当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第9期 > 正文
编号:11256709
Conditional Expression Demonstrates the Role of the Homeodomain Transcription Factor Pdx1 in Maintenance and Regeneration of -Cells in the A
     1 Autoimmunity and Transplantation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia

    2 Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas

    ABSTRACT

    The homeodomain transcription factor Pdx1 is essential for pancreas development. To investigate the role of Pdx1 in the adult pancreas, we employed a mouse model in which transcription of Pdx1 could be reversibly repressed by administration of doxycycline. Repression of Pdx1 in adult mice impaired expression of insulin and glucagon, leading to diabetes within 14 days. Pdx1 repression was associated with increased cell proliferation predominantly in the exocrine pancreas and upregulation of genes implicated in pancreas regeneration. Following withdrawal of doxycycline and derepression of Pdx1, normoglycemia was restored within 28 days; during this period, Pdx1+/Ins+ and Pdx+/InseC cells were observed in association with the duct epithelia. These findings confirm that Pdx1 is required for -cell function in the adult pancreas and indicate that in the absence of Pdx1 expression, a regenerative program is initiated with the potential for Pdx1-dependent -cell neogenesis.

    Pdx1 is a homeodomain transcription factor essential for pancreatic development (1eC3). Early in mouse development (embryonic day 8.5), Pdx1 is highly expressed in a region of the posterior foregut endoderm, from which the dorsal and ventral pancreatic buds arise. Its expression then becomes progressively restricted to endocrine cells (4), and in the adult pancreas Pdx1 regulates several genes including insulin, glucose transporter-2, and glucokinase (5eC7) integral to -cell function. The adult pancreas retains the capacity to increase its -cell mass in response to physiological requirements, e.g., as in pregnancy (8) or in response to injury (9). -Cell neogenesis is presumed to depend on the existence of progenitor -cells in the adult pancreas, although the origin of progenitors from ductal (10eC12), islet (13,14), exocrine (15eC19), or multilineage (20) cells remains contentious (21,22). In addition to its key role in pancreas development, Pdx1 is implicated in -cell neogenesis, being expressed in duct and duct-associated cells in models of pancreas regeneration (20,23,24). It is unclear, however, whether Pdx1 is required early for the activation of a regenerative program or later for the differentiation of putative progenitor cells. Investigating the role of Pdx1 in the adult pancreas is precluded by the fact that pancreas development is arrested following specification of the foregut endoderm in mice lacking Pdx1 (1eC3). To investigate the role of Pdx1 postdevelopment, we created a mouse model (25) in which the expression of Pdx1 can be reversibly repressed by administering the tetracycline analog, doxycycline. Here, we employ this model to investigate the requirement for Pdx1 in -cell function and regeneration in the adult pancreas.

    RESEARCH DESIGN AND METHODS

    Generation of knock-in and transgenic mice.

    The basic elements of the Pdx1 tet-off inducible gene repression system are shown schematically in Fig. 1. Pdx1tTA knock-in and TgPdx1 transgenic mice were generated as previously described (25). To obtain mice in which Pdx1 could be conditionally repressed, the Pdx1tTA/+ knock-in mice were crossed with TgPdx1 mice and progeny intercrossed to derive Pdx1tTA/tTA;TgPdx1 mice as well as genotype control mice (see RESULTS).

    Animal care and treatment.

    Animals were housed under standard 12-h light/dark conditions and fed and watered ad libitum. Experiments were approved by the Royal Melbourne Hospital Campus Animal Research Ethics Committee. Groups of mice were age and sex matched within each experiment. Doxycycline was administered as a single intraperitoneal dose of 100 mg/kg at time t = 0 and maintained by addition to drinking water at 0.5 mg/ml. BrdU (100 mg/kg) was administered intraperitoneally as a single dose 16 h before the mice were killed.

    Glucose tolerance test.

    Mice were fasted overnight and challenged with 2 g/kg i.p. glucose. Glucose levels in retroorbital venous blood samples were measured at 0, 15, 30, 60, 120, and 180 min postchallenge with an Accu-chek Advantage glucometer (Roche Diagnostics, Castle Hill, NSW, Australia). Statistical analyses (Mann-Whitney tests) were performed using Graphpad Prism (GraphPad Software, San Diego, CA).

    Immunocytochemistry and immunofluorescence.

    Adult pancreata were fixed in Histochoice (Sigma, St. Louis, MO) or 4% paraformaldehyde for 3 h before processing, embedding in paraffin, and sectioning (5 e). The following primary antibodies were used: guinea pig anti-human insulin (Dako, Carpinteria, CA) at 1:1,000, rabbit anti-human amylase (Sigma) at 1:1,000, rabbit anti-human glucagon (Dako) at 1:200, rabbit anti-human somatostatin (Dako) at 1:100, rabbit anti-GLUT2 (a gift from Bernard Thorens, Universitee de Lausanne, Lausanne, Switzerland) at 1:100, and rabbit anti-human filtrin (Alpha Diagnostic, San Antonio, TX) at 1:20. Rabbit anti-Pdx1 (a gift from Christopher Wright, Vanderbilt University Medical Center, Nashville, TN) was used at 1:500 and rabbit anti-Pdx1 antibody (purified IgG 1:25) was raised against a glutathione S-transferase fusion protein containing amino acids 14eC284 of murine Pdx1. Fluorescein isothiocyanateeCconjugated anti-guinea pig (ICN Biomedicals, Aurora, OH) or Alexa fluor 568eCconjugated goat anti-rabbit (Molecular Probes, Eugene, OR) immunoglobulins were used for immunofluorescence. Nuclei were counterstained with 4',6'-diamidine-2'-phenylindole dihydrochloride (DAPI). BrdU-labeled cells were detected with a cell proliferation detection kit (Amersham, Buckinhamshire, U.K.) with mouse monoclonal anti-BrdU primary and Alexa fluor 568eCconjugated anti-mouse secondary antibodies. Terminal deoxynucleotide transferase-mediated dUTP nick-end labeling was performed with an apoptag fluorescein apoptosis detection kit (Serologicals, Norfolk, GA). Digital images were captured with an Axiocam camera from an Axioplan2 compound microscope (Carl Zeiss, Gttingen, Germany). Dual-color immunofluorescence images were compiled from two separate images of the same section using fluorochrome-specific filters.

    BrdU-labeling index was determined by imaging 6eC8 fields (200x magnification) of randomly selected pancreas sections from each of three untreated or 21-day doxycyclineeCtreated, or six 14-day doxycyclineeCtreated, mice per group. BrdU+ cells were counted with National Institutes of Health image software (National Institutes of Health, Bethesda, MD). Quantitative analysis of glucagon- and insulin-stained pancreata was performed with Axiovision 4.2 image analysis software (Carl Zeiss). Briefly, 4eC8 nonoverlapping fields containing islets (coimmunostained with separate flourophores for insulin and glucagon and counterstained with DAPI) were imaged from pancreata of 2eC3 animals for each treatment group. Insulin- and glucagon-stained areas were determined by performing identical semiautomated image analysis routines on each of the separate fluorescent image channels. Islet areas were determined by manually delineating the islets, and costained cells were counted manually on the merged immunofluorescent images.

    Differential gene expression and real-time PCR analyses.

    Total RNA was isolated with RNAzol B (Tel-Test, Friendswood, TX) from the pancreata of age- and sex-matched Pdx1tTA/tTA;TgPdx1 mice either untreated (n = 3) or treated (n = 4) with doxycyline for 14 days. Equal amounts of RNA were pooled to create +dox and eCdox groups for comparison on Affymetrix gene chip arrays (Affymetrix, Santa Clara, CA). A total of 30 e total RNA was used to generate fragmented cRNA as per Affymetrix gene chip protocols. cRNA quality was assessed by hybridization of 5 e fragmented cRNA to Affymetrix Test 3 arrays before hybridizing 20 e fragmented cRNA to Affymetrix MGU74Av2 gene chip arrays. Chips were processed and scanned as per protocols in the Affymetrix gene chip expression analysis technical manual. Analysis of the probe data were performed as follows. The perfect match probe intensities were background corrected and normalized across arrays and then summarized for each probe set using robust multichip analysis (26,27). Once expression levels had been calculated, differentially expressed genes were ranked and values expressed as fold change.

    Quantitative real-time RT-PCR was performed on the Roche Lightcycler (Roche Diagnostics). For each gene to be assayed, a template was amplified by PCR and subcloned into a pGEM-T vector (Promega, Madison, WI). The templates were used to generate standard curves for quantitative determination of mRNA expression level in the individual RNAs described above. Expression levels were corrected relative to the expression of the of -actin gene. Sequences of oligonucleotide primers used are available on request.

    Islet area determination.

    Islet area was determined for pancreata of mice that were either untreated or treated with doxycycline for 14 days (four mice per group). Three hematoxylin and eosineCstained sections from each pancreas were sampled, each section separated by 250eC300 e. Nonoverlapping 40x magnification fields were captured with an Axiocam digital camera (Carl Zeiss), and the total tissue and islet area per field was determined using Axiovision 4.2 image analysis software (Carl Zeiss). All sections were scored blind with respect to treatment. Statistical significance was determined using a Mann-Whitney U test (GraphPad Software).

    RESULTS

    Conditional repression of Pdx1.

    Two genetic modifications were required to achieve conditional repression of Pdx1 (25) (Fig. 1). First, the endogenous Pdx1 gene was inactivated by replacing its coding region with that of the tetracycline transactivator (tTA off) by homologous recombination. As a result, transcription of the tTA gene is directed by the endogenous Pdx1 transcriptional regulatory sequences. Mice with one inactivated Pdx1 allele develop normally (1eC3) and can be maintained in the heterozygous state. Second, mice bearing a tTA-responsive Pdx1 gene were created by introducing a bicistronic transgene containing a Pdx1 mini-gene and an enhanced green fluorescence protein reporter gene under the control of a tTA-responsive promoter. When the two mouse lines were crossed, some of the progeny contained both the knocked-in tTA gene and the tTA-responsive transgene. Mice homozygous for the Pdx-tTAeCoff knock-in allele are dependent on tTA-mediated transcription of the transgene for expression of Pdx1. In this system, mice with the Pdx1tTA/tTA;TgPdx1 genotype develop normally because transgenic Pdx1 expression is driven, indirectly, by tTA expressed off the native Pdx1 promoter. Administration of doxycycline prevents the tTA from binding to the tTA-responsive promoter, thereby deactivating transcription of the transgene and depleting Pdx1 (Fig. 1). We previously reported (25) that Pdx1tTA/tTA;TgPdx1 mice survive to adulthood and maintain normal glucose homeostasis.

    Doxycycline-mediated repression of Pdx1 progressively impairs -cell function.

    Adult mice were treated with doxycycline for 0, 7, 14, or 21 days, and -cell function was assessed weekly by intraperitoneal glucose tolerance tests (Fig. 2A). Untreated Pdx1tTA/tTA;TgPdx1 mice had normal fasting blood glucose levels and responded normally to a glucose challenge, recovering to basal levels within 3 h. However, within 7 days of starting doxycycline, the early glucose response of Pdx1tTA/tTA;TgPdx1 mice was increased and their ability to recover after the glucose challenge was impaired, similar to that of the Pdx1tTA/+ mice. By 14 days of doxycycline treatment, Pdx1tTA/tTA;TgPdx1 mice had diabetes (Fig. 2A).

    Although pancreas histology of Pdx1tTA/tTA;TgPdx1 mice treated with doxycycline for 14 days was grossly normal, the islets appeared to be smaller (Fig. 2B). Morphometric analysis confirmed a significant decrease (P < 0.05) in the islet area of treated animals (Fig. 2D). Transferase-mediated dUTP nick-end labeling staining was performed to determine whether impaired -cell function in doxycycline-treated Pdx1tTA/tTA;TgPdx1 mice was associated with -cell apoptosis. Transferase-mediated dUTP nick-end labelingeCpositive cells were detected at a similar low frequency in both control and doxycycline-treated mice, predominantly in the exocrine pancreas, labeled cells being rarely detected in the islets of either treated or untreated mice (data not shown). Staining for filtrin (28), a member of the nephrin family (29) whose expression is restricted to -cells (28,30), revealed that -cells, although lacking insulin (see below), were still present after 14 (Fig. 2C) and 28 days of doxycycline treatment. Quantitative RT-PCR demonstrated a decrease in Pdx1 and insulin transcripts and a corresponding increase in glucagon transcripts in pancreata of doxycycline-treated mice (Fig. 2E). In doxycycline-treated mice, histological analysis of the pancreas revealed total absence of Pdx1 (Fig. 3A, panel f) and a marked reduction of insulin, with only a few cells in each islet staining strongly (Fig. 3A, panel f). Glucagon-positive cells were prominent, as well as cells costaining for glucagon and insulin (Fig. 3A, panel h). Expression of the glucose transporter Glut2 was undetectable in the islets of Pdx1-repressed mice (Fig. 3A, panel i). Amylase was restricted to the exocrine pancreas, although its staining appeared to be reduced (Fig. 3A, panel j), consistent with our previous finding that the level of amylase mRNA is reduced in Pdx1-repressed mice (25). Untreated Pdx1tTA/tTA;TgPdx1 mice (Fig. 3A, panels aeCe) and control Pdx1+/+;TgPdx1 mice (Fig. 3A, panels keCo) had normal expression of Pdx1, insulin, somatostatin, glucagon, Glut2, and amylase.

    Quantitative image analysis of the pancreata of doxycycline-treated mice revealed that the decrease in insulin staining corresponded with an increase in glucagon staining (Fig. 3B) and with an increase in the number of islet cells coexpressing insulin and glucagon (Fig. 3C), confirming the quantitative RT-PCR results. These observations suggest that Pdx1 normally supresses expression of the glucagon gene in -cells.

    Doxycycline-induced diabetes is reversible.

    A key feature of the tet-off system is the ability to reversibly control expression of the tTA-dependent transgene. Therefore, to demonstrate that derepression of the Pdx1 transgene would restore -cell function, we withdrew doxycycline after 14 days administration to Pdx1tTA/tTA;TgPdx1 mice. Blood glucose levels of 8-h fasted mice were then measured every 7 days for 35 days. Blood glucose levels decreased significantly by 14 days and progressively until four of five mice were normoglycemic by 28 days, demonstrating that doxycycline-induced diabetes is reversible (Fig. 4A). Histological analysis revealed Pdx1 protein in some islet cells 7 days after doxycycline withdrawal, although insulin expression remained low (Fig. 4B). However, by 14 days, both Pdx1 and insulin were expressed homogeneously at pretreatment levels throughout the islets (Fig. 4B), and both the glucagon-positive islet area (Fig. 3B) and the number of insulin+/glucagon+ costained cells per islet (Figs. 3C and 4C) had also decreased.

    Repression of Pdx1 initiates proliferation of pancreatic exocrine and duct-associated cells.

    Foci of duct proliferation comprising small ductules embedded in connective tissue, similar to those observed in other models of pancreas regeneration (10,31), were present in mice treated with doxycycline for 14 days and became more prominent the longer the duration of doxycycline administration (Fig. 5A, upper panel). Staining for proliferating cell nuclear antigen confirmed the presence of proliferating cells within these ductal-like structures (Fig. 5A, lower panel). To quantitate cellular proliferation, Pdx1tTA/tTA;TgPdx1 mice either untreated or treated with doxycycline for 14 or 21 days were pulsed with BrdU overnight before they were killed. Immunofluorescence analysis revealed increased nuclear labeling in the pancreata of Pdx1-repressed mice at both 14 (P < 0.0005) and 21 (P < 0.0001) days (Fig. 5B). BrdU-positive cells were more frequent within exocrine tissue, although they were also observed proximate to ducts, within foci of duct proliferation and in islets (Fig. 5B).

    Derepression of Pdx1 leads to the appearance of duct-associated insulin+ cells.

    Following withdrawal of doxycycline, insulin+ cells were primarily detected in islets or smaller islet-like cell clusters (Fig. 5C). Pdx1 was observed at high levels in islets and islet-like cell clusters in insulin+ cells (Fig. 5C). In addition, insulin+ cells expressing Pdx1 were observed embedded in or closely associated with ducts (Fig. 5C). Pdx1+/insulineC cells were also observed associated with ducts (Fig. 5C, panel b), but the Pdx1 staining in these cells was generally of lower intensity than in the insulin+ cells.

    Repression of Pdx1 upregulates genes implicated in pancreas regeneration.

    To further characterize the molecular events associated with Pdx1 repression and potentially identify novel Pdx1-regulated genes, pancreatic RNA was isolated from Pdx1tTA/tTA;TgPdx1 mice that had been treated with doxycycline for 14 days and from untreated mice, and gene expression profiles were compared on Affymetrix MGU74Av2 gene chip arrays. To minimize variation, the gene chip analyses were performed on pooled RNA samples from treated and untreated mice. The experiment was replicated with independent cohorts of mice, and the results obtained were essentially identical. Pdx1 repression was associated with upregulation of 97 transcripts and downregulation of 29 transcripts by at least 1.8-fold (Table 1). Gene ontology associations revealed that >50% of the upregulated genes were involved in metabolism or cell growth and/or maintenance (Fig. 6A). The results were validated by real-time RT-PCR of several genes in individual mouse RNA samples that had been pooled for the gene chip experiments (Figs. 2E and 6B). Despite variation between individual samples, expression levels determined by real-time RT-PCR correlated well with the fold change determined from the gene chip analysis of pooled RNA. Of the downregulated genes, the most notable were the insulin genes ins1 and ins2, validating the results of the gene chip strategy. Upregulated genes notably included several previously identified by differential gene expression screens in models of pancreas regeneration (32,33), in particular the regenerating isleteCderived (Reg) family (Reg2, Reg3, Reg3, and Reg3) that plays a role in the maintenance of progenitor cell populations and endocrine differentiation via the notch signal transduction pathway and transducer of ErbB2.1 (Tob1) involved in the regulation of cell proliferation and transformation. Novel transcripts were also identified in the screen. One of these, 1810015C04Rik (Entrez Gene ID 66270) was confirmed by RT-PCR to be upregulated in all of the Pdx1-repressed mice (Fig. 6B) and found to encode a highly conserved protein of unknown function. It has been previously identified in a number of tissues, including embryonic pancreas (Unigene Mm.25311), suggesting a potential role for this gene in early pancreas development. RT-PCR analysis revealed expression of 1810015C04Rik in two pancreatic -cell lines, Min1046 and TC-3, as well as in kidney and liver but not in the -cell line, TC-1 (Fig. 6C).

    DISCUSSION

    Using genetically modified mice in which Pdx1 transcription can be reversibly repressed, we confirm the critical role of Pdx1 in maintaining -cell function. Furthermore, we provide evidence that Pdx1 normally suppresses glucagon expression in -cells and that repression of Pdx1 leads to enhanced proliferation of presumptive -cell progenitors.

    Mutations of Pdx1 leading to impaired -cell function underlie maturity-onset diabetes of the young type 4 in humans, and several studies (5eC7,34,35) have demonstrated a dose-dependent requirement for Pdx1 in the maintenance of normal glucose homeostasis in mice. These studies initially examined pancreatic function in adult mice with one Pdx1 allele inactivated (Pdx1+/eC) and more recently used transgenic approaches to conditionally inactivate (5) or inducibly repress Pdx1 either with antisense Pdx1 (6) or Pdx1-specific ribozyme (7) strategies. However, these models are limited by their dependence on expression of molecules under the control of the insulin promoter, which is downregulated following repression of Pdx1, resulting in relatively mild impairment of glucose tolerance (6,7) or the late onset of diabetes in the case of RIP-Cre-Pdx1 mice (5). In our model, the coding sequence for the tet-off regulatory protein (tTA) is incorporated into the Pdx1 locus so that normal temporal and spatial regulation of the gene is maintained, thereby overcoming a problem inherent in many transgenic systems that utilize partial regulatory sequences out of their genomic context. The genetically modified mice are morphologically and functionally normal, yet doxycycline treatment of adult mice repressed Pdx1 expression and induced marked hyperglycemia associated with reduced expression of insulin and the glucose transporter, Glut2. These effects were reversible upon withdrawal of doxycycline. This is therefore a truly inducible gene knock-out model in which the role of the gene can be studied at any time during development or in mature animals. -Cells were still present in Pdx1-repressed mice, and some contained residual insulin, which is not surprising as Pdx1 is only one of a number of transcription factors involved in the regulation of insulin transcription (36). Indeed, mice heterozygous for Pdx1 have normal pancreatic insulin content, although their ability to secrete insulin and clear glucose is impaired (5,37). We confirmed that the expression of several genes involved in the maintenance of glucose homeostasis, including insulin (38), Glut2 (39), and glucokinase (40), as is regulated by Pdx1 in the inducible antisense Pdx1 repression model of Lottmann et al. (6). Similarly, we were unable to detect Glut2 in the islets of Pdx1-repressed mice. It is likely that the loss of Glut2 leads to impaired glucose sensing and contributes to the diabetic phenotype of the Pdx1-repressed mice. In addition, we observed a reduction in islet area in Pdx1-repressed mice, indicating loss of the ability to maintain -cell mass in the absence of Pdx1. It has been recently reported that haploinsufficiency of Pdx1 may be responsible for increased apoptosis of islet cells in Pdx1+/eC mice (41). We did not find evidence for increased -cell apoptosis in Pdx1-repressed mice. Nevertheless, as apoptosis is a rapid and transient event, it is possible that a population of apoptosis-sensitive -cells may have been ablated before the 7-day time point at which our first observations were made.

    In addition to the progressive loss of insulin expression in -cells during Pdx1 repression, we observed a concomitant increase in glucagon expression and in the number of cells expressing both insulin and glucagon in islets. These observations are consistent with studies demonstrating that glucagon expression is repressed by Pdx1 in -cell lines (42,43) and that the -cell mass is increased in -celleCspecific Pdx1 knock-down mouse models (5,7). Similarly, ectopic expression of Pdx1 in -cell lines has been shown to repress glucagon gene expression (44), although this probably requires cell lineeCspecific cofactors (45).

    We performed microarray analysis to characterize the response to Pdx1 repression. A number of upregulated genes were involved in metabolism, consistent with studies (37,46) in which defects in mitochondrial metabolism have been linked to diminished expression of Pdx1. Several of the most differentially upregulated genes (Reg2, Reg3, Reg3, and Reg3) were members of the regenerating islet-derived gene family originally identified in models of -cell regeneration (47eC49). In addition, several other upregulated genes, including Numb and Tob, are likely to be involved in islet cell development. Numb is a modulator of the notch signaling pathway involved in the maintenance of embryonic neural progenitor cells (32). It is also expressed in early pancreatic progenitors and mature -cells and has been implicated in the differentiation of -cells from duct cells (33). The epidermal growth factor receptor ErbB2 is expressed in the developing pancreas and is upregulated in a model of duct-cell proliferation and islet neogenesis (50). Tob is an ErbB2 interacting protein implicated in the regulation of cell proliferation and differentiation (51). We also identified a novel highly conserved gene, 1810015C04Rik, which is expressed in the developing pancreas and mature - but not -cells. Further investigation is required to determine the role of 1810015C04Rik and other novel genes identified in this screen in -cell viability and regeneration. In addition, it will also be important to determine whether these changes in gene expression are due to Pdx1 repression per se or are secondary to impaired -cell function and hyperglycemia. Nevertheless, the initiation of a regenerative program by Pdx1 repression is a novel observation and a basis for further understanding -cell neogenesis in the adult pancreas.

    The adult pancreas has the capacity to increase its -cell mass during times of increased physiological need or in response to injury. However, the nature of putative -cell progenitors is a subject of ongoing debate. Numerous studies (9eC11) support the existence of duct-associated progenitors in models of pancreatic injury, whereas a more recent lineage-tracing study (13) indicates that -cell progenitors derive from -cells themselves in islets. These findings are not mutually exclusive and may reflect different means to an end, depending on circumstances. Our model of reversible Pdx1 repression reveals a potential duct-associated progenitor cell population. First, during Pdx1 repression, we observed focal regions of duct proliferation, recognized as a hallmark of pancreas regeneration (10,31). Second, we observed an increase in cell replication, proportional to the duration of Pdx1 repression, primarily in the exocrine pancreas but also associated with duct-like structures as well as in islets. Third, we found that genes known to be associated with -cell regeneration were upregulated during Pdx1 repression. Finally, after reversal of Pdx1 repression we observed cells that were Pdx1+/insulin+ or Pdx1+(low)/InseC closely associated with duct epithelium.

    In rodent models of pancreas injury (20,23,24), Pdx1 expression has been associated with -cell neogenesis. The proliferation we observed in response to repression of Pdx1 could be secondary to hyperglycemia rather than the loss of Pdx1 itself, as observed in other models of induced hyperglycemia (31,52). However, our findings show that proliferation of progenitors occurs in the absence of Pdx1 and that Pdx1 is required for their differentiation potentially into -cells. Gu et al. (53) found that Pdx1 was rarely expressed in the ductal cells of the adult pancreas and were likely to be present only in differentiating cells in the process of delaminating from the ducts. In a separate study (R.J.M., unpublished data), it was shown that Pdx1 is not required for the continued proliferation of duct cells in the developing pancreas but is absolutely required for the differentiation of duct-like cells of the embryonic epithelium into endocrine and exocrine lineages. As suggested by Waguri et al. (20), -cells could arise from both intraislet and ductal progenitors, depending on the circumstance. During periods of acute hyperglycemia, it may be sufficient for -cells to enter the cell cycle and thereby increase the -cell mass. However, following pancreatic injury or chronic hyperglycemia, a more primitive regenerative program may recruit duct-associated -cell progenitors. Possibly consistent with evidence from lineage tracing (13), we also observed occasional islet cells labeled with BrdU in Pdx1-repressed mice but were unable to determine to what extent these cells contribute to -cell regeneration. It is worth noting that in Pdx1-repressed mice, filtrin staining revealed the presence of -cells, but the islet area was decreased, suggesting that Pdx1 may be required for the maintenance of intraislet progenitors. An important caveat, however, is that we have not been able to discriminate between restoration of -cell function in pre-existing -cells and -cell neogenesis.

    Combining the conditional Pdx1 expression model with lineage tracing or cell-specific ablation models could further elucidate the origins of -cell progenitors and contribute to the goal of renewing -cells for the treatment of type 1 diabetes.

    ACKNOWLEDGMENTS

    This work was supported by a Juvenile Diabetes Research Foundation (JDRF) Postdoctoral Fellowship (to A.M.H.) and by a partnership Program Grant from the JDRF and the National Health and Medical Research Foundation of Australia (to L.C.H.). R.J.M. was supported by National Institutes of Health Grant DK55266.

    The authors thank Ken Simpson for technical assistance with microarray analysis and Fang-Xu Jiang for helpful discussions.

    DAPI, 4',6'-diamidine-2'-phenylindole dihydrochloride

    REFERENCES

    Jonsson J, Carlsson L, Edlund T, Edlund H: Insulin-promoter-factor 1 is required for pancreas development in mice. Nature371 :606 eC609,1994

    Ahlgren U, Jonsson J, Edlund H: The morphogenesis of the pancreatic mesenchyme is uncoupled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice. Development122 :1409 eC1416,1996

    Offield MF, Jetton TL, Labosky PA, Ray M, Stein RW, Magnuson MA, Hogan BL, Wright CV: PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development122 :983 eC995,1996

    Guz Y, Montminy MR, Stein R, Leonard J, Gamer LW, Wright CVE, Teitelman G: Expression of murine STF-1, a putative insulin gene transcription factor, in beta cells of pancreas, duodenal epthelium and pancreatic exocrine and endocrine progenitors during ontogeny. Development121 :11 eC18,1995

    Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund H: Beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the beta-cell phenotype and maturity onset diabetes. Genes Dev12 :1763 eC1768,1998

    Lottmann H, Vanselow J, Hessabi B, Walther R: The Tet-On system in transgenic mice: inhibition of the mouse pdx-1 gene activity by antisense RNA expression in pancreatic beta-cells. J Mol Med79 :321 eC328,2001

    Thomas MK, Devon ON, Lee JH, Peter A, Schlosser DA, Tenser MS, Habener JF: Development of diabetes mellitus in aging transgenic mice following suppression of pancreatic homeoprotein IDX-1. J Clin Invest108 :319 eC329,2001

    Parsons JA, Bartke A, Sorenson RL: Number and size of islets of Langerhans in pregnant, human growth hormone-expressing transgenic, and pituitary dwarf mice: effect of lactogenic hormones. Endocrinology136 :2013 eC2021,1995

    Rosenberg L: In vivo cell transformation: neogenesis of beta cells from pancreatic ductal cells. Cell Transplant4 :371 eC383,1995

    Bonner-Weir S, Baxter LA, Schuppin GT, Smith FE: A second pathway for regeneration of adult exocrine and endocrine pancreas: a possible recapitulation of embryonic development. Diabetes42 :1715 eC1720,1993

    Rosenberg L: Induction of islet cell neogenesis in the adult pancreas: the partial duct obstruction model. Microsc Res Tech43 :337 eC346,1998

    Gu D, Sarvetnick N: Epithelial cell proliferation and islet neogenesis in IFN-g transgenic mice. Development118 :33 eC46,1993

    Dor Y, Brown J, Martinez OI, Melton DA: Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature429 :41 eC46,2004

    Guz Y, Nasir I, Teitelman G: Regeneration of pancreatic beta cells from intra-islet precursor cells in an experimental model of diabetes. Endocrinology142 :4956 eC4968,2001

    Rooman I, Heremans Y, Heimberg H, Bouwens L: Modulation of rat pancreatic acinoductal transdifferentiation and expression of PDX-1 in vitro. Diabetologia43 :907 eC914,2000

    Rooman I, Lardon J, Bouwens L: Gastrin stimulates beta-cell neogenesis and increases islet mass from transdifferentiated but not from normal exocrine pancreas tissue. Diabetes51 :686 eC690,2002

    Iovanna JL: Redifferentiation and apoptosis of pancreatic cells during acute pancreatitis. Int J Pancreatol20 :77 eC84,1996

    Bockman DE: Morphology of the exocrine pancreas related to pancreatitis. Microsc Res Tech37 :509 eC519,1997

    Hall PA, Lemoine NR: Rapid acinar to ductal transdifferentiation in cultured human exocrine pancreas. J Pathol166 :97 eC103,1992

    Waguri M, Yamamoto K, Miyagawa JI, Tochino Y, Yamamori K, Kajimoto Y, Nakajima H, Watada H, Yoshiuchi I, Itoh N, Imagawa A, Namba M, Kuwajima M, Yamasaki Y, Hanafusa T, Matsuzawa Y: Demonstration of two different processes of -cell regeneration in a new diabetic mouse model induced by selective perfusion of alloxan. Diabetes46 :1281 eC1290,1997

    Bonner-Weir S, Toschi E, Inada A, Reitz P, Fonseca SY, Aye T, Sharma A: The pancreatic ductal epithelium serves as a potential pool of progenitor cells. Pediatr Diabetes5 (Suppl. 2) :16 eC22,2004

    Holland AM, Gonez LJ, Harrison LC: Progenitor cells in the adult pancreas. Diabetes Metab Res Rev20 :13 eC27,2004

    Kritzik MR, Jones E, Chen Z, Krakowski M, Krahl T, Good A, Wright C, Fox H, Sarvetnick N: PDX-1 and Msx-2 expression in the regenerating and developing pancreas. J Endocrinol163 :523 eC530,1999

    Sharma A, Zangen DH, Reitz P, Taneja M, Lissauer ME, Miller CP, Weir GC, Habener JF, Bonner-Weir S: The homeodomain protein IDX-1 increases after an early burst of proliferation during pancreatic regeneration. Diabetes48 :507 eC513,1999

    Holland AM, Hale MA, Kagami H, Hammer RE, MacDonald RJ: Experimental control of pancreatic development and maintenance. Proc Natl Acad Sci U S A99 :12236 eC12241,2002

    Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP: Summaries of Affymetrix GeneChip probe level data. Nucleic Acid Res31 :e15 ,2003

    Gautier L, Cope L, Bolstad BM, Irizarry RA: affy: analysis of Affymetrix GeneChip data at the probe level. Bioinformatics20 :307 eC315,2004

    Sun C, Kilburn D, Lukashin A, Crowell T, Gardner H, Brundiers R, Diefenbach B, Carulli JP: Kirrel2, a novel immunoglobulin superfamily gene expressed primarily in beta cells of the pancreatic islets. Genomics82 :130 eC142,2003

    Ihalmo P, Palmeen T, Ahola H, Valtonen E, Holthfer H: Filtrin is a novel member of nephrin-like proteins. Biochem Biophys Res Comm300 :364 eC370,2003

    Palmeen T, Ahola H, Palgi J, Aaltonen P, Luimila P, Wang S, Jaakkola I, Knip M, Otonkoski T: Nephrin is expressed in the pancreatic beta cells. Diabetologia44 :1274 eC1280,2001

    Lipsett M, Finegood DT: -Cell neogenesis during prolonged hyperglycemia in rats. Diabetes51 :1834 eC1841,2002

    Petersen PH, Zou K, Hwang JK, Jan YN, Zhong W: Progenitor cell maintenance requires numb and numblike during mouse neurogenesis. Nature419 :929 eC934,2002

    Yoshida T, Tokunaga A, Nakao K, Okano H: Distinct expression patterns of splicing isoforms of mNumb in the endocrine lineage of developing pancreas. Differentiation71 :486 eC495,2003

    Dutta S, Bonner-Weir S, Montminy M, Wright C: Regulatory factor linked to late-onset diabetes (Letter) Nature392 :560 ,1998

    Stoffers DA, Ferrer J, Clarke WL, Habener JF: Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat Genet17 :138 eC139,1997

    Pugliese A, Miceli D: The insulin gene in diabetes. Diabetes Metab Res Rev18 :13 eC25,2002

    Brissova M, Shiota M, Nicholson WE, Gannon M, Knobel SM, Piston DW, Wright CV, Powers AC: Reduction in pancreatic transcription factor PDX-1 impairs glucose-stimulated insulin secretion. J Biol Chem277 :11225 eC11232,2002

    Ohlsson H, Karlsson K, Edlund T: IPF1, a homeodomain-containing transactivator of the insulin gene. EMBO J12 :4251 eC4259,1993

    Waeber G, Thompson N, Nicod P, Bonny C: Transcriptional activation of the GLUT2 gene by the IPF-1/STF-1/IDX-1 homeobox factor. Mol Endocrinol10 :1327 eC1334,1996

    Watada H, Kajimoto Y, Umayahara Y, Matsuoka T, Kaneto H, Fujitani Y, Kamada T, Kawamori R, Yamasaki Y: The human glucokinase gene -celleCtype promoter: an essential role of insulin promoter factor 1/PDX-1 in it’s activation in HIT-T15 cells. Diabetes45 :1478 eC1488,1996

    Johnson JD, Ahmed NT, Luciani DS, Han Z, Tran H, Fujita J, Misler S, Edlund H, Polonsky KS: Increased islet apoptosis in Pdx1+/eC mice. J Clin Invest111 :1147 eC1160,2003

    Wang H, Maechler P, Ritz-Laser B, Hagenfeldt KA, Ishihara H, Philippe J, Wollheim CB: Pdx1 level defines pancreatic gene expression pattern and cell lineage differentiation. J Biol Chem276 :25279 eC25286,2001

    Chakrabarti SK, James JC, Mirmira RG: Quantitative assessment of gene targeting in vitro and in vivo by the pancreatic transcription factor, Pdx1: importance of chromatin structure in directing promoter binding. J Biol Chem277 :13286 eC13293,2002

    Ritz-Laser B, Gauthier BR, Estreicher A, Mamin A, Brun T, Ris F, Salmon P, Halban PA, Trono D, Philippe J: Ectopic expression of the beta-cell specific transcription factor Pdx1 inhibits glucagon gene transcription. Diabetologia46 :810 eC821,2003

    Ritz-Laser B, Estreicher A, Klages N, Saule S, Philippe J: Pax-6 and Cdx-2/3 interact to activate glucagon gene expression on the G1 control element. J Biol Chem274 :4124 eC4132,1999

    Gauthier BR, Brun T, Sarret EJ, Ishihara H, Schaad O, Descombes P, Wollheim CB: Oligonucleotide microarray analysis reveals PDX1 as an essential regulator of mitochondrial metabolism in rat islets. J Biol Chem279 :31121 eC31130,2004

    Terazono K, Yamamoto H, Takasawa S, Shiga K, Yonemura Y, Tochino Y, Okamoto H: A novel gene activated in regenerating islets. J Biol Chem263 :2111 eC2114,1988

    Unno M, Itoh T, Watanabe T, Miyashita H, Moriizumi S, Teraoka H, Yonekura H, Okamoto H: Islet beta-cell regeneration and reg genes. Adv Exp Med Biol321 :61 eC66,1992

    Narushima Y, Unno M, Nakagawara K, Mori M, Miyashita H, Suzuki Y, Noguchi N, Takasawa S, Kumagai T, Yonekura H, Okamoto H: Structure, chromosomal localization and expression of mouse genes encoding type III Reg, RegIII alpha, RegIII beta, RegIII gamma. Gene185 :159 eC168,1997

    Kritzik MR, Krahl T, Good A, Gu D, Lai C, Fox H, Sarvetnick N: Expression of ErbB receptors during pancreatic islet development and regrowth. J Endocrinol165 :67 eC77,2000

    Yoshida Y, Nakamura T, Komoda M, Satoh H, Suzuki T, Tsuzuku JK, Miyasaka T, Yoshida EH, Umemori H, Kunisaki RK, Tani K, Ishii S, Mori S, Suganuma M, Noda T, Yamamoto T: Mice lacking a transcriptional corepressor Tob are predisposed to cancer. Genes Dev17 :1201 eC1206,2003

    Kojima H, Fujimiya M, Matsumura K, Nakahara T, Hara M, Chan L: Extrapancreatic insulin-producing cells in multiple organs in diabetes. Proc Natl Acad Sci U S A101 :2458 eC2463,2004

    Gu G, Dubauskaite J, Melton DA: Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development129 :2447 eC2457,2002(Andrew M. Holland, L. Jor)