Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Oxid Med Cell Longev ; 2022: 6831779, 2022.
Article in English | MEDLINE | ID: mdl-35154570

ABSTRACT

Aging has been deemed the primary factor in erectile dysfunction (ED). Herein, age-related changes in the erectile response and histomorphology were detected, and the relationship between aging and ED was investigated based on gene expression levels. Thirty male Sprague-Dawley (SD) rats were randomly divided into 6 groups, and intracavernous pressure (ICP) and mean arterial pressure (MAP) were measured. Subsequently, the corpus cavernosum (CC) was harvested and prepared for histological examinations of apoptosis, oxidative stress (OS), and fibrosis. Then, the microarray dataset (GSE10804) was analyzed to identify differentially expressed genes (DEGs) in ED progression, and hub genes were selected. In addition, aged CC smooth muscle cells (CCSMCs) were isolated to evaluate the function of the hub gene by siRNA interference, qRT-PCR, immunofluorescence staining, enzyme-linked immunosorbent assay, western blot analysis, CCK-8 assay, EdU staining, and flow cytometry approaches. The ICP/MAP and smooth muscle cell (SMC)/collagen ratios declined with aging, while apoptosis and OS levels increased with aging. The enriched functions and pathways of the DEGs were investigated, and 15 hub genes were identified, among which IGFBP3 was significantly upregulated. The IGFBP3 upregulation was verified in the CC of aging rats. Furthermore, aged CCSMCs were transfected with siRNA to knock down IGFBP3 expression. The viability and proliferation of the CCSMCs increased, while apoptosis, OS, and fibrosis decreased. Our findings demonstrate that the erectile response of SD rats declines in parallel with enhanced CC apoptosis, OS, and fibrosis with aging. Upregulation of IGFBP3 plays an important role; furthermore, downregulation of IGFBP3 improves the viability and proliferation of CCSMCs and alleviates apoptosis, OS, and fibrosis. Thus, IGFBP3 is a potential therapeutic target for age-related ED.


Subject(s)
Aging/metabolism , Apoptosis/genetics , Erectile Dysfunction/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Oxidative Stress/genetics , Signal Transduction/genetics , Up-Regulation/genetics , Animals , Cells, Cultured , Disease Models, Animal , Down-Regulation/genetics , Fibrosis , Gene Knockdown Techniques/methods , Insulin-Like Growth Factor Binding Protein 3/genetics , Male , Penile Erection/genetics , Rats , Rats, Sprague-Dawley , Transfection
2.
Zhonghua Nan Ke Xue ; 26(3): 223-227, 2020 Mar.
Article in Chinese | MEDLINE | ID: mdl-33346961

ABSTRACT

OBJECTIVE: To investigate the expression of the kallistatin gene in the corpus cavernosum and search for some new molecular targets for the regulation of penile erectile function and treatment of ED. METHODS: Using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR), Western blot and immunofluorescence staining, we detected the expression of kallistatin in the rat corpus cavernosum and compared it with that in the aorta. RESULTS: The results of RT-qPCR and Western blot revealed both mRNA and protein expressions of kallistatin in the rat corpus cavernosal tissue, with no statistically significant difference from those in the aorta (P > 0.05). Immunofluorescence staining showed that kallistatin was expressed in both endothelial and smooth muscle cells in the corpus cavernosum and localized in the cytoplasm, with no statistically significant difference from its expression in the aorta (P > 0.05) either. CONCLUSIONS: The kallistatin gene is highly expressed in the corpus cavernosum and localized in cavernosal endothelial and smooth muscle cells, suggestive of its involvement in the cellular function of cavernosal endothelial and smooth muscle cells and its participation in the regulation of penile erectile function.


Subject(s)
Penile Erection/genetics , Penis/metabolism , Serpins/genetics , Animals , Aorta , Blotting, Western , Endothelial Cells/metabolism , Erectile Dysfunction/genetics , Male , Myocytes, Smooth Muscle/metabolism , Rats
3.
Urology ; 133: 249.e1-249.e7, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31476348

ABSTRACT

OBJECTIVE: To investigate the relationship between the upregulated expression of sphingosine-1-phosphate receptor 1 (S1P1) in the corpus cavernosum and erectile function in spontaneously hypertensive rats (SHRs). METHODS: Twelve-week-old healthy male Wistar-Kyoto rats (WKY) and SHR rats were randomly divided into 4 groups: WKY, SHR, WKY transfection, and SHR transfection (n = 5). A lentiviral vector carrying the S1P1 gene was injected into the corpus cavernosum penis of rats in the transfection groups (1 × 109 TU/mL, 20 µL). After 1 week, the maximum penile intracavernous pressure/mean arterial pressure (ICPmax/MAP), nitric oxide (NO) content, and the expression of eNOS, P-eNOS, ROCK1, ROCK2, and S1P1 in the corpus cavernosum penis of rats in each group were measured. RESULTS: The ICPmax/MAP value was significantly higher in the SHR transfection group than in the SHR group under 3-V and 5-V electrical stimulations (P <.01). The expression of S1P1 and P-eNOS proteins significantly increased (P <.01), while that of ROCK1 and ROCK2 proteins significantly decreased (P <.01) in the SHR transfected group compared with the SHR group. The NO content was significantly higher in the SHR transfection group than in the SHR group (P <.01). CONCLUSION: The upregulated expression of S1P1 in SHR corpus cavernosum penis may improve the SHR erectile function by upregulating the P-eNOS/eNOS ratio and inhibiting the RhoA/Rho kinase signaling pathway.


Subject(s)
Penile Erection/genetics , Sphingosine-1-Phosphate Receptors/genetics , Animals , Male , Random Allocation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Transfection
4.
Stem Cell Res Ther ; 10(1): 208, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31311594

ABSTRACT

BACKGROUND: Stem cell therapy has revealed a promising future for treating erectile dysfunction (ED), but the fate and curative mechanism of intracavernosal transplanted stem cells are under further exploration. This study aimed to demonstrate the effects of myocardin gene modification on improving erectile function and prolonging the retention of implanted adipose-derived stem cells (ASCs) using in vivo small animal imaging. METHODS: ASCs were isolated, cultured, and identified by flow cytometry and osteogenic and adipogenic induction. The effects of gene modification on cell proliferation, apoptosis, and contraction were determined by CCK-8, EdU, flow cytometry, and collagen gel lattice contraction assays as well as confocal microscopy. A total of 20 normal and 60 diabetes mellitus ED to (DMED) Sprague-Dawley rats were recruited to the 7 day and 21 day groups. Each group contained subgroups of 10 rats each: the negative control (NC), DMED + ASCs plus Ad-Luc-Myocardin, DMED + ASCs plus Ad-Luc, and DMED + phosphate buffer solution (PBS) groups. Erectile function was evaluated with the intracavernosal pressure/mean arterial pressure (△ICP/MAP) ratio. In vivo small animal imaging and an EdU cell tracking strategy were introduced to detect the transplanted ASCs, and IHC and WB were performed to assess smooth muscle cell protein levels. RESULTS: The ASCs expressed high CD29 and CD90 and scant CD45, while the multi-induction potential was verified by oil red O and alizarin red staining. Gene transfection of myocardin had no significant influence on ASC apoptosis but inhibited cell proliferation and promoted cell contraction. Myocardin combined with ASCs enhanced the therapeutic potential of ASCs for improving the △ICP/MAP ratio as well as α-SMA and calponin expression. In vivo imaging confirmed that ASCs resided within the cavernous body in 21 days, while only a few red EdU dots were detected. CONCLUSIONS: Myocardin induced ASC differentiation towards smooth muscle-like cells and enhanced the therapeutic potential of ASCs for ameliorating ED in STZ-induced diabetic rats. Notably, in vivo small animal tracking was an effective strategy for monitoring the implanted stem cells, and this strategy might have advantages over traditional EdU assays.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Erectile Dysfunction/therapy , Mesenchymal Stem Cell Transplantation , Nuclear Proteins/genetics , Trans-Activators/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Erectile Dysfunction/genetics , Erectile Dysfunction/pathology , Humans , Male , Mesenchymal Stem Cells/metabolism , Muscle, Smooth/metabolism , Nuclear Proteins/therapeutic use , Penile Erection/genetics , Penile Erection/physiology , Rats , Rats, Sprague-Dawley , Trans-Activators/therapeutic use
5.
Clin Sci (Lond) ; 132(20): 2175-2188, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30232174

ABSTRACT

Increased production of reactive oxygen species (ROS) and inflammation are major contributors to the development and progression of diabetes-associated erectile dysfunction (DMED). As an endogenous antioxidant and anti-inflammatory factor, the potential implication of pigment epithelium-derived factor (PEDF) in DMED has not been revealed. To assess the potential antioxidant and anti-inflammatory functions of PEDF in DMED, we first demonstrated that PEDF was significantly decreased at the levels of the mRNA and protein in the penis of diabetic rats compared with normal controls. To test the hypothesis that decreased the penile levels of PEDF are associated with oxidative stress and inflammation in DMED, an adenovirus expressing PEDF (Ad-PEDF) or the same titer of control virus (Ad-GFP) was intracavernously administered at 2 weeks after diabetic onset. After 6 weeks of treatment, we found that administration of Ad-PEDF could significantly increase erectile response to cavernosal nerve stimulation in the diabetic rats by restoring the endothelial NO synthase (eNOS), P-eNOS, and neuronal NO synthase (nNOS) protein levels to the standard levels represented in normal rats and by suppressing the levels of tumor necrosis factor-α (TNF-α) and oxidative stress. In conclusion, the present data indicated that the antioxidant and anti-inflammatory potential of PEDF plays important role in restoring erectile function by the inhibition of oxidative stress and TNF-α production.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Eye Proteins/genetics , Nerve Growth Factors/genetics , Penile Erection/genetics , Penis/metabolism , Serpins/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Down-Regulation , Eye Proteins/metabolism , Gene Expression Regulation , Male , Nerve Growth Factors/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Serpins/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Andrology ; 6(6): 865-873, 2018 11.
Article in English | MEDLINE | ID: mdl-30101547

ABSTRACT

BACKGROUND: No study has assessed the possible involvement of endothelial nitric oxide synthase (eNOS) T-786C and G894T and G-protein ß3 subunit (GNB3) C825T polymorphisms with susceptibility to diabetic vasculogenic erectile dysfunction (VED) in North African subjects. OBJECTIVES: Our aim was to evaluate the interaction and association between these gene polymorphisms and this disorder. MATERIALS AND METHODS: A total of 164 type 2 diabetes patients with VED diagnosed with penile color Doppler ultrasonography and 148 age-matched healthy volunteers were genotyped for the rs1799983 (G894T) and rs2070744 (T-786C) of the eNOS gene and the rs5443 (C825T) of the GNB3 gene using the PCR-RFLP method. RESULTS: A significant association of the eNOS G894T (p = 0.005) and T-786C (p = 0.02) with altered susceptibility to VED was observed. The risk also holds for the G894T and T-786C eNOS gene polymorphisms when excluding patients with dyslipidemia and cardiovascular diseases (p = 1.7·10-4 and p = 3.2·10-5 , respectively). The univariate odds ratio associated with CC alleles of the eNOS T-786C revealed a four times increased risk for VED (OR = 4.04; 95% CI = 1.53-10.67; p = 0.006). VED risk was also associated with the G894T variant under dominant model (p = 0.002) and the T-786C variant under recessive model (p = 0.004). Furthermore, the concomitant presence of the combined genotypes of the 894T and 786T strongly affected the predisposition to VED (p = 0.007). DISCUSSION AND CONCLUSION: Our study gave a comprehensive insight into functional interaction between GNB3 and eNOS gene polymorphisms and suggests that the eNOS G894T and T-786C variants are strong predisposing factors of VED susceptibility within men with type 2 diabetes.


Subject(s)
Diabetic Angiopathies/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Impotence, Vasculogenic/genetics , Nitric Oxide Synthase Type III/genetics , Penile Erection/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Diabetic Angiopathies/diagnostic imaging , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/physiopathology , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Heredity , Humans , Impotence, Vasculogenic/diagnostic imaging , Impotence, Vasculogenic/enzymology , Impotence, Vasculogenic/physiopathology , Male , Middle Aged , Phenotype , Risk Assessment , Risk Factors , Tunisia , Ultrasonography, Doppler, Color
7.
Andrology ; 6(5): 766-774, 2018 09.
Article in English | MEDLINE | ID: mdl-29939496

ABSTRACT

Previously, we have demonstrated that human tissue kallikrein 1 (hKLK1) improves age-related erectile dysfunction (ED). Autophagy has been implicated in age-related diseases, including ED. However, the molecular mechanisms underlying hKLK1-mediated amelioration of age-related ED via regulation of autophagy remains unknown. To explore the potential mechanism, male wild-type Sprague-Dawley rats (WTR) and transgenic rats harboring human KLK1 (TGR) were bred till 4 or 18 months of age and divided into three groups: young WTR (yWTR) as the control group, aged WTR (aWTR) group, and aged TGR (aTGR) group. The erectile function of each rat was evaluated using cavernous nerve electrostimulation. The ratio of intracavernous pressure/mean arterial pressure (ICP/MAP) and total ICP were also measured. Western blotting, immunohistochemistry, and transmission electron microscopy were performed to detect the levels of autophagy. The expression levels of related signaling pathways were determined by western blotting and immunohistochemistry. We found that hKLK1 improved the impaired erectile function of aged rats. Compared to the yWTR and aTGR groups, the aWTR group showed reduced smooth muscle/collagen ratio, fewer autophagosomes, and lower expression of Beclin 1 and LC3-II, which indicate impaired smooth muscle function and low level of autophagy in the smooth muscle cells. Moreover, the PI3K/Akt/mTOR signaling pathway, which is considered to be a negative regulator of autophagy, was upregulated in the aWTR group. hKLK1 may partially restore erectile function in aged transgenic rats by upregulating protective autophagy via the PI3K/Akt/mTOR pathway. These observations indicate that hKLK1 is a potential gene therapy candidate for age-related ED.


Subject(s)
Erectile Dysfunction/genetics , Genetic Therapy , Penile Erection/genetics , Tissue Kallikreins/genetics , Animals , Autophagy , Erectile Dysfunction/therapy , Humans , Male , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tissue Kallikreins/therapeutic use
8.
PLoS One ; 12(1): e0170427, 2017.
Article in English | MEDLINE | ID: mdl-28103290

ABSTRACT

Our previous studies had reported that Human Tissue Kallikrein 1 (hKLK1) preserved erectile function in aged transgenic rats, while the detailed mechanism of hKLK1 protecting erectile function in aged rats through activation of cGMP and cAMP was not mentioned. To explore the latent mechanism, male wild-type Sprague-Dawley rats (WTR) and transgenic rats harboring the hKLK1 gene (TGR) were fed to 4 and 18 months old and divided into four groups: young WTR (yWTR) as the control, aged WTR (aWTR), aged TGR (aTGR) and aged TGRs with HOE140 (aTGRH). Erectile function of all rats was evaluated by cavernous nerve electrostimulation method and measured by the ratio of intracavernous pressure/ mean arterial pressure (ICP/MAP) in rats. Expression levels of cAMP and cGMP were assessed, and related signaling pathways were detected by western blot, immunohistochemistry and RT-PCR. Our experiment results showed erectile function of the aWTR group and aTGRH group was lower compared with those of other two groups. Also, expression levels of cAMP and cGMP were significantly lower than those of other two groups. Moreover, expressions of related signaling pathways including DDAH/ADMA/NOS/cGMP and COX-2/PTGIS/cAMP were also downregulated in the corpus cavernosum of rats in aWTR group. Our finding revealed hKLK1 played a protective role in age-related ED. The DDAH/ADMA/NOS/cGMP and COX-2/PTGIS/cAMP pathways that were linked to the mechanism hKLK1 could increase the levels of cGMP and cAMP, which might provide novel therapy targets for age-related ED.


Subject(s)
Aging/physiology , Penile Erection/physiology , Tissue Kallikreins/physiology , Aging/genetics , Amidohydrolases/metabolism , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Erectile Dysfunction/genetics , Erectile Dysfunction/metabolism , Erectile Dysfunction/physiopathology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Male , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Penile Erection/genetics , Penis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tissue Kallikreins/genetics
9.
Investig Clin Urol ; 57(4): 286-97, 2016 07.
Article in English | MEDLINE | ID: mdl-27437539

ABSTRACT

PURPOSE: To investigate the effects of cavernous nerve injury (CNI) on gene expression profiles in the cavernosal tissue of a CNI-induced erectile dysfunction (ED) model and to provide a basis for future investigations to discover potential target genes for ED treatment. MATERIALS AND METHODS: Young adult rats were divided randomly into 2 groups: sham operation and bilateral CN resection. At 12 weeks after CNI we measured erectile responses and performed microarray experiments and gene set enrichment analysis to reveal gene signatures that were enriched in the CNI-induced ED model. Alterations in gene signatures were compared with those in the diabetes-induced ED model. The diabetic-induced ED data is taken from GSE2457. RESULTS: The mean ratio of intracavernosal pressure/blood pressure for the CNI group (0.54±0.4 cmH2O) was significantly lower than that in the sham operation group (0.73±0.8 cmH2O, p<0.05). Supervised and unsupervised clustering analysis showed that the diabetes- and CNI-induced ED cavernous tissues had different gene expression profiles from normal cavernous tissues. We identified 46 genes that were upregulated and 77 genes that were downregulated in both the CNI- and diabetes-induced ED models. CONCLUSIONS: Our genome-wide and computational studies provide the groundwork for understanding complex mechanisms and molecular signature changes in ED.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Erectile Dysfunction/genetics , Penis/metabolism , Peripheral Nerve Injuries/genetics , Transcriptome , Animals , Cluster Analysis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Down-Regulation/physiology , Erectile Dysfunction/etiology , Erectile Dysfunction/metabolism , Gene Expression Profiling/methods , Genome-Wide Association Study , Male , Penile Erection/genetics , Penile Erection/physiology , Penis/innervation , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/metabolism , Up-Regulation/physiology
10.
Andrologia ; 48(1): 59-64, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25879169

ABSTRACT

It was investigated whether short hairpin ribonucleic acid constructs targeting insulin-like growth factor binding protein-3 (IGFBP-3 shRNA) can rehabilitate dyslipidaemia in streptozotocin-induced diabetic rats. After 12 weeks of intracavernous administration of IGFBP-3 shRNA, intracavernous pressure responses to electrical stimulation of cavernous nerves were evaluated. The concentrations of serum low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglyceride and cavernous cyclic guanosine monophosphate were all detected by enzyme-linked immunosorbent assay. The per cent of smooth muscle in corpus cavernous tissue was also evaluated. It was found that the cavernosal pressure was significantly increased in the IGFBP-3 shRNA treatment group compared to the diabetic control group after 12 weeks of intracavernous administration of IGFBP-3 shRNA (P < 0.01). The concentrations of serum low-density lipoprotein cholesterol and triglyceride were significantly decreased in the IGFBP-3 shRNA treatment group compared to the diabetic control group, while no significant changes of serum high-density lipoprotein cholesterol concentration were found (P < 0.01). At the same time, cavernous cyclic guanosine monophosphate concentrations and the percentage of cavernosal smooth muscle were both significantly increased in the IGFBP-3 shRNA treatment group compared to the diabetic control group (P < 0.01). This study indicated that IGFBP-3 shRNA might rehabilitate erectile function via a decrease in concentrations of serum low-density lipoprotein and triglyceride, an increase in the percentage of cavernosal smooth muscle and an improvement in the nitric oxide-cyclic guanosine monophosphate signalling activities in streptozotocin-induced diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Dyslipidemias/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Penile Erection/genetics , Penis/metabolism , Animals , Cyclic GMP/metabolism , Dyslipidemias/metabolism , Enzyme-Linked Immunosorbent Assay , Erectile Dysfunction/genetics , Erectile Dysfunction/metabolism , Gene Knockdown Techniques , Lipoproteins, HDL , Lipoproteins, LDL/metabolism , Male , Muscle, Smooth/pathology , Nitric Oxide/metabolism , Penis/physiopathology , RNA, Small Interfering/genetics , Rats , Triglycerides/metabolism
11.
Genet Mol Res ; 14(3): 10482-9, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26400279

ABSTRACT

We examined the relationship between chronic hypoxia and erectile dysfunction in rat and its possible pathogenic mechanism. Forty-eight white male adult Sprague-Dawley rats were randomly divided into a test group and a control group. In accordance with the experimental time (2, 6, and 10 weeks), each group was divided into 3 subgroups, with 8 rats in each subgroup. Rats in the test group were fed in an airtight hypoxia cabin, while rats in the control group were maintained in a normal environment, with other conditions kept the same. At 2, 6, and 10 weeks, the rats in each group were observed for erectile function. Affinity purification was used to detect neural nitric oxide synthase (nNOS)-positive nerve fibers and endothelial nitric oxide synthase (eNOS) expression. After hypoxia, erectile frequency decreased significantly compared to before hypoxia (P < 0.001). Comparison of the test group and control group revealed a significant difference in the quantity of nNOS-positive nerve fiber and eNOS protein expression (P < 0.01). Hypoxia may influence erectile function and nNOS and eNOS expression in rats. The decrease in the quantity of nNOS nerve fibers and expression of eNOS may contribute to erectile dysfunction under hypoxic conditions in rats.


Subject(s)
Erectile Dysfunction/genetics , Hypoxia/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type I/genetics , Oxygen/pharmacology , Penile Erection/drug effects , Animals , Erectile Dysfunction/metabolism , Erectile Dysfunction/physiopathology , Gene Expression Regulation , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Nerve Fibers/drug effects , Nerve Fibers/metabolism , Nerve Fibers/pathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Penile Erection/genetics , Penis/blood supply , Penis/drug effects , Penis/physiology , Rats , Rats, Sprague-Dawley
13.
Zhonghua Nan Ke Xue ; 21(2): 99-106, 2015 Feb.
Article in Chinese | MEDLINE | ID: mdl-25796680

ABSTRACT

Erectile dysfunction (ED), as a common male disease, seriously affects the patients' sexual life quality. Most ED patients benefit from phosphodiesterase type 5 (PDE5) inhibitors, but some refractory ED sufferers fail to respond to them. With the rapid development of molecular biology, the relevant molecular signaling pathways of penile erection and molecular pathogenesis of ED have been gradually clarified, and attempts have been made at a better management or a complete cure of ED with advanced molecular biological methods such as the gene therapy. This article presents an overview on the research progress in the molecular signaling pathways, molecular pathogenesis, and gene therapy of ED.


Subject(s)
Biomedical Research , Erectile Dysfunction/genetics , Penile Erection/genetics , Erectile Dysfunction/therapy , Genetic Therapy , Humans , Male , Phosphodiesterase 5 Inhibitors/therapeutic use , Quality of Life , Signal Transduction
14.
J Pharmacol Exp Ther ; 353(2): 330-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25740897

ABSTRACT

Type 2 diabetes mellitus (DM2) and obesity are major risk factors for erectile dysfunction (ED). In diabetes, increased oxidative stress leads to decreased nitric oxide (NO) bioavailability, and diabetic patients appear to be less responsive to conventional therapy with phosphodiesterase type 5 inhibitors. We investigated whether the soluble guanylyl cyclase stimulator BAY 41-2272 (5-cyclopropyl-2-[1-(2-fluoro-benzyl)-1H-pyrazolo[3,4-b]pyridine-3-yl]pyrimidin-4ylamine) is effective in improving impaired corpus cavernosum (CC) relaxation in obese DM2 mice by reducing oxidative stress. Adult db/db(-/-) mice or their lean db(/+) littermates were used to assess vascular function, cGMP levels, antioxidant status, NADPH oxidase expression, and superoxide formation in the absence or presence of BAY 41-2272. Results showed that BAY 41-2272 (10(-8) to 10(-5) M) potently relaxed CC from db(/+) or db/db(-/-) mice in a similar manner. BAY 41-2272 significantly enhanced both endothelium-dependent and nitrergic relaxation induced by electrical field stimulation (EFS), and improved the impaired relaxation to acetylcholine and EFS in the diabetic animals in a concentration-dependent manner (10(-8) to 10(-7) M). BAY 41-2272 increased cGMP levels and potentiated relaxation responses to exogenous NO in CC. Total antioxidant status was reduced in plasma and urine whereas expression of vascular NADPH oxidase subunits (gp91phox, p22phox, and p47phox) was increased in the CC of db/db(-/-) mice, suggesting a state of oxidative stress. These effects were prevented by BAY 41-2272 in a concentration-dependent manner. These results suggest that BAY 41-2272 improves CC relaxation in db/db(-/-) mice by increasing cGMP and augmenting antioxidant status, making this drug is a potential novel candidate to treat ED.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Guanylate Cyclase/metabolism , Obesity/physiopathology , Penile Erection/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Blood Glucose/metabolism , Cyclic GMP/metabolism , Drug Interactions , Epithelium/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Lipids/blood , Male , Mice , Mice, Obese , Muscle Relaxation/drug effects , NADPH Oxidases/metabolism , Nitroprusside/pharmacology , Oxidative Stress/drug effects , Penile Erection/drug effects , Penis/drug effects , Penis/metabolism , Penis/pathology , Penis/physiopathology , Soluble Guanylyl Cyclase , Vasodilation/drug effects
15.
Andrologia ; 46(9): 997-1003, 2014.
Article in English | MEDLINE | ID: mdl-24428436

ABSTRACT

We aimed to evaluate the effect of acetylsalicylic acid (ASA) treatment on diabetes-induced erectile dysfunction. Adult male Sprague-Dawley rats were divided into four groups as follows: (i) control (C), (ii) diabetic (D), (iii) ASA-treated control (C+ASA) and (iv) ASA-treated diabetic (D+ASA) groups. In groups 2 and 4, diabetes was induced by injection of 35 mg kg(-1) streptozotocin. ASA (100 mg kg(-1) day(-1) , orally) was administrated to rats in groups 3 and 4 for 8 weeks. Both intracavernosal pressure (ICP) and mean arterial blood pressure (MAP) were measured in in vivo studies. In organ bath, the relaxation responses to acetylcholine (ACh), electrical field stimulation (EFS) and sodium nitroprusside were tested in corpus cavernosum (CC) strips. The mRNA expression for neuronal nitric oxide synthase (nNOS) was calculated using reverse transcription polymerase chain reaction technique. In in vivo experiments, diabetic rats displayed reduced ICP/MAP values, which were normalised with ASA treatment. The relaxant response to high-dose ACh and EFS at low frequencies (1-8 Hz) in CC strips from the D+ASA group were significantly higher when compared to the D group. Treatment with ASA normalised the raised mRNA expressions of nNOS in diabetic penile tissues. ASA may be involved in mRNA of protein synthesis of NO released from nonadrenergic and noncholinergic cavernosal nerve in diabetes.


Subject(s)
Aspirin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Penile Erection/drug effects , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Diabetes Complications/genetics , Diabetes Complications/physiopathology , Diabetes Complications/prevention & control , Diabetes Mellitus, Experimental/genetics , Electric Stimulation , Erectile Dysfunction/genetics , Erectile Dysfunction/physiopathology , Erectile Dysfunction/prevention & control , Male , Nitric Oxide Synthase Type I/genetics , Nitroprusside/pharmacology , Penile Erection/genetics , Penile Erection/physiology , Penis/blood supply , Penis/drug effects , Penis/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
Eur J Clin Invest ; 42(7): 729-37, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22224829

ABSTRACT

BACKGROUND: Imbalance in nitric oxide (NO), an atheroprotective vasodilator, is associated with endothelial dysfunction, cardiovascular diseases (CVD) and diabetic complications. Various endothelial NO synthase (eNOS) polymorphisms may affect NO bioavailability, thereby promoting adverse cardiovascular milieu. MATERIALS AND METHODS: To analyze glucose homeostasis, cardiometabolic phenotype, and micro- and macroangiopathies associated with eNOS G894T gene polymorphism in type 2 diabetes (T2DM). 210 T2DM outpatients (mean age (1SD) 70 (12); diabetes duration 19 (9) years; males:females 64:36%; metabolic syndrome 87%) had insulin sensitivity and b-cell function modelled with HOMA, alongside routine laboratory and endothelin measurements. RESULTS: GG, GT and TT genotypes represented 48% (n = 100), 39% (n = 83) and 13% (n = 27). Overall microangiopathy (retinopathy, neuropathy and/or nephropathy) was present in 74%, and overall macroangiopathy (CAD, PAD and/or TIA/stroke) in 45%. The TT genotype did not translate into a more severe vascular phenotype, as TT patients carrying the proposed risk genotype did not suffer a higher rate of micro- and macrovascular complications. On the other hand, erectile dysfunction, present in 60% of males (n = 135), was much more prevalent in TT males: 57% [GG & GT] vs. 93% in TT (p 0.0088). Ocular hypertension/glaucoma frequency (18% of the whole group) was also markedly different, albeit in opposing directions, between eNOS G894T gene polymorphism subgroups: 21% [GG & GT] vs. 0% prevalence (TT; p 0.0057). CONCLUSIONS: eNOS G894T gene polymorphism in homozygous TT carriers translates into opposing effects on erectile function (detrimental) and ocular hypertension/glaucoma (protective) in T2DM, without affecting glucose homeostasis determinants or the presence of micro- and macrovascular complications.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Intraocular Pressure/genetics , Nitric Oxide Synthase Type III/genetics , Penile Erection/genetics , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Cross-Sectional Studies , Erectile Dysfunction/genetics , Female , Glucose/metabolism , Humans , Male , Middle Aged , Penile Erection/physiology , Polymorphism, Genetic/genetics , Risk Factors , Severity of Illness Index
17.
J Sex Med ; 8(11): 3075-85, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21883953

ABSTRACT

INTRODUCTION: Elastin fibers confer passive recoil to many tissues including the lung, skin, and arteries. In the penis, elastin is present in sinusoids, arterioles, and in the tunica albuginea. Although decreased penile elastin has been reported in men with erectile dysfunction, the exact role of elastin in physiologic processes integral to erection remains speculative. AIM: The aim of this study was to characterize erectile function in elastin-deficient mice. METHODS: Elastin haploinsufficient mice (Eln(+/-) ) and aged match Eln(+/+) (Wt) mice were used. Cavernosum was removed from some mice for quantification of elastin, collagen, and smooth muscle actin. Ex vivo assessment of contractile force generation was performed by myography. In vivo assessment of intracorporal pressure normalized to mean arterial pressure in response to electrical stimulation of the cavernosal nerve was measured. Veno-occlusive function was determined by cavernosography. MAIN OUTCOME MEASURES: The main outcome measures of this study were the in vitro and in vivo assessment of cavernosal vasoreactivity, veno-occlusive function and erection in mice deficient in elastin. RESULTS: Eln (+/-) mice exhibited ∼33% less penile elastin than Wt mice, with no change in collagen. Cavernosal tissue from Eln(+/-) mice has a significantly heightened contractile response, explained in part by increased smooth muscle cell content. Veno-occlusive function was significantly altered in Eln(+/-) mice. Interestingly, erectile function was impaired only at submaximal voltage (1 V) stimulation (there was no impairment during the higher 2-V stimulus). CONCLUSIONS: Eln (+/-) mice display a cavernosal phenotype consistent with developmental changes attributable to the loss of elastin. These alterations confer a degree of altered erectile function that is able to be overridden by maximal stimulatory input. Altogether, these data suggest that elastin is important for erectile function.


Subject(s)
Elastin/deficiency , Haploinsufficiency/physiology , Penile Erection/physiology , Actins/analysis , Actins/physiology , Animals , Blotting, Western , Collagen/analysis , Collagen/physiology , Elastin/analysis , Elastin/genetics , Haploinsufficiency/genetics , Male , Mice , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/physiology , Penile Erection/genetics , Penis/anatomy & histology , Penis/chemistry , Penis/physiology , Real-Time Polymerase Chain Reaction , Vasodilation/physiology
18.
J Sex Med ; 8(8): 2181-90, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21595839

ABSTRACT

INTRODUCTION: Previous studies have confirmed the gene transfer of insulin-like growth factor-1 (IGF-1) and the IGF-1 protein can improve the erectile function in aging rats. IGF binding protein (BP)-3 can regulates the availability of IGF-I. The higher expression of IGFBP-3 may play an important role in erectile dysfunction (ED). AIM: The study aimed to investigate the mRNA and protein expression of IGFBP-3 in young and old rat penile tissues and assess the alteration of the penile structure and the NO-guanosine 3',5'-cyclic-monophosphate (cGMP) signaling pathways-related marker in ED associated with aging. MAIN OUTCOME MEASURES: The main outcome measures for this study were the expression of IGFBP-3, morphological changes, NO-cGMP signaling pathways-related marker, erectile responses were determined. METHODS: Traditional reverse transcriptase polymerase chain reaction (RT-PCR) and real-time PCR were performed to examine the mRNA expression of the IGFBP-3. The Western blot was used to confirm the protein expression. Immunohistochemistry was also performed to identify the cellular localization of the encoded protein. The percentage of smooth muscle in corpus cavernosum tissue, the activity of nitric oxide synthase (NOS), and concentration of cGMP in penile tissue were also analyzed. RESULTS: The expression levels of IGFBP-3 of mRNA and protein were greatly increased in aging rats compared with young control rats, which is confirmed by traditional RT-PCR, real-time PCR, and Western blot (P < 0.01, respectively). Increased IGFBP-3 protein was localized to the epithelium of the urethra, penile endothelium, and smooth muscle in the corpus cavernosum. Significant depletion of the smooth muscle density relative to the connective tissue was also observed in the penis of the aged rats, and the lower activity of NOS and lower concentration of cGMP was also demonstrated accompanied with a significant reduction in the intracavernous pressure. CONCLUSIONS: Our data suggest that the increased mRNA and protein expression of IGFBP-3 in old rats may play a role in ED.


Subject(s)
Erectile Dysfunction/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Penile Erection/genetics , RNA, Messenger/genetics , Age Factors , Aging/genetics , Animals , Disease Models, Animal , Erectile Dysfunction/metabolism , Gene Expression , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Male , Penis/chemistry , Penis/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction
19.
Andrologia ; 42(4): 218-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20629643

ABSTRACT

Erectile dysfunction (ED) is often associated with cardiovascular disorders such as hypertension, coronary heart disease, hypercholesterolaemia and diabetes mellitus. The genotypes in the GNB3 C825T, the ACE I/D and the eNOS G894T polymorphisms have been identified as genetic risk factors for cardiovascular disorders. The association between the genotypes in these polymorphisms and the risk to develop ED was analysed. In 455 German ED patients and 111 age-matched healthy controls genotyping in the candidate polymorphisms was performed after DNA extraction from whole blood. Association studies between the genotype distribution in the control group in comparison with the ED-group and age of onset of the disease as well as erectile response to intracorporal prostaglandin injection in dependence of candidate polymorphism genotype were performed using the SPSS-Software(R). Genotype distribution of the GNB3 C825T, the ACE I/D and the eNOS G894T polymorphisms was similar in the ED population and the healthy control group. The age of onset of the disease as well as the erectile response to intracorporal prostaglandin injection was independent of the genotypes in the three candidate polymorphisms. In contrast to the previous studies in this analysis, the risk to develop ED is not influenced by the genotypes in the GNB3 C825T, the ACE I/D and the eNOS G894T polymorphisms.


Subject(s)
Erectile Dysfunction/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Nitric Oxide Synthase Type III/genetics , Peptidyl-Dipeptidase A/genetics , Age of Onset , Alprostadil/therapeutic use , Erectile Dysfunction/drug therapy , Genetic Association Studies , Genotype , Germany , Humans , Male , Middle Aged , Penile Erection/genetics , Polymorphism, Genetic
20.
Expert Opin Biol Ther ; 10(9): 1305-14, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20662742

ABSTRACT

IMPORTANCE OF THE FIELD: Erectile dysfunction (ED) is a major men's health problem. Although the high success rate of treating ED by phosphodiesterase 5 (PDE5) inhibitors has been reported, there are a significant number of ED patients who do not respond to currently available treatment modalities. AREAS COVERED IN THIS REVIEW: To elucidate the current status of gene therapy applications for ED, gene therapy approaches for ED treatment are reviewed. WHAT THE READER WILL GAIN: Gene therapy strategies that can enhance nitric oxide (NO) production or NO-mediated signaling pathways, growth factor-mediated nerve regeneration or K(+) channel activity in the smooth muscle could be promising approaches for the treatment of ED. Although the majority of gene therapy studies are still in the preclinical phase, the first clinical trial using non-viral gene transfer of Ca(2+)-activated, large-conductance K(+) channels into the corpus cavernosum of ED patients showed positive results. TAKE HOME MESSAGE: Gene therapy represents an exciting future treatment option for ED, especially for people with severe ED unresponsive to current first-line therapies such as PDE5 inhibitors although the long-term safety of both viral and non-viral gene therapies should be established.


Subject(s)
Erectile Dysfunction/therapy , Genetic Therapy , Penile Erection , Penis/physiopathology , Animals , Erectile Dysfunction/genetics , Erectile Dysfunction/metabolism , Erectile Dysfunction/physiopathology , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Nitric Oxide/metabolism , Penile Erection/genetics , Penis/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Signal Transduction , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...