Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Sex Med ; 20(1): 1-13, 2023 01 14.
Article in English | MEDLINE | ID: mdl-36897236

ABSTRACT

BACKGROUND: Sex steroids have been demonstrated as important modulators of vaginal function. The RhoA/ROCK calcium-sensitizing pathway plays a role in genital smooth muscle contractile mechanism, but its regulation has never been elucidated. AIM: This study investigated the sex steroid regulation of the vaginal smooth muscle RhoA/ROCK pathway using a validated animal model. METHODS: Ovariectomized (OVX) Sprague-Dawley rats were treated with 17ß-estradiol (E2), testosterone (T), and T with letrozole (T + L) and compared with intact animals. Contractility studies were performed to test the effect of the ROCK inhibitor Y-27632 and the nitric oxide (NO) synthase inhibitor L-NAME. In vaginal tissues, ROCK1 immunolocalization was investigated; mRNA expression was analyzed by semiquantitative reverse transcriptase-polymerase chain reaction; and RhoA membrane translocation was evaluated by Western blot. Finally, rat vaginal smooth muscle cells (rvSMCs) were isolated from the distal vagina of intact and OVX animals, and quantification of the RhoA inhibitory protein RhoGDI was performed after stimulation with NO donor sodium nitroprusside, with or without administration of the soluble guanylate cyclase inhibitor ODQ or PRKG1 inhibitor KT5823. OUTCOMES: Androgens are critical in inhibiting the RhoA/ROCK pathway of the smooth muscle compartment in the distal vagina. RESULTS: ROCK1 was immunolocalized in the smooth muscle bundles and blood vessel wall of the vagina, with weak positivity detected in the epithelium. Y-27632 induced a dose-dependent relaxation of noradrenaline precontracted vaginal strips, decreased by OVX and restored by E2, while T and T + L decreased it below the OVX level. In Western blot analysis, when compared with control, OVX significantly induced RhoA activation, as revealed by its membrane translocation, with T reverting it at a level significantly lower than in controls. This effect was not exerted by E2. Abolishing NO formation via L-NAME increased Y-27632 responsiveness in the OVX + T group; L-NAME had partial effects in controls while not modulating Y-27632 responsiveness in the OVX and OVX + E2 groups. Finally, stimulation of rvSMCs from control animals with sodium nitroprusside significantly increased RhoGDI protein expression, counteracted by ODQ and partially by KT5823 incubation; no effect was observed in rvSMCs from OVX rats. CLINICAL IMPLICATIONS: Androgens, by inhibiting the RhoA/ROCK pathway, could positively contribute to vaginal smooth muscle relaxation, favoring sexual intercourse. STRENGTHS AND LIMITATIONS: This study describes the role of androgens in maintaining vaginal well-being. The absence of a sham-operated animal group and the use of the only intact animal as control represented a limitation to the study.


Subject(s)
Androgens , Testosterone , Female , Rats , Animals , Humans , Rats, Sprague-Dawley , Nitroprusside , NG-Nitroarginine Methyl Ester , Estradiol/pharmacology , Letrozole , Vagina/physiology , Enzyme Inhibitors , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , Ovariectomy , rhoA GTP-Binding Protein/metabolism
2.
Int J Mol Sci ; 24(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36674598

ABSTRACT

Adiponectin (ADPN), a hormone produced by adipose tissue, facilitates gastric relaxation and can be a satiety signal in the network connecting peripheral organs and the central nervous system for feeding behavior control. Here, we performed preclinical research by morpho-functional analyses on murine gastric fundus smooth muscle to add insights into the molecular mechanisms underpinning ADPN action. Moreover, we conducted a clinical study to evaluate the potential use of ADPN as a biomarker for eating disorders (ED) based on the demonstrated gastric alterations and hormone level fluctuations that are often associated with ED. The clinical study recruited patients with ED and healthy controls who underwent blood draws for ADPN dosage and psychopathology evaluation tests. The findings of this basic research support the ADPN relaxant action, as indicated by the smooth muscle cell membrane pro-relaxant effects, with mild modifications of contractile apparatus and slight inhibitory effects on gap junctions. All of these actions engaged the ADPN/nitric oxide/guanylate cyclase pathway. The clinical data failed to unravel a correlation between ADPN levels and the considered ED, thus negating the potential use of ADPN as a valid biomarker for ED management for the moment. Nevertheless, this adipokine can modulate physiological eating behavior, and its effects deserve further investigation.


Subject(s)
Adiponectin , Gastric Fundus , Humans , Animals , Mice , Adiponectin/metabolism , Adipose Tissue/metabolism , Muscle, Smooth/metabolism , Biomarkers/metabolism
3.
Int J Mol Sci ; 22(4)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557413

ABSTRACT

Metabolic syndrome (MetS) is known to be associated to inflammation and alteration in the hypothalamus, a brain region implicated in the control of several physiological functions, including energy homeostasis and reproduction. Previous studies demonstrated the beneficial effects of testosterone treatment (TTh) in counteracting some MetS symptoms in both animal models and clinical studies. This study investigated the effect of TTh (30 mg/kg/week for 12 weeks) on the hypothalamus in a high-fat diet (HFD)-induced animal model of MetS, utilizing quantitative RT-PCR and immunohistochemical analyses. The animal model recapitulates the human MetS features, including low testosterone/gonadotropin plasma levels. TTh significantly improved MetS-induced hypertension, visceral adipose tissue accumulation, and glucose homeostasis derangements. Within hypothalamus, TTh significantly counteracted HFD-induced inflammation, as detected in terms of expression of inflammatory markers and microglial activation. Moreover, TTh remarkably reverted the HFD-associated alterations in the expression of important regulators of energy status and reproduction, such as the melanocortin and the GnRH-controlling network. Our results suggest that TTh may exert neuroprotective effects on the HFD-related hypothalamic alterations, with positive outcomes on the circuits implicated in the control of energy metabolism and reproductive tasks, thus supporting a possible role of TTh in the clinical management of MetS.


Subject(s)
Diet, High-Fat/adverse effects , Disease Models, Animal , Hypothalamus/drug effects , Metabolic Syndrome/drug therapy , Neuroprotective Agents/pharmacology , Testosterone/pharmacology , Animals , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Rabbits
4.
Eur Eat Disord Rev ; 29(4): 588-599, 2021 07.
Article in English | MEDLINE | ID: mdl-33939220

ABSTRACT

OBJECTIVE: The recent conceptualization of ghrelin as a stress hormone suggested that its chronic alterations may have a role in maintaining overeating behaviors in subjects with eating disorders (EDs) reporting childhood traumatic experiences. The aim of this study was to investigate the alterations of ghrelin levels in patients with EDs, their associations with early trauma, binge and emotional eating, and possible moderation/mediation models. METHOD: Sixty-four patients with EDs and 42 healthy controls (HCs) had their plasma ghrelin levels measured and completed questionnaires evaluating general and ED-specific psychopathology, emotional eating, and childhood traumatic experiences. RESULTS: Participants with anorexia nervosa had higher ghrelin levels than HCs in body mass index (BMI)-adjusted comparisons. Moreover, patients reporting a history of childhood trauma had higher ghrelin levels. Childhood sexual abuse (CSA), BMI, and self-induced vomiting were independent predictors of ghrelin levels. Moderation analyses showed that ghrelin levels were associated with binge and emotional eating only for higher levels of childhood trauma. Elevated ghrelin was a significant mediator for the association of CSA with binge eating. CONCLUSIONS: These results support the hypothesis that chronic alterations in ghrelin levels following childhood traumatic experiences could represent a neurobiological maintaining factor of pathological overeating behaviors in EDs.


Subject(s)
Binge-Eating Disorder , Bulimia , Feeding and Eating Disorders , Binge-Eating Disorder/psychology , Biomarkers , Bulimia/psychology , Ghrelin , Humans
5.
Int J Mol Sci ; 21(17)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854421

ABSTRACT

TNFα is the main proinflammatory cytokine implicated in the pathogenesis of neurodegenerative disorders, but it also modulates physiological functions in both the developing and adult brain. In this study, we investigated a potential direct role of TNFα in determining phenotypic changes of a recently established cellular model of human basal forebrain cholinergic neuroblasts isolated from the nucleus basalis of Meynert (hfNBMs). Exposing hfNBMs to TNFα reduced the expression of immature markers, such as nestin and ß-tubulin III, and inhibited primary cilium formation. On the contrary, TNFα increased the expression of TNFα receptor TNFR2 and the mature neuron marker MAP2, also promoting neurite elongation. Moreover, TNFα affected nerve growth factor receptor expression. We also found that TNFα induced the expression of DNA-methylation enzymes and, accordingly, downregulated genes involved in neuronal development through epigenetic mechanisms, as demonstrated by methylome analysis. In summary, TNFα showed a dual role on hfNBMs phenotypic plasticity, exerting a negative influence on neurogenesis despite a positive effect on differentiation, through mechanisms that remain to be elucidated. Our results help to clarify the complexity of TNFα effects in human neurons and suggest that manipulation of TNFα signaling could provide a potential therapeutic approach against neurodegenerative disorders.


Subject(s)
Basal Forebrain/cytology , Basal Nucleus of Meynert/cytology , DNA Methylation , Tumor Necrosis Factor-alpha/metabolism , Basal Forebrain/drug effects , Basal Forebrain/metabolism , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Cell Line , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Humans , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Neuronal Plasticity/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Tumor Necrosis Factor-alpha/pharmacology , Whole Genome Sequencing
6.
Am J Physiol Endocrinol Metab ; 316(3): E519-E535, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30645174

ABSTRACT

Metabolic syndrome (MetS) clusters cardiovascular and metabolic risk factors along with hypogonadism and erectile dysfunction. Lifestyle modifications including physical exercise (PhyEx) are well-known treatments for this condition. In this study, we analyzed the effect of PhyEx on hypothalamic-pituitary-testis axis and erectile function by use of an animal MetS model, previously established in rabbits fed a high-fat diet (HFD). Rabbits fed a regular diet (RD) were used as controls. A subset of both groups was trained on a treadmill. HFD rabbits showed typical MetS features, including HG (reduced T and LH) and impairment of erectile function. PhyEx in HFD rabbits completely restored plasma T and LH and the penile alterations. At testicular and hypothalamic levels, an HFD-induced inflammatory status was accompanied by reduced T synthesis and gonadotropin-releasing hormone (GnRH) immunopositivity, respectively. In the testis, PhyEx normalized HFD-related macrophage infiltration and increased the expression of steroidogenic enzymes and T synthesis. In the hypothalamus, PhyEx normalized HFD-induced gene expression changes related to inflammation and glucose metabolism, restored GnRH expression, particularly doubling mRNA levels, and regulated expression of molecules related to GnRH release (kisspeptin, dynorphin). Concerning MetS components, PhyEx significantly reduced circulating cholesterol and visceral fat. In multivariate analyses, cholesterol levels resulted as the main factor associated with MetS-related alterations in penile, testicular, and hypothalamic districts. In conclusion, our results show that PhyEx may rescue erectile function, exert anti-inflammatory effects on hypothalamus and testis, and increase LH levels and T production, thus supporting a primary role for lifestyle modification to combat MetS-associated hypogonadism and erectile dysfunction.


Subject(s)
Erectile Dysfunction/metabolism , Hypogonadism/metabolism , Metabolic Syndrome/metabolism , Physical Conditioning, Animal , Animals , Blood Glucose/metabolism , Cholesterol/metabolism , Dynorphins/genetics , Erectile Dysfunction/physiopathology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Kisspeptins/genetics , Luteinizing Hormone/metabolism , Macrophages , Male , Metabolic Syndrome/physiopathology , Rabbits , Testis/metabolism , Testis/pathology , Testosterone/metabolism , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
J Sex Med ; 13(12): 1858-1871, 2016 12.
Article in English | MEDLINE | ID: mdl-27914563

ABSTRACT

INTRODUCTION: Female sexual response is a complex phenomenon in which psychological, neurologic, and vascular mechanisms and hormonal factors interact. During the arousal phase, they cooperate to increase genital blood flow, thus inducing engorgement of the clitoris and lubrication of the vagina. Regulation of vascular and non-vascular smooth muscle tone is the crucial event in the erectile process. Preclinical studies have suggested that nitric oxide (NO) is the main vasodilator neurotransmitter modulating, through the second messenger cyclic guanosine monophosphate (cGMP), clitoral flow vessels. AIM: To investigate the effects of sexual steroid hormones on pro-erectile and relaxant (mediated by NO and cGMP) and anti-erectile and contractile (mediated by ras homolog gene family member A [RhoA] and Rho-associated protein kinase [ROCK]) mechanisms in the clitoris using a validated animal model of female ovariectomized Sprague-Dawley rats. METHODS: Subgroups of ovariectomized rats were treated with 17ß-estradiol, progesterone, testosterone, or testosterone and letrozole for 6 weeks. The experimental groups were compared with a control group of intact rats. MAIN OUTCOME MEASURES: Sex steroids plasma levels were assessed and in vitro contractility studies were carried out in order to investigate the effect of ovariectomy and in vivo treatments on clitoris smooth muscle activity. Smooth muscle cells (SMCs) from rat clitoral biopsies were isolated and characterized. RhoA activity was determined in SMCs cell cultures. RNA from tissues and cells was analyzed by quantitative real-time RT-PCR. RESULTS: Using real-time polymerase chain reaction, testosterone treatment upregulated the expression of NO-mediated pathway genes (endothelial and neuronal NO synthase, guanylate cyclase soluble subunit-α3, guanylate cyclase soluble subunit-ß3, cGMP-dependent protein kinase 1, and phosphodiesterase type 5). Conversely, estrogen replacement upregulated the expression of calcium-sensitizing RhoA-ROCK pathway genes. In vitro contractility studies were performed on phenylephrine pre-contracted clitoris strips. Ovariectomy resulted in a decreased responsiveness to Y-27632, a ROCK inhibitor, which was fully restored by 17ß-estradiol supplementation. To further examine the effect of 17ß-estradiol on the RhoA-ROCK pathway, smooth muscle cells were isolated from rat clitoris and their migration capacity was evaluated. CONCLUSION: Collectively, these data demonstrate that testosterone improves the relaxation of vascular smooth muscle cells through the NO-cGMP pathway, and that testosterone and 17ß-estradiol are necessary to maintain a functional contractile and relaxant machinery in the clitoris. This new concept might provide support for the concomitant use of estrogen and testosterone during the treatment of sexual arousal disorders related to hormonal imbalance or insufficiency.


Subject(s)
Clitoris/drug effects , Estradiol/pharmacology , Nitriles/pharmacology , Testosterone/pharmacology , Triazoles/pharmacology , Amides/pharmacology , Animals , Cyclic GMP/metabolism , Female , Letrozole , Muscle, Smooth/drug effects , Nitric Oxide/metabolism , Ovariectomy , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
8.
J Sex Med ; 11(12): 2890-902, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25243860

ABSTRACT

INTRODUCTION: Estrogen receptor (ER) α is critical in mediating the harmful effects of hyperestrogenism in fetal or neonatal life on the developing penis. In contrast, little is known on the impact of an excess of estrogens on penile function in adulthood. AIM: To investigate the effect of estrogens on metabolic syndrome (MetS)-associated erectile dysfunction (ED). METHODS: We employed a recently established animal model of high fat diet (HFD)-induced MetS. Subgroups of MetS rabbits were dosed with either testosterone (T) or tamoxifen. We evaluated penile responsiveness to acetylcholine (Ach) as well as the expression of genes related to penile smooth muscle relaxation and contractility. MAIN OUTCOME MEASURE: Associations between MetS-induced penile alterations and sex steroids were investigated in an animal model of HFD-induced MetS. To understand the role of either androgen deficiency or estrogen excess on ED, we treated subgroups of MetS rabbits with either T or tamoxifen, a classical ER antagonist. RESULTS: Feeding an HFD-induced MetS was associated to elevated estradiol (E2) and low T levels. E2, but not T, was independently and negatively associated with genes able to affect penile erection. Smooth muscle-related markers decreased as a function of E2 and were positively associated with all the variables investigated. Increasing concentrations of circulating E2 were negatively associated with Ach-induced relaxation. In HFD rabbits, in vivo T dosing significantly improved MetS and completely normalized circulating E2. Conversely, in vivo tamoxifen dosing reduced visceral adiposity and partially restored T level. Ach-induced relaxation was severely impaired by HFD and significantly restored, up to the control level, by both tamoxifen and T dosing. In rabbit smooth muscle cells cultures 17ß-E2 (1 nM) significantly reduced the expression of α-smooth muscle actin, transgelin, and phosphodiesterase type 5. The effects of 17ß-E2 were completely reverted by tamoxifen (100 nM). CONCLUSIONS: This study demonstrates, for the first time, that HFD-induced ED is more associated with a high E2, rather than to a low T, milieu. HFD-induced ED is partially restored by in vivo treatment not only with T but also with the nonsteroidal ER antagonist, tamoxifen.


Subject(s)
Diet, High-Fat/adverse effects , Erectile Dysfunction/metabolism , Estrogens/metabolism , Metabolic Syndrome/complications , Penile Erection/drug effects , Animals , Dietary Fats/adverse effects , Disease Models, Animal , Male , Penis/drug effects , Rabbits
9.
J Sex Med ; 11(5): 1159-72, 2014 May.
Article in English | MEDLINE | ID: mdl-24612540

ABSTRACT

INTRODUCTION: Metabolic syndrome (MetS) and lower urinary tract symptoms (LUTS) are often associated. Bladder detrusor hyper-contractility-a major LUTS determinant-is characterized by increased Ras homolog gene family, member A/Rho-associated protein kinase (RhoA/ROCK) signaling, which is often upregulated in MetS. AIM: This study investigated the effects of tadalafil dosing on RhoA/ROCK signaling in bladder, in a rabbit model of high-fat diet (HFD)-induced MetS. METHODS: Adult male rabbits feeding a HFD for 12 weeks. A subset of HFD animals was treated with tadalafil (2 mg/kg/day, 1 week: the last of the 12 weeks) and compared with HFD and control (feeding a regular diet) rabbits. MAIN OUTCOME MEASURES: In vitro contractility studies to evaluate the relaxant effect of the selective ROCK inhibitor, Y-27632, in carbachol precontracted bladder strips. Evaluation of RhoA activation by its membrane translocation. Immunohistochemistry for ROCK expression has been performed to evaluate ROCK expression in bladder from the different experimental groups. mRNA expression of inflammation, pro-fibrotic markers by quantitative RT-PCR has been performed to evaluate the effect of tadalafil on MetS-induced inflammation and fibrosis within the bladder. The in vitro effect of tadalafil on RhoA/ROCK signaling in bladder smooth muscle cells was evaluated by using chemotaxis assay. RESULTS: Bladder strips from HFD rabbits showed hyper-responsiveness to Y-27632, indicating RhoA/ROCK overactivity in HFD bladder compared with matched controls. Accordingly, the fraction of activated (translocated to the membrane) RhoA as well as ROCK expression are increased in HFD bladder. Tadalafil dosing normalized HFD-induced bladder hypersensitivity to Y-27632, by reducing RhoA membrane translocation and ROCK overexpression. Tadalafil dosing reduced mRNA expression of inflammatory, pro-fibrotic, and hypoxia markers. A direct inhibitory effect of tadalafil on RhoA/ROCK signaling in bladder smooth muscle cell was demonstrated by using chemotaxis assay. Pre-treatment with tadalafil inhibited both basal and PDGF-induced migration of bladder smooth muscle cells. CONCLUSIONS: Tadalafil dosing reduced RhoA/ROCK signaling and smooth muscle overactivity in an animal model of MetS-associated bladder alterations. Our findings suggest a novel mechanism of action of tadalafil in alleviating LUTS in MetS patients.


Subject(s)
Carbolines/pharmacology , Metabolic Syndrome/drug therapy , Urinary Bladder Diseases/drug therapy , Urological Agents/pharmacology , Amides/pharmacology , Animals , Diet, High-Fat , Enzyme Inhibitors/pharmacology , Male , Metabolic Syndrome/genetics , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Pyridines/pharmacology , Rabbits , Signal Transduction/drug effects , Tadalafil , Up-Regulation , rho-Associated Kinases/antagonists & inhibitors
10.
J Sex Med ; 11(7): 1694-708, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24844317

ABSTRACT

INTRODUCTION: In subjects with erectile dysfunction responding poorly to sildenafil, metformin was reported to improve erections. AIMS: The aim of this study is to investigate metformin's mechanism of action on erectile function, particularly focusing on adenosine (ADO) and nitric oxide (NO) signaling in an animal model of high-fat diet (HFD)-induced metabolic syndrome. METHODS: In vitro contractility studies of penile strips. Penile expression of genes related to ADO or NO signaling was also evaluated. MAIN OUTCOME MEASURE: In vitro contractility studies were used to investigate the effect of in vivo and ex vivo metformin administration on ADO- or acetylcholine (Ach)-induced relaxation of penile strips from HFD as compared with animals fed a regular diet (RD). RESULTS: Expression of ADO receptor type 3 (A3 R), ADO deaminase (ADA), AMP deaminase type 1 (AMPD1), and 2 (AMPD2) was decreased in HFD as compared with RD. Accordingly, in HFD the ADO relaxant effect was potentiated as compared with RD (P < 0.02). In vivo metformin treatment in both RD and HFD significantly increased the ADO relaxing effect (P < 0.0001 and P < 0.01, respectively, vs. relative untreated groups) although to a different extent. In fact, the half-maximal inhibitory concentration (IC50 )/IC50 ratio in RD increased fourfold vs. HFD (RD IC50 ratio = 13.75 ± 2.96; HFD IC50 ratio = 2.85 ± 0.52). In corpora cavernosa (CC) from HFD, in vivo metformin (i) normalized A3 R, ADA, and AMPD1; (ii) further decreased AMPD2; (iii) increased dimethylarginine dimethylamino-hydrolase; and (iv) partially restored impaired Ach-induced relaxation. Ex vivo metformin time and dose dependently increased the relaxant effect of ADO in RD. The potentiating effect of metformin on ADO-induced relaxation was significantly reduced by preincubation with NO synthase inhibitor N(ω) -Nitro-L-arginine methyl ester hydrochloride (L-NAME). Interestingly, in vivo testosterone supplementation in HFD rabbits (i) increased penile expression of endothelial NO synthase and AMPD2 and (ii) restored metformin's potentiating effect on ADO-induced relaxation up to RD level. CONCLUSION: Metformin in vivo and ex vivo increases ADO signaling in CC, most probably interfering with NO formation and ADO breakdown.


Subject(s)
Adenosine/metabolism , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Penile Erection/drug effects , AMP Deaminase/metabolism , Animals , Diet, High-Fat , Enzyme Inhibitors/pharmacology , Erectile Dysfunction/drug therapy , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Penis/drug effects , Penis/metabolism , Rabbits , Receptors, Purinergic P1/metabolism , Signal Transduction/drug effects , Testosterone/therapeutic use
11.
Prostate ; 73(4): 428-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22996758

ABSTRACT

BACKGROUND: Phosphodiesterase type 5 (PDE5) inhibitors improve benign prostatic hyperplasia (BPH)-related lower urinary tract symptoms (LUTS), often associated with metabolic syndrome (MetS). This study investigated the effects of PDE5 inhibition in the prostate of rabbits fed a high fat diet (HFD) for 12 weeks. HFD-rabbits develop the most important features of human MetS (glucose intolerance, dyslipidemia, increased abdominal adiposity, and hypertension), along with hypogonadism and LUT abnormalities (prostate and bladder inflammation/tissue remodeling). METHODS: Gene expression was evaluated by quantitative RT-PCR. Prostate morphological changes and oxygenation were evaluated by immunohistochemistry. RESULTS: HFD prostates showed increased PDE5 expression, suggesting a peculiar sensitivity of prostate to the action of PDE5 inhibitors during MetS. Accordingly, prostate PDE5 mRNA was negatively associated to plasma testosterone/estradiol ratio, whose reduction characterizes MetS, and positively with the expression in prostate of several genes exploring pathogenetic processes for BPH/LUTS, such as inflammation, leukocyte infiltration, and fibrosis/myofibroblast activation. Most of these genes was up-regulated by HFD, and significantly reduced by PDE5 inhibition, through either chronic (12 weeks) or, at a lower extent, acute (1-week) tadalafil dosing. Tadalafil was also able to reduce blood pressure and visceral fat in HFD rabbits, without changing any other MetS parameter. Interestingly, 1-week tadalafil administration to HFD rabbits, significantly blunted prostate inflammation (increased CD45 immunopositivity), fibrosis (reduced muscle/fiber ratio) and hypo-oxygenation, thus suggesting a potential curative effect of PDE5 inhibition on MetS-related prostate alterations. CONCLUSIONS: Our data provide the experimental evidences to support the multiple potentiality of PDE5 inhibitors as a useful therapeutic tool in LUTS.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Disease Models, Animal , Metabolic Syndrome/drug therapy , Metabolic Syndrome/enzymology , Phosphodiesterase 5 Inhibitors/pharmacology , Prostate/drug effects , Prostate/enzymology , Animals , Diet, High-Fat/adverse effects , Male , Metabolic Syndrome/pathology , Phosphodiesterase 5 Inhibitors/therapeutic use , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/enzymology , Prostatic Hyperplasia/pathology , Rabbits , Up-Regulation/drug effects , Up-Regulation/physiology
12.
Prostate ; 73(8): 789-800, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23620238

ABSTRACT

BACKGROUND: Metabolic syndrome (MetS) and benign prostate hyperplasia (BPH) are often comorbid. Chronic inflammation, a determinant pathogenic factor for BPH, is a putative link between the two conditions. METHODS: In a multi-center cohort of BPH patients (n = 244) who underwent prostatectomy, we evaluated whether MetS is associated with prostatic inflammation in BPH specimens. In addition, we investigated the in vitro inflammatory effects of metabolic insults on human prostatic myofibroblastic cells (hBPH). RESULTS: Inflammatory infiltrates score (IS) in prostatectomy specimens showed a step-wise association with the number of MetS factors present (P = 0.001). After adjusting for age, reduced HDL cholesterol, and elevated triglycerides were the only factors significantly associated with IS. Increased IS was also significantly associated with hypogonadism. In an age- and testosterone (T)-adjusted model, dyslipidemia was still associated with IS. To investigate whether metabolic factors could directly trigger prostate inflammation, we performed preliminary studies in myofibroblastic hBPH. Among the different factors, oxidized low-density lipoprotein (oxLDL) showed the highest secretion of IL-8 (>10-fold)-a surrogate marker of prostate inflammation--as well as IL-6, and bFGF. Co-treatment with DHT significantly inhibited oxLDL-induced secretion of IL-8, whilst an AR-antagonist, bicalutamide, reversed DHT effects. DHT suppresses oxLDL receptor (LOX-1) expression. CONCLUSIONS: Our data suggest that fats and insulin could have a detrimental effect on prostate health, boosting inflammation, a key pathogenic factor in BPH. Conversely, beneficial effects of DHT in counteracting lipid- and insulin-induced prostatic alterations, suggest that T-via its conversion into DHT-may have unexpected beneficial effects on prostate health.


Subject(s)
Metabolic Syndrome/metabolism , Prostatic Hyperplasia/metabolism , Prostatitis/metabolism , Aged , Cell Line , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cohort Studies , Cytokines/genetics , Cytokines/metabolism , Histocytochemistry , Humans , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Male , Metabolic Syndrome/blood , Metabolic Syndrome/pathology , Middle Aged , Prostatectomy/methods , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/surgery , Prostatitis/blood , Prostatitis/pathology , RNA/chemistry , RNA/genetics , Receptors, Androgen/blood , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Triglycerides/blood , Triglycerides/metabolism
13.
Prostate ; 73(13): 1391-402, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23765639

ABSTRACT

BACKGROUND: Metabolic syndrome (MetS) and benign prostate hyperplasia (BPH)/low urinary tract symptoms (LUTS) are often comorbid. Chronic inflammation is one of the putative links between these diseases. Phosphodiesterase type 5 inhibitors (PDE5i) are recognized as an effective treatment of BPH-related LUTS. One proposed mechanism of action of PDE5 is the inhibition of intraprostatic inflammation. In this study we investigate whether PDE5i could blunt inflammation in the human prostate. METHODS: Evaluation of the effect of tadalafil and vardenafil on secretion of interleukin 8 (IL-8, a surrogate marker of prostate inflammation) by human myofibroblast prostatic cells (hBPH) exposed to different inflammatory stimuli. We preliminary evaluate histological features of prostatic inflammatory infiltrates in BPH patients enrolled in a randomized, double bind, placebo controlled study aimed at investigating the efficacy of vardenafil (10 mg/day, for 12 weeks) on BPH/LUTS. RESULTS: In vitro treatment with tadalafil or vardenafil on hBPH reduced IL-8 secretion induced by either TNFα or metabolic factors, including oxidized low-density lipoprotein, oxLDL, to the same extent as a PDE5-insensitive PKG agonist Sp-8-Br-PET-cGMP. These effects were reverted by the PKG inhibitor KT5823, suggesting a cGMP/PKG-dependency. Treatment with tadalafil or vardenafil significantly suppressed oxLDL receptor (LOX-1) expression. Histological evaluation of anti-CD45 staining (CD45 score) in prostatectomy specimens of BPH patients showed a positive association with MetS severity. Reduced HDL-cholesterol and elevated triglycerides were the only MetS factors significantly associated with CD45 score. In the MetS cohort there was a significant lower CD45 score in the vardenafil-arm versus the placebo-one.


Subject(s)
Carbolines/pharmacology , Imidazoles/pharmacology , Interleukin-8/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Prostate/drug effects , Prostatic Hyperplasia/metabolism , Aged , Aged, 80 and over , Carbolines/therapeutic use , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Double-Blind Method , Humans , Imidazoles/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Lower Urinary Tract Symptoms/drug therapy , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/pathology , Male , Middle Aged , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphodiesterase 5 Inhibitors/therapeutic use , Pilot Projects , Piperazines/therapeutic use , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/pathology , Sulfones/pharmacology , Sulfones/therapeutic use , Tadalafil , Treatment Outcome , Triazines/pharmacology , Triazines/therapeutic use , Vardenafil Dihydrochloride
14.
Environ Pollut ; 317: 120766, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36460192

ABSTRACT

The neuroendocrine control of reproduction is strictly coordinated at the central level by the pulsatile release of gonadotropin-releasing hormone (GnRH) by the hypothalamic GnRH neurons. Alterations of the GnRH-network, especially during development, lead to long-term reproductive and systemic consequences, also causing infertility. Recent evidence shows that benzo[a]pyrene (BaP), a diffuse pollutant that can play a role as an endocrine disruptor, affects gonadal function and gamete maturation, whereas data demonstrating its impact at hypothalamic level are very scarce. This study investigated the effects of BaP (10 µM) in a primary cell culture isolated from the human fetal hypothalamus (hfHypo) and exhibiting a clear GnRH neuron phenotype. BaP significantly decreased gene and protein expression of both GnRH and kisspeptin receptor (KISS1R), the master regulator of GnRH neuron function. Moreover, BaP exposure increased phospho-ERK1/2 signaling, a well-known mechanism associated with KISS1R activation. Interestingly, BaP altered the electrophysiological membrane properties leading to a significant depolarizing effect and it also significantly increased GnRH release, with both effects being not affected by kisspeptin addition. In conclusion, our findings demonstrate that BaP may alter GnRH neuron phenotype and function, mainly interfering with KISS1R signaling and GnRH secretion and therefore with crucial mechanisms implicated in the central neuroendocrine control of reproduction.


Subject(s)
Gonadotropin-Releasing Hormone , Kisspeptins , Humans , Receptors, Kisspeptin-1/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Benzo(a)pyrene/toxicity , Benzo(a)pyrene/metabolism , Reproduction/physiology , Neurons
15.
J Sex Med ; 9(12): 3028-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23057807

ABSTRACT

INTRODUCTION: The efficacy of phosphodiesterase type 5 inhibitors (PDE5i) in treating lower urinary tract symptoms is supported by the extremely high expression and activity of PDE5 in male bladder. Although bladder function regulation is similar among genders, no data are available on PDE5 expression and activity in female bladder. AIM: To investigate sex differences in PDE5 expression and biological activity in female bladder, as opposed to the male counterpart. MAIN OUTCOME MEASURE: Gene and protein expression and enzymatic activity of PDE5. METHODS: We studied gene and protein expression, and enzymatic activity of PDE5 in bladder of male and female rats. A subgroup of female rats was ovariectomized and alternatively replaced with estradiol (E2), progesterone, and testosterone (T) alone or in combination with letrozole to completely abrogate T-induced E formation. As a readout of PDE5 activity, we studied vardenafil efficacy in potentiating sodium nitroprusside (SNP)-induced relaxation in bladder of the different experimental groups. RESULTS: SNP was three-log unit less potent in relaxing the male bladder than the female one. On the contrary, the PDE5-resistant cyclic guanosine monophosphate (cGMP) analog (Bromo-ß-phenyl-1, N(2) -ethenoguanosine-3', 5'-cyclic monophosphorothioate, Sp-isomer [SP-8-Br-PET-cGMPS]) was equipotent in relaxing male and female bladder. Vardenafil was more effective in potentiating SNP-induced bladder relaxation in male than in female. Accordingly, the cGMP-hydrolyzing activity of PDE5 was higher in male vs. female homogenates. In ovariectomized female rats, with or without sex-steroid replacement, vardenafil activity in potentiating SNP-induced bladder relaxation was associated with an increased T/E2 ratio. In particular, masculinization of ovariectomized rats--by the administration of T + letrozole--dramatically increased vardenafil capacity to potentiate SNP-induced relaxation. CONCLUSION: In this study, we demonstrated that PDE5 activity is more pronounced in male as compared with female bladder and that T/E ratio positively regulates responsiveness to PDE5i, thus suggesting that male bladder is a more suitable target for PDE5i than the female counterpart.


Subject(s)
Estradiol/blood , Imidazoles/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Testosterone/blood , Urinary Bladder/drug effects , Androgens/pharmacology , Animals , Aromatase Inhibitors/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Female , Gene Expression , Letrozole , Male , Nitric Oxide Donors/pharmacology , Nitriles/pharmacology , Nitroprusside/pharmacology , Ovariectomy , Progesterone/pharmacology , Progestins/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sex Factors , Sulfones/pharmacology , Testosterone/pharmacology , Triazines/pharmacology , Triazoles/pharmacology , Urinary Bladder/metabolism , Vardenafil Dihydrochloride
16.
Expert Rev Endocrinol Metab ; 17(6): 533-545, 2022 11.
Article in English | MEDLINE | ID: mdl-36447418

ABSTRACT

INTRODUCTION: Hyperprolactinemia has been proven to induce hypogonadism and metabolic derangements in both genders, while the consequences of prolactin (PRL) deficiency have been poorly investigated. AREAS COVERED: To systematically review and analyze data from clinical studies focusing on the metabolic consequences of abnormally high prolactin levels (HPRL) and low prolactin levels (LPRL). In addition, data from preclinical studies about underlying pathophysiological mechanisms were summarized and discussed. EXPERT OPINION: PRL contributes to providing the correct amount of energy to support the mother and the fetus/offspring during pregnancy and lactation, but it also has a homeostatic role. Pathological PRL elevation beyond these physiological conditions, but also its reduction, impairs metabolism and body composition in both genders, increasing the risk of diabetes and cardiovascular events. Hence, hypoprolactinemia should be avoided as much as possible during treatment with dopamine agonists for prolactinomas. Patients with hypoprolactinemia, because of endogenous or iatrogenic conditions, deserve, as those with hyperprolactinemia, careful metabolic assessment.


Subject(s)
Hyperprolactinemia , Pituitary Neoplasms , Prolactinoma , Male , Pregnancy , Humans , Female , Prolactin
17.
J Sex Med ; 8(10): 2746-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812935

ABSTRACT

INTRODUCTION: In humans, prostate phosphodiesterase type 5 inhibitors (PDE5) expression was prominently localized in the endothelial and smooth muscle cells of the vascular bed, suggesting a possible action of PDE5 inhibitors (PDE5i) on prostate blood flow. AIM: To investigate PDE5 expression in human and rat lower urinary tract (LUT) tissues, including vasculature, and determine the effects of PDE5 inhibition with tadalafil on prostatic blood perfusion. MAIN OUTCOME MEASURES: Human vesicular-deferential arteries (which originate from the inferior vesical artery, the main arterial source of blood supply to the bladder and prostate) were analyzed for PDE5 expression and activity. The effects of tadalafil on prostate oxygenation were studied in spontaneously hypertensive rats (SHR), characterized by ischemia/hypoxia of the genitourinary tract. METHODS: PDE5 expression was evaluated by quantitative reverse transcription-polymerase chain reaction and immunohistochemistry. SHR were treated with tadalafil (2 mg/kg/day) for 1, 7, or 28 days and compared with untreated SHR and the unaffected counterpart Wistar-Kyoto (WKY) rats. Prostate oxygenation was detected by Hypoxyprobe-1 and hypoxia markers (hypoxia-inducible factor-1α[HIF-1α] and endothelin-1 type B [ETB]) immunostaining. RESULTS: Human vesicular-deferential artery expressed high levels of PDE5, similar to corpora cavernosa, immunolocalized in the endothelial and smooth muscle layer. In these arteries, tadalafil inhibited cyclic guanosine monophosphate breakdown (half maximal inhibitory concentration (IC(50) ) in the low nanomolar range, as in corpora cavernosa) and increased the relaxant response to sodium nitroprusside. SHR prostate resulted markedly hypoxic (hypoxyprobe immunopositivity) and positive for HIF-1α and ETB, while tadalafil treatment restored oxygenation to WKY level at each time point. The mRNA expression of the HIF-1α target gene, BCL2/adenovirus E1B 19 kDa interacting protein 3, was significantly increased in SHR prostate and partially restored to WKY level by tadalafil. CONCLUSION: Human vesicular-deferential artery is characterized by a high expression and activity of PDE5, which was inhibited by tadalafil in vitro. In SHR, tadalafil increases prostate tissue oxygenation, thus suggesting a possible mechanism through which PDE5i exert beneficial effects on LUT symptoms.


Subject(s)
Carbolines/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Prostate/drug effects , Urinary Tract/enzymology , Animals , Endothelin-1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Nitroprusside/pharmacology , Oxygen/metabolism , Prostate/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Tadalafil , Urinary Bladder/blood supply , Urinary Bladder/enzymology , Urinary Bladder/metabolism , Urinary Tract/blood supply , Urinary Tract/drug effects , Urinary Tract/metabolism
18.
J Sex Med ; 8(1): 57-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20955313

ABSTRACT

INTRODUCTION: The farnesoid X receptor (FXR) is critically involved in the regulation of the hepato-biliary system. Recent data suggest a role for FXR in modulating other metabolic pathways and vascular function. AIM: To investigate whether long-term administration of the selective FXR agonist INT-747 ameliorates erectile function, we tested it in two animal models of metabolic derangements: a rabbit model of high-fat diet (HFD)-induced metabolic syndrome (MetS) and a rat model of streptozotocin (STZ)-induced type 1 diabetes. METHODS: HFD rabbit or STZ rats with or without chronic INT-747 dosing (10 mg/kg/day for 12 weeks). INT-747 addition to rabbit penile smooth muscle cells (rpSMCs). MAIN OUTCOME MEASURE: Effects of INT-747 on metabolic features and erectile function in animal models and clarification of mechanism of action in isolated cells. RESULTS: INT-747 dosing normalized visceral adiposity and glucose intolerance in HFD rabbits. INT-747 increased penile FXR expression and partially restored endothelial nitric oxide synthase and dimethylarginine dimethylaminohydrolase 1 expression as well as impaired nitric oxide (NO)-dependent relaxation (improved responsiveness to acetylcholine and electrical field stimulation). INT-747 was also effective in regulating NO downstream events, as shown by increased sodium nitroprusside-induced relaxation. Because phosphodiesterase type 5 and protein kinase G (PKG) were unaltered by INT-747, we analyzed the calcium-sensitizing RhoA/ROCK pathway. HFD increased, and INT-747 normalized, RhoA membrane translocation/activation. RhoA/ROCK signaling inhibition by INT-747 was confirmed in rpSMCs by confocal microscopy, MYPT1-phosphorylation, cytoskeleton remodeling, cell migration, and smooth muscle-related genes expression. In STZ rats, FXR penile expression was not altered but was significantly upregulated by INT-747 dosing. In this model, INT-747 improved penile erection induced by electrical stimulation of cavernous nerve and hypersensitivity to intracavernous injection of a ROCK-inhibitor, Y-27632, without improving hyperglycemia. CONCLUSION: In HFD rabbits, INT-747 dosing improved glucose sensitivity and MetS-associated erectile dysfunction, via upregulation of NO transmission and inhibition of RhoA/ROCK pathway. In STZ rats, INT-747 restored in vivo penile erection and sensitivity to ROCK inhibition, independently of effects on glycemia.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Erectile Dysfunction/drug therapy , Metabolic Syndrome/drug therapy , Receptors, Cytoplasmic and Nuclear/therapeutic use , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Erectile Dysfunction/complications , Erectile Dysfunction/metabolism , Male , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Muscle, Smooth/drug effects , Penis/blood supply , Penis/cytology , Rabbits , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism
19.
Eur J Histochem ; 65(s1)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34459573

ABSTRACT

Benzo[a]pyrene (BaP) is a widespread pollutant that can act as an endocrine disrupting compound (EDC) and interferes with reproductive function. The central regulatory network of the reproductive system is mediated by gonadotropin-releasing hormone (GnRH) neurons, which originate in the olfactory placode and, during ontogenesis, migrate into the hypothalamus. Given the importance of the migratory process for GnRH neuron maturation, we investigated the effect of BaP (10 µM for 24 h) on GnRH neuroblasts isolated from the human fetal olfactory epithelium (FNCB4). BaP exposure significantly reduced the mRNA level of genes implicated in FNCB4 cell migration and affected their migratory ability. Our findings demonstrate that BaP may interfere with the central neuronal network controlling human reproduction affecting GnRH neuron maturation.


Subject(s)
Benzo(a)pyrene/adverse effects , Cell Movement , Fetus/pathology , Gonadotropin-Releasing Hormone/metabolism , Neural Stem Cells/pathology , Neurons/pathology , Olfactory Mucosa/pathology , Fetus/drug effects , Fetus/metabolism , Humans , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Olfactory Mucosa/drug effects , Olfactory Mucosa/metabolism
20.
Endocrinology ; 162(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33247714

ABSTRACT

In this study, we investigated steroidogenic gene mRNA expression in human vaginas and verified the ability of human vagina smooth muscle cells (hvSMCs) to synthesize androgens from upstream precursor dehydroepiandrosterone (DHEA). As a readout for androgen receptor (AR) activation, we evaluated the mRNA expression of various androgen-dependent markers. hvSMCs were isolated from vagina tissues of women undergoing surgery for benign gynecological diseases. In these cells, we evaluated mRNA expression of several steroidogenic enzymes and sex steroid receptors using real time reverse transcription-polymerase chain reaction. Androgen production was quantified with liquid chromatography tandem-mass spectrometry (LC-MS/MS). In vaginal tissues, AR mRNA was significantly less expressed than estrogen receptor α, whereas in hvSMCs, its mRNA expression was higher than progestin and both estrogen receptors. In hvSMCs and in vaginal tissue, when compared to ovaries, the mRNA expression of proandrogenic steroidogenic enzymes (HSD3ß1/ß2, HSD17ß3/ß5), along with 5α-reductase isoforms and sulfotransferase, resulted as being more abundant. In addition, enzymes involved in androgen inactivation were less expressed than in the ovaries. The LC-MS/MS analysis revealed that, in hvSMCs, short-term DHEA supplementation increased Δ4-androstenedione levels in spent medium, while increasing testosterone and DHT secretion after longer incubation. Finally, androgenic signaling activation was evaluated through AR-dependent marker mRNA expression, after DHEA and T stimulation. This study confirmed that the human vagina is an androgen-target organ with the ability to synthesize androgens, thus providing support for the use of androgens for local symptoms of genitourinary syndrome in menopause.


Subject(s)
Androgens/metabolism , Menopause/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Steroid/metabolism , Vagina/metabolism , Aged , Aged, 80 and over , Dehydroepiandrosterone , Female , Gene Expression , Humans , Middle Aged , Myocytes, Smooth Muscle/cytology , Primary Cell Culture , Testosterone , Vagina/cytology
SELECTION OF CITATIONS
SEARCH DETAIL