Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Biol Sci ; 19(9): 2663-2677, 2023.
Article in English | MEDLINE | ID: mdl-37324943

ABSTRACT

As a peripheral nerve injury disease, cavernous nerve injury (CNI) caused by prostate cancer surgery and other pelvic surgery causes organic damage to cavernous blood vessels and nerves, thereby significantly attenuating the response to phosphodiesterase-5 inhibitors. Here, we investigated the role of heme-binding protein 1 (Hebp1) in erectile function using a mouse model of bilateral CNI, which is known to promote angiogenesis and improve erection in diabetic mice. We found a potent neurovascular regenerative effect of Hebp1 in CNI mice, demonstrating that exogenously delivered Hebp1 improved erectile function by promoting the survival of cavernous endothelial-mural cells and neurons. We further found that endogenous Hebp1 delivered by mouse cavernous pericyte (MCP)-derived extracellular vesicles promoted neurovascular regeneration in CNI mice. Moreover, Hebp1 achieved these effects by reducing vascular permeability through regulation of claudin family proteins. Our findings provide new insights into Hebp1 as a neurovascular regeneration factor and demonstrate its potential therapeutic application to various peripheral nerve injuries.


Subject(s)
Diabetes Mellitus, Experimental , Erectile Dysfunction , Extracellular Vesicles , Peripheral Nerve Injuries , Animals , Humans , Male , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Erectile Dysfunction/drug therapy , Erectile Dysfunction/etiology , Extracellular Vesicles/metabolism , Heme-Binding Proteins/pharmacology , Nerve Regeneration , Penis/blood supply , Penis/innervation , Penis/surgery , Pericytes/metabolism , Peripheral Nerve Injuries/therapy
2.
Investig Clin Urol ; 62(1): 100-110, 2021 01.
Article in English | MEDLINE | ID: mdl-33258327

ABSTRACT

PURPOSE: Penile erection requires integrative interactions between vascular endothelial cells, pericytes, smooth muscle cells, and autonomic nerves. Furthermore, the importance of the role played by pericytes in the pathogenesis of angiopathy has only recently been appreciated. However, global gene expression in pericytes in diabetes mellitus-induced erectile dysfunction (DMED) remains unclear. We aimed to identify potential target genes related to DMED in mouse cavernous pericytes (MCPs). MATERIALS AND METHODS: Mouse cavernous tissue was allowed to settle under gravity in collagen I-coated dishes, and sprouted cells were subcultivated for experiments. To imitate diabetic conditions, MCPs were treated with normal-glucose (NG, 5 mM) or high-glucose (HG, 30 mM) media for 3 days. Microarray technology was used to evaluate gene expression profiles, and RT-PCR was used to validate sequencing data. Histological examinations and Western blot were used to validate final selected target genes related to DMED. RESULTS: Decreased tube formation and increased apoptosis were detected in MCPs exposed to the HG condition. As shown by microarray analysis, the gene expression profiles of MCPs exposed to the NG or HG condition differed. A total of 2,523 genes with significantly altered expression were classified into 15 major gene categories. After further screening based on gene expression and RT-PCR and histologic results, we found that Hebp1 gene expression was significantly diminished under the HG condition and in DM mice. CONCLUSIONS: This gene profiling study provides new potential targets responsible for diabetes in MCPs. Validation studies suggest that Hebp1 may be a suitable biomarker for DMED.


Subject(s)
Diabetic Angiopathies/genetics , Erectile Dysfunction/genetics , Gene Expression/drug effects , Glucose/pharmacology , Heme-Binding Proteins/genetics , Pericytes/physiology , Animals , Apoptosis/drug effects , Biomarkers , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Erectile Dysfunction/etiology , Gene Expression Profiling , Gene Ontology , Male , Mice , Mice, Inbred C57BL , Penis/blood supply , Pericytes/drug effects , Pericytes/pathology , Primary Cell Culture , Streptozocin
3.
World J Mens Health ; 38(1): 123-131, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30929324

ABSTRACT

PURPOSE: To establish a simple and nonenzymatic technique to isolate endothelial cells (ECs) and pericytes from human corpus cavernosum tissue and to evaluate the angiogenic ability of the human cavernous EC or pericytes for the study of high glucose-induced angiopathy. MATERIALS AND METHODS: For primary human cavernous EC culture, cavernous tissues were implanted into Matrigel in dishes. For primary human cavernous pericyte culture, cavernous tissues were settled by gravity into dishes. We performed immunocytochemistry and Western blot to determine phenotype and morphologic changes from passage 1 to 5. The primary cultured cells were exposed to a normal-glucose (5 mmol/L) or a high-glucose (30 mmol/L) condition, and then tube formation assay was done. RESULTS: We successfully isolated high-purity EC and pericytes from human corpus cavernosum tissue. Primary cultured EC showed highly positive staining for von Willebrand factor, and pericyte revealed positive staining for NG2 and platelet-derived growth factor receptor-ß. Primary cultured EC and pericytes maintained their cellular characteristics up to passage 2 or 3. However, we observed significant changes in their typical phenotype from the passage 4 and morphological characteristics from the passage 3. Human cavernous EC or pericytes formed well-organized capillary-like structures in normal-glucose condition, whereas severely impaired tube formation was detected in high-glucose condition. CONCLUSIONS: This study provides a simple and nonenzymatic method for primary culture of human cavernous EC and pericytes. Our study will aid us to understand the pathophysiology of diabetic erectile dysfunction, and also be a valuable tool for determining the efficacy of candidate therapeutic targets.

4.
Asian J Androl ; 20(5): 465-472, 2018.
Article in English | MEDLINE | ID: mdl-29667617

ABSTRACT

Men with diabetic erectile dysfunction (ED) respond poorly to the currently available oral phosphodiesterase-5 inhibitors. Therefore, functional therapies for diabetic ED are needed. Stromal vascular fraction (SVF) and the adenovirus-mediated cartilage oligomeric matrix angiopoietin-1 (Ad-COMP-Ang1) gene are known to play critical roles in penile erection. We previously reported that SVF and Ad-COMP-Ang1 have only a short-term effect in restoring erectile function. Further improvements to ED therapy are needed for long-lasting effects. In the present study, we aimed to test if the combination of SVF and Ad-COMP-Ang1 could extend the erection effect in diabetic ED. We found that the combination therapy showed a long-term effect in restoring erectile function through enhanced penile endothelial and neural cell regeneration. Combination therapy with SVF and Ad-COMP-Ang1 notably restored cavernous endothelial cell numbers, pericyte numbers, endothelial cell-cell junctions, decreased cavernous endothelial cell permeability, and promoted neural regeneration for at least 4 weeks in diabetic mice. In summary, this is an initial description of the long-term effect of combination therapy with SVF and Ad-COMP-Ang1 in restoring erectile function through a dual effect on endothelial and neural cell regeneration. Such combination therapy may have therapeutic potential for the treatment of diabetic ED.


Subject(s)
Angiopoietin-1/genetics , Diabetes Mellitus, Experimental/complications , Erectile Dysfunction/therapy , Genetic Therapy/methods , Mesenchymal Stem Cell Transplantation , Penile Erection/physiology , Animals , Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Erectile Dysfunction/etiology , Erectile Dysfunction/metabolism , Intercellular Junctions/metabolism , Male , Mice , Permeability
5.
Diabetes ; 67(6): 1149-1161, 2018 06.
Article in English | MEDLINE | ID: mdl-29559443

ABSTRACT

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.


Subject(s)
Angiopoietin-1/agonists , Diabetes Mellitus, Type 1/metabolism , Endothelium, Vascular/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Penis/metabolism , Pericytes/metabolism , Receptor, TIE-2/agonists , Adult , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Crosses, Genetic , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/innervation , Endothelium, Vascular/pathology , Erectile Dysfunction/complications , Erectile Dysfunction/drug therapy , Erectile Dysfunction/metabolism , Erectile Dysfunction/pathology , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/therapeutic use , Male , Mice, Inbred C57BL , Mice, Transgenic , Penis/blood supply , Penis/innervation , Penis/pathology , Pericytes/drug effects , Pericytes/pathology , Receptor, TIE-2/metabolism , Wnt Signaling Pathway , Young Adult
7.
World J Surg Oncol ; 15(1): 158, 2017 Aug 23.
Article in English | MEDLINE | ID: mdl-28835249

ABSTRACT

BACKGROUND: The efficacy of exercise training in patients with lung cancer after lung resection has not been well established yet. Therefore, we performed a meta-analysis to investigate the efficiency of exercise training in patients with lung cancer after lung resection. METHODS: Several databases were searched for eligible randomised controlled trials (RCTs). The primary outcome was quality of life, and the secondary outcomes included 6-min walk distance (6MWD), forced expiratory volume in 1 s (FEV1) and postoperative complications (POCs). Weighted mean differences (WMDs) and relative risks (RRs) with 95% confidence intervals (CIs) were calculated by random-effects model. RESULTS: Six RCTs involving 438 patients were enrolled in this meta-analysis. The pooled WMDs of the scores were 2.41 (95% CI = -5.20 to 10.02; P = 0.54) and -0.46 (95% CI = -20.52 to 19.61; P = 0.96) for the physical and mental components of the 36-item short-form scale, respectively. The pooled WMDs were 23.50 m (95% CI = -22.04 to 69.03; P = 0.31) for 6MWD and 0.03 L (95% CI = -0.19 to 0.26; P = 0.76) for FEV1. Finally, the pooled RRs were 0.79 (95% CI = 0.41 to 1.53; P = 0.49) for POCs. CONCLUSIONS: Insufficient evidence is available to support the efficacy of exercise training in patients with lung cancer after lung resection. Further studies must confirm our findings and investigate the long-term effects of exercise training on patients with lung cancer following lung resection.


Subject(s)
Exercise , Lung Neoplasms/surgery , Pneumonectomy/adverse effects , Postoperative Complications/epidemiology , Forced Expiratory Volume , Humans , Postoperative Complications/etiology , Prognosis , Quality of Life , Randomized Controlled Trials as Topic , Walk Test
8.
Sci Rep ; 5: 9222, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25783805

ABSTRACT

Despite the advent of oral phosphodiesterase-5 inhibitors, curative treatment for erectile dysfunction (ED) remains unavailable. Recently, the link between ED and cardiovascular disease was unveiled and the main etiology of ED was found to be vasculogenic. Therefore, neovascularization is a promising strategy for curing ED. Angiopoietin-1 (Ang1) is an angiogenic growth factor that promotes the generation of stable and functional vasculature. Here, we demonstrate that local delivery of the soluble, stable, and potent Ang1 variant, COMP-Ang1 gene or protein, into the penises of hypercholesterolemic mice increases cavernous angiogenesis, eNOS phosphorylation, and cGMP expression, resulting in full recovery of erectile function and cavernous blood flow up to 8 weeks after treatment. COMP-Ang1-induced promotion of cavernous angiogenesis and erectile function was abolished in Nos3(-/-) mice and in the presence of the NOS inhibitor, L-NAME. COMP-Ang1 also restored the integrity of endothelial cell-cell junction by down-regulating the expression of histone deacetylase 2 in the penis of hypercholesterolemic mice and in primary cultured mouse cavernous endothelial cells. These findings constitute a new paradigm toward curative treatment of both cavernous angiopathy and ED.


Subject(s)
Hypercholesterolemia/pathology , Penile Erection , Penis/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cells, Cultured , Cyclic GMP/metabolism , Diet, High-Fat , Disease Models, Animal , Down-Regulation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Histone Deacetylase 2/metabolism , Hypercholesterolemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Neovascularization, Pathologic , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Penile Erection/drug effects , Penis/blood supply , Phosphorylation , Recombinant Fusion Proteins/genetics , Regional Blood Flow , Tight Junction Proteins/metabolism
9.
J Sex Med ; 11(8): 1962-73, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24902866

ABSTRACT

INTRODUCTION: Erectile dysfunction (ED) is a major complication of radical prostatectomy. Men with radical prostatectomy-induced ED respond less positively to oral phosphodiesterase-5 inhibitors. AIM: The study aims to examine whether and how stromal vascular fraction (SVF) restores erectile function in mice with cavernous nerve injury (CNI). METHODS: Twelve-week-old male C57BL/6J mice were used and the animals were distributed into five groups: sham operation group and CNI group receiving a single intracavernous injection of phosphate-buffered saline (PBS) or SVF (1 × 10(4) , 1 × 10(5) , or 3 × 10(5) cells/20 µL, respectively). SVF was isolated from epididymal adipose tissues of green fluorescence protein transgenic mice. MAIN OUTCOME MEASURES: Two weeks after injection, erectile function was measured by cavernous nerve stimulation. The penis was stained with antibodies to platelet/endothelial cell adhesion molecule-1, phosphohistone H3, and phosphorylated endothelial nitric oxide synthase (phospho-eNOS). We also performed Western blot for angiopoietin-1 (Ang-1), vascular endothelial growth factor-A, hepatocyte growth factor, phospho-eNOS, and eNOS in the corpus cavernosum tissue. RESULTS: Local delivery of SVF restored erectile function in a dose-dependent manner in CNI mice. The highest erectile response was noted at a dose of 3 × 10(5) cells, for which the response was comparable with that in the sham operation group. Local delivery of SVF significantly increased the expression of angiogenic factor proteins and induced cavernous endothelial cell proliferation and eNOS phosphorylation compared with that in the PBS-treated CNI group. SVF-induced promotion of cavernous angiogenesis and erectile function was diminished in the presence of soluble antibody to Tie2, a receptor tyrosine kinase of Ang-1. CONCLUSION: Secretion of angiogenic factors from SVF is an important mechanism by which SVF induces cavernous endothelial regeneration and restores erectile function. These findings suggest that cavernous endothelial regeneration by using SVF may represent a promising treatment strategy for radical prostatectomy-induced ED.


Subject(s)
Erectile Dysfunction/therapy , Stromal Cells/transplantation , Trauma, Nervous System/physiopathology , Adipose Tissue/cytology , Angiogenesis Inducing Agents/metabolism , Angiopoietin-1/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Erectile Dysfunction/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Penis/blood supply , Penis/innervation , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Regeneration , Vascular Endothelial Growth Factor A/metabolism
10.
J Sex Med ; 11(1): 51-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24433558

ABSTRACT

INTRODUCTION: Men with erectile dysfunction (ED) respond poorly to oral phosphodiesterase-5 inhibitors following radical prostatectomy. Recent studies have reported that up-regulation of transforming growth factor-ß1 (TGF-ß1) and activation of the Smad signaling pathway play important roles in cavernous fibrosis and in the deterioration of erectile function in a mouse model of cavernous nerve injury (CNI) and in patients with spinal cord injury. The mothers against decapentaplegic homolog 7 (Smad7) is known to inhibit the phosphorylation of Smad2 and Smad3. AIM: To investigate the effectiveness of adenoviruses encoding Smad7 gene (Ad-Smad7) on erectile function in a mouse model of CNI. METHODS: Twelve-week-old C57BL/6J mice were used and distributed into 7 groups: sham operation group, untreated CNI group, and CNI groups receiving a single intracavernous injection of adenovirus encoding LacZ (1 × 10(8) virus particles [vp]/20 µL) or adenovirus encoding Smad7 (Ad-Smad7; 1 × 10(7), 1 × 10(8), 2 × 10(8), or 1 × 10(9) vp/20 µL). MAIN OUTCOME MEASURES: Two weeks after bilateral cavernous nerve crushing and treatment, erectile function was measured by electrical stimulation of the cavernous nerve. The penis was harvested for histologic examinations and Western blot analysis. RESULTS: The highest erectile response was noted in CNI mice treated with Ad-Smad7 at a dose of 1 × 10(8) vp, which reached up to 82-85% of sham control values. Local delivery of Ad-Smad7 significantly decreased endothelial cell apoptosis and the production of extracellular matrix proteins, including plasminogen activator inhibitor-1, fibronectin, collagen I, and collagen IV, and induced endothelial nitric oxide synthase phosphorylation in the corpus cavernosum tissue of CNI mice. CONCLUSION: The adenovirus-mediated gene transfer of Smad7 successfully restored erectile function by enhancing endothelial cell function and through antifibrotic effects. These findings suggest that inhibition of the TGF-ß signaling pathway by use of Smad7 may represent a promising therapeutic strategy for ED induced by radical prostatectomy.


Subject(s)
Erectile Dysfunction/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Peripheral Nerve Injuries/therapy , Smad7 Protein/genetics , Adenoviridae , Animals , Apoptosis/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Erectile Dysfunction/etiology , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Nitric Oxide Synthase Type III/metabolism , Penile Erection , Penis/innervation , Penis/pathology , Penis/surgery , Peripheral Nerve Injuries/complications , Phosphorylation , Signal Transduction , Transforming Growth Factor beta1/metabolism , Up-Regulation
11.
J Sex Med ; 10(12): 2912-27, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23937122

ABSTRACT

INTRODUCTION: Erectile dysfunction (ED) is a highly prevalent complication of diabetes, and the severity of endothelial dysfunction is one of the most important factors in reduced responsiveness to oral phosphodiesterase type 5 inhibitors. AIM: To study the effects of human angiopoietin-4 (Ang-4) protein on erectile function in diabetic mice. METHODS: Diabetes was induced by intraperitoneal injection of streptozotocin into 8-week-old C57BL/6J male mice. At 8 weeks after the induction of diabetes, the animals were divided into four groups: control nondiabetic mice and diabetic mice receiving two successive intracavernous injections of phosphate buffered saline (days -3 and 0), a single intracavernous injection of Ang-4 protein (day 0), or two successive intracavernous injections of Ang-4 protein (days -3 and 0). MAIN OUTCOME MEASURES: One week after treatment, we measured erectile function by electrical stimulation of the cavernous nerve. The penis was harvested and stained with hydroethidine or antibodies to Ang-4, platelet/endothelial cell adhesion molecule-1, and phosphorylated endothelial nitric oxide synthase (eNOS). We also determined the differential expression of Ang-4 in cavernous tissue in the control and diabetic mice. The effect of Ang-4 protein on the phosphorylation of Tie-2, Akt, and eNOS was determined in human umbilical vein endothelial cells (HUVECs) by Western blot. RESULTS: The cavernous expression of Ang-4 was downregulated in diabetic mice; Ang-4 was mainly expressed in endothelial cells. Local delivery of Ang-4 protein significantly increased cavernous endothelial content, induced eNOS phosphorylation, and decreased the generation of superoxide anion and apoptosis in diabetic mice. Ang-4 protein strongly increased the phosphorylation of Tie-2, Akt, and eNOS in HUVECs. Repeated intracavernous injections of Ang-4 induced significant restoration of erectile function in diabetic mice (87% of control values), whereas a single intracavernous injection of Ang-4 protein elicited modest improvement. CONCLUSIONS: Cavernous endothelial regeneration by use of Ang-4 protein may have potential for the treatment of vascular disease-induced ED, such as diabetic ED.


Subject(s)
Angiopoietins/administration & dosage , Diabetes Mellitus, Experimental/complications , Erectile Dysfunction/drug therapy , Penile Erection/drug effects , Penis/drug effects , Angiopoietin-1/metabolism , Angiopoietin-1/pharmacology , Angiopoietin-1/therapeutic use , Angiopoietins/metabolism , Animals , Diabetes Mellitus, Experimental/physiopathology , Erectile Dysfunction/etiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type III/physiology , Penile Erection/physiology , Penis/blood supply , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/drug effects
12.
J Sex Med ; 10(6): 1488-501, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23551591

ABSTRACT

INTRODUCTION: Radical prostatectomy for prostate cancer can not only induce cavernous nerve injury (CNI) but also result in structural changes in the cavernous tissues. Nerve injury-induced protein 1, Ninjurin-1 (Ninj1), is known to be involved in neuroinflammatory processes and to be related to vascular regression during the embryonic period. AIM: The study aims to determine whether and how Ninj1 neutralizing antibody (Ninj1-Ab) restores erectile function in mice with CNI. METHODS: Twelve-week-old C57BL/6J mice were used and distributed into four groups: sham operation group and CNI groups receiving a single intracavernous injection of immunoglobulin G (IgG) control antibody, low-dose Ninj1-Ab (1.0 µg/20 µL), or high-dose Ninj1-Ab (2.5 µg/20 µL). MAIN OUTCOME MEASURES: One week after bilateral cavernous nerve crush, erectile function was measured by electrical stimulation of the cavernous nerve. The penis was harvested for histologic examinations and Western blot analysis. RESULTS: The cavernous expression of Ninj1 protein was upregulated up to 7 days after CNI and returned to baseline levels thereafter. Local delivery of Ninj1-Ab significantly increased penile neuronal nitric oxide synthase and neurofilament contents, induced cavernous endothelial proliferation and phosphorylation of Akt and endothelial nitric oxide synthase, and decreased endothelial cell apoptosis in the CNI mice by upregulating angiopoietin-1 and downregulating angiopoietin-2. High-dose Ninj1-Ab induced profound restoration of erectile function in the CNI mice (91% of sham control values), whereas low-dose Ninj1-Ab elicited partial improvement. CONCLUSION: The dual neurotrophic and angiogenic effects of Ninj1 blockade may provide a good opportunity for treating erectile dysfunction resulting from radical prostatectomy.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Erectile Dysfunction/drug therapy , Nerve Growth Factors/antagonists & inhibitors , Penile Erection/drug effects , Penis/drug effects , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Antibodies, Neutralizing/administration & dosage , Cell Adhesion Molecules, Neuronal/immunology , Cell Proliferation/drug effects , Disease Models, Animal , Electric Stimulation , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Erectile Dysfunction/metabolism , Fibrosis , Injections , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Nerve Crush , Nerve Growth Factors/immunology , Nerve Regeneration/drug effects , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Penis/innervation , Penis/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
13.
J Sex Med ; 10(12): 2928-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23578329

ABSTRACT

INTRODUCTION: Much attention has recently been focused on therapeutic angiogenesis as a treatment for erectile dysfunction (ED). The apelin and apelin receptor (APJ) system is known to cause endothelium-dependent vasodilatation and to be involved in angiogenesis. AIM: To examine the differential expression of apelin and APJ in animal models of vasculogenic ED and to determine whether and how enhancement of apelin-APJ signaling restores erectile function in hypercholesterolemic mice. METHODS: Acute cavernous ischemia was induced in C57BL/6J mice by bilateral occlusion of internal iliac arteries, and chronic vasculogenic ED was induced by feeding a high-cholesterol diet or by intraperitoneal injection of streptozotocin. MAIN OUTCOME MEASURES: Messenger RNA (mRNA) levels of apelin and APJ were determined in cavernous tissue of each vasculogenic ED model by semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR). We evaluated erectile function by electrical stimulation of the cavernous nerve in hypercholesterolemic mice 1, 3, 7, and 14 days after a single intracavernous injection of apelin protein (5 µg/20 µL). The penis was harvested for histologic examinations and Western blot analysis. RESULTS: The cavernous mRNA expression of apelin and APJ was up-regulated in acute ischemia model and down-regulated in chronic vasculogenic ED models. A significant restoration of erectile function was noted 1 day after injection of apelin protein into the penis of hypercholesterolemic mice; however, erectile function returned to baseline values thereafter. The beneficial effects of apelin on erectile function resulted mainly from an activation of endothelial nitric oxide synthase and increase in nitric oxide bioavailability through reduction in reactive oxygen species-mediated endothelial apoptosis rather than through direct endothelial cell proliferation. CONCLUSION: These findings suggest that apelin-APJ signaling is a potential therapeutic target in the treatment of vasculogenic ED. Further studies are needed to develop a potent agonist for APJ and to determine the role of repeated dosing of apelin on long-term recovery of erectile function.


Subject(s)
Impotence, Vasculogenic/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Penile Erection , Penis/blood supply , Receptors, G-Protein-Coupled/biosynthesis , Adipokines , Animals , Apelin , Apelin Receptors , Cell Proliferation , Disease Models, Animal , Endothelium, Vascular/metabolism , Impotence, Vasculogenic/drug therapy , Intercellular Signaling Peptides and Proteins/genetics , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Up-Regulation
14.
J Urol ; 190(2): 779-89, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23454152

ABSTRACT

PURPOSE: Erectile dysfunction is often a harbinger of cardiovascular disease. We sought to gain mechanistic insight at the cellular and molecular levels into why erectile dysfunction precedes the clinical consequences of cardiovascular disease. MATERIALS AND METHODS: Diabetes was induced by intraperitoneal streptozotocin injection in 8-week-old C57BL/6J mice. At 8 weeks after diabetes induction, we determined the expression of endothelial cell-cell junction proteins and vascular endothelial permeability in the penis, heart and hind limb by systemic injection of various vascular space markers (350 Da to 2,000 kDa) or by immunohistochemical staining with antibody to oxidized low density lipoprotein. We also investigated the effect of recombinant Ang1 protein on cavernous endothelial permeability. RESULTS: Alterations in the integrity of the endothelial cell-cell junction, including a decrease in endothelial cell-cell junction proteins and an increase in vascular permeability to fluorescent tracers or oxidized low density lipoprotein, were prominent in the cavernous tissue of diabetic mice. In contrast, no significant changes in endothelial cell-cell junction proteins or vascular permeability were noted in heart or hind limb tissue according to the diabetic condition. Intracavernous injection of Ang1 protein, an anti-permeability factor, significantly decreased cavernous endothelial permeability to oxidized low density lipoprotein by restoring endothelial cell-cell junction proteins in diabetic mice. CONCLUSIONS: The incompetent cavernous endothelial cell-cell junction in the diabetic condition provides an important clue to why erectile dysfunction is highly prevalent and often precedes other systemic vascular diseases.


Subject(s)
Cardiovascular Diseases/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Endothelium, Vascular/physiopathology , Erectile Dysfunction/physiopathology , Intercellular Junctions/physiology , Analysis of Variance , Angiopoietin-1/pharmacology , Animals , Blotting, Western , Coronary Circulation , Hindlimb/blood supply , Male , Mice , Mice, Inbred C57BL , Penis/blood supply , Statistics, Nonparametric
15.
J Sex Med ; 9(12): 3051-65, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23088258

ABSTRACT

INTRODUCTION: Men with diabetic erectile dysfunction (ED) often have severe endothelial dysfunction and respond poorly to oral phosphodiesterase-5 inhibitors. AIM: To examine whether and how freshly isolated stromal vascular fraction (SVF) promotes cavernous endothelial regeneration and restores erectile function in diabetic animals. METHODS: Eight-week-old C57BL/6J mice were used. Diabetes was induced by intraperitoneal injection of streptozotocin. SVF was isolated from epididymal adipose tissues of green fluorescence protein transgenic mice. At 8 weeks after the induction of diabetes, the animals were divided into six groups: controls, diabetic mice, and diabetic mice treated with a single intracavernous injection of phosphate-buffered saline (PBS) or SVF (1 × 10(4) cells, 1 × 10(5) cells, or 2 × 10(5) cells/20 µL, respectively). MAIN OUTCOME MEASURES: Two weeks later, erectile function was measured by cavernous nerve stimulation. The penis was stained with antibodies to CD31, CD34, phosphohistone H3, phospho-endothelial nitric oxide synthase (eNOS), and vascular endothelial growth factor-A (VEGF-A). We also performed Western blot for phospho-eNOS and eNOS, and determined cyclic guanosine monophosphate (cGMP) concentration in the corpus cavernosum tissue. RESULTS: Significant improvement in erectile function was noted in diabetic mice treated with SVF at concentrations of 1 × 10(5) and 2 × 10(5) cells, which reached up to 82% of the control values. Local delivery of SVF significantly increased cavernous endothelial cell proliferation, eNOS phosphorylation, and cGMP expression compared with that in the untreated group and the PBS-treated diabetic group. Intracavernous injection of SVF increased cavernous VEGF-A expression and induced recruitment of CD34(+)CD31(-) progenitor cells. Some SVF underwent differentiation into cavernous endothelial cells. SVF-induced promotion of cavernous angiogenesis and erectile function was abolished in the presence of VEGF-Trap, a soluble VEGF-A neutralizing antibody. CONCLUSION: The results support the concept of cavernous endothelial regeneration by use of SVF as a curative therapy for diabetic ED.


Subject(s)
Adipose Tissue/transplantation , Endothelium, Vascular/physiology , Penile Erection/physiology , Penis/surgery , Regeneration , Stromal Cells/transplantation , Adipose Tissue/cytology , Animals , Antigens, CD34/metabolism , Cell Differentiation , Cell Proliferation , Cyclic GMP/metabolism , Diabetes Mellitus, Experimental , Endothelial Cells/cytology , Epididymis/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/physiology , Penis/blood supply , Penis/metabolism , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
J Sex Med ; 9(7): 1760-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22548733

ABSTRACT

INTRODUCTION: A proper cavernous endothelial cell culture system would be advantageous for the study of the pathophysiologic mechanisms involved in endothelial dysfunction and erectile dysfunction (ED). AIM: To establish a nonenzymatic technique, which we termed the "Matrigel-based sprouting endothelial cell culture system," for the isolation of mouse cavernous endothelial cells (MCECs) and an in vitro model that mimics in vivo situation for diabetes-induced ED. METHODS: For primary MCEC culture, mouse cavernous tissue was implanted into Matrigel and sprouting cells from the tissue were subcultivated. To establish an in vitro model for diabetes-induced ED, the primary cultured MCECs were exposed to a normal-glucose (5 mmoL) or a high-glucose (30 mmoL) condition for 48 hours. MAIN OUTCOME MEASURES: The purity of isolated cells was determined by fluorescence-activated cell sorting analysis. MCECs incubated under the normal- or the high-glucose condition were used for Western blot, cyclic guanosine monophosphate (cGMP) quantification, and in vitro angiogenesis assay. RESULTS: We could consistently isolate high-purity MCECs (about 97%) with the Matrigel-based sprouting endothelial cell culture system. MCECs were subcultured up to the fifth passage and no significant changes were noted in endothelial cell morphology or purity. The phosphorylation of Akt and eNOS and the cGMP concentration were significantly lower in MCECs exposed to high glucose than in those exposed to normal glucose. MCECs exposed to the normal-glucose condition formed well-organized capillary-like structures, whereas derangements in tube formation were noted in MCECs exposed to high glucose. The protein expression of transforming growth factor-ß1 (TGF-ß1) and phospho-Smad2 was significantly increased by exposure to high glucose. CONCLUSION: The Matrigel-based sprouting endothelial cell culture system is a simple, technically feasible, and reproducible technique for isolating pure cavernous endothelial cells in mice. An in vitro model for diabetic ED will be a valuable tool for evaluating the angiogenic potential of novel endogenous or synthetic modulators.


Subject(s)
Erectile Dysfunction/physiopathology , Glucose/pharmacology , Penis/cytology , Animals , Apoptosis/drug effects , Cells, Cultured , Culture Media , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Erectile Dysfunction/chemically induced , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Penis/drug effects , Superoxides/metabolism
17.
J Androl ; 33(5): 845-53, 2012.
Article in English | MEDLINE | ID: mdl-22403284

ABSTRACT

Cavernous hypoxia is an important factor in the pathogenesis of vasculogenic erectile dysfunction (ED). Therefore, the hypoxia-inducible gene expression system can be exploited as gene therapy for vasculogenic ED. This study was undertaken to examine the effectiveness of a hypoxia-inducible gene expression system, namely, the RTP801 promoter or the erythropoietin enhancer, in a mouse model of hypercholesterolemic ED in vivo and in primary cultured mouse cavernous endothelial cells in vitro. Two-month-old male C57BL/6 mice were fed a diet containing 4% cholesterol and 1% cholic acid, and age-matched control animals were fed a normal diet for 3 months. Mouse cavernous endothelial cells were isolated and cultured under normoxic or hypoxic conditions. After treatment of animals or endothelial cells with pSV-Luc, pRTP801-Luc, or pEpo-SV-Luc vector, gene expression was evaluated by luciferase assay, and the gene expression area was evaluated by immunohistochemistry. Plasmids pRTP801-Luc and pEpo-SV-Luc induced gene expression significantly in the hypercholesterolemic mice and in cavernous endothelial cells under hypoxia, and the highest gene expression was noted in the group treated with pEpo-SV-Luc. Gene expression was higher for more than 7 days in the hypercholesterolemic mice injected with pEpo-SV-Luc than in mice injected with pSV-Luc. As shown by immunohistochemistry, the gene expression area was also greater in the pEpo-SV-Luc group than in the pSV-Luc group, but the difference was not as great as that in luciferase activity. The hypoxia-specific gene expression system could be a valuable tool for facilitating gene delivery into ischemic corpus cavernosum tissue resulting from vascular causes.


Subject(s)
Cholesterol, Dietary , Enhancer Elements, Genetic , Erythropoietin/genetics , Genetic Therapy , Hypercholesterolemia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Impotence, Vasculogenic/therapy , Penile Erection , Penis/blood supply , Penis/physiopathology , Adaptor Proteins, Signal Transducing , Animals , Blood Vessels/metabolism , Cell Hypoxia/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Genes, Reporter , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Impotence, Vasculogenic/etiology , Impotence, Vasculogenic/genetics , Impotence, Vasculogenic/metabolism , Impotence, Vasculogenic/physiopathology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Time Factors , Transcription Factors/genetics , Transfection , Up-Regulation
18.
Diabetes ; 60(3): 969-80, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21270241

ABSTRACT

OBJECTIVE: Patients with diabetic erectile dysfunction often have severe endothelial dysfunction and respond poorly to oral phosphodiesterase-5 inhibitors. We examined the effectiveness of the potent angiopoietin-1 (Ang1) variant, cartilage oligomeric matrix protein (COMP)-Ang1, in promoting cavernous endothelial regeneration and restoring erectile function in diabetic animals. RESEARCH DESIGN AND METHODS: Four groups of mice were used: controls; streptozotocin (STZ)-induced diabetic mice; STZ-induced diabetic mice treated with repeated intracavernous injections of PBS; and STZ-induced diabetic mice treated with COMP-Ang1 protein (days -3 and 0). Two and 4 weeks after treatment, we measured erectile function by electrical stimulation of the cavernous nerve. The penis was harvested for histologic examinations, Western blot analysis, and cGMP quantification. We also performed a vascular permeability test. RESULTS: Local delivery of the COMP-Ang1 protein significantly increased cavernous endothelial proliferation, endothelial nitric oxide (NO) synthase (NOS) phosphorylation, and cGMP expression compared with that in the untreated or PBS-treated STZ-induced diabetic group. The changes in the group that received COMP-Ang1 restored erectile function up to 4 weeks after treatment. Endothelial protective effects, such as marked decreases in the expression of p47(phox) and inducible NOS, in the generation of superoxide anion and nitrotyrosine, and in the number of apoptotic cells in the corpus cavernosum tissue, were noted in COMP-Ang1-treated STZ-induced diabetic mice. An intracavernous injection of COMP-Ang1 completely restored endothelial cell-cell junction proteins and decreased cavernous endothelial permeability. COMP-Ang1-induced promotion of cavernous angiogenesis and erectile function was abolished by the NOS inhibitor, N-nitro-L-arginine methyl ester, but not by the NADPH oxidase inhibitor, apocynin. CONCLUSIONS: These findings support the concept of cavernous endothelial regeneration by use of the recombinant Ang1 protein as a curative therapy for diabetic erectile dysfunction.


Subject(s)
Angiopoietin-1/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Impotence, Vasculogenic/drug therapy , Penis/metabolism , Recombinant Fusion Proteins/therapeutic use , Analysis of Variance , Angiopoietin-1/metabolism , Animals , Blotting, Western , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Endothelium/metabolism , Immunohistochemistry , Impotence, Vasculogenic/etiology , Impotence, Vasculogenic/metabolism , Male , Mice , Nitric Oxide Synthase Type III/metabolism
19.
J Sex Med ; 8(5): 1454-62, 2011 May.
Article in English | MEDLINE | ID: mdl-20946170

ABSTRACT

INTRODUCTION: Transforming growth factor-ß1 (TGF-ß1) is implicated in bladder fibrosis after spinal cord injury (SCI) and in the fibrosis in the corpus cavernosum tissue after cavernous nerve injury. AIM: We investigated the differential expression of TGF-ß1 and the Smad transcription factor, the key molecule for the initiation of TGF-ß-mediated fibrosis, in cavernous tissue from SCI patients. METHODS: After obtaining informed consent and approval from the patients and our institutional review board, we enrolled 5 patients with psychogenic erectile dysfunction (ED) (mean age 36.8 years; range 20-50 years) and 10 patients with neurogenic ED from SCI (mean age 38.8 years; range 18-50 years). Cavernous tissues were obtained by percutaneous biopsy and stained with Masson trichrome, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL), or antibodies to TGF-ß1 and phospho-Smad2. MAIN OUTCOME MEASURES: Semi-quantitative analysis of TGF-ß1 and phospho-Smad2 was performed, and the numbers of apoptotic cells were counted. We also quantified the cavernous collagen area with the use of an image analyzer system. RESULTS: The expression of TGF-ß1 and phospho-Smad2 protein was significantly higher in the SCI group than in the psychogenic group. The TUNEL assay revealed a higher apoptotic index in the SCI group than in the psychogenic group. Higher TGF-ß1 and phospho-Smad2 expression and more apoptotic cells were noted mainly in endothelial cells, smooth muscle cells, and fibroblasts of the SCI group. Double labeling of cavernous tissue with TUNEL and antibody to phospho-Smad2 revealed that most TUNEL-positive cells showed immunoreactivity to phospho-Smad2 staining. Cavernous collagen content was significantly greater in the SCI group than in the psychogenic group. CONCLUSION: Upregulation of TGF-ß1 and activation of the Smad signaling pathway may play important roles in SCI-induced cavernous fibrosis and deterioration of erectile function, which warrants early pharmacological intervention to protect erectile tissue from irreversible damage.


Subject(s)
Erectile Dysfunction/etiology , Penis/metabolism , Signal Transduction , Smad2 Protein/metabolism , Spinal Cord Injuries/complications , Transforming Growth Factor beta1/metabolism , Adolescent , Adult , Erectile Dysfunction/metabolism , Humans , In Situ Nick-End Labeling , Male , Middle Aged , Penis/chemistry , Smad2 Protein/analysis , Transforming Growth Factor beta1/analysis , Young Adult
20.
J Sex Med ; 7(10): 3351-64, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20646178

ABSTRACT

INTRODUCTION: With the advent of genetically engineered mice, it seems important to develop a mouse model of cavernous nerve injury (CNI). AIM: To establish a mouse model of CNI induced either by nerve crushing or by neurectomy and to evaluate time-dependent derangements in penile hemodynamics in vivo and subsequent histologic alterations in the cavernous tissue. METHODS: Twelve-week-old C57BL/6J mice were divided into 4 groups (N=36 per group): control, sham operation, bilateral cavernous nerve crush, and bilateral cavernous neurectomy group. MAIN OUTCOME MEASURES: Three days and 1, 2, 4, 8, and 12 weeks after CNI, erectile function was measured by electrical stimulation of the cavernous nerve. The penis was then harvested and TUNEL was performed. Immunohistochemical analysis was performed assaying for caspase-3, transforming growth factor-ß1 (TGF-ß1), phospho-Smad2, PECAM-1, factor VIII, and smooth muscle α-actin. The numbers of apoptotic cells and phospho-Smad2-immunopositive cells in endothelial cells or smooth muscle cells were counted. RESULTS: Erectile function was significantly less in the cavernous nerve crushing and neurectomy groups than in the control or sham group. This difference was observed at the earliest time point assayed (day 3) and persisted up to 4 weeks after nerve crushing and to 12 weeks after neurectomy. The apoptotic index peaked at 1 or 2 weeks after CNI and decreased thereafter. Cavernous TGF-ß1 and phospho-Smad expression was also increased after CNI. The numbers of apoptotic cells and phospho-Smad2-immunopositive cells in cavernous endothelial cells and smooth muscle cells were significantly greater in the cavernous nerve crush and cavernous neurectomy groups than in the control or sham group. Conclusion. The mouse is a useful model for studying pathophysiologic mechanisms involved in erectile dysfunction after CNI. Early intervention to prevent apoptosis in smooth muscle cells and endothelial cells or to inhibit cavernous tissue fibrosis is required to restore erectile function.


Subject(s)
Erectile Dysfunction/etiology , Penis/innervation , Animals , Apoptosis/physiology , Blotting, Western , Disease Models, Animal , Erectile Dysfunction/metabolism , Erectile Dysfunction/pathology , Erectile Dysfunction/physiopathology , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Penile Erection/physiology , Penis/injuries , Penis/metabolism , Penis/pathology , Penis/physiopathology , Smad2 Protein/metabolism , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...