Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 619
Filter
1.
Continence (Amst) ; 62023 Jun.
Article in English | MEDLINE | ID: mdl-37389025

ABSTRACT

This review summarizes the presentations made to a workshop entitled "Targeting Neurotrophin and Nitric Oxide Signaling to Promote Recovery and Ameliorate Neurogenic Bladder Dysfunction following Spinal Cord Injury - Mechanistic Concepts and Clinical Implications" at the International Continence Society (ICS) 2022 Vienna Meeting. Spinal cord injury (SCI; T8-T9 contusion/transection) causes impaired mobility, neurogenic detrusor overactivity (NDO), detrusor sphincter dyssynergia (DSD) and subsequent decreased quality of life. This workshop discussed the potential of future therapeutic agents that manage the lesion and its consequences, in particular possibilities to reduce the lesion itself and manage pathophysiological changes to the lower urinary tract (LUT). Attenuation of the spinal cord lesion itself was discussed with respect to the potential of a trio of agents: LM11A-3, a p75 neurotrophin receptor modulator to counter activation of local apoptotic pathways; LM22B-10 to promote neuronal growth by targeting tropomyosin-related kinase (Trk) receptors; and cinaciguat, a soluble guanylate cyclase (sGC) activator as an agent promoting angiogenesis at the injury site. The workshop also discussed targets on the bladder to block selectivity sites associated with detrusor overactivity and poor urinary filling profiles, such as purinergic pathways controlling excess contractile activity and afferent signaling, as well as excess fibrosis. Finally, the importance of increased mechanosensitive signaling as a contributor to DSD was considered, as well as potential drug targets. Overall, an emphasis was placed on targets that help restore function and reduce pathological LUT consequences, rather than downregulate normal function.

2.
Continence (Amst) ; 62023 Jun.
Article in English | MEDLINE | ID: mdl-37389026

ABSTRACT

This review summarises the presentations during a workshop session entitled "The Use of Soluble Guanylate Cyclase Activators to Treat Benign Prostatic Hyperplasia, Obstruction and Fibrosis - Mechanistic Concepts and Clinical Implications" at the International Continence Society (ICS) 2021 Melbourne Virtual meeting. Benign prostatic hyperplasia (BPH) is a highly prevalent condition that can result in bladder outflow obstruction (BOO) and development of lower urinary tract symptoms (LUTS), and by 80 years of age is present in about 75% of men. Current pharmacological therapies include α-adrenoceptor antagonists, 5α-reductase inhibitors, and the phosphodiesterase type 5 (PDE5) inhibitor, tadalafil. The efficacy of tadalafil suggests a role for nitric oxide (NO•) through activation of soluble guanylate cyclase (sGC) and production of cyclic guanosine 3'5'-monophosphate (cGMP), a cyclic nucleotide that relaxes smooth muscle, reduces neurotransmitter release and also acts as an antifibrotic agent. Patient refractoriness to tadalafil may be, for example, due to sGC inactivation due to oxidative stress. The workshop discussed the superiority of cinaciguat, an sGC activator that functions even when the enzyme is oxidised, over PDE5 inhibitors, and potentially its use in combination with agents that reduce formation of reactive oxygen species.

3.
Physiol Res ; 68(5): 785-792, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31424252

ABSTRACT

The former perception of the urothelium as an impermeable barrier has been revised during the last decade, as increasing evidence of changes in urine composition during its passage of the urinary tract has been presented. Since differences in urothelial permeability between upper and lower urinary tract have been found, our aim is to demonstrate whether changes in urine composition occur during passage through the ureter. We studied consecutive urine samples from both renal pelvises in six pigs and compared them to samples from the bladder and distal ureter. We further sampled urine during storage in the bladder at a fixed volume. All samples were analysed by measuring osmolality and pH, along with the concentration of the following parameters: Na(+), K(+), Cl(-), creatinine, urea. Urine alkalinity increased significantly during passage of the ureter. Creatinine concentration, pH and K(+) increased significantly during the passage from pelvis to the bladder. All other parameters increased non-significantly during the passage to the bladder. The increase in concentration was more pronounced at low concentrations in the pelvis. During storage in the bladder, there was a significant increase in urea concentration. Changes in the composition of urine occur during its passage from the renal pelvis to the bladder and during storage in the bladder. Despite the brief transit time, significant changes in alkalinity were found already during passage through the ureter.


Subject(s)
Kidney Concentrating Ability , Ureter/metabolism , Urinary Bladder/metabolism , Urine/chemistry , Animals , Chlorides/urine , Creatinine/urine , Female , Hydrogen-Ion Concentration , Osmolar Concentration , Potassium/urine , Sodium/urine , Sus scrofa , Time Factors , Urea/urine
4.
Br J Pharmacol ; 175(13): 2554-2565, 2018 07.
Article in English | MEDLINE | ID: mdl-29667180

ABSTRACT

The discovery of the nitric oxide/cGMP pathway was the basis for our understanding of many normal physiological functions and the pathophysiology of several diseases. Since the discovery and introduction of sildenafil, inhibitors of PDE5 have been the first-line therapy for erectile dysfunction (ED). The success of sildenafil in the treatment of ED stimulated research in the field of PDE5 inhibition and led to many new applications, such as treatment of lower urinary symptoms, and pulmonary arterial hypertension, which are now approved indications. However, PDE5 inhibitors have also been used in several other disorders not discussed in this review, and the fields of clinical use are increasing. In the present review, the pharmacological basis of the NO/cGMP pathway and the rationale and clinical use of PDE5 inhibitors in different diseases are discussed.


Subject(s)
Cyclic GMP/antagonists & inhibitors , Drug Discovery , Nitric Oxide/antagonists & inhibitors , Phosphodiesterase 5 Inhibitors/pharmacology , Sildenafil Citrate/pharmacology , Animals , Cyclic GMP/metabolism , Humans , Nitric Oxide/metabolism
5.
Physiol Res ; 67(2): 283-292, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29303600

ABSTRACT

The expression of aquaporins (AQPs) in the fetal porcine urinary tract and its relation to gestational age has not been established. Tissue samples from the renal pelvis, ureter, bladder and urethra were obtained from porcine fetuses. Samples were examined by RT-PCR (AQPs 1-11), QPCR (AQPs positive on RT-PCR), and immunohistochemistry. Bladder samples were additionally examined by Western blotting. RNA was extracted from 76 tissue samples obtained from 19 fetuses. Gestational age was 60 (n=11) or 100 days (n=8). PCR showed that AQP1, 3, 9 and 11 mRNA was expressed in all locations. The expression of AQP3 increased significantly at all four locations with gestational age, whereas AQP11 significantly decreased. AQP1 expression increased in the ureter, bladder and urethra. AQP9 mRNA expression increased in the urethra and bladder, but decreased in the ureter. AQP5 was expressed only in the urethra. Immunohistochemistry showed AQP1 staining in sub-urothelial vessels at all locations. Western blotting analysis confirmed increased AQP1 protein levels in bladder samples during gestation. Expression levels of AQP1, 3, 5, 9 and 11 in the urinary tract change during gestation, and further studies are needed to provide insights into normal and pathophysiological water handling mechanisms in the fetus.


Subject(s)
Aquaporins/biosynthesis , Urinary Tract/embryology , Urinary Tract/metabolism , Adult , Animals , Female , Fetus/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Humans , Pregnancy , Sus scrofa , Swine , Ureter/embryology , Ureter/metabolism , Urethra/embryology , Urethra/metabolism , Urinary Bladder/embryology , Urinary Bladder/metabolism
7.
Br J Pharmacol ; 172(16): 4158-72, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26013995

ABSTRACT

BACKGROUND AND PURPOSE: T16A(inh)-A01, CaCC(inh)-A01 and MONNA are identified as selective inhibitors of the TMEM16A calcium-activated chloride channel (CaCC). The aim of this study was to examine the chloride-specificity of these compounds on isolated resistance arteries in the presence and absence (±) of extracellular chloride. EXPERIMENTAL APPROACH: Isolated resistance arteries were maintained in a myograph and tension recorded, in some instances combined with microelectrode impalement for membrane potential measurements or intracellular calcium monitoring using fura-2. Voltage-dependent calcium currents (VDCC) were measured in A7r5 cells with voltage-clamp electrophysiology using barium as a charge carrier. KEY RESULTS: Rodent arteries preconstricted with noradrenaline or U46619 were concentration-dependently relaxed by T16A(inh) -A01 (0.1-10 µM): IC50 and maximum relaxation were equivalent in ±chloride (30 min aspartate substitution) and the T16A(inh) -A01-induced vasorelaxation ±chloride were accompanied by membrane hyperpolarization and lowering of intracellular calcium. However, agonist concentration-response curves ±chloride, with 10 µM T16A(inh) -A01 present, achieved similar maximum constrictions although agonist-sensitivity decreased. Contractions induced by elevated extracellular potassium were concentration-dependently relaxed by T16A(inh)-A01 ±chloride. Moreover, T16A(inh) -A01 inhibited VDCCs in A7r5 cells in a concentration-dependent manner. CaCC(inh) -A01 and MONNA (0.1-10 µM) induced vasorelaxation ±chloride and both compounds lowered maximum contractility. MONNA, 10 µM, induced substantial membrane hyperpolarization under resting conditions. CONCLUSIONS AND IMPLICATIONS: T16A(inh) -A01, CaCC(inh) -A01 and MONNA concentration-dependently relax rodent resistance arteries, but an equivalent vasorelaxation occurs when the transmembrane chloride gradient is abolished with an impermeant anion. These compounds therefore display poor selectivity for TMEM16A and inhibition of CaCC in vascular tissue in the concentration range that inhibits the isolated conductance.


Subject(s)
Chloride Channels/antagonists & inhibitors , Pyrimidines/pharmacology , Thiazoles/pharmacology , Thiophenes/pharmacology , Vasodilator Agents/pharmacology , ortho-Aminobenzoates/pharmacology , Animals , Cell Line , Cerebral Arteries/drug effects , Cerebral Arteries/physiology , Chloride Channels/physiology , Female , In Vitro Techniques , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mice, Inbred C57BL , Rats, Wistar
8.
Auton Neurosci ; 191: 78-81, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25979768

ABSTRACT

It is well established that in most species, exocytotic vesicular release of ATP from parasympathetic neurons contributes to contraction of the bladder. However, ATP is released not only from parasympathetic nerves, but also from the urothelium. During bladder filling, the urothelium is stretched and ATP is released from the umbrella cells thereby activating mechanotransduction pathways. ATP release can also be induced by various mediators present in the urine and and/or released from nerves or other components of the lamina propria. Urothelial release of ATP is mainly attributable to vesicular transport or exocytosis and, to a smaller extent, to pannexin hemichannel conductive efflux. After release, ATP acts on P2X3 and P2X2/3 receptors on suburothelial sensory nerves to initiate the voiding reflex and to mediate the sensation of bladder filling and urgency. ATP also acts on suburothelial interstitial cells/myofibroblasts generating an inward Ca(2+) transient that via gap junctions could provide a mechanism for long-distance spread of signals from the urothelium to the detrusor muscle. ATP release can be affected by urological diseases, e.g., interstitial cystitis and both the mechanisms of release and the receptors activated by ATP may be targets for future drugs for treatment of lower urinary tract disorders.


Subject(s)
Receptors, Purinergic/metabolism , Urinary Bladder/metabolism , Adenosine Triphosphate/metabolism , Animals , Humans
9.
Neurourol Urodyn ; 34(8): 787-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25230878

ABSTRACT

AIMS: To assess the urodynamic effects of soluble guanylyl cyclase (sGC) stimulator, BAY 41-2272, and activator, BAY 60-2770, (which both are able to induce cGMP synthesis even in the absence of nitric oxide (NO)) alone or in combination with a phosphodiesterase type 5 (PDE5) inhibitor, vardenafil, in a model of partial urethral obstruction (PUO) induced bladder overactivity (BO). METHODS: Fifty-six male Sprague-Dawley rats were used, 31 of them underwent PUO. Fourteen rats were used for Western blots to assess PDE5 and sGC expression. For drug evaluation cystometry without anesthesia was performed three days following bladder catheterization. RESULTS: Obstructed rats showed higher micturition frequency and bladder pressures than non-obstructed animals (Intermicturition Interval, IMI, 2.28 ± 0.55 vs. 3.60 ± 0.60 min (± standard deviation, SD); maximum micturition pressure, MMP, 70.1 ± 8.0 vs. 48.8 ± 7.2 cmH2O; both P < 0.05). In obstructed rats vardenafil, BAY 41-2272, and BAY 60-2770 increased IMI (2.77 ± 1.12, 2.62 ± 0.52, and 3.22 ± 1.04 min; all P < 0.05) and decreased MMP (54.4 ± 2.8, 61.5 ± 11.3, and 51.2 ± 6.3 cmH2O; all P < 0.05). When vardenafil was given following BAY 41-2272 or BAY 60-2770 no further urodynamic effects were observed. PDE5 as well as sGC protein expression was reduced in obstructed bladder tissue. CONCLUSIONS: Targeting sGC via stimulators or activators, which increase the levels of cGMP independent of endogenous NO, is as effective as vardenafil to reduce urodynamic signs of BO. Targeting the NO/cGMP pathway via compounds acting on sGC might become a new approach to treat BO.


Subject(s)
Benzoates/therapeutic use , Biphenyl Compounds/therapeutic use , Hydrocarbons, Fluorinated/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Urethral Obstruction/drug therapy , Urinary Bladder, Overactive/drug therapy , Urinary Bladder/drug effects , Animals , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Disease Models, Animal , Drug Therapy, Combination , Guanylate Cyclase/metabolism , Hydrocarbons, Fluorinated/pharmacology , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Urethral Obstruction/complications , Urethral Obstruction/metabolism , Urinary Bladder/metabolism , Urinary Bladder, Overactive/etiology , Urinary Bladder, Overactive/metabolism
10.
Ann Phys Rehabil Med ; 57(5): 321-328, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24954496

ABSTRACT

A majority of patients diagnosed with multiple sclerosis (MS) will develop lower urinary tract symptoms (LUTS) during the course of the disease. Even if antimuscarinic (anticholinergic) treatment is currently the mainstay of conservative treatment of neurogenic detrusor overactivity (NDO), including MS-induced NDO, extensive data regarding their effectiveness and safeness are lacking. When antimuscarinic medications fail to prove efficacious, a further option is intradetrusor injections of onabotulinumtoxin A. In several studies, more than half (and up 76%) of the patients treated with onabotulinumtoxin A experienced significant improvement in symptoms or even achieved complete continence. Cannabis extracts have shown some promise but has still not gained wide acceptance as an effective treatment. Over the last few years many new disease-modifying drugs that have been approved and introduced for treatment of MS. These drugs may have effects not only on the MS disease process, but also on the disease symptoms, including LUTS. However, MS is not primarily a bladder disease and treatment of the underlying pathophysiology should be the main goal of treatment. Since most of the urology drugs are targeting LUTS, these drugs should be regarded as "adds on" to treatments modifying the underlying disorder. Considering that most of these drugs have not been studied specifically with respect to efficacy on LUTS, and since they are not without significant side effects, it seems important that if and when they are going to be used for treatment of bladder symptoms should be a joint decision between the neurologist and urologist taking care of the patient.


Subject(s)
Urinary Bladder, Neurogenic/drug therapy , Urinary Bladder, Overactive/drug therapy , Forecasting , Humans , Multiple Sclerosis/complications , Urinary Bladder, Neurogenic/etiology , Urinary Bladder, Overactive/etiology
11.
J Urol ; 191(1): 253-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24018240

ABSTRACT

PURPOSE: We evaluated the efficacy, safety and tolerability of the EP1 receptor antagonist ONO-8539 in patients with overactive bladder syndrome. MATERIALS AND METHODS: This was a 12-week, randomized, double-blind, placebo controlled, parallel group, multicenter study with a 2-week single blind placebo run-in phase. The 435 patients were randomized to receive twice daily ONO-8539 (30, 100 or 300 mg), placebo or once daily tolterodine (4 mg). RESULTS: At the end of the 12-week treatment no statistically significant difference was found between ONO-8539 and placebo in the change from baseline in the number of micturitions per 24 hours. The primary end points for 30, 100 and 300 mg ONO-8539, and placebo were -1.02, -1.53, -1.31 and -1.40, respectively. There was no statistically significant difference between any ONO-8539 group and placebo in the change from baseline in the number of urgency or urinary urgency incontinence episodes per 24 hours, or the mean volume voided per micturition, which were secondary end points. Statistically significant differences for tolterodine vs placebo were observed in the change from baseline in the number of micturitions (p = 0.045), urgency episodes (p = 0.04) and mean volume voided per micturition (p <0.001). The incidence of adverse events was 54.1% in the placebo group, 43.0% to 54.0% in the ONO-8539 groups and 46.6% in the tolterodine group. The intensity of adverse events was similar among the treatment groups. Similar to other treatments, the most frequently reported adverse events after ONO-8539 were nasopharyngitis and diarrhea. CONCLUSIONS: The results of this study, which to our knowledge represents the first evaluation of ONO-8539 in patients with overactive bladder, suggest a minimal role for EP1 receptor antagonism in the management of overactive bladder syndrome.


Subject(s)
Benzhydryl Compounds/therapeutic use , Cresols/therapeutic use , Phenylpropanolamine/therapeutic use , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Urinary Bladder, Overactive/drug therapy , Urological Agents/therapeutic use , Adult , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Tolterodine Tartrate , Treatment Outcome
12.
Urologe A ; 52(2): 204-11, 2013 Feb.
Article in German | MEDLINE | ID: mdl-23417046

ABSTRACT

Phosphodiesterase-5 (PDE5) inhibitors, such as sildenafil, tadalafil and vardenafil are first line treatment for erectile dysfunction (ED). These PDE5 inhibitors are known to increase cyclic guanosine monophosphate (cGMP) concentrations in the smooth muscle cells of the corpora cavernosa penis by inhibiting PDE5, leading to smooth muscle relaxation. This mode of action is also believed to result in prostatic smooth muscle relaxation and to improve lower urinary tract symptoms (LUTS). Randomized controlled trials have shown beneficial effects on LUTS and on objective parameters such as maximum urinary flow rate (tadalafil). Based on these data tadalafil was recently approved for treatment of patients with male LUTS; however, the mechanisms leading to improvement of symptoms are still under debate.


Subject(s)
Phosphodiesterase 5 Inhibitors/therapeutic use , Prostatic Hyperplasia/drug therapy , Carbolines/adverse effects , Carbolines/therapeutic use , Controlled Clinical Trials as Topic , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Erectile Dysfunction/drug therapy , Erectile Dysfunction/pathology , Humans , Imidazoles/adverse effects , Imidazoles/therapeutic use , Lower Urinary Tract Symptoms/diagnosis , Lower Urinary Tract Symptoms/drug therapy , Lower Urinary Tract Symptoms/pathology , Male , Phosphodiesterase 5 Inhibitors/adverse effects , Piperazines/adverse effects , Piperazines/therapeutic use , Prostatic Hyperplasia/diagnosis , Prostatic Hyperplasia/pathology , Purines/adverse effects , Purines/therapeutic use , Sildenafil Citrate , Sulfones/adverse effects , Sulfones/therapeutic use , Tadalafil , Triazines/adverse effects , Triazines/therapeutic use , Urodynamics/drug effects , Vardenafil Dihydrochloride
13.
Acta Physiol (Oxf) ; 207(1): 16-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23033838

ABSTRACT

The autonomic nervous system plays an important role in the regulation of the urinary bladder function. Under physiological circumstances, noradrenaline, acting mainly on ß(3) -adrenoceptors in the detrusor and on α(1) (A) -adrenoceptors in the bladder outflow tract, promotes urine storage, whereas neuronally released acetylcholine acting mainly on M(3) receptors promotes bladder emptying. Under pathophysiological conditions, however, this system may change in several ways. Firstly, there may be plasticity at the levels of innervation and receptor expression and function. Secondly, non-neuronal acetylcholine synthesis and release from the urothelium may occur during the storage phase, leading to a concomitant exposure of detrusor smooth muscle, urothelium and afferent nerves to acetylcholine and noradrenaline. This can cause interactions between the adrenergic and cholinergic system, which have been studied mostly at the post-junctional smooth muscle level until now. The implications of such plasticity are being discussed.


Subject(s)
Autonomic Nervous System/physiology , Neuronal Plasticity/physiology , Urinary Bladder Diseases/physiopathology , Urinary Bladder/innervation , Urinary Bladder/physiology , Animals , Humans , Receptors, Adrenergic, beta/physiology , Receptors, Muscarinic/physiology
14.
Br J Pharmacol ; 166(6): 1926-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22364229

ABSTRACT

BACKGROUND AND PURPOSE α(1) -Adrenoceptor-induced contraction of prostate smooth muscle is mediated by calcium- and Rho kinase-dependent mechanisms. In addition, other mechanisms, such as activation of c-jun N-terminal kinase (JNK) may be involved. Here, we investigated whether JNK participates in α(1)-adrenoceptor-induced contraction of human prostate smooth muscle. EXPERIMENTAL APPROACH Prostate tissue was obtained from patients undergoing radical prostatectomy. Effects of the JNK inhibitors SP600125 (50 µM) and BI-78D3 (30 µM) on contractions induced by phenylephrine, noradrenaline and electric field stimulation (EFS) were studied in myographic measurements. JNK activation by noradrenaline (30 µM) and phenylephrine (10 µM), and the effects of JNK inhibitors of c-Jun phosphorylation were assessed by Western blot analyses with phospho-specific antibodies. Expression of JNK was studied by immunohistochemistry and fluorescence double staining. KEY RESULTS The JNK inhibitors SP600125 and BI-78D3 reduced phenylephrine- and noradrenaline-induced contractions of human prostate strips. In addition, SP600125 reduced EFS-induced contraction of prostate strips. Stimulation of prostate tissue with noradrenaline or phenylephrine in vitro resulted in activation of JNK. Incubation of prostate tissue with SP600125 or BI-78D3 reduced the phosphorylation state of c-Jun. Immunohistochemical staining demonstrated the expression of JNK in smooth muscle cells of human prostate tissue. Fluorescence staining showed that α(1A)-adrenoceptors and JNK are expressed in the same cells. CONCLUSIONS AND IMPLICATIONS Activation of JNK is involved in α(1)-adrenoceptor-induced prostate smooth muscle contraction. Models of α(1)-adrenoceptor-mediated prostate smooth muscle contraction should include this JNK-dependent mechanism.


Subject(s)
Anthracenes/pharmacology , Dioxanes/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth/drug effects , Prostate/drug effects , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Aged , Humans , In Vitro Techniques , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth/physiology , Prostate/physiology , Receptors, Adrenergic, alpha-1/physiology
15.
Int J Impot Res ; 24(3): 91-100, 2012.
Article in English | MEDLINE | ID: mdl-22205244

ABSTRACT

Erectile dysfunction (ED) is a prevalent medical condition affecting 18 million men and their sexual partners in the United States alone. In the majority of patients, ED is related to alterations in the flow of blood to or from the penis. Undeniably, significant progress has been made in understanding the multifactorial mechanisms that modulate erectile capacity and predispose one to ED, and this, in turn, has led to the availability of more effective treatment options. Nonetheless, all current therapies have untoward side effects, and moreover, there are still no satisfactory treatments for many patients with ED. Further enhancements in the treatment of ED would logically result from both early intervention and more detailed mechanistic insight into the characteristics of the disease process per se. This fact underscores the importance of improved understanding of the initiation, development and progression of ED. However, to do so requires longitudinal studies on animal models that more closely approximate the corresponding clinical features and time course of human disease. The goal of this report is twofold. First, to provide a brief general overview of the applicability of commonly used animal models for the study of ED. The second and primary goal is to highlight the scientific rationale for using non-human primates to evaluate the impact of atherosclerosis-induced vascular disease on the penile and systemic circulatory systems. This latter goal seems especially relevant in light of the recent literature documenting a link between ED and systemic vascular disease, a finding that has major implications in an aging US male population consuming a high fat diet.


Subject(s)
Atherosclerosis/complications , Disease Models, Animal , Impotence, Vasculogenic/etiology , Primates , Adult , Aged , Animals , Coronary Disease , Diet, High-Fat/adverse effects , Humans , Male , Mice , Middle Aged , Penis/blood supply , Rabbits , Rats , Risk Factors
16.
Pharmacol Rev ; 63(4): 811-59, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21880989

ABSTRACT

Erection is basically a spinal reflex that can be initiated by recruitment of penile afferents, both autonomic and somatic, and supraspinal influences from visual, olfactory, and imaginary stimuli. Several central transmitters are involved in the erectile control. Dopamine, acetylcholine, nitric oxide (NO), and peptides, such as oxytocin and adrenocorticotropin/α-melanocyte-stimulating hormone, have a facilitatory role, whereas serotonin may be either facilitatory or inhibitory, and enkephalins are inhibitory. The balance between contractant and relaxant factors controls the degree of contraction of the smooth muscle of the corpora cavernosa (CC) and determines the functional state of the penis. Noradrenaline contracts both CC and penile vessels via stimulation of α1-adrenoceptors. Neurogenic NO is considered the most important factor for relaxation of penile vessels and CC. The role of other mediators, released from nerves or endothelium, has not been definitely established. Erectile dysfunction (ED), defined as the "inability to achieve or maintain an erection adequate for sexual satisfaction," may have multiple causes and can be classified as psychogenic, vasculogenic or organic, neurologic, and endocrinologic. Many patients with ED respond well to the pharmacological treatments that are currently available, but there are still groups of patients in whom the response is unsatisfactory. The drugs used are able to substitute, partially or completely, the malfunctioning endogenous mechanisms that control penile erection. Most drugs have a direct action on penile tissue facilitating penile smooth muscle relaxation, including oral phosphodiesterase inhibitors and intracavernosal injections of prostaglandin E1. Irrespective of the underlying cause, these drugs are effective in the majority of cases. Drugs with a central site of action have so far not been very successful. There is a need for therapeutic alternatives. This requires identification of new therapeutic targets and design of new approaches. Research in the field is expanding, and several promising new targets for future drugs have been identified.


Subject(s)
Alprostadil/therapeutic use , Erectile Dysfunction/drug therapy , Penile Erection/drug effects , Phosphodiesterase Inhibitors/therapeutic use , Alprostadil/pharmacology , Erectile Dysfunction/etiology , Erectile Dysfunction/physiopathology , Humans , Male , Nervous System Physiological Phenomena , Neurotransmitter Agents/pharmacology , Neurotransmitter Agents/therapeutic use , Penile Erection/physiology , Phosphodiesterase Inhibitors/pharmacology
17.
Actas urol. esp ; 35(7): 373-388, jul.-ago. 2011. tab, graf
Article in Spanish | IBECS | ID: ibc-90149

ABSTRACT

Contexto: Las primeras directrices sobre incontinencia de la European Association of Urology (EAU) se publicaron en 2001. Dichas directrices se han actualizado con regularidad en los últimos años. Objetivo: El objetivo de este artículo es ofrecer un resumen de la actualización de las directrices sobre incontinencia urinaria (IU) de la EAU realizada en 2009. Recogida de evidencias: El comité de trabajo de la EAU formó parte de la IV Consulta Internacional sobre Incontinencia (ICI) y, con permiso de la ICI, llevó a cabo la extracción de la información de relevancia. La metodología de la IV ICI consistió en una amplia revisión de la literatura por parte de expertos internacionales y en la creación de un nivel de consenso. Asimismo, el nivel de evidencia se calificó de acuerdo con un sistema Oxford modificado y los grados de recomendación se atribuyeron en consonancia. Resumen de evidencias: Está disponible una versión completa de las directrices de la EAU sobre incontinencia urinaria en formato impreso (ampliada y en formato reducido), así como en formato de CD-ROM, pudiendo solicitarse a la oficina de la EAU o en línea en la dirección (http://www.uroweb.org/guidelines/online-guidelines/). La amplitud e invasividad de la evaluación de la IU depende de la gravedad y/o complejidad de los síntomas y signos clínicos, y es diferente para varones, mujeres, personas mayores de salud delicada, niños y pacientes con neuropatías. En el nivel de tratamiento inicial se aplican pruebas básicas de diagnóstico para descartar enfermedades o problemas subyacentes, tales como infecciones del tracto urinario. El tratamiento suele ser conservador (intervenciones en los hábitos de vida, fisioterapia, terapia física, farmacoterapia) y es de naturaleza empírica. En el nivel de tratamiento especializado (cuando haya fracasado la terapia inicial, ante un diagnóstico incierto o si los síntomas y señales son complejos o graves) suele ser necesaria una evaluación más elaborada, incluyendo técnica de imagen, endoscopia y urodinámica. Entre las opciones de tratamiento se incluyen intervenciones invasivas y la cirugía. Conclusiones: Las opciones de tratamiento de la IU están creciendo en número con rapidez, y estas guías de la EAU proporcionan una gradación de las evidencias (orientada por la medicina basada en la evidencia), así como una escala de recomendaciones para que la valoración sea la adecuada y las opciones de tratamiento estén en consonancia, aplicándose así una perspectiva clínica (AU)


Context: The first European Association of Urology (EAU) guidelines on incontinence were published in 2001. These guidelines were periodically updated in past years. Objective: The aim of this paper is to present a summary of the 2009 update of the EAU guidelines on urinary incontinence (UI). Evidence acquisition: The EAU working panel was part of the 4th International Consultation on Incontinence (ICI) and, with permission of the ICI, extracted the relevant data. The methodology of the 4th ICI was a comprehensive literature review by international experts and consensus formation. In addition, level of evidence was rated according to a modified Oxford system and grades of recommendation were given accordingly. Evidence summary: A full version of the EAU guidelines on urinary incontinence is available as a printed document (extended and short form) and as a CD-ROM from the EAU office or online from the EAU Web site (http://www.uroweb.org/guidelines/online-guidelines/). The extent and invasiveness of assessment of UI depends on severity and/or complexity of symptoms and clinical signs and is different for men, women, frail older persons, children, and patients with neuropathy. At the level of initial management, basic diagnostic tests are applied to exclude an underlying disease or condition such as urinary tract infection. Treatment is mostly conservative (lifestyle interventions, physiotherapy, physical therapy, pharmacotherapy) and is of an empirical nature. At the level of specialised management (when primary therapy failed, diagnosis is unclear, or symptoms and/or signs are complex/severe), more elaborate assessment is generally required, including imaging, endoscopy, and urodynamics. Treatment options include invasive interventions and surgery. Conclusions: Treatment options for UI are rapidly expanding. These EAU guidelines provide ratings of the evidence (guided by evidence-based medicine) and graded recommendations for the appropriate assessment and according treatment options and put them into clinical perspective (AU)


Subject(s)
Humans , Male , Female , Urinary Incontinence, Stress/diagnosis , Urinary Incontinence, Stress/drug therapy , Urinary Incontinence, Stress/therapy , Urinary Incontinence, Urge/diagnosis , Urinary Incontinence, Urge/drug therapy , Urinary Incontinence, Urge/therapy , Urinary Incontinence/epidemiology , Urinary Incontinence/etiology , Urinary Incontinence, Stress/surgery , Urinary Bladder, Overactive/epidemiology , Deamino Arginine Vasopressin/therapeutic use , Estrogens/therapeutic use , Muscarinic Antagonists/therapeutic use , Adrenergic alpha-Antagonists/therapeutic use
18.
Neurourol Urodyn ; 30(7): 1388-91, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21717496

ABSTRACT

BACKGROUND: Bombesin (BOM) and gastrin releasing peptide (GRP) have been located to the lower urinary tract (LUT). However, there is a paucity of data demonstrating the impact of these endogenous peptides. OBJECTIVES: The aim of the present study was to investigate the contractile actions of BOM and GRP on the female rat urethra in vitro and in vivo. Female Sprague-Dawley rats (n = 37) weighing approximately 225 g were used. Intraurethral pressure was recorded by a catheter placed at the maximum pressure zone corresponding to the intrinsic urethral sphincter. MEASUREMENTS: In vitro, changes in intraurethral pressure was conducted on perfused intact urethral/bladder preparations and are expressed as percentages of sphincteric intraurethral pressure achieved with noradrenaline. In vivo, changes in intraurethral pressure was conducted in anesthetized subjects and compared with the baseline intraurethral pressure and sham controls. RESULTS: In vitro, the increase in intraurethral pressure induced by BOM was 23.6 ± 3.2 cmH(2)O, exceeding the pressure evoked with NA by 9.6 cmH(2) O or 174.4% whereas GRP induced a maximum pressure of 10.7 ± 1.6 cmH(2) O, an increase of 2.2 ± 0.5 cmH(2) O or 82.9% (P < 0.05) of the NA evoked pressure. In vivo, the mean baseline pressure was 22.9 ± 1.4 cmH(2) O. The intraurethral pressure evoked by BOM was 50.6 ± 6.3 cmH(2) O (P < 0.05), and for GRP, the evoked intraurethral pressure was 56.2 ± 13.4 cmH(2) O (P < 0.05). CONCLUSIONS: The present data suggest that both BOM and GRP may contribute to the control of continence by their contractile action on the sphincters of the LUT outflow region.


Subject(s)
Bombesin/administration & dosage , Gastrin-Releasing Peptide/administration & dosage , Muscle Contraction/drug effects , Urethra/drug effects , Animals , Dose-Response Relationship, Drug , Female , Norepinephrine/administration & dosage , Pressure , Rats , Rats, Sprague-Dawley , Urethra/innervation
19.
Neurourol Urodyn ; 30(5): 723-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21661020

ABSTRACT

AIMS: Detrusor underactivity (DU) is defined by the International Continence Society as a contraction of reduced strength and/or duration resulting in prolonged or incomplete emptying of the bladder but has yet received only little attention. The purpose of this report is to summarize the ICI-RS meeting in Bristol in 2010 exploring current knowledge on DU and outline directions for future research. METHODS: A think tank discussion was held and the summary of discussions was presented to all ICI-RS participants. This report is based on the final discussions. RESULTS: The understanding of the pathophysiology, epidemiology, assessment, and treatment of DU remains rudimentary. DU is defined by pressure-flow analysis but no consensus exists regarding which of the available formulae should be used for quantification of detrusor work. DU is likely to be multifactorial. Aging causes a decay in detrusor activity but other concomitant causes, either myogenic or neurogenic, may aggravate the problem resulting in decrease of detrusor contractility. No effective pharmacotherapy for the condition exists. Only a few surgical therapeutic strategies have been explored, such as neuromodulation and skeletal muscle myoplasties. Consequently, the management of affected individuals remains unsatisfactory. CONCLUSIONS: Future directions recommended by the ICI-RS panel include assessment of pathogenesis by developing novel animal models in addition to new non-invasive tests allowing longitudinal trials. Furthermore, optimizing the existing evaluation algorithms to support standard testing for DU and further epidemiological studies to quantify the size of the problem are required for the development of future treatment modalities.


Subject(s)
Urinary Bladder Diseases/physiopathology , Urinary Bladder/physiopathology , Urinary Retention/physiopathology , Urination , Animals , Biomedical Research , Humans , Pressure , Risk Factors , Terminology as Topic , Treatment Outcome , Urinary Bladder/innervation , Urinary Bladder Diseases/classification , Urinary Bladder Diseases/diagnosis , Urinary Bladder Diseases/epidemiology , Urinary Bladder Diseases/therapy , Urinary Retention/classification , Urinary Retention/diagnosis , Urinary Retention/epidemiology , Urinary Retention/therapy , Urodynamics
20.
Actas Urol Esp ; 35(7): 373-88, 2011.
Article in Spanish | MEDLINE | ID: mdl-21600674

ABSTRACT

CONTEXT: The first European Association of Urology (EAU) guidelines on incontinence were published in 2001. These guidelines were periodically updated in past years. OBJECTIVE: The aim of this paper is to present a summary of the 2009 update of the EAU guidelines on urinary incontinence (UI). EVIDENCE ACQUISITION: The EAU working panel was part of the 4th International Consultation on Incontinence (ICI) and, with permission of the ICI, extracted the relevant data. The methodology of the 4th ICI was a comprehensive literature review by international experts and consensus formation. In addition, level of evidence was rated according to a modified Oxford system and grades of recommendation were given accordingly. EVIDENCE SUMMARY: A full version of the EAU guidelines on urinary incontinence is available as a printed document (extended and short form) and as a CD-ROM from the EAU office or online from the EAU Web site (http://www.uroweb.org/guidelines/online-guidelines/). The extent and invasiveness of assessment of UI depends on severity and/or complexity of symptoms and clinical signs and is different for men, women, frail older persons, children, and patients with neuropathy. At the level of initial management, basic diagnostic tests are applied to exclude an underlying disease or condition such as urinary tract infection. Treatment is mostly conservative (lifestyle interventions, physiotherapy, physical therapy, pharmacotherapy) and is of an empirical nature. At the level of specialised management (when primary therapy failed, diagnosis is unclear, or symptoms and/or signs are complex/severe),more elaborate assessment is generally required, including imaging, endoscopy, and urodynamics. Treatment options include invasive interventions and surgery. CONCLUSIONS: Treatment options for UI are rapidly expanding. These EAU guidelines provide ratings of the evidence (guided by evidence-based medicine) and graded recommendations for the appropriate assessment and according treatment options and put them into clinical perspective.


Subject(s)
Urinary Incontinence/diagnosis , Urinary Incontinence/therapy , Algorithms , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...