<<

Page 1 of 47 Diabetes

Pancreatic pericytes support beta-cell function in a Tcf7l2-

dependent manner

Lina Sakhneny1, Eleonor Rachi1, Alona Epshtein1, Helen C. Guez1, Shane WaldAltman2, Michal Lisnyansky1, Laura KhalifaMalka1, Adina Hazan3, Daria Baer1, Avi Priel3, Miguel Weil2 and Limor Landsman1*

1Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel 2Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel 3Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel

Running title: Pericytic Tcf7l2 in betacell function

* Address for Correspondence: Limor Landsman, PhD Department of Cell and Developmental Biology Sackler Faculty of Medicine Tel Aviv University Ramat Aviv, Tel Aviv 69978 Israel (T): 9723640 6149 (F): 9723640 7432 Email: [email protected]

Abstract: 195 words Total word count: 3996 words References: 50 Tables: 0 Figures: 6

Conflict of Interest: The authors have declared that no conflict of interest exists.

1

Diabetes Publish Ahead of Print, published online December 15, 2017 Diabetes Page 2 of 47

Abstract Polymorphism in TCF7L2, a component of the canonical , has a strong association with βcell dysfunction and through a yet to be defined mechanism. βCells 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 βcell function and glucosestimulated secretion. Inactivation of pericytic Tcf7l2 was associated with impaired expression of required for βcell function and maturity in isolated islets. In addition, we identified Tcf7l2dependent pericytic expression of secreted factors shown to promote βcell function, including BMP4. Finally, we show that exogenous BMP4 is sufficient to rescue the impaired glucosestimulated insulin secretion of transgenic mice, pointing to a potential mechanism through which pericytic Tcf7l2 activity impacts βcells. To conclude, we suggest that pancreatic pericytes produce secreted factors, including BMP4, in a Tcf7l2 dependent manner to support βcell function. Our findings thus propose a potential cellular mechanism through which abnormal TCF7L2 activity predisposes individuals to diabetes, and implicate abnormalities in the islet microenvironment in this disease.

2 Page 3 of 47 Diabetes

Introduction 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 the TCF7L2 (TCF4) is associated with increased risk to diabetes (3). This encodes a

member of TCF/LEF (Tcell factor/lymphoid enhancer factor) factors family,

which functions downstream of the canonical Wnt signaling pathway by recruiting βcatenin

to target genes (4). Diabetesassociated alleles of TCF7L2 (such as the Tallele of the

singlenucleotide polymorphism (SNP) 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 rs7903146 variant was predicted to result in an

inactive lacking its DNAbinding 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 βcell dysfunction has produced conflicting

results. As opposed to humans, hepatic phenotypes dominate the abnormal glucose levels

observed upon bodywide deregulation of Tcf7l2 expression in mice (1012). βCell specific

inference with Tcf7l2 activity using mouse genetic tools yielded discrepant results, with some

studies showing reduced βcell mass and glucose intolerance, while others showing normal

glucose response (1217). This contradiction could partially stem from the use of different

approaches to interfere with Tcf7l2 activity, i.e., knockdown of the endogenous gene

(12,14,15) versus overexpressing a dominantnegative (DN) form (13,16). Recently, Tcf7

(Tcf1), a member of the TCF/LEF family with high homology to Tcf7l2, was shown to play a

central role in maintaining βcell mass (18), raising the possibility that overexpressing an

DN Tcf7l2 interferes with the activity of other TCF/LEF in βcells. Interestingly, while

βcell selective deletion of Tcf7l2 resulted in their reduced mass (15), selective deletion of

this DNAbinding domain affected neither βcell function nor mass (12).

3 Diabetes Page 4 of 47

Accordingly, nonautonomous roles of Tcf7l2 in regulating βcell function were suggested

(17).

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

Profiling 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 βcell function. To test our hypothesis, we selectively expressed an inactive form of this transcription factor in these cells by combining two transgenic mouse lines: Tcf7l2flox, which allows Cremediated deletion of this transcription factor DNAbinding domain (24), and Nkx3.2Cre (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 βcell function and maturity upon inactivation of pericytic

Tcf7l2. Lastly, we linked Tcf7l2dependent pericytic expression of Bone Morphogenetic

Protein 4 (BMP4) to glucose regulation. To conclude, our results indicate that pericytic

Tcf7l2 activity is required for βcell function and glucose homeostasis. Our findings further point to the contribution of abnormal pericytes activity to diabetes progression.

4 Page 5 of 47 Diabetes

Research Design and Methods Mice

All experiments were performed according to protocols approved by the Committee on

Animal Research at Tel Aviv University. Nkx3.2Cre (Nkx3–2tm1(cre)Wez)(25) and Tcf7l2flox

(Tcf7l2tm2.1Cle)(24) mice were generous gifts from Warren Zimmer (Texas A&M University,

TX) and Hans Clevers (Hubrecht Institute, Utrecht, The Netherlands), respectively. R26YFP

(Gt(ROSA)26Sortm1(EYFP)Cos) mice were obtained from The Jackson Laboratory. For diet

induced obesity, mice were placed on a high fat diet (HFD; 60% fat [kCal]; Teklad) at 6

weeks of age. Mice were intraperitoneally injected with dextrose (Sigma), insulin (Lilly), or

mouse recombinant BMP (rBMP4; R&D) when indicated. For analysis of functional

vasculature, fluoresceinlabeled tomato lectin (Vector) was injected intravenously and

allowed to circulate for 5 minutes before the animal was euthanized.

Flow cytometry

Cell isolation were performed as described (26). Cells were stained with primary antibodies

(Supplementary Table 1) when indicated, and either collected using FACS Aria (BD) or

analyzed using Gallios flowcytometer (Beckman Coulter) and Kaluza software (Beckman

Coulter).

Hormone detection

Islets were isolated according to standard protocols (21). For insulin secretion, following their

overnight culture, isolated islets were incubated in RPMI medium supplemented with

glucose for 1 hour. 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 GLP1 (736) Active Elisa kit

(Millipore).

Immunofluorescence

Dissected tissues were fixed in paraformaldehyde (4%), followed by cryosectioning. Tissue

section were stained with primary antibodies (Supplementary Table 1) followed by

secondary fluorescent antibodies (AlexaFluor, Invitrogen). Following staining with antiTcf7l2

5 Diabetes Page 6 of 47

antibody, TSA Fluorescein System (PerkinElmer) was used. For TUNEL assay, In Situ Cell

Death Detection Kit (Roche) was used. Images were acquired using Keyence BZ9000

(Biorevo) and SP8 confocal (Leica) Microscopes.

Morphometric analysis

Analysis of islet vasculature was performed as described (21). For measurement of βcell mass, immunostained paraffinembedded tissue sections were counterstained with HCS

CellMask Stain (Invitrogen) to label the whole tissue sections. Sections were automatically imaged using InCell 2000 analyzer (GE) and analyzed by developer software (GE).

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 (http://www.ebi.ac.uk/arrayexpress/arrays/EMTAB5325). For qPCR 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 twotailed Student’s t test.

6 Page 7 of 47 Diabetes

Results

Pancreatic pericytes express the transcription factor Tcf7l2

We recently showed that pericytes support βcell function (21). Here, we set to elucidate the

molecular basis of pericyte activity. To this end, we employed the Nkx3.2Cre 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, Nkx3.2Cre line targets mural

cells, including islet pericytes and vSMCs (vascular smooth muscle cells), but no other

pancreatic cell types (including epithelial and endothelial cells) ((21) and Supplementary Fig.

1). Pericytes (identified by expression of NG2 [Neural/Glial antigen 2] and desmin) in the

exocrine pancreas were also targeted by this Cre, as apparent fluorescent labeling of these

cells in the pancreas of Nkx3.2Cre;R26YFP mice (Supplementary Fig. 1). Of note, hepatic

pericytes, which were shown to regulate insulin response (28), are not targeted by this

mouse line (Supplementary Fig. 1). Thus, our analysis indicated that Nkx3.2Cre line targets

mural cells in both the endocrine and exocrine pancreas.

Next, we characterized pancreatic mural cells by profiling their gene expression. To

this end, cells were sorted from pancreatic tissues of Nkx3.2Cre;R26YFP mice based on

their fluorescent labeling (Supplementary Fig. 1). Of note, vast majority of labeled cells

express PDGFRβ (PlateletDerived 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). (GO) 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 βcell dysfunction and diabetes, we focused our

analysis on this transcription factor.

7 Diabetes Page 8 of 47

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 5fold higher than in islets, and 22fold 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, E), but not in the nuclei of vSMCs (identified by expression of

αSMA [α Smooth Muscle Actin] 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 βcells 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 diabetesassociated T allele of rs7903146 variant was predicted to result in an inactive Tcf7l2 protein lacking its DNAbinding domain

(9). We therefore employed a previously described transgenic mouse line, Tcf7l2flox, allowing

Cremediated deletion of the endogenous Tcf7l2 DNAbinding 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 Nkx3.2Cre line. To verify recombination of the Tcf7l2 locus, we analyzed its transcript levels by employing primers providing detection of wildtype

Tcf7l2, but not its recombined form (12,24). To allow isolation of pancreatic mural cells by flowcytometry (as described in Supplementary Fig. 1), a R26YFP transgene was included to generate Nkx3.2Cre;R26YFP;Tcf7l2flox/+ and Nkx3.2Cre;R26YFP;Tcf7l2flox/flox mice, as well as Nkx3.2Cre;R26YFP control mice. As shown in Figure 2A, wildtype Tcf7l2 was nearly absent from pancreatic mural cells of homozygous mice, and was significantly reduced in cells of heterozygous mice as compared to control mice. Of note, the expression

8 Page 9 of 47 Diabetes

levels of wildtype Tcf7l2 in islets isolated from homozygous and heterozygous mice were

comparable to those of controls (Fig. 2B).

To analyze mice glucose response, we performed intraperitoneal glucose tolerance

tests (IPGTT) on three groups of mice: homozygous for the inactive Tcf7l2 allele (Nkx3.2

Cre;Tcf7l2flox/flox), heterozygous for this allele (Nkx3.2Cre;Tcf7l2flox/+), and nontransgenic

controls (Cre negative: Tcf7l2flox/+ or Tcf7l2flox/flox). Of note, mice expressing the Nkx3.2Cre

transgene by itself (i.e., do not carry the Tcf7l2flox transgene) displayed comparable glucose

response to Crenegative control mice (Supplementary Fig. 3). As shown in Figure 2C, our

analysis revealed that 13week old homozygous, but not heterozygous male mice, display

an impaired glucose response as compared to littermate controls (Fig. 2C). TCF7L2

rs7903146 Tallele was shown to have a modest effect on βcell function, which becomes

more evident when insulin action decreases (34). To test if metabolic stress aggravates

glucose intolerance of transgenic mice, we placed mice on 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, while 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 both

women and men (8). However, we did not observe differences in glucose response of

transgenic and control female mice (Supplementary Fig. 3). Female and male mice differ in

their glucose metabolism (35). Thus, sexdependent differences may underlie the distinct

phenotype observed in female and male Nkx3.2Cre;Tcf7l2flox/flox mice.

The Nkx3.2Cre 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 Nkx3.2Cre;Tcf7l2flox/flox and Nkx3.2Cre;Tcf7l2flox/+ mice that could

contribute to their glucose intolerance. The three analyzed mouse groups show comparable

body weight under both regular chow and HFD, indicating normal food uptake and digestion

9 Diabetes Page 10 of 47

(Supplementary Fig. 4). Next, we analyzed for GLP1 (Glucagonlike peptide1) production by analyzing gut expression of Pcsk1 and Gcg (encoding prohormone convertases 1/3 and proglucagon, respectively) and measuring serum GLP1 levels, and found them comparable in Nkx3.2Cre;Tcf7l2flox/flox and control mice (Supplementary Fig. 4). Finally, insulin sensitivity was comparable between Nkx3.2Cre;Tcf7l2flox/flox, Nkx3.2Cre;Tcf7l2flox/+, and control (Cre negative) male mice, on both 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 set to analyze whether Tcf7l2 inactivation in pancreatic pericytes interferes with islet vascularization. To this end, Nkx3.2Cre;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.

βββ-Cell dysfunction upon pericytic Tcf7l2 inactivation

We previously showed that islet pericytes support βcell function (21). We therefore set to determine if βcell dysfunction underlies the observed glucose intolerance upon loss of

Tcf7l2 activity in pancreatic pericytes. To test if the impaired glucose response is associated with insufficient GSIS, we measured glucosestimulated serum insulin levels and found them to be lower in Nkx3.2Cre;Tcf7l2flox/flox mice (Fig. 4A). Next, we analyze if βcell mass and/or function are affected in Tcf7l2deficient mice. Our analysis indicated normal islet morphology

10 Page 11 of 47 Diabetes

and comparable pancreatic and βcell mass in transgenic mice (Fig. 4BD). In agreement

with these findings, we observed neither βcell death nor expression of genes associated

with βcell stress in islets of transgenic mice (i.e., Chop, Atf4; Supplementary Fig. 5). To test

for potential changes in βcell function we analyzed GSIS of isolated islets, and found that

those of transgenic mice secrete less insulin in response to high glucose levels, as

compared to control islets (Fig. 4E). Thus, our analysis points to βcell dysfunction upon

inactivation of pericytic Tcf7l2.

Next, we measured insulin content in isolated islets and pancreatic tissues from

Nkx3.2Cre;Tcf7l2flox/flox and control animals, and found them to be significantly reduced in

homozygous animals (Fig. 4F and G). While 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). While we detected a

statistically significant reduction in expression levels of Pcsk1, required for posttranslational

processing of insulin, in Nkx3.2Cre;Tcf7l2flox/flox islets (Fig. 4H), the ratio between proinsulin

and insulin levels were comparable in transgenic and control islets (Fig. 4I). In addition, Gcg

levels were comparable in Nkx3.2Cre;Tcf7l2flox/flox and control islets (Fig. 4J). To conclude,

we observed reduced islet and pancreatic insulin content upon inactivation of pericytic

Tcf7l2.

Nkx3.2Cre;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 analyze if 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 Figure 4K, all three genes were expressed at lower levels in Nkx3.2

Cre;Tcf7l2flox/flox islets as compared to control. In contrast, expression of Glpr1, encoding the

GLP1 receptor, was comparable in transgenic and control islets (Fig. 4L). βCell function

and gene expression have been shown to depend on transcription factors, including MafA,

11 Diabetes Page 12 of 47

Pdx1 and NeuroD1 (2). We observed reduced transcripts level of all three in islets from

Nkx3.2Cre;Tcf7l2flox/flox mice as compared to control (Fig. 4M). Thus, inactivation of pericytic

Tcfl72 is associated with impaired expression of βcell genes.

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

Tcf7l2 regulates pericytic expression of ligands shown to support βββ-cell function

Our findings point to a Tcf7l2dependent activity of pancreatic pericytes in supporting βcell function and gene expression. We therefore set to analyze potential changes in pancreatic pericytes upon inactivation of Tcf7l2. Our immunofluorescence analysis revealed that pericytes are localized in proximity to endothelial cells in both control and transgenic islets

(Fig. 5A). However, morphometric analysis revealed that pericyte coverage was mildly reduced in islets of Nkx3.2Cre;Tcf7l2flox/flox mice as compared to control (by 7%; Fig. 5B).

Next, we analyzed whether loss of Tcf7l2 activity affect their gene expression. To this end, pancreatic mural cells were purified by flowcytometry (as described in Supplementary

Fig. 1) from Nkx3.2Cre;R26YFP;Tcf7l2flox/flox and control Nkx3.2Cre;R26YFP mice, and islets were isolated from nontransgenic mice, and their RNA was extracted and sequenced.

We hypothesized that pericytes secrete factors to support βcells under normal conditions, and the expression of some of these factors is Tcf7l2dependent. 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 4). The expression levels of seven of these genes were significantly lower in Tcf7l2transgenic pancreatic mural cells (Fig. 5C and Supplementary Table 5): Bmp4, Ccl2, Ccl7, C7, Il6, Fam150b and Nmb.

Tcf7l2 was previously shown to directly regulate the expression of first three genes (3638).

Importantly, βcells were shown to express the receptors for BMP4, Neuromedin B, and IL6

12 Page 13 of 47 Diabetes

(encoded by Bmp4, Nmb, and Il6, respectively), and these three factors were implicated in

βcell function (3941).

To conclude, our analysis showed that lack of Tcf7l2 activity in pancreatic pericytes

lead to mildly reduced islet pericyte density, and further resulted in impaired expression of

secreted ligands by these cells, some of which were implicated in βcell function.

Impaired glucose response in Tcfl72-deficient mice is rescued by exogenous

BMP4

βCells 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 mice

glucose response and promoted the mature βcell 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 Tcf7l2

dependent manner supports βcell function. To test our hypothesis, we first validated

pericytic Bmp4 expression by qPCR analysis, which revealed reduced transcript levels in

Tcf7l2deficient mural cells to a third of the level found in control cells (Fig. 6A).

To study the contribution of reduced BMP4 production by pericytes to the observed

phenotype in Nkx3.2Cre;Tcf7l2flox/flox mice, we intraperitoneally injected rBMP4 to

homozygous mice (40). As shown in Figure 6, glucose tolerance and GSIS of rBMP4treated

animals were significantly improved as compared to untreated transgenic animals.

Importantly, glucose response and insulin secretion of rBMP4treated Nkx3.2

Cre;Tcf7l2flox/flox mice were comparable to that of nontransgenic control animals (Fig. 6B and

C).

To analyze if the improved GSIS upon rBMP4 treatment of Nkx3.2Cre;Tcf7l2flox/flox

mice is associated with improvement in their βcell 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 hours promoted the expression of Pdx1 and MafA,

13 Diabetes Page 14 of 47

encoding transcription factors associated with mature βcell 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 wildtype islets (Supplementary Fig. 6).

To conclude, our analysis showed that extrinsic BMP4 was sufficient to rescue glucose response, GSIS and mature βcell gene expression in mice deficient of pericytic

Tcf7l2. Thus, our findings suggest that pericytes support βcell function through Tcf7l2 depedent production of BMP4.

14 Page 15 of 47 Diabetes

Discussion Here, we show that pericytes support βcell 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 DNAbinding domain,

in pancreatic pericytes. Loss of pericytic Tcf7l2 activity interfered with mice glucose

regulation due to impaired βcell function (Fig. 2 and 4). Our analysis further indicated

Tcf7l2dependent pericytic expression of secreted factors implicated in βcell function,

including BMP4 (Fig. 5). Finally, we showed that treatment of Tcf7l2deficient 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

Tcf7l2dependent manner to support βcell function and glucose response. Our findings

propose that impaired pericytic activity perturbs βcell function, thus potentially contributing

to disrupted glucose regulation and diabetes progression.

Abnormalities in islet pericytes were implicated in obesity and diabetes (23). Others

and we showed that pericytes directly support βcell function and glucose regulation (21,22).

Interestingly, SORCS1, a type 2 diabetesassociated 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 diabetesassociated 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 βcell dysfunction and loss of

glucose regulation.

Our analysis revealed that pericytes produce BMP4 in Tcf7l2dependent manner to

support glucose response (Fig. 5 and 6). Treatment of Nkx3.2Cre;Tcf7l2flox/flox mice with

rBMP4 was sufficient to rescue their glucose tolerance, strengthening the importance of this

factor in supporting proper βcell function. While the BMP4BMPR1A pathway was shown to

promote βcell function and gene expression in vivo (40), treating isolated islets with rBMP4

15 Diabetes Page 16 of 47

either did not affect βcells or impaired their survival and function (43,44), pointing to the requirement of regulated BMP4 levels for proper βcell function. Loss of BMPR1A function in

βcells resulted in a more severe glucose intolerance phenotype than the one observed by us upon loss of pericytic Tcf7l2 (compare Reference (40) and Fig. 2). These differences might reflect the presence of residual pericytic BMP4 in transgenic mice and/or additional pancreatic BMP4 sources, as 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 / proinsulin ratio, this observation indicates an impaired insulin biosynthesis in

Nkx3.2Cre;Tcf7l2flox/flox mice. While Ins1 and Ins2 transcript levels were on average lower in transgenic islets (by 39 and 33%, respectively; Fig. 4), this reduction was not statistical 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 βcell function in a cellautonomous manner (14,15).

However, whether impaired Tcf7l2 activity in βcells is sufficient to drive diabetes progression remains controversial (12,17,47,48). Accordingly, Tcf7l2 was suggested to regulate βcell 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/βcell axis.

Polymorphism in TCF7L2 has a strong correlation to type 2 diabetes in humans

(3,5,6). In particular, the T allele of rs7903146 was shown by multiple studies to increase the risk of developing this disease (by 30%)(5). Individuals harboring one or two copies of this allele display reduced basal and glucosestimulated insulin secretion, but maintain hepatic function (6,7,34). As shown in this study, while mice lacking Tcf7l2 activity in their pancreatic pericytes were glucose intolerant, they did not develop diabetes, even when

16 Page 17 of 47 Diabetes

subjected to a HFD (Fig. 2). These discrepancies could reflect the evident difference in

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, while impaired TCF7L2 activity increases the risk of

developing diabetes, additional genetic and environmental factors contribute to disease

progression (1). Nevertheless, impaired βcell function upon loss of pericytic TCF7L2 activity

proposes a cellular mechanism through which mutations in this transcription factor

predispose individuals to diabetes.

17 Diabetes Page 18 of 47

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

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

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

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, acquired and analyzed data, S.W.A. analyzed data, M.L., L.M.K., A.H., and D.B. conducted experiments and acquired data, A.P. and

M.W. provided reagents, and 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 This study was presented at the 2nd Joint Meeting of the European Association for the

Study of Diabetes (EASD) Islet Study Group and Workshop, Dresden, Germany,

711 May 2017 and the American Diabetes Association (ADA) 77th Scientific Session, San

Diego, CA, 913 June 2017.

18 Page 19 of 47 Diabetes

References

1. McCarthy MI. Genomics, type 2 diabetes, and obesity. N Engl J Med. 2010 Dec 9;363(24):2339–50.

2. Doria A, Patti ME, Kahn CR. The emerging genetic architecture of type 2 diabetes. Cell Metabolism. 2008 Sep;8(3):186–200.

3. Grant SFA, Thorleifsson G, Reynisdottir I, Benediktsson R, Manolescu A, Sainz J, et al. Variant of transcription factor 7like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet. 2006 Mar;38(3):320–3.

4. Nusse R, Clevers H. Wnt/βCatenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell. 2017 Jun 1;169(6):985–99.

5. Helgason A, Pálsson S, Thorleifsson G, Grant SFA, Emilsson V, Gunnarsdottir S, et al. Refining the impact of TCF7L2 gene variants on type 2 diabetes and adaptive evolution. Nat Genet. 2007 Feb;39(2):218–25.

6. Lyssenko V, Lupi R, Marchetti P, Del Guerra S, OrhoMelander M, Almgren P, et al. Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. Journal of Clinical Investigation. 2007 Aug;117(8):2155–63.

7. Varghese RT, Viegas I, Barosa C, Marques C, Shah M, Rizza RA, et al. Diabetes Associated Variation in TCF7L2 Is Not Associated With Hepatic or Extrahepatic Insulin Resistance. Diabetes. 2016 Apr;65(4):887–92.

8. Zhang C, Qi L, Hunter DJ, Meigs JB, Manson JE, van Dam RM, et al. Variant of Transcription Factor 7Like 2 (TCF7L2) Gene and the Risk of Type 2 Diabetes in Large Cohorts of U.S. Women and Men. Diabetes. 2006 Sep 1;55(9):2645–8.

9. Locke JM, da Silva Xavier G, Rutter GA, Harries LW. An alternative polyadenylation signal in TCF7L2 generates isoforms that inhibit T cell factor/lymphoidenhancer factor (TCF/LEF)dependent target genes. Diabetologia. 2011;54(12):3078–82.

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 Sep;21(9):1417–25.

11. Oh KJ, Park J, Kim SS, Oh H, Choi CS, Koo SH. TCF7L2 modulates glucose homeostasis by regulating CREB and FoxO1dependent transcriptional pathway in the liver. PLoS Genet. 2012 Sep;8(9):e1002986.

12. Boj SF, Van Es JH, Huch M, Li VSW, José A, Hatzis P, et al. Diabetes Risk Gene and Wnt Effector Tcf7l2/TCF4 Controls Hepatic Response to Perinatal and Adult Metabolic Demand. Cell. 2012 Dec 21;151(7):1595–607.

13. Takamoto I, Kubota N, Nakaya K, Kumagai K, Hashimoto S, Kubota T, et al. TCF7L2 in mouse pancreatic beta cells plays a crucial role in glucose homeostasis by regulating beta cell mass. Diabetologia. 2014 Mar;57(3):542–53.

14. da Silva Xavier G, Mondragon A, Sun G, Chen L, McGinty JA, French PM, et al. Abnormal glucose tolerance and insulin secretion in pancreasspecific Tcf7l2null mice. Diabetologia. 2012 Oct;55(10):2667–76.

19 Diabetes Page 20 of 47

15. Mitchell RK, Mondragon A, Chen L, McGinty JA, French PM, Ferrer J, et al. Selective disruption of Tcf7l2 in the pancreatic β cell impairs secretory function and lowers β cell mass. Hum Mol Genet. 2015 Mar 1;24(5):1390–9.

16. Shao W, Xiong X, Ip W, Xu F, Song Z, Zeng K, et al. The expression of dominant negative TCF7L2 in pancreatic beta cells during the embryonic stage causes impaired glucose homeostasis. Molecular Metabolism. 2015 Apr 1;4(4):344–52.

17. Bailey KA, Savic D, Zielinski M, Park SY, Wang LJ, Witkowski P, et al. Evidence of nonpancreatic beta celldependent roles of Tcf7l2 in the regulation of glucose metabolism in mice. Hum Mol Genet. 2015 Mar 15;24(6):1646–54.

18. Campbell JE, Ussher JR, Mulvihill EE, Kolic J. TCF1 links GIPR signaling to the control of beta cell function and survival. Nat Med. 2015;22(1):84–90.

19. Eberhard D, Lammert E. The pancreatic betacell in the islet and organ community. Curr Opin Genet Dev. 2009 Oct;19(5):469–75.

20. Brissova M, Aamodt K, Brahmachary P, Prasad N, Hong JY, Dai C, et al. Islet microenvironment, modulated by vascular endothelial growth factorA signaling, promotes β cell regeneration. Cell Metabolism. 2014 Mar 4;19(3):498–511.

21. Sasson A, Rachi E, Sakhneny L, Baer D, Lisnyansky M, Epshtein A, et al. Islet Pericytes Are Required for βCell Maturity. Diabetes. 2016 Oct;65(10):3008–14.

22. Houtz J, Borden P, Ceasrine A, Minichiello L, Kuruvilla R. Neurotrophin Signaling Is Required for GlucoseInduced Insulin Secretion. Dev Cell. 2016 Nov 7;39(3):329–45.

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 Jun;31(3):343–63.

24. Van Es JH, Haegebarth A, Kujala P, Itzkovitz S, Koo BK, Boj SF, et al. A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic selfrenewal. Mol Cell Biol. 2012 May;32(10):1918–27.

25. Verzi MP, Stanfel MN, Moses KA, Kim BM, Zhang Y, Schwartz RJ, et al. Role of the homeodomain transcription factor Bapx1 in mouse distal stomach development. Gastroenterology. 2009 May;136(5):1701–10.

26. Epshtein A, Sakhneny L, Landsman L. Isolating and Analyzing Cells of the Pancreas Mesenchyme by Flow Cytometry. JoVE. 2017 Jan 28;(119):e55344–4.

27. Tribioli C, Lufkin T. Molecular cloning, chromosomal mapping and developmental expression of BAPX1, a novel human containing gene homologous to Drosophila bagpipe. Gene. 1997 Dec 12;203(2):225–33.

28. Raines SM, Richards OC, Schneider LR, Schueler KL, Rabaglia ME, Oler AT, et al. Loss of PDGFB activity increases hepatic vascular permeability and enhances insulin sensitivity. Am J Physiol Endocrinol Metab. 2011 Sep;301(3):E517–26.

29. Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011 Aug 16;21(2):193–215.

20 Page 21 of 47 Diabetes

30. Segerstolpe Å, Palasantza A, Eliasson P, Andersson EM, Andréasson AC, Sun X, et al. SingleCell Transcriptome Profiling of Human Pancreatic Islets in Health and Type 2 Diabetes. Cell Metabolism. 2016 Oct 11;24(4):593–607.

31. Xin Y, Kim J, Okamoto H, Ni M, Wei Y, Adler C, et al. RNA Sequencing of Single Human Islet Cells Reveals Type 2 Diabetes Genes. Cell Metabolism. 2016 Oct 11;24(4):608–15.

32. Columbus J, Chiang Y, Shao W, Zhang N, Wang D, Gaisano HY, et al. Insulin treatment and highfat diet feeding reduces the expression of three Tcf genes in rodent pancreas. Journal of Endocrinology. 2010 Oct;207(1):77–86.

33. Pujadas G, Cervantes S, Tutusaus A, Ejarque M, Sanchez L, García A, et al. Wnt9a deficiency discloses a repressive role of Tcf7l2 on endocrine differentiation in the embryonic pancreas. Sci Rep. 2016;6:19223.

34. Shah M, Varghese RT, Miles JM, Piccinini F, Dalla Man C, Cobelli C, et al. TCF7L2 Genotype and αCell Function in Humans Without Diabetes. Diabetes. 2016 Feb;65(2):371–80.

35. Macotela Y, Boucher J, Tran TT, Kahn CR. Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes. 2009 Apr;58(4):803–12.

36. van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A, et al. The betacatenin/TCF4 complex imposes a crypt progenitor phenotype on cells. Cell. 2002 Oct 18;111(2):241–50.

37. Mestdagt M, Polette M, Buttice G, Noël A, Ueda A, Foidart JM, et al. Transactivation of MCP1/CCL2 by betacatenin/TCF4 in human breast cancer cells. Int J Cancer. 2006 Jan 1;118(1):35–42.

38. Fujita M, Furukawa Y, Nagasawa Y, Ogawa M, Nakamura Y. DownRegulation of Monocyte Chemotactic Protein3 by Activated βCatenin. Cancer Res. 2000 Dec 1;60(23):6683–7.

39. Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, Meier DT, et al. Interleukin6 enhances insulin secretion by increasing glucagonlike peptide1 secretion from L cells and alpha cells. Nat Med. 2011 Oct 30;17(11):1481–9.

40. Goulley J, Dahl U, Baeza N, Mishina Y, Edlund H. BMP4BMPR1A signaling in beta cells is required for and augments glucosestimulated insulin secretion. Cell Metabolism. 2007 Mar;5(3):207–19.

41. Paula GSM, Souza LL, Bressane NOS, Maravalhas R, Wilieman M, BentoBernardes T, et al. Mice with Deletion of Neuromedin B Receptor Exhibit Decreased Oral GlucoseStimulated Insulin Release. Horm Metab Res. 2016 Dec;48(12):854–61.

42. Whissell G, Montagni E, Martinelli P, HernandoMomblona X, Sevillano M, Jung P, et al. The transcription factor GATA6 enables selfrenewal of colon adenoma stem cells by repressing BMP gene expression. Nat Cell Biol. 2014 Jun 22;16(7):695–707.

43. Christensen GL, Jacobsen MLB, Wendt A, Mollet IG, Friberg J, Frederiksen KS, et al. Bone morphogenetic protein 4 inhibits insulin secretion from rodent beta cells through regulation of calbindin1 expression and reduced voltagedependent calcium currents. Diabetologia. 2015 Jun;58(6):1282–90.

21 Diabetes Page 22 of 47

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 Nov 1;3(6):367–75.

45. Clee SM, Yandell BS, Schueler KM, Rabaglia ME, Richards OC, Raines SM, et al. Positional cloning of Sorcs1, a type 2 diabetes quantitative trait locus. Nat Genet. 2006 Jun;38(6):688–93.

46. Wicksteed B, Alarcon C, Briaud I, Lingohr MK, Rhodes CJ. Glucoseinduced translational control of proinsulin biosynthesis is proportional to preproinsulin mRNA levels in islet betacells but not regulated via a positive feedback of secreted insulin. J Biol Chem. 2003 Oct 24;278(43):42080–90.

47. Nobrega MA. TCF7L2 and Glucose Metabolism: Time to Look Beyond the Pancreas. Diabetes. 2013 Mar 1;62(3):706–8.

48. McCarthy MI, Rorsman P, Gloyn AL. TCF7L2 and diabetes: a tale of two tissues, and of two species. Cell Metabolism. 2013 Feb 5;17(2):157–9.

49. Shao W, Wang D, Chiang YT, Ip W, Zhu L, Xu F, et al. The Wnt Signaling Pathway Effector TCF7L2 Controls Gut and Brain Proglucagon Gene Expression and Glucose Homeostasis. Diabetes. 2013 Mar 1;62(3):789–800.

50. da Silva Xavier G, Mondragon A, Mourougavelou V, CrucianiGuglielmacci C, Denom J, Herrera PL, et al. Pancreatic alpha cellselective deletion of Tcf7l2 impairs glucagon secretion and counterregulatory responses to hypoglycaemia in mice. Diabetologia. 2017 Mar 25;14(Pt 3):551.

22 Page 23 of 47 Diabetes

Figure legends Figure 1: Expression of Tcf7l2 gene and protein by pancreatic pericytes

A) Bar diagram shows number of genes (xaxis) clustered to Gene Ontology (GO) terms (y

axis) enriched in RNA sequencing analysis of purified pancreatic mural cells, FACS sorted

from Nkx3.2Cre;R26EYFP 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 per million aligned

fragments [FPKM]) of TCF/LEF family members in RNA sequencing analysis of pancreatic

mural cells, as described in A’.

C) Boxandwhisker plots showing relative levels of Tcf7l2 transcript by qPCR analysis. RNA

was extracted from bulk pancreatic tissue (left; ‘Pancreas’), isolated islets (middle; ‘Islets’;

average was set to ‘1’) and purified pancreatic mural cells (right) and gene expression level

was analyzed. N = 45. ***, p<0.005.

(DF) Immunofluorescence analysis of pancreatic tissue sections from adult wildtype

mouse. (D) Sections were stained for NG2 (red) to label mural cells and Tcf7l2 (green), and

were counterstained with DAPI (blue). Middle and right panels, higher magnification of the

area framed in the white box in left panel. Note the presence of nuclear Tcf7l2 in pericytes.

(E) Sections were stained for insulin (white), NG2 (red) and Tcf7l2 (green). Right panel,

higher magnification of the area framed in the white box in left panel. Note the presence of

nuclear Tcf7l2 in isletassociated pericytes. (F) Sections were stained for αSMA (white) to

label vSMCs, PECAM1 (red) to label endothelial cells and Tcf7l2 (green). Note Tcf7l2 is

absent from nuclei of vSMCs. Shown are representative fields.

23 Diabetes Page 24 of 47

Figure 2: Male mice with a disrupted pericytic Tcf7l2 activity are glucose intolerant

A) Bar diagram showing relative levels of wildtype Tcf7l2 transcript in mural cells by qPCR analysis. RNA was extracted from purified pancreatic mural cells (as described in Fig. 2a) of

Nkx3.2Cre;R26EYFP;Tcf7l2flox/flox (red), Nkx3.2Cre;R26EYFP;Tcf7l2flox/+ (orange) and

Nkx3.2Cre;R26EYFP (gray; set to ‘1’) mice and gene expression was analyzed. N = 34.

***, p<0.005, as compared to Nkx3.2Cre;R26EYFP control mice.

B) Bar diagram showing relative levels of wildtype Tcf7l2 transcript in islets by qPCR analysis. RNA was extracted from isolated islets of Nkx3.2Cre;Tcf7l2flox/flox (red), Nkx3.2

Cre;Tcf7l2flox/+ (orange) and nontransgenic (Crenegative; ‘non tg’; gray; set to ‘1’) mice and gene expression was analyzed. N=4.

C,D) Intraperitoneal Glucose tolerance test (IPGTT) on Nkx3.2Cre;Tcf7l2flox/flox (red circles),

Nkx3.2Cre;Tcf7l2flox/+ (orange triangles) and nontransgenic littermate (Crenegative; ‘non tg’; gray squares) male mice at 13 weeks of age. Mice were fed either regular chow (C; N =

812) or highfat diet (HFD)(D; N = 69), when these fed HFD and reaching a body weight of

>32 grams at 13 weeks of age were considered obese and used for analysis. After overnight fasting, mice were intraperitoneally injected with dextrose (2 mg/g body weight) and tail vein blood glucose levels were measured at indicated time points. Left and middle, shown are mean (± SEM) blood glucose levels. Right, Boxandwhisker plot showing area under the curve (AUC) of glucose responses shown in left and middle panels. *, p<0.05, ***, p<0.005,

NS = not significant, as compared to nontransgenic control.

24 Page 25 of 47 Diabetes

Figure 3: Functional islet vasculature upon inactivation of pericytic Tcf7l2

Nkx3.2Cre;Tcf7l2flox/flox transgenic (red bars and right panels) and nontransgenic control

(Crenegative, ‘non tg’; gray bars and left panel) mice were analyzed.

A,B) Mice were intravenously injected with tomato lectin (1 mg/ml; red) to label function

vessels. Pancreas was harvested from treated mice, and tissue sections were stained for

insulin (blue). A) Images showing representative islets. Upper panels: shown are lectin

labeling and antiinsulin staining. Lower panels: shown is lectin labeling alone. White lines

demark the outer border of Insulin+ area, as shown in upper panels. B) Bar diagrams (mean

± SD) showing quantification of intraislet vascular density. The relative ratio of lectin+ and

insulin+ area per each islet was calculated. 30 islets per mouse, from sections at least 100

m apart, were analyzed. N = 3. NS = not significant, as compared to nontransgenic

control.

C) Boxandwhisker plot showing 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.

25 Diabetes Page 26 of 47

Figure 4: Impaired βββ-cell function and gene expression in Nkx3.2-Cre;Tcf7l2flox/flox mice

Nkx3.2Cre;Tcf7l2flox/flox transgenic (red) and nontransgenic control (Crenegative, ‘non tg’; gray) 13week old mice were analyzed.

A) Glucosestimulated serum insulin levels. After overnight fast, dextrose (2 mg/g body weight) was intraperitoneally injected, and tail vein blood was collected at indicated time points from transgenic (red circles) and control (gray squares) mice. Shown are mean (±

SEM) blood insulin levels. N = 45. B) Pancreatic tissues of transgenic (left) and non transgenic (right) mice were stained for insulin (green) and glucagon (red). Shown are representative islets.

C) Bar diagrams (mean ± SD) showing pancreatic weight. N = 3. D) Bar diagrams (mean ±

SD) showing estimated βcell mass. For each analyzed mouse, Insulin+ and pancreatic

(labeled by HCS CellMask Stain) areas were measured in sections at least 100 m 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, E’) Bar diagram (mean ± SD) shows impaired glucosestimulated insulin secretion by transgenic islets. Isolated islets from transgenic and control mice (N = 4) were incubated with either low

(1.67 mM) or high (16.7 mM) glucose, and 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 either normalized to islets insulin content (E’) or not normalized

(E).

F, G, I) Bar diagrams (mean ± SD) showing reduced insulin content in pancreatic tissues (F) and isolated islets (G) of transgenic mice, but comparable islet proinsulin/insulin ratio (I).

Insulin and proinsulin content of isolated islets were analyzed after their overnight culture.

Pancreatic insulin content was normalized to total pancreatic protein content. N = 4.

H, J-M) Boxandwhisker plots showing impaired gene expression in Nkx3.2Cre;Tcf7l2flox/flox islets. RNA was extracted from islets isolated from transgenic and control mice (N = 45) and expression of indicated genes was analyzed by qPCR, when average levels in non

26 Page 27 of 47 Diabetes

transgenic islets were set to ‘1’. *, p<0.05; **, p<0.01; ***, p<0.005; NS = nonsignificant, as

compared to nontransgenic control.

27 Diabetes Page 28 of 47

Figure 5: Reduced expression of pericytic secreted ligands upon Tcf7l2 inactivation.

A, B) Islet pericyte density in mildly reduced upon loss of Tcf7l2 activity. Pancreatic tissues of Nkx3.2Cre;Tcf7l2flox/flox transgenic (red bars and right panel) and nontransgenic control mice (Crenegative, non tg; gray bars; left panel) were stained for NG2 (red) to label pericytes, PECAM1 (green) to label endothelial cells, and insulin to label βcells. A) Images of representative islets showing normal distribution of pericytes in transgenic islets. White lines demark the outer border of Insulin+ area. B) Bar diagrams (mean ± SD) showing 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. 50 islets per mouse, from sections at least 100 m apart, were analyzed. N = 3. *, p < 0.05, as compared to nontransgenic control.

C, D) Analysis of Tcf7l2dependent expression of secreted factors by pancreatic mural cell.

Mural cells were isolated from pancreatic tissues of 10week old Nkx3.2Cre;R26REYFP

(non tg; middle panel) and Nkx3.2Cre;R26REYFP;Tcf7l2flox/flox (‘Tcf7l2f/f’; right panel), as described in Supplementary Fig. 1. RNA was extracted from pancreatic mural cells and islets isolated from agematched nontransgenic mice (left panels) and subjected to deep sequencing. Heat map shows levels of indicated transcripts in FPKM.

*, p<0.05; ***, p<0.005; NS = nonsignificant, as compared to nontransgenic control.

28 Page 29 of 47 Diabetes

Figure 6: Exogenous BMP4 rescues glucose intolerance of Tcf7l2-deficient mice

A) Analysis of Tcf7l2dependent expression of Bmp4 by pancreatic mural cells. Nkx3.2/YFP+

mural cells were isolated by flowcytometry from pancreatic tissues of 10week old Nkx3.2

Cre;R26REYFP (control; gray) and Nkx3.2Cre;R26REYFP;Tcf7l2flox/flox (red). 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, as compared to control.

B, C) IPGTT and GSIS after treatment of homozygous mice with exogenous BMP4. Nkx3.2

Cre;Tcf7l2flox/flox mice were intraperitoneally injected twice daily with either PBS or

recombinant BMP4 (rBMP4; 20 ng/g body weight) for three consecutive days, and a final

treatment 30 min before analysis. rBMP4treated Nkx3.2Cre;Tcf7l2flox/flox (empty red circles,

broken red lines and striped boxes, N = 34), PBStreated Nkx3.2Cre;Tcf7l2flox/flox (full red

circles and boxes, and continuous red lines, N = 56), and nontransgenic littermate (non tg;

gray squares, lines and boxes, N = 58) male mice at 13 weeks of age were analyzed. After

overnight fasting, mice were intraperitoneally injected with dextrose (2 mg/g body weight)

and tail vein blood glucose and insulin levels were measured at indicated time points. B)

IPGTT. Left, shown are mean (± SEM) blood glucose levels. Right, Boxandwhisker plot

showing area under the curve (AUC) of glucose responses, as shown in left panel. *, p<0.05,

**, p<0.01, ***, p<0.005, NS = not significant; Left panel, as compared to untreated

transgenic controls; Right panel, as indicated by horizontal bars. C) GSIS. Insulin levels

were measured by ELISA. *, p<0.05, ***, p<0.005, as compared to untreated transgenic

controls.

D) Boxandwhisker plot showing expression of βcell genes upon treatment of transgenic

islets with rBMP4 ex vivo. Islets isolated from untreated Nkx3.2Cre;Tcf7l2flox/flox mice were

cultured in media supplemented with 6 ng/ml rBMP4 (‘+rBMP4’; striped red boxes) or

unsupplemented media (red boxes) for 24 hours. RNA was extracted and expression levels

of indicated genes were analyzed by qPCR, when average levels in untreated islets were set

29 Diabetes Page 30 of 47

to ‘1’. *, p<0.05, ***, p<0.005, as compared to untreated control. N=6. Representative of three independent experiments.

30 Page 31 of 47 Diabetes

Figure 1: Expression of Tcf7l2 gene and protein by pancreatic pericytes

180x87mm (300 x 300 DPI)

Diabetes Page 32 of 47

Figure 2: Male mice with a disrupted pericytic Tcf7l2 activity are glucose intolerant

177x123mm (300 x 300 DPI)

Page 33 of 47 Diabetes

Figure 3: Functional islet vasculature upon inactivation of pericytic Tcf7l2

89x103mm (300 x 300 DPI)

Diabetes Page 34 of 47

Figure 4: Impaired βcell function and gene expression in Nkx3.2Cre;Tcf7l2flox/flox mice

174x154mm (300 x 300 DPI)

Page 35 of 47 Diabetes

Figure 5: Reduced expression of pericytic secreted ligands upon Tcf7l2 inactivation

74x122mm (300 x 300 DPI)

Diabetes Page 36 of 47

Figure 6: Exogenous BMP4 rescues glucose intolerance of Tcf7l2-deficient mice

179x83mm (300 x 300 DPI)

Page 37 of 47 Diabetes

Supplemental Information

Pancreatic pericytes support beta-cell function in a Tcf7l2 dependent manner

Lina Sakhneny, Eleonor Rachi, Alona Epshtein, Helen C. Guez, Shane Wald-Altman, Michal Lisnyansky, Laura Khalifa-Malka, Adina Hazan, Daria Baer, Avi Priel, Miguel Weil and Limor Landsman

Diabetes Page 38 of 47

Sakhneny et al.

Supplementary Figure 1: Nkx3.2-Cre mouse line drives gene expression in pancreatic mural cells. (A) Tissue sections from adult Nkx3.2-Cre;R26-YFP mice were stained for YFP (green), NG2 (red) and insulin (white) and were counterstained with DAPI (blue). Right panels, higher magnification of the areas framed in white boxes in left panels, when ‘*’, ‘#’ and ‘@’ mark the different boxes, corresponding to islets, exocrine pancreas and pancreatic large blood vessels, respectively. Note the presence of YFP+ mural cells in all three pancreatic areas. ‘i’ = islet; ‘v’=blood vessel. Shown is a representative field. B, C) Tissue sections from adult Nkx3.2-Cre;R26-YFP mice were stained for YFP (green) and NG2 (red; B) or desmin (red; C). Right panel, green channel of the field shown in left panel. Note YFP+ cells express the mural cell markers NG2 and desmin. Shown is a representative field. D) Hepatic tissues from adult Nkx3.2-Cre;R26-YFP mice were analyzed. Tissue sections were stained for YFP (green) and NG2 (red) and counterstained with DAPI. Right panel, higher

2 Page 39 of 47 Diabetes

Sakhneny et al.

magnification of the area framed in a white box in left panel, when only the red and green channels are shown. Note NG2+ cells are not marked with YFP. Shown is a representative field. E) Flow cytometry analysis of digested pancreatic tissue from adult Nkx3.2-Cre;R26-YFP mice. Left, dotplot showing the presence of yellow fluorescent cell population (gated cells) in Nkx3.2-Cre;R26-YFP pancreas. Right, green histograms (‘+Primary antibody’) showing staining of YFP+ cells (as gated in left panel) for the pericytic marker PDGFRb. Gray histogram showing Nkx3.2/YFP+ cells without addition of primary antibody (‘Staining control’). Number represents the percentage of PDGFRb-stained cells out of YFP+ cell population (as indicated by horizontal line). Note that majority of YFP+ cells express the pericyte marker PDGFRb.

3 Diabetes Page 40 of 47

Sakhneny et al.

mTcf7l2-transfected HEK293

Mural cells

Islets Blank

82 Tcf7l2 64 kDa Actin

Supplementary Figure 2: Western blot analysis detects Tcf7l2 protein in pancreatic mural cells Protein extract from purified pancreatic mural cells (3 µg), isolated islets (20 µg) and mouse Tcf7l2 (mTcf7l2) transfected HEK293 cells (3 µg) were analyzed using antibodies against Tcf7l2 (upper panels) and pan-actin (lower panels). Pancreatic mural cells were FACS sorted from Nkx3.2-Cre;R26-EYFP pancreatic tissue based on their yellow fluorescent labeling (as shown in Supplementary Fig. 1). Islets were isolated from wild type mice. HEK293 cells were transfected with 1 µg plasmid containing mTcf7l2 gene for 48 hours prior to analysis. Note the presence of multiple Tcf7l2 products in pancreatic mural cells.

Western blot analysis: Immunoblotting was performed by homogenizing cells in RIPA buffer. Protein samples were separated on 10% SDS/PAGE gel (Invitrogen) and transferred to a nitrocellulose membrane (Invitrogen). Membrane was stained with primary antibodies (Supplementary Table 1), followed by incubation with HRP-conjugated secondary antibodies (Abcam). Signal was detected using ECL reagents (SignalFire Elite; Cell Signaling).

4 Page 41 of 47 Diabetes

Sakhneny et al.

A B IPGTT, Males Wild type IPGTT, Females Non tg 400 Nkx3.2-Cre 400 Nkx3.2-Cre;Tcf7l2f/+ 300 300 Nkx3.2-Cre;Tcf7l2f/f

200 200

100 100

0 0 Blood glucose (mg/dL) 0 30 60 90 120 Blood glucose (mg/dL) 0 30 60 90 120 Time (min) Time (min)

Supplementary Figure 3: Intact glucose tolerance in Nkx3.2-Cre male mice and Tcf7l2 transgenic female mice A) Intraperitoneal Glucose tolerance test (IPGTT) on Nkx3.2-Cre (gray squares) and wild type (black circles) age-matched male mice. After overnight fasting, mice were intraperitoneally injected with dextrose (2 mg/g body weight) and tail vein blood glucose levels were measured at indicated time points. N = 5-7 B) IPGTT on 13-weeks old Nkx3.2-Cre;Tcf7l2flox/flox (red circles), Nkx3.2-Cre;Tcf7l2flox/+ (orange triangles) and non-transgenic littermate (‘non tg’; gray squares) female mice. After overnight fasting, mice were intraperitoneally injected with dextrose (2 mg/g body weight) and tail vein blood glucose levels were measured at indicated time points. N = 9

5 Diabetes Page 42 of 47

Sakhneny et al.

Supplementary Figure 4: Body weight, GLP-1 production and insulin sensitivity are maintained in Nkx3.2-Cre;Tcf7l2flox/flox mice Nkx3.2-Cre;Tcf7l2flox/flox (red circles), Nkx3.2-Cre;Tcf7l2flox/+ (orange triangles) and non- transgenic littermate (Cre-negative; ‘non tg’; gray squares) mice were analyzed. A,B) Box-and-whisker plots show body weight in 13-weeks males. Mice were fed either regular chow (B; N = 8-12) or high-fat diet (HFD)(C; N = 6-9), when these fed HFD and reaching a body weight of >32 grams at 13 weeks of age were considered obese and used for analysis. C,D) Bar diagrams (mean ± SD) show relative expression of Gcg (D) and Pcsk1 (E) in gut of transgenic and control mice. RNA was extracted from indicated regions of the gut, and gene expression was analyzed by qPCR, when average levels in non-transgenic tissues were set to ‘1’. N = 3. E) Bar diagrams (mean ± SD) show levels of serum GLP-1. After an overnight fast, blood was collected from mouse tails, and GLP-1 levels were analyzed by ELISA. N = 4. F,G) Intraperitoneal insulin tolerance test (ITT). Insulin (0.75 U/kg body weight) was intraperitoneally injected after a 6-hour fast to 13-weeks male mice, and tail vein blood glucose levels (mean ± SEM) were analyzed at indicated time points. N = 6 - 9.

6 Page 43 of 47 Diabetes

Sakhneny et al.

Supplementary Figure 5: Loss of pericytic Tcf7l2 does not result in b-cell apoptosis Nkx3.2-Cre;Tcf7l2flox/flox (red) and non-transgenic littermate (‘non tg’; gray) mice were analyzed at 13 weeks of age. A) Left and middle panels, pancreatic tissues of transgenic (middle panel) and non-transgenic (left panel) mice were subjected to TUNEL assay (green) to identify dying cells, and were stained for insulin (red) to identify b-cells. Right panel, non-transgenic pancreatic tissue pre- treated with DNase to induce DNA breaks, which served as a positive control of the TUNEL assay (‘staining control’). Shown are representative fields. B) Box-and-whisker plot showing gene expression analysis of isolated islets. RNA was extracted and expression of the b-cell stress genes Chop and Atf4 was analyzed by qPCR, when average levels in non-transgenic islets were set to ‘1’. N = 5.

7 Diabetes Page 44 of 47

Sakhneny et al.

2 untreated +rBMP4 NS NS Cultured islets, wild type

1

Relative expression 0 MafA Pdx1

Supplementary Figure 6: Culturing wild type islets in the presence of rBMP4 does not affect expression of b-cell genes Islets isolated from wild type mice were cultured in media supplemented with 6 ng/ml recombinant BMP4 (‘+rBMP4’; striped bars) or unsupplemented media (untreated; gray bars) for 24 hours. RNA was extracted and expression levels of indicated genes were analyzed by qPCR, when average levels in untreated islets were set to ‘1’. NS = non-significant, as compared to untreated control. N=4. Representative of two independent experiments.

8 Page 45 of 47 Diabetes

Sakhneny et al.

Supplementary Table 1: List of primary antibodies

Host Catalog Antigen Manufacturer Application species number Actin Mouse Millipore MAB1501 Western blot αSMA Mouse Sigma-Aldrich A5228 Immunofluorescence Desmin Mouse Sigma-Aldrich D1033 Immunofluorescence Glucagon Rabbit Millipore AB932 Immunofluorescence GFP / YFP Chicken Abcam AB13970 Immunofluorescence Insulin Guinea pig Dako A0564 Immunofluorescence NG2 Mouse Millipore MAB5384 Immunofluorescence NG2 Rabbit Millipore AB5320 Immunofluorescence PDGFRβ, Rat Affymetrix 13-1402 Flow cytometry biotinylated PECAM1 Rat BD 553370 Immunofluorescence Immunofluorescence, Tcf7l2 Rabbit Cell Signaling 2569 Western blot

9 Diabetes Page 46 of 47

Sakhneny et al.

Supplementary Table 2: List of qPCR probes

Gene Method Primers Atf4 Taqman Applied Biosystem expression assay Bmp4 Taqman Applied Biosystem expression assay Chop Taqman Applied Biosystem expression assay TGCCGCCAGTGCCATT Cyclophilin SYBR green TCACAGAATTATTCCAGGATT TGCACCACCAACTGCTTAG Gapdh Taqman GGATGCAGGGATGATGTTC Probe: CAGAAGACTGTGGATGGCCCCTC Gcg Taqman Applied Biosystem expression assay Glpr1 Taqman Applied Biosystem expression assay Glut2 Taqman Applied Biosystem expression assay Hif1a Taqman Applied Biosystem expression assay GGGTCGAGGTGGGCC Ins1 SYBR green CTGCTGGCCTCGCTTGC GGCTGCGTAGTGGTGGGTCTA Ins2 SYBR green CCTGCTCGCCCTGCTCTT Kir6.2 Taqman Applied Biosystem expression assay GCTGGTATCCATGTCCGTGC MafA SYBR green TGTTTCAGTCGGATGACCTCC NeuroD1 Taqman Applied Biosystem expression assay Pcsk1 Taqman Applied Biosystem expression assay Pdx1 Taqman Applied Biosystem expression assay Sur1 Taqman Applied Biosystem expression assay GCCATCAACCAGATTCTC Tcf7l2 SYBR green TTCTTCTTCCCATAGTTATCC

10 Page 47 of 47 Diabetes

Sakhneny et al.

Supplementary Table 3: RNA sequencing data To characterize gene expression of pancreatic mural cells, cells were purified by flow- cytometry (as described in Supplementary Fig. 1) from pancreatic tissue of Nkx3.2- Cre;R26-YFP 10-week old mice (N = 3). To characterize Tcf7l2-dependent gene expression of pancreatic mural cells, cells were further purified by flow-cytometry (as described in Supplementary Fig. 1) from pancreatic tissue of Nkx3.2-Cre;R26- YFP;Tcf7l2flox/flox 10-week old mice (N = 3) and islets were isolated from age-matched non-transgenic mice (N= 3). RNA was extracted from isolated cells and subjected to RNA sequencing to obtain 1.5-2 x 107 reads from each sample. Gene expression data have been deposited in ArrayExpress, and files can be accessed at: http://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-5325

11