Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
World J Urol ; 38(11): 2915-2921, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31996980

ABSTRACT

PURPOSE: To examine phosphodiesterase type 5 (PDE5) expression in the anterior fibromuscular stroma (AFMS) of the prostate. Although PDE5 expression was identified in the human prostate, differences in PDE5 expression in intra-prostatic regions are unknown. The AFMS in the prostate has peculiar innervations that could contribute to voiding function. Here, we examined regional differences in PDE5 expression in the prostate with special reference to the AFMS. METHODS: A total 18 human prostate and bladder specimens were obtained. Tissue specimens were processed by hematoxylin-eosin (H&E) staining and immunohistochemistry for PDE5. Immunoreactivity with PDE5 was evaluated using computer-assisted image analysis in the following regions: the AFMS, bladder neck, stromal hyperplasia in the transition zone, glandular hyperplasia in the transition zone (TZ gland), and the peripheral zone (PZ). The correlation between PDE5 expression in the AFMS and clinical data was analysed. RESULTS: Image analysis revealed that the median ratio of the PDE5-immunoreactive area to smooth muscle area by H&E staining was 74.7% in the AFMS. There was significantly higher PDE5 expression in the AFMS than in the TZ gland (p = 0.034) and PZ (p = 0.002). PDE5 expression in the AFMS was not significantly correlated with age, prostate volume, transition zone volume, or transition zone index. However, older men had a tendency to have higher PDE5 expression in the AFMS. CONCLUSIONS: We found higher PDE5 expression in the AFMS compared with other prostatic regions, which suggested that the AFMS is a target region of PDE5 inhibitors in the prostate.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Prostate/metabolism , Aged , Cyclic Nucleotide Phosphodiesterases, Type 5/analysis , Humans , Immunohistochemistry , Male , Middle Aged , Prostate/anatomy & histology , Prostate/chemistry
2.
Andrology ; 8(2): 427-433, 2020 03.
Article in English | MEDLINE | ID: mdl-31433119

ABSTRACT

BACKGROUND: Type 5 phosphodiesterase (PDE5) expression in the normal and pathological prostate is controversial. OBJECTIVES: This study aimed at identifying the cell type/s, if any, expressing PDE5 in human healthy or pathological prostate sections in order to further validate the rationale of PDE5 inhibitor (PDE5i) treatment of benign prostatic hyperplasia (BPH) and their safety in the treatment of erectile dysfunction following prostate cancer (PCa) surgery. MATERIALS AND METHODS: By immunohistochemical analysis, we studied PDE5 expression in tissue microarrays containing sections obtained from healthy, BPH, and PCa samples. RESULTS: Our results showed that PDE5 is barely expressed in the epithelial or stromal compartment of normal human prostates, but it is highly expressed in the stromal compartment of BPH sections. We also found that a low but significant number of PCa samples (22%) expressed PDE5 in the epithelial cancer cells but not in stromal cells and that such expression was not correlated with the tumor aggressiveness, according to their Gleason score. DISCUSSION AND CONCLUSION: PDE5 overexpression in the stromal compartment of BPH samples supports the rationale of PDE5 as a target in lower urinary tract symptoms of BPH. PDE5 expression in a significant percentage of PCa samples but the lack of correlation with the Gleason score suggests that this enzyme is not correlated with tumor aggressiveness; however, a role of PDE5 in the minimal residual disease of PCa cannot be excluded.


Subject(s)
Adenocarcinoma/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Prostate/enzymology , Prostatic Hyperplasia/enzymology , Prostatic Neoplasms/enzymology , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cyclic Nucleotide Phosphodiesterases, Type 5/analysis , Humans , Male , Middle Aged , Prostate/pathology , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , Young Adult
3.
Sci Rep ; 9(1): 12206, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434939

ABSTRACT

Aneurysms and dissections affecting thoracic aorta are associated with smooth muscle cell (SMC) dysfunction. NO/cGMP signaling pathway in smooth muscle cells has been shown to be affected in sporadic thoracic aortic aneurysms. We analyzed the mRNA levels of PDE5, a cGMP-hydrolyzing enzyme highly expressed in aortic SMCs, that regulates arterious vascular tone by lowering cGMP levels. We found that aortic tissue obtained from Marfan, tricuspid and bicuspid thoracic aneurysms expressed lower levels of PDE5 mRNA compared to control aortas. In particular, we found that affected aortas showed lower levels of all the PDE5A isoforms, compared to control aortas. Transfection of vascular SMCs (VSMCs) with NOTCH3 activated domain (NICD3) induced the expression of PDE5A1 and A3 protein isoforms, but not that of the corresponding mRNAs. VSMC stimulation with GSNO, a nitric oxide analogue or with 8-br-cGMP, but not with 8-br-cAMP, up-regulated PDE5 and NOTCH-3 protein levels, indicating a negative feedback loop to protect the arterial wall from excessive relaxation. Finally, we found that PDE5 is expressed early during human aorta development, suggesting that if loss of function mutations of PDE5 occur, they might potentially affect aortic wall development.


Subject(s)
Aortic Aneurysm, Thoracic/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Gene Expression Regulation, Enzymologic , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Adult , Aged , Aortic Aneurysm, Thoracic/pathology , Female , Humans , Isoenzymes/biosynthesis , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology
4.
World Neurosurg ; 132: e795-e801, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31404697

ABSTRACT

OBJECTIVE: To examine changes of expression and activity of phosphodiesterase V (PDE V) in the basilar artery following cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH) in a rabbit model. METHODS: A rabbit model of CVS after SAH was constructed by double blood injection into the cisterna magna. Subjects were divided into 3 groups: blank control group, normal saline group, and SAH group. Transcranial Doppler and selective vertebrobasilar digital subtraction angiography were performed to identify changes of CVS. Changes of PDE V expression and activity were examined. RESULTS: Mean basilar arterial blood flow rate measured by transcranial Doppler was significantly increased in the SAH group compared with the blank control group and normal saline group. Mean basilar artery diameter measured by digital subtraction angiography in the SAH group was narrower than in the other 2 groups. Compared with the other 2 groups, the expression of PDE V in the SAH group was significantly upregulated, and the activity was significantly enhanced. CONCLUSIONS: The rabbit model of SAH-induced CVS was successfully constructed through double blood injection method. Increased basilar artery blood flow, narrowing of the basilar artery, increased PDE V expression, and enhanced PDE V activity in the basilar artery were detected in the CVS rabbits, suggesting that PDE V has the potential to be used as a target for CVS therapy.


Subject(s)
Basilar Artery/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Vasospasm, Intracranial/enzymology , Angiography, Digital Subtraction , Animals , Basilar Artery/diagnostic imaging , Cerebrovascular Circulation , Cisterna Magna , Disease Models, Animal , Immunohistochemistry , Male , Rabbits , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/enzymology , Ultrasonography, Doppler, Transcranial , Vasospasm, Intracranial/diagnostic imaging , Vasospasm, Intracranial/etiology
5.
Urology ; 85(3): 703.e7-13, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25733302

ABSTRACT

OBJECTIVE: To investigate the level and location of phosphodiesterase 5 (PDE5) expression in rat prostate. METHODS: The ventral, dorsal, and lateral lobes of rat prostate were examined for PDE5 expression by Western blotting. Intact rat urogenital complex, including the urinary bladder and accessory reproductive glands, was examined for PDE5 expression by immunohistochemistry. Individual prostatic lobes were further examined by immunofluorescence for expression of PDE5, α-smooth muscle actin, and rat endothelial cell antigen. RESULTS: Western blot analysis showed that PDE5 was expressed at a significantly lower level in dorsal lobe (DL) than in ventral lobe (VL) or lateral lobe (LL). Immunohistochemistry and immunofluorescence analyses showed that PDE5 was expressed in both acinar epithelium and periacinar smooth muscle. However, although similar levels of smooth muscle PDE5 expression were observed in all 3 prostatic lobes, significantly lower level of epithelial PDE5 expression was found in DL compared with VL or LL. In prostatic blood vessels, PDE5 expression was clearly visible in the endothelium but not as easily detectable in the smooth muscle. CONCLUSION: PDE5 was expressed in the acinar epithelium and periacinar smooth muscle of rat prostate. However, the epithelial PDE5 expression was significantly less in DL than in VL or LL. Regardless, the acinar wall, not the blood vessel wall, is the predominant PDE5 expression site in rat prostate.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Prostate/metabolism , Animals , Male , Prostate/anatomy & histology , Rats , Rats, Sprague-Dawley
6.
J Cell Physiol ; 230(9): 2281-98, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25704960

ABSTRACT

We determined whether the multi-kinase inhibitor sorafenib or its derivative regorafenib interacted with phosphodiesterase 5 (PDE5) inhibitors such as Viagra (sildenafil) to kill tumor cells. PDE5 and PDGFRα/ß were over-expressed in liver tumors compared to normal liver tissue. In multiple cell types in vitro sorafenib/regorafenib and PDE5 inhibitors interacted in a greater than additive fashion to cause tumor cell death, regardless of whether cells were grown in 10 or 100% human serum. Knock down of PDE5 or of PDGFRα/ß recapitulated the effects of the individual drugs. The drug combination increased ROS/RNS levels that were causal in cell killing. Inhibition of CD95/FADD/caspase 8 signaling suppressed drug combination toxicity. Knock down of ULK-1, Beclin1, or ATG5 suppressed drug combination lethality. The drug combination inactivated ERK, AKT, p70 S6K, and mTOR and activated JNK. The drug combination also reduced mTOR protein expression. Activation of ERK or AKT was modestly protective whereas re-expression of an activated mTOR protein or inhibition of JNK signaling almost abolished drug combination toxicity. Sildenafil and sorafenib/regorafenib interacted in vivo to suppress xenograft tumor growth using liver and colon cancer cells. From multiplex assays on tumor tissue and plasma, we discovered that increased FGF levels and ERBB1 and AKT phosphorylation were biomarkers that were directly associated with lower levels of cell killing by 'rafenib + sildenafil. Our data are now being translated into the clinic for further determination as to whether this drug combination is a useful anti-tumor therapy for solid tumor patients.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Phosphodiesterase 5 Inhibitors/administration & dosage , Piperazines/administration & dosage , Sulfonamides/administration & dosage , Apoptosis/drug effects , Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Drug Synergism , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Hep G2 Cells , Humans , Neoplasm Proteins/biosynthesis , Neoplasms/genetics , Neoplasms/pathology , Niacinamide/administration & dosage , Purines/administration & dosage , Signal Transduction/drug effects , Sildenafil Citrate , Sorafenib , Xenograft Model Antitumor Assays
7.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L868-76, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25326578

ABSTRACT

Excess superoxide has been implicated in pulmonary hypertension (PH). We previously found lung overexpression of the antioxidant extracellular superoxide dismutase (EC-SOD) attenuates PH and pulmonary artery (PA) remodeling. Although comprising a small fraction of total SOD activity in most tissues, EC-SOD is abundant in arteries. We hypothesize that the selective loss of vascular EC-SOD promotes hypoxia-induced PH through redox-sensitive signaling pathways. EC-SOD(loxp/loxp) × Tg(cre/SMMHC) mice (SMC EC-SOD KO) received tamoxifen to conditionally deplete smooth muscle cell (SMC)-derived EC-SOD. Mice were exposed to hypobaric hypoxia for 35 days, and PH was assessed by right ventricular systolic pressure measurements and right ventricle hypertrophy. Vascular remodeling was evaluated by morphometric analysis and two-photon microscopy for collagen. We examined cGMP content and soluble guanylate cyclase expression and activity in lung, lung phosphodiesterase 5 (PDE5) expression and activity, and expression of endothelial nitric oxide synthase and GTP cyclohydrolase-1 (GTPCH-1), the rate-limiting enzyme in tetrahydrobiopterin synthesis. Knockout of SMC EC-SOD selectively decreased PA EC-SOD without altering total lung EC-SOD. PH and vascular remodeling induced by chronic hypoxia was augmented in SMC EC-SOD KO. Depletion of SMC EC-SOD did not impact content or activity of lung soluble guanylate cyclase or PDE5, yet it blunted the hypoxia-induced increase in cGMP. Although total eNOS was not altered, active eNOS and GTPCH-1 decreased with hypoxia only in SMC EC-SOD KO. We conclude that the localized loss of PA EC-SOD augments chronic hypoxic PH. In addition to oxidative inactivation of NO, deletion of EC-SOD seems to reduce eNOS activity, further compromising pulmonary vascular function.


Subject(s)
Hypertension, Pulmonary/therapy , Hypoxia/therapy , Superoxide Dismutase/genetics , Animals , Blood Pressure , Cyclic GMP/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Estrogen Antagonists/pharmacology , GTP Cyclohydrolase/biosynthesis , Guanylate Cyclase/biosynthesis , Hypertrophy, Right Ventricular/physiopathology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/biosynthesis , Pulmonary Artery/pathology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Signal Transduction , Soluble Guanylyl Cyclase , Tamoxifen/pharmacology
9.
J Urol ; 192(1): 267-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24316094

ABSTRACT

PURPOSE: We determined the effects of low androgens in the neonatal period on biomarkers of smooth muscle cell differentiation, Myh11 and Acta2, and on Pde5A expression in the penis. MATERIALS AND METHODS: One-day-old pups were treated daily with the gonadotropin-releasing hormone antagonist antide with or without dihydrotestosterone for 1 to 6 days. Tissues were collected at age day 7 and at adulthood at age 120 days. Penes were examined by quantitative reverse transcriptase-polymerase chain reaction, Western blot and immunohistochemistry. Testes were assayed for the intratesticular testosterone and steroidogenic enzymes Cyp17α1 and StAR. RESULTS: Gonadotropin-releasing hormone antagonist exposure suppressed the neonatal testicular testosterone surge 70% to 80%. Quantitative reverse transcriptase-polymerase chain reaction revealed 80% to 90% reductions in Cyp17α1 and StAR protein, and 40% to 60% reductions in Myh11 and ACTA2 as a result of gonadotropin-releasing hormone antagonist compared to controls. Dihydrotestosterone co-administration mitigated these decreases. Western blot confirmed the Myh11 decrease at the protein level. Immunohistochemistry of Acta2 confirmed cavernous smooth muscle cell loss at the tissue level. Also, gonadotropin-releasing hormone antagonist exposure decreased Pde5a expression and dihydrotestosterone co-administration mitigated the decrease. Comparison of data between 2 parts of the penis body (corpora cavernosa and corpus spongiosum) showed that antagonist induced decreases in Myh11, Acta2 and Pde5a expression occurred only in the corpora cavernosa, implying that the latter is the target site of low androgen action. CONCLUSIONS: As evidenced by gonadotropin-releasing hormone antagonist induced suppression of the neonatal testosterone surge and reduced steroidogenesis, low androgens in the neonatal period altered gene expression of biomarkers of smooth muscle cell differentiation. This led to loss of cavernous smooth muscle cells and consequently to penile maldevelopment.


Subject(s)
Androgens/deficiency , Cell Differentiation , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Myosin Heavy Chains/biosynthesis , Penis/abnormalities , Penis/growth & development , Actins/biosynthesis , Animals , Animals, Newborn , Biomarkers , Gene Expression Regulation , Male , Rats , Rats, Sprague-Dawley
10.
J Urol ; 190(4): 1430-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23545097

ABSTRACT

PURPOSE: Phosphodiesterase type 5 inhibitors were recently introduced as a new treatment option for men with lower urinary tract symptoms. Safety and clinical effectiveness are well documented but the mode of action is still unclear. We determined and compared the expression of phosphodiesterase type 5 in the spinal cord of normal (sham operated) rats and rats with partial urethral obstruction induced bladder overactivity. We also assessed the urodynamic effects of intravenously and intrathecally administered sildenafil in the rats to determine whether phosphodiesterase type 5 inhibitors exert effects on the sacral spinal cord. MATERIALS AND METHODS: A total of 65 male Sprague-Dawley rats were used for molecular/morphological and functional experiments. Bladder overactivity was induced via surgical partial urethral obstruction in 39 of 65 rats. Spinal phosphodiesterase type 5 expression was assessed by histology and polymerase chain reaction. The effects of sildenafil administered intravenously or intrathecally were studied urodynamically. RESULTS: Phosphodiesterase type 5 was expressed in various regions of the lumbosacral spinal cord, including the sacral regions of micturition control. Expression was similar in normal rats and rats with partial urethral obstruction/bladder overactivity. In normal rats intravenous and intrathecal sildenafil had no urodynamic effect. When administered intravenously and intrathecally to rats with partial urethral obstruction/bladder overactivity, sildenafil decreased micturition frequency and bladder pressure. Doses tested intrathecally had no effect when given intravenously. CONCLUSIONS: Phosphodiesterase type 5 is expressed in the rat spinal cord. Intravenous sildenafil may exert part of its urodynamic effect in rats with partial urethral obstruction/bladder overactivity via an effect on the sacral spinal cord.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Spinal Cord/metabolism , Urinary Bladder Neck Obstruction/complications , Urinary Bladder, Overactive/etiology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/analysis , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Spinal Cord/chemistry , Spinal Cord/drug effects , Sulfones/pharmacology
11.
Urology ; 81(3): 480-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23333001

ABSTRACT

Both clinical and preclinical studies have mostly shown beneficial effects for Phosphodiesterase-5 (PDE-5) inhibitors in the treatment of lower urinary tract symptoms. Molecular studies have consistently shown abundant PDE-5 expression in bladder smooth muscle. Data concerning urethral PDE-5 expression have been surprising because striated muscle was not only positively identified, but also found to express more PDE-5 than the smooth muscle. In the prostate, highly variable results have been obtained. For PDE-5 expression, the data have ranged from extremely low to highly abundant. PDE-5 has been found in the glandular epithelium, vascular smooth muscle, endothelium, and fibromuscular stroma.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Urethra/physiology , Urinary Bladder/physiology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Humans
12.
Pediatr Pulmonol ; 47(11): 1088-96, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22328320

ABSTRACT

RATIONALE AND OBJECTIVES: Little is known about molecular changes in lungs of fetal rabbits with surgically induced diaphragmatic hernia (DH). Therefore, we examined in this model gene expressions of pivotal molecules for the developing lung. METHODS: At day 23 of gestation, DH was created in 12 fetuses from 4 does. Both lungs from six live DH fetuses and from six unoperated controls were harvested and weighed at term. Transcription of 15 genes involved in alveolarization, angiogenesis, regulation of vascular tone, or epithelial maturation was investigated by real-time quantitative polymerase chain reaction. MAIN RESULTS: DH decreased lung-to-body weight ratio (P < 0.001). A bilateral downregulation was seen for genes encoding for tropoelastin (P < 0.01), lysyl oxidase (P < 0.05), fibulin 5 (P < 0.05), and cGMP specific phosphodiesterase 5 (P < 0.05). Lower mRNA levels for endothelial nitric oxide synthase occurred in the ipsilateral lung (P < 0.05). CONCLUSIONS: Experimental DH in fetal rabbits disrupted transcription of genes implicated in lung growth and function. Similarities with the human disease make this model appropriate for investigation of new prenatal therapies.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hernias, Diaphragmatic, Congenital , Lung/growth & development , Lung/metabolism , Signal Transduction/physiology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Disease Models, Animal , Female , Fetal Organ Maturity/physiology , Fetus/metabolism , Gene Expression Profiling , Hernia, Diaphragmatic/metabolism , Hernia, Diaphragmatic/surgery , Humans , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Pregnancy , Protein-Lysine 6-Oxidase/biosynthesis , Protein-Lysine 6-Oxidase/genetics , Rabbits , Tropoelastin/biosynthesis , Tropoelastin/genetics
13.
J Neurotrauma ; 29(3): 528-38, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-21970599

ABSTRACT

Nitric oxide (NO) is an important regulator of vasodilation and angiogenesis in the central nervous system (CNS). Signaling initiated by the membrane receptor CD47 antagonizes vasodilation and angiogenesis by inhibiting synthesis of cyclic guanosine monophosphate (cGMP). We recently found that deletion of CD47 led to significant functional locomotor improvements, enhanced angiogenesis, and increased epicenter microvascular perfusion in mice after moderate contusive spinal cord injury (SCI). We tested the hypothesis that improving NO/cGMP signaling within the spinal cord immediately after injury would increase microvascular perfusion, angiogenesis, and functional recovery, with an acute, 7-day administration of the cGMP phosphodiesterase 5 (PDE5) inhibitor sildenafil. PDE5 expression is localized within spinal cord microvascular endothelial cells and smooth muscle cells. While PDE5 antagonism has been shown to increase angiogenesis in a rat embolic stroke model, sildenafil had no significant effect on angiogenesis at 7 days post-injury after murine contusive SCI. Sildenafil treatment increased cGMP concentrations within the spinal cord and improved epicenter microvascular perfusion. Basso Mouse Scale (BMS) and Treadscan analyses revealed that sildenafil treatment had no functional consequence on hindlimb locomotor recovery. These data support the hypothesis that acutely improving microvascular perfusion within the injury epicenter by itself is an insufficient strategy for improving functional deficits following contusive SCI.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Hindlimb/blood supply , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/therapeutic use , Spinal Cord Injuries/drug therapy , Sulfones/therapeutic use , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Capillaries/metabolism , Cyclic GMP/physiology , Endothelial Cells/metabolism , Female , Hindlimb/drug effects , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Immunohistochemistry , Locomotion/physiology , Mice , Mice, Inbred C57BL , Microcirculation/physiology , Nitric Oxide/physiology , Purines/therapeutic use , Recovery of Function/drug effects , Regional Blood Flow/drug effects , Sildenafil Citrate , Spinal Cord Injuries/physiopathology
14.
Am J Physiol Renal Physiol ; 302(1): F216-25, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22031848

ABSTRACT

Lithium (Li)-treated patients often develop urinary concentrating defect and polyuria, a condition known as nephrogenic diabetes insipidus (NDI). In a rat model of Li-induced NDI, we studied the effect that sildenafil (Sil), a phosphodiesterase 5 (PDE5) inhibitor, has on renal expression of aquaporin-2 (AQP2), urea transporter UT-A1, Na(+)/H(+) exchanger 3 (NHE3), Na(+)-K(+)-2Cl(-) cotransporter (NKCC2), epithelial Na channel (ENaC; α-, ß-, and γ-subunits), endothelial nitric oxide synthase (eNOS), and inducible nitric oxide synthase. We also evaluated cGMP levels in medullary collecting duct cells in suspension. For 4 wk, Wistar rats received Li (40 mmol/kg food) or no treatment (control), some receiving, in weeks 2-4, Sil (200 mg/kg food) or Li and Sil (Li+Sil). In Li+Sil rats, urine output and free water clearance were markedly lower, whereas urinary osmolality was higher, than in Li rats. The cGMP levels in the suspensions of medullary collecting duct cells were markedly higher in the Li+Sil and Sil groups than in the control and Li groups. Semiquantitative immunoblotting revealed the following: in Li+Sil rats, AQP2 expression was partially normalized, whereas that of UT-A1, γ-ENaC, and eNOS was completely normalized; and expression of NKCC2 and NHE3 was significantly higher in Li rats than in controls. Inulin clearance was normal in all groups. Mean arterial pressure and plasma arginine vasopressin did not differ among the groups. Sil completely reversed the Li-induced increase in renal vascular resistance. We conclude that, in experimental Li-induced NDI, Sil reduces polyuria, increases urinary osmolality, and decreases free water clearance via upregulation of renal AQP2 and UT-A1.


Subject(s)
Diabetes Insipidus, Nephrogenic/physiopathology , Lithium Compounds/adverse effects , Piperazines/therapeutic use , Polyuria/drug therapy , Sulfones/therapeutic use , Animals , Aquaporin 2/biosynthesis , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Diabetes Insipidus, Nephrogenic/chemically induced , Drinking/drug effects , Epithelial Sodium Channels/biosynthesis , Glomerular Filtration Rate/drug effects , Kidney/metabolism , Kidney Medulla/enzymology , Male , Membrane Transport Proteins/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Purines/therapeutic use , Rats , Sildenafil Citrate , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/biosynthesis , Sodium-Potassium-Chloride Symporters/biosynthesis , Solute Carrier Family 12, Member 1 , Urea Transporters
15.
Anticancer Res ; 31(6): 2319-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21737658

ABSTRACT

In former studies, raised urine cGMP levels have been reported to predict adverse outcome in cervical cancer. The main objective of the present study was to investigate the value of nitric oxide synthase (iNOS) and other members of the cGMP pathway as potential biomarkers for prognosis of cervical carcinoma. Tissue samples from 85 patients surgically treated for early-stage cervical carcinoma were immunohistochemically stained for iNOS, soluble guanylyl cyclase subunits α1 (sGC-α1), soluble guanylyl cyclase α2 (sGC-α2), and phosphodiesterase 5, each sample evaluated microscopically by a semi-quantitative score. Results were correlated to recurrence and FIGO stage (depth of tumour cell infiltration). Correlation was found between high expression of iNOS in tumour cells and low risk of recurrence (p=0.019, p=0.05, p=0.022 and p=0.025). High expression of iNOS, sGC-α1 and sGC-α2 also correlated to superficial tumour growth. Our results demonstrate that iNOS expression in cervical tumour tissue is a robust prognostic marker for cervical cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Uterine Cervical Neoplasms/enzymology , Carcinoma, Squamous Cell/pathology , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Female , Guanylate Cyclase/biosynthesis , Humans , Immunohistochemistry , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Signal Transduction , Uterine Cervical Neoplasms/pathology
16.
Cell Signal ; 23(3): 594-601, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21118717

ABSTRACT

The Schizosaccharomyces pombe fbp1 gene is transcriptionally repressed by protein kinase A (PKA) that is activated by extracellular glucose via a cAMP-signaling pathway. We previously used an fbp1-ura4 reporter that places uracil biosynthesis under the control of the glucose-sensing pathway to identify mutations in genes of the cAMP pathway. More recently, this reporter has been used in high throughput screens for small molecule inhibitors of heterologously-expressed cyclic nucleotide phosphodiesterases (PDEs) that hydrolyse cAMP to 5' AMP. Here we show that strains lacking the adenylyl cyclase gene respond to either exogenous cAMP or cGMP to activate PKA, thus regulating fbp1-ura4 expression and other PKA-regulated processes such as conjugation and the nuclear export of an Rst2-GFP fusion protein. Expression of cGMP-specific PDEs or ones that hydrolyse both cAMP and cGMP increases the amount of exogenous cGMP required to activate PKA in order to repress fbp1-ura4 expression, creating conditions that allow detection of inhibitors of these PDEs. As proof of this concept, we screened a collection of compounds previously identified as inhibitors of cAMP-specific PDE4 or PDE7 enzymes for their ability to inhibit the mammalian cGMP-specific PDE5A enzyme. We identified compound BC76, which inhibits PDE5A in an in vitro enzyme assay with an IC(50) of 232nM. Further yeast-based assays show that BC76 inhibits PDE1, PDE4, PDE5, PDE8, PDE10 and PDE11, thus demonstrating the utility of this system for detecting and characterising inhibitors of either cAMP- or cGMP-metabolising PDEs.


Subject(s)
Benzimidazoles/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Drug Evaluation, Preclinical/methods , Genes, Reporter , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrroles/pharmacology , Recombinant Proteins/biosynthesis , Schizosaccharomyces/genetics , Animals , Cattle , Humans , Mice
17.
J Urol ; 184(2): 769-74, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20639054

ABSTRACT

PURPOSE: We investigated phosphodiesterase 5 distribution and activity in the urethra. MATERIALS AND METHODS: Rat tissues were examined for phosphodiesterase 5 and alpha-smooth muscle actin expression. Urethral phosphodiesterase 5 activity was examined by tissue bath in the presence of sildenafil (Pfizer, New York, New York). RESULTS: Anti-alpha-smooth muscle actin antibody (Abcam) stained all known smooth muscles in all tested tissues and revealed a few smooth muscle fibers in the levator ani muscle. Anti-phosphodiesterase 5 antibody (Abcam) stained smooth muscle in the penis and bladder but not striated leg muscle. However, it stained predominantly striated muscle in the urethra and the levator ani muscle. In the urethra the amount of phosphodiesterase 5 in striated muscle was 6 times that in smooth muscle. In urethral striated muscle phosphodiesterase 5 expression was localized to Z-band striations. Smooth and striated muscle intermingling was clearly visible on the inner and outer rims of the circularly arranged striated muscle layer. Relaxation of precontracted urethral tissues by sodium nitroprusside (Sigma-Aldrich) was enhanced by sildenafil, indicating phosphodiesterase 5 activity, which was primarily located in the striated muscle according to phosphodiesterase 5 staining. CONCLUSIONS: Despite its presumed smooth muscle specificity phosphodiesterase 5 was predominantly expressed in the striated muscle of the urethra and in the levator ani muscle. Results are consistent with earlier studies in which these striated muscles were developmentally related to smooth muscle. They also suggest that these striated muscles are possibly regulated by phosphodiesterase 5.


Subject(s)
Actins/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Muscle, Striated/enzymology , Urethra/enzymology , Animals , Female , Male , Pelvis , Rats , Rats, Sprague-Dawley
18.
Int J Androl ; 33(5): 717-29, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20039971

ABSTRACT

The role of the structural complexity of the testis and the nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signalling pathway was analysed in adult male rats exposed to acute and repeated immobilization stress (IMO). In whole testis preparations, exposure to acute and repeated IMO caused an increase in NO production. In contrast, NO production was inhibited in interstitial cell preparations after exposure to all types of stress. In purified Leydig cell preparations, NO production was inhibited only after exposure to prolonged IMO. These findings indicate that biologically active compounds released from various testicular compartments exert both stimulatory and inhibitory effects on NO production. TaqMan Low Density Array of rat phosphodiesterases revealed a decrease in the expression of cGMP-specific phosphodiesterase 5 (PDE5) in Leydig cells of animals exposed to repeated IMO. In contrast, the expression of cGMP-dependent protein kinase type I (PKG I), total and phosphorylated steroidogenic acute regulatory protein (StAR), and PKG I/StAR immunoprecipitated complex was increased during repeated exposure to IMO. The increase in both total and phosphorylated StAR formation was effectively blocked by inhibition of PKG I in vitro. Thus, increased expressions of PKG I and StAR complex, accompanied by decreased PDE5 activity, suggest that the NO-cGMP signalling pathway and consequent activation of the StAR protein regulate the adaptive response of Leydig cells to repeated IMO stress.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Leydig Cells/metabolism , Nitric Oxide/physiology , Phosphoproteins/physiology , Signal Transduction/physiology , Testis/metabolism , Androgens/biosynthesis , Animals , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Male , Progesterone/biosynthesis , Rats , Rats, Wistar , Restraint, Physical , Stress, Physiological
20.
Curr Pharm Des ; 15(30): 3506-20, 2009.
Article in English | MEDLINE | ID: mdl-19860697

ABSTRACT

This review study refers to the possibility to employ PDE5 inhibitors as an adjunct tool for the therapeutic management of male infertility. The literature tends to suggest that PDE5 inhibitors enhance the Leydig cell secretory function and play a role in the regulation of the contractility of the tunica albuginea and the epididymis. In addition, the literature suggests that PDE5 inhibitors increase the prostatic secretory function that results in an improvement in sperm motility in several cases. Some studies additionally demonstrate a role of PDE5 inhibitors in the regulation of sperm capacitation process. Additional placebo-controlled, randomized, blind studies are necessary to unequivocally suggest a therapeutic role of PDE5 inhibitors in the alleviation of semen disorders and male infertility.


Subject(s)
Epididymis/physiopathology , Genital Diseases, Male/complications , Genital Diseases, Male/drug therapy , Infertility, Male/drug therapy , Infertility, Male/etiology , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/therapeutic use , Testicular Diseases/complications , Testicular Diseases/drug therapy , Acrosome Reaction/drug effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/biosynthesis , Genital Diseases, Male/physiopathology , Genitalia, Male/metabolism , Humans , Infertility, Male/enzymology , Leydig Cells/metabolism , Male , Semen/cytology , Semen/drug effects , Seminiferous Tubules/metabolism , Sertoli Cells/metabolism , Sperm Capacitation/drug effects , Spermatozoa/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL