• Aucun résultat trouvé

Pdx1 level defines pancreatic gene expression pattern and cell lineage differentiation

N/A
N/A
Protected

Academic year: 2022

Partager "Pdx1 level defines pancreatic gene expression pattern and cell lineage differentiation"

Copied!
9
0
0

Texte intégral

(1)

Article

Reference

Pdx1 level defines pancreatic gene expression pattern and cell lineage differentiation

WANG, Haiyan, et al.

Abstract

The absence of Pdx1 and the expression of brain-4 distinguish alpha-cells from other pancreatic endocrine cell lineages. To define the transcription factor responsible for pancreatic cell differentiation, we employed the reverse tetracycline-dependent transactivator system in INS-I cell-derived subclones INSralphabeta and INSrbeta to achieve tightly controlled and conditional expression of wild type Pdx1 or its dominant-negative mutant, as well as brain-4. INSralphabeta cells express not only insulin but also glucagon and brain-4, while INSrbeta cells express only insulin. Overexpression of Pdx1 eliminated glucagon mRNA and protein in INSralphabeta cells and promoted the expression of beta-cell-specific genes in INSrbeta cells. Induction of dominant-negative Pdx1 in INSralphabeta cells resulted in differentiation of insulin-producing beta-cells into glucagon-containing alpha-cells without altering brain4 expression. Loss of Pdx1 function alone in INSrbeta cells, which do not express endogenous brain-4 and glucagon, was also sufficient to abolish the expression of genes restricted to beta-cells and to cause alpha-cell [...]

WANG, Haiyan, et al . Pdx1 level defines pancreatic gene expression pattern and cell lineage differentiation. Journal of Biological Chemistry , 2001, vol. 276, no. 27, p. 25279-86

DOI : 10.1074/jbc.M101233200 PMID : 11309388

Available at:

http://archive-ouverte.unige.ch/unige:35182

Disclaimer: layout of this document may differ from the published version.

1 / 1

(2)

Pdx1 Level Defines Pancreatic Gene Expression Pattern and Cell Lineage Differentiation*

Received for publication, February 8, 2001, and in revised form, March 21, 2001 Published, JBC Papers in Press, April 17, 2001, DOI 10.1074/jbc.M101233200

Haiyan Wang‡§, Pierre Maechler‡, Beate Ritz-Laser, Kerstin A. Hagenfeldt‡, Hisamitsu Ishihara‡, Jacques Philippe, and Claes B. Wollheim‡§

From the‡Division of Clinical Biochemistry and theDiabetes Unit, Department of Internal Medicine, Geneva University Medical Center, CH-1211 Geneva 4, Switzerland

The absence of Pdx1 and the expression of brain-4 distinguish-cells from other pancreatic endocrine cell lineages. To define the transcription factor responsible for pancreatic cell differentiation, we employed the re- verse tetracycline-dependent transactivator system in INS-I cell-derived subclones INSr␣␤ and INSrto achieve tightly controlled and conditional expression of wild type Pdx1 or its dominant-negative mutant, as well as brain-4. INSr␣␤cells express not only insulin but also glucagon and brain-4, while INSrcells express only insulin. Overexpression of Pdx1 eliminated glucagon mRNA and protein in INSr␣␤ cells and promoted the expression of-cell-specific genes in INSrcells. Induc- tion of dominant-negative Pdx1 in INSr␣␤cells resulted in differentiation of insulin-producing-cells into glu- cagon-containing-cells without altering brain4 expres- sion. Loss of Pdx1 function alone in INSrcells, which do not express endogenous brain-4 and glucagon, was also sufficient to abolish the expression of genes re- stricted to-cells and to cause-cell differentiation. In contrast, induction of brain-4 in INSrcells initiated detectable expression of glucagon but did not affect

-cell-specific gene expression. In conclusion, Pdx1 con- fers the expression of pancreatic-cell-specific genes, such as genes encoding insulin, islet amyloid polypep- tide, Glut2, and Nkx6.1. Pdx1 defines pancreatic cell lineage differentiation. Loss of Pdx1 function rather than expression of brain4 is a prerequisite for-cell differentiation.

The pancreatic islets of Langerhans are composed of four different endocrine cell types: glucagon- (␣), insulin- (␤), soma- tostatin- (␦), and pancreatic polypeptide-producing cells (1).

These four endocrine cell lineages are differentiated from a common neurogenin3-expressing precursor (2– 6). Targeted disruption of mouse genes encoding various transcription fac- tors has demonstrated their importance in the control of islet cell development and differentiation. The homozygous deletion of thePdx1/Ipf1/Idx1/Stf1gene in mice (7, 8) and in a patient

(9) causes pancreas agenesis. InneuroD-null mice, the endo- crine (␣, ␤, and ␦) cell mass is markedly reduced (10). In Isl1-deficient mice, the dorsal pancreatic mesenchyme and four hormone-producing endocrine cell lineages are deleted (11).

Homozygous disruption of theNkx2.2gene in mice results in reduced ␣ and pancreatic polypeptide cell mass as well as defective ␤-cell differentiation (12). Pax6 is indispensable for

␣-cell development (13, 14), whereas homozygous Pax4-null mice lack ␤- and ␦-cells (15). Nkx6.1 is required for mature

␤-cell differentiation and␤-cell neogenesis during the second- ary transition (16).

The␤-cell-specific inactivation of thePdx1gene in mice has revealed that Pdx1 is required for maintaining the␤-cell phe- notype by positively regulating insulin expression and by re- pressing glucagon expression (17). Furthermore, brain4 has been suggested to confer the pancreatic␣-cell specificity (18).

The absence of Pdx1 and the expression of brain4 are charac- teristics of the mature islet ␣-cell lineage (5, 6, 8, 18 –20).

Nkx6.1 is exclusively expressed in islet␤-cells after embryonic day 13 (17, 21, 22) and is required for mature␤-cell differen- tiation (16). Pax4, which is expressed only transiently in the fetal pancreas and functions as a transcriptional repressor of the glucagon promoter, plays a significant role in the develop- ment and differentiation of islet ␤/␦ cells (15, 23–25). It has been demonstrated that Pdx1 binds to the promoters of the Nkx6.1 and Pax4, and Pdx1 is also necessary for Nkx6.1 ex- pression (17, 25, 26). However, the correlation of Pdx1 function with the expression of brain4 and Pax4 in the transcriptional hierarchy has not been well defined.

The pluripotent property of islet tumor cells has been de- scribed and used as a model to study islet cell differentiation and to identify islet cell-specific transcription factors (18, 20, 21, 27–29). INS-1 cells, which express endogenous pancreatic transcription factors Pdx1, neuroD, Isl1, Pax4, Pax6, Nkx2.2, Nkx6.1, HNF1␣, and HNF4␣(30) and display differentiation plasticity, would be suitable for elucidating the function of transcription factors in islet cell differentiation. The present study was designed to define the role of Pdx1 and brain4 in the regulation of pancreatic cell lineage differentiation using INS-1-derived stable cell lines expressing Pdx1, its dominant- negative mutant (DN1-Pdx1), or brain4 under the control of the reverse tetracycline-dependent transactivator (rtTA) (31). The established INS-1 subclones would also allow us to pinpoint the Pdx1-specific downstream target genes.

* This work was supported by Swiss National Science Foundation Grant Number 32-49755.96, the Institute for Human Genetics and Biochemistry, the Berger Foundation, the Carlos and Elsie de Reuters Foundation, and the Juvenile Diabetes Foundation International, New York. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked

“advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

§ To whom correspondence may be addressed: Division de Biochimie Clinique et de Diabe´tologie Expe´rimentale, De´partment de Me´decine Interne, Center Me´dical Universitaire, CH-1211 Geneva 4, Switzer- land. Tel.: 41 22 702 5548; Fax: 41 22 702 5543; E-mail: Haiyan.

Wang@medicine.unige.ch or Claes.Wollheim@medicine.unige.ch.

1The abbreviations used are: DN, dominant-negative; rtTA, reverse tetracycline-dependent transactivator; PCR, polymerase chain reac- tion; EMSA, electrophoretic mobility shift assay; IAPP, islet amyloid polypeptide; DOX, doxycycline.

© 2001 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org

25279

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(3)

EXPERIMENTAL PROCEDURES

Cloning of the Rat Brain4 cDNA, Construction of Plasmids, and Generation of Stable Cell Lines—The rat insulinoma INS-1 cell line- derived stable clones were cultured in RPMI 1640 in 11.2 mMglucose (30), unless otherwise indicated. The first-step stable clones INSr␣␤

and INSr␤, which express the reverse tetracycline-dependent transac- tivator, were reported previously as INS-r3 and INS-r9, respectively (32). Plasmids used in the secondary stable transfection were con- structed by subcloning the cDNAs encoding the mouse Pdx1 (kindly supplied by Dr. H. Edlund, Umea, Sweden), its dominant-negative mutant (DN-Pdx1), and the rat brain4 into the expression vector PUHD10 –3 (a kind gift from Dr. H. Bujard). DN-Pdx1 (truncated mu- tation lacking the first 79 amino acids) was PCR-amplified from Pdx1 using the following primers: 5⬘-gcaggatccgctcacctccaccaccaccttccagct-3⬘ and 5⬘-ggcagatctggccattagcttggcatcagaagc-3⬘. The PCR fragment was subcloned into modified pcDNA3.1myc (Invitrogene) and sequenced.

The rat brain4 cDNA was cloned by reverse transcription-PCR using RNA from freshly isolated rat islets and the following primers: 5⬘- gaccatggccacagctgcctc-3⬘and 5⬘-tgcagcgggccacctccttg-3⬘. The PCR prod- uct was inserted into the pGEM-T Easy Vector (Promega/Catalys) ac- cording to the manufacturer’s protocol and sequenced. The stable transfection and the clone selection and screening procedures were described previously (32).

Immunoblot and Immunofluorescence—Immunoblotting procedures were performed as described previously using enhanced chemilumines- cence (Pierce) for detection (30). The dilution for antibody against Pdx1 C terminus (a kind gift from Dr. H. Edlund) was 1:5,000.

For immunofluorescence cells were grown on polyornithine-treated glass coverslips for 1 day prior to 4 days of treatment with 500 ng/ml doxycycline. The cells were then washed, fixed in 4% paraformaldehyde, and permeabilized with 0.1% Triton X-100 in phosphate-buffered saline containing 1% bovine serum albumin. The preparation was then blocked with phosphate-buffered saline-bovine serum albumin before incubating with the first antibodies mouse monoclonal anti-human insulin (1:1,000 dilution; from Sigma) and rabbit polyclonal anti-por- cine glucagon (1:2,500 dilution) (33), followed by the second antibody labeling. The resultant immunofluorescence was viewed using a Zeiss laser scan confocal 410 microscope (Zurich, Switzerland).

Nuclear Extract Preparation and Electrophoretic Mobility Shift As- say (EMSA)—Nuclear extracts from INS-1 cells grown in culture me- dium with or without 500 ng/ml doxycycline for 24 h and from BHK-21 cells transfected with various expression plasmids were prepared according to Schreiberet al.(34). The following double-stranded oligo- nucleotides were used as probes: the rat insulin I FLAT element 5⬘- GATCTTGTTAATAATCTAATTACC-3⬘(35, 36) and the rat glucagon G1 element (37). EMSA procedures including conditions for probe la- beling, binding reactions, unlabeled probe competition, and antibody supershift were performed as previously reported (37). Polyclonal anti- bodies raised against Pdx1 and Pax-6 were generously provided by H.

Edlund and S. Saule, respectively.

Cell Extract Fractionation—Cells in 10-cm diameter dishes were cultured with or without 500 ng/ml doxycycline for 24 h at 2.5, 6, 12, and 24 mMglucose. After washing twice with ice-cold phosphate-buffered saline, the cells were suspended and allowed to swell for 15 min at 4 °C in 400␮l of hypotonic buffer composed of 20 mMTris (pH 7.4), 5 mM

EDTA, 2 mMdithiothreitol, and 0.2 mMphenylmethylsulfonyl fluoride.

After 3 cycles of freeze-thaw, the cytosolic proteins (supernatant) were separated from the nuclear fraction (pellet) by centrifugation. The nu- clear proteins were further isolated from the pellet according to Schre- iberet al.(34).

Total RNA Isolation and Northern Blotting—Cells in 10-cm diameter dishes were cultured in normal (11.2 mM) glucose medium with or without 500 ng/ml doxycycline for the indicated times, followed by an additional 8 h in culture medium with 2.5, 6, 12, and 24 mMglucose.

Total RNA was extracted and blotted to nylon membranes as described previously (32). The membrane was prehybridized and then hybridized to32P-labeled random primer cDNA probes as previously described (38).

To ensure equal RNA loading and even transfer, all membranes were stripped and rehybridized with “house-keeping gene” probes such as

␤-actin or cyclophilin. cDNA fragments used as probes for glucokinase, Glut2, insulin, Pdx1, and brain4 mRNA detection were digested from corresponding plasmids. cDNA probes for rat islet amyloid polypeptide (IAPP), somatostatin, glucagon, Nkx6.1, Nkx2.2, Pax4, Pax6, Isl1, and

␤2/NeuroD were prepared by reverse transcription-PCR and confirmed by sequencing.

Transient Transfection and Luciferase Assay—Transient transfec- tion experiments and luciferase reporter enzyme assays were carried

out as previously reported (39). To overexpress transcription factors for EMSA, non-islet Syrian baby hamster kidney (BHK-21) cells were transfected by the calcium phosphate precipitation using 10 ␮g of expression plasmids encoding quail Pax6 (S. Saule, Institut Curie, Paris, France), hamster Cdx-2/3 (M. S. German, University of Califor- nia, San Francisco, CA), and mouse Pdx1 or DN-Pdx1.

Cellular Insulin and Glucagon Content—Cells in 24-well dishes were cultured with or without 500 ng/ml doxycycline for 4 days. The content of insulin and glucagon was determined after extraction with acid ethanol or 0.2% Tween 20 in phosphate-buffered saline containing 6 milliunits/ml aprotinin, respectively, following the procedures of Wang et al. (39). Insulin was detected by radioimmunoassay using rat insulin as standard (39), and glucagon was measured with a glucagon assay kit (Linco, St. Charles, MO).

Insulin Secretion—Cells in 24-well dishes were cultured for 2 days in INS-1 medium followed by 5 h of equilibration in 2.5 mM medium.

Insulin secretion was measured in the presence of 2.5, 6, 12, or 24 mM

glucose using Krebs-Ringer bicarbonate HEPES buffer as previously described (39).

RESULTS

Characterization of INS-1-derived Subclones Expressing the rtTA and Establishment of Secondary Stable Lines Overex- pressing Pdx1, DN-Pdx1, or Brain4 —The rat insulinoma INS-1 cells were used as the parental line for stable transfection of an expression plasmid encoding the rtTA (31, 32). A clone termed INSr␤, which maintains the INS-1 ␤-cell phenotype, and an- other clone called INSr␣␤, which represents a hybrid of␣- and

␤-cells, were used for the present study. As shown in Fig. 1, INS-r␤and INS-1 cells express predominantly the␤-cell-spe- cific markers insulin and IAPP. In contrast, INSr␣␤cells ex- press not only insulin and IAPP but also glucagon and brain4 (Fig. 1). Somatostatin mRNA was abundantly expressed in freshly isolated islets but not detected in parental INS-1 cells or derived clones (Fig. 1). Pdx1 mRNA was present in both INSr␤and INSr␣␤lines (Fig. 1).

To study whether the production of glucagon in INSr␣␤cells is correlated with the expression of brain4, we generated an INSr␤-derived stable cell line expressing brain4 in a doxycy- cline-inducible manner. The cDNA encoding brain4 was ob- tained by reverse transcription-PCR using RNA from freshly isolated rat islets, cloned into pGEM-T vector, and sequenced.

To delineate the Pdx1-specific target genes and the Pdx1-reg- ulated islet cell lineage differentiation, we also established both INSr␤- and INSr␣␤-derived stable clones expressing Pdx1 or DN-Pdx1 under the control of rtTA. DN-Pdx1 represents the epitope Myc-tagged truncated Pdx1 mutant protein lacking the N-terminal transactivation domain (the first 79 amino acids) FIG. 1.Characterization of INS-1-derived INSrand INSr␣␤

subclones by Northern blot analysis of mRNA expression.Total RNAs were extracted from freshly isolated rat islets, parental INS-1 cells, and rtTA-expressing subclones INSr␤and INSr␣␤. 20-␮g RNA samples were analyzed by hybridizing with the indicated32P-labeled cDNA probes.

Pdx1 Defines Pancreatic Cell Lineage 25280

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(4)

(40). The clones, named r␤-brain4-119, r␤-Pdx1-6, r␤-DN- Pdx1-28, r␣␤-Pdx1-21, and r␣␤-DN-Pdx1-59, which expressed the transgenes at the highest level after induction and showed only background expression under non-induced conditions, were selected for the present study.

Both Pdx1 and DN-Pdx1 Proteins Translocated from the Cytoplasm to the Nucleus in a Glucose-dependent Manner but Did Not Change Molecular Mass—It has been reported that glucose stimulates translocation of Pdx1 from the cytoplasm or nuclear periphery to the nucleus in human islets and MIN6 cells (41– 43). Macfarlaneet al.(41) also demonstrated that the Pdx1 translocation is concomitant with a shift in molecular mass from 31 to 46 kDa, whereas Rafiqet al.(42) reported no such change. We investigated the intracellular location of both endogenous Pdx1 and induced Pdx1 using cytoplasmic and nuclear fractions prepared from r␣␤-Pdx1-21 cells incubated in 2.5, 6, 12, and 24 mMglucose, respectively, for 24 h (Fig. 2A).

The majority of Pdx1 protein was present in the cytosolic frac- tion when r␣␤-Pdx1-21 cells were maintained at 2.5 mMextra- cellular glucose concentration; however, in 6 mMglucose, Pdx1 predominantly translocated to the nuclear fraction (Fig. 2A).

Similar results were obtained using the r␤-Pdx1-6 line (data not shown). The 31-kDa form of Pdx1 described by Macfarlane et al.(41) was not detected by Western blotting of the cytoplas- mic fraction (Fig. 2A) or whole cell extracts (data not shown) from the INS-1-derived clones. Nevertheless, the endogenous Pdx1 protein in Min6-m9 cells (44) also translocated from the cytoplasm to the nucleus in a glucose-dependent manner but did not change molecular mass (Fig. 2B). However, there is a clear difference in glucose responsiveness in INS-1-derived cells compared with MIN6-m9 cells, because Pdx1 transloca- tion was maximally stimulated at glucose concentrations be- tween 2.5 and 6 mMin INS-1-derived clones (Fig. 2A), whereas glucose-regulated Pdx1 translocation occurred over the full range from 2.5 to 24 mM glucose in MIN6-m9 cells (Fig. 2B).

The induced DN-Pdx1 mutant protein lacking the N-terminal transactivation domain translocated in a way similar to endog- enous Pdx1 in both r␣␤-DN-Pdx1-59 (Fig. 2C) and r␤-DN- Pdx1-28 (Fig. 2D) cells, in agreement with the identification of the nuclear localization signal in the Pdx1 homeodomain (45).

It is noteworthy that the glucose responsiveness, in terms of both Pdx1 translocation and insulin secretion in INSr␣␤-de- rived cells, was equivalent to that in INSr␤-derived cells (Fig.

2, A,C, and D and Table I). The right shift of the glucose- induced Pdx1 translocation in MIN6-m9 cells (Fig. 2B) corre- sponds to the right shift of the glucose-stimulated insulin secretion in these cells (44) relative to INS-1-derived cells (Fig.

2,A,C, andDand Table I). Thus, half-maximal insulin secre- tion in MIN6-m9 cells was observed at 15 mM glucose (44), whereas the value is 6 mMin the INS-1-derived clones (Table I).

Whether or not there is a direct correlation between the glu- cose-induced Pdx1 translocation and the glucose-stimulated insulin secretion remains to be established.

Induction of Pdx1 Eliminates Glucagon Expression in INSr␣␤Cells and Promotes-Cell-specific Gene Expression in INSrCells—Quantitative Northern blotting was employed to study the impact of induction of Pdx1 on the expression of INS-1 mRNAs at extracellular glucose concentrations of 2.5, 6, 12, and 24 mM. As demonstrated in Fig. 3A, glucagon mRNA expression in INSr␣␤cells was no longer detectable after in- duction of Pdx1 for 48 h, whereas brain4 mRNA remained constant. Overexpression of Pdx1 in r␣␤-Pdx1-21 cells slightly raised the mRNA level of Nkx6.1 but had no significant effect on the mRNA expression of Nkx2.2, Pax4, Pax6, Isl1, and

␤2/NeuroD (Fig. 1A). We also performed EMSA experiments with a probe corresponding to the rat insulin I FLAT element,

which contains the Pdx1-binding site (35, 36). Using nuclear extracts from r␣␤-Pdx1-21 cells cultured with or without 500 ng/ml doxycycline for 24 h, we found that induction of Pdx1 resulted in a dramatic increase (⬎20-fold) in its binding activ- ity to the rat insulin promoter (data not shown). Unexpectedly, we did not see a concomitant rise in the mRNA levels of insulin or IAPP (Fig. 3A). Interestingly, the insulin content in r␣␤- Pdx1-21 cells was increased by more than 2-fold (⫺Dox, 33.65

⫾1.37 ng/mg or proteinversus⫹Dox, 74.55⫾17.23 ng/mg of protein;p⬍0.005) after 4 days induction of Pdx1, despite the fact that the insulin mRNA was reduced by 32.6% under the same condition. In contrast, the glucagon content in r␣␤- FIG. 2.Cellular translocation of Pdx1 and DN-Pdx1 proteins in response to incremental extracellular glucose concentrations.

Nuclear extracts and cytosolic proteins were prepared from r␣␤- Pdx1-21 (A), MIN6-m9 (B), r␣␤-DN-Pdx1-59 (C), and r␤-DN-Pdx1-28 (D) cells cultured with or without 500 ng/ml doxycycline for 24 h at the indicated glucose concentrations. 10 ␮g of protein from the nuclear extracts or 100␮g of protein from the cytosolic fraction were resolved via 9% SDS-polyacrylamide gel electrophoresis, transferred to nitrocel- lulose, and immunoblotted with an antibody against the Pdx1 C termi- nus.NE, nuclear extracts from cells in 11.2 mMglucose;kD, kilodaltons.

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(5)

Pdx1-21 cells dropped by 94% (⫺Dox, 8.09 ⫾ 1.51 ng/mg of proteinversus⫹Dox, 0.48⫾0.11 ng/mg of protein;p⬍0.001) after induction of Pdx1 for 4 days. Consistently, the immuno- fluorescence double staining of r␣␤-Pdx1-21 cells showed that the glucagon expression was no longer detectable, whereas the insulin level was concomitantly increased after doxycycline induction for 4 days (Fig. 3B).

As shown in Fig. 3C, the graded overexpression of Pdx1 in r␤-Pdx1-6 cells caused a stepwise increase in the expression of Nkx6.1 mRNA. The expression of another ␤-cell-specific marker, Glut2, was also slightly increased by induction of Pdx1 (Fig. 3C). Overexpression of Pdx1 alone is not sufficient to raise the endogenous insulin and IAPP mRNA levels (Fig. 3C). How- ever, overexpression of Pdx1 may facilitate the biosynthesis of insulin or the formation (or maturation) of insulin granules, because doxycycline treatment for 4 days also significantly increased the insulin content in r␤-Pdx1-6 cells without alter- ing insulin mRNA expression.2

Loss of Pdx1 Function Converts INSr␣␤and INSrCells into Glucagon-predominant-Cells Independent of Brain4 Expres- sion—We predicted that DN-Pdx1, which lacks the transacti- vation domain but preserves an intact DNA-binding domain (40), would exert its dominant-negative function by competing with endogenous Pdx1 for binding to the cognate site in the rat insulin promoter. Indeed, our EMSA experiments using nu- clear extracts from r␣␤-DN-Pdx1-59 cells showed that the bind- ing activity of endogenous Pdx1 to the rat insulin I FLAT element was abolished over 90% after 24 h of induction of DN-Pdx1 (data not shown). The expression of ␤-cell-specific genes encoding insulin, IAPP, Glut2, and Nkx6.1 was drasti- cally reduced but not completely eliminated after treatment of r␣␤-DN-Pdx1-59 cells with doxycycline for 4 days (Fig. 4A).

Concomitantly, the glucagon transcript level in these cells in- creased 5- and 10-fold after induction of DN-Pdx1 for 2 and 4 days, respectively, whereas brain4 mRNA expression was not altered (Fig. 4A). Similarly, immunofluorescence confocal mi- croscopy studies in r␣␤-DN-Pdx1-59 cells showed that the in- sulin-staining cells were lost by 90%, and the glucagon-staining cells accounted for over 90% of the whole cell population after treatment with doxycycline for 4 days (Fig. 4B). Accordingly, the insulin content in these cells decreased by 72% (48.15 ⫾ 8.81versus13.68⫾1.56 ng/mg of protein;p⬍0.001), whereas the glucagon content increased over 2-fold (35.77⫹7.69versus 75.07 ⫹ 6.12 ng/mg of protein; p ⬍ 0.001). Induction of DN-Pdx1 had no significant effect on the mRNA expression of

␤2/NeuroD, Pax4, Pax6, Nkx2.2, and Isl-1 (Fig. 4A), suggesting that these pancreatic transcription factors are not Pdx1-spe- cific target genes.

We also performed quantitative Northern blot analysis to investigate the consequences of DN-Pdx1 induction on the gene

expression patterns of r␤-DN-Pdx1-28 cells that do not express detectable endogenous brain4. As shown in Fig. 4C, the expres- sion of islet␤-cell-specific markers, insulin, IAPP, Glut2, and Nkx6.1 in r␤-DN-Pdx1-28 cells was dramatically decreased in a time-dependent manner (Fig. 4C). In contrast, glucagon mRNA

2H. Wang and C. B. Wollheim, unpublished data.

TABLE I

Glucose-stimulated insulin secretion in INSr␣␤- and INSr-derived clones

The amount of insulin released over 30 min from INSr␣␤-derived r␣␤-DN-Pdx1-59 clone and INSr␤-derived r␤-Pdx1-6 clone is presented as percent of insulin content. Data from six independent experiments are shown as means⫾S.E.

Glucose r␣␤-DN-Pdx1-59 r-Pdx1-6

mM

2.5 0.117⫾0.014 0.311⫾0.097

6 0.215⫾0.034 0.581⫾0.090

12 0.376⫾0.034 0.808⫾0.271

24 0.423⫾0.0839 0.894⫾0.203

FIG. 3.Induction of Pdx1 eliminates endogenous glucagon ex- pression in r␣␤-Pdx1-21 cells and promotes␤-cell-specific gene expression in r-Pdx1-6 cells.A, Northern blot analysis of gene expression in r␣␤-Pdx1-21 cells induced with 500 ng/ml doxycycline and cultured first in normal (11.2 mM) glucose medium for 24 or 48 h and then incubated further for 8 h at the indicated glucose concentrations.

20␮g of total RNA samples were analyzed by hybridizing with indi- cated cDNA probes.B, double immunofluorescence staining with anti- insulin serum (green) and anti-glucagon serum (red) in r␣␤-Pdx1-21 cells cultured in the absence (⫺Dox) or the presence (⫹Dox) of 500 ng/ml doxycycline for 4 days.C, Northern blotting quantification of gene expression in r␤-Pdx1-6 cells induced with indicated concentrations of doxycycline and cultured in normal (11.2 mM) glucose medium for 48 h.

Total RNA samples extracted from two independent experiments were blotted in parallel to demonstrate the consistency of the data. 20␮g of RNA per lane were loaded and analyzed by hybridizing with the indi- cated cDNA probes.GK, glucokinase.

Pdx1 Defines Pancreatic Cell Lineage 25282

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(6)

expression in r␤-DN-Pdx1-28 cells (Fig. 4C) rose from an un- detectable background to a level comparable with that of fully induced r␣␤-DN-Pdx1-59 cells (Fig. 4A), whereas brain4 mRNA remained undetectable (data not shown) after induction of

DN-Pdx1. These results demonstrate that dominant-negative suppression of Pdx1 function alone is sufficient to differentiate the insulin-predominant␤-cell lineage to the glu- cagon-predominant␣-cell phenotype. The brain4 expression is not required for this effect, although there is a time delay in glucagon gene expression comparing r␤-DN-Pdx1-28 (Fig. 4C) with r␣␤-DN-Pdx1-59 cells (Fig. 4A).

Although it has been proposed that Pdx1 regulates the hu- man glucokinase promoter activity (46), we found that the endogenous rat glucokinase mRNA expression was unrespon- sive to Pdx1 function in r␤-Pdx1-6 (Fig. 3C), r␤-DN-Pdx1-28 (Fig. 4C), r␣␤-Pdx1-21 (Fig. 3A), and r␣␤-DN-Pdx1-59 (Fig. 4A) cells.

Induction of Brain4 Is Sufficient for Initiating a Detectable Level of Glucagon Expression in INS-rCells but Not Mature

-Cell Differentiation—To investigate whether loss of Pdx1 function is necessary for␣-cell differentiation, we examined the gene expression patterns in r␤-brain4-119 cells using Northern blot analysis. After induction of brain4 at an extremely high level for 4 days, glucagon mRNA expression was initiated to a detectable level (Fig. 5) that is, however, 10 –20-fold lower compared with that of r␤-DN-Pdx1-28 (Fig. 4C) and r␣␤-DN- Pdx1-59 (Fig. 4A) cells under similar conditions. Unlike induc- tion of DN-Pdx1, however, forced expression of brain4 did not alter the mRNA levels of islet ␤-cell-specific genes encoding insulin, IAPP, Nkx6.1, and Glut2 (Fig. 5). Pdx1 is not the up-stream transcriptional regulator of brain4 (Figs. 3A and 4A); conversely induction of brain4 did not affect the Pdx1 expression either (Fig. 5).

Pdx1 Represses the Glucagon Promoter Activity, Binds the G1 Element, but Does Not Compete with Pax6 for DNA Bind- ing—To investigate whether Pdx1 suppresses the glucagon promoter activity, we performed transient transfection experi- ments in r␣␤-Pdx1-21 and r␣␤-DN-Pdx1-59 lines using a re- porter plasmid consisting of the firefly luciferase gene driven by the rat glucagon promoter (⫺350GlucagonLuc). As expected, induction of Pdx1 for 24 h caused a 65% reduction (⫺Dox, 5.13

⫾1.68 light units/mg of proteinversus⫹Dox, 1.81⫾0.4 light units/mg of protein;p⬍0.005; six independent experiments) in the luciferase reporter enzyme activity in r␣␤-Pdx1-21 cells transfected with⫺350GlucagonLuc. Consistently, induction of DN-Pdx1 for 24 h in r␣␤-DN-Pdx1-59 cells resulted in a 5.8-fold increase (⫺Dox, 3.66 ⫾1.07 light units/mg of proteinversus FIG. 4.Induction of DN-Pdx1 in r␣␤-DN-Pdx1-59 and r-DN-

Pdx1-28 cells converted the insulin-producing-cell lineage into the glucagon predominant␣-cell phenotype.A, quantitative evaluation by Northern blotting of the gene expression profile in r␣␤- DN-Pdx1-59 cells induced with 500 ng/ml doxycycline and cultured first in normal (11.2 mM) glucose medium for 2 or 4 days and then incubated further for 8 h at the indicated glucose concentrations. 20␮g of total RNA samples were analyzed by hybridizing with indicated cDNA probes.B, double immunofluorescence staining with anti-insulin serum (green) and anti-glucagon serum (red) in r␣␤-DN-Pdx1-59 cells cultured with or without 500 ng/ml doxycycline for 4 days. C, Northern blot analysis of gene expression patterns in r␤-DN-Pdx1-28 cells induced with 500 ng/ml doxycycline and cultured first in normal (11.2 mM) glucose medium for 4 or 7 days and then incubated further for 8 h at the indicated glucose concentrations. 20 ␮g of total RNA samples were analyzed by hybridizing with the indicated cDNA probes.GK, glucoki- nase;WT, wild type.

FIG. 5.Induction of brain4 in the r␤-brain4-119 clone triggered only a detectable level of glucagon expression but did not sup- press the-cell-specific gene expression.Northern blot analysis of gene expression in r␤-brain4-119 cells induced with 500 ng/ml doxycy- cline and cultured first in normal (11.2 mM) glucose medium for 4 days and then incubated further for 8 h at the indicated glucose concentra- tions. 20␮g of total RNA samples were analyzed by hybridizing with the indicated cDNA probes.GK, glucokinase.

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(7)

⫹Dox, 21.4 ⫾ 4.98 light units/mg of protein;p ⬍0.001; six separate experiments) in the glucagon promoter activity.

To explore the possible mechanism underlying the repressive function of Pdx1 on glucagon gene promoter activity, we per- formed EMSA experiments using nuclear extracts from r␣␤- Pdx1-21 and r␣␤-DN-Pdx1-59 cells and the G1 element of the rat glucagon promoter that confers ␣cell specificity (37). As shown in Fig. 6A, Pdx1 overexpressed in r␣␤-Pdx1-21 cells formed a complex on G1 that migrated similarly to the paired homeodomain protein Pax-6, a major G1-binding factor. Simi- larly, DN-Pdx1 from doxycycline-induced r␣␤-DN-Pdx1-59 cells interacted with the G1 element, and anti-Pdx1 antibodies specifically recognized both Pdx1 protein complexes (Fig. 6A).

To assess the affinity of DN-Pdx1 and Pax-6 for G1, we per- formed EMSA competition experiments. When BHK-21 nuclear extracts containing Pax-6 were mixed with DN-Pdx-1- containing extracts, Pdx1 formed a weaker complex on G1-56 as compared with the individual binding reaction (Fig. 6B).

Furthermore, a 10 times greater excess of cold G1-56 was required to compete for DN-Pdx1 than for Pax-6, indicating a better affinity of Pax-6 for G1 as compared with DN-Pdx1. It has previously been shown that Pdx1 has a lower affinity for G1 when compared with Pax-6 but an affinity similar to that of Cdx-2/3, a homeodomain protein interacting synergistically with Pax-6 on G1.3Similarly, Cdx-2/3 and DN-Pdx1 displayed comparable affinity for G1 (Fig. 6C). However, because Pdx1 and DN-Pdx1 had opposite effects on glucagon promoter activ- ity, it is unlikely that Pdx1 exerts transcriptional repression through its binding to the G1 element. Its ability to interact with other pancreatic transcription factors at the protein level could be an alternative explanation, because we found that Pdx1 suppressed the transactivation of Pax6 through protein- protein interaction independent of DNA binding.3

DISCUSSION

Establishment of an in Vitro Model of Islet Cell Lineage Differentiation—Overexpression of a transcription factor, CCAAT/enhancer-binding protein␤, in a pancreatic tumor cell line has recently been demonstrated to provoke the transdif- ferentiation toward the hepatocyte phenotype (47). In fact, the differentiation potential and the multihormonal phenotype of islet tumor cells have been known for a long time (27, 28). We found that rat insulinoma INS-1 cells also display high plas- ticity of differentiation, and we could actually detect by reverse transcription-PCR (data not shown) the mRNAs of four islet hormones but not the exocrine marker, p48 (48). In addition, we demonstrated that INS-1 cells express multiple endogenous pancreatic transcription factors including Pdx1, Nkx6.1, Nkx2.2, Pax4, Pax6,␤2/NeuroD, and Isl1 (30), which are im- portant for regulation of islet cell differentiation and gene expression (49). Therefore, the INS-1 cells represent neither pancreatic precursor cells nor mature differentiated mono-hor- mone-producing islet cells. We defined, by quantitative North- ern blot analysis, two rtTA-expressing INS-1-derived subclones termed INSr␤, expressing predominantly insulin, and INSr␣␤, producing both insulin and glucagon. However, we did not obtain any clone showing predominantly the ␣-cell, ␦-cell, or pancreatic polypeptide cell phenotype. We implemented the doxycycline-inducible system (Tet-on) (31) in INSr␤ and INSr␣␤ lines to study the role of Pdx1 and brain4 in the regulation of islet cell lineage differentiation and gene expres- sion. Using the inducible cell line rather than two distinct stable clones enabled us to tightly control the level of trans- genes and also to precisely quantify the gene expression pat- terns under induced and non-induced conditions. It is impor-

tant to note that doxycycline itself, or the induction of an irrelevant protein, had no effect on mRNA expression, insulin secretion, and insulin content in INS-1 cells (30, 32). We also demonstrated the differentiation plasticity of these cell lines by immunofluorescence double staining with antibodies against insulin and glucagon.

Glucose Regulates the Cellular Translocation of Pdx1 Protein

3B. Ritz-Laser, submitted for publication.

FIG. 6.Pdx1 and DN-Pdx1 bind to the glucagon gene promoter element G1.A, EMSA using nuclear extracts from r␣␤-Pdx1-21 and r␣␤-DN-Pdx1-59 cells incubated for 24 h in the absence or presence of 500 ng/ml doxycycline. Both Pdx1 and DN-Pdx1 were able to bind G1.

Px6andPx1indicate the addition of anti-Pax-6 and anti-Pdx1 antibod- ies, respectively.BandC, competition experiments analyzing the rel- ative affinity of DN-Pdx1 and Pax-6 or Cdx-2/3 for the G1 element.

Protein-DNA complexes formed with nuclear extracts from BHK-21 cells overexpressing DN-Pdx1, Pax-6, or Cdx-2/3 were competed for by the indicated molar excess of cold G1-56 oligonucleotides.

Pdx1 Defines Pancreatic Cell Lineage 25284

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(8)

without Altering Its Molecular Mass—The present study sup- ports the previous reports that glucose stimulates transloca- tion of Pdx1 from the cytoplasm or nuclear periphery to the nuclei (41, 42). In addition, we found that the majority of Pdx1 protein is located in the nucleus at physiological concentrations of glucose (6 mM). During starvation associated with hypogly- cemia, Pdx1 would be transported out of the nuclei, which may prevent the␤-cell from producing excessive insulin. Although Macfarlaneet al.(41) reported a shift in the molecular mass of Pdx1 (from 31 to 46 kDa) concomitant with the glucose-stimu- lated Pdx1 translocation, our Western blotting data demon- strated that Pdx1 protein migrated at a constant molecular mass of 46 kDa irrespective of the glucose concentration. Our results are in agreement with another study by Rafiqet al.(42).

The nuclear localization signal of Pdx1 has been identified as part of the homeodomain (45). This explains why the translo- cation of DN-Pdx1 lacking the N-terminal transactivation do- main resembled that of wild type Pdx1.

Pdx1 Paradox—Pdx1 exerts paradoxical effects on insulin mRNA expression and insulin promoter activity in insulin- producing cells and non-␤-cells (40, 50 –56). The ectopic expres- sion of Pdx1 in non-␤-cells resulted in the induction of insulin generation (29, 50) or transactivation of insulin promoter activity (40, 51–55). Consistent with previous studies in insu- lin-producing ␤-cells (52, 56), we demonstrated that overex- pression of Pdx1 did not increase, but even reduced, the insulin mRNA levels in INS-1-derived subclones. It has been hypoth- esized that the suppressive effect of Pdx1 in ␤-cells is due to cooperative interactions between Pdx1 and other transcription factors (53–57). The present study showed that overexpression of Pdx1 in INS-1 cells also raised the mRNA level of the␤-cell- restricted transcription factor Nkx6.1, known as a potent tran- scriptional repressor of the intact insulin promoter (22, 25).

The negative feedback mechanism of Nkx6.1 provides an alter- native explanation for the inhibitory effect of Pdx1 in␤-cells.

The relevance of the increase of Nkx6.1 may also explain the paradoxical effect of Pdx1 on glucagon promoter activity. We showed that induction of Pdx1 in INSr␣␤cells dramatically suppressed the glucagon promoter activity and eventually eliminated the expression of glucagon mRNA and protein. In contrast, the ectopic expression of Pdx1 in␣TC1 cells, predom- inantly expressing glucagon, did not inhibit glucagon gene transcription.3 Nkx6.1 is expressed in the insulin-producing

␤-cell lines, including INS-1-derived subclones, but not in the

␣-cell lines such as␣TC1 cells (21, 25). It is therefore likely that Nkx6.1, induced by Pdx1 (17, 25, and the present study), may indeed function as a potent transcriptional repressor (22) and contribute to the inhibitory effect of Pdx1 on glucagon gene transcription. However, this hypothesis cannot explain why targeted disruption of mouse Nkx6.1gene did not cause in- creased␣-cell mass (16). Although we found that Pdx1 is capa- ble of binding the glucagon G1 element, the binding activity is not required for the suppressive function of Pdx1 on the gluca- gon promoter.3 Protein-protein interactions with other tran- scription factors including Pax6 could also contribute to the transcriptional repression of glucagon.3Pax4, a transcriptional repressor of the glucagon promoter (23, 24), is essential for

␤/␦-cell development (15) and is expressed transiently in the differentiating fetal␤-cells but not in the differentiated adult

␤-cells (25). Although we found that Pdx1 alone did not have any significant effect on the Pax4 mRNA expression, we cannot rule out the possibility that Pdx1 suppresses glucagon pro- moter activity through synergistic interaction with Pax4.

Overexpression of Pdx1 Raises Cellular Insulin Content with- out Increasing Insulin mRNA Expression—Another intriguing function of Pdx1 is the increase in cellular insulin content,

without affecting insulin mRNA levels. We therefore suggest that Pdx1 may also regulate the biosynthesis of insulin or the formation or maturation of insulin granules.

Pdx1 Is Required for Maintaining Insulin Expression—The ectopic expression of Pdx1 alone in glucagonoma cellsin vitro (29) and in mouse hepatocytesin vivo(50) resulted in induction of insulin production, suggesting thatPdx1is a master gene in the regulation of insulin expression. We demonstrated here that dominant-negative suppression of Pdx1 function in INS-1 cells caused a drastic reduction, but not complete elimination, of the insulin mRNA expression, insulin immunofluorescence staining, and cellular insulin content. Our results are in agree- ment with the previousin vivostudy of Ahlgrenet al.(17), who showed that␤-cell-specific inactivation of the mousePdx1gene resulted in up to 90% reduction in the pancreatic insulin con- tent. We thus conclude that Pdx1 is required for maintaining insulin expression.

Loss of Pdx1 Function rather than Expression of Brain4 Is the Prerequisite for-Cell Differentiation—The absence of Pdx1 and the specific expression of brain4 are the hallmarks of differentiated ␣-cells. The present study provided profound evidence that loss of Pdx1 function rather than expression of brain4 is the determining factor for␣-cell differentiation. We showed that induction of DN-Pdx1 in INSr␣␤and INSr␤cells not only markedly repressed insulin expression but also caused pronounced expression of glucagon. We also performed immu- nofluorescence double labeling to show that dominant-negative suppression of Pdx1 function converted the insulin-producing

␤-cell lineage to the ␣-cell-dominated phenotype. In contrast, induction of brain4 in INSr␤ cells initiated only detectable amounts of glucagon, without inhibiting the␤-cell-specific gene pattern. The expression of endogenous brain4 in INSr␣␤cells may partially explain the glucagon production.

The␤-cell-specific disruption of Pdx1in vivoshifted the␤/␣ cell ratio from 5:1 to 1:1 (17), which seems moderate in com- parison to the 90% conversion of␤-cells to␣-cells in our study.

The discrepancy may be due to the slower proliferation and neogenesis or to less differentiation plasticity of islet␤-cellsin vivo.

Pdx1-specific Patterns of Gene Expression—We found that the␤-cell genes encoding insulin, IAPP, Glut2, and Nkx6.1 are Pdx1-specific target genes. Dominant-negative suppression of Pdx1 function drastically and selectively reduced the expres- sion of these mRNA species. Although it has been reported that Pdx1 binds to the Pax4 promoter (26), we showed that Pdx1 alone is not sufficient to regulate Pax4 mRNA expression.

Glucokinase expression is not restricted to islet ␤-cells al- though it is less abundant in␣-cells (58). We demonstrated that the mRNA expression of glucokinase is completely unrespon- sive to Pdx1 regulation. This concurs with a previous study on transgenic mice (17). These results contrast with the earlier claims that Pdx1 regulates the glucokinase gene (46).

Conclusion—We conclude that Pdx1 is required for main- taining the expression of␤-cell-specific genes and␤-cell lineage differentiation. Loss of Pdx1 function rather than expression of brain4 is the prerequisite for␣-cell differentiation.

Acknowledgments—We are grateful to D. Cornut-Harry, Y. Dupre, G.

Chaffard, C. Bartley, and E.-J. Sarret for expert technical assistance.

We are indebted to Drs. P. B. Iynedjian (glucokinase cDNA and INS-r3 and -r9 cells), E. Edlund (Pdx1 cDNA and antibody), S. Saule (Pax6 cDNA), M. S. German (Cdx-2/3 cDNA), B. Thorens (GLUT-2 cDNA), H.

Bujard (PUHD 10-3 vector), and N. Quintrell (pTKhygro plasmid). We also thank Dr. S. Seino for kindly providing the MIN6-m9 clone.

REFERENCES 1. Orci, L. (1982)Diabetes31,538 –565

2. Apelqvist, A., Li, H., Sommer, L., Beatus, P., Anderson, D. J., Honjo, T., Hrabe de Angelis, M., Lendahl, U., and Edlund, H. (1999)Nature400,877– 881 3. Gradwohl, G., Dierich, A., LeMeur, M., and Guillemot, F. (2000)Proc. Natl.

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

(9)

Acad. Sci. U. S. A.97,1607–1611

4. Herrera, P. L. (2000)Development127,2317–2322

5. Jensen, J., Heller, R. S., Nielsen, T. F., Pedersen, E. E., Lindsell, C., Weinmaster, G., Madsen, O. D., and Serup, P. (2000)Diabetes49,163–176 6. Schwitzgebel, V. M., Scheel, D. W., Conners, J. R., Kalamaras, J., Lee, J. E., Anderson, D. J., Sussel, L., Johnson, J. D., and German, M. S. (2000) Development127,3533–3542

7. Jonsson, J., Carlsson, L., and Edlund, H. (1994)Nature371,606 – 609 8. Offield, M. F., Jetton, T. L., Labosky, P. A., Ray, M., Stein, R., Magnuson,

M. A., Hogan, B. L. M., and Wright, C. V. E. (1996)Development122, 983–995

9. Stoffers, D. A., Zinkin, N. T., Stanojevic, V., Clarke, W. L., and Habener, J. F.

(1997)Nat. Genet.15,106 –110

10. Naya, F. J., Huang, H. P., Qui, Y., Mutoh, H., DeMayo, F. J., Leiter, A. B., and Tsai, M. J. (1997)Genes Dev.11,2323–2334

11. Ahlgren, U., Pfaff, S. L., Jessell, T. M., Edlund, T., and Edlund, H. (1997) Nature385,257–260

12. Sussel, L., Kalamaras, J., Hartigan-O’Connor, D. J., Meneses, J. J., Pedersen, R. A., Rubenstein, J. L., and German, M. S. (1998)Development 125, 2213–2221

13. Sander, M., Neubuser, A., Kalamaras, J., Ee, H. C., Martin, G. R., and German, M. S. (1997)Genes Dev.11,1662–1673

14. St-Onge, L., Sosa-Pineda, B., Chowdhury, K., Mansouri, A., and Gruss, P.

(1997)Nature387,406 – 409

15. Sosa-Pineda, B., Chowdhury, K., Torres, M., Oliver, G., and Gruss, P. (1997) Nature386,399 – 402

16. Sander, M., Sussel, L., Conners, J., Scheel, D., Kalamaras, J., Dela Cruz, F., Schwitzgebel, V., Hayes-Jordan, A., and German, M. (2000)Development 127,5533–5540

17. Ahlgren, U., Jonsson, J., Jonsson, L., Simu, K., and Edlund, H. (1998)Genes Dev.12,1763–1768

18. Hussain, M. A., Lee, J., Miller, C. P., and Habener, J. F. (1997)Mol. Cell. Biol.

17,7186 –7194

19. Larsson, L. I., Madsen, O. D., Serup, P., Jonsson, J., and Edlund, H. (1996) Mech. Dev.60,175–184

20. Serup, P., Petersen, H. V., Pedersen, E. E., Edlund, H., Leonard, J., Petersen, J. S., Larsson, L. I., and Madsen, O. D. (1995)Biochem. J.310,997–1003 21. Jensen, J., Serup, P., Karlsen, C., Nielsen, T. F., and Madsen, O. D. (1996)

J. Biol. Chem.271,18749 –18758

22. Mirmira, R. G., Watada, H., and German, M. S. (2000)J. Biol. Chem.275, 14743–14751

23. Fujitani, Y., Kajimoto, Y., Yasuda, T., Matsuoka, T.-A., Kaneto, H., Umayahara, Y., Fujita, N., Watada, H., Miyazaki, J.-I., Yamasaki, Y., and Hori, M. (1999)Mol. Cell. Biol.19,8281– 8291

24. Smith, S. B., Ee, H. C., Conners, J. R., and German, M. S. (1999)Mol. Cell.

Biol.19,8272– 8280

25. Watada, H., Mirmira, R. G., Leung, J., and German, M. S. (2000)J. Biol.

Chem.275,34224 –34230

26. Smith, S. B., Watada, H., Scheel, D. W., Mrejen, C., and German, M. S. (2000) J. Biol. Chem.275,36910 –36919

27. Madsen, O. D., Andersen, L. C., Michelsen, B., Owerback, D., Larsson, L.-I., Lernmark, A., and Steiner, D. F. (1988)Proc. Natl. Acad. Sci. U. S. A.85, 6652– 6656

28. Madsen, O. D., Karlsen, C., Nielsen, E., Lund, K., Kofod, H., Welinder, B., Rehfeld, J. F., Larsson, L.-I., Steiner, D. F., Holst, J. J., and Michelsen, B. K. (1993)Endocrinology133,2022–2030

29. Serup, P., Jensen, J., Andersen, F., Jorgensen, M. C., Blume, N., Holst, J. J., and Madsen, O. D. (1996)Proc. Natl. Acad. Sci. U. S. A.93,9015–9020 30. Wang, H., Maechler, P., Antinozzi, P. A., Hagenfeldt, K. A., and Wollheim,

C. B. (2000)J. Biol. Chem.17,35953–35959

31. Gossen, M., Freundlieb, S., Bender, G., Mu¨ller, G., Hillen, W., and Bujard, H.

(1995)Science268,1766 –1769

32. Wang, H., and Iynedjian, P. B. (1997)Proc. Natl. Acad. Sci. U. S. A.94, 4372– 4377

33. Trimble, E. R., Halban, P. A., Wollheim, C. B., and Renold, A. E. (1982)J. Clin.

Invest.69,405– 413

34. Schreiber, E., Matthias, P., Muller, M. M., and Schaffner, W. (1988)EMBO J.

7,4221– 4229

35. German, M. S., Moss, L. G., Wang, J., and Rutter, W. J. (1992)Mol. Cell. Biol.

12,1777–1788

36. German, M. S., Wang, J., Chadwick, R. B., and Rutter, W. J. (1992)Genes Dev.

6,2165–2176

37. Ritz-Laser, B., Estreicher, A., Klages, N., Saule, S., and Philippe, J. (1999) J. Biol. Chem.274,4124 – 4132

38. Wang, H., Antinozzi, P. A., Hagenfeldt, K. A., Maechler, P., and Wollheim, C. B. (2000)EMBO J.19,4257– 4264

39. Wang, H., Maechler, P., Hagenfeldt, K. A., and Wollheim, C. B. (1998)EMBO J.17,6701– 6713

40. Peshavaria, M., Henderson, E., Sharma, A., Wright, C. V. E., and Stein, R.

(1997)Mol. Cell. Biol.17,3987–3996

41. Macfarlane, W. M., McKinnon, C. M., Felton-Edkins, Z. A., Cragg, H., James, R. F. L., and Docherty, K. (1999)J. Biol. Chem.274,1011–1016 42. Rafiq, I., da Silva Xavier, G., Hooper, S., and Rutter, G. A. (2000)J. Biol.

Chem.275,15977–15984

43. Rafiq, I., Kennedy, H. J., and Rutter, G. A. (1998) J. Biol. Chem. 273, 23241–23247

44. Minami, K., Yano, H., Miki, T., Nagashima, K., Wang, C.-Z., Tanaka, H., Miyazaki, J.-I., and Seino, S. (2000)Am. J. Physiol.279,E773–E781 45. Hessabi, B., Ziegler, P., Schmidt, I., Hessabi, C., and Walther, R. (1999)Eur.

J. Biochem.263,170 –177

46. Watada, H., Kajimoto, Y., Umayahara, Y., Matzuoka, T., Kaneto, H., Fujitani, Y., Kamada, T., Kawamori, R., and Yamasaki, Y. (1996) Diabetes45, 1478 –1488

47. Shen, C.-N., Slack, J. M. W., and Tosh, D. (2000)Nat. Cell Biol.2,879 – 887 48. Krapp, A., Knofler, M., Frutiger, S., Hughes, G. J., Hagenbuchle, O., and

Wellauer, P. K. (1996)EMBO J.15,4317– 4329 49. Edlund, H. (1998)Diabetes47,1817–1823

50. Ferber, S., Halkin, A., Cohen, H., Ber, I., Einav, V., Goldberg, I., Barshack, I., Seijffers, R., Kopolovic, J., Kaiser, N., and Karasik, A. (2000)Nat. Med.6, 568 –572

51. Glick, E., Leshkowitz, D., and Walker, M. D. (2000) J. Biol. Chem.275, 2199 –2204

52. Marshak, S., Totary, H., Cerasi, E., and Melloul, D. (1996)Proc. Natl. Acad.

Sci. U. S. A.93,15057–15062

53. Marshak, S., Benshushan, E., Shoshkes, M., Havin, L., Cerasi, E., and Melloul, D. (2000)Mol. Cell. Biol.20,7583–7590

54. Ohneda, K., Mirmira, R. G., Wang, J., Johnson, J. D., and German, M. S.

(2000)Mol. Cell. Biol.20,900 –911

55. Petersen, H. V., Serup, P., Leodard, J., Michelsen, B. K., and Madsen, O. D.

(1994)Proc. Natl. Acad. Sci. U. S. A.91,10465–10469

56. Seijffers, R., Ben-David, O., Cohen, Y., Karasik, A., Berezin, M., Newgard, C. B., and Ferber, S. (1999)Endocrinology140,3311–3317

57. Peers, B., Leonard, J., Sharma, S., Teitelman, G., and Montminy, M. R. (1994) Mol. Endocrinol.8,1798 –1806

58. Heimberg, H., De Vos, A., Moens, K., Quartier, E., Bouwens, L., Pipeleers, D., Van Schaftingen, E., Madsen, O., and Schuit, F. (1996)Proc. Natl. Acad.

Sci. U. S. A.93,7036 –7041

Pdx1 Defines Pancreatic Cell Lineage 25286

at Bibliotheque Faculte Medecine Geneve, on October 1, 2012www.jbc.orgDownloaded from

Références

Documents relatifs

In the ThingWorld system [39, 35], rather than describing high-frequency surface details using a finite element model with as many degrees of freedom as there are

S'ils se prenaient pour des Rambo c'était leur problème, mais certainement pas le sien et elle, elle n'était pas sur terre pour se faire chier, surtout dans ce quartier de

supérieure doit être à au moins 2 cm en dessous du volet. Le viseur fixé sur la prothèse permet de réaliser le perçage des trous. La longueur des vis est mesurée et les vis

Access and use of this website and the material on it are subject to the Terms and Conditions set forth at Some unusual insects in houses..

Thus a high-frequency tetanus produces a blockade of the spontaneous activities of STN neurons as a result of a strong depres- sion of intrinsic voltage-gated currents

2) A l’aide d’un algorithme, donner, au jour près, le temps nécessaire pour que le plant de maïs atteigne une hauteur supérieure à 1, 5 m.. 3) On s’intéresse à la vitesse

Anionic phospholipids represent only minor fraction of cell membranes lipids but they are critically important for many membrane-related processes, including membrane identity,

À ton tour de faire apparaître cet animal dans ces deux tableaux de Hodler grâce aux autocollants de la planche n°5!. Hodler représente parfois le même animal, la même femme,