Neurogenin3 Activates the Islet Differentiation Program while Repressing Its Own Expression (original) (raw)

Journal Article

,

1Diabetes Center (S.B.S., H.W., M.S.G.), San Francisco, California 94143-0534

Search for other works by this author on:

,

1Diabetes Center (S.B.S., H.W., M.S.G.), San Francisco, California 94143-0534

Search for other works by this author on:

2Hormone Research Institute and Department of Medicine (M.S.G.), University of California San Francisco, San Francisco, California 94143-0534

*Address all correspondence and requests for reprints to: Michael S. German, Hormone Research Institute, University of California San Francisco, 513 Parnassus Avenue, San Francisco, California 94143-0534.

Search for other works by this author on:

Received:

04 February 2003

Accepted:

29 September 2003

Published:

01 January 2004

Cite

Stuart B. Smith, Hirotaka Watada, Michael S. German, Neurogenin3 Activates the Islet Differentiation Program while Repressing Its Own Expression, Molecular Endocrinology, Volume 18, Issue 1, 1 January 2004, Pages 142–149, https://doi.org/10.1210/me.2003-0037
Close

Navbar Search Filter Mobile Enter search term Search

Abstract

Expression of the proendocrine factor Neurogenin3 determines which progenitor cells in the developing pancreas will differentiate into the endocrine cells of the islets of Langerhans. To better understand how Neurogenin3 directs endocrine differentiation, we examined the mechanisms by which Neurogenin3 regulates the promoters of three transcription factor genes expressed in endocrine precursor cells: the nkx2.2 gene, the PAX4 gene, and the NEUROG3 gene, the human gene encoding Neurogenin3 itself. The function of all three promoters depends on at least one critical E box, a common DNA sequence that forms a binding site for basic helix-loop-helix proteins like Neurogenin3. Neurogenin3 bound to and effectively activated transcription through the nkx2.2 and PAX4 E boxes. In contrast, Neurogenin3 strongly repressed the NEUROG3 promoter, although a proximal E box was required for activity in the absence of Neurogenin3, suggesting that a ubiquitous transcriptional activator may bind to this site, and that Neurogenin3 could act as a competitive inhibitor of this activator. This hypothesis was supported by the lack of evidence for significant intrinsic transcriptional repression capacity in the Neurogenin3 protein, and by the ability of isolated DNA-binding basic helix-loop-helix domains to repress the NEUROG3 promoter. Neurogenin3 produced additional repression, however, when the protein included an intact transcriptional activation domain, suggesting that it may also induce the expression of a downstream transcriptional repressor. In summary, while Neurogenin3 orchestrates islet cell differentiation by activating islet cell transcription factor genes, it simultaneously represses its own promoter.

AS THE PANCREAS develops from a cluster of multipotent epithelial progenitor cells into the distinct populations of mature cells that form the adult pancreas, a diverse group of transcription factors orchestrate the determination of cell-type fates and the progression of these cells through specific lineages (for review see Ref. 1). Key among these factors is the basic helix-loop-helix (bHLH) factor Neurogenin3. The expression of Neurogenin3 is both necessary and sufficient (24) to drive undifferentiated progenitor cells to an endocrine fate, but only initiates the islet differentiation program because it is extinguished before final differentiation of the cells (3, 5). Additional factors, such as the bHLH factor neuroD1 (6) and the homeodomain factors Pax4 (7), Pax6 (8, 9), nkx2.2 (10), nkx6.1 (11), isl1 (12), and PDX1 (1315) are also necessary for this process of differentiation and for maintenance of the mature, differentiated cells.

Subsequent to the identification of Neurogenin3 as a pancreatic proendocrine gene, establishing the factors that lie directly upstream and downstream has stimulated keen interest. Studies of the human NEUROG3 promoter have demonstrated that fragments of the promoter down to the proximal few hundred base pairs are highly active in transformed cell lines irrespective of their tissue of origin, demonstrating the presence of universal activators working through the proximal promoter. In contrast, longer promoter constructs can direct expression of a transgene specifically to progenitor cells in the pancreas, gut, and neural tissues (16). Pancreatic transcriptional activators of the HNF6 (17), HNF1 and HNF3/FoxA (16) families all bind to this longer NEUROG3 promoter. In pancreatic cells not fated to become islet cells, signals through the notch receptor activate the expression of the transcriptional repressor HES-1, which binds to and prevents the activation of the NEUROG3 promoter (4, 16, 18). The mechanisms by which the expression of Neurogenin3 is extinguished in progenitor cells before final differentiation are unknown. In addition, the downstream genes through which Neurogenin3 activates the islet cell differentiation program are largely unknown. Proposed targets for Neurogenin3 include the genes encoding the islet transcription factors NeuroD1 (19), Pax4 (20, 21), and Nkx2.2 (22). Neurogenin3 has been shown to cooperate with HNF1 homeodomain factors in activating both the PAX4 promoter and the chromosomal pax4 gene (21), and with FoxA winged-helix factors in activating the nkx2.2 1A promoter (22), but otherwise little is known about how Neurogenin3 may activate these genes.

In the current study, we set out to understand more clearly how Neurogenin3 expression is controlled, and to understand the function of Neurogenin3 on a molecular level. We found that Neurogenin3 can function as a potent transcriptional activator when bound to E elements from the PAX4 and nkx2.2 gene promoters; and one hybrid analysis mapped this activation capacity to the carboxyl terminus of the Neurogenin3 protein but did not detect significant intrinsic transcriptional repression capacity. The proximal NEUROG3 promoter itself contains a critical E box; but surprisingly, exogenously expressed bHLH activating factors including Neurogenin3 itself repress the activity of the NEUROG3 promoter. We propose that Neurogenin3 can act as a transcriptional repressor of its own promoter by competing with a ubiquitous transcriptional activator, possibly in combination with the induction of a transcriptional repressor.

RESULTS

A previous study demonstrated by transient transfection assay that NEUROG3 promoter constructs as short as 520 bp are active in a variety of cell lines including nonpancreatic lines (16). To identify important promoter elements within this proximal promoter, we constructed additional 5′ promoter deletions driving expression of a luciferase reporter gene. A promoter containing only 207 bp upstream of the transcription start site still maintained a significant level of activity. This promoter contains a potentially important E box (a sequence element with the consensus CANNTG that binds to transcription factors in the bHLH family, including Neurogenin3 itself) located at −149 bp that is also conserved in the mouse promoter. To test the importance of the −149-bp E box, we introduced a 2-bp mutation in essential bases of the E box consensus and found that the mutation abolished the activity of the proximal promoter in all cell lines tested (Fig. 1).

NEUROG3 Promoter Activity Is Dependent upon an E Box Located at −149 bp The three cell lines shown were transfected with reporter plasmids containing the firefly luciferase gene under the control of the either the wild-type −207-bp NEUROG3 promoter, or the −207-bp NEUROG3 promoter containing a 2-bp mutation in the E box (−207-bp ME). Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the promoterless backbone vector (pFOXluc1). Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Fig. 1.

NEUROG3 Promoter Activity Is Dependent upon an E Box Located at −149 bp The three cell lines shown were transfected with reporter plasmids containing the firefly luciferase gene under the control of the either the wild-type −207-bp NEUROG3 promoter, or the −207-bp NEUROG3 promoter containing a 2-bp mutation in the E box (−207-bp ME). Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the promoterless backbone vector (pFOXluc1). Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Given the importance of this conserved E box within the proximal promoter, we tested whether Neurogenin3, alone or in conjunction with its heterodimeric partner E47, could influence the activity of its own promoter (Fig. 2). Surprisingly, Neurogenin3 and E47 repressed the NEUROG3 promoter constructs, but not the control RSV promoter or promoterless constructs. The promoter construct with the mutant E box was not repressed by Neurogenin3 (data not shown), but this promoter was already inactive in the absence of Neurogenin3. In addition, two copies of a short minienhancer containing the proximal NEUROG3 E box also was active in both βTC3 and NIH 3T3 cells, dependent on the E box sequence, and repressed by Neurogenin3 (Fig. 3 and data not shown).

Neurogenin3 Represses Its Own Promoter NIH3T3 cells were transfected with reporter plasmids containing the firefly luciferase gene either with no promoter or under the control of the various length NEUROG3 promoters indicated or the Rous sarcoma virus LTR (RSV). Cells were cotransfected with expression plasmids containing the cytomegalovirus (CMV) early gene promoter driving the expression of either no cDNA or the cDNAs for Neurogenin3 and its heterodimeric partner E47. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the promoterless backbone vector (pFOXluc1) cotransfected with the expression plasmid containing no cDNA. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Fig. 2.

Neurogenin3 Represses Its Own Promoter NIH3T3 cells were transfected with reporter plasmids containing the firefly luciferase gene either with no promoter or under the control of the various length NEUROG3 promoters indicated or the Rous sarcoma virus LTR (RSV). Cells were cotransfected with expression plasmids containing the cytomegalovirus (CMV) early gene promoter driving the expression of either no cDNA or the cDNAs for Neurogenin3 and its heterodimeric partner E47. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the promoterless backbone vector (pFOXluc1) cotransfected with the expression plasmid containing no cDNA. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

E Box Activity Is Conferred upon a Heterologous Promoter βTC3 cells were transfected with reporter plasmids containing the firefly luciferase gene under the control of the herpes simplex virus thymidine kinase minimal promoter either by itself (TK) or linked to two copies of the NSE minienhancer which contains the sequences from −105 to −158 bp from the NEUROG3 promoter including the proximal E box or, in panel A, two copies of the N3mE minienhancer with a 2-bp mutation of the E box. In panel B, cells were cotransfected with either a control plasmid expressing no cDNA or two plasmids expressing the E47 and Neurogenin3 cDNAs under the control of the cytomegalovirus (CMV) early gene promoter. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity in cells transfected with the vector with the isolated TK promoter (TK). Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Fig. 3.

E Box Activity Is Conferred upon a Heterologous Promoter βTC3 cells were transfected with reporter plasmids containing the firefly luciferase gene under the control of the herpes simplex virus thymidine kinase minimal promoter either by itself (TK) or linked to two copies of the NSE minienhancer which contains the sequences from −105 to −158 bp from the NEUROG3 promoter including the proximal E box or, in panel A, two copies of the N3mE minienhancer with a 2-bp mutation of the E box. In panel B, cells were cotransfected with either a control plasmid expressing no cDNA or two plasmids expressing the E47 and Neurogenin3 cDNAs under the control of the cytomegalovirus (CMV) early gene promoter. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity in cells transfected with the vector with the isolated TK promoter (TK). Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Next, we tested the affinity of the pancreatic bHLH proteins Neurogenin3, neuroD1, and E47 for the E box of the NEUROG3 promoter. EMSAs were performed using isolated E boxes from the NEUROG3, nkx2.2, PAX4, and rat insulin I gene promoters as probes in conjunction with in vitro produced proteins Neurogenin3, NeuroD1, and their dimeric partner E47 (Fig. 4). E47 bound to each of the elements with comparable affinity; and, unexpectedly for class B bHLH factors, homodimers of neuroD1 and Neurogenin3 bound to the E box of the nkx2.2 promoter. Heterodimers of E47 with Neurogenin3 or neuroD1 bound to all four probes tested, with differing relative binding affinities in the order from the NEUROG3 E box (weakest), to the nkx2.2 E3 element, the rat Insulin I E2 element, and the PAX4 E box (strongest).

bHLH Factors Bind Pancreatic Promoter E Boxes EMSA were used to test the ability of E47, Neurogenin3, and neuroD1 to bind to labeled, double-stranded oligonucleotides containing the E box sequences from the rat insulin I and mouse nkx2.2 promoters (A) or the human PAX4 and NEUROG3 promoters (B). One microliter of each in vitro-translated protein was incubated with the indicated probes, either individually or in the combination shown. Results are typical of experiments done on three occasions.

Fig. 4.

bHLH Factors Bind Pancreatic Promoter E Boxes EMSA were used to test the ability of E47, Neurogenin3, and neuroD1 to bind to labeled, double-stranded oligonucleotides containing the E box sequences from the rat insulin I and mouse nkx2.2 promoters (A) or the human PAX4 and NEUROG3 promoters (B). One microliter of each _in vitro_-translated protein was incubated with the indicated probes, either individually or in the combination shown. Results are typical of experiments done on three occasions.

To test the function of the different E boxes from the four genes in isolation, six copies of each E box were ligated upstream of the minimal herpes simplex virus (HSV)-thymidine kinase (TK) promoter driving the luciferase reporter gene. The activity of the resulting heterologous promoter was determined in the absence and presence of cotransfected factors (Fig. 5). Unlike the other three constructs, the NEUROG3 E box construct was not activated by any of the transcription factor combinations. It should be noted that in contrast to this minienhancer construct made from six copies of a 16-bp E box sequence, the larger, 54-bp NEUROG3 E box minienhancer used in Fig. 3 was active, and was repressed by Neurogenin3. The higher activity of the larger minienhancer suggests that the ubiquitous activator that requires the NEUROG3 E box either requires a larger binding site, or interacts with factor(s) binding adjacent to the E box.

The NEUROG3 E Box Is Not Activated by the Proendocrine bHLH Factors NIH3T3 cells were transfected with reporter plasmids containing the firefly luciferase gene under the control of the herpes simplex virus thymidine kinase minimal promoter either by itself (TK) or linked to six copies of the indicated E boxes. Cells were cotransfected with expression plasmids containing the cytomegalovirus (CMV) early gene promoter driving the expression of either no cDNA, the E47 cDNA or the cDNA combinations shown. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the vector with the isolated TK promoter (TK) cotransfected with the expression plasmid containing no cDNA. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Fig. 5.

The NEUROG3 E Box Is Not Activated by the Proendocrine bHLH Factors NIH3T3 cells were transfected with reporter plasmids containing the firefly luciferase gene under the control of the herpes simplex virus thymidine kinase minimal promoter either by itself (TK) or linked to six copies of the indicated E boxes. Cells were cotransfected with expression plasmids containing the cytomegalovirus (CMV) early gene promoter driving the expression of either no cDNA, the E47 cDNA or the cDNA combinations shown. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the vector with the isolated TK promoter (TK) cotransfected with the expression plasmid containing no cDNA. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

To determine whether transcriptional repression is an inherent property of Neurogenin3 and whether Neurogenin3 thereby could repress its own promoter by a direct mechanism, one hybrid analysis was used to study the transactivating properties of the Neurogenin3 protein (Fig. 6). Various regions of the protein were fused to the DNA binding domain of GAL4, and the ability of the resulting protein to affect transcription from a promoter containing five GAL4 binding sites was determined. Several regions of the Neurogenin3 protein were able to stimulate transcription, the most potent activation coming from regions encompassing the carboxyl terminus of the protein (constructs 128–214 and 190–214), which was a more potent effect than that exhibited by the pax6 activation domain that served as a positive control. Only one region of the protein, the isolated bHLH domain, was able to weakly repress transcription. A similar construct containing the equivalent region of the neuroD1 protein acted in a comparable manner, suggesting that this weak repression is likely to be a common characteristic of an isolated bHLH domain and not an idiosyncrasy of Neurogenin3.

One Hybrid Analysis Maps Activation Domains within Neurogenin3 In panel A, a low background reporter construct comprised of five copies of the Gal4 consensus binding site (UAS) ligated upstream of the E1b viral promoter driving luciferase expression was transfected into NIH3T3 cells. The reporter construct was cotransfected with a plasmid expressing a fusion protein comprised of the Gal4 DNA binding domain and the indicated portion of Neurogenin3 protein; a similar construct containing the previously characterized pax6 activation domain was included as a positive control. In panel B, a high background reporter construct comprised of five copies of the Gal4 UAS ligated upstream of the HSV-TK promoter driving luciferase was transfected into NIH3T3 cells. The reporter construct was cotransfected with a plasmid expressing the GAL4 DNA binding domain fused to the bHLH domain of Neurogenin3 or NeuroD1/β2 or to the pax6 activation domain. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the luciferase vector cotransfected with the expression plasmid containing the Gal4 DNA binding domain alone. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions. In panel C, a diagram outlines the functional domains of the Neurogenin3 protein.

Fig. 6.

One Hybrid Analysis Maps Activation Domains within Neurogenin3 In panel A, a low background reporter construct comprised of five copies of the Gal4 consensus binding site (UAS) ligated upstream of the E1b viral promoter driving luciferase expression was transfected into NIH3T3 cells. The reporter construct was cotransfected with a plasmid expressing a fusion protein comprised of the Gal4 DNA binding domain and the indicated portion of Neurogenin3 protein; a similar construct containing the previously characterized pax6 activation domain was included as a positive control. In panel B, a high background reporter construct comprised of five copies of the Gal4 UAS ligated upstream of the HSV-TK promoter driving luciferase was transfected into NIH3T3 cells. The reporter construct was cotransfected with a plasmid expressing the GAL4 DNA binding domain fused to the bHLH domain of Neurogenin3 or NeuroD1/β2 or to the pax6 activation domain. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the luciferase vector cotransfected with the expression plasmid containing the Gal4 DNA binding domain alone. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions. In panel C, a diagram outlines the functional domains of the Neurogenin3 protein.

Due to the apparent inability of Neurogenin3 to act directly as a transcriptional repressor, we hypothesized that Neurogenin3 may repress its own promoter by competing for binding with another activator or by inducing the expression of a transcriptional repressor that in turn represses the promoter. If the mechanism was purely the first model, then any factor able to bind the E box should be able to compete and thereby repress in a similar manner. The second model would necessitate that the protein contain a transcriptional activation domain to induce expression of a downstream repressor. To test these two possibilities, we cotransfected the −325-bp NEUROG3 promoter reporter construct with plasmids expressing various E box-binding bHLH proteins. We used the −325-bp NEUROG3 promoter because longer promoters contain additional E boxes.

All combinations repressed the NEUROG3 promoter, with the single exception of E47 in combination with the muscle bHLH protein myoD (Fig. 7). Consistent with the competition model for transcriptional repression of the NEUROG3 promoter, a truncated version of E47 [E47(Δ1–598)] containing only the DNA-binding bHLH domain and lacking any activation domain repressed the promoter as efficiently as the wild-type protein, thus suggesting that E47 repression is not due to the activation of an additional gene, but may simply result from competition with an activator for binding to the E box. In contrast, the isolated Neurogenin3 bHLH domain, which should not bind by itself to the NEUROG3 E box (Fig. 4), did not repress the NEUROG3 promoter. Consistent with the downstream repressor model, however, inclusion of the activation domain allowed Neurogenin3 to repress the NEUROG3 promoter. Because Neurogenin3 probably normally exits as a heterodimer with E47 or other ubiquitous class A bHLH proteins, the heterodimer may both activate a downstream repressor and compete with an activator. Consistent with this possibility, the greatest repression of the NEUROG3 promoter was produced by the combination of E47 and Neurogenin3.

Pancreatic bHLH Factors Repress the NEUROG3 Promoter NIH3T3 cells were transfected with a reporter plasmids containing the firefly luciferase gene under the control of the −325-bp NEUROG3 promoter. Cells were cotransfected with expression plasmids containing the cytomegalovirus (CMV) early gene promoter driving the expression of either no cDNA or the wild-type or truncated bHLH cDNAs indicated. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the luciferase vector cotransfected with the expression plasmid containing no cDNA. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

Fig. 7.

Pancreatic bHLH Factors Repress the NEUROG3 Promoter NIH3T3 cells were transfected with a reporter plasmids containing the firefly luciferase gene under the control of the −325-bp NEUROG3 promoter. Cells were cotransfected with expression plasmids containing the cytomegalovirus (CMV) early gene promoter driving the expression of either no cDNA or the wild-type or truncated bHLH cDNAs indicated. Luciferase activities of all samples were determined 48 h after transfection and are expressed relative to the activity of the luciferase vector cotransfected with the expression plasmid containing no cDNA. Results are expressed as the mean ± sem of data from experiments performed in triplicate on at least three separate occasions.

DISCUSSION

Neurogenin3 activates a cascade of genes involved in islet cell differentiation but is itself inactivated before differentiation, so that it initiates but does not complete the differentiation program. The closely related bHLH gene neuroD1 is activated by Neurogenin3 (19) and persists in the mature islet cells where it plays a role in completing and maintaining the differentiated state (6) by driving the expression of such genes as insulin (23) and glucagon (24). To prevent persistent expression of Neurogenin3 once its task of initiating differentiation is achieved, some mechanism must limit the expression of Neurogenin3 in the differentiating cells. Our data suggest that autorepression by Neurogenin3 of its own expression contributes at least part of this mechanism.

Other genes also utilize autorepression to limit or modulate their own expression. The pancreatic transcription factors pax4 (20) and HES1 (25) bind to and directly inhibit their own promoters. Both pax4 and HES1 are themselves potent transcriptional repressors, and binding to their own promoters presumably results in direct transcriptional repression through the recruitment of corepressors and inhibition of the transcription complex. Because Neurogenin3 lacks a significant transcriptional repression domain, it must work by a different mechanism. Interestingly, the NEUROG3 promoter contains a critical E box that neither Neurogenin3 nor neuroD1 can activate. Instead, it appears that the pancreatic bHLH proteins may compete with some ubiquitous activator that binds to the E box, so that expression of Neurogenin3 and neuroD1 in the differentiating progenitor could displace that activator and inactivate the NEUROG3 gene.

The NEUROG3 E box is a perfect consensus binding site for the basic leucine zipper protein adaptor protein complex-4 (26) or a close match to the site of bHLH protein cMyc (27), both of which function as transcriptional activators. A role for cMyc as the ubiquitous activator is particularly appealing, as its expression is associated with cell division, as is seen in the Neurogenin3-expressing precursors, but not in the differentiated, neuroD1-expressing islet cells (3, 5).

In addition, however, Neurogenin3 may do more than simply displace an activator from the NEUROG3 promoter. In combination with E47, Neurogenin3 is a more potent repressor of the NEUROG3 promoter than the other bHLH proteins tested, and this additional repression depends on the transcriptional activation domain of the Neurogenin3 protein. These data suggest that Neurogenin3 may activate the transcription of some other gene that in turn inhibits the NEUROG3 promoter. The identity of this hypothetical downstream repressor is unknown, but members of the HES family of transcriptional repressors are possible candidates given the remarkable ability of HES1 to extinguish the NEUROG3 promoter (16). In studies in vitro, we have found that Neurogenin3 does not increase HES1 expression, but it does induce the expression of other members of the HES family (Gasa, R., C. Mrejen, and M. S. German, unpublished data). Furthermore, it must be considered that the intact NEUROG3 gene, which certainly contains important regulatory domains outside the limited promoter constructs used in these studies, may have additional mechanisms for silencing.

Finally, it must be noted that the NEUROG3 gene remains off after Neurogenin3 disappears from mature endocrine cells. Our data do not directly address how the NEUROG3 gene stays off because the mechanisms that silence an active gene may be quite different from those that maintain gene silencing. Neurogenin3 alters the expression of many genes, and these changes persist after Neurogenin3 disappears. For example, the nkx2.2 and neuroD1 genes, both direct targets of Neurogenin3, remain active after Neurogenin3 is gone. NeuroD1, although not as potent a repressor of the NEUROG3 promoter as Neurogenin3 itself, may be sufficient to keep the promoter off. Similarly, a downstream repressor could also persist after Neurogenin3 is gone. In addition, the gene expression changes initiated by Neurogenin3 could include the permanent loss of activators of the NEUROG3 promoter such as HNF6 (17).

In summary, the proendocrine factor Neurogenin3 functions as a transcriptional activator, initiating the cascade of gene expression events that leads to the differentiation of pluripotent progenitor cells into mature islet cells in the pancreas. Once this chain of events is initiated, however, Neurogenin3 represses its own expression, possibly by both competing with an activator and activating a repressor, allowing differentiation to proceed autonomously.

MATERIALS AND METHODS

EMSAs

Single-stranded oligonucleotides were 5′-end labeled using (γ-32P)ATP and T4 polynucleotide kinase. An excess of complementary strand was then annealed to form a duplex strand that was column purified. EMSA buffers and electrophoresis conditions were as described previously (28) using 500 ng of poly(deoxyinosine-deoxycytidine):poly(deoxyinosine-deoxycytidine) per 10 μl binding mix. For _in vitro_-produced protein, 1 μl of the 50-μl total reaction volume was used per binding mix.

Oligonucleotides used were as follows (coding strand shown from each double-stranded pair): NEUROG3 promoter E element, 5′-ctttgtccggaatccagctgtgccctgcgggggag-3′; rat insulin I promoter E2 element, 5′-ctgcttcatcaggccatctggccccttgttaataa-3′; PAX4 promoter E element, 5′-tgtataattgtgagcagatggcgggggctggcggc-3′; nkx2.2 promoter E3 element 5′-ttattaccgctgaacatatggccaatattttgact-3′. EMSA results are representative of those seen on at least three occasions.

In Vitro Protein Production

The cloning and construction of in vitro expression vectors containing the cDNAs encoding E47, Neurogenin3, and neuroD1 ligated downstream of the T7 phage promoter have been previously described (20). Proteins were produced using the TNT-coupled reticulocyte lysate system (Promega, Madison, WI) according to the manufacturer’s instructions to provide a total reaction volume of 50 μl from 1 μg of DNA template.

Luciferase Reporter Constructs

The longer NEUROG3 promoter luciferase constructs have been described previously (16). The shorter promoter fragments were generated by PCR and ligated upstream of the luciferase gene in the plasmid pFOXLuc1. A 2-bp mutation was introduced into the proximal NEUROG3 promoter E box in the intact −207-bp promoter by a PCR-based technique whereby two complementary primers corresponding to the region, and containing the mutation, were used as PCR primers to amplify the entire plasmid (29) using Pfu Turbo polymerase (Stratagene, La Jolla, CA). The positive strand primer sequence (mutation in bold type) was 5′-gccctttgtccggaatctggctgtgccctgcggggga-3′.

The minienhancers containing two copies of the proximal E box used in Fig. 3 were ligated upstream of the TK minimal promoter in the plasmid pFOXLuc1TK (30). Oligonucleotides used were as follows (coding strand shown from each double-stranded pair): N3E (−105 to −158 of NEUROG3 promoter) 5′-gatcttccggaatccagctgtgccctgcgggggaggagcgggctcgcgtggcgcggcccg-3′, N3mE 5′-gatcttccggaatctggctgtgccctgcgggggaggagcgggctcgcgtggcgcggcccg-3′. The minienhancers used in Fig. 5 contained six copies of a 16-bp repeat containing each E box, and were constructed by ligating two copies of the following oligonucleotides containing three copies of the respective E boxes (16 bp) upstream of the TK minimal promoter in pFOXLuc1TK: Pax4 5′-gatctgtgagcagatggcggggtgagcagatggcggggtgagcagatggcggg-3′; Nkx2.2 5′-gatctctgaacatatggccaactgaacatatggccaactgaacatatggccaag- 3′; Ngn3 5′-gatctgaatccagctgtgcccgaatccagctgtgcc cgaatccagctgtgcccg-3′.

One-Hybrid Analysis

One-hybrid expression vectors were constructed by amplifying the appropriate coding fragments of Neurogenin3 by PCR and then ligating into the _Eco_RI and _Bam_HI sites of the Gal4DBD vector (CLONTECH, Palo Alto, CA). Two reporter vectors were constructed carrying DNA binding sites for the GAL4 protein. The low background vector to test for activation was constructed from pFOXluc1 (31) with five copies of the GAL4 upstream activating sequence (UAS) ligated upstream of the adenovirus E1b promoter driving the expression of firefly luciferase. The high background vector used to test for repression was constructed from pFOXluc2 (30) with five copies of the GAL4 UAS ligated upstream of the HSV-TK promoter driving expression of firefly luciferase. Two micrograms of reporter construct and 200 ng of the GAL4DBD vector were transfected into one million cultured cells using Transfast lipid reagent (Promega) according to the manufacturer’s instructions and luciferase activity was determined 48 h after transfection using the Promega assay system according to the manufacturer’s instructions.

Cell Culture and Transfection

The mouse β-cell line, βTC3, and the mouse α-cell line, αTC1.6, were grown in DMEM supplemented with 2.5% fetal bovine serum and 15% horse serum. NIH3T3 mouse fibroblast cells were grown in DMEM supplemented with 10% fetal bovine serum. In preparation for transfection, cells were split into six-well plates 24 h before transfection, one million cells per well were used for αTC1.6 and βTC3 transfection and 50 thousand per well for NIH3T3 cells. Two micrograms of reporter construct were used per well, and 50 ng of any cotransfected transcription factor cDNA was used per well. Transfast (Promega) cationic lipid agent was used for all transfections according to the manufacturer’s instructions. Cells were harvested 48 h after transfection and luciferase assays performed with 5 μg of total protein as previously described (20). Transfections were performed on at least three occasions, all data are expressed as mean ± sem.

Acknowledgments

We thank members of German, Hebrok, and Vaisse laboratories for helpful comments.

This work was supported by a grant from the Nora Eccles Treadwell Foundation and NIH Grants DK553401 and DK21344. S.S. and H.W. are both recipients of Juvenile Diabetes Research Foundation International Postdoctoral Fellowships and Juvenile Diabetes Research Foundation International Advanced Postdoctoral Fellowships.

Present address for H.W.: Department of Medicine, Metabolism and Endocrinology, Juntendo University School of Medicine 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.

Abbreviations:

1

Wilson

ME

,

Scheel

D

,

German

MS

2003

Gene expression cascades in pancreatic development.

Mech Dev

120

:

65

80

2

Gradwohl

G

,

Dierich

A

,

LeMeur

M

,

Guillemot

F

2000

Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas.

Proc Natl Acad Sci USA

97

:

1607

1611

3

Schwitzgebel

VM

,

Scheel

DW

,

Conners

JR

,

Kalamaras

J

,

Lee

JE

,

Anderson

DJ

,

Sussel

L

,

Johnson

JD

,

German

MS

2000

Expression of neurogenin3 reveals an islet cell precursor population in the pancreas.

Development

127

:

3533

3542

4

Apelqvist

A

,

Li

H

,

Sommer

L

,

Beatus

P

,

Anderson

DJ

,

Honjo

T

, Hrabe de

Angelis

M

,

Lendahl

U

,

Edlund

H

1999

Notch signaling controls pancreatic cell differentiation.

Nature

400

:

877

881

5

Jensen

J

,

Heller

RS

,

Funder-Nielsen

T

,

Pedersen

EE

,

Lindsell

C

,

Weinmaster

G

,

Madsen

OD

,

Serup

P

2000

Independent development of pancreatic α- and β-cells from neurogenin3-expressing precursors: a role for the notch pathway in repression of premature differentiation.

Diabetes

49

:

163

176

6

Naya

FJ

,

Huang

HP

,

Qiu

Y

,

Mutoh

H

,

DeMayo

FJ

,

Leiter

AB

,

Tsai

MJ

1997

Diabetes, defective pancreatic morphogenesis, and abnormal enteroendocrine differentiation in BETA2/neuroD-deficient mice.

Genes Dev

11

:

2323

2334

7

Sosa-Pineda

B

,

Chowdhury

K

,

Torres

M

,

Oliver

G

,

Gruss

P

1997

The Pax4 gene is essential for differentiation of insulin-producing β cells in the mammalian pancreas.

Nature

386

:

399

402

8

Sander

M

,

Neubuser

A

,

Kalamaras

J

,

Ee

HC

,

Martin

GR

,

German

MS

1997

Genetic analysis reveals that PAX6 is required for normal transcription of pancreatic hormone genes and islet development.

Genes Dev

11

:

1662

1673

9

St-Onge

L

,

Sosa-Pineda

B

,

Chowdhury

K

,

Mansouri

A

,

Gruss

P

1997

Pax6 is required for differentiation of glucagon-producing α-cells in mouse pancreas.

Nature

387

:

406

409

10

Sussel

L

,

Kalamaras

J

,

Hartigan-O’Connor

DJ

,

Meneses

JJ

,

Pedersen

RA

,

Rubenstein

JL

,

German

MS

1998

Mice lacking the homeodomain transcription factor Nkx2.2 have diabetes due to arrested differentiation of pancreatic β cells.

Development

125

:

2213

2221

11

Sander

M

,

Sussel

L

,

Conners

J

,

Scheel

D

,

Kalamaras

J

, Dela

Cruz

F

,

Schwitzgebel

V

,

Hayes-Jordan

A

,

German

M

2000

Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of β-cell formation in the pancreas.

Development

127

:

5533

5540

12

Ahlgren

U

,

Pfaff

SL

,

Jessell

TM

,

Edlund

T

,

Edlund

H

1997

Independent requirement for ISL1 in formation of pancreatic mesenchyme and islet cells.

Nature

385

:

257

260

13

Ahlgren

U

,

Jonsson

J

,

Edlund

H

1996

The morphogenesis of the pancreatic mesenchyme is uncoupled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice.

Development

122

:

1409

1416

14

Offield

MF

,

Jetton

TL

,

Labosky

PA

,

Ray

M

,

Stein

RW

,

Magnuson

MA

,

Hogan

BL

,

Wright

CV

1996

PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum.

Development

122

:

983

995

15

Jonsson

J

,

Carlsson

L

,

Edlund

T

,

Edlund

H

1994

Insulin-promoter-factor 1 is required for pancreas development in mice.

Nature

371

:

606

609

16

Lee

JC

,

Smith

SB

,

Watada

H

,

Lin

J

,

Scheel

D

,

Wang

J

,

Mirmira

RG

,

German

MS

2001

Regulation of the pancreatic pro-endocrine gene neurogenin3.

Diabetes

50

:

928

936

17

Jacquemin

P

,

Durviaux

SM

,

Jensen

J

,

Godfraind

C

,

Gradwohl

G

,

Guillemot

F

,

Madsen

OD

,

Carmeliet

P

,

Dewerchin

M

,

Collen

D

,

Rousseau

GG

,

Lemaigre

FP

2000

Transcription factor hepatocyte nuclear factor 6 regulates pancreatic endocrine cell differentiation and controls expression of the proendocrine gene ngn3.

Mol Cell Biol

20

:

4445

4454

18

Jensen

J

,

Pedersen

EE

,

Galante

P

,

Hald

J

,

Heller

RS

,

Ishibashi

M

,

Kageyama

R

,

Guillemot

F

,

Serup

P

,

Madsen

OD

2000

Control of endodermal endocrine development by Hes-1.

Nat Genet

24

:

36

44

19

Huang

HP

,

Liu

M

,

El-Hodiri

HM

,

Chu

K

,

Jamrich

M

,

Tsai

MJ

2000

Regulation of the pancreatic islet-specific gene BETA2 (neuroD) by neurogenin 3.

Mol Cell Biol

20

:

3292

3307

20

Smith

SB

,

Watada

H

,

Scheel

DW

,

Mrejen

C

,

German

MS

2000

Autoregulation and maturity onset diabetes of the young transcription factors control the human PAX4 promoter.

J Biol Chem

275

:

36910

36919

21

Smith

SB

,

Gasa

R

,

Watada

H

,

Wang

J

,

Griffen

SC

,

German

MS

2003

Neurogenin3 and hepatic nuclear factor 1 cooperate in activating pancreatic expression of Pax4.

J Biol Chem

278

:

38254

38259

22

Watada

H

,

Scheel

DW

,

Leung

J

,

German

MS

2003

Distinct gene expression programs function in progenitor and mature islet cells.

J Biol Chem

278

:

17130

17140

23

Naya

FJ

,

Stellrecht

CM

,

Tsai

MJ

1995

Tissue-specific regulation of the insulin gene by a novel basic helix-loop-helix transcription factor.

Genes Dev

9

:

1009

1019

24

Dumonteil

E

,

Laser

B

,

Constant

I

,

Philippe

J

1998

Differential regulation of the glucagon and insulin I gene promoters by the basic helix-loop-helix transcription factors E47 and BETA2.

J Biol Chem

273

:

19945

19954

25

Takebayashi

K

,

Sasai

Y

,

Sakai

Y

,

Watanabe

T

,

Nakanishi

S

,

Kageyama

R

1994

Structure, chromosomal locus, and promoter analysis of the gene encoding the mouse helix-loop-helix factor HES-1. Negative autoregulation through the multiple N box elements.

J Biol Chem

269

:

5150

5156

26

Aranburu

A

,

Carlsson

R

,

Persson

C

,

Leanderson

T

2001

Transcription factor AP-4 is a ligand for immunoglobulin-kappa promoter E-box elements.

Biochem J

354

:

431

438

27

Swanson

HI

,

Yang

JH

1999

Specificity of DNA binding of the c-Myc/Max and ARNT/ARNT dimers at the CACGTG recognition site.

Nucleic Acids Res

27

:

3205

3212

28

German

MS

,

Moss

LG

,

Wang

J

,

Rutter

WJ

1992

The insulin and islet amyloid polypeptide genes contain similar cell-specific promoter elements that bind identical β-cell nuclear complexes.

Mol Cell Biol

12

:

1777

1788

29

Weiner

MP

,

Costa

GL

,

Schoettlin

W

,

Cline

J

,

Mathur

E

,

Bauer

JC

1994

Site-directed mutagenesis of double-stranded DNA by the polymerase chain reaction.

Gene

151

:

119

123

30

Mirmira

RG

,

Watada

H

,

German

MS

2000

β-Cell differentiation factor Nkx6.1 contains distinct DNA binding interference and transcriptional repression domains.

J Biol Chem

275

:

14743

14751

31

Watada

H

,

Mirmira

RG

,

Kalamaras

J

,

German

MS

2000

Intramolecular control of transcriptional activity by the NK2-specific domain in NK-2 homeodomain proteins.

Proc Natl Acad Sci USA

97

:

9443

9448

Copyright © 2004 by The Endocrine Society

Citations

Views

Altmetric

Metrics

Total Views 862

606 Pageviews

256 PDF Downloads

Since 1/1/2017

Month: Total Views:
January 2017 3
February 2017 13
March 2017 8
May 2017 1
July 2017 2
August 2017 1
September 2017 4
November 2017 1
December 2017 6
January 2018 3
February 2018 8
March 2018 18
April 2018 9
May 2018 13
June 2018 5
July 2018 10
August 2018 6
September 2018 5
October 2018 3
November 2018 16
December 2018 8
January 2019 9
February 2019 15
March 2019 19
April 2019 13
May 2019 8
June 2019 15
July 2019 22
August 2019 6
September 2019 17
October 2019 21
November 2019 13
December 2019 7
January 2020 17
February 2020 10
March 2020 8
April 2020 7
May 2020 4
June 2020 10
July 2020 5
August 2020 13
September 2020 10
October 2020 5
November 2020 7
December 2020 6
January 2021 8
February 2021 6
March 2021 16
April 2021 7
May 2021 5
June 2021 19
July 2021 7
August 2021 6
September 2021 5
October 2021 6
November 2021 10
December 2021 1
January 2022 3
February 2022 15
March 2022 8
April 2022 6
May 2022 3
June 2022 7
July 2022 17
August 2022 7
September 2022 29
October 2022 8
November 2022 2
December 2022 16
January 2023 10
February 2023 5
March 2023 18
April 2023 11
May 2023 11
June 2023 3
July 2023 4
August 2023 10
September 2023 8
October 2023 12
November 2023 10
December 2023 9
January 2024 17
February 2024 15
March 2024 17
April 2024 11
May 2024 9
June 2024 16
July 2024 12
August 2024 5
September 2024 14
October 2024 8

Citations

61 Web of Science

×

Email alerts

More on this topic

Citing articles via

More from Oxford Academic