Page 1 of 84 Diabetes

Pericyte-derived Dickkopf2 regenerates damaged penile neurovasculature through an angiopoietin-1-Tie2 pathway

Guo Nan Yin1,*, HaiRong Jin1,2,*, MinJi Choi1, Anita Limanjaya1, Kalyan Ghatak1, 1 1 1 1 3 Nguyen Nhat Minh , Jiyeon Ock , MiHye Kwon , KangMoon Song , Heon Joo Park , Ho Min Kim4, YoungGuen Kwon5, JiKan Ryu1,6, †, JunKyu Suh1, †

1National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea; 2Department of Urology, Yuhuangding Hospital, Yantai 264000, Shandong Province, China; 3Hypoxia related Disease Research Center, Inha University College of Medicine, Incheon 22212, Republic of Korea; 4Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; 5Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; 6Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon 22212, Republic of Korea

*These authors contributed equally to this work.

†Correspondence and requests for materials should be addressed to J.K.S. or J.K.R. (email: [email protected] or [email protected])

JunKyu Suh, MD, PhD National Research Center for Sexual Medicine and Department of Urology Inha University College of Medicine 7206, 3rd St, ShinheungDong, JungGu Incheon 22332, Republic of Korea Tel: 82328903441, Fax: 82328903097 Email: [email protected]

Diabetes Publish Ahead of Print, published online March 20, 2018 Diabetes Page 2 of 84

JiKan Ryu, MD, PhD National Research Center for Sexual Medicine and Department of Urology Inha University College of Medicine 7206, 3rd ST, ShinheungDong, JungGu, Incheon 22332 Republic of Korea Tel: 82328903505; Fax: 82328903099 Email: [email protected]

Running title: Dickkopf2 regenerates damaged penile neurovasculature Word count (abstract): 140 Word count (main text): 4549 Number of Figures: 6 Number of Tables: 0 Number of Supplemental Figures: 12 Number of Supplemental Tables: 7

Page 3 of 84 Diabetes

Abstract

Penile erection requires wellcoordinated interactions between vascular and nervous

system. Penile neurovascular dysfunction is a major cause of erectile dysfunction (ED)

in patients with diabetes, which causes poor response to oral phosphodiesterase5

inhibitors. Dickkopf2 (DKK2), a Wnt antagonist, is known to promote angiogenesis.

Here, using DKK2Tg mice or DKK2 administration, we demonstrate that

overexpression of DKK2 in diabetic mice enhances penile angiogenesis and neural

regeneration and restores erectile function. Transcriptome analysis revealed that

angiopoietin1 and angiopoietin2 are target for DKK2. Using an endothelial

cellpericyte coculture system and ex vivo neurite sprouting assay, we found that

DKK2mediated juxtacrine signaling in pericyteendothelial cell interactions promotes

angiogenesis and neural regeneration though an angiopoietin1Tie2 pathway, rescuing

erectile function in diabetic mice. The dual angiogenic and neurotrophic effects of

DKK2, especially as a therapeutic protein, will open new avenues to treating diabetic

ED.

Keywords: Dickkopf2, erectile dysfunction, diabetes mellitus, pericyte, angiopathy,

neuropathy Diabetes Page 4 of 84

Penile erection is a neurovascular phenomenon that requires wellcoordinated interactions between vascular endothelial cells, smooth muscle cells, pericytes, and neuronal cells (1, 2). Erectile dysfunction (ED) affects more than half of men aged 40 to 70 years (3). A variety of pathological conditions, including vascular risk factors or diseases, neurological abnormalities, and hormonal disturbances, are involved in penile neurovascular dysfunction (4).

Phosphodiesterase type 5 (PDE5) inhibitors enhance the nitric oxide (NO) cGMP pathway and are currently utilized as a firstline therapy for ED (1). The reduced responsiveness to PDE5 inhibitors in patients with neuropathy, severe angiopathy, or both, such as diabetes, may be related to a decrease in the endogenous

NO released from the nerve terminal and/or endothelial cells of erectile tissue (5, 6).

Therefore, curative therapy for advanced ED, including diabetic ED, requires a new therapeutic strategy that reestablishes structural and functional penile neurovasculature and augments endogenous NO bioactivity.

Several have been effective in targeting angiogenesis of the penis in diabetic or hypercholesterolemic animal models of ED, including vascular endothelial growth factor (VEGF), angiopoietin1 (Ang1), and angiopoietin4 (714). Targeting neural regeneration, derived neurotrophic factor (BDNF) or neurotrophin3 (NT

3) have shown some effectiveness by restoring neuronal nitric oxide synthasepositive

(nNOS+) neurons (15, 16). However, new therapeutics are far from development because of incomplete effects, potential adverse effects, such as inflammation, and difficulties engineering protein for medicine. Moreover, there is no single candidate protein that can solve the complicated underlying pathology, both angiopathy and neuropathy, in diabetic ED. Therefore, the development of a treatment modality Page 5 of 84 Diabetes

targeting these complicated pathologies in refractory ED would be ideal.

Dickkopf2 (DKK2) is a secreted protein containing two cysteinerich regions

that acts as a Wnt antagonist by binding lowdensity lipoprotein (LDL) related

protein 5/6 (1719). Recently, DKK2 was found to improve recovery from hind limb

ischemia and myocardial infarction by enhancing angiogenesis (20). Moreover, in a

corneal angiogenesis assay, DKK2induced capillaries had more coverage of

endothelial cells by pericytes and were less leaky than VEGFinduced vessels (20),

which suggests that DKK2 promotes mature and stable blood vessel formation.

However, the role of DKK2 in the penis has not yet been explored. Therefore, we

hypothesized that DKK2 may be a potential target for therapeutic angiogenesis, which

ultimately leads to the restoration of physiologic erection. We also determined the role

of DKK2 in penile neural regeneration.

Here, we demonstrate that overexpression of DKK2 in DKK2Tg mice that

express mouse DKK2 under the control of the endothelial cellspecific Tie2

promoter/enhancer, or in wildtype (WT) mice via local administration of DKK2

protein into the penis, rescues erectile function under diabetic conditions. This

recovery was accompanied by enhanced proliferation of cavernous endothelial cells

and pericytes; phosphorylation of endothelial NOS (eNOS), restoration of the integrity

of endothelial cellcell junctions, and decreased cavernous vascular permeability; and

enhanced neural regeneration through the secretion of neurotrophic factors.

Transcriptome analysis of DKK2 target genes in primary cultures of mouse cavernous

endothelial cells (MCECs) revealed that Ang1 expression is downregulated, and the

expression of angiopoietin2 (Ang2), an endogenous antagonist of Ang1, upregulated

by high glucose (HG). This effect was reversed by treatment with DKK2 protein. Diabetes Page 6 of 84

Using an MCECmouse cavernous pericyte (MCP) coculture system and an ex vivo neurite sprouting assay, we also found that DKK2mediated juxtacrine signaling in

MCECsMCPs promotes angiogenesis and neural regeneration though an Ang1Tie2 pathway. The recovery of erectile function mediated by DKK2 was abolished by inhibition of Ang1Tie2 signaling with soluble Tie2 protein (sTie2Fc). Page 7 of 84 Diabetes

Research design and Methods

Study design

The primary aim of the present study was to investigate the mechanisms through which

DKK2 restores diabetesinduced ED. We used DKK2Tg mice and administered

DKK2 protein into the penis of diabetic mice. Detailed mechanisms were evaluated

with WT or DKK2Tg mice and primary cultures of MCECs, MCPs, and Neuro2A

cells. All parameters of genetically modified mice and diabetic mice were compared to

those of littermate controls.

Animals and treatments

Eightweekold male C57BL/6 (Orient Bio) and DKK2Tg mice (provided by Young

Guen Kwon, Yonsei University, Korea) were used in this study. DKK2Tg mice were

backcrossed with C57BL/6 for at least seven generations (20). The experiments were

approved by the Institutional Animal Care and Use Committee of Inha University

(Assurance Number: INHA 1401102671). Diabetes was induced by intraperitoneal

injection of multiple low doses of STZ (50 mg/kg body weight in 0.1 M citrate buffer,

pH 4.5) for 5 consecutive days as described previously (21). Eight weeks after diabetes

was induced, the mice were anesthetized with intramuscular injections of ketamine

(100 mg/kg) and xylazine (5 mg/kg) and placed supine on a thermoregulated surgical

table.

For the DKK2Tg study, the mice were distributed into four groups: WT

controls, DKK2Tg mice, WT mice receiving STZ (50 mg/kg body weight for 5 days),

and DKK2Tg mice receiving STZ (50 mg/kg body weight for 5 days). Eight weeks Diabetes Page 8 of 84

after the induction of diabetes, we measured erectile function during electrical stimulation of the cavernous nerve.

For the inhibition study with sTie2Fc, the mice were distributed into four groups: DKK2Tg mice, STZinduced WT diabetic mice, and STZinduced DKK2Tg diabetic mice receiving subcutaneous injection of dimericFc or sTie2Fc (4 µg/20 µl;

R&D Systems). The dose of sTie2Fc was determined based on our previous report

(22). sTie2Fc or dimericFc was administered 8 weeks after the induction of diabetes.

Two weeks after treatment, we measured erectile function, and then the penis was harvested for histological examination.

To test the efficacy of DKK2 protein, the mice were distributed into three groups: agematched controls and STZinduced WT diabetic mice receiving repeated intracavernous injections of PBS or DKK2 protein (days 3 and 0; 6 µg/20 µl; A&R

Therapeutics). For the inhibition study with sTie2Fc, the mice were distributed into four groups: agematched controls and STZinduced WT diabetic mice receiving repeated intracavernous injections of PBS, DKK2 protein (days 3 and 0; 6 µg/20 µl) and dimericFc, or DKK2 protein (days 3 and 0; 6 µg/20 µl) and sTie2Fc (4 µg/20

µl). sTie2Fc or dimericFc was administered immediately before the injection of

DKK2 protein. We evaluated erectile function by cavernous nerve electrical stimulation 2 weeks after treatment. The penis was harvested for histological examination and biochemical study.

To examine the effect of insulin treatment on erectile function, the mice were distributed into three groups: agematched controls and STZinduced WT diabetic mice receiving repeated intraperitoneal injection of PBS or insulin (4 IU/day; Sigma

Aldrich) (23,24). Insulin treatment started 1 week after STZ injection (9 weeks of age) Page 9 of 84 Diabetes

and continued for 9 weeks (18 weeks of age). We measured erectile function during

electrical stimulation of the cavernous nerve.

Fasting and postprandial blood glucose levels were determined by an Accu

check blood glucose meter (Roche Diagnostics) before the mice were sacrificed. We

also measured glycosylated hemoglobin (HbA1c; A1C Now System, PTS Diagnostics)

and serum insulin levels by using a mouse insulin ELISA (Mercodia).

Human corpus cavernosum tissue

Human corpus cavernosum tissues were obtained from a 21yearold patient with

congenital penile curvature who had normal erectile function during reconstructive

penile surgery and a 56yearold patient with diabetic ED (type 2 DM; duration of DM,

2 22 years; HbA1c level, 8.8% [73 mmol/mol]; BMI, 23.1 kg/m ; comorbidity,

hypertension; medications, subcutaneous insulin and oral metformin) during penile

prosthesis implantation. All tissue donors provided informed consent, and the

experiments were approved by the internal review board of Inha University.

Measurement of erectile function

Measurement of erectile function was performed as described previously (21). Details

can be found in ‘Supplemental Methods’.

Cell culture experiments

The MCECs and MCPs were prepared and maintained as described previously (2, 25,

26). Tube formation assay, scratch woundhealing assay, transfection assay, RTPCR,

and cDNA microarray were performed as described in ‘Supplemental Methods’. Diabetes Page 10 of 84

Aortic ring assay

Aortas were harvested from 8weekold C57BL/6 WT mice. The aortic rings were placed in the 8well Nunc LabTek Chamber Slide System (SigmaAldrich) and sealed in place with an overlay of 50 µl matrigel. The aortic rings were cultured in medium

199 with 20 ng/ml of basic fibroblast growth factor and 1% penicillin/streptomycin for

5 days. The aortic segments and sprouting cells were fixed in 4% paraformaldehyde for at least 30 minutes and used for immunofluorescent staining.

Ex vivo neurite sprouting assay

The mouse major pelvic ganglion (MPG) tissues were prepared and maintained as described previously (27), with minor modifications. The MPG tissues were isolated from male mice using a microscope, transferred into sterile vials containing Hank’s balanced salt solution (Gibco), and then rinsed and washed twice in PBS. The MPG tissues were cut into small pieces and the samples plated on polyDlysine hydrobromidecoated (SigmaAldrich) 12well plate. The whole MPG tissue was covered with matrigel and the culture plate placed on ice for 5 minutes prior to incubation at 37°C for 1015 minutes in a 5% CO2 atmosphere. We added 1 ml of complete Neurobasal medium (Gibco) supplemented with 2% serumfree B27

(Gibco) and 0.5 nM GlutaMAX™I (Gibco). The dishes were then incubated at 37°C in a 5% CO2 atmosphere. Three days after incubation, we evaluated neurite outgrowth.

Establishment of in vitro or ex vivo experimental systems that mimic diabetic ED

To mimic an in vivo or ex vivo condition for diabetesinduced angiopathy and Page 11 of 84 Diabetes

neuropathy, primary cultures or tissues were serumstarved for 24 hours and then

exposed to normal glucose (NG, 5 mmol) or HG (30 mmol; SigmaAldrich) conditions

for 2 days (MCECs, MCPs, and Neuro2A cells), 3 days (MPG tissue), or 5 days (aortic

ring).

Preparation of conditioned medium

To examine the effect of pericytederived DKK2 on endothelial cells, the conditioned

medium (CM) derived from MCPs in the presence or absence of DKK2 depletion was

transferred to MCECs. In addition, CM derived from MCECs in the presence of

absence of DKK2 depletion was transferred to MCPs to determine the effect of

endothelial cellderived DKK2 on pericytes. To do this, MCECs and MCPs were

grown in 60mm dishes until 80% confluence and the medium changed for an

additional 2 days. The culture supernatants were collected and centrifuged for 10

minutes at 200 × g to remove cell debris. For DKK2 depletion, an immunoprecipitation

protocol was used. Briefly, the CM was incubated with DKK2 antibody (1:200; Abcam,

Cat. Ab95274) or control rabbit IgG (1:100; Santa Cruz Biotechnology, Cat. sc2027)

for 12 hours at 4°C. Protein Gcoupled Sepharose beads (Millipore) were added to the

medium and incubated for an additional 12 hours at 4°C to remove DKK2 antibody

and protein. MCP or MCEC complement medium was used as a control.

We also determined the effect of CM derived from MCECMCP coculture on

neurite sprouting from MPG tissue. To do this, MCECs and MCPs were cultured and

treated under the following conditions: NG; HG + PBS; HG + DKK2 protein (200

ng/ml) + scrambled siRNA; HG + DKK2 protein (200 ng/ml) + Ang1 siRNA (200

pmol) transfection in MCECs; HG + DKK2 protein (200 ng/ml) + Ang1 siRNA (200 Diabetes Page 12 of 84

pmol) transfection in MCPs; and HG + DKK2 protein + Ang1 siRNA (200 pmol) transfection in both MCECs and MCPs. The culture supernatants were collected and centrifuged for 10 minutes at 200 × g to remove cell debris and then transferred to

MPG tissue.

Histological examinations

Histologic examinations and BrdU labeling were performed as described in

‘Supplemental Methods’.

Western blot

Western blot analysis was performed as described in ‘Supplemental Methods’.

Statistical analysis

The results are expressed as mean ± SE. Intergroup comparisons were made by Mann

Whitney U test or KruskalWallis test. Pvalues < 5% were considered significant. We used SigmaStat 3.11 software (Systat Software) for statistical analyses. Page 13 of 84 Diabetes

Results

Metabolic variables

The fasting and postprandial blood glucose concentrations as well as HbA1c levels in

streptozotocin (STZ)treated WT or DKK2Tg diabetic mice were significantly higher

than in control mice. In addition, body weight and serum insulin levels were

significantly lower in the STZinduced diabetic mice than in the controls. However, the

body weight and blood glucose levels of the diabetic mice did not differ significantly

regardless of treatment (Supplementary Table S1S4).

DKK2 is mainly expressed in pericytes

The expression of DKK2 mRNA and protein was significantly higher in primary

cultures of MCPs and human brain microvascular pericytes (HBMPs) than in MCECs

and human umbilical vein endothelial cells (HUVECs), respectively (Fig. 1AD and

Supplementary Fig. 1). Similarly, immunohistochemical staining of normal human or

mouse erectile tissue revealed that a significant proportion of the DKK2 expression

overlapped with pericytes and partially colocalized with endothelial cells (Fig. 1E to

H).

Cavernous expression of DKK2 is decreased under diabetic conditions

Western blot analysis revealed a decrease in DKK2 expression in the penis tissue of

diabetic mice in vivo and in MCECs or MCPs exposed to HG conditions in vitro (Fig.

1IN). Immunofluorescent staining also revealed that the expression of DKK2 protein

was significantly lower in cavernous tissue from diabetic patients or diabetic mice than

in controls (Fig. 1OQ). These findings gave us a rationale to use DKK2 for the Diabetes Page 14 of 84

treatment of diabetic ED.

Overexpression of DKK2 preserves the regenerative potential of endothelial cells and pericytes under diabetic conditions

Eight weeks after the injection of STZ into WT or DKK2Tg mice and the induction of diabetes, the cavernous endothelial cell and pericyte content was significantly lower in

WT mice that received STZ than in untreated WT mice, whereas cavernous endothelial cell and pericyte content was relatively preserved in DKK2Tg mice that received STZ

(Fig. 2A and G). Moreover, DKK2 profoundly enhanced the proliferation of endothelial cells and pericytes under diabetic conditions both in vivo and in vitro (Fig.

2B and H and Supplementary Fig. 2). Overexpression of DKK2 significantly induced the phosphorylation of Akt and eNOS (Supplementary Fig. 3), restored cavernous endothelial cellcell junction proteins claudin5 and vascular endothelial (VE)cadherin

(Supplementary Fig. 4), and decreased the extravasation of oxidized LDL

(Supplementary Fig. 5) under diabetic conditions compared to WT littermates that received STZ.

We further examined the role of DKK2 in MCEC and MCP monoculture or direct mixed coculture. The mixture of MCECs and MCPs formed wellorganized capillarylike structures at the ratio of 3:1 (Supplementary Fig. 6). In vitro matrigel assays revealed impaired tube formation in MCEC and MCP monoculture or coculture exposed to HG, and these impairments were completely restored by treatment with

DKK2 protein (200 ng/ml) (Fig. 2 C and I). DKK2 protein also promoted MCEC, MCP,

HUVEC, and HBMP migration under HG conditions (Fig. 2D and J and

Supplementary Fig. 7). In an ex vivo aortic ring assay, the average length and branch Page 15 of 84 Diabetes

number of outgrowing microvessels were significantly lower in aortic segments

exposed to HG than segments exposed to NG conditions. Moreover, DKK2 protein

significantly enhanced the outgrowth of microvessels from aortic rings under HG

conditions (Fig. 2E and K). We observed higher DKK2 expression in the sprouting

front than the aortic ring (Fig. 2F).

Overexpression of DKK2 decreases cavernous ROS production under diabetic

conditions

Cavernous inducible NOS (iNOS) protein expression was significantly higher in the

PBStreated diabetic mice than in the agematched controls. Repeated intracavernous

injections of DKK2 protein (days 3 and 0; 6 µg/20 µl) significantly decreased

cavernous iNOS expression in the diabetic mice (Supplementary Fig. 8A-C). We also

performed immunohistochemical localization of nitrotyrosine to determine

peroxynitrite generation, which is derived from NO and superoxide anion, and in situ

analysis of superoxide anion production. Nitrotyrosine expression and the fluorescent

products of oxidized hydroethidine in endothelial cells of the corpus cavernosum were

significantly higher in WT mice that received STZ than in untreated WT mice, whereas

the generation of superoxide anion and nitrotyrosine was profoundly decreased in

DKK2Tg mice that received STZ. DKK2 protein also significantly decreased

cavernous ROS production in diabetic condition (Supplementary Fig. 8D to I).

Transmigration of DKK2 from pericytes to endothelial cells promotes

angiogenesis

Immunocytochemical staining revealed higher DKK2 expression in MCPs than Diabetes Page 16 of 84

MCECs. In contrast, after cultivation of MCECs and MCPs using an indirect non contact coculture system, DKK2 expression was higher in MCECs than in MCPs (Fig.

3A). To confirm whether pericytederived DKK2 migrates into the endothelial cells,

MCPs were transfected with DKK2RFP DNA. We observed DKK2RFP expression in

MCECs after coculture with DKK2RFPtransfected MCPs (Fig. 3B).

To test the functional role of pericytederived DKK2 on endothelial cells,

MCECs were treated with CM derived from MCPs in the presence or absence of

DKK2. We observed enhanced tube formation in MCECs treated with MCPCM compared to cells treated with complement medium for MCPs. However, MCECs treated with DKK2depleted MCPCM had profoundly impaired tube formation (Fig.

3C and D). Although DKK2depleted MCECCM slightly decreased tube formation in

MCPs compared to cells treated with DKK2 containing CM, it was not significant (Fig.

3E and F).

Overexpression of DKK2 preserves neurotrophic function under diabetic conditions

The expression of , βIII , and nNOS in dorsal nerve bundle or corpus cavernosum was significantly lower in WT mice that received STZ than in untreated WT mice, whereas the neuronal cell content was completely restored in

DKK2Tg mice that received STZ (Fig. 4A, D, E, F). DKK2 protein also significantly enhanced neurite sprouting in an ex vivo MPG tissue culture exposed to HG (Fig. 4B and G).

Next, we asked whether the effects of DKK2 were mediated by the production of neurotrophic factors and their receptors. The cavernous expression of nerve growth Page 17 of 84 Diabetes

factor (NGF), BDNF, and TrkA was significantly higher in diabetic mice receiving

DKK2 protein than in PBStreated diabetic mice and comparable to the level found in

agematched controls. We observed similar results in mouse albino neuroblastoma

(Neuro2A) cells in vitro (Fig. 4C, H, I, J, K). The expression of TrkB and TrkC was not

detectable in the penis or Neuro2A cells.

Overexpression of DKK2 preserves erectile function under diabetic conditions

In accordance with DKK2mediated angiogenesis and neural regeneration, the ratio of

maximal intracavernous pressure (ICP) or total ICP to mean systolic blood pressure

(MSBP) was significantly lower in WT mice that received STZ than in untreated WT

mice, whereas erectile function was relatively preserved in DKK2Tg mice that

received STZ (Supplementary Fig. 9A, C, D).

Two weeks after treatment, repeated intracavernous injections of DKK2

protein (days 3 and 0; 6 µg/20 µl) significantly induced recovery of erection

parameters in WT mice that received STZ (Supplementary Fig. 9B, E, F). No

detectable differences in MSBP were found among the experimental groups

(Supplementary Table S1 and S2). The dosage of DKK2 protein was determined based

on our pilot study. We obtained the highest erectile function recovery at a

concentration of 6 µg/20 µl (Supplementary Fig. 10).

Transcriptome analysis of DKK2 target genes in MCECs

To identify the genes regulated by DKK2, microarray analysis was performed. We

selected genes for which the ratios changed more than 2fold in both conditions, i.e.,

MCECs exposed to NG conditions compared to those exposed to HG conditions + PBS, Diabetes Page 18 of 84

and MCECs exposed to HG conditions + DKK2 protein compared to those exposed to

HG conditions + PBS. After filtering data from 39,429 genes, 201 genes were changed more than 2fold. Of these genes, only three were downregulated under HG conditions compared to NG conditions. These levels were reversed after treatment with DKK2 protein (Supplementary Table S5 and S6). These genes included Angpt1 (Ang1) and

Angpt2 (Ang2) (Fig. 5A).

DKK2-mediated angiogenesis and neural regeneration and the recovery of erectile function are dependent on the Ang1-Tie2 signaling pathway

We further confirmed that Ang1 mRNA and protein expression were significantly lower and Ang2 expression significantly higher in MCECs or MCPs exposed to HG conditions than in cells exposed to NG conditions. The expression of both Ang1 and

Ang2 returned to baseline values after treatment with DKK2 protein (Fig. 5BD).

Physiological erection studies revealed that inhibition of the Ang1Tie2 pathway by sTie2Fc (4 µg/20 µl) abolished DKK2mediated erectile function recovery in both diabetic DKK2Tg mice and diabetic WT mice treated with DKK2 protein (Fig. 5EJ). No detectable differences in MSBP were found among the experimental groups (Supplementary Table S3 and S4).

Immunofluorescent staining of penis tissue revealed that the enhanced cavernous angiogenesis and neural regeneration were profoundly diminished in STZ injected DKK2Tg mice treated with sTie2Fc (Fig. 5K, N, O). Treatment of MCEC

MCP coculture or MPG tissue with sTie2Fc also abolished the DKK2mediated enhancement of tube formation and neurite sprouting under HG conditions (Fig. 5I, M,

P, Q). Page 19 of 84 Diabetes

We also examined the cellular sources of Ang1 that may play a major role in

DKK2mediated angiogenesis and neural regeneration. Compared to MCECs

transfected with Ang1 siRNA, the transfection of Ang1 siRNA into MCPs or both

MCECs and MCPs profoundly diminished the DKK2mediated enhancement of tube

formation in mixed MCECMCP coculture exposed to HG (Supplementary Fig. 11A

and B). We also determined whether CM derived from MCECs or MCPs cultivated

under the aforementioned conditions affects neural regeneration. Similar to the results

of the tube formation assays, DKK2mediated neurite sprouting was more impaired in

MPG tissue treated with CM derived from Ang1 siRNAtransfected MCPs than in the

tissue treated with CM derived from Ang1 siRNAtransfected MCECs (Supplementary

Fig. 11C and D). These findings suggest that pericytederived Ang1 plays a crucial role

in DKK2mediated angiogenesis and neural regeneration.

Insulin treatment does not prevent the deterioration of erectile function under

diabetic conditions

We finally examined whether insulin treatment rescues erectile function in diabetic

condition. The treatment of diabetic mice with insulin did not restore erectile function

(Supplementary Fig. 12AF). Insulin treatment also failed to restore cavernous DKK2

expression in the diabetic mice (Supplementary Fig. 12G and H). Metabolic and

physiologic variables, including body weight, blood glucose concentrations, and

systemic blood pressure, are summarized in Supplementary Table S7. Diabetes Page 20 of 84

Discussion

Here, we investigated whether DKK2 plays a role as a positive regulator of angiogenesis and neural regeneration, exerting beneficial effects in diabetic ED.

DKK2mediated interactions between pericytes and endothelial cells promoted angiogenesis and neural regeneration through an Ang1Tie2 pathway, rescuing erectile function in diabetic animals. The detailed mechanisms of action by which DKK2 restores erectile function are illustrated in Fig. 6.

To test whether DKK2 induces cavernous angiogenesis under pathological conditions, we immunohistochemically evaluated the expression of CD31 and platelet derived growth factor receptorβ (PDGFRβ). Similar to the results of previous studies in STZinduced diabetic rats (28, 29) and mice (2, 9, 11), the cavernous endothelial cell and pericyte area was significantly smaller in WT diabetic mice than in control mice. BrdU labeling revealed increased endothelial cell and pericyte proliferation, and these cellular contents were relatively well preserved in STZtreated DKK2Tg mice.

DKK2 protein also promoted tube formation, proliferation, and migration by endothelial cells and pericytes, and enhanced microvessel sprouting from the aortic ring under HG conditions.

Endothelial cellcell junctions serve as a barrier by regulating paracellular permeability and play a crucial role in vascular formation, the vascular network, and remodeling of blood vessels (30). Diabetes mellitus promotes LDL oxidation and extravasation, which in turn induces vascular inflammatory responses and endothelial cell apoptosis (31). We recently revealed impaired cavernous endothelial cellcell junctions and increased cavernous endothelial permeability to oxidized LDL in diabetic mice (32). In the present study, cavernous endothelial cellcell junction Page 21 of 84 Diabetes

proteins were well preserved and less oxidized LDL extravasated in STZinjected

DKK2Tg mice than in STZinjected WT littermates. The restoration of endothelial

cellcell junctions and enhanced pericyte coverage on endothelial cells by DKK2 may

be attributable to a decrease in cavernous vascular permeability.

The interaction between endothelial cells and pericytes plays a crucial role in

blood vessel formation and vascular maturation (33). The close anatomical relationship

between endothelial cells and pericytes implicates paracrine or juxtacrine signaling, i.e.,

endothelial cellpericyte signaling. Using human and mouse erectile tissues in vivo,

and endothelial cells (MCECs and HUVECs) and pericytes (MCPs and HBMPs) in

vitro, we found that pericytes are the major source of DKK2 expression. In indirect

noncontact MCEC and DKK2RFPtransfected MCP coculture experiments, we

confirmed that pericytederived DKK2 migrates into endothelial cells. Moreover, we

found profound tube formation impairment in MCECs treated with DKK2depleted

MCPCM, whereas the treatment of MCPs with DKK2depleted MCECCM did not

significantly affect tube formation. These findings suggest that pericytederived DKK2

may play a crucial role in DKK2mediated angiogenesis.

The number of functional nNOS+ neurons is critical for physiological penile

erection (34). In the present study, DKK2Tg mice that received STZ had preserved

nNOS, neurofilament, and βIII tubulin expression in the corpus cavernosum and dorsal

nerve bundle. Moreover, direct administration of DKK2 protein or DKK2 protein

treated CM derived from MCECMCP coculture profoundly enhanced neurite

sprouting in MPG tissue under HG conditions. In penis tissue from WT diabetic mice

and Neuro2A cells exposed to HG, the expression of NGF, BDNF, and TrkA was

greatly restored by treatment with DKK2 protein. Given that the activation of Wnt Diabetes Page 22 of 84

signaling has neuroprotective effects (35, 36), the Wnt signaling antagonist DKK2 may exert its neurotrophic effects independent of the Wnt pathway.

Transcriptome analysis showed that Ang1 and Ang2 are target genes of DKK2.

Inhibition of the Ang1Tie2 pathway with sTie2Fc diminished DKK2mediated angiogenesis and neural regeneration in STZinjected DKK2Tg mice and MCECs or

MPG tissue exposed to HG. Pretreatment with sTie2Fc also abolished DKK2 mediated erectile function recovery in both STZinjected DKK2Tg mice and STZ injected WT mice treated with DKK2 protein. These findings suggest that the Ang1

Tie2 pathway is crucial for DKK2mediated restoration of the penile neurovascular structure and erectile function. Although the Ang1Tie2 pathway is well known to play an important role in generating a stable and functional vasculature (37), its role in the nervous system is largely unknown. However, Ang1 has been reported to promote neurite outgrowth in dorsal root ganglion cells positive for Tie2 receptor through transactivation of TrkA receptor (38).

For clinical applications, a GLP preclinical study of DKK2 protein has begun.

An interim report revealed no toxicity in C.B17 SCID mice (i.e., no unscheduled death, clinical signs, changes in body weight, or abnormal necropsy findings) after a single intravenous injection of DKK2 protein at a dosage of up to 90 mg/kg body weight (Supporting Data 1). Also, no tumorigenesis related to DKK2 protein was noted in cell lines (MDAMB231, MCF7) or a lung cancer cell line (A549) in vitro (Supporting Data 2) or in vivo after implantation of a lung cancer cell line (A549) in nude mice (Supporting Data 3). It was reported that DKK2Tg mice exhibited increased retinal blood vessel formation during developmental period (20). DKK2Tg mice also showed increased blood vessel density and decreased hemorrhage in the Page 23 of 84 Diabetes

oxygeninduced retinopathy model (39), suggesting clinical utility of DKK2 in diabetic

retinopathy.

Our study has some limitations. First, this study did not explain how DKK2

regulates the expression of Ang1 and Ang2. Second, the mechanisms by which the

Ang1Tie2 pathway is involved in neural regeneration need to be documented in detail.

Finally, we measured systemic blood pressure by use of a noninvasive tailcuff system,

not by direct carotid artery cannulation, because of concerns about the viability of

animals and the reliability of the ICP results with significant bleeding during the carotid

artery cannulation. Systemic blood pressure and ICP were not measured simultaneously,

because vibrations occurring from electrical stimulation of the cavernous nerve can

impede the accurate measurement of systemic blood pressure by use of the tailcuff

system. Thus, we measured systemic blood pressure immediately before electrical

stimulation of the cavernous nerve.

Taken together, our findings reveal a unique function of DKK2 in

reprogramming damaged erectile tissue toward neurovascular repair through an Ang1

Tie2 pathway. Dual angiogenic and neurotrophic effects of DKK2, especially in the

form of locally injectable protein, may provide a paradigm shift in the development of

novel therapeutics not only for ED, but also for other ischemic vascular diseases or

neurological disorders. Diabetes Page 24 of 84

Acknowledgements. This work was supported by a grant from the Korean Health

Technology R&D Project, Ministry of Health & Welfare, Republic of Korea (J.K.S.,

A110076; J.K.R. and H.M.K., H15C0508), by a Medical Research Center Grant (J.

K.R. and H.J.P., 2014R1A5A2009392), and by the National Research Foundation of

Korea (NRF) grant (J.K.R., 2016R1A2B2010087) funded by the Korean government

(Ministry of Science, ICT and Future Planning). Professor JiKan Ryu 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.

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

Author contributions. G.N.Y., H.R.J., J.K.R., and J.K.S. designed the experiments and wrote the manuscript; G.N.Y., H.R.J., M.J.C., A.L., K.G., N.N.M., J.O., K.M.H., and K.M.S. performed the experiments; Y.G.K. and H.M.K. contributed essential reagents; H.J.P. analyzed and critically discussed the data; and J.K.R. and J.K.S. supervised the project. Page 25 of 84 Diabetes

References

1. Andersson KE. Mechanisms of penile erection and basis for pharmacological treatment of erectile dysfunction. Pharmacol Rev 2011;63:811859 2. Yin GN, Das ND, Choi MJ, Song KM, Kwon MH, Ock J, Limanjaya A, Ghatak K, Kim WJ, Hyun JS, Koh GY, Ryu JK, Suh JK. The pericyte as a cellular regulator of penile erection and a novel therapeutic target for erectile dysfunction. Sci Rep 2015;5:10891 3. Feldman HA, Goldstein I, Hatzichristou DG, Krane RJ, McKinlay JB. Impotence and its medical and psychosocial correlates: results of the Massachusetts Male Aging Study. J Urol 1994;151:5461 4. Lue TF. Erectile dysfunction. N Engl J Med 2000;342:18021813 5. Angulo J, GonzalezCorrochano R, Cuevas P, Fernandez A, La Fuente JM, Rolo F, Allona A, Saenz de Tejada I. Diabetes exacerbates the functional deficiency of NO/cGMP pathway associated with erectile dysfunction in human corpus cavernosum and penile arteries. J Sex Med 2010;7:758768 6. Musicki B, Burnett AL. Endothelial dysfunction in diabetic erectile dysfunction. Int J Impot Res 2007;19:129138 7. Dall'Era JE, Meacham RB, Mills JN, Koul S, Carlsen SN, Myers JB, Koul HK. Vascular endothelial growth factor (VEGF) therapy using a nonviral gene delivery system improves erectile function in a diabetic rat model. Int J Impot Res 2008;20:307314 8. Gholami SS, Rogers R, Chang J, Ho HC, Grazziottin T, Lin CS, Lue TF. The effect of vascular endothelial growth factor and adenoassociated virus mediated brain derived neurotrophic factor on neurogenic and vasculogenic erectile dysfunction induced by hyperlipidemia. J Urol 2003;169:15771581 9. Jin HR, Kim WJ, Song JS, Piao S, Choi MJ, Tumurbaatar M, Shin SH, Yin GN, Koh GY, Ryu JK, Suh JK. Intracavernous delivery of a designed angiopoietin 1 variant rescues erectile function by enhancing endothelial regeneration in the streptozotocininduced diabetic mouse. Diabetes 2011;60:969980 10. Jin HR, Kim WJ, Song JS, Piao S, Tumurbaatar M, Shin SH, Choi MJ, Tuvshintur B, Song KM, Kwon MH, Yin GN, Koh GY, Ryu JK, Suh JK. Intracavernous delivery of synthetic angiopoietin1 protein as a novel therapeutic strategy for erectile dysfunction in the type II diabetic db/db mouse. J Sex Med 2010;7:36353646 11. Kwon MH, Ryu JK, Kim WJ, Jin HR, Song KM, Kwon KD, Batbold D, Yin Diabetes Page 26 of 84

GN, Koh GY, Suh JK. Effect of intracavernous administration of angiopoietin 4 on erectile function in the streptozotocininduced diabetic mouse. J Sex Med 2013;10:29122927 12. Rogers RS, Graziottin TM, Lin CS, Kan YW, Lue TF. Intracavernosal vascular endothelial growth factor (VEGF) injection and adenoassociated virus mediated VEGF gene therapy prevent and reverse venogenic erectile dysfunction in rats. Int J Impot Res 2003;15:2637 13. Ryu JK, Kim WJ, Koh YJ, Piao S, Jin HR, Lee SW, Choi MJ, Shin HY, Kwon MH, Jung K, Koh GY, Suh JK. Designed angiopoietin1 variant, COMP angiopoietin1, rescues erectile function through healthy cavernous angiogenesis in a hypercholesterolemic mouse. Sci Rep 2015;5:9222 14. Yamanaka M, Shirai M, Shiina H, Tanaka Y, Enokida H, Tsujimura A, Matsumiya K, Okuyama A, Dahiya R. Vascular endothelial growth factor restores erectile function through inhibition of apoptosis in diabetic rat penile crura. J Urol 2005;173:318323 15. Bakircioglu ME, Lin CS, Fan P, Sievert KD, Kan YW, Lue TF. The effect of adenoassociated virus mediated brain derived neurotrophic factor in an animal model of neurogenic impotence. J Urol 2001;165:21032109 16. Bennett NE, Kim JH, Wolfe DP, Sasaki K, Yoshimura N, Goins WF, Huang S, Nelson JB, de Groat WC, Glorioso JC, Chancellor MB. Improvement in erectile dysfunction after neurotrophic factor gene therapy in diabetic rats. J Urol 2005;173:18201824 17. Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human to treatments. Nat Med 2013;19:179192 18. Kawano Y, Kypta R. Secreted antagonists of the Wnt signalling pathway. J Cell Sci 2003;116:26272634 19. Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A, Niehrs C. LDLreceptor related protein 6 is a receptor for Dickkopf proteins. Nature 2001;411:321325 20. Min JK, Park H, Choi HJ, Kim Y, Pyun BJ, Agrawal V, Song BW, Jeon J, Maeng YS, Rho SS, Shim S, Chai JH, Koo BK, Hong HJ, Yun CO, Choi C, Kim YM, Hwang KC, Kwon YG. The WNT antagonist Dickkopf2 promotes angiogenesis in and human endothelial cells. J Clin Invest 2011;121:18821893 21. Jin HR, Kim WJ, Song JS, Choi MJ, Piao S, Shin SH, Tumurbaatar M, Tuvshintur B, Nam MS, Ryu JK, Suh JK. Functional and morphologic Page 27 of 84 Diabetes

characterizations of the diabetic mouse corpus cavernosum: comparison of a multiple lowdose and a single highdose streptozotocin protocols. J Sex Med 2009;6:32893304 22. Yin GN, Choi MJ, Kim WJ, Kwon MH, Song KM, Park JM, Das ND, Kwon KD, Batbold D, Oh GT, Koh GY, Kim KW, Ryu JK, Suh JK. Inhibition of Ninjurin 1 restores erectile function through dual angiogenic and neurotrophic effects in the diabetic mouse. Proc Natl Acad Sci U S A 2014;111:E27312740 23. Yashida MH, Da Silva Faria AL, Caldeira EJ. Estrogen and insulin replacement therapy modulates the expression of insulinlike growth factorI receptors in the salivary glands of diabetic mice. Anat Rec 2011;294:1930 1938 24. Baba H, Kurano M, Nishida T, Hatta H, Hokao R, Tsuneyama K. Facilitatory effect of insulin treatment on hepatocellular carcinoma development in diabetes. BMC Res Notes 2017;10:478 25. Neng L, Zhang W, Hassan A, Zemla M, Kachelmeier A, Fridberger A, Auer M, Shi X. Isolation and culture of endothelial cells, pericytes and perivascular resident macrophagelike melanocytes from the young mouse ear. Nat Protoc 2013;8:709720 26. Yin GN, Ryu JK, Kwon MH, Shin SH, Jin HR, Song KM, Choi MJ, Kang DY, Kim WJ, Suh JK. Matrigelbased sprouting endothelial cell culture system from mouse corpus cavernosum is potentially useful for the study of endothelial and erectile dysfunction related to highglucose exposure. J Sex Med 2012;9:17601772 27. Lin G, Chen KC, Hsieh PS, Yeh CH, Lue TF, Lin CS. Neurotrophic effects of vascular endothelial growth factor and neurotrophins on cultured major pelvic ganglia. BJU Int 2003;92:631635 28. Burchardt T, Burchardt M, Karden J, Buttyan R, Shabsigh A, de la Taille A, Ng PY, Anastasiadis AG, Shabsigh R. Reduction of endothelial and smooth muscle density in the corpora cavernosa of the streptozotocin induced diabetic rat. J Urol 2000;164:18071811 29. Zhang LW, Piao S, Choi MJ, Shin HY, Jin HR, Kim WJ, Song SU, Han JY, Park SH, Mamura M, Kim SJ, Ryu JK, Suh JK. Role of increased penile expression of transforming growth factorbeta1 and activation of the Smad signaling pathway in erectile dysfunction in streptozotocininduced diabetic rats. J Sex Med 2008;5:23182329 Diabetes Page 28 of 84

30. Liebner S, Cavallaro U, Dejana E. The multiple languages of endothelial cell tocell communication. Arterioscler Thromb Vasc Biol 2006;26:14311438 31. Artwohl M, Graier WF, Roden M, Bischof M, Freudenthaler A, Waldhausl W, BaumgartnerParzer SM. Diabetic LDL triggers apoptosis in vascular endothelial cells. Diabetes 2003;52:12401247 32. Ryu JK, Jin HR, Yin GN, Kwon MH, Song KM, Choi MJ, Park JM, Das ND, Kwon KD, Batbold D, Lee T, Gao ZL, Kim KW, Kim WJ, Suh JK. Erectile dysfunction precedes other systemic vascular diseases due to incompetent cavernous endothelial cellcell junctions. J Urol 2013;190:779789 33. Armulik A, Genove G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 2011;21:193 215 34. Hurt KJ, Musicki B, Palese MA, Crone JK, Becker RE, Moriarity JL, Snyder SH, Burnett AL. Aktdependent phosphorylation of endothelial nitricoxide synthase mediates penile erection. Proc Natl Acad Sci U S A 2002;99:4061 4066 35. Gao K, Wang YS, Yuan YJ, Wan ZH, Yao TC, Li HH, Tang PF, Mei XF. Neuroprotective effect of rapamycin on spinal cord injury via activation of the Wnt/beta signaling pathway. Neural Regen Res 2015;10:951957 36. Harvey K, Marchetti B. Regulating Wnt signaling: a strategy to prevent neurodegeneration and induce regeneration. J Mol Cell Biol 2014;6:12 37. Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC, Yancopoulos GD. Isolation of angiopoietin1, a ligand for the TIE2 receptor, by secretiontrap expression cloning. Cell 1996;87:11611169 38. Kosacka J, Figiel M, Engele J, Hilbig H, Majewski M, SpanelBorowski K. Angiopoietin1 promotes neurite outgrowth from dorsal root ganglion cells positive for Tie2 receptor. Cell Tissue Res 2005;320:1119 39. Park H, Jung HY, Choi HJ, Kim DY, Yoo JY, Yun CO, Min JK, Kim YM, Kwon YG. Distinct roles of DKK1 and DKK2 in tumor angiogenesis. Angiogenesis 2014;17:221234

Page 29 of 84 Diabetes

Figure legends

Figure 1. Decreased DKK2 expression under diabetic conditions. A and B:

Representative RTPCR and Western blots for DKK2 in mouse cavernous endothelial

cells (MCECs) and mouse cavernous pericytes (MCPs). C and D: Normalized band

intensity values (n = 4). *P < 0.05 vs. MCEC group. The relative ratio of the MCEC

group was arbitrarily set to 1. E and F: VWF (green)/CD31(green) and DKK2 (red) or

PDGFRβ (green) and DKK2 (red) staining in normal human or mouse cavernous tissue.

Nuclei were labeled with DAPI (blue). Scale bar = 100 µm. G and H: The DKK2

immunopositive area in cavernous endothelial cells and pericytes was quantified by

Image J. n = 1 human sample; n = 6 mouse samples. In human tissue, images were

obtained for four different regions. *P < 0.01 vs. VWF or CD31 group. I-K:

Representative Western blots for DKK2 in agematched control and diabetic mouse

penis, and in MCECs and MCPs exposed to normal glucose (NG) or high glucose (HG)

conditions for 48 hours. L-N: Normalized band intensity values (n = 4). *P< 0.05 vs.

Control or NG group. O: DKK2 (red) staining in cavernous tissue from a patient with

diabetic erectile dysfunction or diabetic mice and agematched controls. Scale bar =

100 µm. P and Q: The DKK2immunopositive area was quantified by Image J. n = 1

human sample; n = 4 mouse samples. In human tissues, images were obtained for four

different regions per group. *P < 0.01 vs. control group. The Pvalues were determined

by MannWhitney U test. Data in graphs are presented as mean ± SE. The relative ratio

in the control or NG groups was arbitrarily set to 1. DM, diabetes mellitus; PDGFRβ,

plateletderived growth factor receptorβ; VWF, von Willebrand factor.

Diabetes Page 30 of 84

Figure 2. DKK2 overexpression is resistant to diabetesinduced angiopathy. A: CD31

(green) and PDGFRβ (red) staining in cavernous tissue from wildtype (WT) and

DKK2Tg mice, WT mice receiving STZ, and DKK2Tg mice receiving STZ. Scale bar = 100 µm. B: CD31 (red) and BrdU (green, arrow head) staining in penis tissue from each group. Nuclei were labeled with DAPI (blue). Scale bars = 50 µm (merged) and 25 µm (magnification). C: Tube formation assay in mouse cavernous endothelial cell (MCEC) or mouse cavernous pericyte (MCP) monoculture and MCECMCP mixed coculture exposed to normal glucose (NG) or high glucose (HG) conditions for

48 hours and treated with PBS or DKK2 protein (200 ng/ml). 40× magnification. D:

Scratch woundhealing assay in MCECs 24 hours after treatment. 40× magnification. E and F: Ex vivo aortic ring assay. The images were taken 5 days after treatment. 40× magnification. (F) DKK2 (red) staining in aortic ring. Nuclei were labeled with DAPI

(blue). The sprouting front and aortic tissue are demarcated by the white dashed line.

100× magnification. Results were similar in three independent experiments. G:

Quantification of cavernous endothelial cell and pericyte content by Image J (n = 6).

*P < 0.001 vs. WT and DKK2 Tg groups. #P < 0.001 vs. WT + STZ group. The relative ratio of the WT or DKK2Tg group was arbitrarily set to 1. H: Number of

BrdUpositive endothelial cells per highpower field (n = 6). *P < 0.05 vs. WT and

DKK2Tg groups. #P < 0.01 vs. WT + STZ group. I: Number of branch points per highpower field (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBStreated group.

J: Number of migrated endothelial cells (n = 6). *P < 0.001 vs. NG group. #P < 0.001 vs. PBStreated group. K: Area of outgrowing microvessels from aortic ring (n = 6). *P

< 0.05 vs. NG group. #P < 0.05 vs. PBStreated group. Pvalues were determined by

KruskalWallis test. Data in graphs are presented as mean ± SE. PDGFRβ, platelet Page 31 of 84 Diabetes

derived growth factor receptorβ; STZ, streptozotocin.

Figure 3. Pericytederived DKK2 migrates into endothelial cells and regulates

angiogenesis. A: DKK2 (red) staining in mouse cavernous endothelial cell (MCEC)

and mouse cavernous pericyte (MCP) monoculture and indirect noncontact MCEC

MCP coculture. Nuclei were labeled with DAPI (blue). Note the increased DKK2

expression in MCECs after coculture. Scale bar = 50 µm. B: MCPs were transfected

with DKK2RFP DNA then cocultured with MCECs. Note the DKK2RFP expression

in MCECs after coculture. Images are representative of four independent experiments.

Scale bar = 100 µm. C and D: Tube formation assay in MCECs exposed to MCP

complement medium (control) or MCPconditioned culture medium (CM) with or

without deletion of DKK2 with neutralizing antibody. 40× magnification. Number of

tubes per highpower field (n = 4). *P < 0.05 vs. control group. #P < 0.05 vs. untreated

CM group. E and F: Tube formation assay in MCPs exposed to MCEC complement

medium (control) or MCECconditioned CM with or without deletion of DKK2 with

neutralizing antibody. 40× magnification. Number of tubes per highpower field (n =

4). *P < 0.001 vs. control group. Pvalues were determined by KruskalWallis test.

Data in graphs are presented as mean ± SE. RFP, red fluorescent protein.

Figure 4. DKK2 overexpression is resistant to diabetesinduced neuropathy. A:

Neurofilament (NF; red), neuronal nitric oxide synthase (nNOS; red in dorsal nerve

and green in cavernosum), and βIII tubulin (red) staining in penis tissue from wildtype

(WT) and DKK2Tg mice, WT mice receiving streptozotocin (STZ), and DKK2Tg

mice receiving STZ. Nuclei were labeled with DAPI (blue). Scale bars = 25 m (dorsal Diabetes Page 32 of 84

nerve bundle) or 50 m (cavernosum). B: βIII tubulin (red) staining in mouse pelvic ganglion (MPG) tissue culture exposed to normal glucose (NG) or high glucose (HG) conditions for 72 hours and treated with PBS or DKK2 protein (200 ng/ml). Scale bar

= 200 µm. C: Representative Western blot for neurotrophic factors (nerve growth factor [NGF], brainderived neurotrophic factor [BDNF], and neurotrophin3 [NG3]) and receptor kinase A (TrkA) in penis tissue from agematched control or diabetic mice 2 weeks after repeated intracavernous injections of PBS (20 µl) or DKK2 protein (days 3 and 0; 6 µg/20 µl), and in Neuro2A cells exposed to NG or HG conditions for 48 hours and treated with PBS or DKK2 protein (200 ng/ml). D-F: The

NF, nNOS, and βIII tubulin immunopositive areas quantified in dorsal nerve bundle or cavernous tissue by Image J (n = 6). *P < 0.05 vs. WT and DKK2Tg groups. #P <

0.05 vs. WT + STZ group. G: Quantification of neurite length by Image J (n = 4). *P <

0.001 vs. NG group. #P < 0.001 vs. PBStreated group. H-K: Normalized band intensity values (n = 4). *P < 0.05 vs. control (C) and NG groups. #P < 0.05 vs. PBS treated groups. Pvalues were determined by KruskalWallis test. Data in graphs are presented as mean ± SE. The relative ratio in the WT, C, or NG group was arbitrarily set to 1.

Figure 5. DKK2mediated recovery of erectile function is dependent on the Ang1Tie2 signaling pathway. A: Microarray analysis using total RNA from mouse cavernous endothelial cells (MCECs) exposed to normal glucose (NG) or high glucose (HG) conditions for 48 hours and treated with PBS or DKK2 protein (200 ng/ml). B:

Representative RTPCR and Western blot for Ang1 and Ang2 in MCECs and mouse cavernous pericytes (MCPs) exposed to NG or HG conditions for 48 hours and treated Page 33 of 84 Diabetes

with PBS or DKK2 protein (200 ng/ml). C and D: Normalized band intensity values (n

= 4). *P < 0.05 vs. NG group. #P < 0.05 vs. PBStreated group. The relative ratio in the

NG group was arbitrarily set to 1. E-G: Representative intracavernous pressure (ICP)

responses for the DKK2Tg mice, wildtype (WT) mice receiving streptozotocin (STZ),

DKK2Tg mice receiving STZ and dimericFc (4 µg), and DKK2Tg mice receiving

STZ and soluble Tie2 antibody (sTie2; 4 µg) (n = 6). *P < 0.001 vs. DKK2Tg group.

#P < 0.001 vs. WT + STZ group. †P < 0.001 vs. DKK2Tg + STZ + Fc group. H-J:

Representative ICP responses for agematched control (C) or diabetic mice stimulated

2 weeks after repeated intracavernous injections of PBS (20 µl), DKK2 protein (days

3 and 0; 6 µg/20 µl) and dimericFc, or DKK2 protein and sTie2 (n = 5). *P < 0.001 vs.

control group. #P < 0.001 vs. PBStreated group. †P < 0.001 vs. DKK2 + Fc group. K:

CD31 (green), PDGFRβ (red), and βIII tubulin (red) staining in penis tissue from each

group (n = 6). Scale bar = 100 µm (top), 25 µm (middle), and 50 µm (bottom). L: Tube

formation assay in MCECs exposed to NG or HG conditions for 48 hours and treated

with PBS, DKK2 protein (200 ng/ml), or DKK2 protein + sTie2 (100 ng/ml). 40×

magnification. M: βIII tubulin (red) staining in mouse pelvic ganglion (MPG) tissue

culture. N and O: The CD31, PDGFRβ, and βIII tubulin immunopositive areas

quantified by Image J (n = 6). *P < 0.05 vs. DKK2Tg group. #P < 0.05 vs. WT + STZ

group. †P < 0.01 vs. DKK2Tg + STZ + Fc group. The relative ratio in the DKK2Tg

group was arbitrarily set to 1. P: Number of tubes per highpower field (n = 4). *P <

0.001 vs. NG group. #P < 0.01 vs. PBStreated group. †P < 0.01 vs. DKK2 + Fc group.

Q: Quantification of neurite length by Image J (n = 4). *P < 0.001 vs. NG group. #P <

0.001 vs. PBStreated group. †P < 0.001 vs. DKK2 + Fc group. The relative ratio in the

NG group was arbitrarily set to 1. Pvalues were determined by KruskalWallis test. Diabetes Page 34 of 84

Data in graphs are presented as mean ± SE. PDGFRβ, plateletderived growth factor receptorβ.

Figure 6. Schematic diagram of a proposed mechanism in which DKK2 preserves erectile function in diabetic mice. Pericytederived DKK2 migrates into endothelial cells (ECs). The DKK2mediated interaction between pericytes and endothelial cells then promotes angiogenesis and neural regeneration through an Ang1Tie2 pathway, rescuing erectile function in diabetic condition. Ang, angiopoietin; BDNF, brain derived neurotrophic factor; ECJ, endothelial cellcell junction; eNOS, endothelial nitric oxide synthase; MCECs, mouse cavernous endothelial cells; MCPs, mouse cavernous pericytes; NGF, nerve growth factor; nNOS, neuronal nitric oxide synthase;

NT3, neurotrophin3; PC, pericyte; sTie2 = soluble Tie2 protein. Pagea 35 of 84 b Diabetesi j k MCECs MCPs MCECs MCPs Control DM NG HG NG HG DKK2 DKK2 DKK2 Penis WB MCPs  -  MCECs RT-PCR GAPDH -actin

l 1.5 m 1.5 n 1.5 c 2.5 d 4 * * 2.0 1.0 1.0 1.0

3 -actin) -actin) -actin)   

1.5 -actin)

 2 * 1.0 0.5 * 0.5 * 0.5 Relative ratio ratio Relative 1 ratio Relative Relative ratio ratio Relative (DKK2 (DKK2 / 0.5 (DKK2 / (DKK2 (DKK2 / Relative ratio ratio Relative Relative ratio ratio Relative 0.0 0.0 0.0 (DKK2 (DKK2 / GAPDH) 0.0 (DKK2 / 0 C DM NG HG NG HG MCECs MCPs MCECs MCPs Mouse MCECs MCPs o DKK2 DAPI Merged Magnification e VWF / Control Human Human DKK2 PDGFRβ DM / DKK2 f CD31 / Control Mouse Mouse DKK2 DM PDGFRβ / DKK2 g h p q 70 60 1.5 1.5 60 * 50 * 50 40 1.0 1.0 40 30 30 20 0.5 0.5 20 * /Cavernosum /Cavernosum DKK2 DKK2 (+) area 10 DKK2 (+) area

10 (%) cavernosum

cavernosum (%) cavernosum *

Merged area/Mouse area/Mouse Merged 0 0.0 0.0 Merged area/Human area/Human Merged 0 VWF PDGFRβ CD31 PDGFRβ C DM C DM Human Mouse a g b Endothelium or Pericyte Magnification Merged BrdU CD31 Merged PDGFRβ CD31 area/Cavernosum 0.0 0.5 1.0 1.5 TWT WT WT WT PDGFRβ CD31 DKK2 -Tg * * STZ DKK2 # -Tg DKK2-Tg # DKK2-Tg h No.BrdU (+) endothelial

cells/HPF 10 0 2 4 6 8 TWT WT DKK2 -Tg WT+STZ DKK2-Tg+ STZ DKK2-Tg+ WT+STZ WTSTZ + * STZ DKK2 -Tg # i

Branch points/Field STZ + DKK2-Tg 100 20 40 60 80 0 MCECs-MCPs MCPs MCECs GPSDKK2 PBS NG Diabetes * * c * e f d HG Aortic ring Aortic ring MCEC-MCP mix

# assay assay MCECs ed co-culture MCPs MCECs Sprouting Sprouting # front # DKK2 NG NG NG

Number of j migrated cells 100 150 200

50 Aortic 0 ring GPSDKK2 PBS NG * HG + PBS HG + DKK2 + HG PBS + HG HG + PBS HG + DKK2 + HG PBS + HG HG+PBS HG DAPI # k Vascular area (%) 0.5 1.0 1.5 2.0 2.5 0 GPSDKK2 PBS NG HG+DKK2 Merged * HG 100x # 40x 40x 40x Page 36of84 Page 37of84 b a e c MCPs MCECs DKK2/DAPI MCPs Lipofectamine2000 MCEC complementMCEC MCPcomplement MCECs MCECs MCPs medium medium DKK2-RFPtransfection Mono-culture MCPs MCP-conditionedculture MCEC-conditioned DKK2-RFP culturemedium MCPs MCPs medium MCPs Diabetes MCECs DKK2 (-) MCEC-conditioned(-) DKK2 Lipofectamine2000 DKK2 (-) MCP-conditioned(-) DKK2 Co-culturewith MCPs for 24 hours culturemedium MCECs culturemedium MCECs MCPs Co-culture MCECs MCECs f d

Branch points/Field Branch points/Field DKK2-RFP 100 120 100 120 20 40 60 80 20 40 60 80 MCECs MCPs 0 MCPs 0 MCPs C NT DKK2 (-) DKK2 NT C C NT DKK2 (-) DKK2 NT C * * CM CM # a c Cavernosum Cavernosum Dorsal nerve Dorsal nerve Dorsal nerve βIII tubulin nNOS/DAPI βIII tubulin nNOS/DAPI NF/DAPI oto PSDKK2 PBS Control WT Cavernosum STZ K2T T+SZDKK2-TgSTZ + WTSTZ + DKK2-Tg GPSDKK2 PBS NG N2Acell HG -actin TrkA NT-3 BDNF NGF Diabetes i f Relative ratio βIII tubulin (+) area/Dorsal d

(BDNF/-actin) NF (+) area/Dorsal nerve b 0.0 0.5 1.0 1.5 0.0 0.5 1.0 1.5 2.0 nerve or Cavernosum 0.0 0.5 1.0 1.5 2.0 2.5 Merged Phase image βIII tubulin G P M /GPSDKK2 PBS C/NG TWT WT WT N2A cell N2A Cavernosum Cavernosum DNB DKK2 -Tg * DKK2 NG -Tg STZ/HG * * STZ # * # DKK2 WT -Tg # * STZ DKK2 # -Tg G P M j

Relative ratio # (NT-3/-actin) 0.0 0.5 1.0 1.5 2.0 G+PSHGDKK2 + HGPBS + g nNOS (+) area/Dorsal e Neurite length 0.0 0.5 1.0 1.5 nerve or Cavernosum /GPSDKK2 PBS C/NG 0.0 0.5 1.0 1.5 2.0 N2A cell N2A Cavernosum GPBS NG * WT Cavernosum DNB STZ/HG * * HG DKK2 DKK2 -Tg # G P M h * k Relative ratio Relative ratio WT

(TrkA/-actin) (NGF/-actin) * 0.0 0.5 1.0 1.5 2.0 0.0 0.5 1.0 1.5 2.0 STZ DKK2 # -Tg /GPSDKK2 PBS C/NG /GPBS C/NG N2A cell N2A Cavernosum N2A cell N2A Cavernosum # * * Page 38of84 STZ/HG STZ/HG * * DKK2 # # # # Page 39 of 84 Diabetes Diabetes Page 40 of 84

MCPs Ang1 DKK2 Cavernous nerve Ang2 DKK2

MCECs Ang1 DKK2 Ang2

Neurotrophic factors (NGF, BDNF, NT-3) EC regeneration sTie2 PC regeneration • Proliferation • Proliferation Neural regeneration sTie2 • Migration • Migration

eNOS activation Penile erection sTie2 Vessel ECJ proteins permeability MCPs-DKK2 Ang1 Page 41 of 84 Diabetes

Supplemental Materials, Tables, and Figures

Pericyte-derived Dickkopf2 regenerates damaged penile neurovasculature through an angiopoietin-1-Tie2 pathway

Guo Nan Yin1,*, HaiRong Jin1,2,*, MinJi Choi1, Anita Limanjaya1, Kalyan Ghatak1, Nguyen Nhat Minh1, Jiyeon Ock1, MiHye Kwon1, KangMoon Song1, Heon Joo 3 4 5 1,6, † 1, † Park , Ho Min Kim , YoungGuen Kwon , JiKan Ryu , JunKyu Suh

1National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Republic of Korea; 2Department of Urology, Yuhuangding Hospital, Yantai 264000, Shandong Province, China; 3Hypoxiarelated Disease Research Center, Inha University College of Medicine, Incheon 22212, Republic of Korea; 4Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; 5Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; 6Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon 22212, Republic of Korea

*These authors contributed equally to this work.

†Correspondence and requests for materials should be addressed to J.K.S. or J.K.R. (email: [email protected] or [email protected])

JunKyu Suh, MD, PhD National Research Center for Sexual Medicine and Department of Urology Inha University College of Medicine 7206, 3rd St, ShinheungDong, JungGu Incheon 22332, Republic of Korea Tel: 82328903441, Fax: 82328903097 Diabetes Page 42 of 84

Email: [email protected]

JiKan Ryu, MD, PhD National Research Center for Sexual Medicine and Department of Urology Inha University College of Medicine 7206, 3rd ST, ShinheungDong, JungGu, Incheon 22332 Republic of Korea Tel: 82328903505; Fax: 82328903099 Email: [email protected] Page 43 of 84 Diabetes

Supplemental Materials

Measurement of erectile function

Bipolar platinum wire electrodes were placed around the cavernous nerve.

Stimulation parameters were 5 V at a frequency of 12 Hz, a pulse width of 1 ms, and

duration of 1 minute. The maximal intracavernous pressure (ICP) was recorded

during tumescence. The total ICP was determined by the area under the curve from

the beginning of cavernous nerve stimulation to a point 20 s after stimulus

termination. Two electrostimulations were replicated at intervals of 10 minutes.

Systemic blood pressure was measured using a noninvasive tailcuff system

(Visitech systems). The validity of this system was demonstrated previously (1).

Systemic blood pressure was measured prior to the measurement of ICP, because

vibration during electrical stimulation the cavernous nerve did not allow accurate

assessment of blood pressure. The ratio of maximal ICP and total ICP to mean

systolic blood pressure was calculated to adjust for variations in systemic blood

pressure.

Cell culture

Penis tissue was harvested and transferred into sterile vials containing Hank’s

balanced salt solution (Gibco) and washed twice in PBS. The glans penis, urethra,

and dorsal neurovascular bundle were removed from the penis, and only the corpus

cavernosum tissue was used for primary cell culture. For primary culture of mouse

cavernous endothelial cells (MCEC), the corpus cavernosum tissue was cut into two

or three pieces and the samples plated on matrigelcoated (Becton Dickinson) 60 Diabetes Page 44 of 84

mm cell culture dishes. The matrigel was polymerized with a 5minute incubation period at 37°C; 3 ml of complement medium 199 (Gibco) supplemented with 20% fetal bovine serum (FBS), 1% penicillin/streptomycin, 0.5 mg/ml heparin (Sigma

Aldrich), and 5 ng/ml vascular endothelial growth factor (VEGF, R&D Systems) was added to the cell culture dish. The dishes were incubated at 37°C in a 5% CO2 atmosphere. After the cells were confluent and spread over the bottom of the dish

(~2 to 3 weeks after the start of culture), only sprouting cells were used for subcultivation. The sprouting cells were seeded onto dishes coated with 0.2% gelatin

(SigmaAldrich).

For primary culture of mouse cavernous pericytes (MCP), the corpus cavernosum tissue was cut into several 1mm pieces and the fragmented pieces settled by gravity into collagen Icoated 35mm cell culture dishes (BD Biosciences).

After 30 minutes of incubation at 37°C with 300 µl complement Dulbecco’s modified Eagle Medium (DMEM; Gibco) supplemented with 10% FBS, 1% penicillin/streptomycin, and 10 nM human pigment epitheliumderived factor

(PEDF; SigmaAldrich), we added an additional 900 µl complement medium and incubated the samples at 37°C in a 5% CO2 atmosphere. The medium was changed every 2 days. After the cells were confluent and spread over the bottom of the dish

(~2 weeks after the start of culture), only sprouting cells were used for subcultivation. The sprouting cells were seeded onto dishes coated with 50 µl/ml collagen I (Advanced BioMatrix). Cells between passages 2 and 3 were used for experiments.

Human umbilical vein endothelial cells (HUVEC) were cultured in complement medium 199 (Gibco) supplemented with 20% FBS, 1% Page 45 of 84 Diabetes

penicillin/streptomycin, 0.5 mg/ml heparin (SigmaAldrich), and 5 ng/ml VEGF

(R&D Systems). Human brain microvascular pericytes (HBMPs) and Neuro2A cells

were cultured in DMEM supplemented with 10% FBS and 1%

penicillin/streptomycin.

Establishment of MCEC-MCP co-culture system

We established indirect noncontact and direct mixed coculture systems to examine

the interaction between endothelial cells and pericytes. For the indirect noncontact

coculture system, the inside of the insert (1.0 µm pore size Transwells; Becton

Dickinson) was coated with 0.2% gelatin and the bottom of the culture plate with 50

µl collagen I. MCECs were then plated on the inside of the insert and MCPs on the

bottom of the culture plate until cells were confluent. To determine the optimal ratio

of MCECs and MCPs for direct mixed coculture, the cells were cultivated at

different ratios (MCEC:MCP = 10:1, 5:1, 3:1, 2:1, 1:1) in 50% DMEM and 50%

M199 complement medium.

Tube formation assay

Approximately 50 µl of growth factorreduced matrigel (Collaborative Biomedical

Products) was dispensed into 96well tissue culture plates at 4°C. After gelling at

37°C for at least 30 minutes, the MCECs or MCPs were seeded onto the gel at 2 ×

104 cells/well in 200 µl of M199 or DMEM medium. The assay was performed in a

CO2 incubator and the plates incubated at 37°C for 24 hours. Images were obtained

with a phasecontrast microscope and the number of tubes in each well of the plate

counted at a screen magnification of ×40. Only integrated tubes were counted. Diabetes Page 46 of 84

Scratch wound-healing assay

MCECs, MCPs, HVECs, or HBMPs were plated on 60mm culture dishes at 95% confluence, wounded with a 2mmwide razor blade, and marked at the injury line.

After wounding, the cultures were washed with serumfree medium and further incubated in M199 media or DMEM with 2% FBS for 24 hours. Phasecontrast images were taken 24 hours after scratching. Migration was quantitated by counting the number of cells that moved beyond the reference line.

DNA transfection

The mouse DKK2 (NM_020265.3) ORF mammalian expression plasmid (C

OFPSpark/RFP tag cDNA) was purchased from Sino Biological. Inc. (SB, Beijing,

China). Transient transfection was performed using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. MCPs were transfected with 1 µg of

DKK2RFP at a density of 5 × 105 cells/wells.

RNA interference

MCECs or MCPs at a density of 2 × 106 cells/well were transfected with siRNA against Ang1 (200 pmol; Santa Cruz Biotechnology) using Lipofectamine 2000

(Invitrogen).

RT-PCR

Total RNA was extracted from cultured cells using Trizol (Thermo Fisher Scientific) following the manufacturer’s protocols. Reverse transcription was performed using 1 Page 47 of 84 Diabetes

µg of RNA in 20 µl of reaction buffer with oligo dT primer and AccuPower RT

Premix (Bioneer). We used the following primers to measure relative changes in

mRNA levels: mouse Dkk2 forward, 5′TGA TGG TGG AGA GCT CAC AG3′;

mouse Dkk2 reverse, 5′GTA GGC ATG GGT CTC CTT CA3′; mouse Ang1 forward,

5′AAA CAG CAA ATG GGA ACA GG3′; mouse Ang1 reverse, 5′GGG CAG GTG

AAC TCC ACT AA3′; mouse Ang2 forward, 5′CAA GGC ACT GAG AGA CAC

CA3′; mouse Ang2 reverse, 5′CTG AAC TCC CAC GGA ACA TT3′; human

DKK2 forward, 5′TAG AGA TTG AGT TTG AGC CT3′; human DKK2 reverse, 5′

AAA GGG TGG ACA TAA GAA A3′; GAPDH forward, 5′CCA CTG GCG TCT

TCA CCA C3′; GAPDH reverse, 5′CCT GCT TCA CCA CCT TCT TG3′. The

PCR reaction was performed with denaturation at 94°C for 30 s, annealing at 60°C

for 30 s, and extension at 72°C for 1 minute in a DNA Engine Tetrad Peltier Thermal

Cycler (MJ Research). For the analysis of PCR products, 10 µl of each PCR reaction

was electrophoresed on 1% agarose gel and detected under ultraviolet . GAPDH

was used as an internal control.

cDNA microarray

For the microarray study, the MCECs were cultured and treated (n = 3 per group)

with normalglucose (NG, 5 mmol), highglucose (HG, 30 mmol) and PBS, or HG

(30 mmol) and DKK2 protein (200 ng/ml). The cDNA microarray analysis was

performed 48 hours after treatment. For the RNAs isolated from the three groups, the

synthesis of target cRNA probes and hybridization were performed using Agilent’s

Low RNA Input Linear Amplification kit (Agilent Technology) according to the

manufacturer’s instructions. The hybridized images were scanned using an Agilent Diabetes Page 48 of 84

DNA microarray scanner and quantified by Feature Extraction Software (Agilent

Technology). All data normalization and gene selection were performed using

GeneSpringGX 7.3 (Agilent Technology). The average normalized ratios were calculated by dividing the average normalized signal channel intensity by the average normalized control channel intensity. Functional gene annotation was performed according to the Gene OntologyTM Consortium

(http://www.geneontology.org/index.shtml) using GeneSpringGX 7.3. gene classification based on searches performed by BioCarta (http://www.biocarta.com/),

GenMAPP (http://www.genmapp.org/), DAVID (http://david.abcc.ncifcrf.gov/), and

Medline databases (http://www.ncbi.nlm.nih.gov/). The cDNA microarray data have been deposited in the Omnibus database

(www.ncbi.nlm.nih.gov/geo accession no. GSE95178).

Histological examinations

The penis tissue and cultured major pelvic ganglion or aortic ring were fixed in 4% paraformaldehyde for 24 hours at 4°C and frozen tissue sections (8m [thincut] or 60

m [thickcut]) incubated with antibodies to DKK2 (1:100; Abcam, Cat. ab95274),

DKK2 (1:50; Santa Cruz Biotechnology, Cat. sc25517), CD31 (1:50; Millipore, Cat.

MAB1398Z), von Willebrand factor (VWF; 1:50; Santa Cruz Biotechnology, Cat. sc

59810), fluorescein isothiocyanate (FITC)conjugated antibody to smooth muscle α actin (1:200; SigmaAldrich, Cat. F3777), PDGFRβ (1:50; Santa Cruz Biotechnology,

Cat. sc1627), neurofilament (1:100; SigmaAldrich, Cat. N5389), nNOS (1:100;

Santa Cruz Biotechnology, Cat. sc5302), βIII tubulin (1:200; Abcam, Cat. ab107216),

5′bromo2′deoxyuridine (BrdU, 1:50; AbD Serotec, Cat. MCA2060), phosphoeNOS Page 49 of 84 Diabetes

(1:100; Cell Signaling, Cat. 9571), claudin5 (1:100; Thermo Fisher Scientific, Cat.

352500), VEcadherin (1:100; Millipore, Cat. AB1955), nitrotyrosine (1:50; Millipore,

Cat. 06284), or oxidized LDL (1:400; Abcam, Cat. ab14519) at 4°C overnight.

Control sections were incubated without the primary antibody at this step. After

several washes with PBS, the sections were incubated with tetramethyl rhodamine

isothiocyanate (TRITC)conjugated (Jackson ImmunoResearch, Inc., Cat. 127025

160 for Hamster; Cat. 711026152 for Rabbit; Cat. 315025003 for Mouse; Cat.

705025147 for Goat; Cat. 703025155 for Chicken.) or FITCconjugated

secondary antibodies (Jackson ImmunoResearch, Inc., Cat. 127095160 for

Hamster; Cat. 711096152 for Rabbit; Cat. 715545150 for Mouse; Cat. 705545

147 for Goat) or DyLight TM 405 (Jackson ImmunoResearch, Inc., Cat. 127475

160) for 2 hours at room temperature. Signals were visualized and digital images

obtained with a confocal microscope (FV1000, Olympus). For BrdU labeling

experiments, an additional antigen retrieval step was performed. Briefly, following

fixation, the sections were washed with PBS three times and incubated for 10

minutes in 1M HCl on ice and for 20 minutes in 2M HCl at 37°C to allow DNA

denaturation. The sections were then washed three times (5 minutes each) in 0.1M

sodium borate buffer (Na2B4O7). Histological examinations were quantitated using

an image analyzer system (National Institutes of Health [NIH] Image J 1.34,

http://rsbweb.nih.gov/ij/).

In situ detection of superoxide anion

Hydroethidine (1:5000; Molecular Probes, Cat. D23107), an oxidative fluorescent

dye, was used to evaluate levels of superoxide anion in situ as previously described Diabetes Page 50 of 84

(2, 3). After immunofluorescent staining with antibody to CD31 and FITC conjugated secondary antibodies, hydroethidine (2 µmol/l) was applied to each tissue section and the samples were coverslipped. The slides were incubated in a light protected humidified chamber at 37°C for 30 minutes. The number of ethidium bromide fluorescencepositive endothelial cells was counted at a screen magnification of ×400 in 6 or 8 different regions. Values were expressed per high power field.

BrdU labeling

The mice from each group were given intraperitoneal injections of BrdU (50 mg/kg of body weight; SigmaAldrich) once a day for three consecutive days and sacrificed

1 day after BrdU injection. The MCECs, MCPs, HUVECs, or HBMPs were incorporated with BrdU at a final concentration of 10 µM at 37°C for 1 hour. An additional antigen retrieval step was performed as described above. The number of

BrdUpositive endothelial cells or pericytes was counted at a screen magnification of

400× in six or eight different regions. Values were expressed per highpower field.

Western blot

The collected penile tissues were immediately stored within liquid nitrogen. The tissues were then homogenized with small volume of liquid nitrogen. This experimental step was repeated several times till changing to the complete powder of tissues. And then, the RAPI buffer (100 l, SigmaAldrich) was added into the homogenates. After incubating on ice for 60 minutes, the homogenates were centrifuged at 13,000 g for 20 minutes at 4°C and the concentrations of each protein Page 51 of 84 Diabetes

in the supernatants were measured using the Universal Microplate Reader ELx800G

(BioTek Instruments Inc.). Equal amounts of protein (50 µg per lane) were

electrophoresed on 8% to 15% sodium dodecylsulfatepolyacrylamide gels and

transferred to nitrocellulose membranes. After blocking with 5% nonfat dry milk for

1 hour at room temperature, the membrane was incubated at room temperature with

antibodies to DKK2 (1:500; Abcam, Cat. ab95274), BDNF (1:500; Santa Cruz

Biotechnology, Cat. sc546), NGF (1:500; Santa Cruz Biotechnology, Cat. sc548),

NT3 (1:500; Santa Cruz Biotechnology, Cat. sc547), TrkA (1:500; Abcam, Cat.

ab76291), Ang1 (1:500; NOVUS Biologicals, Cat. NB600629), Ang2 (1:500;

NOVUS Biologicals, Cat. NB11085467), iNOS (1:1000; Becton Dickinson, Cat.

610332), Akt (1:1000; Cell Signaling, Cat. 9272), phosphoAkt (1:1000; Cell

Signaling, Cat. 9271), eNOS (1:1000; Becton Dickinson, Cat. 610297), phospho

eNOS (1:1000; Cell Signaling, Cat. 9571), or βactin (1:6000; Abcam, Cat. ab8226)

for 2 hour and washed 3 times with PBST. The signals were visualized by using an

ECL (Amersham Pharmacia Biotech.) detection system. The results were quantified

by densitometry.

Immunoprecipitation

We performed additional experiments to confirm the Western blot data. Shortly,

immunoprecipitation was performed in lysates prepared from normal mice penile

tissues (500 g of total protein) with antibody to eNOS (1:1000; Becton Dickinson,

Cat. 610297) or diabetic mice penile tissues with antibody to iNOS (1:1000; Becton

Dickinson, Cat. 610332) at 4°C overnight. Then, the proteinantibody complex was

incubated with 50 µL Protein Gcoupled Sepharose beads (Millipore) for 2 hours at Diabetes Page 52 of 84

4°C with gentle rotation. The beads were washed three times with lysis buffer. The pellet was resuspended with 30 µL lysis buffer and the sample was heated at 100°C for 10 minutes and microcentrifuged for 1 minute at 14,000 g. The sample was electrophoresed on 8% SDSPAGE gels and transferred to nitrocellulose membranes.

After blocking with 5% nonfat dry milk for 1 hour at room temperature, the membrane was incubated at room temperature with antibodies to phosphoeNOS

(1:1000; Cell Signaling, Cat. 9571), eNOS (1:1000; Becton Dickinson, Cat. 610297), or iNOS (1:1000; Becton Dickinson, Cat. 610332). The signals were visualized by using an ECL detection system (Amersham Pharmacia Biotech.). Page 53 of 84 Diabetes

Supplemental References

1. Krege JH, Hodgin JB, Hagaman JR, Smithies O. A noninvasive computerized tailcuff system for measuring blood pressure in mice. Hypertension 1995;25:11111115 2. Bivalacqua TJ, Usta MF, Kendirci M, Pradhan L, Alvarez X, Champion HC, Kadowitz PJ, Hellstrom WJ. Superoxide anion production in the rat penis impairs erectile function in diabetes: influence of in vivo extracellular superoxide dismutase gene therapy. J Sex Med 2005;2:187197 3. Jin HR, Kim WJ, Song JS, Choi MJ, Piao S, Shin SH, Tumurbaatar M, Tuvshintur B, Nam MS, Ryu JK, Suh JK. Functional and morphologic characterizations of the diabetic mouse corpus cavernosum: comparison of a multiple lowdose and a single highdose streptozotocin protocols. J Sex Med 2009;6:32893304

Diabetes Page 54 of 84

Supplementary Tables:

Supplementary Table S1. Physiological and metabolic parameters 8 weeks after induction of diabetes with streptozotocin (STZ) in wildtype (WT) and DKK2Tg mice. WT DKK2Tg WT + STZ DKK2Tg + STZ Body weight (g) 30.7±1.1 29.7±0.8 25.5±0.9* 25.4±0.5* Fasting glucose (mg/dl) 114.3±6.6 111.3±9.8 393.5±51.1* 376.3±49.0* Postprandial glucose (mg/dl) 157.3±7.1 151.5±5.5 558.8±53.4* 556.5±23.4* MSBP (mmHg) 99.3±5.2 97.1±6.2 100.2±4.8 97.2±7.1

HbA1c (%) 5.0±0.2 4.9±0.3 11.2±0.1* 9.5±1.3*

HbA1c (mmol/mol) 31 30 98* 81* Insulin (µg/l) 2.61±0.21 2.75±0.08 0.18±0.02* 0.20±0.0.03* Values are presented as the mean ± SE for n = 45 animals per group. *P < 0.01 vs. WT and DKK2Tg mice. MSBP, mean systolic blood pressure. Page 55 of 84 Diabetes

Supplementary Table S2. Physiological and metabolic parameters 2 weeks after treatment with DKK2 protein. Control DM+PBS DM+DKK2 (6 µg) Body weight (g) 32.2±2.0 23.7±2.4* 23.6±2.6* Fasting glucose (mg/dl) 105.2±5.0 545.3±36.3* 504.6±54.3* Postprandial glucose (mg/dl) 153.0±13.9 594.5±6.6* 585.8±30.6* MSBP (mmHg) 99.5±4.7 98.6±4.1 91.5±2.9 Values are presented as the mean ± SE for n = 6 animals per group. *P < 0.01 vs. Control. DM, diabetes mellitus; PBS, phosphatebuffered saline; MSBP, mean systolic blood pressure.

Diabetes Page 56 of 84

Supplementary Table S3. Physiological and metabolic parameters 8 weeks after induction of diabetes with streptozotocin (STZ) in wildtype (WT) and DKK2Tg mice.

DKK2Tg WT+STZ DKK2Tg+STZ+Fc DKK2Tg + STZ+sTie2 Body weight (g) 33.4±1.4 24.9±2.2* 24.7±1.8* 22.9±1.7* Fasting glucose (mg/dl) 104.5±6.4 483.0±52.5* 466.8±13.3* 499.5±52.6* Postprandial glucose (mg/dl) 159.2±5.3 594.0±6.1* 579.5±25.9* 585.5±25.0* MSBP (mmHg) 98.8±10.6 101.1±13.6 99.8±10.3 103.0±5.5 Values are presented as the mean ± SE for n = 5 animals per group. *P < 0.01 vs. DKK2Tg mice. Fc, dimeric Fcprotein; MSBP, mean systolic blood pressure; sTie2, soluble Tie2 antibody.

Page 57 of 84 Diabetes

Supplementary Table S4. Physiological and metabolic parameters 2 weeks after treatment with DKK2 protein or soluble Tie2 antibody (sTie2). Control DM+PBS DM+DKK2+Fc DM+DKK2+sTie2 Body weight (g) 32.4±1.9 22.2±1.2* 22.0±1.3* 22.8±0.6*

Fasting glucose (mg/dl) 104.3±5.2 557.8±17.4* 508.5±61.9* 497.5±42.0*

Postprandial glucose (mg/dl) 155.5±14.7 597.8±4.5* 582.3±34.2* 565.5±33.7* MSBP (mmHg) 100.0±5.7 98.4±3.8 100.8±6.9 99.1±1.8 Values are the mean ± SE for n = 5 animals per group. *P < 0.01 vs. Control. DM, diabete mellitus; Fc, dimeric Fc protein; MSBP, mean systolic blood pressure; PBS, phosphatebuffered saline.

Diabetes Page 58 of 84

Supplementary Table S5. Summary of selected genes decreased in the HG + PBS group compared to the NG group and restored by DKK2 treatment. HG + PBS/ HG + DKK2/ Gene Product GenBank NG* HG + PBS* Angpt1 Angiopoietin 1 0.2 2.0 NM_009640 Sh3bgrl2 SH3 domain binding rich protein like 0.4 3.9 NM_172507 Chmp4b Chromatin modifying protein 4B 0.4 2.2 NM_029362 * MCECs were exposed to normal glucose (NG) or high glucose (HG) conditions and treated with phosphatebuffered saline (PBS) or DKK2 protein. We selected genes that changed more than 2fold for both HG + PBS/NG and HG + DKK2/HG + PBS.

Page 59 of 84 Diabetes

Supplementary Table S6. Summary of selected genes that increased in the HG + PBS group compared to the NG group and restored by DKK2 treatment. HG + HG + DKK2/ Gene Product GenBank PBS/ NG* HG + PBS* Ptpn13 Protein tyrosine phosphatase, nonreceptor type 13 2.0 0.5 NM_011204 Sema7a Sema domain, immunoglobulin domain (Ig), and GPI membrane anchor 2.0 0.5 NM_011352 Lxn Latexin 2.0 0.5 NM_016753 Nhsl2 NHSlike 2 2.0 0.5 NM_001163610 Fabp4 Fatty acid binding protein 4 2.0 0.4 NM_024406 Tsr2 TSR2, 20S rRNA accumulation, homolog 2.0 0.4 NM_001164578 Odz4 Odd Oz/tenm homolog 4 2.0 0.4 NM_011858 Lhx8 LIM homeobox protein 8 2.0 0.4 NM_010713 Fam164a Family with sequence similarity 164, member A 2.0 0.4 NM_173181 Zfr2 Zinc finger RNA binding protein 2 2.0 0.5 NM_001034895 Kcnip3 Kv channel interacting protein 3, 2.0 0.5 NM_001111331 Fzd4 homolog 4 (Drosophila) 2.0 0.2 NM_008055 C1rb Complement component 1, r subcomponent B 2.0 0.3 NM_001113356 Klf15 Kruppellike factor 15 2.0 0.4 NM_023184 Arl4d ADPribosylation factorlike 4D 2.0 0.4 NM_025404 Nkd1 Naked cuticle 1 homolog (Drosophila) 2.0 0.2 NM_027280 Diabetes Page 60 of 84

Synm , protein 2.0 0.4 NM_201639 Fam149a Family with sequence similarity 149, member A 2.0 0.5 NM_153535 Zfp82 Zinc finger protein 82 2.0 0.4 NM_177889 Fign Mus musculus fidgetin (Fign), mRNA [NM_021716] 2.0 0.4 NM_021716 Rnf125 Ring finger protein 125 2.1 0.5 NM_026301 Xrcc6bp1 XRCC6 binding protein 1 2.1 0.4 NM_026858 Lpl Lipoprotein lipase 2.1 0.3 NM_008509 Entpd2 Ectonucleoside triphosphate diphosphohydrolase 2 2.1 0.4 NM_009849 Amz2 Archaelysin family metallopeptidase 2 2.1 0.5 NM_025275 Jag1 Jagged 1 2.1 0.3 NM_013822 Aspn Asporin 2.1 0.3 NM_025711 Dnahc7b , axonemal, heavy chain 7B 2.1 0.5 NM_001160386 Gm1661 Predicted gene 1661 2.1 0.2 NM_001145637 Ankrd29 repeat domain 29 2.1 0.5 NM_001190371 Susd2 Sushi domain containing 2 2.1 0.2 NM_027890 Scara5 Scavenger receptor class A, member 5 2.1 0.2 NM_028903 Plscr2 Phospholipid scramblase 2 2.1 0.3 NM_008880 Tfpi pathway inhibitor 2.1 0.4 NM_011576 Tspyl3 TSPYlike 3 2.1 0.4 NM_198617 Zmat1 Zinc finger, matrin type 1 2.1 0.4 NM_175446 Gulp1 GULP, engulfment adaptor PTB domain containing 1 2.1 0.4 NM_027506 Page 61 of 84 Diabetes

Ccdc80 Coiledcoil domain containing 80 2.1 0.5 NM_026439 Adh7 Alcohol dehydrogenase 7 (class IV), mu or sigma polypeptide 2.1 0.2 NM_009626 Pcdh10 Protocadherin 10 2.1 0.4 NM_001098172 Igdcc4 Immunoglobulin superfamily, DCC subclass, member 4 2.1 0.3 NM_020043 Reps2 RALBP1associated Eps domaincontaining protein 2 2.1 0.3 NM_178256 Phxr4 Perhexamer repeat gene 4 2.1 0.4 NM_008835 Mcc Mutated in colorectal cancers 2.1 0.3 NM_001085373 Cxcl14 Chemokine (CXC motif) ligand 14 2.1 0.3 NM_019568 Zfp467 Zinc finger protein 467 2.1 0.3 NM_001085417 Prr24 Prolinerich 24 2.1 0.5 NM_001136270 N6amt1 N6 adeninespecific DNA methyltransferase 1 2.1 0.4 NM_026366 Tfpi Tissue factor pathway inhibitor 2.1 0.5 NM_011576 Mrvi1 MRV integration site 1 2.1 0.3 NM_194464 Dpep1 Dipeptidase 1 (renal) 2.1 0.2 NM_007876 Rab3d RAB3D, member RAS oncogene family 2.1 0.4 NM_031874 Pappa Pregnancyassociated plasma protein A 2.1 0.3 NM_021362 Smad9 MAD homolog 9 (Drosophila) 2.1 0.5 NM_019483 Rab6b RAB6B, member RAS oncogene family 2.2 0.4 NM_173781 Lifr Leukemia inhibitory factor receptor 2.2 0.2 NM_013584 C1ra Complement component 1, r subcomponent A 2.2 0.2 NM_023143 H19 H19 fetal liver mRNA 2.2 0.3 NR_001592 Diabetes Page 62 of 84

BC026782 cDNA sequence BC026782 2.2 0.3 NM_001025575 Vcam1 Vascular cell adhesion molecule 1 2.2 0.5 NM_011693 Rbms3 RNA binding motif, singlestranded interacting protein 2.2 0.4 NM_001172123 Arhgef17 Rho guanine nucleotide exchange factor (GEF) 17 2.2 0.4 XM_133692 Bace2 Betasite APPcleaving enzyme 2 2.2 0.4 NM_019517 Rnd3 Rho family GTPase 3 2.2 0.4 NM_028810 Gabra1 Gammaaminobutyric acid (GABA) A receptor, subunit alpha 1 2.2 0.2 NM_010250 Sspn Sarcospan 2.2 0.4 NM_010656 Clca2 calcium activated 2 2.2 0.4 NM_030601 Ap2b1 Adaptorrelated protein complex 2, beta 1 subunit 2.2 0.5 NM_027915 Msmp Microseminoprotein, prostate associated 2.2 0.2 NM_001099314 Ccl27a Chemokine (CC motif) ligand 27A 2.2 0.5 NM_001048179 Fh1 Fumarate hydratase 1 2.2 0.5 NM_010209 Abca5 ATPbinding cassette, subfamily A (ABC1), member 5 2.2 0.5 NM_147219 Myh8 , heavy polypeptide 8, , perinatal 2.2 0.1 NM_177369 Epyc Epiphycan 2.2 0.3 NM_007884 Tnfrsf25 Tumor necrosis factor receptor superfamily, member 25 2.2 0.3 NM_033042 Kctd12 tetramerization domain containing 12 2.2 0.3 NM_177715

Kcnab1 Potassium voltagegated channel, related subfamily, beta member 1 2.2 0.5 NM_010597

Crispld2 Cysteinerich secretory protein LCCL domain containing 2 2.2 0.3 NM_030209 C1qtnf7 C1q and tumor necrosis factorrelated protein 7 2.2 0.4 NM_001135172 Page 63 of 84 Diabetes

Art3 ADPribosyltransferase 3 2.2 0.2 NM_181728 Ebf2 Early Bcell factor 2 2.2 0.2 NM_010095 Ctla2a Cytotoxic T lymphocyteassociated protein 2 alpha 2.2 0.4 NM_007796 Marcks Myristoylated alaninerich protein kinase C substrate 2.2 0.5 NM_008538 Ctps2 Cytidine 5'triphosphate synthase 2 2.2 0.3 NM_018737 Art3 ADPribosyltransferase 3 2.2 0.4 NM_181728 Sgce Sarcoglycan, epsilon 2.2 0.5 NM_011360 Zscan18 Zinc finger and SCAN domain containing 18 2.2 0.5 NM_001017955 Eif2c4 Eukaryotic translation initiation factor 2C, 4 2.2 0.4 NM_153177 Col5a2 Collagen, type V, alpha 2 2.3 0.3 NM_007737 Raver2 Ribonucleoprotein, PTBbinding 2 2.3 0.2 NM_183024 Gabrd Gammaaminobutyric acid (GABA) A receptor, subunit delta 2.3 0.3 NM_008072 Radil Ras association and DIL domains 2.3 0.5 NM_178702 Ptpru Protein tyrosine phosphatase, receptor type, U 2.3 0.3 NM_001083119 Lgi4 Leucinerich repeat LGI family, member 4 2.3 0.2 NM_144556 Mum1l1 Melanomaassociated antigen (mutated) 1like 1 2.3 0.4 NM_175541 Postn Periostin, osteoblastspecific factor 2.3 0.2 NM_015784 Nsun7 NOL1/NOP2/Sun domain family, member 7 2.3 0.5 NM_027602 Me3 Malic enzyme 3, NADP(+)dependent, mitochondrial 2.3 0.2 NM_181407 Calml4 like 4 2.3 0.3 NM_138304 Bicc1 Bicaudal C homolog 1 (Drosophila) 2.3 0.3 NM_031397 Diabetes Page 64 of 84

Fhl2 Four and a half LIM domains 2 2.3 0.3 NM_010212 Caprin2 Caprin family member 2 2.3 0.5 NM_181541 Rspo2 Rspondin 2 homolog 2.3 0.3 NM_172815 Rufy4 RUN and FYVE domain containing 4 2.3 0.4 NM_001034060 Sipa1l1 Signalinduced proliferationassociated 1 like 1 2.3 0.4 NM_001167983 Flrt3 Fibronectin leucinerich transmembrane protein 3 2.3 0.4 NM_001172160 Ddx4 EEAD (AspGluAlaAsp) box polypeptide 4 2.3 0.1 NM_010029 Sparcl1 SPARClike 1 2.3 0.2 NM_010097 Gstm1 Glutathione Stransferase, mu 1 2.3 0.5 NM_010358 Acrbp Proacrosin binding protein 2.3 0.4 NM_016845 Myl1 Myosin, light polypeptide 1 2.3 0.0 NM_021285 Man1a2 Mannosidase, alpha, class 1A, member 2 2.3 0.5 NM_010763 Nrbp2 Nuclear receptor binding protein 2 2.4 0.3 NM_144847 Aqp11 11 2.4 0.5 NM_175105 Rasgrp2 RAS, guanyl releasing protein 2 2.4 0.4 NM_011242 Ccl27a Chemokine (CC motif) ligand 27A 2.4 0.5 NM_011336 Ppara Peroxisome proliferatoractivated receptor alpha 2.4 0.5 NM_011144 Abi3bp ABI gene family, member 3 (NESH) binding protein 2.4 0.3 NM_001014423 Dyx1c1 Dyslexia susceptibility 1 candidate 1 homolog (human) 2.4 0.4 NM_026314 Pde2a Phosphodiesterase 2A, cGMPstimulated 2.4 0.4 NM_001008548 Mosc1 MOCO sulfurase Cterminal domain containing 1 2.4 0.3 NM_001081361 Page 65 of 84 Diabetes

Lrrn3 Leucinerich repeat protein 3, neuronal 2.4 0.4 NM_010733 Cxcl12 Chemokine (CXC motif) ligand 12 2.4 0.5 NM_021704 Il17rd Interleukin 17 receptor D 2.5 0.4 NM_13443 Dlc1 Deleted in liver cancer 1 2.5 0.4 XM_486096 Mtap7d3 MAP7 domain containing 3 2.5 0.4 NM_177293 Nxnl2 Nucleoredoxinlike 2 2.5 0.3 NM_029173 Chadl Chondroadherinlike 2.5 0.4 NM_001164320 Spry1 Sprouty homolog 1 (Drosophila) 2.5 0.2 NM_011896 Ogn Osteoglycin 2.5 0.1 NM_008760 Clca1 Chloride channel calcium activated 1 2.5 0.4 NM_009899 Amy1 Amylase 1, salivary 2.5 0.2 NM_007446 Prss12 Protease, serine, 12 neurotrypsin 2.5 0.4 NM_008939 Fam71e1 Family with sequence similarity 71, member E1 2.5 0.5 NM_028169 Uba7 Ubiquitinlike modifier activating enzyme 7 2.5 0.5 NM_023738 Ctla2b Cytotoxic T lymphocyteassociated protein 2 beta 2.5 0.5 NM_007797 Dnahc2 Dynein, axonemal, heavy chain 2 2.5 0.2 NM_001081330 Adig Adipogenin 2.6 0.2 NM_145635 Zfpm1 Zinc finger protein, multitype 1 2.6 0.5 NM_009569 Thbs4 Thrombospondin 4 2.6 0.3 NM_011582 Ccdc46 Coiledcoil domain containing 46 2.6 0.5 NM_029606 Sfrs18 Splicing factor, arginine/serinerich 18 2.6 0.4 NM_025669 Diabetes Page 66 of 84

Ggt5 Gammaglutamyltransferase 5 2.6 0.5 NM_011820 Scai Suppressor of cancer cell invasion 2.6 0.5 NM_178778 Lpl Lipoprotein lipase 2.6 0.3 NM_008509 Zfpm1 Zinc finger protein, multitype 1 2.6 0.5 NM_009569 Ccdc106 Coiledcoil domain containing 106 2.6 0.4 NM_146178 Avpr1a Arginine receptor 1A 2.6 0.2 NM_016847 Ccdc80 Coiledcoil domain containing 80 2.7 0.3 NM_026439 Igf1 Insulinlike growth factor 1 2.7 0.3 NM_010512 Ankrd29 Ankyrin repeat domain 29 2.7 0.4 NM_001190371 Gnb1l Guanine nucleotide binding protein (G protein), beta polypeptide 1like 2.7 0.5 NM_001081682 EG665756 Predicted gene, EG665756 2.7 0.5 XM_979235 Agtr1a Angiotensin II receptor, type 1a 2.7 0.2 NM_177322 Cd200 CD200 antigen 2.7 0.3 NM_010818 Txnrd3 Thioredoxin reductase 3 2.7 0.4 NM_153162 Epas1 Endothelial PAS domain protein 1 2.8 0.4 NM_010137 Cacna1g , voltagedependent, T type, alpha 1G subunit 2.8 0.3 NM_009783 Gprasp2 G proteincoupled receptor associated sorting protein 2 2.8 0.3 XM_142154 Gpihbp1 GPIanchored HDLbinding protein 1 2.8 0.5 NM_026730 Lrrtm1 Leucinerich repeat transmembrane neuronal 1 2.8 0.3 NM_028880 Pcsk6 Proprotein convertase subtilisin/kexin type 6 2.8 0.5 XM_355911 Id3 Inhibitor of DNA binding 3 2.8 0.5 NM_008321 Page 67 of 84 Diabetes

Cfh Complement component factor h 2.8 0.3 NM_009888 Wisp2 WNT1 inducible signaling pathway protein 2 2.8 0.1 NM_016873 Plekha4 Pleckstrin homology domain containing, family A member 4 2.8 0.3 NM_148927 Clstn2 Calsyntenin 2 2.8 0.4 NM_022319 Aldh1a3 Aldehyde dehydrogenase family 1, subfamily A3 2.8 0.2 NM_022319 Apoc1 Apolipoprotein CI 2.8 0.3 NM_007469 Myl9 Myosin, light polypeptide 9, regulatory 2.9 0.3 XM_485171 Cdh6 Cadherin 6 2.9 0.3 NM_007666 Kif5a family member 5A 2.9 0.4 NM_001039000 Crygs Crystallin, gamma S 3.0 0.1 NM_009967 Cyp1a1 Cytochrome P450, family 1, subfamily a, polypeptide 1 3.0 0.4 NM_009992 Lgr5 Leucinerich repeat containing G proteincoupled receptor 5 3.0 0.1 NM_010195 Crispld2 Cysteinerich secretory protein LCCL domain containing 2 3.0 0.3 NM_030209 Kcnip4 Kv channel interacting protein 4 3.1 0.4 NM_030265 Fat4 FAT tumor suppressor homolog 4 (Drosophila) 3.1 0.4 XM_619892 Gpd1 Glycerol3phosphate dehydrogenase 1 3.1 0.2 NM_010271 Kera Keratocan 3.2 0.5 NM_008438 Scube1 Signal peptide, CUB domain, EGFlike 1 3.2 0.4 NM_022723 Sh3d19 SH3 domain protein D19 3.2 0.4 NM_001082414 Ptpn13 Protein tyrosine phosphatase, nonreceptor type 13 3.3 0.4 NM_011204 Omd Osteomodulin 3.4 0.2 NM_012050 Diabetes Page 68 of 84

Adrb3 , beta 3 3.6 0.2 NM_013462 Itga11 Integrin alpha 11 3.7 0.2 NM_176922 G0s2 G0/G1 switch gene 2 3.9 0.2 NM_008059 Tspan18 Tetraspanin 18 3.9 0.1 NM_183180 Aoc3 Amine oxidase, copper containing 3 4.1 0.1 NM_009675 Ccny Cyclin Y 4.2 0.5 NM_026484 Cfd Complement factor D (adipsin) 4.3 0.1 NM_013459 Cdcp1 CUB domain containing protein 1 4.4 0.4 NM_133974 Tmed9 Transmembrane emp24 protein transport domain containing 9 4.8 0.4 NM_026211 Ptx3 Pentraxinrelated gene 5.3 0.2 NM_008987 Trio Triple functional domain (PTPRF interacting) 5.4 0.4 NM_001081302 Angpt2 Angiopoietin 2 5.5 0.5 NM_007426 Saa3 Serum amyloid A 3 5.8 0.3 NM_011315 Hp Haptoglobin 9.7 0.1 NM_017370

* MCECs were exposed to normal glucose (NG) or high glucose (HG) conditions and treated with phosphatebuffered saline (PBS) or DKK2 protein. We selected genes that changed more than 2fold for both HG + PBS/NG and HG + DKK2/HG + PBS.

Page 69 of 84 Diabetes

Supplementary Table S7. Physiological and metabolic parameters 10 weeks after induction of diabetes with streptozotocin (STZ) with or without treatment of insulin. Control STZ+PBS STZ+Insulin Body weight (g) 31.6±0.8 23.3±0.5* 26.9±1.1*

Fasting glucose (mg/dl) 104.4±3.2 492.8±12.6* 122.6±13.2#

Postprandial glucose (mg/dl) 160.6±3.5 591.4±5.3* 190.0±20.0# MSBP (mmHg) 106.1±2.2 102.6±1.7 104.2±2.2 Values are the mean ± SE for n = 45 animals per group. *P < 0.01 vs. Control. #P < 0.01 vs. STZ+PBS. MSBP, mean systolic blood pressure; PBS, phosphatebuffered saline.

Diabetes Page 70 of 84

Supplemental Figures:

Supplementary Figure 1. DKK2 is mainly expressed in pericytes.

A and B: Representative RTPCR and Western blots for DKK2 in human umbilical vein endothelial cells (HUVECs) and human brain microvascular pericytes (HBMPs).

C and D: Normalized band intensity values (n = 4). *P < 0.05 vs. HUVECs, Mann

Whitney U test. Data are presented as mean ± SE. The relative ratio in the HUVEC group was arbitrarily set to 1.

Page 71 of 84 Diabetes

Supplementary Figure 2. DKK2 overexpression enhances proliferation of

endothelial cells and pericytes. A: PDGFRβ (red) and BrdU (green) staining in penis

tissue from wildtype (WT) and DKK2Tg mice, WT mice receiving streptozotocin

(STZ), and DKK2Tg mice receiving STZ. Nuclei were labeled with DAPI (blue).

Scale bars = 50 µm. B: BrdU (green) staining in mouse cavernous endothelial cells

(MCECs), mouse cavernous pericytes (MCPs), human umbilical vein endothelial

cells (HUVECs), and human brain microvascular pericytes (HBMPs). The cells were

exposed to normal glucose (NG) or high glucose (HG) conditions for 48 hours and

treated with PBS or DKK2 protein (200 ng/ml). C: Number of BrdU immunopositive

pericytes per highpower field (n = 4). *P < 0.05 vs. WT and DKK2Tg groups. #P <

0.001 vs. WT + STZ group. D to G: Number of BrdU immunopositive endothelial

cells or pericytes per highpower field (n = 4). *P < 0.05 vs. NG group. #P < 0.01 vs. Diabetes Page 72 of 84

HG group. Pvalues were determined by KruskalWallis test. Data in graphs are presented as mean ± SE. PDGFRβ, plateletderived growth factor receptorbeta.

Supplementary Figure 3. Preserved eNOS phosphorylation in DKK2Tg mice under diabetic conditions. A: CD31 (red) and phosphoeNOS (Ser1177; green) staining of cavernous tissue from wildtype (WT) and DKK2Tg mice, WT mice receiving streptozotocin (STZ), and DKK2Tg mice receiving STZ. Scale bar = 100 Page 73 of 84 Diabetes

µm. B: Quantification of the phosphoeNOS immunopositive area in cavernous

tissue by Image J (n = 4). *P < 0.001 vs. WT and DKK2Tg groups. #P < 0.001 vs.

WT + STZ group. C: Representative Western blots for phosphoAkt and Akt or

phosphoeNOS and eNOS in agematched control and diabetic mice 2 weeks after

repeated intracavernous injections of PBS (20 µl) or DKK2 protein (days 3 and 0; 6

µg/20 µl). Arrows represent target bands. D and E: Normalized band intensity values

(n = 4). *P < 0.05 vs. control (C) group. #P < 0.05 vs. PBStreated group. F and G:

Immunoprecipitation (IP) to confirm the phosphoeNOS and eNOS Western blot

data. Pvalues were determined by KruskalWallis test. Data in graphs are presented

as mean ± SE. The relative ratio in the WT or C group was arbitrarily set to 1.

Diabetes Page 74 of 84

Supplementary Figure 4. Preserved endothelial cellcell junction proteins in

DKK2Tg mice under diabetic conditions. A and B: CD31 (red) and claudin5 (green) staining of cavernous tissue from wildtype (WT) and DKK2Tg mice, WT mice receiving streptozotocin (STZ), and DKK2Tg mice receiving STZ. Scale bar = 100

µm. Quantification of the claudin5 immunopositive area in cavernous tissue by

Image J (n = 4). *P < 0.001 vs. WT and DKK2Tg groups. #P < 0.001 vs. WT + STZ group. C and D: CD31 (red) and VEcadherin (green) staining of cavernous tissue.

Scale bar = 100 µm. Quantification of the VEcadherin immunopositive area (n = 4). Page 75 of 84 Diabetes

*P < 0.01 vs. WT and DKK2 Tg groups. #P < 0.001 vs. WT + STZ group. Pvalues

were determined by KruskalWallis test. Data in graphs are presented as mean ± SE.

The relative ratio in the WT group was arbitrarily set to 1. VE, vascular endothelial.

Supplementary Figure 5. DKK2Tg mice decrease the extravasation of oxidized

LDL under diabetic conditions. A and B: CD31 (red) and oxidizedLDL (green)

staining of cavernous tissue from wildtype (WT) and DKK2Tg mice, WT mice

receiving streptozotocin (STZ), and DKK2Tg mice receiving STZ. Scale bar = 100

µm. Quantification of the oxidizedLDL immunopositive area in cavernous tissue by

Image J (n = 4). *P < 0.05 vs. WT and DKK2Tg groups. #P < 0.05 vs. WT + STZ

group. Pvalues were determined by KruskalWallis test. Data are presented as mean

± SE. The relative ratio in the WT group was arbitrarily set to 1. LDL, lowdensity

lipoprotein.

Diabetes Page 76 of 84

Supplementary Figure 6. Establishment of a direct mixed MCECMCP coculture system. To determine the optimal ratio of MCECs and MCPs for direct mixed cell culture, the cells were cultivated at different ratios (10:1, 5:1, 3:1, 2:1, 1:1). Tube formation assays were performed on matrigel in 96well plates and images taken at

24 and 48 hours. 40× magnification. MCEC, mouse cavernous endothelial cell; MCP, mouse cavernous pericyte.

Page 77 of 84 Diabetes

Supplementary Figure 7. DKK2 overexpression promotes migration of endothelial

cells and pericytes. A to C: Scratch woundhealing assay in mouse cavernous

pericytes (MCPs), human umbilical vein endothelial cells (HUVECs), and human

brain microvascular pericytes (HBMPs) exposed to normal glucose (NG) or high

glucose (HG) conditions for 48 hours and treated with PBS or DKK2 protein (200

ng/ml). The images were taken 24 hours after scratching. 40× magnification. D to F:

Number of migrated endothelial cells and pericytes (n = 4). *P < 0.001 vs. NG group.

#P < 0.001 vs. PBStreated group. Pvalues were determined by KruskalWallis test.

Data in graphs are presented as mean ± SE. Diabetes Page 78 of 84

Supplementary Figure 8. DKK2 overexpression decreases cavernous reactive oxygen species (ROS) production under diabetic conditions. A: Representative

Western blots for iNOS in agematched control and diabetic mice 2 weeks after repeated intracavernous injections of PBS (20 µl) or DKK2 protein (days 3 and 0; 6

µg/20 µl). B: Normalized band intensity values (n = 4). *P < 0.01 vs. control (C) group. #P < 0.01 vs. PBStreated group. C: Immunoprecipitation (IP) to confirm the iNOS Western blot data. D: In situ detection of superoxide anion in cavernous endothelial cells of each group of animals. Corpus cavernosum tissue was incubated with hydroethidine (red), an oxidative fluorescent dye used to detect superoxide anion, and antibody to CD31 (green). E and F: Number of ethidium bromide Page 79 of 84 Diabetes

fluorescencepositive endothelial cells (ECs) per highpower field (screen

magnification ×400) (n = 4). (E) *P < 0.05 vs. WT and DKK2Tg groups. #P < 0.05

vs. WT + STZ group. (F) *P < 0.05 vs. C group. #P < 0.05 vs. PBStreated group. G:

Nitrotyrosine (red) and CD31 (green) staining in cavernous endothelial cell of each

group of animals (n = 4). H and I: The nitrotyrosineimmunopositive area quantified

in cavernous endothelial cells by Image J. (H) *P < 0.05 vs. WT and DKK2Tg

groups. #P < 0.05 vs. WT + STZ group. (I) *P < 0.05 vs. C group. #P < 0.05 vs. PBS

treated group. Pvalues were determined by KruskalWallis test. Data in graphs are

presented as mean ± SE. The relative ratio in the C group was arbitrarily set to 1.

Supplementary Figure 9. DKK2 overexpression is resistant to diabetesinduced Diabetes Page 80 of 84

erectile dysfunction. A: Representative intracavernous pressure (ICP) responses for wildtype (WT) and DKK2Tg mice, WT mice receiving streptozotocin (STZ), and

DKK2Tg mice receiving STZ. The cavernous nerve was stimulated at 5 V. The stimulus interval is indicated by a solid bar. The ICP was measured 8 weeks after

STZ injection. B Representative ICP responses for agematched control and diabetic mice stimulated 2 weeks after repeated intracavernous injections of PBS (20 µl) or

DKK2 protein (days 3 and 0; 6 µg/20 µl). C to F: Ratios of mean maximal ICP and total ICP (area under the curve) to mean systolic blood pressure (MSBP) were calculated for each group (n = 58). (C and D) *P < 0.001 vs. WT and DKK2Tg groups. #P < 0.001 vs. WT + STZ group. (E and F) *P < 0.001 vs. control (C) group.

#P < 0.001 vs. PBStreated group. Pvalues were determined by KruskalWallis test.

Data are presented as mean ± SE.

Page 81 of 84 Diabetes

Supplementary Figure 10. Determination of optimal dosage of DKK2 protein to

induce maximal erectile function recovery under diabetic conditions. A:

Representative ICP responses for agematched control and diabetic mice stimulated

2 weeks after repeated intracavernous injections of PBS (20 µl) or DKK2 protein

(days 3 and 0; 1 µg/20 µl, 6 µg/20 µl, and 10 µg/20 µl, respectively). The cavernous

nerve was stimulated at 5 V. The stimulus interval is indicated by a solid bar. B and

C: Ratios of mean maximal ICP and total ICP (area under the curve) to mean systolic

blood pressure (MSBP) were calculated for each group (n = 510). *P < 0.05 vs.

control (C) group. #P < 0.05 vs. PBStreated group. Pvalues were determined by

KruskalWallis test. Data are presented as mean ± SE.

Diabetes Page 82 of 84

Supplementary Figure 11. Angiopoietin1 (Ang1) derived from mouse cavernous pericytes (MCPs) rather than mouse cavernous endothelial cells (MCECs) plays a crucial role in DKK2mediated angiogenesis and neural regeneration. A and B: Tube formation assay in MCECMCP mixed coculture exposed to normal glucose (NG) or high glucose (HG) conditions for 48 hours and treated with DKK2 protein (200 ng/ml). MCECs and MCPs were transfected with scrambled or Ang1 siRNA (200 pmol/l). (A) 40× magnification. (B) Number of tubes per highpower field (n = 4).

*P < 0.05 vs. NG group; #P < 0.05 vs. HG group. †P < 0.05 vs. HG + DKK2 group. Page 83 of 84 Diabetes

C and D: Conditioned medium derived from MCECMCP mixed coculture regulates

neurite sprouting in mouse pelvic ganglion (MPG) tissue culture. (C) βIII tubulin

(red) staining in MPG tissue culture exposed to NG or HG conditions for 72 hours

and treated with conditioned medium from each group. (D) Quantification of neurite

length by Image J (n = 4). *P < 0.001 vs. NG group; #P < 0.001 vs. HG group. †P <

0.001 vs. HG + DKK2 group. Pvalues were determined by KruskalWallis test. Data

in graphs are presented as mean ± SE. The relative ratio in the NG group was

arbitrarily set to 1.

Supplementary Figure 12. Insulin treatment does not rescue diabetesinduced

erectile dysfunction. A: Representative intracavernous pressure (ICP) responses for Diabetes Page 84 of 84

agematched control and diabetic mice treated with PBS or insulin (4 IU/day).

Insulin treatment started 1 week after STZ injection (9 weeks of age) and continued for 9 weeks (18 weeks of age). The cavernous nerve was stimulated at 5 V. The stimulus interval is indicated by a solid bar. B and C: Ratios of mean maximal ICP and total ICP (area under the curve) to mean systolic blood pressure (MSBP) were calculated for each group (n = 5). *P < 0.001 vs. control (C) group. D:

Representative Western blots for DKK2 in each group of animals. E: Normalized band intensity values (n = 4). *P < 0.01 vs. C group. Pvalues were determined by

KruskalWallis test. Data in graphs are presented as mean ± SE. The relative ratio in the C group was arbitrarily set to 1.