead expressed glucagon. At E12.5, Nkx6.1 and PDX-1, twotranscription factors that mark the developing pancreaticepithelium at this stage, are not expressed in the majority ofglucagon+ cells in either wild type or SeyNeu/SeyNeu pancreas(Figs. 3A, B, E, F). It is important to note that at E12.5, PDX-1expression was not affected by Pax6 loss of function. Thus, the

Page 6: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 3. Pax6-deficiency reduces the proportion of MafB+ cells expressing glucagon. Adjacent sections from E12.5 wild type (A–C) and SeyNeu/SeyNeu (E–G) (n=5wild type, 4 mutant) pancreata were stained to detect Nkx6.1, PDX-1 and MafB expression in green and glucagon in red. In addition, sections from E15.5 embryonicpancreata (D, H) were stained for MafB in green and glucagon in red. In E12.5 SeyNeu/SeyNeu pancreata Nkx6.1 and PDX-1 expression was normal, while theexpression of MafB and glucagon expression was reduced at both E12.5 and E15.5. Arrow denotes MafB+ glucagon+ cells in SeyNeu/SeyNeu pancreas at E12.5. Bars:20 μm. (I) Proportion of MafB+ glucagon+ cells to total number of MafB+ or glucagon+ cells were quantified from wild type and SeyNeu/SeyNeu pancreatic sections fromE12.5 and E15.5. Absence of Pax6 function reduces the proportion of glucagon+ cells that express MafB at both E12.5 and E15.5, while the proportion of MafB+ cellsexpressing glucagon is reduced only in E15.5 pancreata.

448 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

reduced PDX-1 in older SeyNeu/SeyNeu embryos (Figs. 1 and 2)may reflect the ability of Pax6 to selectively regulate theinduction of PDX-1 expression (PDX-1high) in insulin+ cellsduring secondary transition.

The quantification of glucagon+ and MafB+ cells at E12.5and 15.5 provides important insights into the roles of Pax6 andMafB in the induction of hormone expression and in thedifferentiation of early endocrine cells. At E12.5 in wild typepancreas, the majority of MafB+ cells co-express glucagon, andconversely the majority of glucagon+ cells are MafB+

(260 MafB+, 251 Glu+ and 239 MafB+ Glu+ cells). Thenumber of MafB+ cells at E12.5 in SeyNeu/SeyNeu pancreata wasextremely low, but all MafB+ cells co-expressed glucagon(7 MafB+, 37 Glu+ and 7 MafB+ Glu+ cells), and a number ofglucagon+ cells did not express MafB (Fig. 3G). At E15.5, withthe induction of insulin expression, nearly 40% MafB+ cells

expressed glucagon in wild type (500 MafB+, 204 Glu+ and198 MafB+ Glu+ cells), and this number decreased to less than10% in SeyNeu/SeyNeu pancreata (216 MafB+, 48 Glu+ and15 MafB+ Glu+ cells) (Fig. 3I). Nearly 65% of MafB+ cells atE15.5 express insulin in wild type pancreas (Fig. 2B),suggesting that most of the MafB+ cells at E15.5 in wild typeeither express insulin or glucagon. Together, these results (Figs.2 and 3) suggest increased numbers of MafB+ cells that do notexpress either insulin or glucagon in SeyNeu/SeyNeu pancreas atE15.5. Deficiency of Pax6 (Heller et al., 2005) and Nkx2.2(possibly via reducing Pax6 expression) (Prado et al., 2004)changes cell-fate of endocrine precursors to ghrelin expressingε-cells at the expense of the other endocrine cell types. Weobserved a similar increase in ghrelin only expressing ε-cells inPax6 deficient mice, but the ghrelin-expressing ε-cells did notexpress MafB (Supplementary Fig. 2).

Page 7: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 4. The krENU allele encodes a reduced ability to activate insulin and glucagon gene expression. (A) Schematics of wild type MafB and its krENU mutant anddeletion derivatives showing activation and DNA binding domains. (B, C) Western blot analyses to detect MafB protein expression from various MafB constructs.Lysates used in transient transfection assays in panels D and E were subjected to Western blot to detect expression of MafB (B) and β-actin (C). (D) Insulin promoter:Luciferase reporter constructs, wild type (−238 WT LUC, red) and insulin promoter with mutation in −121–122 bp (−121–122 m LUC, pink), and (E) glucagonpromoter: Luciferase reporter constructs (GLU LUC, green) were transfected into HeLa cells with the indicated expression plasmids and pSVβ-gal as an internalcontrol. Luciferase and β-gal activities were determined. Results are presented relative to activity of wild-type luciferase construct±S.E. (n=4).

449W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

MafB can activate insulin and glucagon gene expression

To determine whether MafB itself plays an important role inthe differentiation of endocrine cells, we first examined the abilityof ENU induced N248S mutation in the MafB coding region

Fig. 5.MafB deficiency reduces insulin+ and glucagon+ cells. (A, B) Pancreas from Eembryos were stained for glucagon in green and insulin in red. Immunostained sectioareas per pancreatic epithelial area as described in Materials and methods. Bars: 20

(krENU) to activate the insulin and glucagon promoters. Twoplasmid constructs were made from wild-type MafB plasmid(pCMV Sport MafB), one carrying the ENU induced N248Smutation (krENU) and another lacking the C-terminal DNAbinding and bZIP domain (ΔCMafB) (Fig. 4A). All three

15.5 wild type (+/+) and MafB mutant (krENU/krENU) (n=4 wild type, 4 mutant)ns were used to quantify the proportion of insulin (C) and glucagon (D) positiveμm.

Page 8: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 6. MafB deficiency does not inhibit the initiation of endocrine differentiation. Sections from E15.5 pancreata from wild type and krENU/krENU embryos werestained for Ngn3, Nkx2.2 and Pax6 in green and insulin in red. Expression of insulin was reduced in krENU/krENU pancreata, but Ngn3, Nkx2.2 and Pax6 expressionwas similar to that in the wild type embryos. Bars: 20 μm.

450 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

plasmids transfected into HeLa cells expressed a proteinrecognized by anti-MafB antibody in Western blots (Fig. 4B).Quantification with normalization to β-actin (Fig. 4C) indicatedthat krENU is expressed at a level slightly higher than the wild-typeMafB protein. Co-transfection of these plasmids with insulin(Fig. 4D) or glucagon (Fig. 4E) promoter luciferase reporterconstructs demonstrated that wild type MafB activated bothinsulin and glucagon promoters, as previously reported (Nishi-mura et al., 2006; Zhao et al., 2005).ΔCMafB was ineffective ininducing expression of these genes, and krENU showed significantimpairment in its ability to activate insulin and glucagon geneexpression (Figs. 4D, E). Co-transfecting these expressionplasmids with the insulin reporter construct −122.121 m Luc(Harrington and Sharma, 2001) containing a mutated insulinMARE significantly reduced the MafB and krENU mediatedactivation of luciferase, suggesting that krENU retains someresidual activity to regulate insulin expression via its cognatebinding site. Thus, in principal, krENU mice should represent amodel of MafB deficiency, but not absence, of MafB function.

MafB deficiency results in reduction of insulin+ and glucagon+

cells

In adult mouse pancreas MafB expression is restricted to α-cells, but during embryonic development MafB expression is

Fig. 7. MafB deficiency reduces PDX-1, MafA and insulin expression and impairsections from E15.5 wild type and krENU/krENU (n=4 wild type, 3 mutant) pancreataIn krENU/krENU sections the number of cells expressing PDX-1, MafA and insulin wpancreatic sections from wild type and krENU/krENU embryos were quantified to deteproportion of insulin+ cells expressing different transcription factors (J). (I) Proporcells in krENU/krENU were significantly reduced, while all MafA+ cells expressed insnot significantly different between wild type and krENU/krENU. (K) MafB and otherluciferase reporter construct was transfected into HeLa cells with the indicated expresented relative to the activity of a wild-type luciferase construct transfected wit

also seen in insulin+ cells (Nishimura et al., 2006). Since ourresults indicate a correlation between reduced MafB expressionand the number of insulin+ and glucagon+ cells in SeyNeu/SeyNeu mice (Figs. 1–3), we hypothesized that MafB mightregulate the differentiation of both α- and β-cells. To test thishypothesis we examined the effect of MafB deficiency onendocrine differentiation using krENU/krENU mice. At E15.5,MafB deficiency did not affect pancreatic appearance butinsulin+ and glucagon+ cells were drastically reduced (Figs. 5A,B). As a percentage of total epithelial area, insulin+ andglucagon+ area were reduced to nearly 55% and 60%,respectively (Figs. 5C, D). Thus, as in SeyNeu/SeyNeu mice,MafB deficiency results in a reduction, but not complete loss, ofinsulin+ and glucagon+ cells.

MafB deficiency does not affect the initiation of endocrinedifferentiation

We next examined the mechanism underlying the reducednumber of insulin+ and glucagon+ cells in krENU/krENU mice.During pancreatic development, the transcription factor Ngn3marks the progenitors that give rise to endocrine cells, whilePax6 marks cells further along the endocrine differentiationpathway. Nkx2.2 is expressed in early epithelial progenitors andsubsequently becomes restricted to endocrine cells. Pancreatic

s the ability of MafB+ and PDX-1+ cells to express insulin. (A–H) Adjacentwere stained for MafB, Nkx6.1, PDX-1 and MafA in green and insulin in red.ere reduced compared to wild type pancreata. Bars: 20 μm. (I, J) Stained E15.5rmine the proportion of transcription factor+ cells expressing insulin (I), and thetions of MafB+Ins+ cells/total MafB+ cells and PDX-1+Ins+ cells/total PDX-1+

ulin. (J) The proportion of insulin+ cells expressing MafB, PDX-1 or MafAwaslarge Maf factors can activate PDX-1 expression. A −4.5 kb PDX-1 promoter:pression plasmids and pSVβ-gal was used as an internal control. Results areh the pcDNA3.1±S.E. (n=3).

Page 9: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

451W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

sections from E15.5 wild type and krENU/krENU mice wereimmunostained for Ngn3, Nkx2.2 and Pax6 (Fig. 6). AlthoughkrENU/krENU mice have reduced insulin+ cells, the number ofNgn3+ cells was unaffected (Figs. 6A, D), suggesting thatMafB

deficiency does not affect the specification of endocrinedifferentiation. Similarly, MafB deficiency did not affectNkx2.2+ or Pax6+ cell number (Figs. 6B, C, E, F). Theseresults show that the krENU mutation does not affect pancreatic

Page 10: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 8. MafB deficiency does not induce the formation of ε-cells. Sections fromE15.5 wild type (+/+) and krENU/krENU pancreata were stained to detectsomatostatin (Som) or ghrelin (Ghre) in green and a mixture of antibodies thatrecognize insulin and glucagon in red. In spite of reduction in the number ofinsulin and glucagon expressing cells, the number of somatostatin+ and ghrelin+

cells is unchanged in krENU/krENU embryos. Also, ghrelin+ glucagon− ε-cellnumbers were similar in pancreata from wild type and krENU/krENU embryos.

452 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

precursors or formation of endocrine cells and that the reductionin the number of insulin+ and glucagon+ cells (Figs. 5 and 6)occurs in the absence of any change in the levels of Pax6+ cells(Figs. 6C, F).

Effect of MafB deficiency on MafB+, PDX-1high and MafA+

cells

Whether the homozygous krENU mutation affected thematuration of insulin+ cells was addressed next. Since the kr-allele expresses a mutant protein recognized by the MafB

antibody (Fig. 4B), we examined the effect of MafB deficiencyon MafB expression. In the krENU/krENU pancreas at E15.5Nkx6.1+ cells were not affected by MafB deficiency (Figs. 7B,F), insulin+ MafB+, insulin− MafB+ cells and occasionalinsulin+ MafB− cells were seen, and the total number ofMafB+ cells showed a modest increase (∼20%) compared to thewild type (Figs. 7A, E). Unlike the effect ofMafB deficiency onits own expression, the number of PDX-1high and MafA+ cellswere reduced (Figs. 7C, D, G, H), suggesting that deficientMafB function was sufficient to reduce, but not eliminate,cells expressing PDX-1high and MafA. At E15.5, nearly 60% ofMafB+ cells (175 MafB+, 110 Ins+ and 105 MafB+Ins+ cells)expressed insulin in wild type, but this number dropped to∼25% in krENU/krENU (238 MafB+, 68 Ins+ and 65 MafB+Ins+

cells) (Fig. 7I). This observation suggests that a functionallycompromised MafB isoform (krENU) prevents a significantproportion of differentiating endocrine cells from expressinginsulin. The deficiency of MafB function also resulted in areduced proportion of PDX-1high cells co-expressing insulin(128 PDX-1high, 122 Ins+ and 92 PDX1highIns+ cells in wildtype vs. 107 PDX-1high, 90 Ins+ and 65 PDX1highIns+ cells inkrENU/krENU pancreas). This reduced ability of MafB+ andPDX-1high cells to co-express insulin in MafB deficient micewas similar to that observed in SeyNeu/SeyNeu mice (Fig. 2). InkrENU/krENU pancreas all MafA+ cells were insulin+, but not allinsulin+ cells expressed MafA (75 MafA+, 111 Ins+ and75 MafA+Ins+ cells in wild type vs. 60 MafA+, 92 Ins+ and60 MafA+Ins+ cells krENU/krENU pancreas). Interestingly, theproportion of insulin+ cells co-expressing the different tran-scription factors was unaltered in wild type and krENU/krENU

mice (Fig. 7J): nearly 95%, 75% and 65% of insulin+ cells co-expressed MafB, PDX-1high and MafA, respectively.

The ability of MafB to directly activate PDX-1 expressionwas examined by co-transfecting −4.5 kb PDX-1:luciferaseplasmid (Eto et al., 2007) and either MafA, MafB or cMafplasmids in HeLa cells (Fig. 7K). MafA, MafB and cMaf wereall capable of activating PDX-1 expression, consistent with thereported ability of MafA to induce PDX-1 expression (Samaraset al., 2003).

Unlike Pax6, MafB deficiency does not affect the cell-fatedecision of endocrine cells

As stated above, SeyNeu/SeyNeu and krENU/krENU mice showvery similar pattern of reductions of insulin+ and glucagon+

cells. Loss of Pax6 function results in endocrine cells that do

not express insulin or glucagon and that acquire an alternatecell-fate with ghrelin expression (Prado et al., 2004; Heller etal., 2005). However, in E15.5 krENU/krENU the reduction ininsulin+ and glucagon+ cells was not accompanied by anincrease in somatostatin+ or ghrelin+ cells (Fig. 8 and Artner etal., 2007). This finding is consistent with our observation inSeyNeu/SeyNeu mice that the ghrelin+ ε cells do not expressMafB (Supplementary Fig. 2). Thus, MafB deficiency, unlikePax6 deficiency, does not trigger endocrine cells to acquirealternate cell-fates.

An increased proportion of the remaining insulin+ cells inkrENU/krENU pancreata express Hb9

The presence of insulin+ and glucagon+ cells in SeyNeu/SeyNeu and krENU/krENU mice suggests the induction of thesehormones can occur via a pathway independent of Pax6 orMafB function. Two parallel pathways have been proposed totrigger the formation of insulin+ cells: one involving Nkx2.2 andPax4 in regulating the Hb9 and PDX-1 dependent expression ofinsulin, and a second involving Nkx2.2, Pax6 and PDX-1(Wang et al., 2004). Wang and colleagues reported that at E14.5,Pax6 deficiency had no obvious effect on Hb9 expression; wealso observed cells expressing Hb9 in our Pax6 deficient mice atE15.5 (Supplementary Fig. 3). Since our data suggest thatMafBfunctions downstream of Pax6, we examined whether MafBdeficiency differentially affected the formation of insulin+ cellsfrom either the Nkx2.2–Pax6 or Pax4–Hb9 pathways byquantifying immunostained E15.5 wild type and krENU/krENU

pancreata. Here, MafB deficiency increased the proportion ofinsulin+ cells expressing Hb9 but had no effect on the ability of

Page 11: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

Fig. 9. An increased proportion of insulin+ cells in the MafB deficient pancreataco-express Hb9. (A, B) E15.5 sections from wild type and krENU/krENU

pancreata were stained for Hb9 in green and insulin in red. Hb9− insulin+ cellsand Hb9+ insulin+ cells co-exist in the wild type pancreas while the number ofHb9− insulin+ cells is reduced in krENU/krENU pancreas. Bars: 20 μm. (C)Quantification of immunohistochemical data. The proportion of Hb9+ cellsexpressing insulin did not change between the krENU/krENU and wild type mice.However, the proportion of insulin+ cells that co-express Hb9 in these mice issignificantly increased (p=0.02).

453W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

Hb9+ cells to co-express insulin (220 Hb9+, 227 Ins+ and 164Hb9+Ins+ cells in wild type vs. 139 Hb9+, 111 Ins+ and 104Hb9+Ins+ cells krENU/krENU pancreas) (Fig. 9). The increasedproportion of insulin+ cells expressing Hb9 in krENU/krENU

mice suggests that these remaining insulin+ cells may bespecified via the Pax4–Hb9 pathway.

Discussion

Previously we proposed that the maturation of insulin+ cellsduring embryonic development requires a switch from aninsulin+ MafB+ state to an insulin+ MafA+ state via anintermediate PDX-1high stage. Here we use Pax6 and MafBdeficient mice to demonstrate that the deficiency of either Pax6or MafB function results in reduced number of cells expressinginsulin, glucagon, PDX-1 and MafA (Figs. 1, 3, 5, 7).Comparable proportions of Ngn3 and MafB expressing cellsin krENU/krENU and wild type pancreata (Figs. 6 and 7) suggestthat similar to Pax6 deficiency (Ashery-Padan et al., 2004),MafB deficiency does not affect the initiation of endocrine

differentiation, but affects expression of markers of terminaldifferentiation. The reduced function of either Pax6 or MafBresults in an increased proportion of MafB+ insulin− and PDX-1high insulin− cells, while the expression of MafA, althoughreduced, is always restricted to the cells that express insulin(Figs. 2 and 7), suggesting that PDX-1 and MafA functiondownstream of Pax6 and MafB. The loss of Pax6 functionreduces the numbers of MafB expressing cells (Figs. 1 and 2),while the deficiency of MafB activity has no effect on Pax6expression (Fig. 6). Taken together, these results suggest thatMafB can be a downstream mediator of Pax6 function or thatPax6 requires MafB for inducing the expression of insulin andglucagon. However, the conversion of endocrine precursors intoghrelin+ cells does not occur inMafB deficient mice (Fig. 8, andArtner et al., 2007) as observed in the absence of Pax6 function(Heller et al., 2005; Prado et al., 2004, and Supplemenraty Fig.2). Thus, the suppression of ghrelin+ ε-cells occurs at the levelof Pax6 expression, and MafB does not appear to have a directrole in regulating the formation of ε-cells.

Analysis of Nkx2.2, Pax6 and Pax4 knockout micesuggested that specification of insulin+ cells involves twodistinct pathways; one requiring Nkx2.2, Pax4, Hb9 and PDX-1, and a second involving Nkx2.2, Pax6 and PDX-1 (Wang etal., 2004). Deficiency of MafB function in krENU/krENU miceled to reduction of insulin+ and glucagon+ cells (Fig. 5) as inPax6 deficient mice (Sander et al., 1997; Wang et al., 2004;Ashery-Padan et al., 2004; Figs. 1 and 3). These observationssuggest that insulin and glucagon expression in some endocrineprecursors is independent of Pax6 andMafB function. A similarreduction in the proportion of hormone+ cells found in MafBknockout mice (Artner et al., 2007) suggests that the residualactivity of the krENU allele does not regulate the formation ofremaining hormone+ cells. We also observed in MafB deficientmice an increased proportion of remaining insulin+ cellsexpressing Hb9 (Fig. 9). This increase could be due to theselective lack of insulin+ cells derived from the Pax6–MafBpathway in these krENU/krENU mice. Thus, our results providestrong support to the two parallel pathways model proposed bySosa-Pineda and colleagues for the formation of insulin+ cells(Wang et al., 2004).

The presence of increased numbers of MafB+ insulin− cellsin SeyNeu/SeyNeu mice (Fig. 2) suggests that either Pax6function is essential for MafB to activate insulin expression orPax6 initiates insulin expression with MafB required for furthermaturation of insulin+ cells. However, in krENU/krENU mice thenumber of Pax6+ cells was unchanged while that of insulin+

cells was reduced (Fig. 6). Thus, it is most likely that in thepresence of Pax6 (or following its action), MafB initiates theexpression of insulin in some hormone− endocrine precursors.Although Pax6 andMafB are required for the formation of onlysome insulin+ cells, they are expressed in most, if not all,insulin+ cells, suggesting a role for these factors even in insulin+

cells derived from a Pax6–MafB independent pathway. Sincematuration of insulin+ cells accompanies their transition fromMafB to MafA expression (Nishimura et al., 2006), MafB mayhave two distinct roles during endocrine differentiation: oneessential for regulating the differentiation of insulin+ cells via

Page 12: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

454 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

the Pax6–MafB pathway, and the other in the maturation ofPax4–Hb9 derived insulin+ cells. Additional studies are neededto confirm the consequence of a loss of MafB or Pax6 functionon the maturation of these latter insulin+ cells.

The increased proportion of insulin+ Hb9+ cells in krENU/krENU mice would suggest that the induction of insulinexpression in the Pax4–Hb9 pathway does not depend onMafB expression. Interestingly, a recent MafB knockout study(Artner et al., 2007) suggested that MafA was required tospecify the remaining insulin+ cells. However, since MafA isexpressed only in insulin+ cells in krENU/krENU mice and manyinsulin+ cells are MafA− (Fig. 7), it is unlikely that MafAinduces insulin expression in MafB knockout mice. MafA, atthe most, might be responsible for a subpopulation of insulin+

cells (MafA+ cells), while another factor is required to triggerinsulin expression in MafA− cells. It is important to note thatMafA expression remained restricted to insulin+ cells in bothSeyNeu/SeyNeu and krENU/krENU mice, and MafA knockout mice(Zhang et al., 2005) show no reduction in insulin+ cells at birth.It seems most likely that MafA expression is initiated only afterthe induction of insulin expression in endocrine precursors,which suggests that in krENU/krENU mice a factor other thanMafA or MafB induces insulin expression in cells differentiat-ing via the Pax4–Hb9 pathway. While our analysis of MafBdeficient mice generally agrees with a recent characterization ofMafB knockout mice (Artner et al., 2007), our data do notsupport a role for PDX-1high upstream of MafB function nor arole for MafA in inducing insulin expression. Additionally, byanalyzing bothMafB and Pax6 deficient mice, the current studyextends our understanding of the role of MafB in pancreaticdevelopment. Our results strongly suggest that MafB functionsdownstream of Pax6 and is most likely responsible for the Pax6dependent loss of insulin+ and glucagon+ cells.

Analysis of SeyNeu/SeyNeu mice demonstrates a role of Pax6andMafB in the differentiation of glucagon+ cells. The majorityof glucagon positive cells present before the secondarytransition express MafB (Fig. 3), consistent with the expressionof Pax6 in the early glucagon+ cells reported earlier (Heller etal., 2004; Sander et al., 1997). It is likely that the MafB+

glucagon− cells present at E12.5 (Fig. 3) represent the insulin+

MafB+ cells seen at this stage (Nishimura et al., 2006). AtE15.5, due to the increase in insulin+ MafB+ cells, theproportion of MafB+ cells expressing glucagon decreased.Yet, the proportion of glucagon+ cells expressing MafBremained unchanged at 95% in wild type mice (Figs. 2 and3), suggesting that either the induction of glucagon expressionin a small proportion of endocrine precursors in wild type micedoes not require MafB function or that MafB is turned onrapidly after the induction of glucagon.

Loss of Pax6 function significantly reduced the number ofMafB+ cells, resulting in a significant proportion of glucagon+

cells not expressing MafB at both E12.5 and E15.5 (Fig. 3).This finding suggests either the existence of a Pax6–MafBindependent pathway for the induction of glucagon or a lack ofsustainability of MafB in these glucagon+ cells in the absence ofPax6 function. The reduction in the proportion of glucagon+

cells expressing MafB in SeyNeu/SeyNeu mice was opposite of

no effect of Pax6 deficiency on the ability of insulin+ cells toexpress MafB (Figs. 2 and 3). This observation suggestsdifferences in the specification and maturation of glucagon+

cells and insulin+ cells. Since the specification and maturationprocess of glucagon+ cells does not require the function ofPDX-1 and MafA, it is likely that MafB does not regulate thematuration process and only regulates initiation of glucagonexpression. A recent study showing different actions of Nkx2.2in rescuing glucagon and insulin-expressing cells in Nkx2.2knockout mice (Doyle et al., 2007), supports the suggestion thatthe transcription factors that regulate differentiation of bothinsulin and glucagon expression, use distinct mechanisms ineach endocrine cell type. Similarly,MafB may only be involvedin specification of glucagon+ cells whereas it may have a dualrole in the specification and maturation of insulin+ cells.

Inhibiting the proper maturation of insulin+ cells hasfunctional consequences. It has been shown thatMafA knockoutmice develop diabetes due to postnatal reduction in β-cellnumber (Zhang et al., 2005). Inhibition of MafA function ininsulin-producing cells via gene knock-down also resulted inreduced expression of genes involved in insulin synthesis andsecretion (Wang et al., 2007). MafA regulates the expression ofgranuphilin, critical for docking the insulin vesicle to the plasmamembrane (Kato et al., 2006). Furthermore, insulin+ cellsderived from human embryonic stem cells expressed MafB butnot PDX-1 and probably not MafA (D'Amour et al., 2006); thislack could contribute to their inability to secrete insulin inresponse to glucose and further highlights the importance of thematuration process.

Previously we proposed that during maturation, insulin+

cells undergo a switch from an insulin+ MafB+ state to aninsulin+ MafA+ state via an intermediate PDX-1high stage. Inthis study, we observed that nearly 95%, 75% and 65% ofinsulin+ cells in wild type, SeyNeu/SeyNeu, and krENU/krENU

mice expressed MafB, PDX-1 and MafA, respectively (Figs. 2and 7). The observed gradation in expression of these threetranscription factors in insulin+ cells as well as the ability ofMafB to activate PDX-1 expression further supports ourhypothesis that maturation of insulin+ cells proceeds from aMafB+ to MafA+ state after the induction of PDX-1high. Resultsfrom Pax6 and MafB deficient mice further support suchsequential requirement forMafB, PDX-1 and MafA functions inthe maturation process. An analysis of conditional PDX-1knockout in developing endocrine cells will be necessary toconfirm whether the MafB is upstream of PDX-1high functionandMafA is downstream of this gene. Knowledge of the precisefunction of these transcription factors during the maturation ofinsulin+ cells will play a crucial role in generating glucoseresponsive insulin producing cells.

Acknowledgments

We thank Drs. O. Madsen, P. Serup and the NIH-funded BetaCell Biology Consortium for Nkx6.1 and Ngn3 antibodies, Dr.Chris Wright for PDX-1 antibody, Dr. Sam Pfaff for Hb9antibody, Drs. Dan Drucker and Melissa Thomas for glucagonand PDX-1 reporter constructs, respectively, and Dr. Greg Barsh

Page 13: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

455W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

for krENU mice. This study was supported by research grantsfrom NIH (RO1 DK060127) and Harvard Stem Cell Institute toAS, NIH 3RO1 CA95021-04S1 to SMS, NIH (RO1DK065791) and Harvard Stem Cell Institute to RLM, JuvenileDiabetes Research Foundation postdoctoral fellowship (3-2005-74) and Mary K. Iacocca fellowship to WN, CanadianInstitute of Health Research Fellowship to SR, and the Mediaand Advanced Microscopy (Histology and Confocal facilities)Cores of the Joslin Diabetes Endocrinology Research Center(NIH DK-36836).

Appendix A. Supplementary data

Supplementary data associated with this article can be found,in the online version, at doi:10.1016/j.ydbio.2007.12.009.

References

Artner, I., Le Lay, J., Hang, Y., Elghazi, L., Schisler, J.C., Henderson, E., Sosa-Pineda, B., Stein, R., 2006. MafB: an activator of the glucagon geneexpressed in developing islet alpha- and beta-cells. Diabetes 55, 297–304.

Artner, I., Blanchi, B., Raum, J.C., Guo, M., Kaneko, T., Cordes, S., Sieweke,M., Stein, R., 2007. MafB is required for islet beta cell maturation. Proc.Natl. Acad. Sci. U. S. A. 104, 3853–3858.

Ashery-Padan, R., Zhou, X., Marquardt, T., Herrera, P., Toube, L., Berry, A.,Gruss, P., 2004. Conditional inactivation of Pax6 in the pancreas causesearly onset of diabetes. Dev. Biol. 269, 479–488.

Collombat, P., Hecksher-Sorensen, J., Broccoli, V., Krull, J., Ponte, I.,Mundiger, T., Smith, J., Gruss, P., Serup, P., Mansouri, A., 2005. Thesimultaneous loss of Arx and Pax4 genes promotes a somatostatin-producing cell fate specification at the expense of the alpha- and beta-celllineages in the mouse endocrine pancreas. Development 132, 2969–2980.

Collombat, P., Hecksher-Sorensen, J., Serup, P., Mansouri, A., 2006. Specifyingpancreatic endocrine cell fates. Mech. Dev. 123, 501–512.

Collombat, P., Hecksher-Sorensen, J., Krull, J., Berger, J., Riedel, D., Herrera,P.L., Serup, P., Mansouri, A., 2007. Embryonic endocrine pancreas andmature beta cells acquire alpha and PP cell phenotypes upon Arxmisexpression. J. Clin. Invest. 117, 961–970.

Cordes, S.P., Barsh, G.S., 1994. The mouse segmentation gene kr encodes anovel basic domain-leucine zipper transcription factor. Cell 79, 1025–1034.

D'Amour, K.A., Bang, A.G., Eliazer, S., Kelly, O.G., Agulnick, A.D., Smart,N.G., Moorman, M.A., Kroon, E., Carpenter, M.K., Baetge, E.E., 2006.Production of pancreatic hormone-expressing endocrine cells from humanembryonic stem cells. Nat. Biotechnol. 24, 1392–1401.

Doyle, M.J., Loomis, Z.L., Sussel, L., 2007. Nkx2.2-repressor activity issufficient to specify alpha-cells and a small number of beta-cells in thepancreatic islet. Development 134, 515–523.

Edlund, H., 2002. Pancreatic organogenesis—developmental mechanisms andimplications for therapy. Nat. Rev., Genet. 3, 524–532.

Eichmann, A., Grapin-Botton, A., Kelly, L., Graf, T., Le Douarin, N.M.,Sieweke, M., 1997. The expression pattern of the mafB/kr gene in birds andmice reveals that the kreisler phenotype does not represent a null mutant.Mech. Dev. 65, 111–122.

Eto, K., Kaur, V., Thomas, M.K., 2007. Regulation of pancreas duodenumhomeobox-1 expression by early growth response-1. J. Biol. Chem. 282,5973–5983.

Glaser, T., Jepeal, L., Edwards, J.G., Young, S.R., Favor, J., Maas, R.L., 1994.PAX6 gene dosage effect in a family with congenital cataracts, aniridia,anophthalmia and central nervous system defects. Nat. Genet. 7, 463–471.

Gradwohl, G., Dierich, A., LeMuer, M., Guillemot, F., 2000. Neurogenin3 isrequired for the development of the four endocrine cell lineages of thepancreas. Proc. Natl. Acad. Sci. U. S. A. 97, 1607–1611.

Grapin-Botton, A., Melton, D.A., 2000. Endoderm development: frompatterning to organogenesis. Trends Genet. 16, 124–130.

Harrington, R.H., Sharma, A., 2001. Transcription factors recognizingoverlapping C1–A2 binding sites positively regulate insulin geneexpression. J. Biol. Chem. 276, 104–113.

Heller, R.S., Stoffers, D.A., Liu, A., Schedl, A., Crenshaw III, E.B., Madsen,O.D., Serup, P., 2004. The role of Brn4/Pou3f4 and Pax6 in forming thepancreatic glucagon cell identity. Dev. Biol. 268, 123–134.

Heller, R.S., Jenny, M., Collombat, P., Mansouri, A., Tomasetto, C., Madsen,O.D., Mellitzer, G., Gradwohl, G., Serup, P., 2005. Genetic determinants ofpancreatic epsilon-cell development. Dev. Biol. 286, 217–224.

Hill, R.E., Favor, J., Hogan, B.L., Ton, C.C., Saunders, G.F., Hanson, I.M.,Prosser, J., Jordan, T., Hastie, N.D., van Heyningen, V., 1991. Mouse smalleye results from mutations in a paired-like homeobox-containing gene.Nature 354, 522–525.

Jensen, J., 2004. Gene regulatory factors in pancreatic development. Dev. Dyn.229, 176–200.

Jonsson, J., Carlsson, L., Edlund, T., Edlund, H., 1994. Insulin-promoter-factor1 is required for pancreas development in mice. Nature 371, 606–609.

Kataoka, K., Han, S.I., Shioda, S., Hirai, M., Nishizawa, M., Handa, H., 2002.MafA is a glucose-regulated and pancreatic beta-cell-specific transcriptionalactivator for the insulin gene. J. Biol. Chem. 277, 49903–49910.

Kato, T., Shimano, H., Yamamoto, T., Yokoo, T., Endo, Y., Ishikawa, M.,Matsuzaka, T., Nakagawa, Y., Kumadaki, S., Yahagi, N., Takahashi, A.,Sone, H., Suzuki, H., Toyoshima, H., Hasty, A.H., Takahashi, S., Gomi, H.,Izumi, T., Yamada, N., 2006. Granuphilin is activated by SREBP-1c andinvolved in impaired insulin secretion in diabetic mice. Cell Metab. 4,143–154.

Kim, S.K., MacDonald, R.J., 2002. Signaling and transcriptional control ofpancreatic organogenesis. Curr. Opin. Genet. Dev. 12, 540–547.

Leonard, J., Peers, B., Johnson, T., Ferreri, K., Lee, S., Montminy, M.R., 1993.Characterization of somatostatin transactivating factor-1, a novel homeoboxfactor that stimulates somatostating expression in pancreatic islet cells. Mol.Endocrinol. 7, 1275–1283.

Manzanares, M., Cordes, S., Kwan, C.T., Sham, M.H., Barsh, G.S., Krumlauf,R., 1997. Segmental regulation of hoxb-3 by kreisler. Nature 387, 191–195.

Matsuoka, T.A., Zhao, L., Artner, I., Jarrett, H.W., Friedman, D., Means, A.,Stein, R., 2003. Members of the large Maf transcription family regulateinsulin gene transcription in islet beta cells. Mol. Cell. Biol. 23,6049–6062.

Miller, C.P., McGehee, R.E., Habener, J.F., 1994. IDX-1: a new homeodomaintranscription factor expressed in rat pancreatic islets and duodenum thattransactivates the somatostatin gene. EMBO J. 13, 1145–1156.

Moriguchi, T., Hamada, M., Morito, N., Terunuma, T., Hasegawa, K., Zhang,C., Yokomizo, T., Esaki, R., Kuroda, E., Yoh, K., Kudo, T., Nagata, M.,Greaves, D.R., Engel, J.D., Yamamoto, M., Takahashi, S., 2006. MafB isessential for renal development and F4/80 expression in macrophages. Mol.Cell. Biol. 26, 5715–5727.

Murtaugh, L.C., 2007. Pancreas and beta-cell development: from the actual tothe possible. Development 134, 427–438.

Nishimura, W., Salameh, T., Kondo, T., Sharma, A., 2005. Regulation of insulingene expression by overlapping DNA-binding elements. Biochem. J. 392,181–189.

Nishimura, W., Kondo, T., Salameh, T., El Khattabi, I., Dodge, R., Bonner-Weir,S., Sharma, A., 2006. A switch fromMafB to MafA expression accompaniesdifferentiation to pancreatic beta-cells. Dev. Biol. 293, 526–539.

Offield, M.F., Jetton, T.L., Labosky, P., Ray, M., Stein, R., Magnuson, M.,Hogan, B.L.M., Wright, C.V.E., 1996. PDX-1 is required for pancreaticoutgrowth and differentiation of the rostral duodenum. Development 122,983–985.

Ohlsson, H., Karlsson, K., Edlund, T., 1993. IPF1, a homeodomain-containingtransactivator of the insulin gene. EMBO J. 12, 4251–4259.

Olbrot, M., Rud, J., Moss, L.G., Sharma, A., 2002. Identification of beta-cell-specific insulin gene transcription factor RIPE3b1 as mammalian MafA.Proc. Natl. Acad. Sci. U. S. A. 99, 6737–6742.

Prado, C.L., Pugh-Bernard, A.E., Elghazi, L., Sosa-Pineda, B., Sussel, L., 2004.Ghrelin cells replace insulin-producing beta cells in two mouse models ofpancreas development. Proc. Natl. Acad. Sci. U. S. A. 101, 2924–2929.

Reza, H.M., Yasuda, K., 2004. The involvement of neural retina pax6 in lensfiber differentiation. Dev. Neurosci. 26, 318–327.

Page 14: Preferential reduction of β cells derived from Pax6–MafB pathway in MafB deficient mice

456 W. Nishimura et al. / Developmental Biology 314 (2008) 443–456

Reza, H.M., Ogino, H., Yasuda, K., 2002. L-Maf, a downstream target of Pax6,is essential for chick lens development. Mech. Dev. 116, 61–73.

Sadl, V., Jin, F., Yu, J., Cui, S., Holmyard, D., Quaggin, S., Barsh, G., Cordes,S., 2002. The mouse Kreisler (Krml1/MafB) segmentation gene is requiredfor differentiation of glomerular visceral epithelial cells. Dev. Biol. 249,16–29.

Samaras, S.E., Zhao, L., Means, A., Henderson, E., Matsuoka, T.A., Stein, R.,2003. The islet beta cell-enriched RIPE3b1/Maf transcription factorregulates pdx-1 expression. J. Biol. Chem. 278, 12263–12270.

Sander, M., Neubuser, A., Kalamaras, J., Ee, H.C., Martin, G.R., German, M.S.,1997. Genetic analysis reveals that PAX6 is required for normal transcriptionof pancreatic hormone genes and islet development. Genes Dev. 11,1662–1673.

Sharma, A., Stein, R., 1994. Glucose-induced transcription of the insulin gene ismediated by factors required for B-cell-type-specific expression. Mol. Cell.Biol. 14, 871–879.

Sieweke, M.H., Tekotte, H., Frampton, J., Graf, T., 1996. MafB is an interactionpartner and repressor of Ets-1 that inhibits erythroid differentiation. Cell 85,49–60.

Wang, J., Elghazi, L., Parker, S.E., Kizilocak, H., Asano, M., Sussel, L., Sosa-Pineda, B., 2004. The concerted activities of Pax4 and Nkx2.2 are essentialto initiate pancreatic beta-cell differentiation. Dev. Biol. 266, 178–189.

Wang, H., Brun, T., Kataoka, K., Sharma, A.J., Wollheim, C.B., 2007. MAFAcontrols genes implicated in insulin biosynthesis and secretion. Diabetologia50, 348–358.

Zhang, C., Moriguchi, T., Kajihara, M., Esaki, R., Harada, A., Shimohata, H.,Oishi, H., Hamada, M., Morito, N., Hasegawa, K., Kudo, T., Engel, J.D.,Yamamoto, M., Takahashi, S., 2005. MafA is a key regulator of glucose-stimulated insulin secretion. Mol. Cell. Biol. 25, 4969–4976.

Zhao, L., Guo, M., Matsuoka, T.A., Hagman, D.K., Parazzoli, S.D., Poitout, V.,Stein, R., 2005. The islet beta cell-enrichedMafA activator is a key regulatorof insulin gene transcription. J. Biol. Chem. 280, 11887–11894.