Polymorphism in TCF7L2, a component of the canonical Wnt signaling pathway, has a strong association with β-cell dysfunction and type 2 diabetes through a mechanism that has yet to be defined. β-Cells rely on cells in their microenvironment, including pericytes, for their proper function. Here, we show that Tcf7l2 activity in pancreatic pericytes is required for β-cell function. Transgenic mice in which Tcf7l2 was selectively inactivated in their pancreatic pericytes exhibited impaired glucose tolerance due to compromised β-cell function and glucose-stimulated insulin secretion. Inactivation of pericytic Tcf7l2 was associated with impaired expression of genes required for β-cell function and maturity in isolated islets. In addition, we identified Tcf7l2-dependent pericytic expression of secreted factors shown to promote β-cell function, including bone morphogenetic protein 4 (BMP4). Finally, we show that exogenous BMP4 is sufficient to rescue the impaired glucose-stimulated insulin secretion of transgenic mice, pointing to a potential mechanism through which pericytic Tcf7l2 activity affects β-cells. To conclude, we suggest that pancreatic pericytes produce secreted factors, including BMP4, in a Tcf7l2-dependent manner to support β-cell function. Our findings thus propose a potential cellular mechanism through which abnormal TCF7L2 activity predisposes individuals to diabetes and implicates abnormalities in the islet microenvironment in this disease.

Type 2 diabetes has a strong genetic component, with a number of genetic variations associated with an increased risk to develop this disease (1,2). In particular, polymorphism in TCF7L2 (TCF4) is associated with increased risk to diabetes (3). This gene encodes a member of T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factors family, which functions downstream of the canonical Wnt signaling pathway by recruiting β-catenin to target genes (4). Diabetes-associated alleles of TCF7L2, such as the T allele of the single-nucleotide polymorphism in rs7903146, are associated with impaired glucose-stimulated insulin secretion (GSIS) and insulin production but intact hepatic function and insulin sensitivity (3,58). The T allele of the rs7903146 variant was predicted to result in an inactive protein lacking its DNA-binding domain (9). However, how TCF7L2 functions to regulate glucose homeostasis remains an open question.

To date, the use of mouse systems to determine the cellular mechanism(s) through which abnormal Tcf7l2 activity contributes to β-cell dysfunction has produced conflicting results. As opposed to humans, hepatic phenotypes dominate the abnormal glucose levels observed upon body-wide deregulation of Tcf7l2 expression in mice (1012). β-Cell–specific inference with Tcf7l2 activity using mouse genetic tools yielded discrepant results, with some studies showing reduced β-cell mass and glucose intolerance and others showing normal glucose response (1217). This contradiction could partially stem from the use of different approaches to interfere with Tcf7l2 activity, such as knocking down the endogenous gene (12,14,15) versus overexpressing a dominant-negative (DN) form (13,16). Tcf7 (Tcf1), a member of the TCF/LEF family with high homology to Tcf7l2, was recently shown to play a central role in maintaining β-cell mass (18), raising the possibility that overexpressing a DN Tcf7l2 interferes with the activity of other TCF/LEF proteins in β-cells. Interestingly, although β-cell–selective deletion of Tcf7l2 resulted in their reduced mass (15), selective deletion of this transcription factor DNA-binding domain affected neither β-cell function nor mass (12). Accordingly, nonautonomous roles of Tcf7l2 in regulating β-cell function were suggested (17).

β-Cells rely on extrinsic cues, including those provided by cells of the islet microenvironment, for their proper function (19,20). We and others recently showed that pericytes, which together with endothelial cells make the dense islet capillary network, support β-cell function and glucose homeostasis (21,22). Although abnormalities in islet pericytes were implicated in obesity and type 2 diabetes (23), whether impaired pericyte function contributes to β-cell dysfunction and disease progression remains an open question.

Profiling gene expression of pancreatic pericytes revealed the expression of Tcf7l2 in these cells. We hypothesized that Tcf7l2 activity in pancreatic pericytes is required for their ability to properly support β-cell function. To test our hypothesis, we selectively inactivated this transcription factor in these cells by combining two transgenic mouse lines: Tcf7l2flox, which allows Cre-mediated deletion of this transcription factor DNA-binding domain (24), and Nkx3.2-Cre (25), which selectively targets mural cells of the pancreas (21). Our results show an impaired glucose tolerance but intact insulin sensitivity in male mice homozygous for mutated Tcf7l2. Our analysis pointed to impaired GSIS and reduced expression of genes required for β-cell function and maturity upon inactivation of pericytic Tcf7l2. Lastly, we linked Tcf7l2-dependent pericytic expression of bone morphogenetic protein 4 (BMP4) to glucose regulation. To conclude, our results indicate that pericytic Tcf7l2 activity is required for β-cell function and glucose homeostasis. Our findings further point to the contribution of abnormal pericytes activity to diabetes progression.

Mice

All experiments were performed according to protocols approved by the Tel Aviv University Committee on Animal Research. Nkx3.2-Cre (Nkx3–2tm1(cre)Wez) (25) and Tcf7l2flox (Tcf7l2tm2.1Cle) (24) mice were gifts from Warren Zimmer (Texas A&M University, College Station, TX) and Hans Clevers (Hubrecht Institute, Utrecht, the Netherlands), respectively. R26-yellow fluorescent protein (YFP; enhanced [E]YFP) (Gt(ROSA)26Sortm1(EYFP)Cos) mice were obtained from The Jackson Laboratory. For diet-induced obesity, mice were fed a high-fat diet (HFD; 60% fat [kCal]; Teklad) beginning at 6 weeks of age. Mice were intraperitoneally injected with dextrose (Sigma-Aldrich), insulin (Lilly), or mouse recombinant BMP (rBMP4; R&D) when indicated. For analysis of functional vasculature, fluorescein-labeled tomato lectin (Vector) was injected intravenously and allowed to circulate for 5 min before the animal was euthanized.

Flow Cytometry

Cell isolation was performed as described (26). Cells were stained with primary antibodies (Supplementary Table 1) when indicated and collected using FACSAria (BD Biosciences) or analyzed using a Gallios flow cytometer (Beckman Coulter) and Kaluza software (Beckman Coulter).

Hormone Detection

Islets were isolated according to standard protocols (21). For insulin secretion, after overnight culture, isolated islets were incubated in RPMI medium supplemented with glucose for 1 h. Pancreas and islet insulin was extracted by overnight incubation in 1.5% HCl and 70% ethanol mixture. Hormone levels were determined using mouse Ultrasensitive Insulin ELISA (Alpco), mouse Proinsulin ELISA (Alpco), and glucagon-like peptide 1 (GLP-1[7-36]) Active Elisa kit (Millipore).

Immunofluorescence

Dissected tissues were fixed in paraformaldehyde (4%), followed by cryosectioning. Tissue sections were stained with primary antibodies (Supplementary Table 1), followed by secondary fluorescent antibodies (Alexa Fluor; Invitrogen). After tissues were stained with anti-Tcf7l2 antibody, the TSA Fluorescein System (PerkinElmer) was used. The In Situ Cell Death Detection Kit (Roche) was used for TUNEL assay. Images were acquired using BZ-9000 BioRevo (Keyence) and SP8 confocal (Leica) microscopes.

Morphometric Analysis

Analysis of islet vasculature was performed as described (21). For measurement of β-cell mass, immunostained paraffin-embedded tissue sections were counterstained with HCS CellMask Stain (Invitrogen) to label the whole tissue sections. Sections were automatically imaged using IN Cell 2000 analyzer (GE Healthcare) and analyzed by developer software (GE Healthcare).

Gene Expression

RNA was extracted using PureLink RNA Micro kit (Invitrogen). For RNA deep sequencing, amplification, cDNA library preparation, sequencing, and bioinformatics analysis were performed using commercial services (Otogenetics). Gene expression data have been deposited in ArrayExpress (https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-5325/). For quantitative (q)PCR analysis, TaqMan and SYBR Green assays (Invitrogen) (Supplementary Table 2) were used, normalized to GAPDH and cyclophilin expression, respectively.

Statistics

Paired data were evaluated using two-tailed Student t test.

Pancreatic Pericytes Express the Transcription Factor Tcf7l2

We recently showed that pericytes support β-cell function (21). Here, we set to elucidate the molecular basis of pericyte activity. To this end, we used the Nkx3.2-Cre mouse line (25) to manipulate pancreatic mural cells. Nkx3.2 (Bapx1) is expressed in the mesenchymal compartment of the embryonic gut, stomach, and pancreatic buds, as well as in skeletal somites (27). We recently showed that in the adult pancreas, Nkx3.2-Cre line targets mural cells, including islet pericytes and vascular smooth muscle cells (vSMCs), but no other pancreatic cell types, including epithelial and endothelial cells (21) (Supplementary Fig. 1). Pericytes in the exocrine pancreas, identified by expression of neural/glial antigen 2 (NG2) and desmin, were also targeted by this Cre as apparent fluorescent labeling of these cells in the pancreas of Nkx3.2-Cre;R26-YFP mice (Supplementary Fig. 1). Of note, this mouse line does not target hepatic pericytes, which were shown to regulate insulin response (28) (Supplementary Fig. 1); thus, our analysis indicated that the Nkx3.2-Cre line targets mural cells in the endocrine and exocrine pancreas.

We next characterized pancreatic mural cells by profiling their gene expression. To this end, cells were sorted from pancreatic tissues of Nkx3.2-Cre;R26-YFP mice based on their fluorescent labeling (Supplementary Fig. 1). Of note, vast majority of labeled cells express platelet-derived growth factor receptor-β (PDGFR-β), which is expressed by pericytes but not vSMCs (Supplementary Fig. 1) (29). RNA was extracted from sorted cells and subjected to deep sequencing (Supplementary Table 3). Gene Ontology term analysis revealed that pancreatic mural cells were enriched with components of Wnt signaling (Fig. 1A; 126 genes). Interestingly, these cells expressed two of the four mammalian TCF/LEF transcription factors, Tcf7l1 and Tcf7l2 (Fig. 1B). Considering the association of polymorphism in TCF7L2 with β-cell dysfunction and diabetes, we focused our analysis on this transcription factor.

Figure 1

Expression of Tcf7l2 gene and protein by pancreatic pericytes. A: Bar diagram shows the number of genes (x-axis) clustered to Gene Ontology (GO) terms (y-axis) enriched in RNA sequencing analysis of purified pancreatic mural cells, FACS sorted from Nkx3.2-Cre;R26-EYFP pancreatic tissue based on their yellow fluorescent labeling (as shown in Supplementary Fig. 1) (n = 3). B: Heat map shows expression levels (as fragments per kilobase of exon per million aligned fragments [FPKM]) of TCF/LEF family members in RNA sequencing analysis of pancreatic mural cells, as described in panel A. C: Box-and-whisker plot shows relative levels of Tcf7l2 transcript by qPCR analysis. RNA was extracted from bulk pancreatic tissue, isolated islets (average was set to “1”), and purified pancreatic mural cells, and gene expression level was analyzed (n = 4–5). ***P < 0.005. DF: Immunofluorescence analysis of pancreatic tissue sections from adult wild-type mouse. D: Sections were stained for NG2 (red) to label mural cells and Tcf7l2 (green) and counterstained with DAPI (blue). Middle and right panels show higher magnification of the area framed in the white box in the left panel. Note the presence of nuclear Tcf7l2 in pericytes. E: Sections were stained for insulin (white), NG2 (red), and Tcf7l2 (green). Right panel shows higher magnification of the area framed in the white box in the left panel. Note the presence of nuclear Tcf7l2 in islet-associated pericytes. F: Sections were stained for α-SMA (white) to label vSMCs, platelet endothelial cell adhesion molecule 1 (PECAM1; red) to label endothelial cells, and Tcf7l2 (green). Note Tcf7l2 is absent from nuclei of vSMCs. Representative fields are shown.

Figure 1

Expression of Tcf7l2 gene and protein by pancreatic pericytes. A: Bar diagram shows the number of genes (x-axis) clustered to Gene Ontology (GO) terms (y-axis) enriched in RNA sequencing analysis of purified pancreatic mural cells, FACS sorted from Nkx3.2-Cre;R26-EYFP pancreatic tissue based on their yellow fluorescent labeling (as shown in Supplementary Fig. 1) (n = 3). B: Heat map shows expression levels (as fragments per kilobase of exon per million aligned fragments [FPKM]) of TCF/LEF family members in RNA sequencing analysis of pancreatic mural cells, as described in panel A. C: Box-and-whisker plot shows relative levels of Tcf7l2 transcript by qPCR analysis. RNA was extracted from bulk pancreatic tissue, isolated islets (average was set to “1”), and purified pancreatic mural cells, and gene expression level was analyzed (n = 4–5). ***P < 0.005. DF: Immunofluorescence analysis of pancreatic tissue sections from adult wild-type mouse. D: Sections were stained for NG2 (red) to label mural cells and Tcf7l2 (green) and counterstained with DAPI (blue). Middle and right panels show higher magnification of the area framed in the white box in the left panel. Note the presence of nuclear Tcf7l2 in pericytes. E: Sections were stained for insulin (white), NG2 (red), and Tcf7l2 (green). Right panel shows higher magnification of the area framed in the white box in the left panel. Note the presence of nuclear Tcf7l2 in islet-associated pericytes. F: Sections were stained for α-SMA (white) to label vSMCs, platelet endothelial cell adhesion molecule 1 (PECAM1; red) to label endothelial cells, and Tcf7l2 (green). Note Tcf7l2 is absent from nuclei of vSMCs. Representative fields are shown.

Close modal

To validate Tcf7l2 expression, we performed qPCR, Western blot, and immunofluorescence analyses. Tcf7l2 transcript and protein were detected in purified pancreatic mural cells (Fig. 1C and Supplementary Fig. 2). Notably, Tcf7l2 mRNA levels in mural cells were 5-fold higher than in islets and 22-fold higher than in bulk pancreatic tissue (Fig. 1C). Tcf7l2 protein was detected in the nuclei of pancreatic pericytes, including those associated with islets (Fig. 1D and E), but not in the nuclei of vSMCs, identified by expression of α-smooth muscle actin (α-SMA) and localization around large blood vessels (Fig. 1F). In agreement with previous studies reporting low Tcf7l2 transcript and protein levels in pancreatic endocrine cells (3033), we did not detect this transcription factor in β-cells and isolated islets (Fig. 1E and Supplementary Fig. 2). To conclude, our analyses revealed the expression of Tcf7l2 by pancreatic pericytes.

Tcf7l2 Activity in Pancreatic Pericytes Is Required for Glucose Homeostasis

To test the requirement of pericytic Tcf7l2 for glucose regulation, we set to interfere with this transcription factor activity in these cells. The diabetes-associated T allele of rs7903146 variant was predicted to result in an inactive Tcf7l2 protein lacking its DNA-binding domain (9). We therefore used a previously described transgenic mouse line, Tcf7l2flox, allowing Cre-mediated deletion of the endogenous Tcf7l2 DNA-binding domain, rendering it inactive (24). Of note, all splice variants are present in mice carrying this transgene (24). To selectively inactive this transcription factor in pancreatic pericytes, the Tcf7l2flox transgenic mouse line was crossed with the Nkx3.2-Cre line. To verify recombination of the Tcf7l2 locus, we analyzed its transcript levels by using primers providing detection of wild-type Tcf7l2 but not its recombined form (12,24). To allow isolation of pancreatic mural cells by flow cytometry (as described in Supplementary Fig. 1), a R26-YFP transgene was included to generate Nkx3.2-Cre;R26-YFP;Tcf7l2flox/+ and Nkx3.2-Cre;R26-YFP;Tcf7l2flox/flox mice, as well as Nkx3.2-Cre;R26-YFP control mice. As shown in Fig. 2A, wild-type Tcf7l2 was nearly absent from pancreatic mural cells of homozygous mice and was significantly reduced in cells of heterozygous mice compared with control mice. Of note, the expression levels of wild-type Tcf7l2 in islets isolated from homozygous and heterozygous mice were comparable to those of controls (Fig. 2B).

Figure 2

Male mice with a disrupted pericytic Tcf7l2 activity are glucose intolerant. A: Bar diagram shows relative levels of wild-type (wt) Tcf7l2 transcript in mural cells by qPCR analysis. RNA was extracted from purified pancreatic mural cells (Supplementary Fig. 1) of Nkx3.2-Cre;R26-YFP;Tcf7l2flox/flox (f, flox), Nkx3.2-Cre;R26-YFP;Tcf7l2flox/+, and Nkx3.2-Cre;R26-YFP (average was set to “1”) mice, and gene expression was analyzed (n = 3–4).***P < 0.005 vs. Nkx3.2-Cre;R26-YFP control mice. B: Bar diagram shows relative levels of wild-type Tcf7l2 transcript in islets by qPCR analysis. RNA was extracted from isolated islets of Nkx3.2-Cre;Tcf7l2flox/flox, Nkx3.2-Cre;Tcf7l2flox/+, and nontransgenic (Non tg) littermate (Cre-negative; average was set to “1”) mice, and gene expression was analyzed (n = 4). C and D: IPGTT on Nkx3.2-Cre;Tcf7l2flox/flox, Nkx3.2-Cre;Tcf7l2flox/+, and nontransgenic littermate (Cre-negative) male mice at 13 weeks of age. Mice were fed regular chow (n = 8–12) (C) or the HFD (n = 6–9) (D). Mice fed the HFD that reached a body weight of >32 g at 13 weeks of age were considered obese and used for analysis. Mice were fasted overnight and intraperitoneally injected with dextrose (2 mg/g body weight). Tail vein blood glucose levels were measured at indicated times. Left and middle panels show mean ± SEM blood glucose levels. The box-and-whisker plots on the right show the area under the curve (AUC) of the glucose responses shown in the left and middle panels. *P < 0.05; *** P < 0.005; NS, compared with nontransgenic control.

Figure 2

Male mice with a disrupted pericytic Tcf7l2 activity are glucose intolerant. A: Bar diagram shows relative levels of wild-type (wt) Tcf7l2 transcript in mural cells by qPCR analysis. RNA was extracted from purified pancreatic mural cells (Supplementary Fig. 1) of Nkx3.2-Cre;R26-YFP;Tcf7l2flox/flox (f, flox), Nkx3.2-Cre;R26-YFP;Tcf7l2flox/+, and Nkx3.2-Cre;R26-YFP (average was set to “1”) mice, and gene expression was analyzed (n = 3–4).***P < 0.005 vs. Nkx3.2-Cre;R26-YFP control mice. B: Bar diagram shows relative levels of wild-type Tcf7l2 transcript in islets by qPCR analysis. RNA was extracted from isolated islets of Nkx3.2-Cre;Tcf7l2flox/flox, Nkx3.2-Cre;Tcf7l2flox/+, and nontransgenic (Non tg) littermate (Cre-negative; average was set to “1”) mice, and gene expression was analyzed (n = 4). C and D: IPGTT on Nkx3.2-Cre;Tcf7l2flox/flox, Nkx3.2-Cre;Tcf7l2flox/+, and nontransgenic littermate (Cre-negative) male mice at 13 weeks of age. Mice were fed regular chow (n = 8–12) (C) or the HFD (n = 6–9) (D). Mice fed the HFD that reached a body weight of >32 g at 13 weeks of age were considered obese and used for analysis. Mice were fasted overnight and intraperitoneally injected with dextrose (2 mg/g body weight). Tail vein blood glucose levels were measured at indicated times. Left and middle panels show mean ± SEM blood glucose levels. The box-and-whisker plots on the right show the area under the curve (AUC) of the glucose responses shown in the left and middle panels. *P < 0.05; *** P < 0.005; NS, compared with nontransgenic control.

Close modal

We analyzed glucose response in mice by performing intraperitoneal glucose tolerance tests (IPGTTs) on three groups of mice: homozygous for the inactive Tcf7l2 allele (Nkx3.2-Cre;Tcf7l2flox/flox), heterozygous for this allele (Nkx3.2-Cre;Tcf7l2flox/+), and nontransgenic controls (Cre negative: Tcf7l2flox/+ or Tcf7l2flox/flox). Of note, mice expressing the Nkx3.2-Cre transgene by itself (i.e., do not carry the Tcf7l2flox transgene) displayed comparable glucose response to Cre-negative control mice (Supplementary Fig. 3). As shown in Fig. 2C, our analysis revealed that 13-week-old homozygous, but not heterozygous male mice, display an impaired glucose response compared with littermate controls (Fig. 2C). The TCF7L2 rs7903146 T-allele has a modest effect on β-cell function, which becomes more evident when insulin action decreases (34). We tested whether metabolic stress aggravates glucose intolerance of transgenic mice by feeding mice the HFD (60% fat) to induce obesity. Our analysis revealed that heterozygous and homozygous obese animals were glucose intolerant (Fig. 2D). Thus, our findings indicate that reduced levels of active Tcf7l2 in pericytes (in heterozygous mice) were sufficient to maintain glucose response in lean but not obese mice and that its complete loss (in homozygous mice) induced glucose intolerance in both lean and obese animals.

Polymorphism in TCF7L2 is associated with an increased risk of diabetes in women and men (8); however, we did not observe differences in glucose response in transgenic and control female mice (Supplementary Fig. 3). Glucose metabolism differs in female and male mice (35); thus, sex-dependent differences may underlie the distinct phenotype observed in female and male Nkx3.2-Cre;Tcf7l2flox/flox mice.

The Nkx3.2-Cre mouse line has nonpancreatic expression in the gastrointestinal mesenchyme and skeleton (25,27). We therefore analyzed for potential changes in function of these tissues in Nkx3.2-Cre;Tcf7l2flox/flox and Nkx3.2-Cre;Tcf7l2flox/+ mice that could contribute to their glucose intolerance. The three analyzed mouse groups show comparable body weight when fed both regular chow and HFD, indicating normal food uptake and digestion (Supplementary Fig. 4). Next, we analyzed for GLP-1 production by analyzing gut expression of Pcsk1 and Gcg (encoding prohormone convertases 1/3 and proglucagon, respectively) and measuring serum GLP-1 levels, and found them comparable in Nkx3.2-Cre;Tcf7l2flox/flox and control mice (Supplementary Fig. 4). Finally, insulin sensitivity was comparable between Nkx3.2-Cre;Tcf7l2flox/flox, Nkx3.2-Cre;Tcf7l2flox/+, and control (Cre-negative) male mice fed the normal diet and HFD (Supplementary Fig. 4).

To conclude, our results implicate that Tcf7l2 activity in pancreatic pericytes is required for glucose regulation in vivo, without affecting insulin sensitivity. Furthermore, our analysis suggests that the requirement of pericytic Tcf7l2 activity for glucose regulation is more evident upon metabolic stress.

Functional Islet Vasculature in Nkx3.2-Cre;Tcf7l2flox/flox Transgenic Mice

Pericytes support endothelial cell function and blood flow (29). We therefore analyzed whether Tcf7l2 inactivation in pancreatic pericytes interferes with islet vascularization. To this end, Nkx3.2-Cre;Tcf7l2flox/flox and control mice were intravenously injected with tomato lectin to label functional vessels. Our analysis revealed intact islet vascularization distribution and density in transgenic mice (Fig. 3A and B). In agreement, expression levels of the hypoxia gene Hif1a was comparable in islets isolated from the two mouse groups (Fig. 3C). Thus, our analysis indicates that loss of pericytic Tcf7l2 activity did not interfere with functionality of islet vasculature.

Figure 3

Functional islet vasculature upon inactivation of pericytic Tcf7l2. Nkx3.2-Cre;Tcf7l2flox/flox (f, flox) transgenic and nontransgenic (Non tg) control (Cre-negative) mice were analyzed. A and B: Mice were intravenously injected with 1 mg/mL tomato lectin (red) to label function vessels. Pancreata were harvested from treated mice, and tissue sections were stained for insulin (blue). A: Images show representative islets. Upper panels show lectin labeling and anti-insulin staining. Lower panels show lectin labeling alone. The white lines demark the outer border of the insulin+ area, as shown in the upper panels. B: Bar graph diagram (mean ± SD) shows quantification of intraislet vascular density. The relative ratio of lectin+ and insulin+ area per each islet was calculated. Thirty islets per mouse, from sections at least 100 μm apart, were analyzed (n = 3). NS, compared with nontransgenic control. C: Box-and-whisker plot shows gene expression analysis of the hypoxic marker Hif1a in isolated islets. RNA was extracted and gene expression was analyzed by qPCR, when average levels in nontransgenic islets were set to “1” (n = 5).

Figure 3

Functional islet vasculature upon inactivation of pericytic Tcf7l2. Nkx3.2-Cre;Tcf7l2flox/flox (f, flox) transgenic and nontransgenic (Non tg) control (Cre-negative) mice were analyzed. A and B: Mice were intravenously injected with 1 mg/mL tomato lectin (red) to label function vessels. Pancreata were harvested from treated mice, and tissue sections were stained for insulin (blue). A: Images show representative islets. Upper panels show lectin labeling and anti-insulin staining. Lower panels show lectin labeling alone. The white lines demark the outer border of the insulin+ area, as shown in the upper panels. B: Bar graph diagram (mean ± SD) shows quantification of intraislet vascular density. The relative ratio of lectin+ and insulin+ area per each islet was calculated. Thirty islets per mouse, from sections at least 100 μm apart, were analyzed (n = 3). NS, compared with nontransgenic control. C: Box-and-whisker plot shows gene expression analysis of the hypoxic marker Hif1a in isolated islets. RNA was extracted and gene expression was analyzed by qPCR, when average levels in nontransgenic islets were set to “1” (n = 5).

Close modal

β-Cell Dysfunction Upon Pericytic Tcf7l2 Inactivation

We previously showed that islet pericytes support β-cell function (21). We therefore set to determine whether β-cell dysfunction underlies the observed glucose intolerance upon loss of Tcf7l2 activity in pancreatic pericytes. We tested whether the impaired glucose response is associated with insufficient GSIS by measuring glucose-stimulated serum insulin levels and found they were lower in Nkx3.2-Cre;Tcf7l2flox/flox mice (Fig. 4A). Next, we analyzed whether β-cell mass or function, or both, were affected in Tcf7l2-deficient mice. Our analysis indicated normal islet morphology and comparable pancreatic and β-cell mass in transgenic mice (Fig. 4B–D). In agreement with these findings, we observed neither β-cell death nor expression of genes associated with β-cell stress in islets of transgenic mice (i.e., Chop, Atf4) (Supplementary Fig. 5). To test for potential changes in β-cell function, we analyzed GSIS of isolated islets and found that those of transgenic mice secreted less insulin in response to high glucose levels compared with control islets (Fig. 4E). Thus, our analysis points to β-cell dysfunction upon inactivation of pericytic Tcf7l2.

Figure 4

Impaired β-cell function and gene expression in Nkx3.2-Cre;Tcf7l2flox/flox (f, flox) mice. Nkx3.2-Cre;Tcf7l2flox/flox transgenic and nontransgenic (Non tg) control (Cre-negative) 13-week-old mice were analyzed. A: Glucose-stimulated serum insulin levels. After an overnight fast, dextrose (2 mg/g body weight) was intraperitoneally injected, and tail vein blood was collected at the indicated times from transgenic and control mice. Shown are mean ± SEM blood insulin levels (n = 4–5). B: Pancreatic tissues of nontransgenic (left) and transgenic (right) mice were stained for insulin (green) and glucagon (red). Representative islets are shown. Bar diagrams (mean ± SD) show pancreatic weight (n = 3) (C) and estimated β-cell mass (D). For each analyzed mouse, insulin+ and pancreatic (labeled by HCS CellMask Stain) areas were measured in sections at least 100 μm apart from each other, representing 20% of pancreatic area. For each analyzed mouse, insulin+ area was divided by pancreatic area and multiplied by pancreatic weight (n = 3). E and E’: Bar diagrams (mean ± SD) show impaired GSIS by transgenic islets. Isolated islets from transgenic and control mice (n = 4) were incubated with low (1.67 mmol/L) or high (16.7 mmol/L) glucose, and the supernatant was collected and analyzed by ELISA. For each mouse, three groups of 10 islets were analyzed for each condition. Shown are secreted insulin levels normalized to islets insulin content (E’) or not normalized (E). Bar diagrams (mean ± SD) show reduced insulin content in pancreatic tissues (F) and isolated islets (G) of transgenic mice, but comparable islet proinsulin-to-insulin ratio (I). Insulin and proinsulin content of isolated islets were analyzed after overnight culture. Pancreatic insulin content was normalized to total pancreatic protein content (n = 4). H and JM: Box-and-whisker plots show impaired gene expression in Nkx3.2-Cre;Tcf7l2flox/flox islets. RNA was extracted from islets isolated from transgenic and control mice (n = 4–5), and expression of indicated genes was analyzed by qPCR, when average levels in nontransgenic islets were set to “1.” *P < 0.05; **P < 0.01; ***P < 0.005; NS, compared with nontransgenic control.

Figure 4

Impaired β-cell function and gene expression in Nkx3.2-Cre;Tcf7l2flox/flox (f, flox) mice. Nkx3.2-Cre;Tcf7l2flox/flox transgenic and nontransgenic (Non tg) control (Cre-negative) 13-week-old mice were analyzed. A: Glucose-stimulated serum insulin levels. After an overnight fast, dextrose (2 mg/g body weight) was intraperitoneally injected, and tail vein blood was collected at the indicated times from transgenic and control mice. Shown are mean ± SEM blood insulin levels (n = 4–5). B: Pancreatic tissues of nontransgenic (left) and transgenic (right) mice were stained for insulin (green) and glucagon (red). Representative islets are shown. Bar diagrams (mean ± SD) show pancreatic weight (n = 3) (C) and estimated β-cell mass (D). For each analyzed mouse, insulin+ and pancreatic (labeled by HCS CellMask Stain) areas were measured in sections at least 100 μm apart from each other, representing 20% of pancreatic area. For each analyzed mouse, insulin+ area was divided by pancreatic area and multiplied by pancreatic weight (n = 3). E and E’: Bar diagrams (mean ± SD) show impaired GSIS by transgenic islets. Isolated islets from transgenic and control mice (n = 4) were incubated with low (1.67 mmol/L) or high (16.7 mmol/L) glucose, and the supernatant was collected and analyzed by ELISA. For each mouse, three groups of 10 islets were analyzed for each condition. Shown are secreted insulin levels normalized to islets insulin content (E’) or not normalized (E). Bar diagrams (mean ± SD) show reduced insulin content in pancreatic tissues (F) and isolated islets (G) of transgenic mice, but comparable islet proinsulin-to-insulin ratio (I). Insulin and proinsulin content of isolated islets were analyzed after overnight culture. Pancreatic insulin content was normalized to total pancreatic protein content (n = 4). H and JM: Box-and-whisker plots show impaired gene expression in Nkx3.2-Cre;Tcf7l2flox/flox islets. RNA was extracted from islets isolated from transgenic and control mice (n = 4–5), and expression of indicated genes was analyzed by qPCR, when average levels in nontransgenic islets were set to “1.” *P < 0.05; **P < 0.01; ***P < 0.005; NS, compared with nontransgenic control.

Close modal

Next, we measured insulin content in isolated islets and pancreatic tissues from Nkx3.2-Cre;Tcf7l2flox/flox and control animals and found it was significantly reduced in homozygous animals (Fig. 4F and G). Although gene expression analysis indicated that the levels of Ins1 and Ins2 transcripts, encoding insulin, were on average lower in transgenic islets, this reduction was not statistically significant (Fig. 4H). We detected a statistically significant reduction in expression levels of Pcsk1, required for posttranslational processing of insulin, in Nkx3.2-Cre;Tcf7l2flox/flox islets (Fig. 4H), but the ratio between proinsulin and insulin levels was comparable in transgenic and control islets (Fig. 4I). In addition, Gcg levels were comparable in Nkx3.2-Cre;Tcf7l2flox/flox and control islets (Fig. 4J). To conclude, we observed reduced islet and pancreatic insulin content upon inactivation of pericytic Tcf7l2.

Nkx3.2-Cre;Tcf7l2flox/flox islets secrete a smaller portion of their insulin content in response to glucose challenge, indicating an impaired GSIS independent of abrogated insulin production (Fig. 4E’). To determine whether the observed impaired GSIS in Nkx3.2-Cre;Tcf7l2flox/flox islets is associated with changes in expression levels of genes required for glucose sensing and insulin secretion, we analyzed islets for expression of Glut2, Kir6.2, and Sur1. As shown in Fig. 4K, all three genes were expressed at lower levels in Nkx3.2-Cre;Tcf7l2flox/flox islets than in control islets. In contrast, expression of Glpr1, encoding the GLP-1 receptor, was comparable in transgenic and control islets (Fig. 4L). β-Cell function and gene expression have been shown to depend on transcription factors, including MafA, Pdx1, and NeuroD1 (2). We observed reduced transcripts level of all three in islets from Nkx3.2-Cre;Tcf7l2flox/flox mice compared with control islets (Fig. 4M). Thus, inactivation of pericytic Tcfl72 is associated with impaired expression of β-cell genes.

To conclude, our findings indicate that although β-cell mass was unaffected in Tcf7l2 transgenic mice, their functionality was impaired. Furthermore, our analysis suggests that normal expression of β-cell genes associated with their function and maturity is dependent on Tcf7l2 activity in pancreatic pericytes.

Tcf7l2 Regulates Pericytic Expression of Ligands Shown to Support β-Cell Function

Our findings point to a Tcf7l2-dependent activity of pancreatic pericytes in supporting β-cell function and gene expression. We therefore analyzed potential changes in pancreatic pericytes upon inactivation of Tcf7l2. Our immunofluorescence analysis revealed that pericytes are localized in proximity to endothelial cells in control and in transgenic islets (Fig. 5A). However, morphometric analysis revealed that pericyte coverage was mildly reduced in islets of Nkx3.2-Cre;Tcf7l2flox/flox mice compared with control islets (by 7%) (Fig. 5B).

Figure 5

Reduced expression of pericytic secreted ligands upon Tcf7l2 inactivation. A and B: Islet pericyte density is mildly reduced upon loss of Tcf7l2 activity. Pancreatic tissues of Nkx3.2-Cre;Tcf7l2flox/flox (f, flox) transgenic (right panel) and nontransgenic (Non tg; left panel) control mice (Cre-negative) were stained for NG2 (red) to label pericytes, platelet endothelial cell adhesion molecule 1 (PECAM1; green) to label endothelial cells, and insulin to label β-cells. A: Images of representative islets show normal distribution of pericytes in transgenic islets. The white lines demark the outer border of insulin+ area. B: Bar diagram (mean ± SD) shows morphometric analysis of intraislet pericyte density. Pancreatic tissues were stained as described, and the relative ratio of NG2+ and insulin+ area per each islet was calculated. Fifty islets per mouse, from sections at least 100 μm apart, were analyzed (n = 3). *P < 0.05 compared with nontransgenic control. C: Analysis of Tcf7l2-dependent expression of secreted factors by pancreatic mural cells. Mural cells were isolated from pancreatic tissues of 10-week-old Nkx3.2-Cre;R26R-YFP (Non tg; middle panel) and Nkx3.2-Cre;R26R-YFP;Tcf7l2flox/flox (Tcf7l2f/f; right panel), as described in Supplementary Fig. 1. RNA was extracted from pancreatic mural cells and islets isolated from age-matched nontransgenic mice (left panels) and subjected to deep sequencing. Heat map shows levels of indicated transcripts in fragments per kilobase of exon per million aligned fragments (FPKM). *P < 0.05; ***P < 0.005, compared with nontransgenic control.

Figure 5

Reduced expression of pericytic secreted ligands upon Tcf7l2 inactivation. A and B: Islet pericyte density is mildly reduced upon loss of Tcf7l2 activity. Pancreatic tissues of Nkx3.2-Cre;Tcf7l2flox/flox (f, flox) transgenic (right panel) and nontransgenic (Non tg; left panel) control mice (Cre-negative) were stained for NG2 (red) to label pericytes, platelet endothelial cell adhesion molecule 1 (PECAM1; green) to label endothelial cells, and insulin to label β-cells. A: Images of representative islets show normal distribution of pericytes in transgenic islets. The white lines demark the outer border of insulin+ area. B: Bar diagram (mean ± SD) shows morphometric analysis of intraislet pericyte density. Pancreatic tissues were stained as described, and the relative ratio of NG2+ and insulin+ area per each islet was calculated. Fifty islets per mouse, from sections at least 100 μm apart, were analyzed (n = 3). *P < 0.05 compared with nontransgenic control. C: Analysis of Tcf7l2-dependent expression of secreted factors by pancreatic mural cells. Mural cells were isolated from pancreatic tissues of 10-week-old Nkx3.2-Cre;R26R-YFP (Non tg; middle panel) and Nkx3.2-Cre;R26R-YFP;Tcf7l2flox/flox (Tcf7l2f/f; right panel), as described in Supplementary Fig. 1. RNA was extracted from pancreatic mural cells and islets isolated from age-matched nontransgenic mice (left panels) and subjected to deep sequencing. Heat map shows levels of indicated transcripts in fragments per kilobase of exon per million aligned fragments (FPKM). *P < 0.05; ***P < 0.005, compared with nontransgenic control.

Close modal

Next, we analyzed whether loss of Tcf7l2 activity affects their gene expression. To this end, pancreatic mural cells were purified by flow cytometry (as described in Supplementary Fig. 1) from Nkx3.2-Cre;R26-YFP;Tcf7l2flox/flox and control Nkx3.2-Cre;R26-YFP mice, and islets were isolated from nontransgenic mice and their RNA was extracted and sequenced. We hypothesized that pericytes secrete factors to support β-cells under normal conditions and that the expression of some of these factors is Tcf7l2 dependent. We thus focused our analysis on genes that encode secreted ligands that are expressed by nontransgenic pancreatic mural cells but not by isolated islets (Supplementary Table 3). The expression levels of seven of these genes were significantly lower in Tcf7l2-transgenic pancreatic mural cells (Fig. 5C and Supplementary Table 3): Bmp4, Ccl2, Ccl7, C7, Il6, Fam150b, and Nmb. Tcf7l2 was previously shown to directly regulate the expression of first three genes (3638). Importantly, β-cells were shown to express the receptors for BMP4, neuromedin B, and interleukin (IL)-6 (encoded by Bmp4, Nmb, and Il6, respectively), and these three factors were implicated in β-cell function (3941).

To conclude, our analysis showed that lack of Tcf7l2 activity in pancreatic pericytes led to mildly reduced islet pericyte density and further resulted in impaired expression of secreted ligands by these cells, some of which were implicated in β-cell function.

Exogenous BMP4 Rescues Impaired Glucose Response in Tcfl72-Deficient Mice

β-Cells were shown to depend on the activity of the BMP4 receptor, BMPR1A, for their proper function in vivo (40). In addition, elevating BMP4 levels in vivo (by pancreatic expression of a Bmp4 transgene or systemic administration of rBMP4) improved the glucose response in mice and promoted the mature β-cell phenotype (40). Importantly, Tcf7l2 was shown to directly promote Bmp4 expression through its identified binding sites on this gene promotor (36,42). We therefore hypothesized that BMP4 produced by pericytes in a Tcf7l2-dependent manner supports β-cell function. We tested our hypothesis by first validating pericytic Bmp4 expression by qPCR analysis, which revealed reduced transcript levels in Tcf7l2-deficient mural cells to a third of the level found in control cells (Fig. 6A).

Figure 6

Exogenous BMP4 rescues glucose intolerance of Tcf7l2-deficient mice. A: Analysis of Tcf7l2-dependent expression of Bmp4 by pancreatic mural cells. Nkx3.2/YFP+ mural cells were isolated by flow cytometry from pancreatic tissues of 10-week-old Nkx3.2-Cre;R26R-YFP (control) and Nkx3.2-Cre;R26R-YFP;Tcf7l2flox/flox (f, flox) mice. RNA was extracted and gene expression was analyzed by qPCR, when average levels in control samples were set to “1” (n = 4). *P < 0.05 compared with control. B and C: IPGTT and GSIS after treatment of homozygous mice with exogenous BMP4. Nkx3.2-Cre;Tcf7l2flox/flox mice were intraperitoneally injected twice daily with PBS or rBMP4 (20 ng/g body weight) for 3 consecutive days and a final treatment 30 min before analysis. rBMP4-treated Nkx3.2-Cre;Tcf7l2flox/flox (n = 3–4), PBS-treated Nkx3.2-Cre;Tcf7l2flox/flox (n = 5–6), and nontransgenic (Non tg) littermate (n = 5–8) male mice at 13 weeks of age were analyzed. After an overnight fast, mice were intraperitoneally injected with dextrose (2 mg/g body weight), and tail vein blood glucose and insulin levels were measured at indicated times. B: IPGTT. Left panel, blood glucose levels (mean ± SEM). Right panel, box-and-whisker plot shows area under the curve (AUC) of glucose responses, as shown in left panel. *P < 0.05; **P < 0.01; ***P < 0.005; NS, (left panel) compared with untreated transgenic controls and (right panel) as indicated by horizontal bars. C: GSIS. Insulin levels were measured by ELISA. *P < 0.05; ***P < 0.005 compared with untreated transgenic controls. D: Box-and-whisker plot shows expression of β-cell genes upon treatment of transgenic islets with rBMP4 ex vivo. Islets isolated from untreated Nkx3.2-Cre;Tcf7l2flox/flox mice were cultured in media supplemented with 6 ng/mL rBMP4 (+rBMP4) or unsupplemented media for 24 h. RNA was extracted, and expression levels of indicated genes were analyzed by qPCR, when average levels in untreated islets were set to “1” (n = 6). Representative of three independent experiments.*P < 0.05; ***P < 0.005 compared with untreated control.

Figure 6

Exogenous BMP4 rescues glucose intolerance of Tcf7l2-deficient mice. A: Analysis of Tcf7l2-dependent expression of Bmp4 by pancreatic mural cells. Nkx3.2/YFP+ mural cells were isolated by flow cytometry from pancreatic tissues of 10-week-old Nkx3.2-Cre;R26R-YFP (control) and Nkx3.2-Cre;R26R-YFP;Tcf7l2flox/flox (f, flox) mice. RNA was extracted and gene expression was analyzed by qPCR, when average levels in control samples were set to “1” (n = 4). *P < 0.05 compared with control. B and C: IPGTT and GSIS after treatment of homozygous mice with exogenous BMP4. Nkx3.2-Cre;Tcf7l2flox/flox mice were intraperitoneally injected twice daily with PBS or rBMP4 (20 ng/g body weight) for 3 consecutive days and a final treatment 30 min before analysis. rBMP4-treated Nkx3.2-Cre;Tcf7l2flox/flox (n = 3–4), PBS-treated Nkx3.2-Cre;Tcf7l2flox/flox (n = 5–6), and nontransgenic (Non tg) littermate (n = 5–8) male mice at 13 weeks of age were analyzed. After an overnight fast, mice were intraperitoneally injected with dextrose (2 mg/g body weight), and tail vein blood glucose and insulin levels were measured at indicated times. B: IPGTT. Left panel, blood glucose levels (mean ± SEM). Right panel, box-and-whisker plot shows area under the curve (AUC) of glucose responses, as shown in left panel. *P < 0.05; **P < 0.01; ***P < 0.005; NS, (left panel) compared with untreated transgenic controls and (right panel) as indicated by horizontal bars. C: GSIS. Insulin levels were measured by ELISA. *P < 0.05; ***P < 0.005 compared with untreated transgenic controls. D: Box-and-whisker plot shows expression of β-cell genes upon treatment of transgenic islets with rBMP4 ex vivo. Islets isolated from untreated Nkx3.2-Cre;Tcf7l2flox/flox mice were cultured in media supplemented with 6 ng/mL rBMP4 (+rBMP4) or unsupplemented media for 24 h. RNA was extracted, and expression levels of indicated genes were analyzed by qPCR, when average levels in untreated islets were set to “1” (n = 6). Representative of three independent experiments.*P < 0.05; ***P < 0.005 compared with untreated control.

Close modal

To study the contribution of reduced BMP4 production by pericytes to the observed phenotype in Nkx3.2-Cre;Tcf7l2flox/flox mice, we intraperitoneally injected rBMP4 to homozygous mice (40). As shown in Fig. 6, glucose tolerance and GSIS of rBMP4-treated animals were significantly improved compared with untreated transgenic animals. Importantly, glucose response and insulin secretion of rBMP4-treated Nkx3.2-Cre;Tcf7l2flox/flox mice were comparable to that of nontransgenic control animals (Fig. 6B and C).

To analyze whether the improved GSIS upon rBMP4 treatment of Nkx3.2-Cre;Tcf7l2flox/flox mice is associated with improvement in their β-cell mature phenotype, we cultured isolated islets in the presence or absence of this recombinant protein. Treating Nkx3.2-Cre;Tcf7l2flox/flox islets with rBMP4 for 24 h promoted the expression of Pdx1 and MafA, encoding transcription factors associated with mature β-cell phenotype (Fig. 6D). Of note, in agreement with previous studies (43,44), we could not observe changes in the genes expression levels in similarly treated wild-type islets (Supplementary Fig. 6).

To conclude, our analysis showed that extrinsic BMP4 was sufficient to rescue glucose response, GSIS, and mature β-cell gene expression in mice deficient of pericytic Tcf7l2. Thus, our findings suggest that pericytes support β-cell function through Tcf7l2-depedent production of BMP4.

Here, we show that pericytes support β-cell function and glucose response in a Tcf7l2-dependent manner. To analyze the requirement of this transcription factor for pericyte function, we selectively expressed a mutated form of Tcf7l2, lacking its DNA-binding domain, in pancreatic pericytes. Loss of pericytic Tcf7l2 activity interfered with glucose regulation in mice due to impaired β-cell function (Figs. 2 and 4). Our analysis further indicated Tcf7l2-dependent pericytic expression of secreted factors that were previously implicated in β-cell function, including BMP4 (Fig. 5). Finally, we showed that treatment of Tcf7l2-deficient mice with exogenous rBMP4 was sufficient to rescue their glucose intolerance and impaired GSIS phenotype (Fig. 6). Thus, we suggest that pancreatic pericytes express secreted factors in a Tcf7l2-dependent manner to support β-cell function and glucose response. Our findings propose that impaired pericytic activity perturbs β-cell function, thus potentially contributing to disrupted glucose regulation and diabetes progression.

Abnormalities in islet pericytes were implicated in obesity and diabetes (23). We, along with others, showed that pericytes directly support β-cell function and glucose regulation (21,22). Interestingly, SORCS1, a type 2 diabetes–associated gene that encodes a PDGF-binding protein, was suggested to regulate pericyte function (23,45). However, whether impaired pericyte activity contributes to diabetes progression remains an open question. The findings of this study suggest that diabetes-associated changes in pancreatic pericytes lead to impaired glucose regulation. This raises the possibility that abnormalities in the islet microenvironment in individuals with diabetes contribute to β-cell dysfunction and loss of glucose regulation.

Our analysis revealed that pericytes produce BMP4 in a Tcf7l2-dependent manner to support glucose response (Figs. 5 and 6). Treatment of Nkx3.2-Cre;Tcf7l2flox/flox mice with rBMP4 was sufficient to rescue their glucose tolerance, strengthening the importance of this factor in supporting proper β-cell function. Although the BMP4-BMPR1A pathway was shown to promote β-cell function and gene expression in vivo (40), treating isolated islets with rBMP4 did not affect β-cells or impair their survival and function (43,44), pointing to the requirement of regulated BMP4 levels for proper β-cell function. Loss of BMPR1A function in β-cells resulted in a more severe glucose intolerance phenotype than the one we observed upon loss of pericytic Tcf7l2 (40) (Fig. 2). These differences might reflect the presence of residual pericytic BMP4 in transgenic mice or additional pancreatic BMP4 sources, or both, as previously suggested (40).

Loss of pericytic Tcf7l2 activity was associated with a reduction in pancreatic and islet insulin content (by 50 and 38%, respectively) (Fig. 4). Together with the unaffected insulin-to-proinsulin ratio, this observation indicates an impaired insulin biosynthesis in Nkx3.2-Cre;Tcf7l2flox/flox mice. Ins1 and Ins2 transcript levels were an average of 39 and 33%, respectively, lower in transgenic islets (Fig. 4), but this reduction was not statistically significant. Thus, reduced insulin content accompanying the loss of pericytic Tcf7l2 could potentially result from an impaired posttranscriptional regulation of insulin biosynthesis, such as compromised proinsulin translation (46).

Tcf7l2 was proposed to support β-cell function in a cell-autonomous manner (14,15). Whether impaired Tcf7l2 activity in β-cells is sufficient to drive diabetes progression remains controversial, however (12,17,47,48). Accordingly, Tcf7l2 was suggested to regulate β-cell function in a nonautonomous manner (12,17); for example, pancreatic and nonpancreatic incretin production was recently shown to depend on this transcription factor (49,50). Our findings provide additional evidence for a nonautonomous role of Tcf7l2 in β-cell function through regulation the pericyte/β-cell axis.

Polymorphism in TCF7L2 has a strong correlation to type 2 diabetes in humans (3,5,6). In particular, multiple studies have shown that the T allele of rs7903146 increases by 30% the risk of developing this disease (5). Individuals harboring one or two copies of this allele display reduced basal and glucose-stimulated insulin secretion but maintain hepatic function (6,7,34). As shown in this study, mice lacking Tcf7l2 activity in their pancreatic pericytes were glucose intolerant but did not develop diabetes, even when fed the HFD (Fig. 2). These discrepancies could reflect the evident difference in the physiologies of humans and mice. Alternatively, TCF7L2 may act in multiple cell types to regulate glucose homeostasis, and loss of its activity in a single cell type is insufficient to drive diabetes progression. Lastly, although impaired TCF7L2 activity increases the risk of developing diabetes, additional genetic and environmental factors contribute to disease progression (1). Nevertheless, impaired β-cell function upon loss of pericytic TCF7L2 activity proposes a cellular mechanism through which mutations in this transcription factor predispose individuals to diabetes.

Acknowledgments. The authors thank Maya Avraham and Shani Mizrachi (Tel Aviv University) for technical assistance.

This work was performed in partial fulfillment of the requirements for a PhD degree for L.S. from the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.

Funding. This work was supported by European Research Council starting grant (336204) to L.L.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. L.S. conducted experiments, acquired and analyzed data, and wrote the manuscript. E.R., A.E., and H.C.G. conducted experiments and acquired and analyzed data. S.W.-A. analyzed data. M.L., L.K.-M., A.H., and D.B. conducted experiments and acquired data. A.P. and M.W. provided reagents. L.L. designed and supervised research, analyzed data, and wrote the manuscript. L.L. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the 2nd Joint Meeting of the European Association for the Study of Diabetes (EASD) Islet Study Group and Beta Cell Workshop, Dresden, Germany, 7–11 May 2017, and at the 77th Scientific Sessions of the American Diabetes Association, San Diego, CA, 9–13 June 2017.

1.
McCarthy
MI
.
Genomics, type 2 diabetes, and obesity
.
N Engl J Med
2010
;
363
:
2339
2350
[PubMed]
2.
Doria
A
,
Patti
ME
,
Kahn
CR
.
The emerging genetic architecture of type 2 diabetes
.
Cell Metab
2008
;
8
:
186
200
[PubMed]
3.
Grant
SFA
,
Thorleifsson
G
,
Reynisdottir
I
, et al
.
Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes
.
Nat Genet
2006
;
38
:
320
323
[PubMed]
4.
Nusse
R
,
Clevers
H
.
Wnt/β-catenin signaling, disease, and emerging therapeutic modalities
.
Cell
2017
;
169
:
985
999
[PubMed]
5.
Helgason
A
,
Pálsson
S
,
Thorleifsson
G
, et al
.
Refining the impact of TCF7L2 gene variants on type 2 diabetes and adaptive evolution
.
Nat Genet
2007
;
39
:
218
225
[PubMed]
6.
Lyssenko
V
,
Lupi
R
,
Marchetti
P
, et al
.
Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes
.
J Clin Invest
2007
;
117
:
2155
2163
[PubMed]
7.
Varghese
RT
,
Viegas
I
,
Barosa
C
, et al
.
Diabetes-associated variation in TCF7L2 is not associated with hepatic or extrahepatic insulin resistance
.
Diabetes
2016
;
65
:
887
892
[PubMed]
8.
Zhang
C
,
Qi
L
,
Hunter
DJ
, et al
.
Variant of transcription factor 7-like 2 (TCF7L2) gene and the risk of type 2 diabetes in large cohorts of U.S. women and men
.
Diabetes
2006
;
55
:
2645
2648
[PubMed]
9.
Locke
JM
,
Da Silva Xavier
G
,
Rutter
GA
,
Harries
LW
.
An alternative polyadenylation signal in TCF7L2 generates isoforms that inhibit T cell factor/lymphoid-enhancer factor (TCF/LEF)-dependent target genes
.
Diabetologia
2011
;
54
:
3078
3082
[PubMed]
10.
Savic
D
,
Ye
H
,
Aneas
I
,
Park
SY
,
Bell
GI
,
Nobrega
MA
.
Alterations in TCF7L2 expression define its role as a key regulator of glucose metabolism
.
Genome Res
2011
;
21
:
1417
1425
[PubMed]
11.
Oh
KJ
,
Park
J
,
Kim
SS
,
Oh
H
,
Choi
CS
,
Koo
SH
.
TCF7L2 modulates glucose homeostasis by regulating CREB- and FoxO1-dependent transcriptional pathway in the liver
.
PLoS Genet
2012
;
8
:
e1002986
[PubMed]
12.
Boj
SF
,
van Es
JH
,
Huch
M
, et al
.
Diabetes risk gene and Wnt effector Tcf7l2/TCF4 controls hepatic response to perinatal and adult metabolic demand
.
Cell
2012
;
151
:
1595
1607
[PubMed]
13.
Takamoto
I
,
Kubota
N
,
Nakaya
K
, et al
.
TCF7L2 in mouse pancreatic beta cells plays a crucial role in glucose homeostasis by regulating beta cell mass
.
Diabetologia
2014
;
57
:
542
553
[PubMed]
14.
da Silva Xavier
G
,
Mondragon
A
,
Sun
G
, et al
.
Abnormal glucose tolerance and insulin secretion in pancreas-specific Tcf7l2-null mice
.
Diabetologia
2012
;
55
:
2667
2676
[PubMed]
15.
Mitchell
RK
,
Mondragon
A
,
Chen
L
, et al
.
Selective disruption of Tcf7l2 in the pancreatic β cell impairs secretory function and lowers β cell mass
.
Hum Mol Genet
2015
;
24
:
1390
1399
[PubMed]
16.
Shao
W
,
Xiong
X
,
Ip
W
, et al
.
The expression of dominant negative TCF7L2 in pancreatic beta cells during the embryonic stage causes impaired glucose homeostasis
.
Mol Metab
2015
;
4
:
344
352
[PubMed]
17.
Bailey
KA
,
Savic
D
,
Zielinski
M
, et al
.
Evidence of non-pancreatic beta cell-dependent roles of Tcf7l2 in the regulation of glucose metabolism in mice
.
Hum Mol Genet
2015
;
24
:
1646
1654
[PubMed]
18.
Campbell
JE
,
Ussher
JR
,
Mulvihill
EE
, et al
.
TCF1 links GIPR signaling to the control of beta cell function and survival
.
Nat Med
2016
;
22
:
84
90
[PubMed]
19.
Eberhard
D
,
Lammert
E
.
The pancreatic beta-cell in the islet and organ community
.
Curr Opin Genet Dev
2009
;
19
:
469
475
[PubMed]
20.
Brissova
M
,
Aamodt
K
,
Brahmachary
P
, et al
.
Islet microenvironment, modulated by vascular endothelial growth factor-A signaling, promotes β cell regeneration
.
Cell Metab
2014
;
19
:
498
511
[PubMed]
21.
Sasson
A
,
Rachi
E
,
Sakhneny
L
, et al
.
Islet pericytes are required for β-cell maturity
.
Diabetes
2016
;
65
:
3008
3014
[PubMed]
22.
Houtz
J
,
Borden
P
,
Ceasrine
A
,
Minichiello
L
,
Kuruvilla
R
.
Neurotrophin signaling is required for glucose-induced insulin secretion
.
Dev Cell
2016
;
39
:
329
345
[PubMed]
23.
Richards
OC
,
Raines
SM
,
Attie
AD
.
The role of blood vessels, endothelial cells, and vascular pericytes in insulin secretion and peripheral insulin action
.
Endocr Rev
2010
;
31
:
343
363
[PubMed]
24.
van Es
JH
,
Haegebarth
A
,
Kujala
P
, et al
.
A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal
.
Mol Cell Biol
2012
;
32
:
1918
1927
[PubMed]
25.
Verzi
MP
,
Stanfel
MN
,
Moses
KA
, et al
.
Role of the homeodomain transcription factor Bapx1 in mouse distal stomach development
.
Gastroenterology
2009
;
136
:
1701
1710
[PubMed]
26.
Epshtein
A
,
Sakhneny
L
,
Landsman
L.
Isolating and analyzing cells of the pancreas mesenchyme by flow cytometry
.
J Vis Exp
2017
;
119
:
e55344
.
27.
Tribioli
C
,
Lufkin
T
.
Molecular cloning, chromosomal mapping and developmental expression of BAPX1, a novel human homeobox-containing gene homologous to Drosophila bagpipe
.
Gene
1997
;
203
:
225
233
[PubMed]
28.
Raines
SM
,
Richards
OC
,
Schneider
LR
, et al
.
Loss of PDGF-B activity increases hepatic vascular permeability and enhances insulin sensitivity
.
Am J Physiol Endocrinol Metab
2011
;
301
:
E517
E526
[PubMed]
29.
Armulik
A
,
Genové
G
,
Betsholtz
C
.
Pericytes: developmental, physiological, and pathological perspectives, problems, and promises
.
Dev Cell
2011
;
21
:
193
215
[PubMed]
30.
Segerstolpe
Å
,
Palasantza
A
,
Eliasson
P
, et al
.
Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes
.
Cell Metab
2016
;
24
:
593
607
[PubMed]
31.
Xin
Y
,
Kim
J
,
Okamoto
H
, et al
.
RNA equencing of single human islet cells reveals type 2 diabetes genes
.
Cell Metab
2016
;
24
:
608
615
[PubMed]
32.
Columbus
J
,
Chiang
Y
,
Shao
W
, et al
.
Insulin treatment and high-fat diet feeding reduces the expression of three Tcf genes in rodent pancreas
.
J Endocrinol
2010
;
207
:
77
86
[PubMed]
33.
Pujadas
G
,
Cervantes
S
,
Tutusaus
A
, et al
.
Wnt9a deficiency discloses a repressive role of Tcf7l2 on endocrine differentiation in the embryonic pancreas
.
Sci Rep
2016
;
6
:
19223
[PubMed]
34.
Shah
M
,
Varghese
RT
,
Miles
JM
, et al
.
TCF7L2 genotype and α-cell function in humans without diabetes
.
Diabetes
2016
;
65
:
371
380
[PubMed]
35.
Macotela
Y
,
Boucher
J
,
Tran
TT
,
Kahn
CR
.
Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism
.
Diabetes
2009
;
58
:
803
812
[PubMed]
36.
van de Wetering
M
,
Sancho
E
,
Verweij
C
, et al
.
The beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells
.
Cell
2002
;
111
:
241
250
[PubMed]
37.
Mestdagt
M
,
Polette
M
,
Buttice
G
, et al
.
Transactivation of MCP-1/CCL2 by beta-catenin/TCF-4 in human breast cancer cells
.
Int J Cancer
2006
;
118
:
35
42
[PubMed]
38.
Fujita
M
,
Furukawa
Y
,
Nagasawa
Y
,
Ogawa
M
,
Nakamura
Y
.
Down-regulation of monocyte chemotactic protein-3 by activated β-catenin
.
Cancer Res
2000
;
60
:
6683
6687
[PubMed]
39.
Ellingsgaard
H
,
Hauselmann
I
,
Schuler
B
, et al
.
Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells
.
Nat Med
2011
;
17
:
1481
1489
[PubMed]
40.
Goulley
J
,
Dahl
U
,
Baeza
N
,
Mishina
Y
,
Edlund
H
.
BMP4-BMPR1A signaling in beta cells is required for and augments glucose-stimulated insulin secretion
.
Cell Metab
2007
;
5
:
207
219
[PubMed]
41.
Paula
GS
,
Souza
LL
,
Bressane
NO
, et al
.
Mice with deletion of neuromedin B receptor exhibit decreased oral glucose-stimulated insulin release
.
Horm Metab Res
2016
;
48
:
854
861
[PubMed]
42.
Whissell
G
,
Montagni
E
,
Martinelli
P
, et al
.
The transcription factor GATA6 enables self-renewal of colon adenoma stem cells by repressing BMP gene expression
.
Nat Cell Biol
2014
;
16
:
695
707
[PubMed]
43.
Christensen
GL
,
Jacobsen
MLB
,
Wendt
A
, et al
.
Bone morphogenetic protein 4 inhibits insulin secretion from rodent beta cells through regulation of calbindin1 expression and reduced voltage-dependent calcium currents
.
Diabetologia
2015
;
58
:
1282
1290
[PubMed]
44.
Brown
ML
,
Kimura
F
,
Bonomi
LM
,
Ungerleider
NA
,
Schneyer
AL
.
Differential synthesis and action of TGFβ superfamily ligands in mouse and rat islets
.
Islets
2011
;
3
:
367
375
[PubMed]
45.
Clee
SM
,
Yandell
BS
,
Schueler
KM
, et al
.
Positional cloning of Sorcs1, a type 2 diabetes quantitative trait locus
.
Nat Genet
2006
;
38
:
688
693
[PubMed]
46.
Wicksteed
B
,
Alarcon
C
,
Briaud
I
,
Lingohr
MK
,
Rhodes
CJ
.
Glucose-induced translational control of proinsulin biosynthesis is proportional to preproinsulin mRNA levels in islet beta-cells but not regulated via a positive feedback of secreted insulin
.
J Biol Chem
2003
;
278
:
42080
42090
[PubMed]
47.
Nobrega
MA
.
TCF7L2 and glucose metabolism: time to look beyond the pancreas
.
Diabetes
2013
;
62
:
706
708
[PubMed]
48.
McCarthy
MI
,
Rorsman
P
,
Gloyn
AL
.
TCF7L2 and diabetes: a tale of two tissues, and of two species
.
Cell Metab
2013
;
17
:
157
159
[PubMed]
49.
Shao
W
,
Wang
D
,
Chiang
Y-T
, et al
.
The Wnt signaling pathway effector TCF7L2 controls gut and brain proglucagon gene expression and glucose homeostasis
.
Diabetes
2013
;
62
:
789
800
[PubMed]
50.
da Silva Xavier
G
,
Mondragon
A
,
Mourougavelou
V
, et al
.
Pancreatic alpha cell-selective deletion of Tcf7l2 impairs glucagon secretion and counter-regulatory responses to hypoglycaemia in mice
.
Diabetologia
2017
;
60
:
1043
1050
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data