1
|
Cervantes A, Hughes FM, Jin H, Purves JT. Specialized pro-resolution mediators in the bladder: effects of resolvin E1 on diabetic bladder dysfunction in the type 1 diabetic male Akita mouse model. BMC Urol 2024; 24:130. [PMID: 38907230 PMCID: PMC11191353 DOI: 10.1186/s12894-024-01519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND One of the most common, but least studied, diabetic complication is diabetic bladder dysfunction. Current therapies include glucose control and symptom-based interventions. However, efficacy of these therapies is mixed and often have undesirable side effects. Diabetes is now known to be a chronic inflammatory disease. Specialized pro-resolving mediators are a class of compounds that promote the resolution of inflammation and have been shown to be effective in treating chronic inflammatory conditions. In this study we examine the ability of resolvin E1 to improve signs of diabetic bladder dysfunction. METHODS Male Akita mice (Type 1 diabetic) develop hyperglycemia at 4 weeks and signs of bladder underactivity by 15 weeks. Starting at 15 weeks, mice were given one or two weeks of daily resolvin E1 and compared to age-matched wild type and untreated Akita mice. RESULTS Resolvin E1 did not affect diabetic blood glucose after one week, although there was a slight decrease after two weeks. Diabetes decreased body weight and increased bladder weights and this was not affected by resolvin E1. Evan's blue dye extravasation (an indirect index of inflammation) was dramatically suppressed after one week of resolvin E1 treatment, but, surprisingly, had returned to diabetic levels after two weeks of treatment. Using cystometry, untreated Akita mice showed signs of underactivity (increased void volumes and intercontraction intervals). One week of resolvin E1treatment restored these cystometric findings back to control levels. After two weeks of treatment, cystometric changes were changed from controls but still significantly different from untreated levels, indicating a durable treatment effect even in the presence of increased inflammation at 2 weeks. CONCLUSIONS Resolvin E1 has a beneficial effect on diabetic bladder dysfunction in the type 1 diabetic male Akita mouse model.
Collapse
Affiliation(s)
- Anissa Cervantes
- Department of Urology, Duke University Medical Center, P.O. Box 3831, Durham, NC, 27710, USA
| | - Francis M Hughes
- Department of Urology, Duke University Medical Center, P.O. Box 3831, Durham, NC, 27710, USA.
| | - Huixia Jin
- Department of Urology, Duke University Medical Center, P.O. Box 3831, Durham, NC, 27710, USA
| | - J Todd Purves
- Department of Urology, Duke University Medical Center, P.O. Box 3831, Durham, NC, 27710, USA
| |
Collapse
|
2
|
Hudson BN, Purves JT, Hughes FM, Nagatomi J. Enzyme-induced hypoxia leads to inflammation in urothelial cells in vitro. Int Urol Nephrol 2024; 56:1565-1575. [PMID: 38133728 DOI: 10.1007/s11255-023-03900-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE To determine the contributions of different durations of hypoxia to NLRP3 inflammasome activation in urothelial cells and how ischemic changes in bladder tissues is an important chemical que that leads to pathological changes seen in BOO. METHODS A rat urothelial cell line (MYP3) was exposed to either a short duration (2 h) or long duration (6 h) of enzyme-induced hypoxia. Following exposure to a short duration of hypoxia, NO and ATP concentrations were measured from supernatant media and caspase-1 levels were measured from cell lysates. In a separate experiment, cells were fixed following hypoxia exposure and immunostained for HIF-1α stabilization. RESULTS Although short exposure of low oxygen conditions resulted in a hypoxic response in MYP3 cells, as indicated by HIF-1α stabilization and increased NO activity, NLRP3 inflammasome activation was not observed as caspase-1 activity remained unchanged. However, exposure of MYP3 cells to a longer duration of hypoxia resulted in an increase in intracellular caspase-1 activity. Furthermore, treatment with antioxidant (GSH) or TXNIP inhibitor (verapamil) attenuated the hypoxia-induced increase in caspase-1 levels indicating that hypoxia primarily drives inflammation through a ROS-mediated TXNIP/NLRP3 pathway. CONCLUSION We conclude that hypoxia induced bladder damage requires a duration that is more likely related to elevated storage pressures/hypoxia, seen in later stages of BOO, as compared to shorter duration pressure elevation/hypoxia that is encountered in normal micturition cycles or early in the BOO pathology where storage pressures are still normal.
Collapse
Affiliation(s)
- Britney N Hudson
- Department of Bioengineering, 301 Rhodes Engineering Research Center, Clemson University, Clemson, SC, 29634-0905, USA
| | - J Todd Purves
- Department of Bioengineering, 301 Rhodes Engineering Research Center, Clemson University, Clemson, SC, 29634-0905, USA
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Francis M Hughes
- Department of Bioengineering, 301 Rhodes Engineering Research Center, Clemson University, Clemson, SC, 29634-0905, USA
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Jiro Nagatomi
- Department of Bioengineering, 301 Rhodes Engineering Research Center, Clemson University, Clemson, SC, 29634-0905, USA.
| |
Collapse
|
3
|
Yang HJ, Kim DS, Lee KW, Kim YH. The Urinary Microbiome; Axis Crosstalk and Short-Chain Fatty Acid. Diagnostics (Basel) 2022; 12:3119. [PMID: 36553126 PMCID: PMC9777402 DOI: 10.3390/diagnostics12123119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/14/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Our knowledge that "urine is sterile" is no longer accepted after the development of a next-generation sequencing (NGS) test. Using NGS, microbiota in the human body were discovered, and it is expected that this will improve our understanding of human diseases. However, the mechanism involved in the effect of the microbiome on diseases is still poorly understood. Associations of gut microbiome with diseases have been recently reported. Based on such associations, bladder-gut-brain axis, gut-bladder axis, gut-vagina-bladder axis, and gut-kidney axis as novel mechanisms of action of the microbiome have been suggested. Each axis can influence the development and progression of disease through interactions. In these interactions, metabolites of the microbiome including short-chain fatty acids (SCFA) and the inflammasome play an important role. Inflammasomes are multiprotein oligomers that can initiate inflammatory responses. Inflammasomes can trigger inflammation and pyroptosis and ultimately contribute to disease development. SCFAs play an important role in immune cell migration, cytokine production, and maintenance of cellular homeostasis. Associations of inflammasomes with systemic diseases such as obesity and insulin resistance have been reported. The roles of inflammasomes and SCFAs in kidney, bladder, and prostate diseases have also been revealed recently.
Collapse
Affiliation(s)
| | | | | | - Young Ho Kim
- Department of Urology, Soonchunhyang University School of Medicine, Cheonan 31151, Republic of Korea
| |
Collapse
|
4
|
Odom MR, Hughes FM, Jin H, Purves JT. Diabetes causes NLRP3-dependent barrier dysfunction in mice with detrusor overactivity but not underactivity. Am J Physiol Renal Physiol 2022; 323:F616-F632. [PMID: 36135959 PMCID: PMC9705026 DOI: 10.1152/ajprenal.00047.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/24/2022] [Accepted: 09/13/2022] [Indexed: 02/08/2023] Open
Abstract
Approximately half of the patients with diabetes develop diabetic bladder dysfunction (DBD). The initiation and progression of DBD is largely attributed to inflammation due to dysregulated glucose and the production of toxic metabolites that activate the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome. NLRP3 activation leads to the production and release of proinflammatory cytokines and causes urothelial pyroptosis, a form of programmed cell necrosis, which we hypothesize compromises urothelial barrier integrity. Here, we investigated how NLRP3-dependent inflammation impacts barrier function during the progression of diabetes using a type 1 diabetic female Akita mouse model that progresses from an early overactive to a late underactive detrusor phenotype at 15 and 30 wk, respectively. To determine the specific role of NLRP3, Akita mice were crossbred with mice lacking the NLRP3 gene. To determine barrier function, permeability to small molecules was assessed, ex vivo using Evans blue dye and in vivo using sulfo-NHS-biotin. Both ex vivo and in vivo permeabilities were increased in diabetic mice at 15 wk. Expression of uroplakin and tight junction components was also significantly downregulated at 15 wk. Interestingly, diabetic mice lacking the NLRP3 gene showed no evidence of barrier damage or downregulation of barrier genes and proteins. At the 30-wk time point, ex vivo and in vivo barrier damage as well as barrier component downregulation was no longer evident in diabetic mice, suggesting urothelial repair or remodeling occurs between the overactive and underactive stages of DBD. Collectively, these findings demonstrate the role of NLRP3-mediated inflammation in urothelial barrier damage associated with detrusor overactivity but not underactivity.NEW & NOTEWORTHY This is the first study to demonstrate that NLRP3-mediated inflammation is responsible for urothelial barrier damage in type 1 diabetic female Akita mice with an overactive bladder. Eliminating the NLRP3 gene in these diabetic mice prevented barrier damage as a result of diabetes. By the time female Akita mice develop an underactive phenotype, the urothelial barrier has been restored, suggesting that inflammation is a critical causative factor early in the development of diabetic bladder dysfunction.
Collapse
Affiliation(s)
- Michael R Odom
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Francis M Hughes
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Huixia Jin
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - J Todd Purves
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
5
|
Hughes FM, Odom MR, Cervantes A, Purves J. Inflammation triggered by the NLRP3 inflammasome is a critical driver of diabetic bladder dysfunction. Front Physiol 2022; 13:920487. [PMID: 36505062 PMCID: PMC9733912 DOI: 10.3389/fphys.2022.920487] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
Diabetes is a rapidly expanding epidemic projected to affect as many as 1 in 3 Americans by 2050. This disease is characterized by devastating complications brought about high glucose and metabolic derangement. The most common of these complications is diabetic bladder dysfunction (DBD) and estimates suggest that 50-80% of patients experience this disorder. Unfortunately, the Epidemiology of Diabetes Interventions and Complications Study suggests that strict glucose control does not decrease ones risk for incontinence, although it does decrease the risk of other complications such as retinopathy, nephropathy and neuropathy. Thus, there is a significant unmet need to better understand DBD in order to develop targeted therapies to alleviate patient suffering. Recently, the research community has come to understand that diabetes produces a systemic state of low-level inflammation known as meta-inflammation and attention has focused on a role for the sterile inflammation-inducing structure known as the NLRP3 inflammasome. In this review, we will examine the evidence that NLRP3 plays a central role in inducing DBD and driving its progression towards an underactive phenotype.
Collapse
Affiliation(s)
- Francis M. Hughes
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | | | | | | |
Collapse
|
6
|
Yang JH, Zhao Z, Niu W, Choi HP, Azadzoi KM. Formation of Double Stranded RNA Provokes Smooth Muscle Contractions and Structural Modifications in Bladder Ischemia. Res Rep Urol 2022; 14:399-414. [PMID: 36415310 PMCID: PMC9676006 DOI: 10.2147/rru.s388464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/11/2022] [Indexed: 08/30/2023] Open
Abstract
Purpose Growing evidence suggests that ischemia provokes detrusor overactivity and degenerative responses in the bladder. Underlying mechanisms appear to involve modification of smooth muscle contractile rudiments by hypoxia, redox, cellular stress and cell survival signaling. Downstream pathways of cellular stress and stress response molecules eliciting bladder dysfunction in ischemia remain largely elusive. Our goal was to define the role of double stranded RNA (dsRNA), a stress response molecule provoked by redox, in ischemia mediated bladder dysfunction. Methods A rat model of pelvic ischemia along with a cell culture hypoxia model were used to investigate the expression levels, functional consequences, structural aspects, and regulatory mechanisms of dsRNA in the bladder. Gene and protein expression were examined by reverse transcription polymerase chain reaction (RT-PCR), dot blot, and Western blotting, respectively. Tissue structure and function were assessed using histological staining and organ bath. Regulatory mechanisms were analyzed in cultured bladder smooth muscle cells. Results The data presented here provide the first evidence of the formation of dsRNA in the overactive bladder. dsRNA is a cellular stress response molecule that sensitizes smooth muscle and regulates inflammatory and degenerative rejoinders. Our data suggest that the production of dsRNA in the bladder is provoked by ischemia. Formation of dsRNA appears to augment bladder smooth muscle contractions and provoke fibrotic and apoptotic responses. Downstream actions of dsRNA in the bladder may involve upregulation of dsRNA-activated protein kinase R (PKR) and caspase-3, the executioner of apoptosis. Conclusion Activation of dsRNA/PKR pathway may play a role in sensitization of bladder smooth muscle cells to contractile stimuli, whereas dsRNA and caspase-3 crosstalk appear to modulate cellular stress and instigate degenerative responses in bladder ischemia. These observations suggest the role of dsRNA in bladder dysfunction and may open new perspectives to overcome overactive smooth muscle contractions and structural damage in the bladder.
Collapse
Affiliation(s)
- Jing-Hua Yang
- Department of Surgery, Boston University School of Medicine and Proteomics Laboratory, VA Boston Healthcare System, Boston, MA, USA
| | - Zuohui Zhao
- Department of Urology, Boston University School of Medicine, Boston, MA, USA
| | - Wanting Niu
- Research Department, VA Boston Healthcare System, Boston, MA, USA
| | - Han-Pil Choi
- Research Department, VA Boston Healthcare System, Boston, MA, USA
| | - Kazem M Azadzoi
- Departments of Urology and Pathology, VA Boston Healthcare System and Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
7
|
Dai XY, Zhu SY, Chen J, Li MZ, Talukder M, Li JL. Role of Toll-like Receptor/MyD88 Signaling in Lycopene Alleviated Di-2-ethylhexyl Phthalate (DEHP)-Induced Inflammatory Response. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10022-10030. [PMID: 35917506 DOI: 10.1021/acs.jafc.2c03864] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Lycopene (Lyc) has anti-inflammatory and antioxidant biological functions. Di-2-ethylhexyl phthalate (DEHP) is an extremely harmful and persistent environmental pollutant and is a threat to animal health. The toll-like receptor (TLR)/MyD88 pathway is an important pathway in the inflammatory response. To illustrate the potential antagonistic action of Lyc against DEHP by the TLR/MyD88 pathway, 140 ICR mice were randomly assigned groups and continuously gavaged with corn oil, distilled water, different DEHP concentrations (500 or 1000 mg/kg BW/day), and/or Lyc (5 mg/kg BW/day) for 28 days. The data show that Lyc effectively attenuates the DEHP-induced activation of the TLR/MyD88 pathway, the upregulation of JNK expression, the content of IL-6 and TNF-α, and the downregulation of the IL-10 content, which eventually inhibit the inflammatory response and mitochondrial injuries. These findings underline the TLR/MyD88 pathway as a potential therapeutic target in DEHP and Lyc as a new therapeutic method to inhibit DEHP toxicity.
Collapse
Affiliation(s)
- Xue-Yan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, P. R. China
| | | | | | | | - Milton Talukder
- Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | | |
Collapse
|
8
|
Hughes FM, Allkanjari A, Odom MR, Jin H, Purves JT. Diabetic bladder dysfunction progresses from an overactive to an underactive phenotype in a type-1 diabetic mouse model (Akita female mouse) and is dependent on NLRP3. Life Sci 2022; 299:120528. [PMID: 35381220 PMCID: PMC9112812 DOI: 10.1016/j.lfs.2022.120528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 01/13/2023]
Abstract
AIMS Diabetic bladder dysfunction (DBD) is a prevalent diabetic complication thought to progress from overactive (OAB) to underactive (UAB) bladder. Previously we found OAB at 15 weeks in the Akita mouse, a genetic model of Type 1 diabetes. The first aim of this study assesses bladder function at 30 weeks to assess progression. In addition, inflammation triggered by the NLRP3 inflammasome is implicated in DBD. In a second aim we assessed a role for NLRP3 by crossing Akita mice with NLRP3-/- mice. MAIN METHODS Akita mice were bred with NLRP3-/- mice. The effect of diabetes was assessed by comparing nondiabetic to diabetic mice (all NLRP3+/+). The effect of diabetes in the absence of the NLRP3 inflammasome was assessed by comparing nondiabetic/NLRP3-/- to diabetic/NLRP3-/- mice. Mice were assessed at 30 weeks for blood glucose (glucometer), inflammation (Evans blue), bladder morphology (histology) and bladder function (urodynamics). KEY FINDINGS At 30 weeks blood glucose of nondiabetics and diabetics was not affected by the presence of absence of NLRP3. Diabetic/NLRP3+/+ mice showed bladder inflammation and detrusor hypertrophy which was blocked in the diabetic/NLRP3-/- mice, clearly showing a role for NLRP3. When bladder function was examined, diabetic/NLRP3+/+ showed an increase in voiding volume and a decrease in frequency, two signs of underactive bladder. However, in the NLRP3-/- mice, diabetes was unable to effectuate these changes, demonstrating that NLRP3-induced inflammation is responsible for UAB symptoms in these mice. SIGNIFICANCE Akita diabetic mice progress from OAB to UAB. NLRP3 is a possible target to treat DBD.
Collapse
Affiliation(s)
- Francis M Hughes
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States of America.
| | - Armand Allkanjari
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| | - Michael R Odom
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| | - Huixia Jin
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| | - J Todd Purves
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, United States of America
| |
Collapse
|
9
|
Hughes FM, Harper SN, Nosé BD, Allkanjari A, Zheng MT, Jin H, Purves JT. Specialized Pro-resolution Mediators in the bladder; Annexin-A1 normalizes inflammation and bladder dysfunction during bladder outlet obstruction. Am J Physiol Renal Physiol 2021; 321:F443-F454. [PMID: 34396790 DOI: 10.1152/ajprenal.00205.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Bladder Outlet Obstruction (BOO) is ultimately experienced by ≈90% of men, most commonly secondary to benign prostatic hyperplasia. Inflammation is a critical driver of BOO pathology in the bladder and can be divided into two critical steps; initiation and resolution. While great strides have been made toward understanding initiation of inflammation in the bladder (through the NLRP3 inflammasome), no studies have examined resolution. Resolution is controlled by 5 classes of compounds known as Specialized Pro-resolving Mediators (SPMs), all of which bind to one or more of 7 different receptors. Using immunocytochemistry, we show the presence of 6 of the known SPM receptors in the bladder of control and BOO rats; the 7th has no rodent homolog. The expression was predominantly localized to the urothelia, often with some expression in the smooth muscle, but little to none in the interstitial cells. We next examined the therapeutic potential of the Annexin-A1 resolution system, also present in control and BOO bladders. Using the peptide mimetic Ac2-26, we blocked inflammation-initiating pathways (NLRP3 activation), diminished BOO-induced inflammation (Evans blue dye extravasation), and normalized bladder dysfunction (urodynamics). Excitingly, Ac2-26 also promoted faster and more complete functional recovery after surgical de-obstruction. Together, the results demonstrate that the bladder expresses a wide variety of potential pro-resolving pathways and that modulation of just one of these pathways can alleviate many detrimental aspects of BOO and speed recovery after de-obstruction. This work establishes a precedent for future studies evaluating SPM effectiveness in resolving the many conditions associated with bladder inflammation.
Collapse
Affiliation(s)
- Francis M Hughes
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Shelby N Harper
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Brent D Nosé
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Armand Allkanjari
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Michael T Zheng
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Huixia Jin
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - J Todd Purves
- Division of Urology, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
10
|
BOO induces fibrosis and EMT in urothelial cells which can be recapitulated in vitro through elevated storage and voiding pressure cycles. Int Urol Nephrol 2021; 53:2007-2018. [PMID: 34232473 DOI: 10.1007/s11255-021-02942-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE To determine the unique contributions from elevated voiding and storage pressures in the development of fibrosis and the epithelial-to-mesenchymal transition (EMT) in urothelial cells, and how progressive BOO pressure cycling is an important mechanical cue leading to these pathological changes. MATERIALS AND METHODS Urothelial cells isolated from control, SHAM, 2 (acute)- or 6 (chronic)-week BOO rats treated with an inflammasome inhibitor or no drug. Total RNA was isolated and RT-PCR was conducted with custom primers for pro-fibrotic and EMT genes. In separate experiments, a rat urothelial cell line was exposed to cyclic pressure regimes characteristic of acute and chronic BOO in the presence or absence of an inflammasome inhibitor. Following exposure, RT-PCR was conducted, collagen content was determined and intracellular caspase-1 activity was measured. RESULTS Urothelial cells isolated from acute and chronic BOO rat models demonstrated expression of pro-fibrotic and EMT genes. Similarly, MYP3 rat urothelial cells subjected to pressure cycling regimes that reflect intravesical pressures in the acute or chronic BOO bladder also demonstrated increased expression of pro-fibrotic and EMT genes, along with elevated soluble collagen. Treatment with inflammasome inhibitors reduced expression of pro-fibrotic genes in the rat model and pressure cycling model but had a limited effect on EMT. CONCLUSION These results indicate that acute and chronic BOO pressure cycling are essential in the initiation and progression of fibrosis in the bladder via the NLRP3 inflammasome, but also provide new evidence that there is also an alternative NLRP3-independent pathway leading to EMT and fibrosis.
Collapse
|
11
|
Harper SN, Leidig PD, Hughes FM, Jin H, Purves JT. Calcium Pyrophosphate And Monosodium Urate Activate The NLRP3 Inflammasome Within Bladder Urothelium Via Reactive Oxygen Species And TXNIP. Res Rep Urol 2019; 11:319-325. [PMID: 31819864 PMCID: PMC6879004 DOI: 10.2147/rru.s225767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/21/2019] [Indexed: 12/24/2022] Open
Abstract
Objective To investigate the in vitro activation of the NLRP3 inflammasome within bladder urothelium by stone-forming components. Further, to describe the contributions of reactive oxygen species (ROS) and thioredoxin-interacting protein (TXNIP), an important structural component of the inflammasome, to this activation. Methods Urothelial cells were harvested and incubated overnight. For agonist studies, cells were treated with varying concentrations of calcium pyrophosphate (CPPD) and monosodium urate (MSU). For inhibitor studies, cells were treated with either N-acetylcysteine (NAC) (1 hr) or Verapamil (4 hrs) prior to incubation with either CPPD (62.5 ug/mL) or MSU (1.25 ug/mL) for 24 hrs. Untreated controls were incubated with ATP (1.25 mM) for 1 hr to maximally stimulate NLRP3 inflammasome activity (measured as caspase-1 cleavage of the fluorogenic substrate Ac-YVAD-AFC). Results are reported as a percentage of maximum ATP response. Results CPPD and MSU activate caspase-1 in urothelial cells in a dose-dependent manner, reaching ~50% and ~25% of the ATP response, respectively. Pre-treatment with the general ROS scavenger NAC reduces this activation in a dose-dependent manner. Additionally, activation was suppressed through treatment with Verapamil, a known downregulator of TXNIP expression. Conclusion The stone components CPPD and MSU activate NLRP3 in an ROS and TXNIP-dependent manner in bladder urothelium. These findings demonstrate the importance of ROS and TXNIP, and suggest that targeting either may be a way to decrease stone-dependent NLRP3 inflammation within the bladder.
Collapse
Affiliation(s)
- Shelby N Harper
- Duke University School of Medicine, Durham, NC, USA.,Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Patrick D Leidig
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Francis M Hughes
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Huixia Jin
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - J Todd Purves
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA.,Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
12
|
Hughes FM, Hirshman NA, Inouye BM, Jin H, Stanton EW, Yun CE, Davis LG, Routh JC, Purves JT. NLRP3 Promotes Diabetic Bladder Dysfunction and Changes in Symptom-Specific Bladder Innervation. Diabetes 2019; 68:430-440. [PMID: 30425063 PMCID: PMC6341307 DOI: 10.2337/db18-0845] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022]
Abstract
The NLRP3 inflammasome senses diabetic metabolites and initiates inflammation implicated in diabetic complications and neurodegeneration. No studies have investigated NLRP3 in diabetic bladder dysfunction (DBD), despite a high clinical prevalence. In vitro, we found that numerous diabetic metabolites activate NLRP3 in primary urothelial cells. In vivo, we demonstrate NLRP3 is activated in urothelia from a genetic type 1 diabetic mouse (Akita) by week 15. We then bred an NLRP3-/- genotype into these mice and found this blocked bladder inflammation and cystometric markers of DBD. Analysis of bladder innervation established an NLRP3-dependent decrease in overall nerve density and Aδ-fibers in the bladder wall along with an increase in C-fiber populations in the urothelia, which potentially explains the decreased sense of bladder fullness reported by patients and overactivity detected early in DBD. Together, the results demonstrate the role of NLRP3 in the genesis of DBD and suggest specific NLRP3-mediated neuronal changes can produce specific DBD symptoms.
Collapse
Affiliation(s)
- Francis M Hughes
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
- Department of Bioengineering, Clemson University, Clemson, SC
| | - Nathan A Hirshman
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
| | - Brian M Inouye
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
| | - Huixia Jin
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
| | - Eloise W Stanton
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
| | - Chloe E Yun
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
| | - Leah G Davis
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
- Duke Cancer Center Biostatistics, Duke University Medical Center, Durham, NC
| | - Jonathan C Routh
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - J Todd Purves
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC
- Department of Bioengineering, Clemson University, Clemson, SC
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| |
Collapse
|
13
|
Tudrej KB, Piecha T, Kozłowska-Wojciechowska M. Role of NLRP3 inflammasome in the development of bladder pain syndrome interstitial cystitis. Ther Adv Urol 2019; 11:1756287218818030. [PMID: 30671141 PMCID: PMC6329030 DOI: 10.1177/1756287218818030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/17/2018] [Indexed: 12/20/2022] Open
Abstract
Although it has been proposed that NOD-like receptor protein 3 (NLRP3) inflammasome activation may have an important contribution to the onset of bladder pain syndrome/interstitial cystitis (BPS/IC), as of today there is still insufficient evidence to accept or to reject this hypothesis. However, taking into consideration that inflammasomes have been already shown as important mediators of cyclophosphamide-induced bladder inflammation and that some studies have also revealed human bladder epithelium expresses high levels of NLRP3, such a hypothesis seems to be reasonable. The purpose of this review is to discuss a scenario that NLRP3 inflammasome is a crucial player in the development of this disease. Identification of a novel mediator of bladder inflammation and pain could lead to emerging new therapeutic strategy and the first causative therapy.
Collapse
Affiliation(s)
- Karol Borys Tudrej
- Medical University of Warsaw, Banacha 1, Warszawa, Mazowieckie, 02-097, Poland
| | - Tomasz Piecha
- Medical University of Warsaw, Warszawa, Mazowieckie, Poland
| | | |
Collapse
|
14
|
Inouye BM, Hughes FM, Jin H, Lütolf R, Potnis KC, Routh JC, Rouse DC, Foo WC, Purves JT. Diabetic bladder dysfunction is associated with bladder inflammation triggered through hyperglycemia, not polyuria. Res Rep Urol 2018; 10:219-225. [PMID: 30533402 PMCID: PMC6247963 DOI: 10.2147/rru.s177633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Diabetes is a grave and progressive condition characterized by debilitating complications. Diabetic bladder dysfunction (DBD) is a very common complication with no specific treatments currently available. Unlike other tissues affected by this disease, the bladder is subjected to two independent insults; 1) polyuria, created by the osmotic effects of glucose in the urine, and 2) hyperglycemia itself. Based on our understanding of inflammation as a major contributor to the underlying organ damage in several other diabetic complications, its presence in the bladder during DBD and the contribution of polyuria and hyperglycemia to its development were assessed. Methods Awake, restrained cystometry was performed on wild type C57BL/6 mice and diabetic (Akita) mice on a C57BL/6 background at 15 weeks of age. A subgroup of the Akita mice were treated with phlorizin, an inhibitor of sodium-glucose linked transporter types 1 and 2 that prevents glucose reabsorption in the kidney. All groups were assessed for serum glucose, 4-hour voiding totals, and inflammation in the bladder (Evans blue assay). Results Akita mice develop cystometrically-defined DBD by 15 weeks of age, as evidenced by an increase in urinary frequency, a decrease in voiding volume, and an increase in post-voiding residual volume. Phlorizin effectively normalized serum glucose in these animals while increasing the urine output. Inflammation in the bladder was present in the diabetic animals at this time point, but not detectable in animals receiving phlorizin. Conclusion Inflammation in the bladder of diabetic mice correlates with the development of DBD and is triggered by hyperglycemia, not polyuria.
Collapse
Affiliation(s)
- Brian M Inouye
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA,
| | - Francis M Hughes
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA, .,Department of Bioengineering, Clemson University, Clemson, SC, USA,
| | - Huixia Jin
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA,
| | - Robin Lütolf
- Department of Health Science and Technology, ETH Zurich, Zürich 8092, Switzerland
| | - Kunal C Potnis
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA,
| | - Jonathan C Routh
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA, .,Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Douglas C Rouse
- Division of Laboratory Animal Medicine, Duke University Medical Center, Durham, NC, USA
| | - Wen-Chi Foo
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - J Todd Purves
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA, .,Department of Bioengineering, Clemson University, Clemson, SC, USA, .,Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
15
|
Dunton CL, Purves JT, Hughes FM, Jin H, Nagatomi J. Elevated hydrostatic pressure stimulates ATP release which mediates activation of the NLRP3 inflammasome via P2X 4 in rat urothelial cells. Int Urol Nephrol 2018; 50:1607-1617. [PMID: 30099658 PMCID: PMC6129973 DOI: 10.1007/s11255-018-1948-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Partial bladder outlet obstruction (pBOO) is a prevalent urological condition commonly accompanied by increased intravesical pressure, inflammation, and fibrosis. Studies have demonstrated that pBOO results in increased NLRP3 inflammasome and caspase-1 activation and that ATP is released from urothelial cells in response to elevated pressure. In the present study, we investigated the role of elevated pressure in triggering caspase-1 activation via purinergic receptors activation in urothelial cells. Rat urothelial cell line, MYP3 cells, was subjected to hydrostatic pressures of 15 cmH2O for 60 min, or 40 cmH2O for 1 min to simulate elevated storage and voiding pressure conditions, respectively. ATP concentration in the supernatant media and intracellular caspase-1 activity in cell lysates were measured. Pressure experiments were repeated in the presence of antagonists for purinergic receptors to determine the mechanism for pressure-induced caspase-1 activation. Exposure of MYP3 cells to both pressure conditions resulted in an increase in extracellular ATP levels and intracellular caspase-1 activity. Treatment with P2X7 antagonist led to a decrease in pressure-induced ATP release by MYP3 cells, while P2X4 antagonist had no effect but both antagonists inhibited pressure-induced caspase-1 activation. Moreover, when MYP3 cells were treated with extracellular ATP (500 µM), P2X4 antagonist inhibited ATP-induced caspase-1 activation, but not P2X7 antagonist. We concluded that pressure-induced extracellular ATP in urothelial cells is amplified by P2X7 receptor activation and ATP-induced-ATP release. The amplified ATP signal then activates P2X4 receptors, which mediate activation of the caspase-1 inflammatory response.
Collapse
Affiliation(s)
- Cody L Dunton
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - J Todd Purves
- Department of Bioengineering, Clemson University, Clemson, SC, USA.,Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, USA.,Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Francis M Hughes
- Department of Bioengineering, Clemson University, Clemson, SC, USA.,Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Huixia Jin
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Jiro Nagatomi
- Department of Bioengineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
16
|
Inouye BM, Hughes FM, Sexton SJ, Purves JT. The Emerging Role of Inflammasomes as Central Mediators in Inflammatory Bladder Pathology. Curr Urol 2017; 11:57-72. [PMID: 29593464 DOI: 10.1159/000447196] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/09/2017] [Indexed: 12/18/2022] Open
Abstract
Irritative voiding symptoms (e.g. increased frequency and urgency) occur in many common pathologic conditions such as urinary tract infections and bladder outlet obstruction, and these conditions are well-established to have underlying inflammation that directly triggers these symptoms. However, it remains unclear as to how such diverse stimuli individually generate a common inflammatory process. Jürg Tschopp provided substantial insight into this conundrum when, working with extracts from THP-1 cells, he reported the existence of the inflammasome. He described it as a structure that senses multiple diverse signals from intracellular/extracellular sources and pathogens and triggers inflammation by the maturation and release of the pro-inflammatory cytokines interleukin-1β and interleukin-18. Recently, many of these sensors were found in the bladder and the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3, has been shown to be a central mediator of inflammation in several urological diseases. In this review, we introduce the nucleotide-binding domain, leucine-rich-containing family, pyrin domaincontaining-3 inflammasome, highlight its emerging role in several common urologic conditions, and speculate on the potential involvement of other inflammasomes in bladder pathology.
Collapse
Affiliation(s)
- Brian M Inouye
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Francis M Hughes
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - Stephanie J Sexton
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| | - J Todd Purves
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
17
|
Abstract
Urinary tract infections (UTIs) cause a huge burden of morbidity worldwide with recurrent UTIs becoming increasingly frequent owing to the emergence of antibiotic-resistant bacterial strains. Interactions between the innate and adaptive immune responses to pathogens colonizing the urinary tract have been the focus of much research. Inflammasomes are part of the innate immune defence and can respond rapidly to infectious insult. Assembly of the multiprotein inflammasome complex activates caspase-1, processes proinflammatory cytokines IL-1β and IL-18, and induces pyroptosis. These effector pathways, in turn, act at different levels to either prevent or resolve infection, or eliminate the infectious agent itself. In certain instances, inflammasome activation promotes tissue pathology; however, the precise functions of inflammasomes in UTIs remain unexplored. An improved understanding of inflammasomes could provide novel approaches for the design of diagnostics and therapeutics for complicated UTIs, enabling us to overcome the challenge of drug resistance.
Collapse
|
18
|
Purves JT, Hughes FM. Inflammasomes in the urinary tract: a disease-based review. Am J Physiol Renal Physiol 2016; 311:F653-F662. [PMID: 27170685 DOI: 10.1152/ajprenal.00607.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/04/2016] [Indexed: 12/28/2022] Open
Abstract
Inflammasomes are supramolecular structures that sense molecular patterns from pathogenic organisms or damaged cells and trigger an innate immune response, most commonly through production of the proinflammatory cytokines IL-1β and IL-18, but also through less understood mechanisms independent of these cytokines. Great strides have been made in understanding these structures and their dysfunction in various inflammatory diseases, lending new insights into urological and renal problems. From a clinical perspective, benign urinary pathology almost universally involves the inflammatory process, and understanding how inflammasomes translate etiological conditions (diabetes, obstruction, stones, urinary tract infections, etc.) into acute and chronic inflammatory responses is critical to understanding these diseases at a molecular level. To date, inflammasome components have been found in the bladder, prostate, and kidney and have been shown to be activated in response to several infectious and noninfectious insults. In this review, we summarize what is known regarding inflammasomes in both the upper and lower urinary tract and describe several common disease states where they potentially play critical roles.
Collapse
Affiliation(s)
- J Todd Purves
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - F Monty Hughes
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
19
|
Hughes FM, Kennis JG, Youssef MN, Lowe DW, Shaner BE, Purves JT. The NACHT, LRR and PYD Domains-Containing Protein 3 (NLRP3) Inflammasome Mediates Inflammation and Voiding Dysfunction in a Lipopolysaccharide-Induced Rat Model of Cystitis. ACTA ACUST UNITED AC 2016; 7. [PMID: 27066297 DOI: 10.4172/2155-9899.1000396] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE NOD-like receptors (NLRs) sense sterile and non-sterile signals and form inflammasomes which trigger an inflammatory response through the activation of caspase-1 and release of IL-1β. Recently we have shown the presence of several NLRs in the bladder urothelia and demonstrated the importance of NLRP3 in bladder outlet obstruction and cyclophosphamide-induced cystitis, both models of sterile inflammation. In this study we explore a role for NLRP3 in mediating the response to LPS, a key antigen of uropathogenic bacteria. METHOD In order to bypass the protective glycosaminoglycan layer lining the urothelium, LPS was directly injected into the bladder wall of Sprague-Dawley rats. Glyburide (a NLRP3 inhibitor) or vehicle was administered orally prior to and after injection. Rats were analyzed 24 h later. Inflammasome activity (caspase-1 activity, IL-1β release) and inflammation (Evan's Blue extravasation, bladder weight) were assessed, as was physiological bladder function (urodynamics). RESULTS Injection of LPS stimulated inflammasome activation (caspase-1 activity) and the release of IL-1β into the urine which was prevented by glyburide. Likewise, LPS increased inflammation, (bladder weight and the extravasation of Evan's blue dye), and this was reversed by glyburide. Functionally, animals injected with saline alone demonstrated decreased voiding volume as measured by urodynamics. In the presence of LPS, additional urinary dysfunction was evident with decreased voiding pressures and threshold pressures. The decrease in voiding pressure was blocked by glyburide but the decrease in threshold pressure was not, suggesting that LPS has significant effects mediated by inflammasome-dependent and -independent mechanisms. CONCLUSION Overall, the results demonstrate the potential importance of inflammasomes in bacterial cystitis as well as the ability of the bladder wall injection technique to isolate the in vivo effects of specific inflammasome ligands to the physiological changes associated with cystitis.
Collapse
Affiliation(s)
- Francis M Hughes
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA; Department of Urology, Medical University of South Carolina, Charleston, SC, USA
| | - James G Kennis
- Department of Urology, Medical University of South Carolina, Charleston, SC, USA
| | - Melissa N Youssef
- Department of Urology, Medical University of South Carolina, Charleston, SC, USA
| | - Danielle W Lowe
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Brooke E Shaner
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - J Todd Purves
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, NC, USA; Department of Urology, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
20
|
The NLRP3 Inflammasome Mediates Inflammation Produced by Bladder Outlet Obstruction. J Urol 2015; 195:1598-1605. [PMID: 26707508 DOI: 10.1016/j.juro.2015.12.068] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2015] [Indexed: 12/11/2022]
Abstract
PURPOSE While bladder outlet obstruction is well established to elicit an inflammatory reaction in the bladder that leads to overactive bladder and fibrosis, little is known about the mechanism by which this is initiated. NLRs (NOD-like receptors) and the structures that they form (inflammasomes) have been identified as sensors of cellular damage, including pressure induced damage, and triggers of inflammation. Recently we identified these structures in the urothelium. In this study we assessed the role of the NLRP3 (NACHT, LRR and PYD domains-containing protein 3) inflammasome in bladder dysfunction resulting from bladder outlet obstruction. MATERIALS AND METHODS Bladder outlet obstruction was created in female rats by inserting a 1 mm outer diameter transurethral catheter, tying a silk ligature around the urethra and removing the catheter. Untreated and sham operated rats served as controls. Rats with bladder outlet obstruction were given vehicle (10% ethanol) or 10 mg/kg glyburide (a NLRP3 inhibitor) orally daily for 12 days. Inflammasome activity, bladder hypertrophy, inflammation and bladder function (urodynamics) were assessed. RESULTS Bladder outlet obstruction increased urothelial inflammasome activity, bladder hypertrophy and inflammation, and decreased voided volume. Glyburide blocked inflammasome activation, reduced hypertrophy and prevented inflammation. The decrease in voided volume was also attenuated by glyburide mechanistically as an increase in detrusor contraction duration and voiding period. CONCLUSION Results suggest the importance of the NLRP3 inflammasome in the induction of inflammation and bladder dysfunction secondary to bladder outlet obstruction. Arresting these processes with NLRP3 inhibitors may prove useful to treat the symptoms that they produce.
Collapse
|