Abstract
PDX1 is a crucial transcription factor in pancreas development and mature β-cell function. However, the regulation of PDX1 expression in larger animals mirroring human pancreas morphogenesis and endocrine maturation remains poorly understood. Therefore, we conducted a comparative analysis to characterize regulatory regions of goat PDX1 gene and assessed their transcriptional activity by transient transfection of several transgenic EGFP constructs in β- and non-β cell lines. We recognized several highly conserved regions encompassing the promoter and cis-regulatory elements (Area I-IV) at 5’ flanking sequence of the genes. Within the promoter, we identified that a key E-box and nearby CAAT element synergistically drive transcription, constituting the basal promoter of goat PDX1 gene. Furthermore, each recognized regulatory area separately enhances this basal promoter activity in β-cells compared to non-β cells; however, cooperatively, they exhibit a bifunctional regulatory effect on transcription. Additionally, the intact ~ 3 kb upstream region (Area I-III) functions as the most efficient reporter transgene in vitro and shows islet-specific expression in native rat pancreas. Together, our findings suggest that the regulation of goat PDX1 gene is governed by conserved regions similar to other mammals, while both structurally and functionally, these regions exhibit a closer resemblance to those found in humans.
Supplementary Information
The online version contains supplementary material available at 10.1038/s41598-024-77614-0.
Keywords: Pancreas development, Islet beta cell (β-cell), PDX1, Tissue‐specific transcription factor, Diabetes, Cis-Regulatory elements
Subject terms: Gene regulation, Gene expression analysis
Introduction
Pancreas organogenesis has been extensively studied in several vertebrate species such as rodents1, humans2,3, and small ruminants (sheep and goat)4–6. In the process of endoderm patterning, inductive signals play a crucial role in subdividing the developing gut into organ-specific areas and influencing the expression of different transcription factors (TFs). These TFs take part in the specification of foregut endodermal epithelium to dorsal (DP) and ventral (VP) pancreatic buds, leading to pancreas formation7,8. Even though the signaling pathway through which TFs regulate pancreas organogenesis is highly conserved, the timeline of cell lineage specification and functional maturation are different between rodents and large animals9. For instance, unlike mice, which undergo three major transition phases during pancreas organogenesis, humans do not have a distinct secondary transition for endocrine differentiation. Instead, human pancreatic development progresses more continuously from the early stages. This smoother progression contrasts with the sharp developmental transitions observed in rodents, highlighting evolutionary differences in pancreatic development3. In human and sheep, functional maturation of insulin-secreting cells mostly occurs before birth, while in rodents it proceeds postnatally5,10–14. Islet formation in sheep and humans involves epithelial-mesenchymal transition (EMT), marked by the co-expression of the mesenchymal marker vimentin and insulin during pancreatogenesis15. This co-expression persists into adulthood in a subset of islet β-cells, unlike in rodents where vimentin expression is absent in adult β-cells. This difference suggests a more extended and sustained islet growth and maturation process in large mammals compared to rodents15. Pancreas development in human, sheep, and mice share conserved molecular mechanisms, however, the expression patterns of key factors show differences across these species. Pancreatic and duodenal homeobox factor 1 (PDX1), a master regulator of pancreas generation, is expressed between 29 and 31 days of gestational age (dGA) in humans and 24–29 dGA in sheep, but it is observed in an earlier embryonic day (E8.5) in mice15. PDX1 plays a key role in early pancreatic specification, as well as in the maturation and function of islet β-cells. Initially, PDX1 is highly expressed in epithelial progenitor cells throughout the pancreatic epithelium, surrounding gut tube, and central nervous system. However, during the EMT phase, its expression decreases as these progenitor cells transition to a mesenchymal state, allowing them to migrate and cluster together to form islets. In adults, PDX1 expression increases again and becomes primarily restricted to β-cells and some δ-cells16. Although the high levels of Pdx1 expression induce β-cell specification, reduced expression of this TF is required for acinar cell differentiation and maturation as well17. Pdx1 knock-out mouse embryos with pancreas deficiency do not stay alive, however, a small dorsal bud with a few glucagon-expressing cells survive without insulin expression18,19.
Developmental studies show that different levels of Pdx1 expression during pancreas specification and in various cell types are regulated by a conserved proximal promoter and several distal enhancers in the 5’ flanking region20. Pdx1 transcription is controlled by a TATA-less promoter, leading to the utilization of multiple transcription start sites, as reported for humans, mice, and rats. Analysis of reporter constructs driven by the Pdx1 promoter in transgenic mice and cultured β-cells revealed that approximately 6.5 kb upstream of the mouse Pdx1 transcriptional start site contains four cis-regulatory regions, Area I-IV, essential for its regulation21. Proximal enhancers including Areas I, II, and III are located between − 2700 and − 1800 bp and control Pdx1 expression during pancreatic development20. Deletion of Area II resulted in serious developmental deficiency and massive failure of endocrine progenitor development. However, the remaining Areas (I, III, and IV) were enough to drive early pancreatic multipotent progenitor specification and complete acinar differentiation22. A complementary study also revealed that the predominant role of Area II in β-cell development is potentiated by Area I, while Area III influences acinar development through PTF1A binding23. Distal Area IV is located between − 6200 to -5670 bp and is only active in postnatal β-cells. In contrast to mutations in neighboring Areas I-III, Area IV–specific mice knockouts did not disturb embryonic or early postnatal β-cell development, although neurogenin3 progenitors and subsequent insulin and somatostatin expressing cells were reduced24. In addition, targeted demethylation of Area IV significantly decreased the function of Pdx1 enhancer as well as the expression of its target genes25. Identical regulatory regions were also found in the human genome between − 2810 and − 1670 of the PDX1 gene, referred to as PH1, PH2, and PH326 but ~ 0.5 kb of human PDX1 Area IV element is located farther between − 8656 to -8155 bp27. These regulatory areas are highly conserved in Pdx1-expressing species, but the PH2/Area II domain is absent in chickens28.
DNase I footprinting analysis showed that several trans-activators, including SP1/3, HNF-3b, HNF-1a, MAFA/B, PAX6, NKX2.2, and PDX1, bind to and modulate the transcriptional activity of four conserved Areas. Additionally, potential cis-acting binding sites for various TFs such as bHLH/bHLH-ZIP proteins (E-box), CTF/NF-1 (CAAT), C/EBP, and upstream stimulatory factor (USF) were identified around the proximal promoter sequences. Mutations and deletions in these sequences severely reduced Pdx1 promoter function, while point mutations within the conserved E-box motif abolished specific transcription in β-cells21. These findings highlight PDX1 as the master TF for pancreas formation and β-cell function.
To date, a detailed analysis of the literature has revealed a noticeable lack of experimental investigations clarifying the function of PDX1 regulatory regions in large animal models. The improved understanding of the function of these regions in goat, sheep and pig whose fetal physiology, organ size, gestational period and especially β-cell proliferation and differentiation resemble the human15,29, raise the possibility of using the animals as research models for pancreas biological studies and even appropriate hosts for generation of human organs30.
In the present study, we identified the chromosomal location and the sequence of goat PDX1 promoter as well as its regulatory elements, through a comparative genomic analysis. We also identified five highly conserved regions between human and goat including the promoter sequence and four cis-regulatory areas. Our findings demonstrate that the proximal sequence could efficiently express the EGFP reporter gene in β-cell lines. The basal activity of the promoter is driven by a functional E-box II (-192) and a nearby CAAT box (-213) within this sequence. Moreover, we discuss changes in the transcriptional activity of this basal promoter when cis-regulatory regions are introduced upstream. Although each of these regulatory areas enhanced the transcriptional activity of the promoter, their bi-functional regulatory role was also observed cooperatively.
Results
Comparative genomic analysis of identified goat PDX1 upstream region
As mentioned previously, critical cis-acting transcription regulatory elements of mouse Pdx1 span around the 6.2 kb of the 5’-flanking sequence of its genomic region20,21. This conserved regulatory region was also characterized in ~ 6.5 and ~ 8.6 kb of the 5’-flanking rat and human Pdx1/PDX1 genes, respectively27. To identify this noncoding regulatory region within the goat genome, approximately 9 kb of PDX1 sequence was obtained from the whole genome shotgun sequence of the Capra hircus (NCBI Reference Sequence: NC_030819.1). This sequence encompasses 8.9 kb of the 5’-flanking region upstream of the transcription start site and continues with the 5’UTR and 0.1 kb related to the ATG start codon (chr12: 54590100–54599100). Comparative analysis of this sequence was conducted using mVISTA with a default identity level of 70% over a 100 bp length and the MLAGAN alignment program. This analysis identified twelve conserved regions within the coordinates of human sequence (Supplementary Table S3). Among them, five sequences were highly similar across rodents (Fig. 1A and Fig. S1). Based on these multiple sequence alignments, VISTA also visualized the genetic diversity between rodents and large animals in the case of PDX1 5’ upstream region by the phylogeny diagram. Rank VISTA analysis indicated that these five noncoding conserved sequences were statistically significant homologous in the alignment (P value < 0.05) and perfectly matched with four identified regulatory areas (Area I-IV) plus the core promoter region of PDX1 (Fig. 1A and Supplementary Table S4).
Fig. 1.
Comparative analysis of goat PDX1 upstream regulatory regions. (A) Schematic diagrams indicate the goat PDX1 chromosome position (upper panel) and comparative analysis of ~ 9 kb upstream regulatory regions among human, mice, and rat (middle panel) by mVISTA. Phylogenic diversity among the species is also displayed in the left panel. (B) Sequence alignment analysis of goat PDX1 upstream regulatory region by UCSC shows several evolutionary conserved sequences among goat, human, and a hundred vertebrates, which are additionally analyzed by some track hubs including EPD, CAGE, and GeneHancer. (C) Transcription initiation region of human PDX1 aligned with several other species and the multiple start sites are reported by FANTOM5 and CAGE (upper panel). The sequencing result of goat PDX1 transcription initiation site also depicts in the bottom panel. (D) Graphical maps of CpG islands within ~ 3.5 kb PDX1 upstream regulatory region of goat, human, mouse, and rat, which are analyzed by MethPrimer browser (right panel) and schematic representation of CpG islands and their interference with regulatory regions (left panel). (E) MatInspector analysis of promoter elements within ~ 1.5 kb (˗1.4 to + 0.1 from the start codon ATG) of goat PDX1 region by Genomatix. Lavender color boxes; four regulatory areas, tan color box; proximal promoter, light blue boxes; CGIs (CpG islands), orange boxes; CEBPs, dark blue boxes; E-boxes, green box; CAAT box, pink boxes; CG-boxes.
Detailed analysis of this sequence by the UCSC Genome Browser revealed a considerable identity among a hundred vertebrate species in the upstream region of their Pdx1 gene. Among studied species, the identity of the goat PDX1 regulatory region with that of human, mouse, and rat was 86.6%, 84.4%, and 83.0%, respectively. A significantly higher score was observed over a longer stretch of the sequence in human BLAT analysis (GRCh37/hg19, Score: 1804, Span: 9704) compared to the best hits in mouse (Score: 557, Span: 4777) and rat (Score: 831, Span: 6151). In accordance with previously reported regulatory sequences of the PDX1 gene, the GeneHancer track set in UCSC analysis shows the interaction between proximal evolutionary conserved enhancers (Area I-III) and PDX1 promoter (Fig. 1B). However, a recent study has revealed a physical interaction between regulatory regions and promoters in islet endocrine cells. This study found that a distal regulatory area (Area IV) also interacts with PDX1 promoter both in β and δ-cells31. Hence, these highly homologous sequences were considered potential candidates for goat PDX1 cis-regulatory elements (CREs), which were followed by sequencing analysis of respective PCR products of goat genomic regions and functional analysis. The absence of a consensus TATA box as a transcription initiator sequence in the 5’-flanking region of the goat PDX1 genomic sequence, similar to that seen in humans and rodents, prompted us to analyze the transcription initiation sites for this gene. RefTSS identifies a transcription initiation peak for human PDX1 located 140 bp upstream of the translation start site. This peak has been validated as a major transcription start site through the analysis of mapped reads using FANTOM5 and CAGE. Additionally, several alternative minor transcription start sites upstream of the ATG start codon were identified (Fig. 1C). Although the transcription start sites identified in this study have nucleotide locations that differ from those previously described32, they are all located in the same DNA region and exhibit highly conserved nucleotides across species (Fig. 1C). In this study, mRNAs were collected from a goat fetal pancreatic sample at 30 dGA and reversely transcribed using PDX1-specific primers (Supplementary Table S1). The sequencing results indicate that goat PDX1 mRNAs are transcribed from an initiation site located 209 bp upstream of the translation start codon, with an 89.2% identity to the corresponding human region, which is higher than that observed in rodents. The NCBI BLAST result confirmed that the obtained sequence was highly similar to several predicted PDX1 mRNA sequences and completely matched the reported Ovis aries PDX1 partial coding sequence (Sequence ID: KP059125.1)33.
CpG islands (CGIs) are typically considered as the sites of transcription initiation and are linked to gene promoters34. MethPrimer browser was used to analyze regions up to 3.5 kb upstream of the PDX1 gene in various species, such as goat, human, mouse, and rat. The borders of CGIs were depicted as light blue boxes in peaks and valleys graphs. We have illustrated these graphical maps in schematics to better illustrate their interaction with regulatory regions. As shown in Fig. 1D, six CpG-rich clusters are distributed throughout the analyzed PDX1 sequences of goat and human, indicating a higher CpG dinucleotide content in the DNA of large animals compared to rodents. Although rats have four CGIs in upstream regulatory region relative to Pdx1 translation start site, the CpG dinucleotides content, particularly those around the promoter are significantly lower. In fact, the increase in the number of CGIs during mammalian evolution is attributed to the rise in chromosome number and recombination rate35, explaining the presence of a higher number of CGIs in the analyzed goat genome in comparison to other species. Subsequent analysis for promoter region identification was performed using Genomatix (Gene2Promoter) to determine the conservation of goat PDX1 sequence with some orthologous promoters via comparative genomic analysis. In addition, MatInspector was utilized to find general core promoter elements such as CEBP, CAAT, GC, and E-boxes. According to Fig. 1E, three CEBPs, one CAAT, two GC-boxes, and six E-boxes were recognized within this promoter region. Some of these elements were consistent with the sequences reported for human and rodent PDX1/Pdx1 promoter sequences21. Therefore, about 1.5 kb fragment (from − 1.4 to + 0.1 kb relative to the translation initiation site) was cloned, sequenced, and functionally characterized by transfection into β and non-β-cell lines.
Sequences necessary for β-cell-specific expression of the goat PDX1 promoter
To experimentally delineate the functional sequences in the 5’ flanking promoter region of goat PDX1, a fragment extending about 1.5 kb was cloned and linked to the EGFP reporter gene. Subsequently, a series of 5’ and 3’ deletion constructs were developed as illustrated in Fig. 2. Seventeen constructs were obtained and transiently transfected into BTC3 and MIN6 cells to evaluate their transcriptional activity using flow cytometry (Fig. 3A, B). The results showed that deletion of up to 608 bp from the full-length promoter (PDX-FR2) did not significantly affect EGFP expression in the PDX-FR1 and PDX-V0 constructs (P > 0.05 for PDX-FR1 and P > 0.9 for PDX1-V0 in both cell lines). The AseI-SbfI restriction deletion shortened this sequence from − 735 to -533 bp that contained the CEBP III and II. This deletion led to a substantial enhancement in the reporter gene activity of PDX-V1 construct in BTC3 cells (P < 0.0001), while it had no effect on reporter gene activity in MIN6 cells (P = 0.6149).
Fig. 2.
Schematic representation of goat PDX1 regulatory elements and their cloning process. The position of primers and restriction enzymes used to construct regulatory fragments is shown relative to the ATG start codon.
Fig. 3.
Functional analysis of recognized motifs in goat PDX1 promoter region. (A) A series of 5’ and 3’ goat PDX1 promoter deletion plasmids (left panel) were constructed and transfected to β-cells including MIN6 and BTC3 (right panel). The activity of each construct was measured by flow cytometry 48 h after transfection. Data are presented as mean ± SEM for three independent transfection experiments. *P < 0.0021 and ****P < 0.0001 for BTC3 cells, ##P = 0.0013, ###P = 0.0002 and ####P < 0.0001 for MIN6 cells, $$$$P ≤ 0.0001 for regulatory constructs and ns; no significance. (B) Transfected β-cells were imaged using a fluorescence microscope (upper panels) 48 h after transfection and their flow cytometry analysis revealed the percentage and intensity of EGFP expression of each construct (bottom panels). pCMV-EGFP; positive control. PL; Promoter-less construct. Scale bar; 200 μm.
Additional deletion of the CEBP I sequence reduced the promoter activity of PDX-V2 constructs by about 40% and 75% in MIN6 and BTC3 cells, respectively (P < 0.0001). In the following, the removal of E-BOX III did not further reduce the promoter activity of PDX-V3 (P > 0.9 for BTC3 and P = 0.001 for MIN6). However, deletion of CAAT element led to a severe reduction (~ 3-fold decrease) in reporter activity of both BTC3 (P = 0.027) and MIN6 (P < 0.0001). Despite the known importance of E-Box II as a crucial proximal element in PDX1 promoter function21, PDX-V4 exhibited minimal reporter expression. Similarly, the transcriptional activity was not affected by additional 5’ deletion of two other tandem elements (GC-box in PDX-V5 and E-BOX I in PDX-V6 constructs), especially when compared to the promoter-less construct with negligible EGFP expression.
In addition to the 5’ deletion analysis, a series of 3’ deletion constructs were generated to investigate which of the examined promoter elements could restore the transcriptional activity of the PDX1 promoter. The PDX-ΔV1, PDX-ΔV2, and PDX-ΔV3 constructs containing the CEBP I-III and E-BOX III elements could not drive the transcription of the PDX1 promoter. In contrast to our previous expectation regarding the CAAT elimination, adding this element did not improve the promoter activity of PDX-V4. Interestingly, this activity was restored when the E-BOX II was located adjacent to CAAT in the PDX-ΔV5 construct. This 10-fold increase (P < 0.0001 for both cell lines) remained unchanged by the promoter sequence expansion up to 50 bp where the GC-box was present in the promoter. The contradictory effect of CAAT and E-BOX II on transcriptional activity was also analyzed by the elimination of these sequences from the full-length promoter. Both individual deletions (PDX-ΔC and PDX-ΔE2) reduced the promoter strength in driving EGFP expression by approximately 70–50% in β-cell lines (P ≤ 0.0001), compared to PDX-FR2, the intact control construct. This data confirms that both elements are required simultaneously for effective regulation of PDX1 expression.
β-cell-specific expression of goat PDX1 through enhancer element sequences
Regulatory areas of the Pdx1 gene have been previously defined as critical cis-regulatory elements for PDX1 expression and thus maturation and maintenance of β-cells23. These four areas within the goat PDX1 upstream region were initially amplified using specific primers as depicted in Fig. 2. Sequencing of these areas showed a significant sequence identity to proximal and distal enhancer areas (Area I-IV) of the human, mouse, and rat PDX1/Pdx1 genes (Fig. S1). To determine their stimulatory properties, these areas were subcloned directly upstream of the obtained V3 goat PDX1 basal promoter (V3) to generate “pA” plasmids. The β-cell-specific expression of these constructs was evaluated by transfection assay in β (BTC3, MIN6) and non-β (NIH3T3, MCFF) cells (Fig. S2) and the results are presented as the expression ratio of β- relative to non-β-cells (Fig. 4 and Fig. S3).
Fig. 4.
Regulatory properties of goat PDX1 areas. (A) Regulatory areas of goat PDX1 were cloned upstream of core promoter pV3 plasmid (left panel) and transfected to β-cells (MIN6 and BTC3) and non β-cells (MCFF) (right panel). The intact ~ 3 kb upstream region was also cloned and analyzed in combination with and without Area IV. The activity of each construct was evaluated by flow cytometry 48 h after transfection. To normalize the activity of the reporter, relative EGFP expression is calculated by dividing the expression of each construct to pV3, and the β-cell specificity results are presented as the relative activity of each construct in β-cells to MCFF. Data are presented as mean ± SEM for three independent transfection experiments. ****P < 0.0001 for BTC3 cells, ####P < 0.0001 for MIN6 cells and $$$$P < 0.0001 for BTC3 vs. MIN6 cells, ++++P < 0.0001 for regulatory constructs and ns; no significance. (B) Images of transfected reporter activity of β-cells using a fluorescence microscope (upper panels) 48 h after transfection and their flow cytometry analysis (bottom panels). Scale bar; 200 μm.
Among four conserved regulatory areas, Area II could drive the highest specific EGFP expression independently in both β-cell lines (P < 0.0001). Area I showed higher activity in β-cells relative to MCFF than Area IV (P < 0.0001) (Fig. 4), while owing to their higher level of expression in NIH3T3 cells, both exhibited equal lower activity (Fig. S3). Area III element directed EGFP expression at a higher level in BTC3 compared to MIN6 cells (P < 0.0001), consistent with earlier findings of human Area III activity in these β-cells28.
In addition to independent analysis of regulatory areas, we assessed their cooperative activity by their different combinations upstream of the V3 core promoter. According to previous studies, Area I/II showed an additive effect in the expression of the reporter gene in the islet β-cells36. In our experiment as shown in Fig. 4, EGFP expression was also effectively driven by pA12V3 compared to other combinations. However, this construct showed lower activity compared to Area II when the expression level was measured relative to non-β-cells (Fig. S2). The addition of either Area III or Area IV to pA12 resulted in pA123 and pA412 constructs, respectively, and led to a decrease in the rate of EGFP expression in MIN6 cells (P < 0.0001 for both construct) (Fig. 4). Nevertheless, combining both of these areas in the pA4123V3 construct restored the initial expression activity of pA12V3 (P > 0.9). To determine whether the physical proximity of Area IV with the intact ~ 3 kb upstream region of goat PDX1 promoter (AR) also contributes to diminishing the promoter activity, this area was cloned upstream of AR fragment (pA4AR). As a result, the addition of Area IV noticeably reduced the EGFP expression level in both transfected β-cell lines (P < 0.0001).
Islet-specific expression of goat PDX1 areas in pancreatic tissue
Previous studies indicated that pancreatic intraductal hydrodynamic injection can be effectively employed for the in vivo evaluation of plasmids37. Thus, we aimed to investigate the expression pattern of pAR plasmid in native pancreatic islets. Initially, the common-bile duct exposure, cannulation, and infusion procedure were optimized using Trypan Blue dye, which gave rise to a uniform distribution of the injected dye in pancreas (Fig. S4A). Moreover, Hematoxylin and Eosin (H&E) staining analysis was performed to examine the impact of intraductal injection on pancreatic tissue damage. Our findings revealed that up to 24 h after injection, the transient expansion of intercellular space returned to the normal size of around 20 μm (Fig. S4B). Next, different injection parameters using pCMV-EGFP plasmid were compared. Isolated islets revealed that an injection volume of approximately 3% of body weight containing about 9 µg plasmid with a flow rate of 1 ml/s was the optimal injection condition with the most islet-targeted reporter expression (Fig. S4C). Employing this injection condition, pAR was used to validate the pancreatic islet-specific EGFP expression. 48 h post-injection, immunofluorescence (IHF) staining of the dissected pancreas verified the expression of EGFP in the islets (Fig. 5A). In addition, EGFP mRNA level was determined in isolated islets by RT-qPCR and revealed that EGFP expression was restricted to the islets of the pancreas but not surrounding tissues (Fig. 5B).
Fig. 5.
Islet-specific expression of goat PDX1 regulatory areas. (A) The pAR-EGFP construct was injected into the male Wistar rat’s pancreas duct. 48 h post-injection the pancreas sections were stained by insulin, EGFP, and DAPI. Scale bars; 200 μm (upper panels) and 20 μm (bottom panels). (B) The relative mRNA levels of EGFP in islets of the pancreas, liver, duodenum, and spleen were analyzed by RT-qPCR in which pAR-EGFP was used as test plasmid and pCMV-EGFP was used as positive control). 18s rRNA was employed as an internal control. Data are presented as mean ± SEM for three independent experiments.
Discussion
In pancreatic development, the coordinated expression of several TFs orchestrates the acquisition of exocrine and endocrine compartments. Mice are frequently employed as models for uncovering the molecular mechanisms behind pancreas formation, however primate islet anatomy and function noticeably differ from rodents9. Small ruminants, such as goat and sheep, have advantages due to their resemblance to humans in pancreas morphogenesis6,15,38,39, and the timing of structural and functional maturation of fetal endocrine cells29,40. In human and sheep the formation of glucose-sensitive cells and insulin secretion occurs before birth, while in rat’s islet maturation proceeds postnatally29. Similarly, β-cell regulatory factors like PDX1 are expressed later in human and sheep than in rodents15,41. Therefore, a better understanding of the molecular basis of pancreas organogenesis in comparative animal models seems essential. Lacking information about PDX1 regulatory regions in large animal models, we decided to identify the goat PDX1 regulatory areas within the 5’ upstream region of this gene.
By comparative genome analysis we identified five closely matched non-coding regions with human PDX1, including the promoter and four regulatory areas (Areas I–IV). The level of identity between goat PDX1 regulatory regions and their homologs in human was 90.3%, 94.6%, 94.6%, 88.8%, and 93.4% for the promoter and Areas I to IV, respectively, which was higher compared to rodents27. Furthermore, within these regulatory regions, we observed a significant overlap of CpG islands between human and goat, indicating more similar tissue-specific methylation dynamics. Research has found that the PDX1 promoter has low methylation levels in beta cells whereas in alpha cells, it is highly methylated42. This promoter-associated methylation status is increased in the islets of diabetic patients43 and pancreatic neuroendocrine tumors44. In addition, in an aberrant metabolic intrauterine environment, Pdx1 is methylated at several CpG sites and permanently silenced, a state that persists after birth and leads to the onset of diabetes in adulthood45. Despite different epigenetic regulation in rodents due to lower CGI density, human and small ruminants likely undergo similar intrauterine growth retardation and epigenetic changes in the PDX1 promoter during development46.
As shown in Fig. 1E, we observed that the goat PDX1 promoter, similar to other TATA-less promoters, contains multiple G/A and G/C sequences along with cis-acting elements such as CEBP, CAAT, GC-, and E-boxes32, 47–49. To investigate the significance of these elements in promoter activity, we generated a series of deletion constructs. Removal of identified elements from the 5’ end affected promoter activity, but deletion of the CAAT box led to a substantial decrease in EGFP expression in β-cells. Further deletion of adjacent E-BOX II did not cause an additional reduction in reporter expression. These observations differ from previous studies that suggested the deletion of E-BOX II as a key element resulted in a significant reduction in mouse Pdx1 promoter activity21. In our analysis of 3’ deletion constructs, the CAAT box alone was insufficient to drive transcription, but the addition of E-BOX II restored promoter activity. Moreover, elimination of each sequences from the full-length promoter resulted in a significant decrease in reporter expression, indicating that both CAAT and E-BOX II are essential for the basal activity of the goat PDX1 promoter. In TATA-less gene regulation, the CAAT box serves as a binding site for nuclear factor Y50–52. NFY can either activate or suppress transcription depending on the promoter context53. The stimulatory mechanism of NFY may rely on its suppressive association with USF-1, which binds to the E-box sequence54. Alternatively, the USF1/PDX1 complex auto-regulates the PDX1 promoter in β-cells, but this feedback loop can be disrupted by SREBP-1c and protein kinase CK248,49. These findings suggest that competition between activators and repressors at PDX1 promoters regulates its expression.
In addition to promoter regulations, transactivation of conserved 5′ cis-regulatory areas is essential for Pdx1 expression, driven by islet-enriched TFs55–57. Therefore, we investigated how the goat PDX1 regulatory areas affect gene expression in β-cell lines. It has been shown that enhancers respond differently to various types of promoters58. Accordingly, goat PDX1 areas were sub-cloned directly upstream of the acquired basal promoter (V3). The results from transient transfection of β-cell lines have demonstrated that all these areas independently enhanced the transcriptional activity of the basal promoter (Fig S2). Among them, Area II exhibited the highest level of expression, which aligns with its known pivotal role in driving Pdx1 transcription22. We also observed subsequent levels of expression in Areas I, emphasizing the functional interactions between Areas I and II in inducing robust Pdx1 expression in β-cells36. In our study, modest reporter expression observed in goat Area III and IV, highlighting their similar regulatory properties in mature β-cells. These regions harbor analogous binding sites for factors that contribute to pancreatic fate determination and β-cell lineage specifications such as FOXA59, PDX160, PTF1A28,61, and EGR162. Area III predominantly regulates Pdx1 expression throughout the pancreatic bud during early development via PTF1A61. Stage-restricted behavior of Area IV was verified by an enhancer-reporter assay during the definitive to pancreatic endoderm transition. This enhancer is primed by acquiring FOXA2, chromatin remodeling, and H3K4me1 deposition, sequentially63. Subsequent activation of this enhancer occurs through the recruitment of PDX1 and histone acetylation during the primitive gut tube stage64. Additionally, experiments showed that low Pdx1 expression directed by Area IV is sufficient for acinar cell development and gut/stomach cell specification independent of other areas65. Area IV-specific knockout had a minor effect on pancreatic development but reduced Ngn3 + cells, leading to a smaller islet β-cell area24. Therefore, the spatiotemporal activity of Areas III and IV seems to be influenced by their epigenetic signature and the stage of differentiation61–64. To understand the interactive behavior of these regulatory modules, we placed them upstream of the Area I-II segment (A12), as the most transcriptionally active enhancer complex in β-cell lines. Addition of Area III or Area IV significantly decreased the β-specific activity of the A12 construct, suggesting their involvement in the dose-dependent regulation of PDX1 as they harbor motifs for both lineage and signal-dependent transcription factors66. This is supported by studies showing that some regulatory elements can function as both enhancers and silencers depending on the cellular context67. Accordingly, we integrated the distant Area IV upstream of the ~ 3 kb goat PDX1 sequence, resulting in a marked reduction of β-cell selective enhancer activity compared to non-β cells. This proximity may eliminate several regulatory regions within the native genomic context, the 3D architecture of enhancer clusters, and the structure of the adjacent islet-specific LncRNA (PLUTO)68. Notably, in a study conducted by Chiou et al., physical interactions between regulatory regions and promoters in primary islets were revealed. Their data showed that the PDX1 promoter in alpha cells links with Areas I-III, while in beta and delta cells, it is controlled by Areas IV and I-III31.
The present study confronts certain limitations. Goats are important species both in agricultural and biomedical studies69, yet their current reference genome is incomplete70 and RNA sequencing is limited to a few numbers of tissues that have been annotated using automated computational analysis. The inadequate comprehensive bioinformatic studies and motif discovery tools has restricted our ability to predict TF binding sites within the PDX1 regulatory region. Future research could address this limitation by creating binding-defective mutants for each TF site and analyzing their regulatory function in detail. The enriched transcription factors in β-cells also influence the specific activity of PDX1 regulatory regions60. Despite sharing a similar T-antigen origin, MIN6 and BTC3 cells exhibit noticeably distinct G1/S expression patterns71, different levels of glucose-stimulated insulin secretion72, diverse ability to form pseudo islets73, and different expression of β-cell specific TFs. Nevertheless, MIN6 cells widely used in pancreatic research73, provided more reliable regulatory expression data in our experiments. On the other hand, the NIH3T3 cell line, often considered as a non-beta cell line, displayed a similar goat PDX1 expression pattern to the applied β-cells (Fig. S2, S3). Likewise, mouse Pdx1 regulatory constructs exhibited a high level of activity in NIH3T3 cells (Fig. S5). As a consequence, the ability of NIH3T3 cells to express PDX1 (74) could inaccurately alter the β-cell specificity of regulatory fragments. Hence, it seems that the functional activity of PDX1 regulatory areas could be more precisely discriminated in MIN6/MCFF cells. In future research, this challenge could be tackled by utilizing primary goat β-cells along with transgenic studies which are the ideal method to unravel how these enhancers operate in a multi-cellular context.
Conclusion
Taken together, our study demonstrates that varying thresholds of PDX1 activity, particularly in relation to the regulatory regions, are crucial for optimal β-cell function. By identifying and analyzing the promoter and non-coding regions of the goat PDX1 gene, we have emphasized the vital role of cis-regulatory elements and their complex interactions in driving β-cell-specific expression. Notably, the conserved nature of these regulatory areas and their functional similarities to human counterparts suggest that goats serve as a valuable model for understanding pancreatic organogenesis and diabetes pathogenesis. However, future studies employing primary β-cells and transgenic models will be crucial in elucidating the precise regulatory dynamics of PDX1.
Materials and methods
Bioinformatics studies in characterization of 5’ flanking regulatory regions of goat PDX1 gene
The sequence of the characterized goat PDX1 gene including the 5’ flanking region was obtained from NCBI’s reference sequence database (NC_030819.1) and blasted with the UCSC genome browser on human (GRCh37/hg19). To locate candidate orthologous regulatory regions, the obtained sequence was also aligned with genomes of related species by VISTA75 (Table S3, S4). Pdx1 core promoter sequences from other species were achieved by the Gene2Promoter database and compared with the goat sequence. Further analysis of this sequence using MatInspector76 depicts core binding sites in promoter elements. MethPrimer browser77 was also employed to identify the CpG islands.
Amplification of 5’ flanking regions of goat PDX1 gene
In this experiment, different sets of primers were designed and used to amplify the characterized PDX1 5’ flanking regulatory regions (Table S1) from goat genomic DNA isolated by DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany). As depicted in schematic Fig. 2, the 2.8 kb fragment encompassing Area I, II, and III; spanning the sequences from − 2779 to + 102 relative to the ATG start codon and proximal core promoter region was amplified using PDX-F and A1-R primers. Within this fragment, the ATG start codon mutation was induced by site-directed mutagenesis (ATG/A-F, ATG/A-R). This fragment was utilized as a PCR template as well as a digestion sample to develop other promoter regions. Similarly, proximal regulatory areas (Area I, Area II, and Area III), in addition to distal Area IV, were also amplified using specific primers. The integrity of all fragments was validated by sequencing and restriction digestion using appropriate enzymes (Thermo Scientific, Waltham, MA). Pfu DNA polymerase (Thermo Scientific) was utilized for all PCR reactions and acquired products were then cloned into pTZ57R/T vector (Takara, Kusatso, Japan) after treatment with Ex Taq DNA polymerase (Takara, Kusatso, Japan).
Construction of reporter plasmid
The Luciferase reporter gene in pGL4.10 plasmid (Promega, Madison, WI, USA) was removed and replaced with EGFP fragment excised from pEGFP-C1 (Clontech Laboratories, California, United States) to construct a basic plasmid (pGL4-EGFP). PDX1 promoter fragments were ligated upstream of the reporter gene in pGL4-EGFP plasmid (PL: Promoter-less construct), and a series of deletion constructs were generated. Regulatory areas were also cloned into pDXV3, constructing regulatory plasmids. In parallel, pGL4-CMV-EGFP (pCMV-EGFP) was constructed as a positive control plasmid.
Cell culture
Mouse insulinoma β-cells (MIN6 and BTC3) were cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 4.5 g/L glucose, supplemented with 15% horse serum, 2.5% heat-inactivated bovine serum, 100 U/ml penicillin-streptomycin, and 2 mM L-Glutamine. NIH3T3 and primary male caprine fetal fibroblasts (MCFF) were cultured in DMEM with 4.5 g/L glucose supplemented with 10% FBS and 100 U/ml penicillin-streptomycin. All cell lines were provided by Royan Institute (Esfahan, Iran) and culture medium and other reagents were purchased from Gibco (Waltham, MA).
Cell transfection
When the cells reached approximately 80% confluence in 24-well plates, they were transfected using 1.5 µl Lipofectamine LTX (Thermo Scientific) according to the manufacturer’s recommendation with 500 ng of pDNAs in 100 ul Opti-MEM I reduced-serum medium (Gibco). 48 h after transfection, EGFP expression was evaluated using an inverted fluorescent microscope (IX71; Olympus, Tokyo, Japan), and the images were acquired with a DP72 digital camera (Olympus).
Flow cytometry
48 h post-transfection, about 5 × 105 transfected cells were detached and resuspended to 500 µl PBS and analyzed by flow cytometer. The mean fluorescence intensity (MFI) was measured by a BD FACSCalibur and data were analyzed with Cell Quest Pro software (Becton-Dickinson, Mountain View, USA).
Animals
10 male Wistar rats (10–12 weeks old with an approximate weight of 300 ± 20 g) were provided by Royan Institute (Esfahan, Iran). All methods were conducted in accordance with the relevant guidelines and regulations. Furthermore, all animal protocols were approved by the Institutional Ethical Committee of the Royan Institute (IR.ACECR.ROYAN.REC.1399.107), which are in compliance with the ARRIVE guidelines.
Intraductal injection
Rats were euthanized under the combined administration of xylazine (5 mg/kg body weight per rat) (Bioveta, Czech Republic) and ketamine (75 mg/kg body weight per rat) (Rotexmedica, Germany). Following euthanasia, a preliminary step was taken to ensure proper positioning and stabilization of the animal prior to the surgical intervention. An incision was made in the abdomen to expose the common bile duct (CBD). The highest part of the CBD was clamped to prevent the leak of DNA solution injected into the gallbladder and liver. A 22-gauge angio-catheter was inserted into CBD through the sphincter of Oddi and another clamp was placed on the injection site to restrict the flow of the injected solution into the duodenum. After the injection, clamps were removed and the injection site of the duodenum was sealed with a medical tissue adhesive (Epiglu, Meyer-Haake GmbH, Germany) and the incision site was sutured. As a primary test, 5% trypan blue saline solution was injected to detect the spreading area of the injected solution. Also, to investigate the optimal injection parameters including dose, injection flow rate, and time for tissue harvest, different amounts of pCMV-EGFP ranging from 1 to 20 µg/ml were injected for 5 and 10 s and animals were studied at 24 and 48 h after injection.
Islet isolation
48 h post-injection, rats were sedated by treatment with CO2, and a laparotomy was performed. The pancreata were exposed and perfused with 10 ml collagenase IV solutions (Sigma-Aldrich, St. Louis, MO) through the common bile duct. The distended pancreata were removed and incubated in a water bath for 15 min at 37 °C with shaking. Once digested, tissues were immediately chilled on ice and 10 ml ice-cold STOP solution was added to each sample. Sand-like disrupted tissues were poured through the 500 μm mesh and centrifuged in a swinging-bucket centrifuge at 250 g for 2 min. Pellets were re-suspended in 5 ml Histopaque-1077 (Sigma-Aldrich) and centrifuged at 1750 g for 20 min. The islets were handpicked and washed 2 times with HBSS (Gibco).
Reverse transcription quantitative PCR (RT-qPCR)
Total RNA was isolated from 10 mg of harvested tissues using TRIzol reagent (Sigma-Aldrich). 1 µg of the total RNAs were treated with DNaseI (Thermo Scientific) and then cDNA synthesis was performed by PrimeScript 1st strand cDNA Synthesis Kit (TaKaRa), both according to manufacturer’s protocols. RT-qPCR was conducted by Applied Biosystems StepOnePlus Real-Time PCR System (Thermo Scientific) in 10 µl reactions using the SYBR Green Master Mix (Takara) and rEGFP, rPDX1, and 18s rRNA primers (Table S2), designed by Beacon Designer 7.9 software (Premier Biosoft International, Palo Alto, CA, USA). All RT-qPCR reactions were performed in triplicate. Relative fold changes in gene expression were normalized to 18s rRNA as internal control and calculated using the 2−ΔΔCt method.
Histological analysis
Pancreatic tissue samples isolated from injected animals were fixed in 4% (v/v) paraformaldehyde, paraffin-embedded, and cut to 6 mm in length. Prior to staining, sections were deparaffinized and rehydrated. Hematoxylin and Eosin (H&E) staining was accomplished following routine protocol. The pancreatic cell distances were measured through Image J software (National Institutes of Health, Bethesda, MD). For IHF staining, sections were permeabilized with 0.2% Triton X-100 for 5 min, blocked in 10% host serum in PBS− for 1 h, and then incubated with primary antibodies (rabbit anti-GFP, 1:1000, ab290, Abcam and goat anti-insulin, 1:200, sc-7839, Santa Cruz) at 4 °C overnight. Conjugated secondary antibodies were used for fluorescent detection (goat anti-rabbit FITC, 1:2000, F1262, Sigma-Aldrich and donkey anti-goat Alexafluor 594, 1:1000 A11057, Invitrogen). DAPI dye solution with 1 µg/ml concentration (D-8417, Sigma-Aldrich) was also employed to detect nuclei in immunofluorescent images, and all of them were collected on a fluorescence microscope (BX51, Olympus) equipped with a DP70 digital camera (Olympus).
Statistical analysis
Statistical significance was determined by Student’s t-test and two-way ANOVA using GraphPad Prism (version 8.0.0 for Windows, GraphPad Software, San Diego, California USA), in which P < 0.032 was considered significant, respectively. All data are presented as mean ± standard error of the mean (SEM) for three independent experiments.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Abbreviations
- CAGE
Cap analysis of gene expression
- CBD
Common bile duct
- CDS
Coding sequence
- C/EBPs
CCAAT/enhancer binding proteins
- CGIs
CpG islands
- CREs
Cis-regulatory elements
- dGA
Days of gestational age
- DP
dorsal pancreatic bud
- E-box
Enhancer box
- EGR1
Early growth response factor 1
- EMT
Epithelial-mesenchymal transition
- FANTOM5
Functional Annotation of the Mammalian Genome
- FOXA
Forkhead Box A
- H&E
Hematoxylin and Eosin
- HNF
Hepatocyte Nuclear Factor
- IHF
Immunofluorescence
- IUGR
Intrauterine growth retardation
- MAF
Musculoaponeurotic fibrosarcoma
- MCFF
Male caprine fetal fibroblast
- MFI
Mean fluorescence intensity
- NF-Y
Nuclear factor Y
- NGN3
Neurogenin-3
- Nkx
Homeobox family of NK genes
- PanNETs
Pancreatic neuroendocrine tumors
- PAX
Paired box
- PDX1
Pancreatic and duodenal homeobox factor 1
- PTF1a
Pancreas associated transcription factor 1a
- SP
Specificity protein
- SREBP-1c
Sterol regulatory element-binding protein-1c
- UCSC
University of California, Santa Cruz
- USF
upstream stimulatory factor
- VP
Ventral pancreatic bud
Author contributions
“N.R. Conceptualization, Methodology, Formal analysis, Investigation, Validation, Visualization, Software, Writing-original draft; K.D. Project administration, Resources, Supervision, Validation, Writing-review & editing; A.K.E. Methodology, Validation; S.M. Methodology, Validation; M.R. Methodology, Validation; F.J. Investigation, Validation; M.H.N.E. Funding acquisition, Investigation, Supervision, Writing-review & editing. “.
Data availability
All data generated or analyzed during this study are included in this published article, its supplementary information files, and publicly available repositories. The datasets generated during the current study are also available in the GenBank repository with the accession numbers of PQ066235, PQ066236 and PQ066237.
Declarations
Competing interests
The authors declare no competing interests.
Ethics statement
The animal study protocol was approved by the Institutional Ethical Committee of the Royan Institute (IR.ACECR.ROYAN.REC.1399.107), which is in compliance with the ARRIVE guidelines. Consent to participate is not applicable.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Kianoush Dormiani, Email: k_dormiani@royaninstitute.org.
Mohammad Hossein Nasr-Esfahani, Email: mh.nasr-esfahani@royaninstitute.org.
References
- 1.Dassaye, R., Naidoo, S. & Cerf, M. E. Transcription factor regulation of pancreatic organogenesis, differentiation and maturation. Islets. 8(1), 13–34 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pan, F. C. & Brissova, M. Pancreas development in humans. Curr. Opin. Endocrinol. Diabetes Obes.21(2), 77 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sarkar, S. et al. Global gene expression profiling and histochemical analysis of the developing human fetal pancreas. Diabetologia. 51, 285–297 (2008). [DOI] [PubMed] [Google Scholar]
- 4.Reddy, S., Bibby, N. & Elliott, R. An immunofluorescent study of insulin-, glucagon‐, pancreatic polypeptide‐and somatostatin‐containing cells in the early ovine fetal pancreas. Q. J. Experimental Physiology: Translation Integr.73(2), 225–232 (1988). [DOI] [PubMed] [Google Scholar]
- 5.Willes, R., Boda, J. & Stokes, H. Cytological localization of insulin and insulin concentration in the fetal ovine pancreas. Endocrinology. 84(3), 671–675 (1969). [DOI] [PubMed] [Google Scholar]
- 6.Reddy, S. & Elliott, R. Insulin, glucagon, pancreatic polypeptide hormone and somatostatin in the goat pancreas: demonstration by immunocytochemistry. Australian J. Biol. Sci.38(1), 59–66 (1985). [PubMed] [Google Scholar]
- 7.Zaret, K. S. & Grompe, M. Generation and regeneration of cells of the liver and pancreas. Science. 322(5907), 1490–1494 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stanger, B. Z. & Hebrok, M. Control of cell identity in pancreas development and regeneration. Gastroenterology. 144(6), 1170–1179 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bakhti, M., Böttcher, A. & Lickert, H. Modelling the endocrine pancreas in health and disease. Nat. Reviews Endocrinol.15(3), 155–171 (2019). [DOI] [PubMed] [Google Scholar]
- 10.Fowden, A. L. Effects of arginine and glucose on the release of insulin in the sheep fetus. J. Endocrinol.85(1), 121–129 (1980). [DOI] [PubMed] [Google Scholar]
- 11.Otonkoski, T., Andersson, S., Knip, M. & Simell, O. Maturation of insulin response to glucose during human fetal and neonatal development: studies with perifusion of pancreatic isletlike cell clusters. Diabetes. 37(3), 286–291 (1988). [DOI] [PubMed] [Google Scholar]
- 12.Scagilia, L., Chahill, C. J., Finegood, D. & Bonnerweir, S. (eds) Apoptosis is part of the remodeling of the endocrine pancreas in the neonatal rat. Diabetes: Amer Diabetes ASSOC 1660 Duke ST, Alexandria, VA 22314 (1995).
- 13.Kassem, S. A., Ariel, I., Thornton, P. S., Scheimberg, I. & Glaser, B. Beta-cell proliferation and apoptosis in the developing normal human pancreas and in hyperinsulinism of infancy. Diabetes. 49(8), 1325–1333 (2000). [DOI] [PubMed] [Google Scholar]
- 14.Piper, K. et al. Beta cell differentiation during early human pancreas development. J. Endocrinol.181(1), 11–24 (2004). [DOI] [PubMed] [Google Scholar]
- 15.Cole, L., Anderson, M., Antin, P. B. & Limesand, S. W. One process for pancreatic β-cell coalescence into islets involves an epithelial-mesenchymal transition. J. Endocrinol.203(1), 19 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Babu, D. A., Deering, T. G. & Mirmira, R. G. A feat of metabolic proportions: Pdx1 orchestrates islet development and function in the maintenance of glucose homeostasis. Mol. Genet. Metab.92(1–2), 43–55 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Roy, N. et al. PDX1 dynamically regulates pancreatic ductal adenocarcinoma initiation and maintenance. Genes Dev.30(24), 2669–2683 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jonsson, J., Carlsson, L., Edlund, T. & Edlund, H. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature. 371, 606–609 (1994). [DOI] [PubMed] [Google Scholar]
- 19.Offield, M. F. et al. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development. 122(3), 983–995 (1996). [DOI] [PubMed] [Google Scholar]
- 20.Gannon, M., Gamer, L. W. & Wright, C. V. Regulatory regions driving developmental and tissue-specific expression of the essential pancreatic gene pdx1. Dev. Biol.238(1), 185–201 (2001). [DOI] [PubMed] [Google Scholar]
- 21.Sharma, S. et al. Pancreatic islet expression of the Homeobox factor STF-1 relies on an E-box motif that binds USF. J. Biol. Chem.271(4), 2294–2299 (1996). [DOI] [PubMed] [Google Scholar]
- 22.Yang, Y-P., Magnuson, M. A., Stein, R. & Wright, C. V. The mammal-specific Pdx1 area II enhancer has multiple essential functions in early endocrine cell specification and postnatal β-cell maturation. Development. 144(2), 248–257 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cox, A. R. & Kushner, J. A. Area IV knockout reveals how Pdx1 is regulated in postnatal β-cell development. Diabetes. 66(11), 2738–2740 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Spaeth, J. M. et al. Defining a novel role for the Pdx1 transcription factor in islet β-cell maturation and proliferation during weaning. Diabetes. 66(11), 2830–2839 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Haldeman, J. M. et al. Creation of versatile cloning platforms for transgene expression and dCas9-based epigenome editing. Nucleic Acids Res.47(4), e23–e (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Marshak, S. et al. Functional conservation of regulatory elements in the pdx-1 gene: PDX-1 and hepatocyte nuclear factor 3β transcription factors mediate β-cell-specific expression. Mol. Cell. Biol.20(20), 7583–7590 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gerrish, K., Van Velkinburgh, J. C. & Stein, R. Conserved transcriptional regulatory domains of the pdx-1 gene. Mol. Endocrinol.18(3), 533–548 (2004). [DOI] [PubMed] [Google Scholar]
- 28.Gerrish, K. et al. Pancreatic β cell-specific transcription of thepdx-1 gene: The role of conserved Upstream Control regions and their hepatic nuclear factor 3β sites. J. Biol. Chem.275(5), 3485–3492 (2000). [DOI] [PubMed] [Google Scholar]
- 29.Morrison, J. L. et al. Improving pregnancy outcomes in humans through studies in sheep. Am. J. Physiology-Regulatory Integr. Comp. Physiol.315(6), R1123–R53 (2018). [DOI] [PubMed] [Google Scholar]
- 30.Suchy, F., Yamaguchi, T. & Nakauchi, H. iPSC-derived organs in vivo: Challenges and promise. Cell. Stem Cell.22(1), 21–24 (2018). [DOI] [PubMed] [Google Scholar]
- 31.Chiou, J. et al. Single-cell chromatin accessibility identifies pancreatic islet cell type–and state-specific regulatory programs of diabetes risk. Nat. Genet.53(4), 455–466 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Melloul, D., Marshak, S. & Cerasi, E. Regulation of insulin gene transcription. Diabetologia. 45, 309–326 (2002). [DOI] [PubMed] [Google Scholar]
- 33.Jaquiery, A. L. et al. Brief neonatal nutritional supplementation has sex-specific effects on glucose tolerance and insulin regulating genes in juvenile lambs. Pediatr. Res.80(6), 861–869 (2016). [DOI] [PubMed] [Google Scholar]
- 34.Deaton, A. M. & Bird, A. CpG islands and the regulation of transcription. Genes Dev.25(10), 1010–1022 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Han, L., Su, B., Li, W-H. & Zhao, Z. CpG island density and its correlations with genomic features in mammalian genomes. Genome Biol.9, 1–12 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Van Velkinburgh, J. C., Samaras, S. E., Gerrish, K., Artner, I. & Stein, R. Interactions between areas I and II direct pdx-1 expression specifically to islet cell types of the mature and developing pancreas. J. Biol. Chem.280(46), 38438–38444 (2005). [DOI] [PubMed] [Google Scholar]
- 37.Yamada, Y., Tabata, M., Abe, J., Nomura, M. & Harashima, H. In vivo transgene expression in the pancreas by the intraductal injection of naked plasmid DNA. J. Pharm. Sci.107(2), 647–653 (2018). [DOI] [PubMed] [Google Scholar]
- 38.Sreeranjini, A. & Ashok, N. Prenatal development of exocrine pancreas in crossbred goats: A histological study. Int. J. Anat. Res.4(2), 2365–2371 (2016). [Google Scholar]
- 39.Singh, D., Prakash, A., Farooqui, M., Singh, S. & Pathak, S. K. Gross anatomical studies on the pancreas in prenatal Goat (Capra hircus). Int. J. Curr. Microbiol. App Sci.6(2), 823–830 (2017). [Google Scholar]
- 40.Banstola, A. & Reynolds, J. N. The sheep as a large animal model for the investigation and treatment of human disorders. Biology. 11(9), 1251 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jennings, R. E. et al. Development of the human pancreas from foregut to endocrine commitment. Diabetes. 62(10), 3514–3522 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Neiman, D. et al. Islet cells share promoter hypomethylation independently of expression, but exhibit cell-type–specific methylation in enhancers. Proc. Natl. Acad. Sci.114(51), 13525–13530 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yang, B. T. et al. Increased DNA methylation and decreased expression of PDX-1 in pancreatic islets from patients with type 2 diabetes. Mol. Endocrinol.26(7), 1203–1212 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chan, C. S. et al. ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nat. Commun.9(1), 4158 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Park, J. H., Stoffers, D. A., Nicholls, R. D. & Simmons, R. A. Development of type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1. J. Clin. Investig.118(6), 2316–2324 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Green, A. S., Rozance, P. J. & Limesand, S. W. Consequences of a compromised intrauterine environment on islet function. J. Endocrinol.205(3), 211–224 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Massari, M. E. & Murre, C. Helix-loop-helix proteins: Regulators of transcription in eucaryotic organisms. Mol. Cell. Biol.20(2), 429–440 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Amemiya-Kudo, M. et al. Suppression of the pancreatic duodenal homeodomain transcription factor-1 (Pdx-1) promoter by sterol regulatory element-binding protein-1c (SREBP-1c). J. Biol. Chem.286(32), 27902–27914 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Spohrer, S. et al. Functional interplay between the transcription factors USF1 and PDX-1 and protein kinase CK2 in pancreatic β-cells. Sci. Rep.7(1), 1–17 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mantovani, R. A survey of 178 NF-Y binding CCAAT boxes. Nucleic Acids Res.26(5), 1135–1143 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nicolás, M., Noé, V. & Ciudad, C. J. Transcriptional regulation of the human Sp1 gene promoter by the specificity protein (sp) family members nuclear factor Y (NF-Y) and E2F. Biochem. J.371(2), 265–275 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hou, Y. et al. Nuclear factor-Y (NF-Y) regulates transcription of mouse Dmrt7 gene by binding to tandem CCAAT boxes in its proximal promoter. Int. J. Biol. Sci.6(7), 655 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bernadt, C. T., Nowling, T., Wiebe, M. S. & Rizzino, A. NF-Y behaves as a bifunctional transcription factor that can stimulate or repress the FGF-4 promoter in an enhancer-dependent manner. Gene Expression J. Liver Res.12(3), 193–212 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Perna, D. et al. Genome-wide mapping of myc binding and gene regulation in serum-stimulated fibroblasts. Oncogene. 31(13), 1695–1709 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gao, T. et al. Pdx1 maintains β cell identity and function by repressing an α cell program. Cell Metabol.19(2), 259–271 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhu, X. et al. Cell cycle regulation of the Pdx1 transcription factor in developing pancreas and insulin-producing β-cells. Diabetes. 70(4), 903–916 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Johnson, J. D. et al. Increased islet apoptosis in Pdx1+/–mice. J. Clin. Investig.111(8), 1147–1160 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Maricque, B. B., Chaudhari, H. G. & Cohen, B. A. A massively parallel reporter assay dissects the influence of chromatin structure on cis-regulatory activity. Nat. Biotechnol.37(1), 90–95 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Geusz, R. J. et al. Sequence logic at enhancers governs a dual mechanism of endodermal organ fate induction by FOXA pioneer factors. Nat. Commun.12(1), 6636 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ebrahim, N., Shakirova, K. & Dashinimaev, E. PDX1 is the cornerstone of pancreatic β-cell functions and identity. Front. Mol. Biosci.9. (2022). [DOI] [PMC free article] [PubMed]
- 61.Wiebe, P. O. et al. Ptf1a binds to and activates area III, a highly conserved region of the Pdx1 promoter that mediates early pancreas-wide Pdx1 expression. Mol. Cell. Biol.27(11), 4093–4104 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Salguero-Aranda, C. et al. Pdx1 is transcriptionally regulated by EGR-1 during nitric Oxide-Induced endoderm differentiation of mouse embryonic stem cells. Int. J. Mol. Sci.23(7), 3920 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lee, K. et al. FOXA2 is required for enhancer priming during pancreatic differentiation. Cell. Rep.28(2), 382–393 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang, A. et al. Epigenetic priming of enhancers predicts developmental competence of hESC-derived endodermal lineage intermediates. Cell. stem cell.16(4), 386–399 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Boyer, D. F. et al. Complementation rescue of Pdx1 null phenotype demonstrates distinct roles of proximal and distal cis-regulatory sequences in pancreatic and duodenal expression. Dev. Biol.298(2), 616–631 (2006). [DOI] [PubMed] [Google Scholar]
- 66.Wortham, M. & Sander, M. Transcriptional mechanisms of pancreatic β-cell maturation and functional adaptation. Trends Endocrinol. Metab.32(7), 474–487 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Segert, J. A. G. S. & Bulyk, M. L. Transcriptional silencers: Driving Gene expression with the brakes on. Trends Genet.37(6), 514–527. 10.1016/j.tig.2021.02.002 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Akerman, I. et al. Human pancreatic β cell lncRNAs control cell-specific regulatory networks. Cell Metabol.25(2), 400–411 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dias, I. E. et al. Mesenchymal stem cell studies in the goat model for biomedical research—a review of the scientific literature. Biology. 11(9), 1276 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Li, R. et al. A near complete genome for goat genetic and genomic research. Genet. Selection Evol.53(1), 1–17 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Cozar-Castellano, I. et al. Lessons from the first comprehensive molecular characterization of cell cycle control in rodent insulinoma cell lines. Diabetes. 57(11), 3056–3068 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Calabrese, A. et al. Connexin 36 controls synchronization of Ca2+ oscillations and insulin secretion in MIN6 cells. Diabetes. 52(2), 417–424 (2003). [DOI] [PubMed] [Google Scholar]
- 73.Drzazga, A., Cichońska, E., Koziołkiewicz, M. & Gendaszewska-Darmach, E. Formation of βTC3 and MIN6 pseudoislets changes the expression pattern of Gpr40, Gpr55, and Gpr119 receptors and improves lysophosphatidylcholines-potentiated glucose-stimulated insulin secretion. Cells. 9(9), 2062 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Dadheech, N. et al. Basal expression of pluripotency-associated genes can contribute to stemness property and differentiation potential. Stem Cells Dev.22(12), 1802–1817 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Frazer, K. A., Pachter, L., Poliakov, A., Rubin, E. M. & Dubchak, I. VISTA: Computational tools for comparative genomics. Nucleic Acids Res.32(suppl_2), W273–W9 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Cartharius, K. et al. MatInspector and beyond: Promoter analysis based on transcription factor binding sites. Bioinformatics. 21(13), 2933–2942 (2005). [DOI] [PubMed] [Google Scholar]
- 77.Li, L-C. & Dahiya, R. MethPrimer: designing primers for methylation PCRs. Bioinformatics. 18(11), 1427–1431 (2002). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data generated or analyzed during this study are included in this published article, its supplementary information files, and publicly available repositories. The datasets generated during the current study are also available in the GenBank repository with the accession numbers of PQ066235, PQ066236 and PQ066237.





