Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Pathol ; 263(3): 300-314, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38606616

ABSTRACT

Steroid 5α reductase 2 (SRD5A2) converts testosterone to dihydrotestosterone and is crucial for prostatic development. 5α reductase inhibitors (5ARI) reduce prostate size in benign prostate hyperplasia (BPH) and ameliorate lower urinary tract symptoms secondary to BPH. However, the mechanisms of 5ARI functioning are still not fully understood. Here, we used a Srd5a2-/- mouse model and employed single-cell RNA sequencing to explore the impact of SRD5A2 absence on prostate cellular heterogeneity. Significant alterations in luminal epithelial cell (LE) populations were observed, alongside an increased proportion and proliferative phenotype of estrogen receptor 1 (ESR1)+ LE2 cells, following an SRD5A2-independent ESR1 differentiation trajectory. LE2 cells exhibited enhanced estrogen response gene signatures, suggesting an alternative pathway for prostate growth when SRD5A2 is absent. Human prostate biopsy analysis revealed an inverse correlation between the expressions of SRD5A2 and LE2 markers (ESR1/PKCα), and an inverse correlation between SRD5A2 and the clinical efficiency of 5ARI. These findings provide insights into 5ARI resistance mechanisms and potential alternative therapies for BPH-related lower urinary tract symptoms. © 2024 The Pathological Society of Great Britain and Ireland.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase , Epithelial Cells , Estrogen Receptor alpha , Membrane Proteins , Mice, Knockout , Prostate , Prostatic Hyperplasia , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Male , Animals , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Prostate/pathology , Prostate/metabolism , Humans , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , 5-alpha Reductase Inhibitors/pharmacology , Cell Proliferation , Disease Models, Animal , Cell Differentiation , Lower Urinary Tract Symptoms/pathology , Lower Urinary Tract Symptoms/metabolism
2.
Article in English | MEDLINE | ID: mdl-38198648

ABSTRACT

BACKGROUND: Older men frequently develop lower urinary tract symptoms attributed to benign prostatic hyperplasia (LUTS/BPH). Risk factors for LUTS/BPH include sedentary lifestyle, anxiety/depression, obesity, and frailty, which all increase with age. Although physical exercise may reduce the progression and/or severity of LUTS/BPH, the age-related mechanisms responsible remain unknown. METHODS: Voiding symptoms, body mass, and frailty were assessed after 4-weeks of voluntary wheel running in 2-month (n = 10) and 24-month (n = 8) old C57Bl/6J male mice. In addition, various social and individual behaviors were examined in these cohorts. Finally, cellular and molecular markers of inflammation and mitochondrial protein expression were assessed in prostate tissue and systemically. RESULTS: Despite running less (aged vs young X¯ = 12.3 vs 30.6 km/week; p = .04), aged mice had reduced voiding symptoms (X¯ = 67.3 vs 23.7; p < .0001) after 1 week of exercise, which was sustained through week 4 (X¯ = 67.3 vs 21.5; p < .0001). Exercise did not affect voiding symptoms in young mice. Exercise also increased mobility and decreased anxiety in both young and aged mice (p < .05). Exercise decreased expression of a key mitochondrial protein (PINK1; p < .05) and inflammation within the prostate (CD68; p < .05 and plasminogen activator inhibitor-1; p < .05) and in the serum (p < .05). However, a frailty index (X¯ = 0.17 vs 0.15; p = .46) and grip strength (X¯ = 1.10 vs 1.19; p = .24) were unchanged after 4 weeks of exercise in aged mice. CONCLUSIONS: Voluntary aerobic exercise improves voiding behavior and mobility, and decreases prostatic mitochondrial protein expression and inflammation in aged mice. This promising model could be used to evaluate molecular mechanisms of aerobic exercise as a novel lifestyle intervention for older men with LUTS/BPH.


Subject(s)
Aging , Lower Urinary Tract Symptoms , Mice, Inbred C57BL , Physical Conditioning, Animal , Animals , Male , Mice , Physical Conditioning, Animal/physiology , Aging/physiology , Lower Urinary Tract Symptoms/physiopathology , Lower Urinary Tract Symptoms/metabolism , Urination/physiology , Prostatic Hyperplasia/metabolism , Frailty/metabolism , Age Factors , Prostate/metabolism , Behavior, Animal/physiology
3.
Am J Pathol ; 194(1): 30-51, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37827216

ABSTRACT

Benign prostate hyperplasia (BPH) is caused by the nonmalignant enlargement of the transition zone of the prostate gland, leading to lower urinary tract symptoms. Although current medical treatments are unsatisfactory in many patients, the limited understanding of the mechanisms driving disease progression prevents the development of alternative therapeutic strategies. The probasin-prolactin (Pb-PRL) transgenic mouse recapitulates many histopathological features of human BPH. Herein, these alterations parallel urodynamic disturbance reminiscent of lower urinary tract symptoms. Single-cell RNA-sequencing analysis of Pb-PRL mouse prostates revealed that their epithelium mainly includes low-androgen signaling cell populations analogous to Club/Hillock cells enriched in the aged human prostate. These intermediate cells are predicted to result from the reprogramming of androgen-dependent luminal cells. Pb-PRL mouse prostates exhibited increased vulnerability to oxidative stress due to reduction of antioxidant enzyme expression. One-month treatment of Pb-PRL mice with anethole trithione (ATT), a specific inhibitor of mitochondrial ROS production, reduced prostate weight and voiding frequency. In human BPH-1 epithelial cells, ATT decreased mitochondrial metabolism, cell proliferation, and stemness features. ATT prevented the growth of organoids generated by sorted Pb-PRL basal and LSCmed cells, the two major BPH-associated, androgen-independent epithelial cell compartments. Taken together, these results support cell plasticity as a driver of BPH progression and therapeutic resistance to androgen signaling inhibition, and identify antioxidant therapy as a promising treatment of BPH.


Subject(s)
Lower Urinary Tract Symptoms , Prostatic Hyperplasia , Male , Humans , Mice , Animals , Aged , Androgens/pharmacology , Androgens/metabolism , Prostate/pathology , Prostatic Hyperplasia/metabolism , Antioxidants/pharmacology , Cell Plasticity , Hyperplasia/pathology , Lead/metabolism , Lead/therapeutic use , Mice, Transgenic , Prolactin/metabolism , Prolactin/therapeutic use , Epithelial Cells/metabolism , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/pathology
4.
Cells ; 11(13)2022 06 28.
Article in English | MEDLINE | ID: mdl-35805135

ABSTRACT

Benign prostatic hyperplasia (BPH) is one of the most common causes of lower urinary tract symptoms (LUTS) in men, which is characterized by a noncancerous enlargement of the prostate. BPH troubles the vast majority of aging men worldwide; however, the pathogenetic factors of BPH have not been completely identified. The heat shock protein 70 (HSP70) subfamily, which mainly includes HSP70, glucose-regulated protein 78 (GRP78) and GRP75, plays a crucial role in maintaining cellular homeostasis. HSP70s are overexpressed in the course of BPH and involved in a variety of biological processes, such as cell survival and proliferation, cell apoptosis, epithelial/mesenchymal transition (EMT) and fibrosis, contributing to the development and progress of prostate diseases. These chaperone proteins also participate in oxidative stress, a cellular stress response that takes place under stress conditions. In addition, HSP70s can bind to the androgen receptor (AR) and act as a regulator of AR activity. This interaction of HSP70s with AR provides insight into the importance of the HSP70 chaperone family in BPH pathogenesis. In this review, we discuss the function of the HSP70 family in prostate glands and the role of HSP70s in the course of BPH. We also review the potential applications of HSP70s as biomarkers of prostate diseases for targeted therapies.


Subject(s)
Lower Urinary Tract Symptoms , Prostatic Hyperplasia , Epithelial-Mesenchymal Transition , HSP70 Heat-Shock Proteins/metabolism , Humans , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/pathology , Male , Prostate/pathology , Prostatic Hyperplasia/metabolism
5.
Life Sci ; 289: 120237, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34922942

ABSTRACT

Tadalafil, a phosphodiesterase-5 (PDE5) inhibitor, shown to exert a protection to heart failure (HF) associated damage or lower urinary tract symptoms (LUTS). Thus, we investigated the contribution of tadalafil chronic treatment in the alterations of LUTS in HF rats. Male rats were subjected to aortocaval fistula model for HF induction. Echocardiography, cystometric, renal function and redox cell balance, as well as concentration-response curves to carbachol, KCl, ATP and frequency-response curves to electrical field stimulation (EFS) were evaluated in Sham, HF, Tadalafil and HF-Tadalafil (12 weeks endpoint) groups. HF group to present increased in left-ventricle (LV) mass and in LV end-diastolic- and LV end-systolic volume, with a decreased ejection fraction. Tadalafil treatment was able to decrease in hypertrophy and improve the LV function restoring cardiac function. For micturition function (in vivo), HF animals shown an increase in basal pressure, threshold pressure, no-voiding contractions and decreased bladder capacity, being that the tadalafil treatment restored the cystometric parameters. Contractile mechanism response (in vitro) to carbachol, KCl, ATP and EFS in the detrusor muscles (DM) were increased in the HF group, when compared to Sham group. However, tadalafil treatment restored the DM hypercontractility in the HF animals. Moreover, renal function as well as the oxidative mechanism was impaired in the HF animals, and the tadalafil treatment improved all renal and oxidative parameters in HF group. Our data shown that tadalafil has potential as multi-therapeutic drug and may be used as a pharmacological strategy for the treatment of cardiovascular, renal and urinary dysfunctions associated with HF.


Subject(s)
Heart Failure , Kidney , Lower Urinary Tract Symptoms , Tadalafil/pharmacology , Urinary Bladder , Animals , Heart Failure/drug therapy , Heart Failure/metabolism , Heart Failure/physiopathology , Kidney/metabolism , Kidney/physiopathology , Lower Urinary Tract Symptoms/drug therapy , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/physiopathology , Male , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Urinary Bladder/metabolism , Urinary Bladder/physiopathology
6.
Prostate ; 81(13): 944-955, 2021 09.
Article in English | MEDLINE | ID: mdl-34288015

ABSTRACT

BACKGROUND: Little is known about how benign prostatic hyperplasia (BPH) develops and why patients respond differently to medical therapy designed to reduce lower urinary tract symptoms (LUTS). The Medical Therapy of Prostatic Symptoms (MTOPS) trial randomized men with symptoms of BPH and followed response to medical therapy for up to 6 years. Treatment with a 5α-reductase inhibitor (5ARI) or an alpha-adrenergic receptor antagonist (α-blocker) reduced the risk of clinical progression, while men treated with combination therapy showed a 66% decrease in risk of progressive disease. However, medical therapies for BPH/LUTS are not effective in many patients. The reasons for nonresponse or loss of therapeutic response in the remaining patients over time are unknown. A better understanding of why patients fail to respond to medical therapy may have a major impact on developing new approaches for the medical treatment of BPH/LUTS. Prostaglandins (PG) act on G-protein-coupled receptors (GPCRs), where PGE2 and PGF2 elicit smooth muscle contraction. Therefore, we measured PG levels in the prostate tissue of BPH/LUTS patients to assess the possibility that this signaling pathway might explain the failure of medical therapy in BPH/LUTS patients. METHOD: Surgical BPH (S-BPH) was defined as benign prostatic tissue collected from the transition zone (TZ) of patients who failed medical therapy and underwent surgical intervention to relieve LUTS. Control tissue was termed Incidental BPH (I-BPH). I-BPH was TZ obtained from men undergoing radical prostatectomy for low-volume, low-grade prostatic adenocarcinoma (PCa, Gleason score ≤ 7) confined to the peripheral zone. All TZ tissue was confirmed to be cancer-free. S-BPH patients divided into four subgroups: patients on α-blockers alone, 5ARI alone, combination therapy (α-blockers plus 5ARI), or no medical therapy (none) before surgical resection. I-BPH tissue was subgrouped by prior therapy (either on α-blockers or without prior medical therapy before prostatectomy). We measured prostatic tissue levels of prostaglandins (PGF2α , PGI2 , PGE2 , PGD2 , and TxA2 ), quantitative polymerase chain reaction levels of mRNAs encoding enzymes within the PG synthesis pathway, cellular distribution of COX1 (PTGS1) and COX2 (PTGS2), and tested the ability of PGs to contract bladder smooth muscle in an in vitro assay. RESULTS: All PGs were significantly elevated in TZ tissues from S-BPH patients (n = 36) compared to I-BPH patients (n = 15), regardless of the treatment subgroups. In S-BPH versus I-BPH, mRNA for PG synthetic enzymes COX1 and COX2 were significantly elevated. In addition, mRNA for enzymes that convert the precursor PGH2 to metabolite PGs were variable: PTGIS (which generates PGI2 ) and PTGDS (PGD2 ) were significantly elevated; nonsignificant increases were observed for PTGES (PGE2 ), AKR1C3 (PGF2α ), and TBxAS1 (TxA2 ). Within the I-BPH group, men responding to α-blockers for symptoms of BPH but requiring prostatectomy for PCa did not show elevated levels of COX1, COX2, or PGs. By immunohistochemistry, COX1 was predominantly observed in the prostatic stroma while COX2 was present in scattered luminal cells of isolated prostatic glands in S-BPH. PGE2 and PGF2α induced contraction of bladder smooth muscle in an in vitro assay. Furthermore, using the smooth muscle assay, we demonstrated that α-blockers that inhibit alpha-adrenergic receptors do not appear to inhibit PG stimulation of GPCRs in bladder muscle. Only patients who required surgery to relieve BPH/LUTS symptoms showed significantly increased tissue levels of PGs and the PG synthetic enzymes. CONCLUSIONS: Treatment of BPH/LUTS by inhibition of alpha-adrenergic receptors with pharmaceutical α-blockers or inhibiting androgenesis with 5ARI may fail because of elevated paracrine signaling by prostatic PGs that can cause smooth muscle contraction. In contrast to patients who fail medical therapy for BPH/LUTS, control I-BPH patients do not show the same evidence of elevated PG pathway signaling. Elevation of the PG pathway may explain, in part, why the risk of clinical progression in the MTOPS study was only reduced by 34% with α-blocker treatment.


Subject(s)
Lower Urinary Tract Symptoms/drug therapy , Prostaglandins/metabolism , Prostate/metabolism , Prostatic Hyperplasia/drug therapy , 5-alpha Reductase Inhibitors/therapeutic use , Adrenergic alpha-Antagonists/therapeutic use , Aged , Humans , Lower Urinary Tract Symptoms/etiology , Lower Urinary Tract Symptoms/metabolism , Male , Middle Aged , Prostatic Hyperplasia/complications , Prostatic Hyperplasia/metabolism , Treatment Failure
7.
Nat Rev Urol ; 18(3): 139-159, 2021 03.
Article in English | MEDLINE | ID: mdl-33536636

ABSTRACT

Disruptions to sensory pathways in the lower urinary tract commonly occur and can give rise to lower urinary tract symptoms (LUTS). The unmet clinical need for treatment of LUTS has stimulated research into the molecular mechanisms that underlie neuronal control of the bladder and transient receptor potential (TRP) channels have emerged as key regulators of the sensory processes that regulate bladder function. TRP channels function as molecular sensors in urothelial cells and afferent nerve fibres and can be considered the origin of bladder sensations. TRP channels in the lower urinary tract contribute to the generation of normal and abnormal bladder sensations through a variety of mechanisms, and have demonstrated potential as targets for the treatment of LUTS in functional disorders of the lower urinary tract.


Subject(s)
Lower Urinary Tract Symptoms/metabolism , Muscle, Smooth/metabolism , Transient Receptor Potential Channels/metabolism , Urinary Bladder/metabolism , Urothelium/metabolism , Visceral Afferents/physiopathology , Female , Humans , Lower Urinary Tract Symptoms/physiopathology , Male , Muscle, Smooth/innervation , Muscle, Smooth/physiopathology , Prostate/metabolism , Prostate/physiopathology , Sensation/physiology , TRPA1 Cation Channel/metabolism , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Urethra/metabolism , Urethra/physiopathology , Urinary Bladder/innervation , Urinary Bladder/physiopathology , Urothelium/innervation
8.
PLoS One ; 16(2): e0246266, 2021.
Article in English | MEDLINE | ID: mdl-33630889

ABSTRACT

Male lower urinary tract symptoms (LUTS) comprise a common syndrome of aging that negatively impacts quality of life. The etiology of LUTS is multifactorial, involving benign prostatic hyperplasia, smooth muscle and neurologic dysfunction, inflammation, sexually transmitted infections, fibrosis, and potentially dysbiosis, but this aspect remains poorly explored. We investigated whether the presence of infectious agents in urine might be associated with LUTS by combining next-generation DNA sequencing for virus discovery, microbiome analysis for characterization of bacterial communities, and mass spectrometry-based metabolomics. In urine from 29 LUTS cases and 9 controls from Wisconsin, we found a statistically significant association between a diagnosis of LUTS and the presence of JC virus (JCV), a common neurotropic human polyomavirus (Polyomaviridae, Betapolyomavirus) linked to severe neurologic disease in rare cases. This association (based on metagenomics) was not borne out when specific polymerase chain reaction (PCR) testing was applied to this set of samples, likely due to the greater sensitivity of PCR. Interestingly, urine metabolomics analysis identified dysregulation of metabolites associated with key LUTS processes. Microbiome analysis found no evidence of microbial community dysbiosis in LUTS cases, but JCV-positive samples contained more Anaerococcus species, which are involved in polymicrobial infections of the urinary tract. Neither age nor body mass index were significantly associated with the presence of urinary JCV-in the initial group or in an additional, regionally distinct group. These data provide preliminary support the hypothesis that viruses such as JCV may play a role in the development or progression of LUTS, together with other infectious agents and host metabolic responses.


Subject(s)
JC Virus , Lower Urinary Tract Symptoms/virology , Polyomavirus Infections/complications , Aged , Case-Control Studies , High-Throughput Nucleotide Sequencing , Humans , JC Virus/genetics , JC Virus/metabolism , JC Virus/pathogenicity , Lower Urinary Tract Symptoms/etiology , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/microbiology , Male , Metabolomics , Middle Aged , Polymerase Chain Reaction , Polyomavirus Infections/virology , Tandem Mass Spectrometry
9.
Prostate ; 81(4): 252-260, 2021 03.
Article in English | MEDLINE | ID: mdl-33465254

ABSTRACT

BACKGROUND: The aim of the present study was to investigate the effects of chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) on bladder function and pathophysiology. METHODS: To create a model for CPPS, rats were intraprostatically injected with zymosan or saline, serving as control. Metabolic cage experiments were performed 7, 14, or 21 days after zymosan injection and after 14 days in the control group. Thereafter, cystometry was performed in which simulated micturition cycles were induced by saline infusion and contractile responses to the cholinergic agonist methacholine and the purinergic agonist ATP were measured. Following cystometry, the prostate and urinary bladder were excised and assessed histopathologically for possible inflammatory changes. RESULTS: Metabolic cage data revealed a significantly increased urinary frequency in zymosan treated rats. Likewise, the volume per micturition was significantly lower in all CPPS groups compared to controls. Cystometry showed a significant increase in the number of nonvoiding contractions, longer voiding time, and a trend towards lower compliance in CPPS rats compared to controls. Induction of CPPS led to significantly reduced cholinergic and purinergic contractile responses. Histopathological analysis demonstrated prostatic inflammation in all CPPS groups, in particular in later stage groups. Both the extent and grade of bladder inflammation were significantly higher in CPPS groups compared to controls. CONCLUSIONS: The current findings demonstrate a potential prostate-to-bladder cross-sensitization leading to symptoms of bladder overactivity and signs of bladder inflammation. Future clinical studies are required to verify the outcomes of the current study and enable advancement of patient care.


Subject(s)
Lower Urinary Tract Symptoms , Pelvic Pain , Prostate , Prostatitis , Urinary Bladder, Overactive , Urinary Bladder , Animals , Cholinergic Agonists/pharmacology , Chronic Pain , Inflammation/metabolism , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/physiopathology , Male , Methacholine Chloride/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Pelvic Pain/etiology , Pelvic Pain/physiopathology , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatitis/complications , Prostatitis/physiopathology , Purinergic Agonists/pharmacology , Rats , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Urinary Bladder, Overactive/metabolism , Urinary Bladder, Overactive/physiopathology , Urination/drug effects , Urination/physiology , Zymosan/pharmacology
10.
Prostate ; 80(16): 1413-1420, 2020 12.
Article in English | MEDLINE | ID: mdl-32941694

ABSTRACT

BACKGROUND: Dutasteride administration reportedly improves lower urinary tract symptoms in patient with chronic, histologically-identified prostatic inflammation, potentially through estrogen receptor ß (ERß), activation of which has anti-inflammatory effects in the prostate tissue. Therefore, we investigated the effect of dutasteride on intraprostatic inflammatory responses and bladder activity using a rat model of chemically induced prostatic inflammation. METHODS: Male Sprague-Dawley rats at 10 weeks old were used. Prostatic inflammation was induced by 5% formalin injection into ventral lobes of the prostate and saline was injected in the control group (control, n = 5). Rats with prostatic inflammation were divided into dutasteride therapy (dutasteride, n = 5) and placebo groups (placebo, n = 5). Dutasteride was administrated at a dose of 0.5 mg/kg daily from 2 days before induction of prostatic inflammation whereas placebo rats received vehicle only. Twenty-eight days later, cystometry was performed in a conscious condition to measure non-voiding contractions (NVCs), intercontraction intervals (ICI) and postvoid residual volume (RV). After cystometry, the prostate was excised for analysis of messenger RNA (mRNA) expression levels of ERα, ERß, interleukin-1ß (IL-1ß), and IL-18 by quantitative polymerase chain reaction. RESULTS: The mean number of NVCs was significantly greater in placebo group than that of control group without prostatic inflammation (p < .05), and ICI were significantly decreased in placebo group compared with control group (p < .05). On the contrary, there was no significant change in NVCs or ICI between control and dutasteride groups. RV was not significantly different among three groups. Gene expression levels of ERα, IL-1ß, and IL-18 was significantly increased in placebo rats compared with control rats (p < .05), but not significantly different between control and dutasteride rats. On the other hand, the mRNA expression level of ERß was significantly decreased in placebo rats (p < .05), but not in dutasteride rats, compared with control rats. CONCLUSION: Dutasteride treatment improved not only prostatic inflammation evident as increased gene expression levels in IL-1ß and IL-18, but also bladder overactivity shown by increased NVCs during bladder filling. These therapeutic effects were associated with the restored expression of anti-inflammatory ERß. Therefore, dutasteride might be effective via ERß modulation for the treatment of prostatic inflammation in addition to its previously known, anti-androgenic effects on benign prostatic hyperplasia.


Subject(s)
5-alpha Reductase Inhibitors/therapeutic use , Dutasteride/therapeutic use , Estrogen Receptor beta/metabolism , Lower Urinary Tract Symptoms/drug therapy , Prostatitis/drug therapy , 5-alpha Reductase Inhibitors/pharmacology , Animals , Disease Models, Animal , Dutasteride/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lower Urinary Tract Symptoms/chemically induced , Lower Urinary Tract Symptoms/metabolism , Male , Prostate/drug effects , Prostate/metabolism , Prostatitis/chemically induced , Prostatitis/metabolism , Rats , Rats, Sprague-Dawley
11.
Neurourol Urodyn ; 39(6): 1700-1707, 2020 08.
Article in English | MEDLINE | ID: mdl-32602164

ABSTRACT

AIMS: Reports link urinary dysfunction and mood disorders, such as depression, but a causative mechanism has never been postulated. Contemporary discoveries demonstrate a local inflammatory response in peripheral organs can trigger inflammation in the brain, particularly the hippocampus, mediated through the NLRP3 inflammasome. Critically, central inflammation causes depressive behavior. Since bladder outlet obstruction (BOO) evokes a local inflammatory response in the bladder, we hypothesize it will induce NLRP3-dependent inflammation in the hippocampus and depressive behavior. METHODS: There were four groups of rats: control, sham, BOO, or BOO + glyburide (an NLRP3 inhibitor). BOO was created by urethral ligation over a 1 mm catheter. Sham was tied loosely. Glyburide was provided by slow-release pellet (subcutaneous 50 mg, 21 day, replaced as needed). Rats were analyzed 12 weeks post-op for: hippocampal inflammation, microglial density, neurogenesis, and depression symptoms (open field and sucrose preference). RESULTS: BOO elicited hippocampal inflammation, accompanied by an increase in activated microglia (22%) and a decrease in neurogenesis (35%), which was blocked by glyburide. In addition, BOO rats displayed anxiety (57% decrease in exploratory behavior in the open field assay) and anhedonia (21% decrease in sucrose preference), two symptoms of depression. Like inflammation, these symptoms were diminished by glyburide to levels not statistically significantly different from controls. CONCLUSIONS: BOO, a bladder-localized event, stimulates NLRP3-dependent inflammation in the rat hippocampus after 12 weeks and this inflammation causes depressive behavior. This is the first mechanistic explanation of the link between BOO and depression and provides evidence for a distinct bladder-brain axis.


Subject(s)
Depression/etiology , Hippocampus/metabolism , Lower Urinary Tract Symptoms/complications , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Urinary Bladder Neck Obstruction/complications , Animals , Depression/metabolism , Disease Models, Animal , Female , Inflammasomes/metabolism , Inflammation/etiology , Inflammation/metabolism , Lower Urinary Tract Symptoms/metabolism , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Urinary Bladder Neck Obstruction/metabolism
12.
Neurourol Urodyn ; 39 Suppl 3: S16-S22, 2020 07.
Article in English | MEDLINE | ID: mdl-32056281

ABSTRACT

Several studies indicate that pelvic ischemia and oxidative stress may play a significant role in lower urinary tract dysfunction (LUTD), including detrusor overactivity (DO)/overactive bladder (OAB) and detrusor underactivity (DU)/underactive bladder (UAB). The present article addresses proposal 1: "Are oxidative stress and ischemia significant causes of bladder damage leading to LUTD?" from the 2019 International Consultation on Incontinence-Research Society (ICI-RS) meeting. Bladder ischemia in animals and humans is briefly described, along with the proposed progression from ischemia to LUTD. Bladder ischemia is compared with ischemia of other organs, and the ongoing development of pelvic ischemia animal models is discussed. In addition, the distribution of blood within the bladder during filling and voiding and the challenges of quantification of blood flow in vivo are described. Furthermore, oxidative stress, including potential biomarkers and treatments, and challenges regarding antioxidant therapy for the treatment of LUTD are discussed. Finally, seven critical research questions and proposed studies to answer those questions were identified as priorities that would lead to major advances in the understanding and treatment of lower urinary tract symptoms (LUTS)/LUTD associated with pelvic ischemia and oxidative stress.


Subject(s)
Ischemia/physiopathology , Lower Urinary Tract Symptoms/physiopathology , Oxidative Stress/physiology , Urinary Bladder/blood supply , Urodynamics/physiology , Animals , Humans , Ischemia/metabolism , Lower Urinary Tract Symptoms/metabolism , Urinary Bladder/metabolism , Urinary Bladder/physiopathology , Urination
13.
Low Urin Tract Symptoms ; 12(1): 47-53, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31407871

ABSTRACT

OBJECTIVE: To evaluate the effects of tadalafil monotherapy on lower urinary tract symptoms, urodynamic parameters, and oxidative stress levels in male patients. METHODS: This prospective study included 53 male patients with urinary symptoms, who met the criteria for overactive bladder (OAB) (≥ 2 points for Q3 [urgency] in the OAB symptom score [OABSS] assessment and ≥ 3 points for the total score). The patients received 5 mg tadalafil orally once daily, and their symptoms were assessed before and after the 12-week treatment. The OABSS and international prostate symptom score (IPSS) were used to evaluate the subjective symptoms. The objective findings were assessed using uroflowmetry. Oxidative stress was assessed by determining urinary levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels with an adjustment for urinary creatinine (CR) concentration. RESULTS: After tadalafil administration, total and individual indices of the OABSS assessment showed significant improvement. In addition, total storage and voiding symptoms that contributed to the IPSS were also significantly improved. The voided volume was increased, and the maximum flow rate was improved after tadalafil treatment (P = .002 and < 0.001, respectively). Urinary 8-OHdG/CR decreased from 12.4 ± 9.7 ng/mg CR to 7.6 ± 11.6 ng/mg CR (P < .001). In patients who showed OAB improvement and did not meet the criteria for OAB after the treatment (44 patients, 83.0%), the urinary 8-OHdG/CR level was significantly decreased from 11.6 ± 8.4 ng/mg CR to 6.4 ± 10.3 ng/mg CR (P < .001). CONCLUSIONS: Tadalafil treatment improves OAB symptoms and urodynamic parameters by decreasing oxidative stress level.


Subject(s)
Lower Urinary Tract Symptoms/drug therapy , Oxidative Stress/physiology , Tadalafil/therapeutic use , Urinary Bladder, Overactive/drug therapy , Urological Agents/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine/urine , Aged , Humans , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/physiopathology , Male , Prospective Studies , Treatment Outcome , Urinary Bladder, Overactive/metabolism , Urinary Bladder, Overactive/physiopathology , Urodynamics
14.
Article in English | MEDLINE | ID: mdl-31589129

ABSTRACT

BACKGROUND: The concept of metabolic syndrome (MetS) as a cluster of risk factors of type 2 diabetes and cardiovascular diseases has undergone some evolutionary transformations over the past years. Integrating the autonomic dysfunction into the pathogenesis of MetS creates the possibility of including a range of nosologies affecting treatment and clinical manifestations of pathologies belonging to MetS into the MetS cluster. The purpose of this work is to determine the involvement of autonomic dysfunction in the pathogenesis of associated pathological conditions in patients and MetS. METHODS: A cross-sectional study was conducted. The sample consisted of 158 patients with metabolic syndrome. The patients underwent a physical examination, including BMI; a blood chemistry test with the determination of the hormonal status (insulin, testosterone, dihydrotestosterone); a 24-hour monitoring of blood pressure (BP); an assessment of heart rate variability; studies showing the presence of gastric reflux (рН-measurement) or its damaging impact (endoscopy); men were tested with the IPSSQOL questionnaire and underwent transrectal ultrasound of the prostate and ultrasound of the bladder. RESULTS: It is revealed that because of MetS, the occurrence of cardiac autonomic neuropathy reaches 37.5%. Some features of gastroesophageal reflux disease in patients with MetS are shown. Regurgitation prevails in the structure of complaints. In case of fibrogastroduodenoscopy, an endoscopynegative form of the disease occurs in 38%. According to the data of daily pH-measurement, when DeMeester score is high, in the supine position, 25% of the time accounts for alkaline reflux (рН > 7). It is found out that young men experience the enlargement of prostate volume and size; according to the IPSS questionnaire, the scores correspond to the initial manifestations of hyperplastic diseases of the prostate gland due to insulin resistance and normal level of androgens. CONCLUSION: The paper demonstrates that the autonomic dysfunction of the nervous system (on a par with insulin resistance) is the main link in the development of MetS. This provides the basis for including the mentioned states - cardiac autonomic neuropathy, lower urinary tract symptoms, and gastroesophageal reflux disease - into the MetS cluster.


Subject(s)
Metabolic Syndrome/epidemiology , Metabolic Syndrome/metabolism , Adult , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/metabolism , Cluster Analysis , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Lower Urinary Tract Symptoms/diagnosis , Lower Urinary Tract Symptoms/epidemiology , Lower Urinary Tract Symptoms/metabolism , Male , Metabolic Syndrome/diagnosis , Middle Aged
15.
Trends Pharmacol Sci ; 40(9): 669-683, 2019 09.
Article in English | MEDLINE | ID: mdl-31395287

ABSTRACT

Transient receptor potential (TRP) channels are a family of functionally diverse and widely expressed cation channels involved in a variety of cell signaling and sensory pathways. Research in the last two decades has not only shed light on the physiological roles of the 28 mammalian TRP channels, but also revealed the involvement of specific TRP channels in a plethora of inherited and acquired human diseases. Considering the historical successes of other types of ion channels as therapeutic drug targets, small molecules that target specific TRP channels hold promise as treatments for a variety of human conditions. In recent research, important new findings have highlighted the central role of TRP channels in chronic pain, lower urinary tract disorders, and type 2 diabetes, conditions with an unmet medical need. Here, we discuss how these advances support the development of TRP-channel-based pharmacotherapies as valuable alternatives to the current mainstays of treatment.


Subject(s)
Chronic Pain/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Lower Urinary Tract Symptoms/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Animals , Chronic Pain/metabolism , Chronic Pain/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/pathology , Molecular Targeted Therapy , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/pathology , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , TRPV Cation Channels/metabolism
16.
Sci Rep ; 9(1): 10869, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31350464

ABSTRACT

Patients with multiple sclerosis (MS) develop a variety of lower urinary tract symptoms (LUTS). We previously characterized a murine model of neurogenic bladder dysfunction induced by a neurotropic strain of a coronavirus. In the present study, we further study the role of long-lasting neurodegeneration on the development of neurogenic bladder dysfunction in mice with corona-virus induced encephalitis (CIE). Long-term follow up study revealed three phenotypes of neurodegenerative symptom development: recovery (REC group), chronic progression (C-PRO group) and chronic disease with relapsing-remitting episodes (C-RELAP group). The levels of IL-1ß in REC group, IL-10 in C-RELAP group, and IL-1ß, IL-6, IL-10 and TNF-α in C-PRO group were diminished in the brain. The levels of TNF-α in REC group and INF-γ, IL-2, TGF-ß and TNF-α in the C-PRO group were also diminished in the urinary bladder. Mice in C-RELAP group showed a delayed recovery of voiding function. In vitro contractility studies determined a decreased basal detrusor tone and reduced amplitude of nerve-mediated contractions in C-RELAP group, whereas C-PRO group had elevated muscle-mediated contractions. In conclusion, mice with CIE developed three phenotypes of neurologic impairment mimicking different types of MS progression in humans and showed differential mechanisms driving neurogenic bladder dysfunction.


Subject(s)
Coronavirus Infections/physiopathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Lower Urinary Tract Symptoms/etiology , Multiple Sclerosis/complications , Murine hepatitis virus , Phenotype , Urinary Bladder, Neurogenic/etiology , Animals , Coronavirus Infections/virology , Cytokines/analysis , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/virology , Enzyme-Linked Immunosorbent Assay , Follow-Up Studies , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/metabolism , Lower Urinary Tract Symptoms/metabolism , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/physiopathology , Urinary Bladder, Neurogenic/metabolism
17.
Am J Physiol Renal Physiol ; 317(1): F197-F206, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31066574

ABSTRACT

Cytotoxic chemotherapy is the foundation for the treatment of the wide variety of childhood malignancies; however, these therapies are known to have a variety of deleterious side effects. One common chemotherapy used in children, doxorubicin (DOX), is well known to cause cardiotoxicity and cardiomyopathy. Recent studies have revealed that DOX impairs skeletal and smooth muscle function and contributes to fatigue and abnormal intestinal motility in patients. In this study, we tested the hypothesis that systemic DOX administration also affects detrusor smooth muscle (DSM) function in the urinary bladder, especially when administered at a young age. The effects on the DSM and bladder function were assessed in BALB/cJ mice that received six weekly intravenous injections of DOX (3 mg·kg-1·wk-1) or saline for the control group. Systemic DOX administration resulted in DSM hypertrophy, increased voiding frequency, and a significant attenuation of DSM contractility, followed by a slower relaxation compared with the control group. Gene expression analyses revealed that unlike DOX-induced cardiotoxicity, the bladders from DOX-administered animals showed no changes in oxidative stress markers; instead, downregulation of large-conductance Ca2+-activated K+ channels and altered expression of myosin light-chain kinase coincided with reduced myosin light-chain phosphorylation. These results indicate that in vivo DOX exposure caused DSM dysfunction by dysregulation of molecules involved in the detrusor contractile-relaxation mechanisms. Collectively, our findings suggest that survivors of childhood cancer treated with DOX may be at increased risk of bladder dysfunction and benefit from followup surveillance of bladder function.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Lower Urinary Tract Symptoms/chemically induced , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Myosin Light Chains/metabolism , Smooth Muscle Myosins/metabolism , Urinary Bladder Diseases/chemically induced , Urinary Bladder/drug effects , Urodynamics/drug effects , Age Factors , Animals , Female , Hypertrophy , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Lower Urinary Tract Symptoms/metabolism , Lower Urinary Tract Symptoms/pathology , Lower Urinary Tract Symptoms/physiopathology , Male , Mice, Inbred BALB C , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , Signal Transduction , Time Factors , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder/physiopathology , Urinary Bladder Diseases/metabolism , Urinary Bladder Diseases/pathology , Urinary Bladder Diseases/physiopathology
18.
Gerontology ; 65(5): 458-464, 2019.
Article in English | MEDLINE | ID: mdl-30943489

ABSTRACT

Benign prostatic hyperplasia (BPH), benign prostatic enlargement (BPE) and lower urinary tract symptoms (LUTS) belong to the most frequent diseases in ageing men. Beyond the 6th decade of life, more than 30% of men suffer from moderate to severe LUTS requiring intervention. The pathophysiology of BPH/BPE is still incompletely understood. The dominant role of the androgen system and the androgen receptor is well defined. Androgen receptors are expressed in BPH tissue in which they are activated by the potent androgen dihydrotestosterone. Synthesis of dihydrotestosterone is under control of the 5α-reductase enzyme, activity of which is antagonized by finasteride and dutasteride. More recently, the impact of prostatic inflammation and metabolic parameters particularly for the development of BPE and LUTS has increasingly been recognized. A better understanding of the pathophysiology is a prerequisite for the development of novel, more effective medical treatment options.


Subject(s)
Aging/metabolism , Lower Urinary Tract Symptoms/metabolism , Prostatic Hyperplasia/metabolism , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , 5-alpha Reductase Inhibitors/therapeutic use , Aging/immunology , Diabetes Mellitus/epidemiology , Diabetes Mellitus/metabolism , Dihydrotestosterone/metabolism , Dutasteride/therapeutic use , Dyslipidemias/epidemiology , Dyslipidemias/metabolism , Finasteride/therapeutic use , Humans , Inflammation/immunology , Lower Urinary Tract Symptoms/drug therapy , Lower Urinary Tract Symptoms/epidemiology , Lower Urinary Tract Symptoms/immunology , Male , Metabolic Syndrome/epidemiology , Metabolic Syndrome/metabolism , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/epidemiology , Prostatic Hyperplasia/immunology , Receptors, Androgen/metabolism
19.
J Urol ; 202(5): 880-889, 2019 11.
Article in English | MEDLINE | ID: mdl-30925127

ABSTRACT

PURPOSE: Lower urinary tract symptoms are prevalent and burdensome, yet methods to enhance diagnosis and appropriately guide therapies are lacking. We systematically reviewed the literature for human studies of biomarkers associated with lower urinary tract symptoms. MATERIALS AND METHODS: PubMed®, EMBASE® and Web of Science® were searched from inception to February 13, 2018. Articles were included if they were in English, performed in benign urological populations without neurological disorders or interstitial cystitis/bladder pain syndrome, and assessed a biomarker's association with or ability to predict specific lower urinary tract symptoms or urological conditions. Bioinformatic pathway analyses were conducted to determine whether individual biomarkers associated with symptoms are present in unifying pathways. RESULTS: Of 6,150 citations identified 125 met the inclusion criteria. Most studies (93.6%) assessed biomarkers at 1 time point and were cross-sectional in nature. Few studies adjusted for potentially confounding clinical variables or assessed biomarkers in an individual over time. No individual biomarkers are currently validated as diagnostic tools for lower urinary tract symptoms. Compared to controls, pathway analyses identified multiple immune response pathways that were enriched in overactive bladder syndrome and cell migration/cytoskeleton remodeling pathways that were enriched in female stress incontinence. CONCLUSIONS: Major deficiencies in the existing biomarker literature include poor reproducibility of laboratory data, unclear classification of patients with lower urinary tract symptoms and lack of adjustment for clinical covariates. Despite these limitations we identified multiple putative pathways in which panels of biological markers need further research.


Subject(s)
Biomarkers/metabolism , Lower Urinary Tract Symptoms/metabolism , Urination/physiology , Humans , Lower Urinary Tract Symptoms/physiopathology
20.
Prostate ; 79(7): 746-756, 2019 05.
Article in English | MEDLINE | ID: mdl-30811062

ABSTRACT

BACKGROUND: Inhibition of prostate smooth muscle contraction by α1 -adrenoceptor antagonists (α1 -blockers) is a first-line medical treatment of lower urinary tract symptoms suggestive of benign prostatic hyperplasia. Increased smooth muscle tone in the hyperplastic prostate may drive urethral obstruction, resulting in bladder outlet obstruction and voiding symptoms. However, efficacy of α1 -blockers is limited, as non-adrenergic mediators including endothelin-1 and thromboxane A2 (TXA2 ) increase prostate smooth muscle tension in parallel to α1 -adrenoceptors. This may maintain urethral obstruction despite therapy with α1 -blockers. Consequently, future treatment options with higher efficacy need to target α1 -adrenergic and non-adrenergic contractions simultaneouly. Recently, several compounds were reported to inhibit adrenergic or neurogenic prostate contractions, however, their effects on non-adrenergic contraction are unknown. Here, we examined effects of inhibitors for Rac-GTPase, Src family kinases (SFKs), and p21-activated kinases (PAKs) on non-adrenergic prostate contractions. METHODS: Prostate tissues were obtained from radical prostatectomy. Contractions were studied in an organ bath. Viability of cultured stromal cells was assessed by CCK-8 assay. RESULTS: Inhibition of α1 -adrenergic contractions by Rac inhibitors EHT1864 (100 µM) and NSC23766 (100 µM), and SFK inhibitors AZM475721 (10 µM) and PP2 (10 µM) was confirmed by inhibition of methoxamine-induced contractions. No effects of the PAK inhibitors FRAX486 (30 µM) and IPA3 (300 µM) on α1 -adrenergic contraction were confirmed by absent effects on methoxamine-inuced contractions. EHT1864 caused inhibition of endothelin-1- and U46619-induced contractions. EHT1864 reduced the viability of stromal cells concentration- and time-dependently. EHT1864 attenuated KCl-induced contractions of prostate strips only slightly, so that toxic effects may not account alone for inhibition of agonist-induced contractions. NSC23766 inhibited U46619-induced contractions, but not endothelin-1-induced contractions. AZM475271 had no effects on endothelin-1- or U46619-induced contractions, while PP2 inhibited U46619- but not endothelin-1-induced contractions. FRAX486 caused inhibition of U46619-induced contractions. IPA3 inhibited U46619-, but not endothelin-1-induced contractions. CONCLUSIONS: Of all six inhibitors, EHT1864 seems to be most promising from a translational point of view, as it inhibited TXA2 - and endothelin-1-induced besides α1 -adrenergic prostate contractions. This reflects divergent pharmacologic profiles of EHT1864 and NSC23766, although both are Rac-GTPase inhibitors. In vivo, urodynamic effects of EHT1864 and possibly of FRAX486 may exceed those of α1 -blockers.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Enzyme Inhibitors/pharmacology , Muscle Contraction/drug effects , Prostate/drug effects , Prostatic Hyperplasia/drug therapy , Humans , Lower Urinary Tract Symptoms/drug therapy , Lower Urinary Tract Symptoms/etiology , Lower Urinary Tract Symptoms/metabolism , Male , Muscle, Smooth/drug effects , Prostate/metabolism , Prostate/surgery , Prostatectomy , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/surgery , Stromal Cells/drug effects , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...