Penile erection requires well-coordinated interactions between vascular and nervous systems. Penile neurovascular dysfunction is a major cause of erectile dysfunction (ED) in patients with diabetes, which causes poor response to oral phosphodiesterase-5 inhibitors. Dickkopf2 (DKK2), a Wnt antagonist, is known to promote angiogenesis. Here, using DKK2-Tg mice or DKK2 protein administration, we demonstrate that the overexpression of DKK2 in diabetic mice enhances penile angiogenesis and neural regeneration and restores erectile function. Transcriptome analysis revealed that angiopoietin-1 and angiopoietin-2 are target genes for DKK2. Using an endothelial cell-pericyte coculture system and ex vivo neurite sprouting assay, we found that DKK2-mediated juxtacrine signaling in pericyte-endothelial cell interactions promotes angiogenesis and neural regeneration through an angiopoietin-1-Tie2 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.

Penile erection is a neurovascular phenomenon that requires well-coordinated interactions among vascular endothelial (VE) cells, smooth muscle cells, pericytes, and neuronal cells (1,2). Erectile dysfunction (ED) affects more than half of men 40–70 years of age (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 inhibitors enhance the nitric oxide (NO)-cyclic guanosine monophosphate pathway and are currently used as a first-line therapy for ED (1). The reduced responsiveness to phosphodiesterase type 5 inhibitors in patients with neuropathy, severe angiopathy, or both, such as patients with 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 re-establishes structural and functional penile neurovasculature and augments endogenous NO bioactivity.

Several proteins have been effective in targeting angiogenesis of the penis in diabetic or hypercholesterolemic animal models of ED, including VE growth factor, angiopoietin (Ang)-1, and Ang4 (714). Targeting neural regeneration, brain-derived neurotrophic factor (BDNF) or neurotrophin-3 (NT-3) has shown some effectiveness by restoring neuronal NO synthase (nNOS)-positive neurons (15,16). However, new therapeutic agents are far from development because of incomplete effects; potential adverse effects, such as inflammation; and difficulties engineering a 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 targeting these complicated pathologies in refractory ED would be ideal.

Dickkopf2 (DKK2) is a secreted protein containing two cysteine-rich regions that act as a Wnt antagonist by binding LDL receptor–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, DKK2-induced capillaries had more coverage of endothelial cells by pericytes and were less leaky than VE growth factor–induced 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 physiological erection. We also determined the role of DKK2 in penile neural regeneration.

Here, we demonstrate that the overexpression of DKK2 in DKK2-Tg mice that express mouse DKK2 under the control of the endothelial cell–specific Tie2 promoter/enhancer, or in wild-type (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 NO synthase, restoration of the integrity of endothelial cell-cell 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 Ang2, an endogenous antagonist of Ang1, is upregulated by high glucose (HG). This effect was reversed by treatment with DKK2 protein. Using an MCEC-mouse cavernous pericyte (MCP) coculture system and an ex vivo neurite sprouting assay, we also found that DKK2-mediated juxtacrine signaling in MCECs-MCPs promotes angiogenesis and neural regeneration though an Ang1-Tie2 pathway. The recovery of erectile function mediated by DKK2 was abolished by the inhibition of Ang1-Tie2 signaling with soluble Tie2 protein (sTie2-Fc).

Study Design

The primary aim of the current study was to investigate the mechanisms through which DKK2 restores diabetes-induced ED. We used DKK2-Tg mice and administered DKK2 protein into the penis of diabetic mice. Detailed mechanisms were evaluated with WT or DKK2-Tg mice and primary cultures of MCECs, MCPs, and mouse albino neuroblastoma (Neuro2A) cells. All parameters of genetically modified mice and diabetic mice were compared with those of littermate controls.

Animals and Treatments

Eight-week-old male C57BL/6 (Orient Bio) and DKK2-Tg mice (provided by Young-Guen Kwon, Yonsei University, Republic of Korea) were used in this study. DKK2-Tg mice were backcrossed with C57BL/6 mice for at least seven generations (20). The experiments were approved by the Institutional Animal Care and Use Committee of Inha University (Assurance Number INHA 140110–267–1). Diabetes was induced by intraperitoneal injection of multiple low doses of streptozotocin (STZ) (50 mg/kg body wt in 0.1 mol/L 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 in the supine position on a thermoregulated surgical table.

For the DKK2-Tg study, the mice were distributed into the following four groups: WT controls, DKK2-Tg mice, WT mice receiving STZ (50 mg/kg body wt for 5 days), and DKK2-Tg mice receiving STZ (50 mg/kg body wt for 5 days). Eight weeks after the induction of diabetes, we measured erectile function during electrical stimulation of the cavernous nerve.

For the inhibition study with sTie2-Fc, the mice were distributed into the following four groups: DKK2-Tg mice, STZ-induced WT diabetic mice, and STZ-induced DKK2-Tg diabetic mice receiving subcutaneous injection of dimeric-Fc or sTie2-Fc (4 µg/20 µL; R&D Systems). The dose of sTie2-Fc was determined based on our previous report (22). sTie2-Fc or dimeric-Fc 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 the following three groups: age-matched controls and STZ-induced 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 sTie2-Fc, the mice were distributed into the following four groups: age-matched controls and STZ-induced WT diabetic mice receiving repeated intracavernous injections of PBS, DKK2 protein (days −3 and 0; 6 µg/20 µL) and dimeric-Fc, or DKK2 protein (days −3 and 0; 6 µg/20 µL) and sTie2-Fc (4 µg/20 µL). sTie2-Fc or dimeric-Fc 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 the following three groups: age-matched controls and STZ-induced WT diabetic mice receiving repeated intraperitoneal injection of PBS or insulin (4 international units/day; Sigma-Aldrich) (23,24). Insulin treatment started 1 week after STZ injection (9 weeks of age) 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 kit (Mercodia).

Human Corpus Cavernosum Tissue

Human corpus cavernosum tissue samples were obtained from a 21-year-old patient with congenital penile curvature who had normal erectile function during reconstructive penile surgery and a 56-year-old patient with diabetic ED (type 2 diabetes mellitus, with a duration of 22 years; HbA1c level 8.8% [73 mmol/mol]; BMI, 23.1 kg/m2; 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 the Supplementary Data.

Cell Culture Experiments

The MCECs and MCPs were prepared and maintained as described previously (2,25,26). Tube formation assay, scratch wound-healing assay, transfection assay, RT-PCR, and cDNA microarray were performed as described in the Supplementary Data.

Aortic Ring Assay

Aortas were harvested from 8-week-old C57BL/6 WT mice. The aortic rings were placed in the eight-well Nunc Lab-Tek Chamber Slide System (Sigma-Aldrich) and sealed in place with an overlay of 50 µL of Matrigel. The aortic rings were cultured in medium 199 with 20 ng/mL 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 min 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, were transferred into sterile vials containing Hank’s balanced salt solution (Gibco), and then rinsed and washed twice in PBS. The MPG tissue was cut into small pieces, and the samples were plated on a poly-d-lysine hydrobromide–coated (Sigma-Aldrich) 12-well plate. The whole MPG tissue sample was covered with Matrigel, and the culture plate was placed on ice for 5 min prior to incubation at 37°C for 10–15 min in a 5% CO2 atmosphere. We added 1 mL of complete Neurobasal Medium (Gibco) supplemented with 2% serum-free B-27 (Gibco) and 0.5 nmol/L 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 diabetes-induced angiopathy and neuropathy, primary cultures or tissues were serum starved for 24 h and then exposed to normal glucose (NG; 5 mmol) or HG (30 mmol; Sigma-Aldrich) 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 pericyte-derived 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 or absence of DKK2 depletion was transferred to MCPs to determine the effect of endothelial cell–derived DKK2 on pericytes. To do this, MCECs and MCPs were grown in 60-mm dishes until 80% confluence, and the medium was changed for an additional 2 days. The culture supernatants were collected and centrifuged for 10 min at 200g to remove cell debris. For DKK2 depletion, an immunoprecipitation protocol was used. Briefly, the CM was incubated with DKK2 antibody (1:200; catalog #Ab95274; Abcam) or control rabbit IgG (1:100; catalog #sc-2027; Santa Cruz Biotechnology) for 12 h at 4°C. Protein G-coupled Sepharose beads (Millipore) were added to the medium and incubated for an additional 12 h 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 MCEC-MCP 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 small interfering RNA (siRNA); HG + DKK2 protein (200 ng/mL) + Ang1 siRNA (200 pmol) transfection in MCECs; HG + DKK2 protein (200 ng/mL) + Ang1 siRNA (200 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 min at 200g to remove cell debris and then transferred to MPG tissue.

Histological Examinations

Histological examinations and BrdU labeling were performed as described in the Supplementary Data.

Western Blot

Western blot analysis was performed as described in the Supplementary Data.

Statistical Analysis

The results are expressed as the mean ± SE. Intergroup comparisons were made by Mann-Whitney U test or Kruskal-Wallis test. P values <5% were considered significant. We used SigmaStat 3.11 software (Systat Software) for statistical analyses.

Metabolic Variables

The fasting and postprandial blood glucose concentrations as well as HbA1c levels in STZ-treated WT or DKK2-Tg diabetic mice were significantly higher than in control mice. In addition, body weight and serum insulin levels were significantly lower in the STZ-induced 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 Tables 14).

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. 1A–D 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–H).

Figure 1

Decreased DKK2 expression under diabetic conditions. A and B: Representative RT-PCR and Western blots for DKK2 in MCECs and 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 ImageJ. 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 age-matched control and diabetic mouse penis and in MCECs and MCPs exposed to NG or HG conditions for 48 h. 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 ED or diabetic mice and age-matched control groups. Scale bar = 100 µm (left) or 50 μm (magnification image). P and Q: The DKK2-immunopositive area was quantified by ImageJ. 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 P values were determined by Mann-Whitney U test. Data in graphs are presented as the mean ± SE. The relative ratio in the control or NG groups was arbitrarily set to 1. C, control; DM, type 2 diabetes for human data and type 1 diabetes for mouse data; VWF, von Willebrand factor; WB, Western blot.

Figure 1

Decreased DKK2 expression under diabetic conditions. A and B: Representative RT-PCR and Western blots for DKK2 in MCECs and 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 ImageJ. 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 age-matched control and diabetic mouse penis and in MCECs and MCPs exposed to NG or HG conditions for 48 h. 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 ED or diabetic mice and age-matched control groups. Scale bar = 100 µm (left) or 50 μm (magnification image). P and Q: The DKK2-immunopositive area was quantified by ImageJ. 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 P values were determined by Mann-Whitney U test. Data in graphs are presented as the mean ± SE. The relative ratio in the control or NG groups was arbitrarily set to 1. C, control; DM, type 2 diabetes for human data and type 1 diabetes for mouse data; VWF, von Willebrand factor; WB, Western blot.

Close modal

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. 1I–N). Immunofluorescent staining also revealed that the expression of DKK2 protein was significantly lower in cavernous tissue from patients with diabetes or diabetic mice than in the control groups (Fig. 1O–Q). These findings gave us a rationale to use DKK2 for the 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 DKK2-Tg 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 DKK2-Tg 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 endothelial NO synthase (Supplementary Fig. 3), restored cavernous endothelial cell-cell junction proteins claudin-5 and VE-cadherin (Supplementary Fig. 4), and decreased the extravasation of oxidized LDL (Supplementary Fig. 5) under diabetic conditions compared with WT littermates that received STZ.

Figure 2

DKK2 overexpression is resistant to diabetes-induced angiopathy. A: CD31 (green) and PDGFRβ (red) staining in cavernous tissue from WT and DKK2-Tg mice, WT mice receiving STZ, and DKK2-Tg 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 (top) and 25 µm (bottom). C: Tube formation assay in MCEC or MCP monoculture and MCEC-MCP mixed coculture exposed to NG or HG conditions for 48 h and treated with PBS or DKK2 protein (200 ng/mL). Original magnification ×40. D: Scratch wound-healing assay in MCECs 24 h after treatment. Original magnification ×40. E and F: Ex vivo aortic ring assay. The images were taken 5 days after treatment. E: Original magnification ×40. 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. Original magnification ×100. Results were similar in three independent experiments. G: Quantification of cavernous endothelial cell and pericyte content by ImageJ (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 DKK2-Tg group was arbitrarily set to 1. H: Number of BrdU-positive endothelial cells per high-power field (HPF) (n = 6). *P < 0.05 vs. WT and DKK2-Tg groups. #P < 0.01 vs. WT + STZ group. I: Number of branch points per high-power field (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated group. J: Number of migrated endothelial cells (n = 6). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated group. K: Area of outgrowing microvessels from aortic ring (n = 6). *P < 0.05 vs. NG group. #P < 0.05 vs. PBS-treated group. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE.

Figure 2

DKK2 overexpression is resistant to diabetes-induced angiopathy. A: CD31 (green) and PDGFRβ (red) staining in cavernous tissue from WT and DKK2-Tg mice, WT mice receiving STZ, and DKK2-Tg 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 (top) and 25 µm (bottom). C: Tube formation assay in MCEC or MCP monoculture and MCEC-MCP mixed coculture exposed to NG or HG conditions for 48 h and treated with PBS or DKK2 protein (200 ng/mL). Original magnification ×40. D: Scratch wound-healing assay in MCECs 24 h after treatment. Original magnification ×40. E and F: Ex vivo aortic ring assay. The images were taken 5 days after treatment. E: Original magnification ×40. 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. Original magnification ×100. Results were similar in three independent experiments. G: Quantification of cavernous endothelial cell and pericyte content by ImageJ (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 DKK2-Tg group was arbitrarily set to 1. H: Number of BrdU-positive endothelial cells per high-power field (HPF) (n = 6). *P < 0.05 vs. WT and DKK2-Tg groups. #P < 0.01 vs. WT + STZ group. I: Number of branch points per high-power field (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated group. J: Number of migrated endothelial cells (n = 6). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated group. K: Area of outgrowing microvessels from aortic ring (n = 6). *P < 0.05 vs. NG group. #P < 0.05 vs. PBS-treated group. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE.

Close modal

We further examined the role of DKK2 in MCEC and MCP monoculture or direct mixed coculture. The mixture of MCECs and MCPs formed well-organized capillary-like structures at a 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. 2C 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 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 Reactive Oxygen Species Production Under Diabetic Conditions

Cavernous inducible NO synthase protein expression was significantly higher in the PBS-treated diabetic mice than in the age-matched controls. Repeated intracavernous injections of DKK2 protein (days −3 and 0; 6 µg/20 µL) significantly decreased cavernous inducible NO synthase 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 DKK2-Tg mice that received STZ. DKK2 protein also significantly decreased cavernous reactive oxygen species production under diabetic conditions (Supplementary Fig. 8D–I).

Transmigration of DKK2 From Pericytes to Endothelial Cells Promotes Angiogenesis

Immunocytochemical staining revealed higher DKK2 expression in MCPs than MCECs. In contrast, after cultivation of MCECs and MCPs using an indirect noncontact coculture system, DKK2 expression was higher in MCECs than in MCPs (Fig. 3A). To confirm whether pericyte-derived DKK2 migrates into the endothelial cells, MCPs were transfected with DKK2-red fluorescent protein (RFP) DNA. We observed DKK2-RFP expression in MCECs after coculture with DKK2-RFP–transfected MCPs (Fig. 3B).

Figure 3

Pericyte-derived DKK2 migrates into endothelial cells and regulates angiogenesis. A: DKK2 (red) staining in MCEC and 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 DKK2-RFP DNA then cocultured with MCECs. Note the DKK2-RFP 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 MCP-conditioned CM with or without deletion of DKK2 with neutralizing antibody. Original magnification ×40. Number of tubes per high-power 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 MCEC-conditioned CM with or without deletion of DKK2 with neutralizing antibody. Original magnification ×40. Number of tubes per high-power field (n = 4). *P < 0.001 vs. control group. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE. C, control; NT, no treatment.

Figure 3

Pericyte-derived DKK2 migrates into endothelial cells and regulates angiogenesis. A: DKK2 (red) staining in MCEC and 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 DKK2-RFP DNA then cocultured with MCECs. Note the DKK2-RFP 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 MCP-conditioned CM with or without deletion of DKK2 with neutralizing antibody. Original magnification ×40. Number of tubes per high-power 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 MCEC-conditioned CM with or without deletion of DKK2 with neutralizing antibody. Original magnification ×40. Number of tubes per high-power field (n = 4). *P < 0.001 vs. control group. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE. C, control; NT, no treatment.

Close modal

To test the functional role of pericyte-derived 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 MCP-CM compared with cells treated with complement medium for MCPs. However, MCECs treated with DKK2-depleted MCP-CM had profoundly impaired tube formation (Fig. 3C and D). Although DKK2-depleted MCEC-CM slightly decreased tube formation in MCPs compared with 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 neurofilament, βIII tubulin, 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 DKK2-Tg mice that received STZ (Fig. 4A and D–F). DKK2 protein also significantly enhanced neurite sprouting in an ex vivo MPG tissue culture exposed to HG (Fig. 4B and G).

Figure 4

DKK2 overexpression is resistant to diabetes-induced neuropathy. A: Neurofilament (NF; red), nNOS (red in dorsal nerve and green in cavernosum), and βIII tubulin (red) staining in penis tissue from WT and DKK2-Tg mice, WT mice receiving STZ, and DKK2-Tg mice receiving STZ. Nuclei were labeled with DAPI (blue). Scale bars = 25 μm (dorsal nerve bundle) or 50 μm (cavernosum). B: βIII tubulin (red) staining in MPG tissue culture exposed to NG or HG conditions for 72 h and treated with PBS or DKK2 protein (200 ng/mL). Scale bar = 200 µm. C: Representative Western blot for neurotrophic factors (NGF, BDNF, and NT-3) and TrkA in penis tissue from age-matched 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 h and treated with PBS or DKK2 protein (200 ng/mL). D–F: The NF, nNOS, and βIII tubulin immunopositive areas were quantified in dorsal nerve bundle (DNB) or cavernous tissue by ImageJ (n = 6). *P < 0.05 vs. WT and DKK2-Tg groups. #P < 0.05 vs. WT + STZ group. G: Quantification of neurite length by ImageJ (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated 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. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE. The relative ratio in the the WT, C, or NG group was arbitrarily set to 1.

Figure 4

DKK2 overexpression is resistant to diabetes-induced neuropathy. A: Neurofilament (NF; red), nNOS (red in dorsal nerve and green in cavernosum), and βIII tubulin (red) staining in penis tissue from WT and DKK2-Tg mice, WT mice receiving STZ, and DKK2-Tg mice receiving STZ. Nuclei were labeled with DAPI (blue). Scale bars = 25 μm (dorsal nerve bundle) or 50 μm (cavernosum). B: βIII tubulin (red) staining in MPG tissue culture exposed to NG or HG conditions for 72 h and treated with PBS or DKK2 protein (200 ng/mL). Scale bar = 200 µm. C: Representative Western blot for neurotrophic factors (NGF, BDNF, and NT-3) and TrkA in penis tissue from age-matched 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 h and treated with PBS or DKK2 protein (200 ng/mL). D–F: The NF, nNOS, and βIII tubulin immunopositive areas were quantified in dorsal nerve bundle (DNB) or cavernous tissue by ImageJ (n = 6). *P < 0.05 vs. WT and DKK2-Tg groups. #P < 0.05 vs. WT + STZ group. G: Quantification of neurite length by ImageJ (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated 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. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE. The relative ratio in the the WT, C, or NG group was arbitrarily set to 1.

Close modal

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 factor (NGF), BDNF, and tropomyosin receptor kinase (Trk) A was significantly higher in diabetic mice receiving DKK2 protein than in PBS-treated diabetic mice and comparable to the level found in age-matched controls. We observed similar results in Neuro2A cells in vitro (Fig. 4C and H–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 DKK2-mediated 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 DKK2-Tg mice that received STZ (Supplementary Fig. 9A, C, and D).

Two weeks after treatment, repeated intracavernous injections of DKK2 protein (days −3 and 0; 6 µg/20 µL) significantly induced the recovery of erection parameters in WT mice that received STZ (Supplementary Fig. 9B, E, and F). No detectable differences in MSBP were found among the experimental groups (Supplementary Tables 1 and 2). The dosage of DKK2 protein was determined based on the findings of 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 twofold in both conditions (i.e., MCECs exposed to NG conditions compared with those exposed to HG conditions + PBS, and MCECs exposed to HG conditions + DKK2 protein compared with those exposed to HG conditions + PBS). After filtering data from 39,429 genes, 201 genes were changed more than twofold. Of these genes, only three were downregulated under HG conditions compared with NG conditions. These levels were reversed after treatment with DKK2 protein (Supplementary Tables 5 and 6). These genes included Angpt1 (Ang1) and Angpt2 (Ang2) (Fig. 5A).

Figure 5

DKK2-mediated recovery of erectile function is dependent on the Ang1-Tie2 signaling pathway. A: Microarray analysis using total RNA from MCECs exposed to NG or HG conditions for 48 h and treated with PBS or DKK2 protein (200 ng/mL). B: Representative RT-PCR and Western blot for Ang1 and Ang2 in MCECs and MCPs exposed to NG or HG conditions for 48 h and treated 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. PBS-treated group. The relative ratio in the NG group was arbitrarily set to 1. E–G: Representative ICP responses for the DKK2-Tg mice, WT mice receiving STZ, DKK2-Tg mice receiving STZ and dimeric-Fc (4 µg), and DKK2-Tg mice receiving STZ and sTie2 antibody (sTie2; 4 µg) (n = 6). *P < 0.001 vs. DKK2-Tg group. #P < 0.001 vs. WT + STZ group. †P < 0.001 vs. DKK2-Tg + STZ + Fc group. H–J: Representative ICP responses for age-matched 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 dimeric-Fc, or DKK2 protein and sTie2 (n = 5). *P < 0.001 vs. control group. #P < 0.001 vs. PBS-treated group. †P < 0.001 vs. DKK2 + Fc group. K: CD31 (green), PDGFR-β (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 h and treated with PBS, DKK2 protein (200 ng/mL), or DKK2 protein + sTie2 (100 ng/mL). Original magnification ×40. M: βIII tubulin (red) staining in MPG tissue culture. N and O: The CD31, PDGFR-β, and βIII tubulin immunopositive areas quantified by ImageJ (n = 6). *P < 0.05 vs. DKK2-Tg group. #P < 0.05 vs. WT + STZ group. †P < 0.01 vs. DKK2-Tg + STZ + Fc group. The relative ratio in the DKK2-Tg group was arbitrarily set to 1. P: Number of tubes per high-power field (n = 4). *P < 0.001 vs. NG group. #P < 0.01 vs. PBS-treated group. †P < 0.01 vs. DKK2 + Fc group. Q: Quantification of neurite length by ImageJ (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated group. †P < 0.001 vs. DKK2 + Fc group. The relative ratio in the NG group was arbitrarily set to 1. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE. DNB, dorsal nerve bundle; WB, Western blot.

Figure 5

DKK2-mediated recovery of erectile function is dependent on the Ang1-Tie2 signaling pathway. A: Microarray analysis using total RNA from MCECs exposed to NG or HG conditions for 48 h and treated with PBS or DKK2 protein (200 ng/mL). B: Representative RT-PCR and Western blot for Ang1 and Ang2 in MCECs and MCPs exposed to NG or HG conditions for 48 h and treated 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. PBS-treated group. The relative ratio in the NG group was arbitrarily set to 1. E–G: Representative ICP responses for the DKK2-Tg mice, WT mice receiving STZ, DKK2-Tg mice receiving STZ and dimeric-Fc (4 µg), and DKK2-Tg mice receiving STZ and sTie2 antibody (sTie2; 4 µg) (n = 6). *P < 0.001 vs. DKK2-Tg group. #P < 0.001 vs. WT + STZ group. †P < 0.001 vs. DKK2-Tg + STZ + Fc group. H–J: Representative ICP responses for age-matched 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 dimeric-Fc, or DKK2 protein and sTie2 (n = 5). *P < 0.001 vs. control group. #P < 0.001 vs. PBS-treated group. †P < 0.001 vs. DKK2 + Fc group. K: CD31 (green), PDGFR-β (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 h and treated with PBS, DKK2 protein (200 ng/mL), or DKK2 protein + sTie2 (100 ng/mL). Original magnification ×40. M: βIII tubulin (red) staining in MPG tissue culture. N and O: The CD31, PDGFR-β, and βIII tubulin immunopositive areas quantified by ImageJ (n = 6). *P < 0.05 vs. DKK2-Tg group. #P < 0.05 vs. WT + STZ group. †P < 0.01 vs. DKK2-Tg + STZ + Fc group. The relative ratio in the DKK2-Tg group was arbitrarily set to 1. P: Number of tubes per high-power field (n = 4). *P < 0.001 vs. NG group. #P < 0.01 vs. PBS-treated group. †P < 0.01 vs. DKK2 + Fc group. Q: Quantification of neurite length by ImageJ (n = 4). *P < 0.001 vs. NG group. #P < 0.001 vs. PBS-treated group. †P < 0.001 vs. DKK2 + Fc group. The relative ratio in the NG group was arbitrarily set to 1. P values were determined by Kruskal-Wallis test. Data in graphs are presented as the mean ± SE. DNB, dorsal nerve bundle; WB, Western blot.

Close modal

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. 5B–D).

Physiological erection studies revealed that inhibition of the Ang1-Tie2 pathway by sTie2-Fc (4 µg/20 µL) abolished DKK2-mediated erectile function recovery in both diabetic DKK2-Tg mice and diabetic WT mice treated with DKK2 protein (Fig. 5E–J). No detectable differences in MSBP were found among the experimental groups (Supplementary Tables 3 and 4).

Immunofluorescent staining of penis tissue revealed that the enhanced cavernous angiogenesis and neural regeneration were profoundly diminished in STZ-injected DKK2-Tg mice treated with sTie2-Fc (Fig. 5K, N, and O). Treatment of MCEC-MCP coculture or MPG tissue with sTie2-Fc also abolished the DKK2-mediated enhancement of tube formation and neurite sprouting under HG conditions (Fig. 5I, M, P, and Q).

We also examined the cellular sources of Ang1 that may play a major role in DKK2-mediated angiogenesis and neural regeneration. Compared with MCECs transfected with Ang1 siRNA, the transfection of Ang1 siRNA into MCPs or both MCECs and MCPs profoundly diminished the DKK2-mediated enhancement of tube formation in mixed MCEC-MCP 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, DKK2-mediated neurite sprouting was more impaired in MPG tissue treated with CM derived from Ang1 siRNA-transfected MCPs than in the tissue treated with CM derived from Ang1 siRNA-transfected MCECs (Supplementary Fig. 11C and D). These findings suggest that pericyte-derived Ang1 plays a crucial role in DKK2-mediated 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 under diabetic conditions. The treatment of diabetic mice with insulin did not restore erectile function (Supplementary Fig. 12A–F). Insulin treatment also failed to restore cavernous DKK2 expression in the diabetic mice (Supplementary Fig. 12G and H). Metabolic and physiological variables, including body weight, blood glucose concentrations, and systemic blood pressure, are summarized in Supplementary Table 7.

Here, we investigated whether DKK2 plays a role as a positive regulator of angiogenesis and neural regeneration, exerting beneficial effects in diabetic ED. DKK2-mediated interactions between pericytes and endothelial cells promoted angiogenesis and neural regeneration through an Ang1-Tie2 pathway, rescuing erectile function in diabetic animals. The detailed mechanisms of action by which DKK2 restores erectile function are illustrated in Fig. 6.

Figure 6

Schematic diagram of a proposed mechanism in which DKK2 preserves erectile function in diabetic mice. Pericyte-derived DKK2 migrates into endothelial cells (ECs). The DKK2-mediated interaction between pericytes and endothelial cells then promotes angiogenesis and neural regeneration through an Ang1-Tie2 pathway, rescuing erectile function under diabetic conditions. ECJ, endothelial cell-cell junction; eNOS, endothelial NO synthase; PC, pericyte.

Figure 6

Schematic diagram of a proposed mechanism in which DKK2 preserves erectile function in diabetic mice. Pericyte-derived DKK2 migrates into endothelial cells (ECs). The DKK2-mediated interaction between pericytes and endothelial cells then promotes angiogenesis and neural regeneration through an Ang1-Tie2 pathway, rescuing erectile function under diabetic conditions. ECJ, endothelial cell-cell junction; eNOS, endothelial NO synthase; PC, pericyte.

Close modal

To test whether DKK2 induces cavernous angiogenesis under pathological conditions, we immunohistochemically evaluated the expression of CD31 and platelet-derived growth factor receptor-β (PDGFRβ). Similar to the results of previous studies in STZ-induced 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 STZ-treated DKK2-Tg 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 cell-cell 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). DM promotes LDL oxidation and extravasation, which in turn induces vascular inflammatory responses and endothelial cell apoptosis (31). We recently revealed impaired cavernous endothelial cell-cell junctions and increased cavernous endothelial permeability to oxidized LDL in diabetic mice (32). In the current study, cavernous endothelial cell-cell junction proteins were well preserved, and less oxidized LDL was extravasated in STZ-injected DKK2-Tg mice than in STZ-injected WT littermates. The restoration of endothelial cell-cell 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 cell-pericyte 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 DKK2-RFP–transfected MCP coculture experiments, we confirmed that pericyte-derived DKK2 migrates into endothelial cells. Moreover, we found profound tube formation impairment in MCECs treated with DKK2-depleted MCP-CM, whereas the treatment of MCPs with DKK2-depleted MCEC-CM did not significantly affect tube formation. These findings suggest that pericyte-derived DKK2 may play a crucial role in DKK2-mediated angiogenesis.

The number of functional nNOS-positive neurons is critical for physiological penile erection (34). In the current study, DKK2-Tg 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 MCEC-MCP 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 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 Ang1-Tie2 pathway with sTie2-Fc diminished DKK2-mediated angiogenesis and neural regeneration in STZ-injected DKK2-Tg mice and MCECs or MPG tissue exposed to HG. Pretreatment with sTie2-Fc also abolished DKK2-mediated erectile function recovery in both STZ-injected DKK2-Tg mice and STZ-injected WT mice treated with DKK2 protein. These findings suggest that the Ang1-Tie2 pathway is crucial for DKK2-mediated restoration of the penile neurovascular structure and erectile function. Although the Ang1-Tie2 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 the transactivation of TrkA receptor (38).

For clinical applications, a Good Laboratory Practice preclinical study of DKK2 protein has begun. An interim report revealed no toxicity in C.B-17 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 wt (Supporting Data 1 in the Supplementary Data). Also, no tumorigenesis related to DKK2 protein was noted in breast cancer cell lines (MDA-MB-231, MCF-7) or a lung cancer cell line (A549) in vitro (Supporting Data 2 in the Supplementary Data) or in vivo after implantation of a lung cancer cell line (A549) in nude mice (Supporting Data 3 in the Supplementary Data). It was reported that DKK2-Tg mice exhibited increased retinal blood vessel formation during the developmental period (20). DKK2-Tg mice also showed increased blood vessel density and decreased hemorrhage in the oxygen-induced retinopathy model (39), suggesting the 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 Ang1-Tie2 pathway is involved in neural regeneration need to be documented in detail. Finally, we measured systemic blood pressure by use of a noninvasive tail-cuff 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 tail-cuff 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.

Funding. This work was supported by a grant from the Korean Health Technology R&D Project; the Ministry of Health & Welfare, Republic of Korea (grants A110076 [to J.-K.S.] and H15C0508 [to J.-K.R. and H.M.K.]); a Medical Research Center Grant (grant 2014R1A5A2009392 [to J.-K.R. and H.J.P.]); and the National Research Foundation of Korea (NRF), funded by the Korean government (Ministry of Science, ICT and Future Planning, grant 2016R1A2B2010087 [to J.-K.R.]).

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

Author Contributions. G.N.Y. and H.-R.J. designed and performed the experiments and wrote the manuscript. M.-J.C., A.L., K.G., N.N.M., J.O., M.-H.K., and K.-M.S. performed the experiments. H.J.P. analyzed and critically discussed the data. H.M.K. and Y.-G.K. contributed essential reagents. J.-K.R. and J.-K.S. designed the experiments, supervised the project, and wrote the manuscript. J.-K.R. 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.

1.
Andersson
KE
.
Mechanisms of penile erection and basis for pharmacological treatment of erectile dysfunction
.
Pharmacol Rev
2011
;
63
:
811
859
[PubMed]
2.
Yin
GN
,
Das
ND
,
Choi
MJ
, et al
.
The pericyte as a cellular regulator of penile erection and a novel therapeutic target for erectile dysfunction
.
Sci Rep
2015
;
5
:
10891
[PubMed]
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
:
54
61
[PubMed]
4.
Lue
TF
.
Erectile dysfunction
.
N Engl J Med
2000
;
342
:
1802
1813
[PubMed]
5.
Angulo
J
,
González-Corrochano
R
,
Cuevas
P
, et al
.
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
:
758
768
[PubMed]
6.
Musicki
B
,
Burnett
AL
.
Endothelial dysfunction in diabetic erectile dysfunction
.
Int J Impot Res
2007
;
19
:
129
138
[PubMed]
7.
Dall’Era
JE
,
Meacham
RB
,
Mills
JN
, et al
.
Vascular endothelial growth factor (VEGF) gene therapy using a nonviral gene delivery system improves erectile function in a diabetic rat model
.
Int J Impot Res
2008
;
20
:
307
314
[PubMed]
8.
Gholami
SS
,
Rogers
R
,
Chang
J
, et al
.
The effect of vascular endothelial growth factor and adeno-associated virus mediated brain derived neurotrophic factor on neurogenic and vasculogenic erectile dysfunction induced by hyperlipidemia
.
J Urol
2003
;
169
:
1577
1581
[PubMed]
9.
Jin
HR
,
Kim
WJ
,
Song
JS
, et al
.
Intracavernous delivery of a designed angiopoietin-1 variant rescues erectile function by enhancing endothelial regeneration in the streptozotocin-induced diabetic mouse
.
Diabetes
2011
;
60
:
969
980
[PubMed]
10.
Jin
HR
,
Kim
WJ
,
Song
JS
, et al
.
Intracavernous delivery of synthetic angiopoietin-1 protein as a novel therapeutic strategy for erectile dysfunction in the type II diabetic db/db mouse
.
J Sex Med
2010
;
7
:
3635
3646
[PubMed]
11.
Kwon
MH
,
Ryu
JK
,
Kim
WJ
, et al
.
Effect of intracavernous administration of angiopoietin-4 on erectile function in the streptozotocin-induced diabetic mouse
.
J Sex Med
2013
;
10
:
2912
2927
[PubMed]
12.
Rogers
RS
,
Graziottin
TM
,
Lin
CS
,
Kan
YW
,
Lue
TF
.
Intracavernosal vascular endothelial growth factor (VEGF) injection and adeno-associated virus-mediated VEGF gene therapy prevent and reverse venogenic erectile dysfunction in rats
.
Int J Impot Res
2003
;
15
:
26
37
[PubMed]
13.
Ryu
JK
,
Kim
WJ
,
Koh
YJ
, et al
.
Designed angiopoietin-1 variant, COMP-angiopoietin-1, rescues erectile function through healthy cavernous angiogenesis in a hypercholesterolemic mouse
.
Sci Rep
2015
;
5
:
9222
[PubMed]
14.
Yamanaka
M
,
Shirai
M
,
Shiina
H
, et al
.
Vascular endothelial growth factor restores erectile function through inhibition of apoptosis in diabetic rat penile crura
.
J Urol
2005
;
173
:
318
323
[PubMed]
15.
Bakircioglu
ME
,
Lin
CS
,
Fan
P
,
Sievert
KD
,
Kan
YW
,
Lue
TF
.
The effect of adeno-associated virus mediated brain derived neurotrophic factor in an animal model of neurogenic impotence
.
J Urol
2001
;
165
:
2103
2109
[PubMed]
16.
Bennett
NE
,
Kim
JH
,
Wolfe
DP
, et al
.
Improvement in erectile dysfunction after neurotrophic factor gene therapy in diabetic rats
.
J Urol
2005
;
173
:
1820
1824
[PubMed]
17.
Baron
R
,
Kneissel
M
.
WNT signaling in bone homeostasis and disease: from human mutations to treatments
.
Nat Med
2013
;
19
:
179
192
[PubMed]
18.
Kawano
Y
,
Kypta
R
.
Secreted antagonists of the Wnt signalling pathway
.
J Cell Sci
2003
;
116
:
2627
2634
[PubMed]
19.
Mao
B
,
Wu
W
,
Li
Y
, et al
.
LDL-receptor-related protein 6 is a receptor for Dickkopf proteins
.
Nature
2001
;
411
:
321
325
[PubMed]
20.
Min
JK
,
Park
H
,
Choi
HJ
, et al
.
The WNT antagonist Dickkopf2 promotes angiogenesis in rodent and human endothelial cells
.
J Clin Invest
2011
;
121
:
1882
1893
[PubMed]
21.
Jin
HR
,
Kim
WJ
,
Song
JS
, et al
.
Functional and morphologic characterizations of the diabetic mouse corpus cavernosum: comparison of a multiple low-dose and a single high-dose streptozotocin protocols
.
J Sex Med
2009
;
6
:
3289
3304
[PubMed]
22.
Yin
GN
,
Choi
MJ
,
Kim
WJ
, et al
.
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
:
E2731
E2740
[PubMed]
23.
Yashida
MH
,
Da Silva Faria
AL
,
Caldeira
EJ
.
Estrogen and insulin replacement therapy modulates the expression of insulin-like growth factor-I receptors in the salivary glands of diabetic mice
.
Anat Rec (Hoboken)
2011
;
294
:
1930
1938
[PubMed]
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
[PubMed]
25.
Neng
L
,
Zhang
W
,
Hassan
A
, et al
.
Isolation and culture of endothelial cells, pericytes and perivascular resident macrophage-like melanocytes from the young mouse ear
.
Nat Protoc
2013
;
8
:
709
720
[PubMed]
26.
Yin
GN
,
Ryu
JK
,
Kwon
MH
, et al
.
Matrigel-based sprouting endothelial cell culture system from mouse corpus cavernosum is potentially useful for the study of endothelial and erectile dysfunction related to high-glucose exposure
.
J Sex Med
2012
;
9
:
1760
1772
[PubMed]
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
:
631
635
[PubMed]
28.
Burchardt
T
,
Burchardt
M
,
Karden
J
, et al
.
Reduction of endothelial and smooth muscle density in the corpora cavernosa of the streptozotocin induced diabetic rat
.
J Urol
2000
;
164
:
1807
1811
[PubMed]
29.
Zhang
LW
,
Piao
S
,
Choi
MJ
, et al
.
Role of increased penile expression of transforming growth factor-beta1 and activation of the Smad signaling pathway in erectile dysfunction in streptozotocin-induced diabetic rats
.
J Sex Med
2008
;
5
:
2318
2329
[PubMed]
30.
Liebner
S
,
Cavallaro
U
,
Dejana
E
.
The multiple languages of endothelial cell-to-cell communication
.
Arterioscler Thromb Vasc Biol
2006
;
26
:
1431
1438
[PubMed]
31.
Artwohl
M
,
Graier
WF
,
Roden
M
, et al
.
Diabetic LDL triggers apoptosis in vascular endothelial cells
.
Diabetes
2003
;
52
:
1240
1247
[PubMed]
32.
Ryu
JK
,
Jin
HR
,
Yin
GN
, et al
.
Erectile dysfunction precedes other systemic vascular diseases due to incompetent cavernous endothelial cell-cell junctions
.
J Urol
2013
;
190
:
779
789
[PubMed]
33.
Armulik
A
,
Genové
G
,
Betsholtz
C
.
Pericytes: developmental, physiological, and pathological perspectives, problems, and promises
.
Dev Cell
2011
;
21
:
193
215
[PubMed]
34.
Hurt
KJ
,
Musicki
B
,
Palese
MA
, et al
.
Akt-dependent phosphorylation of endothelial nitric-oxide synthase mediates penile erection
.
Proc Natl Acad Sci U S A
2002
;
99
:
4061
4066
[PubMed]
35.
Gao
K
,
Wang
YS
,
Yuan
YJ
, et al
.
Neuroprotective effect of rapamycin on spinal cord injury via activation of the Wnt/β-catenin signaling pathway
.
Neural Regen Res
2015
;
10
:
951
957
[PubMed]
36.
Harvey
K
,
Marchetti
B
.
Regulating Wnt signaling: a strategy to prevent neurodegeneration and induce regeneration
.
J Mol Cell Biol
2014
;
6
:
1
2
[PubMed]
37.
Davis
S
,
Aldrich
TH
,
Jones
PF
, et al
.
Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning
.
Cell
1996
;
87
:
1161
1169
[PubMed]
38.
Kosacka
J
,
Figiel
M
,
Engele
J
,
Hilbig
H
,
Majewski
M
,
Spanel-Borowski
K
.
Angiopoietin-1 promotes neurite outgrowth from dorsal root ganglion cells positive for Tie-2 receptor
.
Cell Tissue Res
2005
;
320
:
11
19
[PubMed]
39.
Park
H
,
Jung
HY
,
Choi
HJ
, et al
.
Distinct roles of DKK1 and DKK2 in tumor angiogenesis
.
Angiogenesis
2014
;
17
:
221
234
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data