Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(14)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708907

ABSTRACT

Human stem cell therapy for type 2 diabetes/obesity (T2D/O) complications is performedwith stem cell autografts, exposed to the noxious T2D/O milieu, often with suboptimal results.We showed in the Obese Zucker (OZ) rat model of T2D/O that when their muscle-derived stemcells (MDSC) were from long-term T2D/O male rats, their repair ecacy for erectile dysfunctionwas impaired and were imprinted with abnormal gene- and miR-global transcriptional signatures(GTS). The damage was reproduced in vitro by short-term exposure of normal MDSC to dyslipidemicserum, causing altered miR-GTS, fat infiltration, apoptosis, impaired scratch healing, and myostatinoverexpression. Similar in vitro alterations occurred with their normal counterparts (ZF4-SC) fromthe T2D/O rat model for female stress urinary incontinence, and with ZL4-SC from non-T2D/O leanfemale rats. In the current work we studied the in vitro eects of cholesterol and Na palmitate aslipid factors on ZF4-SC and ZL4-SC. A damage partially resembling the one caused by the femaledyslipidemic serum was found, but diering between both lipid factors, so that each one appears tocontribute specifically to the stem cell damaging eects of dyslipidemic serum in vitro and T2D/Oin vivo, irrespective of gender. These results also confirm the miR-GTS biomarker value forMDSC damage.


Subject(s)
Cholesterol/metabolism , Diabetes Mellitus, Type 2/pathology , Obesity/pathology , Palmitic Acid/metabolism , Stem Cells/pathology , Urinary Incontinence, Stress/pathology , Animals , Apoptosis , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Female , Obesity/metabolism , Rats , Rats, Zucker , Stem Cells/metabolism , Urinary Incontinence, Stress/metabolism
2.
Int J Mol Sci ; 20(16)2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31430893

ABSTRACT

Female stress urinary incontinence (FSUI) is prevalent in women with type 2 diabetes/obesity (T2D/O), and treatment is not optimal. Autograph stem cell therapy surprisingly has poor efficacy. In the male rat model of T2D/O, it was demonstrated that epigenetic changes, triggered by long-term exposure to the dyslipidemic milieu, led to abnormal global transcriptional signatures (GTS) of genes and microRNAs (miR), and impaired the repair capacity of muscle-derived stem cells (MDSC). This was mimicked in vitro by treatment of MDSC with dyslipidemic serum or lipid factors. The current study aimed to predict whether these changes also occur in stem cells from female 12 weeks old T2D/O rats, a model of FSUI. MDSCs from T2D/O (ZF4-SC) and normal female rats (ZL4-SC) were treated in vitro with either dyslipidemic serum (ZFS) from late T2D/O 24 weeks old female Zucker fatty (ZF) rats, or normal serum (ZLS) from 24 weeks old female Zucker lean (ZL) rats, for 4 days and subjected to assays for fat deposition, apoptosis, scratch closing, myostatin, interleukin-6, and miR-GTS. The dyslipidemic ZFS affected both female stem cells more severely than in the male MDSC, with some gender-specific differences in miR-GTS. The changes in miR-GTS and myostatin/interleukin-6 balance may predict in vivo noxious effects of the T2D/O milieu that might impair autograft stem cell (SC) therapy for FSUI, but this requires future studies.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Dyslipidemias/pathology , Stem Cells/pathology , Urinary Incontinence/pathology , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Dyslipidemias/blood , Female , Male , Rats , Rats, Zucker , Stem Cell Transplantation
3.
J Sex Med ; 15(12): 1678-1697, 2018 12.
Article in English | MEDLINE | ID: mdl-30527052

ABSTRACT

BACKGROUND: Previous work showed that muscle-derived stem cells (MDSCs) exposed long-term to the milieu of uncontrolled type 2 diabetes (UC-T2D) in male obese Zucker (OZ) rats, were unable to correct the associated erectile dysfunction and the underlying histopathology when implanted into the corpora cavernosa, and were also imprinted with a noxious gene global transcriptional signature (gene-GTS), suggesting that this may interfere with their use as autografts in stem cell therapy. AIM: To ascertain the respective contributions of dyslipidemia and hyperglycemia to this MDSC damage, clarify its mechanism, and design a bioassay to identify the damaged stem cells. METHODS: Early diabetes MDSCs and late diabetes MDSCs were respectively isolated from nearly normal young OZ rats and moderately hyperglycemic and severely dyslipidemic/obese aged rats with erectile dysfunction. Monolayer cultures of early diabetic MDSCs were incubated 4 days in DMEM/10% fetal calf serum + or - aged OZ or lean Zucker serum from non-diabetic lean Zucker rats (0.5-5%) or with soluble palmitic acid (PA) (0.5-2 mM), cholesterol (CHOL) (50-400 mg/dL), or glucose (10-25 mM). MAIN OUTCOME MEASURE: Fat infiltration was estimated by Oil red O, apoptosis by TUNEL, protein expression by Western blots, and gene-GTS and microRNA (miR)-GTS were determined in these stem cells' RNA. RESULTS: Aged OZ serum caused fat infiltration, apoptosis, myostatin overexpression, and impaired differentiation. Some of these changes, and also a proliferation decrease occurred with PA and CHOL. The gene-GTS changes by OZ serum did not resemble the in vivo changes, but some occurred with PA and CHOL. The miR-GTS changes by OZ serum, PA, and CHOL resembled most of the in vivo changes. Hyperglycemia did not replicate most alterations. CLINICAL IMPLICATIONS: MDSCs may be damaged in long-term UC-T2D/obese patients and be ineffective in autologous human stem cell therapy, which may be prevented by excluding the damaged MDSCs. STRENGTH & LIMITATIONS: The in vitro test of MDSCs is innovative and fast to define dyslipidemic factors inducing stem cell damage, its mechanism, prevention, and counteraction. Confirmation is required in other T2D/obesity rat models and stem cells (including human), as well as miR-GTS biomarker validation as a stem cell damage biomarker. CONCLUSION: Serum from long-term UC-T2D/obese rats or dyslipidemic factors induces a noxious phenotype and miR-GTS on normal MDSCs, which may lead in vivo to the repair inefficacy of late diabetic MDSCs. This suggests that autograft therapy with MDSCs in long-term UT-T2D obese patients may be ineffective, albeit this may be predictable by prior stem cell miR-GTS tests. Masouminia M, Gelfand R, Kovanecz I, et al. Dyslipidemia Is a Major Factor in Stem Cell Damage Induced by Uncontrolled Long-Term Type 2 Diabetes and Obesity in the Rat, as Suggested by the Effects on Stem Cell Culture. J Sex Med 2018;15:1678-1697.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Dyslipidemias/complications , Erectile Dysfunction/etiology , Stem Cell Transplantation , Animals , Cell Differentiation , Diabetes Mellitus, Experimental/therapy , Dyslipidemias/physiopathology , Erectile Dysfunction/physiopathology , Humans , Male , Obesity/complications , Penis/physiopathology , Rats , Rats, Zucker
4.
J Sex Med ; 13(5): 786-97, 2016 05.
Article in English | MEDLINE | ID: mdl-27114192

ABSTRACT

INTRODUCTION: Muscle-derived stem cells (MDSCs) and other SCs implanted into the penile corpora cavernosa ameliorate erectile dysfunction in type 1 diabetic rat models by replenishing lost corporal smooth muscle cells (SMCs) and decreasing fibrosis. However, there are no conclusive data from models of type 2 diabetes (T2D) and obesity. AIM: To determine whether MDSCs from obese Zucker (OZ) rats with T2D at an early stage of diabetes (early diabetic SCs isolated and cultured in low-glucose medium [ED-SCs]) counteract corporal veno-occlusive dysfunction and corporal SMC loss or lipo-fibrosis when implanted in OZ rats at a late stage of diabetes and whether MDSCs from these OZ rats with late diabetes (late diabetic SCs isolated and cultured in high-glucose medium [LD-SC]) differ from ED-SCs in gene transcriptional phenotype and repair capacity. METHODS: ED-SCs and LD-SCs were compared by DNA microarray assays, and ED-SCs were incubated in vitro under high-glucose conditions (ED-HG-SC). These three MDSC types were injected into the corpora cavernosa of OZ rats with late diabetes (OZ/ED, OZ/LD, and OZ/ED-HG rats, respectively). Untreated OZ and non-diabetic lean Zucker rats functioned as controls. Two months later, rats were subjected to cavernosometry and the penile shaft and corporal tissues were subjected to histopathology and DNA microarray assays. MAIN OUTCOME MEASURES: In vivo erectile dysfunction assessment by Dynamic Infusion Cavernosometry followed by histopathology marker analysis of the penile tissues. RESULTS: Implanted ED-SCs and ED-HG-SCs improved corporal veno-occlusive dysfunction, counteracted corporal decreases in the ratio of SMCs to collagen and fat infiltration in rats with long-term T2D, and upregulated neuronal and endothelial nitric oxide. LD-SCs acquired an inflammatory, pro-fibrotic, oxidative, and dyslipidemic transcriptional phenotype and failed to repair the corporal tissue. CONCLUSION: MDSCs from pre-diabetic rats injected into the corpora cavernosa of rats with long-term T2D improve corporal veno-occlusive dysfunction and the underlying histopathology. In contrast, MDSCs from rats with long-term uncontrolled T2D are imprinted by the hyperglycemic and dyslipidemic milieu with a noxious phenotype associated with an impaired tissue repair capacity. SCs affected by diabetes could lack tissue repair efficacy as autografts and should be reprogrammed in vitro or substituted by SCs from allogenic non-diabetic sources.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Erectile Dysfunction/therapy , Stem Cell Transplantation , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Endothelium/pathology , Erectile Dysfunction/physiopathology , Male , Myocytes, Smooth Muscle , Penis/physiopathology , Rats , Rats, Zucker , Stem Cells
5.
J Sex Med ; 12(2): 313-27, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25496134

ABSTRACT

INTRODUCTION: The success of medical therapies for Peyronie's disease (PD) has not been optimal, possibly because many of them went directly to clinical application without sufficient preclinical scientific research. Previous studies revealed cellular and molecular pathways involved in the formation of the PD plaque and in particular the role of the myofibroblast. AIMS: The current work aimed to determine under normal and fibrotic conditions what differentiates PD cells from tunica albuginea (TA) and corpora cavernosa (CC) cells by defining their global transcriptional signatures and testing in vivo whether PD cells can generate a PD-like plaque. METHODS: Human TA, PD, and CC cells were grown with transforming growth factor beta 1 (TGFß1; TA+, PD+, CC+) or without it (TA-, PD-, CC-) and assayed by (i) immunofluorescence, Western blot and RT-PCR for myofibroblast, smooth muscle cell and stem cell markers; (ii) collagen content; and (iii) DNA microarray analysis. The ability of PD+ cells to induce a PD-like plaque in an immuno-suppressed rat model was assessed by Masson trichrome and Picrosirius Red stainings. MAIN OUTCOMES MEASURES: Fibroproliferative features of PD cells and identification of related key genes as novel targets to reduce plaque size. RESULTS: Upon TGFß1stimulation, collagen levels were increased by myofibroblasts in the PD+ but not in the CC+ cells. The transcriptional signature of the PD- cells identified fibroproliferative, myogenic (myofibroblasts), inflammatory, and collagen turnover genes that differentiate them from TA- or CC- cells and respond to TGFß1 with a PD+ fibrotic phenotype, by upregulation of IGF-1, ACTG2, MYF5, ACTC1, PSTN, COL III, MMP3, and others. The PD+ cells injected into the TA of the rat induce a PD-like plaque. CONCLUSIONS: This suggests a novel combination therapy to eliminate a PD plaque by targeting the identified genes to (i) improve collagenase action by stimulating endogenous metalloproteinases specific to key collagen types and (ii) counteract fibromatosis by inhibiting myofibroblast generation, proliferation, and/or apoptosis.


Subject(s)
Penile Induration/drug therapy , Transforming Growth Factor beta1/pharmacology , Animals , Apoptosis , Cell Culture Techniques , Collagen/biosynthesis , Humans , Male , Metalloproteases , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Oligonucleotide Array Sequence Analysis , Penile Induration/physiopathology , Penis/metabolism , RNA, Messenger , Rats , Stem Cells/metabolism
6.
Int J Mol Sci ; 15(10): 17204-20, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25264738

ABSTRACT

Erectile dysfunction is a common complication for patients undergoing surgeries for prostate, bladder, and colorectal cancers, due to damage of the nerves associated with the major pelvic ganglia (MPG). Functional re-innervation of target organs depends on the capacity of the neurons to survive and switch towards a regenerative phenotype. PDE5 inhibitors (PDE5i) have been successfully used in promoting the recovery of erectile function after cavernosal nerve damage (BCNR) by up-regulating the expression of neurotrophic factors in MPG. However, little is known about the effects of PDE5i on markers of neuronal damage and oxidative stress after BCNR. This study aimed to investigate the changes in gene and protein expression profiles of inflammatory, anti-inflammatory cytokines and oxidative stress related-pathways in MPG neurons after BCNR and subsequent treatment with sildenafil. Our results showed that BCNR in Fisher-344 rats promoted up-regulation of cytokines (interleukin- 1 (IL-1) ß, IL-6, IL-10, transforming growth factor ß 1 (TGFß1), and oxidative stress factors (Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, Myeloperoxidase (MPO), inducible nitric oxide synthase (iNOS), TNF receptor superfamily member 5 (CD40) that were normalized by sildenafil treatment given in the drinking water. In summary, PDE5i can attenuate the production of damaging factors and can up-regulate the expression of beneficial factors in the MPG that may ameliorate neuropathic pain, promote neuroprotection, and favor nerve regeneration.


Subject(s)
Ganglia/metabolism , Oxidative Stress/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Sulfonamides/pharmacology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Ganglia/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Male , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nerve Tissue/injuries , Nitric Oxide Synthase Type II/metabolism , Penile Erection/drug effects , Penile Erection/physiology , Penis/innervation , Purines/pharmacology , Rats , Rats, Inbred F344 , Sildenafil Citrate , Transcriptome , Up-Regulation/drug effects
7.
J Sex Med ; 10(12): 2952-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24134786

ABSTRACT

INTRODUCTION: Bisphenol A (BPA), released from plastics and dental sealants, is a suspected endocrine disruptor and reproductive toxicant. In occupationally exposed workers, BPA has been associated with erectile dysfunction (ED). AIMS: To determine whether long-term exposure to high doses of BPA in the rat affects serum levels of testosterone (T) and estradiol (E2), and induces corporal histopathology and resultant ED. METHODS: Young rats were injected intraperitoneal (IP) injection daily with BPA at 25 mg/kg/day or vehicle (n = 8/group). Erectile function was measured at 3 months by cavernosometry and electrical field stimulation (EFS). BPA was assayed in serum, urine, and penile tissue, and serum T and E2 were determined. Quantitative Masson trichrome, terminal deoxynucleotidyl transferase dUTP nick end labeling, Oil Red O, immunohistochemistry for calponin, α-smooth muscle actin, and Oct 4 were applied to penile tissue sections. Protein markers were assessed by Western blots and 2-D minigels, and RNA by DNA microarrays. MAIN OUTCOME MEASURES: Erectile function, histological, and biochemical markers in corporal tissue. RESULTS: In the BPA-treated rats, total and free BPA levels were increased in the serum, urine, and penile tissue while serum T and E2 levels were reduced. In addition, the corpora cavernosa demonstrated a reduction in smooth muscle (SM) content, SM/collagen ratio, together with an increase in myofibroblasts, fat deposits, and apoptosis, but no significant change in collagen content or stem cells (nuclear/perinuclear Oct 4). In the penile shaft, BPA induced a downregulation of Nanog (stem cells), neuronal nitric oxide synthase (nitrergic terminals), and vascular endothelial growth factor (angiogenesis), with genes related to SM tone and cytoskeleton upregulated 5- to 50-fold, accompanied by changes in the multiple protein profile. However, both cavernosometry and EFS were unaltered by BPA. CONCLUSIONS: While rats treated chronically with a high IP dose of BPA developed hypogonadism and a corporal histo- and molecular-pathology usually associated with ED, no changes were detected in erectile function as measured by EFS and cavernosometry. Further studies using alternate routes of BPA administration with various doses and length of exposure are needed to expand these findings.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Penile Erection/drug effects , Penis/drug effects , Phenols/toxicity , Animals , Immunohistochemistry , Male , Muscle, Smooth/metabolism , Nanog Homeobox Protein , Nitric Oxide Synthase Type I/metabolism , Penis/metabolism , Penis/pathology , Rats , Rats, Inbred F344 , Stem Cells/metabolism , Transcription Factors/metabolism
8.
Stem Cell Res Ther ; 4(1): 4, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23295128

ABSTRACT

INTRODUCTION: Stimulating the commitment of implanted dystrophin+ muscle-derived stem cells (MDSCs) into myogenic, as opposed to lipofibrogenic lineages, is a promising therapeutic strategy for Duchenne muscular dystrophy (DMD). METHODS: To examine whether counteracting myostatin, a negative regulator of muscle mass and a pro-lipofibrotic factor, would help this process, we compared the in vitro myogenic and fibrogenic capacity of MDSCs from wild-type (WT) and myostatin knockout (Mst KO) mice under various modulators, the expression of key stem cell and myogenic genes, and the capacity of these MDSCs to repair the injured gastrocnemius in aged dystrophic mdx mice with exacerbated lipofibrosis. RESULTS: Surprisingly, the potent in vitro myotube formation by WT MDSCs was refractory to modulators of myostatin expression or activity, and the Mst KO MDSCs failed to form myotubes under various conditions, despite both MDSC expressing Oct 4 and various stem cell genes and differentiating into nonmyogenic lineages. The genetic inactivation of myostatin in MDSCs was associated with silencing of critical genes for early myogenesis (Actc1, Acta1, and MyoD). WT MDSCs implanted into the injured gastrocnemius of aged mdx mice significantly improved myofiber repair and reduced fat deposition and, to a lesser extent, fibrosis. In contrast to their in vitro behavior, Mst KO MDSCs in vivo also significantly improved myofiber repair, but had few effects on lipofibrotic degeneration. CONCLUSIONS: Although WT MDSCs are very myogenic in culture and stimulate muscle repair after injury in the aged mdx mouse, myostatin genetic inactivation blocks myotube formation in vitro, but the myogenic capacity is recovered in vivo under the influence of the myostatin+ host-tissue environment, presumably by reactivation of key genes originally silenced in the Mst KO MDSCs.


Subject(s)
Muscle Development/genetics , Muscle, Skeletal/metabolism , Myostatin/genetics , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Dystrophin/genetics , Dystrophin/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Myostatin/metabolism
9.
BJU Int ; 111(1): 159-70, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22672418

ABSTRACT

OBJECTIVE: To determine the gene expression profile of pelvic ganglia neurones after bilateral cavernosal nerve resection (BCNR) and subsequent treatment with sildenafil in relation to neurotrophic-related pathways. MATERIALS AND METHODS: Fisher rats aged 5 months were subjected to BCNR or sham operation and treated with or without sildenafil (20 mg/kg body-weight in drinking water) for 7 days. Total RNA isolated from pelvic ganglia was subjected to reverse transcription and then to quantitative reverse transcriptase-polymerase chain reaction (PCR) with the RAT-neurotrophic array. Results were corroborated by real-time PCR and western blotting. Another set of animals were injected with a fluorescent tracer at the base of the penis, 7 days before BCNR or sham operation, and were sacrificed 7 days after surgery. Sections of pelvic ganglia were used for immunohistochemistry with antibodies against neurturin, neuronal nitric oxide synthase, tyrosine hydroxylase and glial cell line-derived neurotrophic factor receptor α2. RESULTS: A down-regulation of the expression of neuronal nitric oxide synthase accompanied by changes in the level of cholinergic neurotrophic factors, such as neurturin and its receptor glial cell line-derived neurotrophic factor receptor α2, artemin, neurotrophin-4 and cilliary neurotrophic factor, was observed 7 days after BCNR in pelvic ganglia neurones. Treatment with sildenafil, starting immediately after surgery, reversed all these changes at a level similar to that in sham-operated animals. CONCLUSIONS: Sildenafil treatment promotes changes in the neurotrophic phenotype, leading to a regenerative state of pelvic ganglia neurones. The present study provides a justification for the use of phosphodiesterase 5 inhibitors as a neuroprotective agent after BCNR.


Subject(s)
Ganglia, Autonomic/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Neuroprotective Agents/pharmacology , Penis/innervation , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Ganglia, Autonomic/metabolism , Gene Expression/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Male , Neurons/metabolism , Neurturin/metabolism , Nitric Oxide Synthase Type I/metabolism , Organ Sparing Treatments/methods , Pelvis/innervation , Penis/drug effects , Penis/surgery , Purines/pharmacology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sildenafil Citrate
10.
J Sex Med ; 9(11): 2814-26, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22974131

ABSTRACT

INTRODUCTION: Long-term daily administration of phosphodiesterase type 5 (PDE5) inhibitors in the rat prevents or reverses corporal veno-occlusive dysfunction (CVOD) and smooth muscle cell (CSMC) loss and fibrosis, in both aging and bilateral cavernosal nerve resection (BCNR) models for erectile dysfunction. In the aging rat model, corporal implantation of skeletal muscle-derived stem cells (MDSC) reverses CVOD. Nitric oxide (NO) and cyclic guanosine monophosphate can modulate stem cell lineage. AIM: To investigate in the BCNR model the effects of sildenafil at lower doses, alone or in combination with MDSC or the NO donor molsidomine, on CVOD and the underlying corporal histopathology. MAIN OUTCOMES MEASURES: CVOD, histological, and biochemical markers in rat corporal tissue. Methods. Rats subjected to BCNR were maintained for 45 days either untreated, or received sildenafil in the water or retrolingually at 10, 2.5, and 1.25 mg/kg/day (medium, low, and very low doses), or intraperitoneal molsidomine, or MDSC implantation into the corpora cavernosa separately or in combination. Cavernosometry evaluated CVOD. Histopathology was assessed on penile sections by Masson trichrome, immunohistochemistry for α-smooth muscle actin (ASMA), or immunofluorescence for neuronal nitric oxide synthase (nNOS)/neurofilament 70, and in fresh tissue by Western blot for various markers and picrosirius red for collagen. RESULTS: All treatments normalized erectile function (drop rate), and most increased the CSMC/collagen ratio and ASMA expression in corporal tissue sections, and reduced collagen content in the penile shaft. MDSC also increased nNOS and brain-derived neurotrophic factor. The combination treatment was not superior to MDSC or sildenafil given alone, and upregulated PDE5. CONCLUSIONS: Lowering the dose of a continuous long-term sildenafil administration still maintained the prevention of CVOD in the BCNR rat previously observed, but it was less effective on the underlying histopathology. As in the aging rat model, MDSC also counteracted CVOD, but supplementation with very low-dose sildenafil did not improve the outcome.


Subject(s)
Impotence, Vasculogenic/prevention & control , Impotence, Vasculogenic/physiopathology , Molsidomine/pharmacology , Muscle Denervation , Muscle Fibers, Skeletal/transplantation , Penis/innervation , Piperazines/pharmacology , Stem Cell Transplantation , Sulfones/pharmacology , Vasodilator Agents/pharmacology , Animals , Combined Modality Therapy , Male , Mice , Penile Erection/drug effects , Penile Erection/physiology , Purines/pharmacology , Rats , Rats, Inbred F344 , Sildenafil Citrate
11.
J Transl Med ; 10: 159, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22871104

ABSTRACT

BACKGROUND: Previous studies have shown that long-term oral daily PDE 5 inhibitors (PDE5i) counteract fibrosis, cell loss, and the resulting dysfunction in tissues of various rat organs and that implantation of skeletal muscle-derived stem cells (MDSC) exerts some of these effects. PDE5i and stem cells in combination were found to be more effective in non-MI cardiac repair than each treatment separately. We have now investigated whether sildenafil at lower doses and MDSC, alone or in combination are effective to attenuate LV remodeling after MI in rats. METHODS: MI was induced in rats by ligature of the left anterior descending coronary artery. Treatment groups were: "Series A": 1) untreated; 2) oral sildenafil 3 mg/kg/day from day 1; and "Series B": intracardiac injection at day 7 of: 3) saline; 4) rat MDSC (106 cells); 5) as #4, with sildenafil as in #2. Before surgery, and at 1 and 4 weeks, the left ventricle ejection fraction (LVEF) was measured. LV sections were stained for collagen, myofibroblasts, apoptosis, cardiomyocytes, and iNOS, followed by quantitative image analysis. Western blots estimated angiogenesis and myofibroblast accumulation, as well as potential sildenafil tachyphylaxis by PDE 5 expression. Zymography estimated MMPs 2 and 9 in serum. RESULTS: As compared to untreated MI rats, sildenafil improved LVEF, reduced collagen, myofibroblasts, and circulating MMPs, and increased cardiac troponin T. MDSC replicated most of these effects and stimulated cardiac angiogenesis. Concurrent MDSC/sildenafil counteracted cardiomyocyte and endothelial cells loss, but did not improve LVEF or angiogenesis, and upregulated PDE 5. CONCLUSIONS: Long-term oral sildenafil, or MDSC given separately, reduce the MI fibrotic scar and improve left ventricular function in this rat model. The failure of the treatment combination may be due to inducing overexpression of PDE5.


Subject(s)
Myocardial Infarction/drug therapy , Myocardium/cytology , Phosphodiesterase Inhibitors/therapeutic use , Piperazines/therapeutic use , Stem Cells/drug effects , Sulfones/therapeutic use , Animals , Male , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Purines/therapeutic use , Rats , Rats, Inbred F344 , Sildenafil Citrate , Sulfones/pharmacology
12.
J Sex Med ; 8(4): 1048-60, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21269401

ABSTRACT

INTRODUCTION: It has been shown that phosphodiesterase type 5 (PDE5) inhibitors preserve smooth muscle (SM) content and ameliorate the fibrotic degeneration normally seen in the corpora cavernosa after bilateral cavernosal nerve resection (BCNR). However, the downstream mechanisms by which these drugs protect the corpora cavernosa remain poorly understood. AIM: To provide insight into the mechanism, we aimed to determine the gene expression profile of angiogenesis-related pathways within the penile tissue after BCNR with or without continuous sildenafil (SIL) treatment. METHODS: Five-month-old Fisher rats were subjected to BCNR or sham operation and treated with or without SIL (20 mg/kg/BW drinking water) for 3 days or 45 days (N = 8 rats per group). Total RNAs isolated from the denuded penile shaft and prostate were subjected to reverse transcription and to angiogenesis real-time-polymerase chain reaction arrays (84 genes). Changes in protein expression of selected genes such as epiregulin (EREG) and connective tissue growth factor (CTGF) were corroborated by Western blot and immunohistochemistry. MAIN OUTCOMES MEASURES: Genes modulated by BCNR and SIL treatment. RESULTS: A decreased expression of genes related to SM growth factors such as EREG, platelet-derived growth factor (PDGF), extracellular matrix regulators such as metalloproteinases 3 and 9, endothelial growth factors, together with an upregulation of pro-fibrotic genes such as CTGF and transforming growth factor beta 2 were found at both time points after BCNR. SIL treatment reversed this process by upregulating endothelial and SM growth factors and downregulating pro-fibrotic factors. SIL did not affect the expression of EREG, VEGF, and PDGF in the ventral prostate of BCNR animals. CONCLUSIONS: SIL treatment after BCNR activates genes related to SM preservation and downregulates genes related to fibrosis in the corpora cavernosa. These results provide a mechanistic justification for the use of SIL and other PDE5 inhibitors as protective therapy against corporal SM loss and fibrosis after radical prostatectomy.


Subject(s)
Extracellular Matrix/drug effects , Fibrosis/drug therapy , Gene Expression/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Penis/surgery , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Disease Models, Animal , Endothelium, Vascular/drug effects , Epidermal Growth Factor/drug effects , Epiregulin , Fibrosis/pathology , Gene Expression/genetics , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 3/drug effects , Nerve Tissue/injuries , Penis/blood supply , Penis/innervation , Purines/pharmacology , Rats , Sildenafil Citrate , Transforming Growth Factor beta2/drug effects , Transforming Growth Factor beta2/genetics
13.
Obstet Gynecol ; 114(2 Pt 1): 300-309, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19622991

ABSTRACT

OBJECTIVES: Grafts are used for vaginal repair after prolapse, but their use to carry stem cells to regenerate vaginal tissue has not been reported. In this study, we investigated whether 1) muscle-derived stem cells (MDSC) grown on small intestinal submucosa (SIS) generate smooth-muscle cells (SMC) in vitro and upon implantation in a rat model of vaginal defects; 2) express markers applicable to the in-vivo detection of vaginal endogenous stem cells; and 3) stimulate the repair of the vagina. METHODS: Mouse MDSC grown on monolayer, SIS, or polymeric mesh, were tested for cell differentiation by immunocytochemistry, Western blot and real-time polymerase chain reaction (PCR). Stem cell markers were screened by DNA microarrays followed by real-time PCR, immunocytochemistry, and Western blot. Rats that underwent hysterectomy and partial vaginectomy were left as such or implanted in the vagina with 4',6-Diamidino-2-Phenylindole (DAPI)-labeled MDSC on SIS, or SIS without MDSC, immunosuppressed, and killed at 2-8 weeks. Immunofluorescence, hematoxylin-eosin, and Masson trichrome were applied to tissue sections. RESULTS: Muscle-derived stem cell cultures on monolayer and on scaffolds differentiate into SMC, as shown by alpha-smooth muscle actin (ASMA), calponin, and smoothelin markers. Muscle-derived stem cells express embryonic stem cell markers Oct-4 and nanog. Dual DAPI/ASMA fluorescence indicated MDSC conversion to SMC. Muscle-derived stem cells/SIS stimulated vaginal tissue repair, including keratin-5 positive epithelium formation and prevented fibrosis at 4 and 8 weeks. Oct-4+ putative endogenous stem cells were identified. CONCLUSION: Muscle-derived stem cells/SIS implants stimulate vaginal tissue repair in the rat, thus autologous MDSC on scaffolds may be a promising approach for the treatment of vaginal repair.


Subject(s)
Stem Cell Transplantation/methods , Tissue Scaffolds , Vagina/surgery , Animals , Cells, Cultured , Female , Intestinal Mucosa , Intestine, Small , Mice , Mice, Inbred C57BL , Muscle Cells/cytology , Myocytes, Smooth Muscle/cytology , Rats , Rats, Inbred F344 , Transplantation, Heterologous , Uterine Prolapse/surgery
14.
J Sex Med ; 6(2): 415-28, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19138364

ABSTRACT

INTRODUCTION: Corporal veno-occlusive dysfunction (CVOD), which usually is associated with a loss of smooth muscle cells (SMC) and an increase in fibrosis within the corpora cavernosa, can be induced by an injury to the cavernosal nerves. The corporal tissue expresses inducible nitric oxide synthase (iNOS), presumably as an antifibrotic and SMC-protective response. AIMS: We studied the temporal relationship in the corpora between the expression of iNOS, other histological and biochemical changes, and the development of CVOD, after bilateral cavernosal nerve resection (BCNR) in the rat. METHODS: Rats underwent either BCNR or sham operation. Cavernosometry was performed 1, 3, 7, 15, 30, and 45 days (N = 8/groups) after surgery. Penile tissue sections were subjected to Masson trichrome staining for SMC and collagen, and immunodetection for alpha smooth muscle actin, iNOS, neuronal NOS (nNOS), endothelial NOS (eNOS), proliferating cell nuclear antigen (PCNA), and terminal transferase dUTP nick end labeling (TUNEL). Quantitative western blot analysis was done in homogenates. MAIN OUTCOME MEASURES: Time course on the development of fibrosis and CVOD. RESULTS: Following BCNR, CVOD was detectable 30 days later, and it became more pronounced by 45 days. In contrast, the SMC/collagen ratio in the BCNR corpora was reduced at 7 days and bottomed at 30 and 45 days, due in part to the reduction of SMC, presumably caused by an increase in apoptosis peaking at 3 days. PCNA also peaked at 3 days, but then decayed. nNOS was reduced early (3-7 days) and disappeared at 30 days, whereas eNOS was not affected. iNOS was induced at day 3, and steadily increased peaking at 30 days. CONCLUSIONS: CVOD develops in the BCNR rat as a result of the early loss of corporal SMC by the neuropraxia-induced apoptosis, which the initial cell replication response cannot counteract, followed by fibrosis. The time course of iNOS induction supports the antifibrotic role of iNOS.


Subject(s)
Fibrosis/pathology , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Penis , Peripheral Nerves/pathology , Peripheral Nerves/physiopathology , Actins/metabolism , Animals , Blotting, Western , Endothelium/metabolism , In Situ Nick-End Labeling , Male , Muscle, Smooth/metabolism , Nitric Oxide Synthase/metabolism , Penis/innervation , Penis/pathology , Penis/physiopathology , Proliferating Cell Nuclear Antigen/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley
15.
BJU Int ; 103(10): 1396-404, 2009 May.
Article in English | MEDLINE | ID: mdl-19154511

ABSTRACT

OBJECTIVES: To determine, in the obese Zucker fa/fa rat (OZR), whether the loss in smooth muscle cells (SMCs) as well as the increase in fibrosis that occurs within the corpora cavernosa accompanying corporal veno-occlusive dysfunction (CVOD), also occurs within the media of the arterial tree. MATERIALS AND METHODS: The penis and aorta from both 7-month-old male diabetic OZR (5 months of diabetes) and aged-matched nondiabetic lean Zucker rats (LZR) rats were harvested (eight per group). The penis and aorta were subjected to histo- or immnohistochemistry, followed by quantitative image analysis (QIA) to determine the contents of SMC, collagen and the pro-fibrotic transforming growth factor (TGF)beta1. The turnover of SMCs was assessed by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labelling (TUNEL) and proliferating cell nuclear antigen (PCNA) assays. Quantitative Western blots determined calponin (SMC marker) and PCNA, and hydroxyproline was used for collagen. In vitro relaxation of corporal strips was measured. RESULTS: In vitro relaxation of corporal tissue from OZR was considerably less than in the LZR. In the media of the penile dorsal artery (PDA) of OZR, there was a considerable reduction in the SMC content and the SMC/collagen ratio, as well as an increase in apoptosis, but there were no changes in PCNA or TGFbeta1 expression, or in the intima-media/lumen ratio. In the aorta of the OZR, in contrast to the PDA, there was a reduction in PCNA as well as a more pronounced decrease in the SMC/collagen ratio, mainly from an increase in collagen, but there were no changes in TGFbeta1 or the wall/lumen morphometry. In the OZR, Western blots of aortic tissue confirmed the decrease in PCNA and a reduction in the SMC marker calponin. CONCLUSIONS: These data show that 5 months after the onset of hyperglycaemia in the OZR, the rats develop both abnormal corporal SMC relaxation and a generalized fibrosis of the arterial media of both the large and small diameter vessels. It is possible that this pan-fibrosis of the media of the arterial system might contribute to the diabetes-related ED that occurs during this period in this rat model.


Subject(s)
Aorta/pathology , Diabetes Mellitus, Type 2/pathology , Impotence, Vasculogenic/pathology , Penis/blood supply , Tunica Media/pathology , Animals , Blotting, Western , Diabetes Mellitus, Type 2/complications , Fibroblasts/pathology , Fibrosis , Immunohistochemistry , Impotence, Vasculogenic/etiology , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Penis/pathology , Rats , Rats, Zucker
16.
BJU Int ; 101(9): 1156-64, 2008 May.
Article in English | MEDLINE | ID: mdl-18294308

ABSTRACT

OBJECTIVE: To determine whether skeletal muscle-derived stem cells (MDSCs) convert into smooth muscle cells (SMCs) both in vitro and in vivo, and in so doing ameliorate the erectile dysfunction (ED) of aged rats, and whether endogenous stem cells are present in the rat corpora cavernosa. MATERIALS AND METHODS: MDSCs were obtained from mouse muscle, and shown by immunocytochemistry for alpha-smooth muscle actin (alpha SMA) to originate in vitro in myofibroblasts and SMCs, discriminating SMCs by calponin 1 expression. In vivo these MDSCs, labelled with 4',6-diamidino-2-phenylindole, were implanted into the corpora cavernosa of young adult (5-month old) and aged (20-month old) rats for 2 and 4 weeks. Histological changes were assessed by immunohistochemistry and quantitative Western blot. Functional changes were determined by electrical field stimulation (EFS) of the cavernosal nerve. RESULTS: The exogenous cells replicated and converted into SMCs, as shown in corporal tissue sections by confocal immunofluorescence microscopy for proliferating cell nuclear antigen (PCNA), alpha SMA, and smoothelin, and also by Western blot for alpha SMA and PCNA. MDSC differentiation was confirmed by the activation of the alpha SMA promoter-linked beta-galactosidase in transfected cells, both in vitro and after implantation in the corpora. Putative endogenous stem cells were shown in corporal tissue sections and Western blots by detecting CD34 and a possible Sca1 variant. EFS showed that implanted MDSCs raised in aged rats the maximal intracavernosal pressure/mean arterial pressure levels above (2 weeks) or up to (4 weeks) those of young adult rats. CONCLUSIONS: MDSCs implanted into the corpora cavernosa of aged rats converted into SMCs and corrected ED, and endogenous cells expressing stem cell markers were also found in untreated tissue. This suggests that exogenous stem cell implantation and/or endogenous stem cell modulation might be viable therapeutic approaches for ageing-related ED.


Subject(s)
Impotence, Vasculogenic/therapy , Muscle, Skeletal/cytology , Myocytes, Smooth Muscle/transplantation , Penis/pathology , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Blotting, Western , Cell Differentiation , Immunohistochemistry , Impotence, Vasculogenic/pathology , Male , Myocytes, Smooth Muscle/cytology , Rats , Rats, Sprague-Dawley
17.
BJU Int ; 101(2): 203-10, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17888043

ABSTRACT

OBJECTIVES: To determine whether a long-term single daily oral dose of a longer half-life phosphodiesterase-5 (PDE5) inhibitor, tadalafil, has a similar effect to that of the shorter half-life PDE5 inhibitors sildenafil and vardenafil, and can prevent the fibrosis and resultant corporal veno-occlusive dysfunction (CVOD) occurring after cavernosal nerve (CN) injury. MATERIALS AND METHODS: Male rats (10 per group) had either a sham operation, unilateral CN resection (CNR) or bilateral CNR, and were left untreated or given retrolingually 5 mg/kg per day of tadalafil. After 45 days, CVOD was assessed via cavernosometry, and the underlying corporal tissue changes were examined by immunohistochemistry and histochemistry (followed by quantitative image analysis), Western blots, and ad hoc methods. RESULTS: Tadalafil treatment normalized the low response to papaverine and high drop rate in the intracavernosal pressure measured by cavernosometry after CNR compared with sham-operated rats. Tadalafil also normalized the increase in penile shaft collagen content, and the reduction in corporal smooth muscle cell (SMC) content, SMC/collagen, and replication index, and improved the lower collagen III/I ratio and the increase in apoptotic index, caused by CNR, compared with sham operation. There were no effects of tadalafil on increased transforming growth factor beta1, inducible nitric oxide synthase and xanthine oxidoreductase levels. CONCLUSIONS: A long-term single daily dose of tadalafil prevented CVOD and the underlying corporal fibrosis in the rat caused by CN damage, as effectively as the previously reported continuous treatment with vardenafil or sildenafil, through a cGMP-related mechanism that appears to be independent of inducible nitric oxide synthase induction.


Subject(s)
Carbolines/administration & dosage , Impotence, Vasculogenic/prevention & control , Penis , Phosphodiesterase Inhibitors/administration & dosage , Prostatectomy/adverse effects , Animals , Blotting, Western , Denervation/methods , Fibrosis/prevention & control , Impotence, Vasculogenic/pathology , Male , Muscle, Smooth/drug effects , Penis/blood supply , Penis/innervation , Penis/pathology , Prostatectomy/methods , Rats , Rats, Sprague-Dawley , Tadalafil
18.
BJU Int ; 100(4): 867-74, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17662078

ABSTRACT

OBJECTIVE: To determine whether ageing-related changes in the penile corpora cavernosa, namely corporal veno-occlusive dysfunction (CVOD), loss of smooth muscle cells (SMCs), and excessive collagen deposition, can be ameliorated by the peroxisome proliferator-activated receptor gamma (PPARgamma) agonist pioglitazone, in a rat model of ageing as we have shown in a rat model of type 2 diabetes. MATERIALS AND METHODS: Male Fischer 344 rats (16-18 months old) were fed chow containing 0%, 0.001% or 0.02% pioglitazone for 2 or 4.5 months, using 5 month old rats as 'young' controls. Functional changes were determined by dynamic-infusion cavernosometry (DIC). Histological changes were assessed by histochemistry and immunohistochemistry followed by quantitative image analysis and/or quantitative Western blot. Reactive oxygen species were estimated in blood. RESULTS: Pioglitazone at both doses reduced the high DIC 'drop rate' present in the untreated aged groups to the level seen in the young rats. The papaverine response was increased to young control levels by short-term high-dose pioglitazone and the long-term low-dose treatment, but not by the short-term low-dose treatment. Pioglitazone at all doses and durations of treatment failed to reverse the decreased corporal SMC/collagen ratio and SMC content, oxidative stress, or the elevated contents of collagen, or transforming growth factor beta1, seen in the aged penis, but did reduce the collagen III/I ratio, and at a high dose increased apoptosis. Both treatments inhibited the Rho-kinase system, by increasing Src homology region 2-containing protein tyrosine phosphatase and reducing Vav. PPARgamma were detected in corporal SMCs. CONCLUSIONS: Pioglitazone ameliorated ageing-related CVOD, possibly by a PPARgamma-mediated inhibition of Rho-kinase and not by a protective effect on the corporal smooth muscle.


Subject(s)
Collagen/drug effects , Impotence, Vasculogenic/prevention & control , PPAR gamma/agonists , Penis/pathology , Thiazolidinediones/administration & dosage , Age Factors , Animals , Blotting, Western , Fibrosis , Immunohistochemistry , Impotence, Vasculogenic/pathology , Male , Oxidative Stress/drug effects , PPAR gamma/metabolism , Penis/drug effects , Pioglitazone , Rats , Rats, Inbred F344 , Reactive Oxygen Species
19.
J Sex Med ; 4(3): 633-643, 2007 May.
Article in English | MEDLINE | ID: mdl-17433082

ABSTRACT

INTRODUCTION: Over-expression of penile neuronal nitric oxide synthase (PnNOS) from a plasmid ameliorates aging-related erectile dysfunction (ED), whereas over-expression of the protein inhibitor of NOS (PIN), that binds to nNOS, increases ED. AIM: To improve this form of gene therapy for ED by comparing the electrical field response of short hairpin RNA (shRNA) for PIN with that of antisense PIN RNA. MAIN OUTCOME MEASURE: Both shRNA and antisense RNA gene therapy vectors increased intracavernosal pressure in aged rats. METHODS: PIN small interfering RNA (siRNA), and plasmid constructs for cytomegalovirus promoter plasmid vector (pCMV-PIN), pCMV-PIN antisense RNA, pSilencer2.1-U6-PIN-shRNA; and pSilencer2.1-U6-randomer-shRNA were prepared and validated by transfection into HEK293 cells, determining the effects on PIN expression by Western blot. Plasmid constructs were then injected, followed by electroporation, into the penile corpora cavernosa of aged (20-month-old) Fisher 344 rats and, 1 month later, the erectile response was measured by intracavernosal pressure increase following electrical field stimulation (EFS) of the cavernosal nerve. PIN was estimated in penile tissue by Western blot and real-time reverse transcriptase-polymerase chain reaction. Cyclic guanosine monophosphate (cGMP) measurements were conducted by competitive enzyme immunoassay (EIA). Immunohistofluorescence detected PIN in corporal tissue sections. RESULTS: In cell culture, PIN siRNA and plasmid-expressed pU6-PIN-shRNA effectively reduced PIN expression from pCMV-PIN. pSilencer2.1-U6-PIN-shRNA corrected the impaired erectile response to EFS in aged rats and raised it above the value for young rats, more efficiently than pCMV-PIN antisense RNA. PIN mRNA expression in the penis was decreased by >70% by the shRNA but remained unaffected by the antisense RNA, whereas PIN protein expression was reduced in both cases, particularly in the dorsal nerve. PIN antisense increased cGMP concentration in treated tissue by twofold. CONCLUSION: pSilencer2.1-U6-PIN-shRNA gene therapy was more effective than the antisense PIN mRNA in ameliorating ED in the aged rat, thereby suggesting that PIN is indeed a physiological inhibitor of nNOS and nitrergic neurotransmission in the penis.


Subject(s)
Aging/genetics , Erectile Dysfunction/drug therapy , Erectile Dysfunction/genetics , Genetic Therapy/methods , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , RNA, Small Interfering/pharmacology , Animals , Blotting, Western , Disease Models, Animal , Male , Nitric Oxide Synthase Type I , Penile Erection/drug effects , Penile Erection/physiology , Penis/enzymology , Penis/metabolism , Rats , Rats, Inbred F344
20.
Urology ; 68(2): 429-35, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16904479

ABSTRACT

OBJECTIVES: Impotence, specifically corporal veno-occlusive dysfunction (CVOD), occurs after radical prostatectomy. It results from the effects of cavernosal nerve damage, which causes smooth muscle (SM) loss and an increase in collagen within the corpora. Recent reports have suggested that long-term treatment with phosphodiesterase-5 inhibitors after radical prostatectomy may prevent such changes. We aimed to determine whether bilateral cavernosal nerve resection (BCNX) in the rat leads to CVOD and whether long-term phosphodiesterase-5 inhibition ameliorates these histologic and functional impairments. METHODS: Rats (n = 7 to 11/group) underwent either the sham operation, BCNX, or BCNX plus 30 mg/L vardenafil in the drinking water. Before the rats were killed 45 days later, CVOD was assessed by dynamic infusion cavernosometry. The corpora underwent histochemistry/immunohistochemistry with quantitative image analysis for SM/collagen ratio, collagen III/I ratio, alpha-SM actin, inducible nitric oxide synthase (iNOS), proliferating cell nuclear antigen, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling as a marker of apoptosis. RESULTS: Compared with the sham group, the BCNX rats demonstrated CVOD as measured by the drop rate, a 60% reduction in the SM/collagen ratio, a twofold increase in iNOS expression, and a threefold increase in intracorporeal apoptosis. Compared with the BCNX group, vardenafil increased both iNOS and proliferating cell nuclear antigen expression (SM cell replication), with normalization of the dynamic infusion cavernosometry drop rate and SM/collagen ratio. CONCLUSIONS: Long-term treatment with vardenafil may prevent CVOD after radical prostatectomy by preserving SM content and inhibiting corporal fibrosis possibly by its effect on iNOS.


Subject(s)
Imidazoles/therapeutic use , Penis/innervation , Penis/pathology , Phosphodiesterase Inhibitors/therapeutic use , Piperazines/therapeutic use , Vascular Diseases/prevention & control , Animals , Denervation/methods , Fibrosis , Male , Muscle, Smooth , Penis/blood supply , Rats , Rats, Inbred F344 , Sulfones/therapeutic use , Triazines/therapeutic use , Vardenafil Dihydrochloride
SELECTION OF CITATIONS
SEARCH DETAIL
...