The gastrointestinal (GI) tract is complex and consists of multiple organs with unique functions. Rare gene variants can cause congenital malformations of the human GI tract, although the molecular basis of these has been poorly studied. We identified a patient with compound-heterozygous variants in RFX6 presenting with duodenal malrotation and atresia, implicating RFX6 in development of the proximal intestine. To identify how mutations in RFX6 impact intestinal patterning and function, we derived induced pluripotent stem cells from this patient to generate human intestinal organoids (HIOs). We identified that the duodenal HIOs and human tissues had mixed regional identity, with gastric and ileal features. CRISPR-mediated correction of RFX6 restored duodenal identity. We then used gain- and loss-of-function and transcriptomic approaches in HIOs and Xenopus embryos to identify that PDX1 is a downstream transcriptional target of RFX6 required for duodenal development. However, RFX6 had additional PDX1-independent transcriptional targets involving multiple components of signaling pathways that are required for establishing early regional identity in the GI tract. In summary, we have identified RFX6 as a key regulator in intestinal patterning that acts by regulating transcriptional and signaling pathways.

Deciphering the factors that control regional patterning during endoderm organogenesis remains a major area of investigation in developmental biology (Thompson et al., 2018; Spence et al., 2011a). The mechanisms that drive specific cell migration and identity require specialized gene regulatory networks at precise times, which are regulated by a combination of signaling pathways and transcription factors (TFs), to give rise to diverse organs. Several major pathways, such as WNT, BMP and Hedgehog, have been shown to have a crucial role in the patterning and formation of endoderm organs (Zorn and Wells, 2007, 2009; Roberts et al., 1995; Thompson et al., 2018). TFs are often responsible for driving organ-specific gene regulatory networks and their loss can result in organ agenesis or abnormal patterning. Regulatory factor X6 (RFX6) is a winged-helix TF that has been shown to have an essential role in endoderm organogenesis and is required for endocrine cell formation in pancreatic islets and enteroendocrine cell (EEC) differentiation in the gastrointestinal (GI) tract (Smith et al., 2010; Piccand et al., 2014). Early in embryonic development, RFX6 is broadly expressed in the epithelium of the foregut and proximal intestine, and becomes restricted to the endocrine pancreas and the proximal small intestine at later stages of development. Reduction or loss of Rfx6 in mouse intestine results in gut malrotation, duodenal atresia, malabsorption, and significant reduction of the enteroendocrine lineage (Soyer et al., 2010; Gehart et al., 2019). In humans, variants of RFX6 result in Mitchell–Riley syndrome, an autosomal-recessive syndrome characterized by neonatal diabetes, small bowel atresia, and malabsorption (Kambal et al., 2019; Concepcion et al., 2014; Trott et al., 2020). Recent studies using an induced pluripotent stem cell (iPSC) line derived from an individual with a similar variant of RFX6 showed reduced pancreatic differentiation (Trott et al., 2020), consistent with previous functional studies showing that Rfx6 is involved in normal pancreas development in mouse (Piccand et al., 2014). However, the molecular mechanisms by which RFX6 regulates the patterning and function of the regions of the human intestine remain unknown.

Another TF that has a crucial role in the development of the pancreas and proximal small intestine is pancreas/duodenal homeobox 1 (PDX1). It has been shown that RFX6 and PDX1 are co-expressed in the developing duodenum and endocrine cells in pancreas and small intestine (Soyer et al., 2010; Smith et al., 2010; Yang et al., 2017). PDX1 has an essential role in pancreas, stomach and duodenum morphogenesis and persists in the adult tissues; Pdx1-null mouse models have shown a lack of mature pancreatic tissue and malformation of the gastroduodenal junction with significantly reduced endocrine cells in the small intestine (Offield et al., 1996; Boyer et al., 2006; Burlison et al., 2008; Chen et al., 2009; Fujita et al., 2008; Fujitani et al., 2006a; Stoffers et al., 1997). In the intestinal tract, PDX1 is maximally expressed in the anterior duodenal region with decreased expression in the distal small intestine. Additionally, PDX1 has been reported to be necessary for the production of gastric inhibitory peptide (GIP)-secreting cells, although multiple cell lineages are lost in PDX1-null models (Chen et al., 2009; Jepeal et al., 2005). Despite an essential role for PDX1 in regulating patterning and development of the duodenum, the factors that regulate PDX1 expression are not known.

Over the past decade, several human organoid models have been developed that enable novel strategies to model diseases modeling and organogenesis (Kechele and Wells, 2019). iPSC-derived organoids are three-dimensional structures that mimic the development of an organ and offer a unique way to study developmental mechanisms and organ functions in human tissue (Carpenter and Rao, 2015). Organoids also possess the unique advantage of being generated from iPSCs, meaning they exhibit the same variant as the individual from whom they are derived, allowing for the study of individual-specific phenotypes at different stages of development. iPSC-derived intestinal organoids (HIOs) can be differentiated in vitro and engrafted under the mouse kidney capsule where they become vascularized and mature to mimic fetal intestine with crypt-villus architecture and the ability to absorb nutrients (Spence et al., 2011b; Watson et al., 2014). We recently used these iPSC-derived human organoids to identify specific pathologies in individuals with a PDX1 variant (Krishnamurthy et al., 2022).

We have identified a patient with a compound-heterozygous variant of RFX6 that caused neonatal diabetes and duodenal malrotation and atresia, resulting in functional loss of the RFX6 protein and abnormal gut patterning. We generated iPSCs and HIOs from this patient to interrogate further the specific roles of RFX6 during intestinal development. This approach allowed us to compare directly the biopsy samples of this patient with the HIOs to discover pathophysiological effects of this variant. Here, we demonstrate that RFX6 is required to maintain duodenal identity and function, and that its loss results in ileal-like tissue. We describe the mechanism by which RFX6 drives PDX1 expression, which activates the gene regulatory network that establishes and maintains duodenal identity. Finally, we largely rescue the phenotypes caused by the RFX6 variants using both CRISPR allele correction and inducible re-introduction of RFX6 or downstream PDX1. This study represents a novel approach to the use of complex individual pathologies to uncover the mechanisms at play in human development.

RFX6 is expressed in the human small intestine and its loss of function leads to gut mispatterning

Several signaling pathways and TFs converge to pattern the small intestine into duodenum, jejunum and ileum. These segments carry out different functions during nutrient absorption, but, importantly, also employ differential gene regulatory networks to generate their identities (Haber et al., 2017). One key TF is the winged helix factor RFX6. Early in development, RFX6 is expressed widely throughout the endoderm but becomes restricted to proximal small intestine and pancreas after embryonic day (E) 13.5 in mouse (Soyer et al., 2010; Smith et al., 2010). A patient presented to Cincinnati Children's Hospital Medical Center (CCHMC) with monogenic diabetes, duodenal atresia, intestinal malrotation, annular pancreas and polyps in the duodenum that were surgically removed for analysis. Exome sequencing of the patient and their parents revealed that this patient carried a compound-heterozygous variant of RFX6, with a frameshift mutation (p. Arg347Lysfs*18) in the paternal allele and a nonsense mutation (p. Gln875*) in the maternal allele (Fig. 1A). Analysis of resected polyps showed them to be gastric in identity expressing the stomach marker CLDN18 with only small regions expressing the intestinal marker CDH17 (Fig. 1B). This developmental phenotype suggested that early stages of organ development were impacted, especially the patterning of the gut tube. However, limited access to human samples has made it challenging to perform deep mechanistic investigation into the cause of these phenotypes. To perform deep phenotyping and investigate the developmental basis of these congenital malformations, we recruited this patient into our study and acquired a blood sample for generation of iPSCs. Analysis of these lines showed them to be karyotypically normal, pathogen free, genetically unique as measured by short tandem repeat analyses, and able to differentiate into definitive endoderm and form early gut spheroids (Fig. S1).

Fig. 1.

RFX6 variants cause abnormal gut patterning and loss of endocrine cells in the intestine. (A) Top: Diagram of the variants in paternal and maternal alleles compared with WT. Bottom: Endoscopy image of polyps (arrows) in the duodenum. Biopsies were taken from polyps and healthy tissue from the same patient. Polyps are used to highlight abnormal patterning. (B) Immunostaining of RFX6 Mut biopsy showing gastric metaplasia (CLDN18), PDX1, SATB2 and MUC2 changes and loss of EECs (CHGA) compared with duodenal biopsy from a visually normal region of the duodenum of the same individual (Ctrl Patient). (C) Expression of RFX6 (green) in duodenal (Duod) and ileal (Ile) HIOs in vitro and in vivo. (D) RFX6 mRNA expression in WT and RFX6 Mut HIOs (n=3). ****P<0.0001 (unpaired t-test). (E) Immunostaining of duodenal and ileal HIOs from RFX6 Mut iPSCs and WT duodenal (Duo) and ileal (Ile) HIO showing duodenum phenotypes, whereas the ileum is normal (excluding loss of EECs). (F) Quantification of HIO staining for patterning and secretory markers (n=3). Significance determined by unpaired t-test with *P<0.05, **P<0.01, ***P<0.001. ns, not significant. Scale bars: 100 μm.

Fig. 1.

RFX6 variants cause abnormal gut patterning and loss of endocrine cells in the intestine. (A) Top: Diagram of the variants in paternal and maternal alleles compared with WT. Bottom: Endoscopy image of polyps (arrows) in the duodenum. Biopsies were taken from polyps and healthy tissue from the same patient. Polyps are used to highlight abnormal patterning. (B) Immunostaining of RFX6 Mut biopsy showing gastric metaplasia (CLDN18), PDX1, SATB2 and MUC2 changes and loss of EECs (CHGA) compared with duodenal biopsy from a visually normal region of the duodenum of the same individual (Ctrl Patient). (C) Expression of RFX6 (green) in duodenal (Duod) and ileal (Ile) HIOs in vitro and in vivo. (D) RFX6 mRNA expression in WT and RFX6 Mut HIOs (n=3). ****P<0.0001 (unpaired t-test). (E) Immunostaining of duodenal and ileal HIOs from RFX6 Mut iPSCs and WT duodenal (Duo) and ileal (Ile) HIO showing duodenum phenotypes, whereas the ileum is normal (excluding loss of EECs). (F) Quantification of HIO staining for patterning and secretory markers (n=3). Significance determined by unpaired t-test with *P<0.05, **P<0.01, ***P<0.001. ns, not significant. Scale bars: 100 μm.

Close modal

We first determined whether we could phenocopy the pathologies of this patient with intestinal (duodenal) organoids generated from their iPSC lines. We first generated HIOs in vitro, at which stage they represent first trimester human duodenum (Spence et al., 2011b). To promote further development and maturation, HIOs were transplanted into immunocompromised mice and allowed to develop for an additional 12 weeks, at which time they are similar to early third trimester human duodenum (Fig. S2A) (Singh et al., 2023). Analysis of HIOs generated from a control (wild-type, WT) iPSC line confirmed them to be duodenal in nature, as we previously reported (Spence et al., 2011b), expressing PDX1 and CDX2 with little-to-no expression of gastric markers (Fig. S2B-E). We found that RFX6 protein was expressed in duodenal HIOs in vitro and those matured in vivo (Fig. 1C, Fig. S3A-D). In contrast, HIOs made from this patient's iPSCs with RFX6 mutations (RFX6 Mut) had low RFX6 expression (Fig. 1D). RFX6 Mut HIOs had large patches of gastric tissue expressing CLDN18, reminiscent of the gastric polyps found in the patient. Further phenotyping of RFX6 Mut HIOs showed a wide range of abnormalities, including increased goblet cells and a decrease in EECs with loss of many subtypes (Fig. 1B,E, Fig. S4A). Interestingly, the HIOs exhibited a loss of PDX1 expression, which is a key driver of the duodenal gene regulatory network (Boyer et al., 2006; Chen et al., 2009). Furthermore, duodenal HIOs had inappropriate expression of the distal intestinal marker SATB2, a TF that has been recently shown to be a driver of ileal-colonic fate (Fig. 1E,F) (Gu et al., 2022). As a control for this regional phenotype, we generated distal HIOs that are ileal in nature by addition of FGF and CHIR99021 which further pattern the developing hindgut (Tsai et al., 2017; Dessimoz et al., 2006). RFX6 is not normally expressed in the distal small intestine, and, as predicted, distal small intestinal organoids were unaffected by the RFX6 variant (Fig. 1E,F).

Having identified multiple previously unappreciated pathologies in RFX6 Mut HIOs, we wanted to investigate whether these were similarly found in tissue from the patient. Given the pathologic nature of the duodenal polyps, we chose to histologically compare HIOs to a biopsy from a visually normal region of the duodenum from this patient. In all cases, the HIO phenotypes were also observed in the biopsy sample, including reduced PDX1, increased SATB2, increased goblet cells, and a reduction in most EEC populations (Fig. 1E,F, Fig. S4B-D). As expected from our immunofluorescence analysis, expression of markers notably reduced in RFX6 Mut duodenal HIOs included several proximally enriched EEC-peptides, such as GIP, ghrelin and motilin (Fig. S4C), suggesting RFX6 has an important role in the differentiation of intestinal EECs in humans (Piccand et al., 2019). This confirmed previous murine studies that had reported loss of EECs in Rfx6-null mice (Gehart et al., 2019; Piccand et al., 2019). These data suggest that RFX6 variants cause mispatterning of the developing GI tract, with loss of duodenal identity and inappropriate expression of distal markers.

Transcriptomic analysis reveals abnormal patterning and loss of duodenal function in mature RFX6 Mut organoids

To investigate more comprehensively the impact of the RFX6 variants on intestinal development, we isolated tissue from transplanted HIOs for RNA sequencing (RNA-seq). These transplanted organoids often display some degree of heterogeneity because they are each grown in a different animal with immunocompromised health; additionally, the different background in the parental line from a healthy to a mutant iPSC line can add some variability. Thus, we mainly focused on genes that were differentially expressed after pooling the replicates together. As a baseline, we determined that in vitro Mut HIOs (35D, i.e. 35 days of culture of the HIO, which is the endpoint in vitro and the time at which they are transplanted into mouse) consistently display intestinal markers (e.g. CDX2), but also exhibit phenotypes seen in biopsies, such as loss of PDX1 and increased expression of SATB2 compared with WT HIOs (Fig. S5A). To establish further the variance pre-transplant, we generated RNA-seq data from in vitro HIOs; principal component analysis (PCA) of transcriptomes from WT and RFX6 Mut duodenal 35D HIOs showed 85% variance between WT and Mut whereas between replicates there was only 6% variance. Additionally, the heatmap showed over 2000 differentially expressed genes (Fig. S5B). In addition, we found altered expression of HOX genes in our in vivo HIOs that are known to regulate downstream targets involved in anterior-posterior (AP) patterning in developing embryos of species ranging from flies to humans (Fig. S5C) (Hajirnis and Mishra, 2021). In the GI tract, HOX genes are expressed in the mesenchyme, the epithelium or both, and follow a proximal-to-distal gradient going from the duodenum to the colon. In RFX6 Mut duodenal HIOs, distal HOX genes, such as HOXC10, HOXA10 and HOXA11, were increased in expression compared with WT duodenal HIOs, suggesting that the duodenum of the patient with the RFX6 variant exhibits more ileal identity. However, known proximal HOX genes did not decrease in RFX6 Mut duodenal HIOs, suggesting that there is a mix of duodenal and ileal-like gene expression patterns in RFX6 Mut HIOs. We analyzed additional mesenchymal markers in our in vivo HIOs to determine whether RFX6 variants impact mesenchyme and showed significantly decreased expression of HAND1 and FOXF1, which have been shown to expressed in the duodenal mesenchyme at these stages of gut tube development in mice (Fig. S5D) (Han et al., 2020).

Gene ontology (GO) term analysis of all the genes downregulated in RFX6 Mut duodenal HIOs revealed loss of biological processes associated with duodenal-specific functions (Fig. 2B). For example, the duodenum is a key segment of the small intestine for lipid uptake and metabolism (DiPatrizio and Piomelli, 2015). RFX6 Mut duodenal HIOs showed decreased expression of MTTP, APOC3 and RBP2, which are involved in lipid metabolism processes, compared with WT duodenal HIOs (Fig. 2A) (Iqbal and Hussain, 2009; Hussain, 2014). We observed a trend toward increased expression of genes involved in ileal-specific functions, such as bile acid absorption and vitamin B12 processing, although they were not significant (Fig. 2A) (Ticho et al., 2019; Thompson and Wrathell, 1977). Given that RFX6 variants impacted the expression of lipid-handling genes, we investigated whether RFX6 Mut HIOs had impaired lipid transport. We exposed the lumen of transplanted organoids to the fluorescent lipid BODIPY and quantified lipid uptake (Carten et al, 2011). We first compared BODIPY uptake in WT duodenal HIOs compared with WT ileal HIOs and, as expected, observed robust uptake of BODIPY into the epithelium of duodenal, but not WT ileal, HIOs. In contrast, RFX6 Mut duodenal HIOs did not efficiently absorb lipids (Fig. 2C). Quantification of lipid uptake showed a two- to threefold reduction in RFX6 Mut duodenal HIOs compared with control HIOs. Taken together, these data suggest that the RFX6 variant affects AP patterning during development of the duodenum, resulting in reduction of proximal small intestine functions, such as lipid absorption, and an enrichment of an ileal signature.

Fig. 2.

Transcriptomic analysis of RFX6 Mut HIOs shows loss of duodenal function and patterning and can be corrected with one WT allele. (A) Genes involved in duodenal functions are downregulated in RFX6 Mut HIOs, whereas genes involved in ileal functions remain unchanged. Results obtained from independent transplanted organoids (n=2). Two HIOs were harvested and samples of ∼2×2 mm were taken from each HIO. Three samples were used for this plot. TPMs, transcripts per million. (B) GO results for Biological Processes by genes significantly downregulated in RFX6 Mut HIOs compared with WT. (C) Fluorescently labeled lipid (BODIPY) uptake in WT compared with RFX6 Mut HIOs (n=3). The organoid was embedded for sectioning and imaged for fluorescence in the epithelium. For C and E, HIOs were used for analysis and a total of five sections were quantified. Diagram created with BioRender.com. (D) Schematic of the proteins encoded by the WT RFX6 allele, the compound-heterozygous variant RFX6 allele and the CRISPR-corrected RFX6 allele. (E) Staining of the CRISPR-corrected and RFX6 Mut compared with WT HIOs showing patterning markers returning to normal and recovery of the EEC population (n=3). Significance determined by unpaired t-test with *P<0.05, **P<0.01, ***P<0.001. ns, not significant. t-test on n=2 done using GraphPad Prism, after DeSeq analysis identifying significant differentially expressed genes. Significant differentially expressed genes from bulk RNA-seq were defined by an adjusted P-value of <0.05. Scale bars: 100 μm.

Fig. 2.

Transcriptomic analysis of RFX6 Mut HIOs shows loss of duodenal function and patterning and can be corrected with one WT allele. (A) Genes involved in duodenal functions are downregulated in RFX6 Mut HIOs, whereas genes involved in ileal functions remain unchanged. Results obtained from independent transplanted organoids (n=2). Two HIOs were harvested and samples of ∼2×2 mm were taken from each HIO. Three samples were used for this plot. TPMs, transcripts per million. (B) GO results for Biological Processes by genes significantly downregulated in RFX6 Mut HIOs compared with WT. (C) Fluorescently labeled lipid (BODIPY) uptake in WT compared with RFX6 Mut HIOs (n=3). The organoid was embedded for sectioning and imaged for fluorescence in the epithelium. For C and E, HIOs were used for analysis and a total of five sections were quantified. Diagram created with BioRender.com. (D) Schematic of the proteins encoded by the WT RFX6 allele, the compound-heterozygous variant RFX6 allele and the CRISPR-corrected RFX6 allele. (E) Staining of the CRISPR-corrected and RFX6 Mut compared with WT HIOs showing patterning markers returning to normal and recovery of the EEC population (n=3). Significance determined by unpaired t-test with *P<0.05, **P<0.01, ***P<0.001. ns, not significant. t-test on n=2 done using GraphPad Prism, after DeSeq analysis identifying significant differentially expressed genes. Significant differentially expressed genes from bulk RNA-seq were defined by an adjusted P-value of <0.05. Scale bars: 100 μm.

Close modal

Finally, we investigated whether expression of jejunal-specific genes were altered in RFX6 Mut organoids. The jejunum-specific transporters SLC2A2, SLC2A5 and SLC34A2 (Anderle et al., 2005), which participate in glucose, fructose and phosphate uptake, were not expressed in WT or Mut HIOs. This could be because HIOs are still fetal even after maturation in vivo, and many transporters are not expressed until exposed to nutrients, which happens postnatally and leads to the functional differences between the segments of the small intestine (Schubert, 2011; Lema et al., 2020).

CRISPR correction of one allele was sufficient to restore a more duodenal phenotype in Rfx6 Mut organoids

The patient in this study carries compound-heterozygous variants in both parental alleles, with the paternal mutation (p. Arg347Lysfs*18) introducing a premature stop codon that deletes 563 amino acids from RFX6. The maternal allele is a nonsense mutation that deletes 52 amino acids. Because the patient's parents did not present with clinical manifestation of RFX6 deficiency, we reasoned that conversion of one mutant allele to a WT allele would be sufficient to restore a more normal duodenal phenotype. Previous reports of RFX6 variants have shown that the larger the deletion, the more severe the symptoms (Kambal et al., 2019). We therefore used CRISPR/Cas9 to convert the paternal mutation into a WT RFX6 allele (Fig. 2D) (Haeussler et al., 2016; Ran et al., 2013; Liang et al., 2017). RNA-seq analysis of the transplanted HIOs revealed that the corrected duodenal HIOs were much more similar to the WT duodenal HIOs than to RFX6 Mut duodenal HIOs (Fig. S5E,F). PCA showed that the primary difference caused by RFX6 variants (PC1;76%) had been completely reversed by CRISPR correction of the paternal allele. However, PC2 showed that there were still differences between control lines and CRISPR-corrected lines. For example, several genes and biological processes specific to the proximal small intestine showed different levels of expression between the WT and corrected HIOs. This could be due to the different genetic backgrounds of the WT control line, or due to reduced RFX6 activity as a result of the maternal variant.

A closer analysis of specific factors that regulate duodenal patterning showed that expression levels of PDX1, GATA4 and several regionally expressed HOX genes was restored in CRISPR-corrected HIOs (Fig. S5G). We also saw restored expression of genes involved in duodenal function, including lipid metabolism (Fig. S5G). CRISPR-corrected duodenal HIOs did not exhibit evidence of gastric polyps, had significantly decreased expression of the ileal-colonic marker SATB2, and goblet cell numbers more comparable to WT duodenal organoids (Fig. 2E). Together, these data show that the rescue of the paternal allele significantly restored a duodenal phenotype to RFX6 compound-heterozygous HIOs, suggesting that RFX6 has a crucial role in establishing duodenal identity and function. However, the expression of some genes was not restored to the levels of control HIOs generated from iPSC lines of different genetic background. This could be due to haploinsufficiency or gene expression variability between organoids generated from diverse genetic backgrounds.

Initiation of duodenal patterning and activity of patterning pathways is deranged in RFX6 Mut endoderm

The AP regional identity of the developing gut tube is established at early stages of development. To investigate the mechanisms leading to the abnormal patterning in RFX6 Mut intestine, we performed RNA-seq analysis during the gut tube patterning stage of differentiation (D7) (Fig. 3A) (Spence et al., 2011a,b). This stage resembles ∼E8.5-9.5 in mice, at which point Rfx6 is already expressed (Soyer et al., 2010; Smith et al., 2010). We first confirmed that RFX6 expression in human cultures initiates in this window of time and found that expression starts at D5 and increases through D7 of differentiation (Fig. S3C); PCA analysis of RNA-seq data displayed a 94% variance between WT and RFX6 Mut and over 4000 differentially expressed genes (Fig. S6A). Most of the differentially expressed genes were downregulated in the RFX6 Mut line, suggesting a failure to activate an RFX6-mediated transcriptional program (Fig. S6A). Lineage-tracing experiments in mice have shown that Rfx6 expression is restricted to the epithelium at this stage of development (Smith et al., 2010). Consistent with this, we observed that RNA counts of crucial endodermal patterning regulators, such as CDX2, GATA4, GATA6, HNF4A, were significantly reduced in the D7 gut tube endoderm cultures from RFX6 Mut iPSC lines (Fig. 3B). Consistent with our observation that the patient duodenum had areas of mixed organ identity, we used a list of organ markers from a published atlas and found that RFX6 Mut cultures lost small intestinal organ markers (Fig. 3C) (Yu et al., 2021).

Fig. 3.

RFX6 Mut D7 HIOs show misregulation of signaling pathways and early hindgut patterning, corrected with one WT allele. (A) Schematic of organoid generation and harvesting time point for bulk RNA-seq. Created with BioRender.com. (B) TFs involved in mid/hindgut patterning are also affected in RFX6 Mut D7 HIOs based on RNA counts (n=4). RNA was isolated from approximately ten D7 spheroids from separate wells (n=4) for RNA-seq. (C) Organ markers from various parts of the GI system showing intestinal markers are downregulated in RFX6 Mut HIOs based on RNA counts (n=4) (Yu et al., 2021). (D) Changes in components of relevant signaling pathways that are affected in RFX6 Mut D7 HIOs (Wnt, BMP and HH) based on RNA counts (n=4). (E) PCA of WT versus RFX6 Mut and CRISPR-corrected D7 HIOs. Venn diagram showing that the percentage of overlap with genes upregulated by CRISPR correction was 18.1%. (F) Upregulated components of several signaling pathways are increased in CRISPR-corrected D7 HIOs based on RNA counts (n=4). (G) Patterning markers return to levels closer to control in CRISPR-corrected D7 HIOs based on RNA counts (n=4). Significance determined by unpaired t-test with *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001. ns, not significant. Significant differentially expressed genes from bulk RNA-seq was defined by an adjusted P-value of <0.05.

Fig. 3.

RFX6 Mut D7 HIOs show misregulation of signaling pathways and early hindgut patterning, corrected with one WT allele. (A) Schematic of organoid generation and harvesting time point for bulk RNA-seq. Created with BioRender.com. (B) TFs involved in mid/hindgut patterning are also affected in RFX6 Mut D7 HIOs based on RNA counts (n=4). RNA was isolated from approximately ten D7 spheroids from separate wells (n=4) for RNA-seq. (C) Organ markers from various parts of the GI system showing intestinal markers are downregulated in RFX6 Mut HIOs based on RNA counts (n=4) (Yu et al., 2021). (D) Changes in components of relevant signaling pathways that are affected in RFX6 Mut D7 HIOs (Wnt, BMP and HH) based on RNA counts (n=4). (E) PCA of WT versus RFX6 Mut and CRISPR-corrected D7 HIOs. Venn diagram showing that the percentage of overlap with genes upregulated by CRISPR correction was 18.1%. (F) Upregulated components of several signaling pathways are increased in CRISPR-corrected D7 HIOs based on RNA counts (n=4). (G) Patterning markers return to levels closer to control in CRISPR-corrected D7 HIOs based on RNA counts (n=4). Significance determined by unpaired t-test with *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001. ns, not significant. Significant differentially expressed genes from bulk RNA-seq was defined by an adjusted P-value of <0.05.

Close modal

In addition to epithelial patterning defects, we observed a surprising number of expression differences in key signaling factors that regulate endodermal patterning through a series of epithelial–mesenchymal interactions (Zorn and Wells, 2007; Walton and Gumucio, 2021; McCarthy et al., 2020) (Fig. 3D). In particular, WNT3, WNT5B, BMP4, IHH and many downstream targets of these pathways were reduced in RFX6 Mut cultures. Given that WNT signaling is involved in the expression of CDX2, the master regulator of intestinal identity (Sherwood et al., 2011; Chawengsaksophak et al., 1997), it is likely that reduced WNT signaling in D7 cultures causes a delay in CDX2 expression. Known CDX2 targets that are involved in early intestinal development (Kumar et al., 2019) were also reduced in the RFX6 Mut line, suggesting that the abnormal patterning is due to low CDX2 expression (Fig. S6B).

Although RFX6 is expressed in the epithelium, RFX6 variants cause patterning changes in both the epithelium and mesenchyme of D7 cultures. To begin to separate the direct effects of the RFX6 variants in the epithelium and indirect effects on the mesenchyme, we looked at published dataset from Han et al. which mapped endoderm versus mesoderm specific markers in early gut development (Han et al., 2020). We identified endoderm- and mesoderm-specific markers and performed GO analysis. Interestingly, the biological processes that were impacted in our RFX6 Mut cultures included intestinal patterning, epithelium development, tube development and morphogenesis (Fig. S6C,D). Together, these data suggest that there is an initial delay of intestinal patterning as observed by low CDX2 expression in RFX6 Mut cultures. However, at later stages of differentiation, CDX2 is expressed in both control and RFX6 Mut HIOs and at those stages duodenal markers are lost and more distal markers are expressed.

Finally, we investigated whether the CRISPR correction of the paternal allele rescued the patterning of the HIOs at D7 using PCA of RNA-seq data. CRISPR correction restored the PC1 component (76% variance) to nearly that of WT, suggesting that correction of one allele allows transcription of many downstream targets (Fig. 3E). There was, however, an 18% variance in PC2 from the WT and the corrected group, due to their background difference. Transcripts that were restored in the CRISPR-corrected cultures included components of the WNT and BMP pathways known to regulate gut tube patterning (Fig. 3F). When comparing the CRISPR-corrected line with both the Mut and WT there were also differentially expressed genes (Fig. 3E, Fig. S6E,F). Lastly, we found that correction of one RFX6 allele was sufficient to rescue the expression of several key TFs involved in hindgut development and patterning, such as CDX2 and HNF4A (Fig. 3G). Taken together, these data suggest that, although RFX6 expression is specific to the epithelium, it functions to regulate patterning and signaling in the epithelium. Additionally, CRISPR correction of the paternal allele is sufficient to restore duodenal patterning in organoids; however, there remain transcriptional differences in organoids that retain the maternal variant allele. This suggests that the maternal allele may not be fully functional.

An evolutionarily conserved role of RFX6 in GI tract patterning and functional characterization of RFX6 variants

During pancreas development, the PDX1 gene is bound by RFX6 protein, suggesting that this gene may be directly regulated by RFX6. In the duodenum, CRISPR correction of one allele in the RFX6 Mut line restored expression of many TF genes, in particular PDX1, which is essential for duodenal patterning (Fig. 4A). We further developed a doxycycline (DOX)-inducible PSC line in which we can temporally induce RFX6 expression in the mutant background. Induction of RFX6 in foregut endoderm was sufficient to induce endogenous PDX1 mRNA and protein expression (Fig. 4A, Fig. S7A-C). Although these findings showed that RFX6 is both necessary and sufficient to regulate PDX1 expression in cultured human foregut endoderm, we wanted to perform similar loss- and gain-of-function experiments in the context of an embryo. To do this, we used the amphibian Xenopus, a vertebrate model in which endoderm development and GI tract organogenesis is highly conserved (Zorn and Wells, 2009; Rankin et al., 2021). Xenopus rfx6 shares a similar expression pattern as observed in mammals (Pearl et al, 2011; Smith et al., 2010), which we verified by in situ hybridization in foregut progenitors at Nieuwkoop and Faber stage (NF) 20 [∼1 day post-fertilization (d.p.f.)] and in pancreas, stomach and duodenum progenitors at NF33 (∼2.5 d.p.f.) (Fig. S8A). Knockdown of rfx6 in Xenopus using a previously validated translation-blocking morpholino (MO; Pearl et al., 2011). We tested the efficiency of the MO knockdown by titrating the MO for its ability to block a tagged Rfx6 protein (FLAG tagged) and showed immunostaining of the FLAG-tagged protein at NF20 (Fig. S8B,C). This MO revealed a loss of pdx1 at NF33 and microdissection of the gut tube at NF43 (∼4 d.p.f.) revealed a hypoplastic, malrotated GI tract with strong reduction of the pdx1 stomach/duodenum domain, a phenotype similar to that of the mouse Rfx6−/− knockout (Fig. 4B, Fig. S8D,E) (Smith et al., 2010). Interestingly, we observed an expansion of the distal hindgut satb2+ domain into the midgut and foregut territories at NF33, suggesting an evolutionarily conserved function for rfx6 in regulating vertebrate GI tract patterning (Fig. 4B). Injection of mRNA encoding human RFX6 was able to rescue these molecular changes at NF33 and NF43 (Fig. 4B).

Fig. 4.

An evolutionarily conserved role of RFX6 in GI tract patterning and functional characterization of RFX6 variants. (A) RT-PCR expression of PDX1 when after correction of one allele of RFX6 in 35D HIOs and PDX1 expression when RFX6 has been induced by DOX in hindgut monolayer (n=3). **P<0.01, ***P<0.001, ****P<0.0001 (unpaired t-test). (B) In situ hybridization of pdx1 and satb2 with control MO and rfx6 MO blocking translation in stage NF33 (2.5 d.p.f.) Xenopus embryos. Numbers in the lower left corner of panel indicate the number of embryos assayed. dp, dorsal pancreas; duo, duodenum; gb, gall bladder; hg, hindgut; st, stomach; vp, ventral pancreas. (C-E) Experimental schematic and human RFX6 TF activity assay in Xenopus. DEX-inducible expression constructs were generated for human RFX6 WT, p.Gln875*stop, and p.Arg347Lys.fs18*stop, as described in the text. (D) RT-qPCR analysis of hindgut endo explants after 8 h of TF (DEX) induction shows that human WT RFX6 and human p.Gln875*stop maintain normal expression of pdx1 and satb2, whereas p.Arg347Lys.fs18*stop causes loss of pdx1 expression and increased satb2. n=3 biological replicates. (E) RT-qPCR of DEX-inducible RFX6 shows that pdx1 is a direct target of RFX6. Red bars indicate expression of endogenous pdx1 in foregut endoderm explants (relative to uninjected, untreated HG endo) as a comparison. n=3 biological replicates. (F) Visualization of an evolutionarily conserved upstream PDX1 enhancer. UCSC genome browser shot of the human PDX1 locus is shown below with tracks from the indicated species, with black bars indicating regions of evolutionary conservation. (G) Experimental schematic of the Xenopus luciferase reporter assay used to test RFX6-dependent regulation of the human PDX1 enhancer. (H,I) RFX6 regulates human PDX1 enhancer activity and mutation of six CisBP-predicted (Weirauch et al., 2014) RFX motifs abolishes RFX6 responsiveness. Each black dot in the luciferase activity graphs represents a pool of n=5 embryos; mean relative luciferase activity±s.d. is shown. *P<0.05, parametric, two-tailed, paired t-test. ns, not significant.

Fig. 4.

An evolutionarily conserved role of RFX6 in GI tract patterning and functional characterization of RFX6 variants. (A) RT-PCR expression of PDX1 when after correction of one allele of RFX6 in 35D HIOs and PDX1 expression when RFX6 has been induced by DOX in hindgut monolayer (n=3). **P<0.01, ***P<0.001, ****P<0.0001 (unpaired t-test). (B) In situ hybridization of pdx1 and satb2 with control MO and rfx6 MO blocking translation in stage NF33 (2.5 d.p.f.) Xenopus embryos. Numbers in the lower left corner of panel indicate the number of embryos assayed. dp, dorsal pancreas; duo, duodenum; gb, gall bladder; hg, hindgut; st, stomach; vp, ventral pancreas. (C-E) Experimental schematic and human RFX6 TF activity assay in Xenopus. DEX-inducible expression constructs were generated for human RFX6 WT, p.Gln875*stop, and p.Arg347Lys.fs18*stop, as described in the text. (D) RT-qPCR analysis of hindgut endo explants after 8 h of TF (DEX) induction shows that human WT RFX6 and human p.Gln875*stop maintain normal expression of pdx1 and satb2, whereas p.Arg347Lys.fs18*stop causes loss of pdx1 expression and increased satb2. n=3 biological replicates. (E) RT-qPCR of DEX-inducible RFX6 shows that pdx1 is a direct target of RFX6. Red bars indicate expression of endogenous pdx1 in foregut endoderm explants (relative to uninjected, untreated HG endo) as a comparison. n=3 biological replicates. (F) Visualization of an evolutionarily conserved upstream PDX1 enhancer. UCSC genome browser shot of the human PDX1 locus is shown below with tracks from the indicated species, with black bars indicating regions of evolutionary conservation. (G) Experimental schematic of the Xenopus luciferase reporter assay used to test RFX6-dependent regulation of the human PDX1 enhancer. (H,I) RFX6 regulates human PDX1 enhancer activity and mutation of six CisBP-predicted (Weirauch et al., 2014) RFX motifs abolishes RFX6 responsiveness. Each black dot in the luciferase activity graphs represents a pool of n=5 embryos; mean relative luciferase activity±s.d. is shown. *P<0.05, parametric, two-tailed, paired t-test. ns, not significant.

Close modal

We next utilized Xenopus to interrogate further the functional impact of the two observed variants in human RFX6. We constructed dexamethasone (DEX)-inducible versions of human WT RFX6 and the two variants, in which the ligand-binding domain of the human glucocorticoid receptor (GR) is fused to the N terminus of RFX6; this GR-Dex induction system has been widely used for decades in Xenopus to control TF activity at a temporal level (Sive and Bradley, 1996; Zorn et al., 1999; Rankin et al., 2021). mRNA encoding the GR-TF fusion is synthesized in vitro and injected into embryos; the mRNA is translated in vivo by the embryo and the GR domain functionally sequesters the TF in the cytoplasm until addition of DEX to the culture buffer, at which point the TF can translocate to the nucleus and regulate target gene expression (Fig. 4C).

We compared the ability of human WT RFX6 and the two variants to induce pdx1 ectopically and suppress satb2 by targeted injections into the Xenopus hindgut endoderm (Fig. 4D), utilizing a hindgut explant dissection assay that also allowed for assessment of direct induction by pre-treatment of explants with cycloheximide (CHX) before temporal DEX TF induction (Fig. 4C,D). WT human RFX6 and the p.Gln875*stop variant were both able to robustly induce pdx1 and suppress satb2 in Xenopus hindgut endoderm, whereas the p.Arg347Lys.fs18*stop variant had no activity (Fig. 4D), suggesting this variant creates a null, non-functional allele. Similarly, in the CHX direct target assay, both the WT and p.Gln875*stop variant were able to induce pdx1 in the presence of CHX, suggesting that pdx1 is a direct target of RFX6 in vivo, whereas again the p.Arg347Lys.fs18*stop variant had no activity (Fig. 4E). As a positive control for CHX effectiveness, we assayed the expression of the pancreas TF gene ptf1a, induction of which by RFX6 was largely blocked by CHX treatment (Fig. S8F). Together, these results suggest that the p.Gln875*stop variant, resulting in a loss of the last 52 amino acids of the C terminus of RFX6, does not affect RFX6 transcriptional activity, whereas the more severe variant, resulting in p.Arg347Lys.fs18*stop, creates a null, non-functional allele in vivo.

During pancreas development, RFX6 has been shown to bind to DNA at sites near to the PDX1 gene; however, no functional analysis of RFX6-binding sites in PDX1 regulatory domains has been performed in any context. To investigate further the transcriptional regulation of pdx1 by RFX6, we bioinformatically examined a known, evolutionarily conserved enhancer upstream of human PDX1, of which the orthologous mouse region is sufficient to drive reporter expression in pancreas, stomach and duodenum (Gannon et al., 2001; Fujitani et al., 2006a), indicating functional enhancer activity. Evolutionary comparison of this human PDX1 upstream enhancer region (hg19_dna range=chr13:28491064-28492299) revealed significant conservation amongst several genomes; using the TF motif searching tool CisBP (Weirauch et al., 2014), six potential RFX-binding motifs were predicted (Table S1). Analysis of published ChIP-seq studies in human PSCs during directed endoderm differentiation for the key endoderm identity TFs FOXA2 and GATA4 (Vinckier et al., 2020), as well as for the midgut/hindgut TF CDX2 (Kumar et al., 2019), revealed binding of these important TFs to this conserved human PDX1 upstream enhancer (Fig. 4F), suggesting it also regulates developmental expression of PDX1 in human endoderm. We thus utilized the Xenopus model to interrogate potential RFX6-dependent regulation of this predicted human PDX1 enhancer. Luciferase reporters were constructed containing the WT human PDX1 upstream enhancer region or a version in which all six CisBP-predicted RFX motifs were mutated (Fig. 4G). Injection of the WT Human PDX1:luc reporter into foregut endoderm (targeting the progenitor domain of the pancreas, stomach, duodenum) revealed robust reporter activity, whereas injection of the enhancer:luc reporter into the satb2+ hindgut domain resulted in negligible activity (Fig. 4H,I) at both NF20 and NF33. Endogenous Xenopus Rfx6 was necessary for reporter activation in the foregut, as Rfx6 knockdown by MO prevented foregut-targeted PDX1 enhancer reporter activity. In support of this, ectopic injection of WT human RFX6 mRNA into the satb2+ hindgut domain resulted in robust PDX1 enhancer activity (Fig. 4I), and mutation of the six CisBP-predicted RFX motifs in the human PDX1 enhancer abolished reporter activity in the foregut domain as well as negated the ability of WT human RFX6 to activate the reporter in the hindgut (Fig. 4I). We further verified that the human p.Gln875*stop RFX6 variant retained ability to activate the human PDX1 enhancer, whereas the p.Arg347Lys.fs18*stop variant had no ability to activate the enhancer (Fig. S8G). Together, these results suggest that RFX6 controls vertebrate GI tract patterning in part by direct activation of PDX1 via a conserved upstream enhancer.

Identifying PDX1-dependent and -independent roles for RFX6 in patterning in D7 gut tube cultures

RFX6 and PDX1 have overlapping expression patterns in the nascent duodenal endoderm and both genes are required for normal duodenal development. Our data show that PDX1 expression is lost in RFX6 variants and suggest that PDX1 is a direct RFX6 target. This suggests that some or all of the phenotypes associated with loss of RFX6 could be due to loss of PDX1 expression. To identify PDX1-dependent versus -independent roles of RFX6, we used RFX6 Mut iPSCs to generate tetracycline-inducible RFX6 or PDX1 constructs whereby we could induce expression of either gene at specific time points (Fig. S7A,B) and identify downstream transcriptional responses. We validated that a 48 h DOX induction of RFX6 was sufficient for PDX1 expression (Fig. S7C). We differentiated RFX6 Mut iPSCs and induced expression of either RFX6 or PDX1 separately after D4 of differentiation and maintained induced expression until harvest at D7, the gut tube stage (Fig. 5A). PCA of gut spheroids with induced RFX6 and PDX1 revealed that gut tube endoderm in which RFX6 had been induced was more similar to that of the WT than to that of cultures in which PDX1 had been induced (Fig. 5B). A comparison of differentially expressed genes in response to RFX6 or PDX1 expression showed that RFX6 expression rescued 58% of the transcripts that were downregulated in the RFX6 Mut cultures. In contrast, tetracycline-induced PDX1 expression rescued only 25% of the transcripts that were lost in the RFX6 Mut cultures (Fig. 5C, Fig. S7D,E). From these comparisons, we identified which differentially expressed genes in the RFX6 Mut gut tube cultures were PDX1 dependent (rescued by PDX1 expression) compared with those that were not rescued by PDX1 expression. GO analysis of genes that appeared to be in the RFX6-PDX1 regulatory axis highlighted duodenal development (Fig. 5D). Expression of either RFX6 or PDX1 in the RFX6 Mut line was able to rescue several key gut-patterning markers, such as CDX2, SATB2 and GATA4, although PDX1 induction was not able to rescue RFX6 expression, supporting the suggestion that PDX1 acts downstream of RFX6 (Fig. 5E). Whereas the majority of PDX1-regulated genes were also regulated by RFX6, 67% of the 2070 differentially expressed genes in the RFX6 Mut samples were not regulated by PDX1, suggesting that these are regulated by RFX6 independently of PDX1 (Fig. 5D, Fig. S7F). In particular, RFX6 appeared to target expression of numerous genes involved in signal transduction pathways that play essential roles in gut tube patterning and development, such as the WNT, BMP and Hedgehog pathways. RFX6 significantly upregulated transcription of multiple ligands, antagonists and receptors, suggesting that RFX6 impacts intestinal development through regulation of these important signaling pathways (Fig. 5F). Lastly, we observed that RFX6 induction was also able to upregulate other members of the RFX family, mainly RFX4-7, whereas PDX1 failed to induce most RFX genes (Fig. 5G). Taken together, these data show that RFX6 is responsible for the correct patterning of the small intestine by activation of PDX1 in the gut tube epithelium as well as by regulating signaling pathway components that are known to be involved in gut tube patterning.

Fig. 5.

RFX6- and PDX1-inducible constructs rescue early hindgut patterning in RFX6 Mut D7 HIOs. (A) Schematic of induction of PDX1 and RFX6 during D4-D7 of HIO differentiation. Created with BioRender.com. (B) PCA of WT versus RFX6 Mut and PDX1-induced (Ind) and RFX6-Ind D7 HIOs (n=4). (C) Genes that are upregulated in RFX6 Mut after induction of PDX1 and RFX6 compared with RFX6 Mut with no induction. (D) Overlap of genes upregulated after induction of PDX1 and RFX6 in RFX6 Mut D7 HIOs. GO terms associated with genes regulated by both PDX1 and RFX6 (ToppGene.com) are shown. (E) Induction of either RFX6 or PDX1 in RFX6 Mut D7 HIOs upregulates expression of TFs that have a role in hindgut patterning. Plotted are RNA counts from bulk RNA-seq on multiple isolated wells of gut spheroids (each dot represents a different well, n=4). (F) Induction of RFX6 upregulates signaling pathways in midgut patterning in RFX6 Mut D7 HIOs whereas induction of PDX1 is less effective. (G) RFX6 induction in RFX6 Mut D7 HIOs upregulates expression of other RFX genes. Significance determined by unpaired t-test with ***P<0.001, ****P<0.0001. Significant differentially expressed genes from bulk RNA-seq was defined by adjusted P-value of <0.05.

Fig. 5.

RFX6- and PDX1-inducible constructs rescue early hindgut patterning in RFX6 Mut D7 HIOs. (A) Schematic of induction of PDX1 and RFX6 during D4-D7 of HIO differentiation. Created with BioRender.com. (B) PCA of WT versus RFX6 Mut and PDX1-induced (Ind) and RFX6-Ind D7 HIOs (n=4). (C) Genes that are upregulated in RFX6 Mut after induction of PDX1 and RFX6 compared with RFX6 Mut with no induction. (D) Overlap of genes upregulated after induction of PDX1 and RFX6 in RFX6 Mut D7 HIOs. GO terms associated with genes regulated by both PDX1 and RFX6 (ToppGene.com) are shown. (E) Induction of either RFX6 or PDX1 in RFX6 Mut D7 HIOs upregulates expression of TFs that have a role in hindgut patterning. Plotted are RNA counts from bulk RNA-seq on multiple isolated wells of gut spheroids (each dot represents a different well, n=4). (F) Induction of RFX6 upregulates signaling pathways in midgut patterning in RFX6 Mut D7 HIOs whereas induction of PDX1 is less effective. (G) RFX6 induction in RFX6 Mut D7 HIOs upregulates expression of other RFX genes. Significance determined by unpaired t-test with ***P<0.001, ****P<0.0001. Significant differentially expressed genes from bulk RNA-seq was defined by adjusted P-value of <0.05.

Close modal

Long-term expression of PDX1 is required to stably maintain duodenal identity in RFX6 Mut HIOs

We identified that PDX1 expression in the RFX6 Mut gut tube endoderm was sufficient to rescue early expression of duodenal patterning markers. However, it was not clear whether early rescue of duodenal patterning was stable at later stages of development. To test this, we used the PDX1 DOX-inducible line to express PDX1 during the early stages of duodenal development that occur in vitro and then we extended development for an additional 10 weeks by transplanting HIOs into immunocompromised mice (Fig. 6A). We found that expression of PDX1 during HIO growth in vitro was not sufficient to maintain duodenal identity during the added 10 weeks of development in vivo (Fig. 6B,C). Moreover, we observed restoration of transcripts involved in proximal small intestine-specific functions, such as lipid metabolism, transport and absorption (Fig. 6D), including genes such as APOB, APOA4 and APOC3 being upregulated in RFX6 Mut HIOs that had DOX-induced PDX1 expression (Fig. 6E), suggesting that this phenotype associated with loss of function of RFX6 could be restored by PDX1. However, if we maintained PDX1 expression in vivo by feeding transplanted mice with DOX-containing chow, we observed restoration of markers of normal duodenal patterning, mainly the lack of SATB2 expression and maintenance of PDX1 expression in the epithelium, and lower numbers of goblet cells (Fig. 6F,G). Although we also saw an increase in the number of EECs in PDX1-expressing HIOs, they were still lower in number compared with controls (Fig. 6F,G). One possible reason is that EECs have higher levels of PDX1 than other epithelial cell types and DOX-containing chow might not induce high enough levels of PDX1 expression to promote robust EEC differentiation. We therefore augmented DOX levels by injecting transplanted mice with an additional 10 μg/kg dose of DOX 5 days before harvesting the HIOs. This resulted in a rescue of the number of EECs in RFX6 Mut HIOs to nearly WT levels (Fig. 6H). Our data suggest that PDX1 not only initiates the patterning of the duodenum and midgut, but is required to be maintained to keep duodenal function and identity (Fig. 6I). Altogether, these data show that expression of PDX1 can partially rescue the phenotypes seen in RFX6 Mut duodenal HIOs and that PDX1 must be actively maintained to preserve duodenal identity.

Fig. 6.

Prolonged induced expression of Pdx1 can partially rescue Rfx6 mutant phenotypes. (A) Schematic of Pdx1 induction by DOX in RFX6 Mut HIOs during differentiation or in vivo via chow. Created with BioRender.com. (B) Levels of Pdx1 mRNA in transplanted RFX6 mut HIOs. HIOs were grown in culture without (No Dox) or with (Dox in vitro) DOX and then transplanted into mice. To maintain PDX1 expression in vivo, a group of animals were fed chow containing DOX (Dox in vivo). Each dot represents a sample from bulk RNA-seq (n=3; in vitro, 3 wells; in vivo, 3 separate organoids). (C) PCA and heatmap of RFX6 Mut HIOs with DOX-inducible PDX1 expression in vitro and in vivo (n=3). (D) GO analysis of biological processes shows rescue of duodenal-specific functions in RFX6 Mut HIOs with Dox-induced PDX1 both in vitro and in vivo. (E) Differentially expressed genes between RFX6 Mut HIO with and without DOX-inducible PDX1 expression in vivo. (F) Comparison of patterning (SATB2, PDX1) and secretory (MUC2, CHGA) markers by immunohistochemistry in RFX6 Mut HIOs with and without induction of Dox PDX1 by DOX in vitro and in vivo. (G) Quantification of patterning and secretory markers in RFX6 Mut HIOs with and without DOX treatment (n=3). Images from a total of six sections from three HIOs were quantified. (H) Additional injection of DOX to induce PDX1 has a bigger effect on EEC differentiation in RFX6 mut HIOs. IP, intraperitoneal. Seven sections in total from three HIOs were quantified. (I) Proposed mechanism of the role of RFX6 in duodenal patterning, acting upstream of PDX1 and repressing distal intestinal features. Green and blue wedges indicate decreased and increased expression of duodenum- and ileum-specific factors, respectively. Significance determined by unpaired t-test with **P<0.01, ****P<0.0001. Significant differentially expressed genes from bulk RNA-seq was defined by adjusted P-value of <0.05. Scale bars: 100 μm.

Fig. 6.

Prolonged induced expression of Pdx1 can partially rescue Rfx6 mutant phenotypes. (A) Schematic of Pdx1 induction by DOX in RFX6 Mut HIOs during differentiation or in vivo via chow. Created with BioRender.com. (B) Levels of Pdx1 mRNA in transplanted RFX6 mut HIOs. HIOs were grown in culture without (No Dox) or with (Dox in vitro) DOX and then transplanted into mice. To maintain PDX1 expression in vivo, a group of animals were fed chow containing DOX (Dox in vivo). Each dot represents a sample from bulk RNA-seq (n=3; in vitro, 3 wells; in vivo, 3 separate organoids). (C) PCA and heatmap of RFX6 Mut HIOs with DOX-inducible PDX1 expression in vitro and in vivo (n=3). (D) GO analysis of biological processes shows rescue of duodenal-specific functions in RFX6 Mut HIOs with Dox-induced PDX1 both in vitro and in vivo. (E) Differentially expressed genes between RFX6 Mut HIO with and without DOX-inducible PDX1 expression in vivo. (F) Comparison of patterning (SATB2, PDX1) and secretory (MUC2, CHGA) markers by immunohistochemistry in RFX6 Mut HIOs with and without induction of Dox PDX1 by DOX in vitro and in vivo. (G) Quantification of patterning and secretory markers in RFX6 Mut HIOs with and without DOX treatment (n=3). Images from a total of six sections from three HIOs were quantified. (H) Additional injection of DOX to induce PDX1 has a bigger effect on EEC differentiation in RFX6 mut HIOs. IP, intraperitoneal. Seven sections in total from three HIOs were quantified. (I) Proposed mechanism of the role of RFX6 in duodenal patterning, acting upstream of PDX1 and repressing distal intestinal features. Green and blue wedges indicate decreased and increased expression of duodenum- and ileum-specific factors, respectively. Significance determined by unpaired t-test with **P<0.01, ****P<0.0001. Significant differentially expressed genes from bulk RNA-seq was defined by adjusted P-value of <0.05. Scale bars: 100 μm.

Close modal

In this study, we generated patient-derived intestinal organoids to model Mitchell–Riley Syndrome, which is caused by a compound-heterozygous variant of RFX6. Mitchell–Riley syndrome is a recessive syndrome characterized by neonatal diabetes, pancreatic hypoplasia, intestinal atresia, chronic diarrhea and malrotation (Concepcion et al., 2014). We uncovered previously unappreciated roles of RFX6 in early patterning of the small intestine and maintenance of duodenal identity, plus we were able to characterize possible downstream targets. We demonstrated that HIOs are an ideal model in which to mimic development and to manipulate genetic rescue experiments to characterize the mechanism of a disease (Spence et al., 2011b; Krishnamurthy et al., 2022). Genetic correction by CRISPR/Cas9 was used to show that one allele of RFX6 was sufficient to pattern the small intestine correctly and maintain duodenal identity. A tetracycline-inducible model also demonstrated that PDX1 can rescue the phenotypes of the RFX6 variant as a downstream target. This study provides additional evidence for the use of iPSC-derived organoids as an alternative to correct variants and generate healthy tissue for future transplantation. Here, we describe the role of RFX6 in intestinal development, patterning and maintenance. This RFX6 variant misregulates several signaling pathways in both the epithelium and mesenchyme, which leads to a delay in CDX2 expression, potentially due to the deranged crosstalk between mesoderm and endoderm. We showed that RFX6 is required to promote a duodenal identity and without it we saw conversion to a more distal pattern as marked by SATB2 and HOX genes. We also showed that the establishment of duodenal identity by RFX6 involves the direct regulation of PDX1 and that PDX1 expression was sufficient to rescue some, but not all, of the RFX6 transcriptional program. At later stages of HIO development, loss of RFX6 led to loss of duodenal function as measured by lipid transporter activity. Lastly, we validated that RFX6 is required for enteroendocrine differentiation as shown in mouse and human (Gehart et al., 2019; Piccand et al., 2019).

Loss of RFX6 caused several phenotypes in the intestinal organoids; however, we wanted to determine whether these could be corrected. CRISPR correction of one RFX6 allele rescued the phenotypes at the hindgut development stage and at the more mature intestine stage, which showed that one WT allele is sufficient to allow RFX6 function in intestinal development and duodenal identity maintenance. We also attempted to rescue the RFX6 variant phenotypes with PDX1. It has been shown that PDX1 is co-expressed in endocrine cells with RFX6, and PDX1 null models have been shown to have intestinal atresia (Chen et al., 2009; Boyer et al., 2006; Fujita et al., 2008; Fujitani et al., 2006a; Yang et al., 2017). A DOX-inducible construct allowed us to introduce PDX1 at different time points, which showed that PDX1 induction can correct the intestinal phenotypes caused by the RFX6 variant as early as the hindgut development stages. Additionally, we showed a significant overlap in differentially expressed genes when inducing RFX6 and PDX1 expression, respectively, which is evidence for redundancy in the roles of these TFs in intestinal maintenance. Finally, we showed that PDX1 not only has to be expressed during the gut developing stage, but is required to be continually expressed to maintain duodenal identity. We were able to identify a plethora of phenotypes caused directly and indirectly by loss of RFX6, which suggests that it has a role in regulating the crosstalk between mesenchyme and epithelium during gut development. We were able to identify the epithelial-specific mechanism of duodenum maintenance via PDX1 and identify candidates that could be involved in the intermediate steps in the regulation of signaling pathways between mesenchyme and epithelium. Additionally, along with evidence from both parents with normal RFX6 function and evidence from our CRISPR correction and Xenopus data, we were able to determine that the maternal allele retains transcriptional activity whereas the paternal allele is equivalent to complete loss of function. This provides an insight into RFX6 function relating to each domain of the protein. Further studies are needed to analyze the structural-functional correlation analysis in more detail; however, our data suggest that the paternal allele, having the DNA-binding domain intact, still loses nearly all RFX6 transcriptional activity.

In summary, we developed an HIO model from a patient with compound-heterozygous variants in RFX6 that led us to identify new pathologies of the duodenum of individuals with such variants. We demonstrated that the paternal allele was null and that CRISPR correction was sufficient to restore duodenal identity, in part through regulation of PDX1, consistent with studies in the developing pancreas in which PDX1 was a target (Trott et al., 2020; Cheng et al., 2019; Piccand et al., 2014). Further to this point, we identified a conserved enhancer in PDX1 that is regulated by RFX6 during gut development in embryos. Consistent with our findings, while this manuscript was in revision, a different paper was published that supports our data of RFX6 having a crucial role in gut patterning (Nakamura et al., 2024); however, their main focus was early endoderm development and did not focus on divergence of downstream transcriptional networks. Some limitations of our study were that there is variability between organoids and specially between parental cell lines; additionally, the differentiation protocol requiring kidney capsule transplantation is technically challenging and lengthy. However, this study helps uncover the spatiotemporal role of RFX6 in the development of the intestine and the maintenance of intestinal function with the characterization of a downstream target in PDX1. We were able to identify a mechanism by which the duodenum is initially patterned and demonstrate how its maintenance is also orchestrated by RFX6, either via PDX1 or independently. This study uses a combination of in vitro, in vivo and ex vivo models including an embryonic model (Xenopus) to validate transcriptional mechanisms and abnormal patterning caused by a variant of RFX6. Finally, our work provides evidence of the translational potential of PSC-derived organoids and genetic technologies that are broadly translatable to other organs, which opens the path for engineered tissue to be an alternative for transplantation after genetic correction.

Pluripotent stem cell culture and directed differentiation of HIOs

The iPSC lines were generated from the patient as previously described (Zhu et al., 2016). Informed consent was obtained from the patient for the collection of whole blood. Peripheral blood mononuclear cells were isolated and used to generate an iPSC line at the Pluripotent Stem Cell Facility at CCHMC. iPSCs were maintained in feeder-free culture. Cells were plated on hESC-qualified Matrigel (BD Biosciences) and maintained at 37°C with 5% CO2 with daily removal of differentiated cells and replacement of mTeSR1 media (STEMCELL Technologies). Cells were passaged routinely every 4 days using Dispase (STEMCELL Technologies). HIOs were generated according to protocols established in our lab (Spence et al., 2011b; Kechele and Wells, 2019; Tsai et al., 2017) and experiments with human iPSCs were approved by the CCHMC ESCRO committee (Protocol #EIPDB2713).

In vivo transplant of HIOs

HIOs were removed from Matrigel 28-35 days after spheroid generation, and were transplanted under the kidney capsule of immune-deficient NOD.Cg-PrkdcscidIl2rgtm1Wjl/Szj (NSG) mice (Watson et al., 2014). NSG mice were maintained on Bactrim chow for a minimum of 2 weeks prior to transplantation and thereafter for the duration of the experiment (8-14 weeks) except for DOX-chow during transplant for specific experiments. Mice received additional intraperitoneal DOX injection (1 ml of 0.5 µg/ml solution per kg body weight) 5 days before harvest.

CRISPR/Cas9 correction

CRISPR/Cas9 was used to correct the mutation in the paternal allele in the RFX6 patient iPSC line. The guide RNA used was ACATAAAAATTGGGAACAGT. A phosphorothioated single-stranded DNA donor oligo (T*A*A*TGCATTTTTTAACAATGAAGCATTTAACACATAGCCTTCTTTGTAGCTTATTAGCAGACATAAGAAATTTTGCTAAgAAcTGGGAgCAaTGGGTTGTTTCATCCTTGGAAAACTTGCCAG*A*A*G; substitutions are underlined and asterisks indicate another nucleotide) was designed to include the nucleotide insertion according to published methods (Liang et al., 2017). The iPSCs were reverse transfected with plasmid and donor oligos using TranIT-LT1 (Mirus). After green fluorescence-activated cell sorting, iPSCs were re-plated in Matrigel. Single clones were manually excised for genotyping, expansion and cryopreservation. Correctly targeted clones were identified by PCR, enzyme digestion, and Sanger sequencing.

Generation of DOX-inducible RFX6 and PDX1 stem cell line

For inducible overexpression, we obtained human RFX6 cDNA in pENTR221 (from ORF Clone Collection ID IOH27199), which was then cloned into lentiviral destination vector pInducer20 (gift from Stephen Elledge, Addgene #44012) using Gateway cloning methods with LR Clonase II (Invitrogen). Successful cloning was confirmed using Sanger sequencing. Lentiviral particles were generated by CCHMC Viral Core. iPSCs were transduced by addition of lentiviral supernatant to the culture medium immediately after passaging, which was then replaced with fresh medium after 24 h of exposure. The cells were selected with G418 (100 μg/ml) for 4 days, beginning on the following day. At this point, the colonies had normal morphology and normal characteristics, whereas mock-transduced cells were dead. The cell line was the maintained under standard culture conditions, with intermittent exposure to G418 to maintain resistant cells. Inducible RFX6 expression was validated by qPCR 24 h after exposure to DOX (0.5 μg/ml) at the pluripotent cell stage. In the same way, a PDX1-inducible line was generated as previously described (Krishnamurthy et al., 2022). They were maintained in mTeSR with intermittent G418 selection.

RNA-seq

The initial amplification step for all samples was carried out using the Ovation RNA-Seq System v2 kit (Tecan Genomics). The assay was used to amplify RNA samples to create double-stranded cDNA. The concentrations were measured using the Qubit dsDNA BR assay. The cDNA size of each sample was determined using an Agilent HS DNA chip. Libraries were then created for all samples. Specifically, the Illumina protocol, the Nextera XT DNA Sample Preparation Kit, was used to create DNA library templates from the double-stranded cDNA. The concentrations were measured using the Qubit dsDNA HS assay. The size of the libraries for each sample was measured using the Agilent HS DNA chip. The samples were placed in a pool. The concentration of the pool was optimized to acquire at least 40 million reads per sample. The investigator requested the use of Paired-End 100 bp Reads, so for sequencing a NovaSeq S1 (200 cycles) v1.5 flow cell was used. Libraries were sequenced on the Illumina NovaSeq 6000 system following the manufacturer's protocol.

Bulk RNA-seq analysis

RNA-seq data were analyzed using Computational Suite for Bioinformaticians and Biologists (CSBB, v3.0.0) using the ‘Process-RNASeq_PairedEnd’ and ‘Generate-TPM_Counts_Matrix’ modules (https://github.com/praneet1988/Computational-Suite-For-Bioinformaticians-and-Biologists). FASTQs had adapters trimmed using a tool called bbduk, and quality was checked using fastqc. Bowtie2+RSEM was used to align and count transcripts, using reference genome (Genome Reference Consortium Human Build 38). From the RSEM results, count/TPM matrices were generated for the genes/isoforms. Further analysis and data visualization was performed using RStudio v4.2.1 with the following package: DESeq2, ggplot2, plyr, ggrepel, EnhancedVolcano, pheatmap (Anders and Huber, 2010; Wickham, 2011).

Immunofluorescence

Tissue was fixed in 4% paraformaldehyde, cryopreserved in 30% sucrose, embedded in OCT, and frozen at −80°C before cryosectioning. Cryosections (8 μm thick) were mounted on Superfrost Plus slides and permeabilized, blocked and stained according to standard protocols (as described by Spence et al., 2011b). Primary antibodies used are listed in Table S2 and all secondary antibodies were conjugated to Alexa Fluor 488, 546/555/568 or 647 (Invitrogen, A-21206, A-21202, A-11055) and used at 1:500. Images were acquired using a Nikon A1 GaAsP LUNV inverted confocal microscope and NIS Elements software (Nikon). Quantification and analysis were carried out using Nikon software (version 4.11.0).

qPCR

RNA was extracted using the Nucleospin RNA extraction kit (Macharey-Nagel) and reverse transcribed into cDNA using Superscript VILO (Invitrogen) according to the manufacturer's instruction. qPCR primers were designed using NCBI PrimerBlast. Primer sequences are listed in Table S3. qPCR was performed using Quantitect SYBR Green PCR kit (QIAGEN) and a QuantStudio 3 Flex RT PCR System (Applied Biosystems). Relative expression was determined using the Delta Cycle method and normalizing to PPIA (cyclophilin A). Samples from at least three independent passages were used for quantification.

Xenopus methods

Xenopus embryos and injections

WT adult X. laevis frogs were purchased from the National Xenopus Resource Center (NXR, Woods Hole, MA, USA; RRID:SCR_013731). Ovulation, in vitro fertilization, embryo de-jellying, and microinjection were performed as described (Sive and Bradley, 1996). Human RFX6 cDNA clone was purchased from Horizon Discovery (OHS6084-202637733) and Gateway sub-cloned from its entry vector pENTR223 into the expression vector pCSf107mT-Gateway-3′myc (Addgene #67617) using clonase (Thermo Fisher Scientific, 11791020) according to the manufacturer's instructions. SacII-linearized plasmid template was used to make mRNA for injection using the Ambion mMessage mMachine SP6 RNA Synthesis Kit (Thermo Fisher Scientific, AM1340) according to the manufacturer's instructions.

The WT human RFX6 coding region (NCBI Reference Sequence: NM_173560.4) and mutated versions (a 1 bp mutation nucleotide C2623T or a 13 bp deletion nucleotides 1040-1052 of the coding sequence) were also commercially synthesized with 5′EcoRV and 3′SpeI ends and cloned into the pCDNA3.1 vector by GenScript. To construct the inducible GR-RFX6 WT or mutants used in CHX direct target assays, the respective RFX6 WT or mutant versions were released from pCDNA3.1 via EcoRV/SpeI digests, gel purified, and cloned 5′EcoRV 3′SpeI into the pT7TS-GR-HA vector. XbaI-linearized plasmid templates were used to make mRNA for injection using the Ambion mMessage mMachine T7 mRNA Synthesis Kit (Thermo Fisher Scientific, AM1344) according to the manufacturer's instructions. One-hundred picograms total mRNA for WT or mutant GR-RFX6 RNAs were injected at the 8-cell stage into ventral-vegetal blastomeres to target the hindgut (50 pg per cell). At stage 20, hindgut explants (containing both endoderm and mesoderm) were microdissected in 1× Modified Barth's Saline (MBS) + 50 μg/ml gentamycin sulfate (gent; MP Biochemicals, 1676045) and cultured in 0.5× MBS+0.2% fatty acid-free bovine serum albumin (Fisher Scientific, BP9704)+50 μg/ml gent with the following concentrations of factors: 1 μM DEX (Sigma-Aldrich D4902); 1 μM CHX (Sigma-Aldrich, C4859). In CHX experiments, explants were treated for 2 h with CHX prior to DEX+CHX treatment for 6 h.

The previously validated translation-blocking MO against Xenopus Rfx6 (Pearl et al., 2011) was purchased from GeneTools and injected at the 8-cell stage (1.5 ng per cell) into each dorsal vegetal blastomere to target the foregut. MO sequences were as follows: Rfx6-MO1: 5′-AATTGGCATTTCACCGGGTTCAGGC-3′; a negative control Rfx6 mismatch (MM MO) with five bases altered: 5′ -AATaGGgATTTgACCcGGTTCAcGC-3′ (mismatch bases indicated in lowercase, bold, underlined).

Xenopus RT-qPCR

Xenopus explants were dissected from embryos of two or three separate fertilization/injection experiments, homogenized and frozen on dry ice in 350 μl of Buffer RA1 containing BME (Macherey-Nagel Nucleospin RNA isolation kit, 740955), and stored at −80°C. RNA was extracted according to the manufacturer's instructions and 500 ng RNA was used in cDNA synthesis reactions using Superscript Vilo Mastermix (Thermo Fisher Scientific, 11755050), also according to the manufacturer's instructions. qPCR reactions were carried out using PowerUp Mastermix (Thermo Fisher Scientific, A25742) on an ABI QuantStudio3 machine. Xenopus RT-qPCR primer sequences were as follows: pdx1.L forward: 5′-GTTCCCTCAGCTGCTTATCG; pdx1.L reverse: 5′-TACCAAGGGGTTGCTGTAGG; ptf1a.L forward: 5'-ATGGAAACGGTCCTGGAGCA; ptf1a.L reverse: 5'-GAGGATGAGAAGGAGAAGTTG. Relative expression, normalized to the ubiquitously expressed odc1, and then set relative to uninjected, untreated hindgut explants, was determined using the 2−ΔΔCt method. Graphs display the average 2−ΔΔCt value±s.d. Statistical significance (P<0.05) was determined using parametric, two-tailed, paired t-tests, relative to uninjected, untreated hindgut explants. Each black dot in the RT-qPCR graphs represents an independent biological replicate containing four explants.

Xenopus in situ hybridization

In situ hybridization of Xenopus embryos was performed as described (Sive and Bradley, 1996) with minor modifications. Briefly, embryos were fixed overnight at 4°C in MEMFA (0.1 M MOPS, 2 mM EGTA, 1 mM MgSO4, 3.7% formaldehyde), washed 2×10 min in MEMFA without formaldehyde, dehydrated directly into 100% ethanol, washed four to six times in 100% ethanol, and stored at −20°C in 100% ethanol for at least 24 h. Proteinase K (Thermo Fisher Scientific, AM2548) on day 1 was used at 2 µg/ml for 10 min on stage NF20 embryos and 5 µg/ml for 10 min on NF33 embryos; hybridization buffer included 0.1% SDS; the RNase step was omitted; and anti-digoxygenin (DIG)-alkaline phosphatase antibody (Sigma-Aldrich, 11093274910) used at 1:5000 in MAB buffer (100 mM maleic acid, 150 mM NaCl, pH 7.5)+20% heat-inactivated lamb serum (Gibco, 16070096)+2% blocking reagent (Sigma-Aldrich, 11096176001). Full-length rfx6.L and pdx1.L anti-sense DIG-labeled in situ probes were generated using linearized plasmid cDNA templates with 10× DIG RNA labeling mix (Sigma-Aldrich, 11277073910) according to the manufacturer's instructions. For immunostaining of the NF20 foregut region (Fig. S6), embryos were fixed for 1 h in MEMFA, dehydrated directly into Dent's post-fixative (80% methanol/20% DMSO), and stored at −20°C. Embryos were serially rehydrated through a methanol series (75%, 40%, 25%), bisected, and blocked for 2 h in PBS+10% normal goat serum (Jackson ImmunoResearch, 017-000-121)+1% DMSO+0.5% Triton X-100. Primary antibody incubation in the same blocking solution was carried out overnight (12-16 h) at 4°C using mouse anti-FLAG M2 (Sigma-Aldrich, F3165, 1:1000) and goat anti-FoxA2 (Santa Cruz Biotechnology, sc-6554, 1:500). After five 1 h washes in PBS+0.1% Triton X-100, embryo halves were incubated in secondary antibodies (donkey anti-mouse Cy2, Jackson ImmunoResearch, 715-225-150, 1:1000; donkey anti-goat Cy5, Jackson ImmunoResearch, 705-175-147, 1:1000) diluted in PBS+0.5% Triton X-100 overnight at 4°C. Embryo halves were then washed five times, 30 min per wash, in PBS+0.1% Triton X-100, dehydrated in 100% methanol, cleared in Murray's clear (80% benzyl benzoate, 20% benzyl alcohol), and imaged with a Nikon A1R confocal microscope.

Xenopus luciferase assays

A 1236 bp sequence of the human PDX1 upstream ‘area I II III’ enhancer (>hg19_dna range=chr13:28491064-28492299) was screened for RFX6 responsiveness because the orthologous mouse Pdx1 enhancer region has previously been shown to drive reporter expression in foregut and duodenum (Gannon et al., 2001; Fujitani et al., 2006b). WT or a mutant version of the human PDX1 enhancer with six CisBP-predicted RFX-binding motifs mutated (Weirauch et al., 2014) were commercially synthesized by GenScript and cloned into the pGL4.23 firefly luc2/miniP vector (Promega, E8411). Embryos (16- to 32-cell stage) were co-injected with 5 pg of pRL-TK:Renilla luciferase plasmid (Promega, E2241)+80 pg of the pGL4.23 luc2/miniP Human PDX1 enhancer:firefly luciferase plasmid (WT or 6RFX motif mutant) into either a D1 blastomere to target foregut or a D4 blastomere to target hindgut. The following amounts of MOs or mRNAs were also injected in enhancer:luciferase assays: 3 ng total of Rfx6-MO1 or 5 bp mismatch-MO; 100 pg WT or mutant human RFX6 mRNAs.

Each biological replicate contained a pool of five embryos, obtained from two or three separate fertilization/injection experiments, which were frozen on dry ice in a minimal volume of 0.1× MBS and stored at −80°C. To assay luciferase activity, samples were lysed in 100 µl of 100 mM TRIS-Cl pH 7.5, centrifuged for 10 min at ∼13,000 g and then 50 µl of the clear supernatant lysate was used separately in firefly (Biotium, 30085-1) and Renilla (Biotium 300821) luciferase assays according to the manufacturer's instructions. Relative luciferase activity was determined by normalizing firefly to Renilla levels for each sample. Graphs show the average relative luciferase activity ±s.d. with black dots showing values of biological replicates. Statistical significance was determined by parametric, two-tailed, paired t-test (*P<0.05).

Statistics

Data are presented as the mean±s.e.m. Significance was determined using appropriate tests in GraphPad Prism, with P>0.05 being considered not significant.

We are thankful for the support provided by the confocal imaging core, gene expression core, pathology core and pluripotent stem cell facility at CCHMC. CCHMC is supported by a Digestive Diseases Research grant (P30 DK078392). We thank all the members of the Wells and Zorn labs for reagents and feedback. All figures were created using Adobe Illustrator, BioRender.com and GraphPad Prism.

Author contributions

Conceptualization: D.O.K., A.M.Z., J.M.W.; Methodology: J.G.S., S.R., E.P., H.A.M., D.O.K., J.R.E., L.L.-F.; Formal analysis: J.G.S.; Investigation: J.G.S., J.M.W.; Resources: S.R., N.-H.J., S.A.W.G., M.K., J.M.W.; Writing - original draft: J.G.S.; Writing - review & editing: S.R., H.A.M., D.O.K., J.R.E., M.K., J.M.W.; Visualization: J.G.S., J.M.W.; Supervision: H.A.M., D.O.K., A.M.Z., M.K., J.M.W.; Project administration: J.M.W.; Funding acquisition: J.M.W.

Funding

This research was supported by the grants from the National Institutes of Health (R01 CA272903 to J.M.W., P01 HD093363 to J.M.W. and A.M.Z., UH3 DK119982 to J.M.W., and R01 DK104942 to S.A.W.G.), the Shipley Foundation (to J.M.W.) and the Allen Foundation (to J.M.W.). Deposited in PMC for release after 12 months.

Data availability

Raw and processed RNA-seq data have been deposited in GEO under accession number GSE245474.

The people behind the papers

This article has an associated 'The people behind the papers' interview with some of the authors.

Anderle
,
P.
,
Sengstag
,
T.
,
Mutch
,
D. M.
,
Rumbo
,
M.
,
Praz
,
V.
,
Mansourian
,
R.
,
Delorenzi
,
M.
,
Williamson
,
G.
and
Roberts
,
M.-A.
(
2005
).
Changes in the transcriptional profile of transporters in the intestine along the anterior-posterior and crypt-villus axes
.
BMC Genomics
6
,
69
.
Anders
,
S.
and
Huber
,
W.
(
2010
).
Differential expression analysis for sequence count data
.
Genome Biol.
11
,
R106
.
Boyer
,
D. F.
,
Fujitani
,
Y.
,
Gannon
,
M.
,
Powers
,
A. C.
,
Stein
,
R. W.
and
Wright
,
C. V. E.
(
2006
).
Complementation rescue of Pdx1 null phenotype demonstrates distinct roles of proximal and distal cis-regulatory sequences in pancreatic and duodenal expression
.
Dev. Biol.
298
,
616
-
631
.
Burlison
,
J. S.
,
Long
,
Q.
,
Fujitani
,
Y.
,
Wright
,
C. V. E.
and
Magnuson
,
M. A.
(
2008
).
Pdx-1 and Ptf1a concurrently determine fate specification of pancreatic multipotent progenitor cells
.
Dev. Biol.
316
,
74
-
86
.
Carpenter
,
M. K.
and
Rao
,
M. S.
(
2015
).
Concise review: making and using clinically compliant pluripotent stem cell lines
.
Stem Cells Transl. Med.
4
,
381
-
388
.
Carten
,
J. D.
,
Bradford
,
M. K.
and
Farber
,
S. A.
(
2011
).
Visualizing digestive organ morphology and function using differential fatty acid metabolism in live zebrafish
.
Dev. Biol.
360
,
276
-
285
.
Chawengsaksophak
,
K.
,
James
,
R.
,
Hammond
,
V. E.
,
Köntgen
,
F.
and
Beck
,
F.
(
1997
).
Homeosis and intestinal tumours in Cdx2 mutant mice
.
Nature
386
,
84
-
87
.
Chen
,
C.
,
Fang
,
R.
,
Davis
,
C.
,
Maravelias
,
C.
and
Sibley
,
E.
(
2009
).
Pdx1 inactivation restricted to the intestinal epithelium in mice alters duodenal gene expression in enterocytes and enteroendocrine cells
.
Am. J. Physiol. Gastrointest. Liver Physiol.
297
,
G1126
-
G1137
.
Cheng
,
C.
,
Lu
,
J.
,
Cao
,
X.
,
Yang
,
F.-Y.
,
Liu
,
J.-Y.
,
Song
,
L.-N.
,
Shen
,
H.
,
Liu
,
C.
,
Zhu
,
X.-R.
,
Zhou
,
J.-B.
et al.
(
2019
).
Identification of Rfx6 target genes involved in pancreas development and insulin translation by ChIP-seq
.
Biochem. Biophys. Res. Commun.
508
,
556
-
562
.
Concepcion
,
J. P.
,
Reh
,
C. S.
,
Daniels
,
M.
,
Liu
,
X.
,
Paz
,
V. P.
,
Ye
,
H.
,
Highland
,
H. M.
,
Hanis
,
C. L.
and
Greeley
,
S. A. W.
(
2014
).
Neonatal diabetes, gallbladder agenesis, duodenal atresia, and intestinal malrotation caused by a novel homozygous mutation in RFX6
.
Pediatr. Diabetes
15
,
67
-
72
.
Dessimoz
,
J.
,
Opoka
,
R.
,
Kordich
,
J. J.
,
Grapin-Botton
,
A.
and
Wells
,
J. M.
(
2006
).
FGF signaling is necessary for establishing gut tube domains along the anterior-posterior axis in vivo
.
Mech. Dev.
123
,
42
-
55
.
DiPatrizio
,
N. V.
and
Piomelli
,
D.
(
2015
).
Intestinal lipid-derived signals that sense dietary fat
.
J. Clin. Invest.
125
,
891
-
898
.
Fujita
,
Y.
,
Chui
,
J. W. Y.
,
King
,
D. S.
,
Zhang
,
T.
,
Seufert
,
J.
,
Pownall
,
S.
,
Cheung
,
A. T.
and
Kieffer
,
T. J.
(
2008
).
Pax6 and Pdx1 are required for production of glucose-dependent insulinotropic polypeptide in proglucagon-expressing L cells
.
Am. J. Physiol. Endocrinol. Metab.
295
,
E648
-
E657
.
Fujitani
,
Y.
,
Fujitani
,
S.
,
Boyer
,
D. F.
,
Gannon
,
M.
,
Kawaguchi
,
Y.
,
Ray
,
M.
,
Shiota
,
M.
,
Stein
,
R. W.
,
Magnuson
,
M. A.
and
Wright
,
C. V. E.
(
2006a
).
Targeted deletion of a cis-regulatory region reveals differential gene dosage requirements for Pdx1 in foregut organ differentiation and pancreas formation
.
Genes Dev.
20
,
253
-
266
.
Fujitani
,
Y.
,
Fujitani
,
S.
,
Luo
,
H.
,
Qiu
,
F.
,
Burlison
,
J.
,
Long
,
Q.
,
Kawaguchi
,
Y.
,
Edlund
,
H.
,
MacDonald
,
R. J.
,
Furukawa
,
T.
,
Fujikado
,
T.
et al.
(
2006b
).
Ptf1a determines horizontal and amacrine cell fates during mouse retinal development
.
Development
133
,
4439
-
4450
.
Gannon
,
M.
,
Gamer
,
L. W.
and
Wright
,
C. V. E.
(
2001
).
Regulatory regions driving developmental and tissue-specific expression of the essential pancreatic gene pdx1
.
Dev. Biol.
238
,
185
-
201
.
Gehart
,
H.
,
van Es
,
J. H.
,
Hamer
,
K.
,
Beumer
,
J.
,
Kretzschmar
,
K.
,
Dekkers
,
J. F.
,
Rios
,
A.
and
Clevers
,
H.
(
2019
).
Identification of enteroendocrine regulators by real-time single-cell differentiation mapping
.
Cell
176
,
1158
-
1173.e16
.
Gu
,
W.
,
Wang
,
H.
,
Huang
,
X.
,
Kraiczy
,
J.
,
Singh
,
P. N. P.
,
Ng
,
C.
,
Dagdeviren
,
S.
,
Houghton
,
S.
,
Pellon-Cardenas
,
O.
,
Lan
,
Y.
et al.
(
2022
).
SATB2 preserves colon stem cell identity and mediates ileum-colon conversion via enhancer remodeling
.
Cell Stem Cell
29
,
101
-
115.e10
.
Haber
,
A. L.
,
Biton
,
M.
,
Rogel
,
N.
,
Herbst
,
R. H.
,
Shekhar
,
K.
,
Smillie
,
C.
,
Burgin
,
G.
,
Delorey
,
T. M.
,
Howitt
,
M. R.
,
Katz
,
Y.
et al.
(
2017
).
A single-cell survey of the small intestinal epithelium
.
Nature
551
,
333
-
339
.
Haeussler
,
M.
,
Schönig
,
K.
,
Eckert
,
H.
,
Eschstruth
,
A.
,
Mianné
,
J.
,
Renaud
,
J.-B.
,
Schneider-Maunoury
,
S.
,
Shkumatava
,
A.
,
Teboul
,
L.
,
Kent
,
J.
et al.
(
2016
).
Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR
.
Genome Biol.
17
,
148
.
Hajirnis
,
N.
and
Mishra
,
R. K.
(
2021
).
Homeotic genes: clustering, modularity, and diversity
.
Front. Cell Dev. Biol.
9
,
718308
.
Han
,
L.
,
Chaturvedi
,
P.
,
Kishimoto
,
K.
,
Koike
,
H.
,
Nasr
,
T.
,
Iwasawa
,
K.
,
Giesbrecht
,
K.
,
Witcher
,
P. C.
,
Eicher
,
A.
,
Haines
,
L.
et al.
(
2020
).
Single cell transcriptomics identifies a signaling network coordinating endoderm and mesoderm diversification during foregut organogenesis
.
Nat. Commun.
11
,
4158
.
Hussain
,
M. M.
(
2014
).
Intestinal lipid absorption and lipoprotein formation
.
Curr. Opin. Lipidol.
25
,
200
-
206
.
Iqbal
,
J.
and
Hussain
,
M. M.
(
2009
).
Intestinal lipid absorption
.
Am. J. Physiol. Endocrinol. Metab.
296
,
E1183
-
E1194
.
Jepeal
,
L. I.
,
Fujitani
,
Y.
,
Boylan
,
M. O.
,
Wilson
,
C. N.
,
Wright
,
C. V.
and
Wolfe
,
M. M.
(
2005
).
Cell-specific expression of glucose-dependent-insulinotropic polypeptide is regulated by the transcription factor PDX-1
.
Endocrinology
146
,
383
-
391
.
Kambal
,
M. A.
,
Al-Harbi
,
D. A.
,
Al-Sunaid
,
A. R.
and
Al-Atawi
,
M. S.
(
2019
).
Mitchell-Riley syndrome due to a novel mutation in RFX6
.
Front. Pediatr.
7
,
243
.
Kechele
,
D. O.
and
Wells
,
J. M.
(
2019
).
Recent advances in deriving human endodermal tissues from pluripotent stem cells
.
Curr. Opin. Cell Biol.
61
,
92
-
100
.
Krishnamurthy
,
M.
,
Kechele
,
D. O.
,
Broda
,
T.
,
Zhang
,
X.
,
Enriquez
,
J. R.
,
McCauley
,
H. A.
,
Sanchez
,
J. G.
,
McCracken
,
K.
,
Palermo
,
J.
,
Bernieh
,
A.
et al.
(
2022
).
Using human induced pluripotent stem cell-derived organoids to identify new pathologies in patients with PDX1 mutations
.
Gastroenterology
163
,
1053
-
1063.e7
.
Kumar
,
N.
,
Tsai
,
Y.-H.
,
Chen
,
L.
,
Zhou
,
A.
,
Banerjee
,
K. K.
,
Saxena
,
M.
,
Huang
,
S.
,
Toke
,
N. H.
,
Xing
,
J.
,
Shivdasani
,
R. A.
et al.
(
2019
).
The lineage-specific transcription factor CDX2 navigates dynamic chromatin to control distinct stages of intestine development
.
Development
146
,
dev172189
.
Lema
,
I.
,
Araújo
,
J. R.
,
Rolhion
,
N.
and
Demignot
,
S.
(
2020
).
Jejunum: the understudied meeting place of dietary lipids and the microbiota
.
Biochimie
178
,
124
-
136
.
Liang
,
X.
,
Potter
,
J.
,
Kumar
,
S.
,
Ravinder
,
N.
and
Chesnut
,
J. D.
(
2017
).
Enhanced CRISPR/Cas9-mediated precise genome editing by improved design and delivery of gRNA, Cas9 nuclease, and donor DNA
.
J. Biotechnol.
241
,
136
-
146
.
McCarthy
,
N.
,
Manieri
,
E.
,
Storm
,
E. E.
,
Saadatpour
,
A.
,
Luoma
,
A. M.
,
Kapoor
,
V. N.
,
Madha
,
S.
,
Gaynor
,
L. T.
,
Cox
,
C.
,
Keerthivasan
,
S.
et al.
(
2020
).
Distinct mesenchymal cell populations generate the essential intestinal BMP signaling gradient
.
Cell Stem Cell
26
,
391
-
402.e5
.
Nakamura
,
T.
,
Fujikura
,
J.
,
Ito
,
R.
,
Keidai
,
Y.
and
Inagaki
,
N.
(
2024
).
Human RFX6 regulates endoderm patterning from at the primitive gut stage
.
PNAS Nexus
3
,
pgae001
.
Offield
,
M. F.
,
Jetton
,
T. L.
,
Labosky
,
P. A.
,
Ray
,
M.
,
Stein
,
R. W.
,
Magnuson
,
M. A.
,
Hogan
,
B. L. M.
and
Wright
,
C. V. E.
(
1996
).
PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum
.
Development
122
,
983
-
995
.
Pearl
,
E. J.
,
Jarikji
,
Z.
and
Horb
,
M. E.
(
2011
).
Functional analysis of Rfx6 and mutant variants associated with neonatal diabetes
.
Dev. Biol.
351
,
135
-
145
.
Piccand
,
J.
,
Strasser
,
P.
,
Hodson
,
D. J.
,
Meunier
,
A.
,
Ye
,
T.
,
Keime
,
C.
,
Birling
,
M.-C.
,
Rutter
,
G. A.
and
Gradwohl
,
G.
(
2014
).
Rfx6 maintains the functional identity of adult pancreatic β cells
.
Cell Rep.
9
,
2219
-
2232
.
Piccand
,
J.
,
Vagne
,
C.
,
Blot
,
F.
,
Meunier
,
A.
,
Beucher
,
A.
,
Strasser
,
P.
,
Lund
,
M. L.
,
Ghimire
,
S.
,
Nivlet
,
L.
,
Lapp
,
C.
et al.
(
2019
).
Rfx6 promotes the differentiation of peptide-secreting enteroendocrine cells while repressing genetic programs controlling serotonin production
.
Mol. Metab.
29
,
24
-
39
.
Ran
,
F. A.
,
Hsu
,
P. D.
,
Wright
,
J.
,
Agarwala
,
V.
,
Scott
,
D. A.
and
Zhang
,
F.
(
2013
).
Genome engineering using the CRISPR-Cas9 system
.
Nat. Protoc.
8
,
2281
-
2308
.
Rankin
,
S. A.
,
Steimle
,
J. D.
,
Yang
,
X. H.
,
Rydeen
,
A. B.
,
Agarwal
,
K.
,
Chaturvedi
,
P.
,
Ikegami
,
K.
,
Herriges
,
M. J.
,
Moskowitz
,
I. P.
and
Zorn
,
A. M.
(
2021
).
Tbx5 drives Aldh1a2 expression to regulate a RA-Hedgehog-Wnt gene regulatory network coordinating cardiopulmonary development
.
eLife
10
,
e69288
.
Roberts
,
D. J.
,
Johnson
,
R. L.
,
Burke
,
A. C.
,
Nelson
,
C. E.
,
Morgan
,
B. A.
and
Tabin
,
C.
(
1995
).
Sonic hedgehog is an endodermal signal inducing Bmp-4 and Hox genes during induction and regionalization of the chick hindgut
.
Development
121
,
3163
-
3174
.
Schubert
,
M. L.
(
2011
).
Stomach and duodenum
.
Curr. Opin Gastroenterol.
27
,
534
-
535
.
Sherwood
,
R. I.
,
Maehr
,
R.
,
Mazzoni
,
E. O.
and
Melton
,
D. A.
(
2011
).
Wnt signaling specifies and patterns intestinal endoderm
.
Mech. Dev.
128
,
387
-
400
.
Singh
,
A.
,
Poling
,
H. M.
,
Chaturvedi
,
P.
,
Thorner
,
K.
,
Sundaram
,
N.
,
Kechele
,
D. O.
,
Childs
,
C. J.
,
McCauley
,
H. A.
,
Fisher
,
G. W.
,
Brown
,
N. E.
et al.
(
2023
).
Transplanted human intestinal organoids: a resource for modeling human intestinal development
.
Development
150
,
dev201416
.
Sive
,
H.
and
Bradley
,
L.
(
1996
).
A sticky problem: the Xenopus cement gland as a paradigm for anteroposterior patterning
.
Dev. Dyn.
205
,
265
-
280
.
Smith
,
S. B.
,
Qu
,
H.-Q.
,
Taleb
,
N.
,
Kishimoto
,
N. Y.
,
Scheel
,
D. W.
,
Lu
,
Y.
,
Patch
,
A.-M.
,
Grabs
,
R.
,
Wang
,
J.
,
Lynn
,
F. C.
et al.
(
2010
).
Rfx6 directs islet formation and insulin production in mice and humans
.
Nature
463
,
775
-
780
.
Soyer
,
J.
,
Flasse
,
L.
,
Raffelsberger
,
W.
,
Beucher
,
A.
,
Orvain
,
C.
,
Peers
,
B.
,
Ravassard
,
P.
,
Vermot
,
J.
,
Voz
,
M. L.
,
Mellitzer
,
G.
et al.
(
2010
).
Rfx6 is an Ngn3-dependent winged helix transcription factor required for pancreatic islet cell development
.
Development
137
,
203
-
212
.
Spence
,
J. R.
,
Lauf
,
R.
and
Shroyer
,
N. F.
(
2011a
).
Vertebrate intestinal endoderm development
.
Dev. Dyn.
240
,
501
-
520
.
Spence
,
J. R.
,
Mayhew
,
C. N.
,
Rankin
,
S. A.
,
Kuhar
,
M. F.
,
Vallance
,
J. E.
,
Tolle
,
K.
,
Hoskins
,
E. E.
,
Kalinichenko
,
V. V.
,
Wells
,
S. I.
,
Zorn
,
A. M.
et al.
(
2011b
).
Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro
.
Nature
470
,
105
-
109
.
Stoffers
,
D. A.
,
Zinkin
,
N. T.
,
Stanojevic
,
V.
,
Clarke
,
W. L.
and
Habener
,
J. F.
(
1997
).
Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence
.
Nat. Genet.
15
,
106
-
110
.
Thompson
,
W. G.
and
Wrathell
,
E.
(
1977
).
The relation between ileal resection and vitamin B12 absorption
.
Can. J. Surg.
20
,
461
-
464
.
Thompson
,
C. A.
,
DeLaForest
,
A.
and
Battle
,
M. A.
(
2018
).
Patterning the gastrointestinal epithelium to confer regional-specific functions
.
Dev. Biol.
435
,
97
-
108
.
Ticho
,
A. L.
,
Malhotra
,
P.
,
Dudeja
,
P. K.
,
Gill
,
R. K.
and
Alrefai
,
W. A.
(
2019
).
Intestinal absorption of bile acids in health and disease
.
Compr. Physiol.
10
,
21
-
56
.
Trott
,
J.
,
Alpagu
,
Y.
,
Tan
,
E. K.
,
Shboul
,
M.
,
Dawood
,
Y.
,
Elsy
,
M.
,
Wollmann
,
H.
,
Tano
,
V.
,
Bonnard
,
C.
,
Eng
,
S.
et al.
(
2020
).
Mitchell-Riley syndrome iPSCs exhibit reduced pancreatic endoderm differentiation due to a mutation in RFX6
.
Development
147
,
dev194878
.
Tsai
,
Y.-H.
,
Nattiv
,
R.
,
Dedhia
,
P. H.
,
Nagy
,
M. S.
,
Chin
,
A. M.
,
Thomson
,
M.
,
Klein
,
O. D.
and
Spence
,
J. R.
(
2017
).
In vitro patterning of pluripotent stem cell-derived intestine recapitulates in vivo human development
.
Development
144
,
1045
-
1055
.
Vinckier
,
N. K.
,
Patel
,
N. A.
,
Geusz
,
R. J.
,
Wang
,
A.
,
Wang
,
J.
,
Matta
,
I.
,
Harrington
,
A. R.
,
Wortham
,
M.
,
Wetton
,
N.
,
Jhala
,
U. S.
et al.
(
2020
).
LSD1-mediated enhancer silencing attenuates retinoic acid signalling during pancreatic endocrine cell development
.
Nat. Commun.
11
,
2082
.
Walton
,
K. D.
and
Gumucio
,
D. L.
(
2021
).
Hedgehog signaling in intestinal development and homeostasis
.
Annu. Rev. Physiol.
83
,
359
-
380
.
Watson
,
C. L.
,
Mahe
,
M. M.
,
Múnera
,
J.
,
Howell
,
J. C.
,
Sundaram
,
N.
,
Poling
,
H. M.
,
Schweitzer
,
J. I.
,
Vallance
,
J. E.
,
Mayhew
,
C. N.
,
Sun
,
Y.
et al.
(
2014
).
An in vivo model of human small intestine using pluripotent stem cells
.
Nat. Med.
20
,
1310
-
1314
.
Weirauch
,
M. T.
,
Yang
,
A.
,
Albu
,
M.
,
Cote
,
A. G.
,
Montenegro-Montero
,
A.
,
Drewe
,
P.
,
Najafabadi
,
H. S.
,
Lambert
,
S. A.
,
Mann
,
I.
,
Cook
,
K.
et al.
(
2014
).
Determination and inference of eukaryotic transcription factor sequence specificity
.
Cell
158
,
1431
-
1443
.
Wickham
,
H.
(
2011
).
The split-apply-combine strategy for data analysis
.
J. Stat. Softw.
40
,
1
-
29
.
Yang
,
Y.-P.
,
Magnuson
,
M. A.
,
Stein
,
R.
and
Wright
,
C. V. E.
(
2017
).
The mammal-specific Pdx1 Area II enhancer has multiple essential functions in early endocrine cell specification and postnatal β-cell maturation
.
Development
144
,
248
-
257
.
Yu
,
Q.
,
Kilik
,
U.
,
Holloway
,
E. M.
,
Tsai
,
Y.-H.
,
Harmel
,
C.
,
Wu
,
A.
,
Wu
,
J. H.
,
Czerwinski
,
M.
,
Childs
,
C. J.
,
He
,
Z.
et al.
(
2021
).
Charting human development using a multi-endodermal organ atlas and organoid models
.
Cell
184
,
3281
-
3298.e22
.
Zhu
,
Z.
,
Li
,
Q. V.
,
Lee
,
K.
,
Rosen
,
B. P.
,
González
,
F.
,
Soh
,
C.-L.
and
Huangfu
,
D.
(
2016
).
Genome editing of lineage determinants in human pluripotent stem cells reveals mechanisms of pancreatic development and diabetes
.
Cell Stem Cell
18
,
755
-
768
.
Zorn
,
A. M.
and
Wells
,
J. M.
(
2007
).
Molecular basis of vertebrate endoderm development
.
Int. Rev. Cytol.
259
,
49
-
111
.
Zorn
,
A. M.
and
Wells
,
J. M.
(
2009
).
Vertebrate endoderm development and organ formation
.
Annu. Rev. Cell Dev. Biol.
25
,
221
-
251
.
Zorn
,
A. M.
,
Butler
,
K.
and
Gurdon
,
J. B.
(
1999
).
Anterior endomesoderm specification in Xenopus by Wnt/beta-catenin and TGF-beta signalling pathways
.
Dev. Biol.
209
,
282
-
297
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information