Next Article in Journal
ACE Phenotyping in Human Blood and Tissues: Revelation of ACE Outliers and Sex Differences in ACE Sialylation
Previous Article in Journal
The Randomized, Multicenter, Open-Label, Controlled POLBOS 3 Trial Comparing Regular Drug-Eluting Stents and the Sirolimus-Eluting BiOSS LIM C Dedicated Coronary Bifurcation Stent: Four-Year Results
Previous Article in Special Issue
Neurosecretory Protein GM–Expressing Neurons Participate in Lipid Storage and Inflammation in Newly Developed Cre Driver Male Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Methylglyoxal and Advanced Glycation End Products (AGEs): Targets for the Prevention and Treatment of Diabetes-Associated Bladder Dysfunction?

by
Akila Lara Oliveira
,
Mariana Gonçalves de Oliveira
,
Fabíola Zakia Mónica
and
Edson Antunes
*
Department of Translational Medicine, Pharmacology Area, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(5), 939; https://doi.org/10.3390/biomedicines12050939
Submission received: 19 March 2024 / Revised: 11 April 2024 / Accepted: 17 April 2024 / Published: 23 April 2024
(This article belongs to the Special Issue Molecular Research in Obesity)

Abstract

:
Methylglyoxal (MGO) is a highly reactive α-dicarbonyl compound formed endogenously from 3-carbon glycolytic intermediates. Methylglyoxal accumulated in plasma and urine of hyperglycemic and diabetic individuals acts as a potent peptide glycation molecule, giving rise to advanced glycation end products (AGEs) like arginine-derived hydroimidazolone (MG-H1) and carboxyethyl-lysine (CEL). Methylglyoxal-derived AGEs exert their effects mostly via activation of RAGE, a cell surface receptor that initiates multiple intracellular signaling pathways, favoring a pro-oxidant environment through NADPH oxidase activation and generation of high levels of reactive oxygen species (ROS). Diabetic bladder dysfunction is a bothersome urological complication in patients with poorly controlled diabetes mellitus and may comprise overactive bladder, urge incontinence, poor emptying, dribbling, incomplete emptying of the bladder, and urinary retention. Preclinical models of type 1 and type 2 diabetes have further confirmed the relationship between diabetes and voiding dysfunction. Interestingly, healthy mice supplemented with MGO for prolonged periods exhibit in vivo and in vitro bladder dysfunction, which is accompanied by increased AGE formation and RAGE expression, as well as by ROS overproduction in bladder tissues. Drugs reported to scavenge MGO and to inactivate AGEs like metformin, polyphenols, and alagebrium (ALT-711) have shown favorable outcomes on bladder dysfunction in diabetic obese leptin-deficient and MGO-exposed mice. Therefore, MGO, AGEs, and RAGE levels may be critically involved in the pathogenesis of bladder dysfunction in diabetic individuals. However, there are no clinical trials designed to test drugs that selectively inhibit the MGO–AGEs–RAGE signaling, aiming to reduce the manifestations of diabetes-associated bladder dysfunction. This review summarizes the current literature on the role of MGO–AGEs–RAGE–ROS axis in diabetes-associated bladder dysfunction. Drugs that directly inactivate MGO and ameliorate bladder dysfunction are also reviewed here.

1. Introduction

Methylglyoxal (MGO) is a highly reactive α-dicarbonyl compound endogenously generated during the glycolytic pathway [1]. Hyperglycemia in diabetic and obese patients markedly elevates the plasma and urine levels of MGO as a consequence of the glycolytic overload [2]. The abnormal accumulation of MGO has been referred to as dicarbonyl stress, which may be implicated in many diseases [3]. Methylglyoxal promotes post-translational modification of peptides or proteins, ultimately leading to the formation of advanced glycation end products (AGEs), the most studied of which include arginine-derived hydroimidazolone (MG-H1) and carboxyethyl-lysine (CEL) [1]. MGO also covalently modifies DNA, leading to nucleic acid AGE formation, consisting mainly of guanine adducts. AGEs bind their cell membrane-anchored ligand receptor, termed RAGE [4], triggering multiple intracellular signaling pathways, including the activation of NADPH oxidase that leads to increased production of reactive oxygen species (ROS), thus contributing to generate a pro-oxidant environment. Diabetic bladder dysfunction (DBD) is a highly prevalent condition that may affect the detrusor, nerve fiber terminals, urothelium, and urethra, and manifests as storage problems such as OAB and urge incontinence, and voiding problems such as decreased sensation and increased capacity [5,6]. DBD may progress from detrusor overactivity at initial stages to detrusor underactivity at advanced stages of this disease, a condition defined by the International Continence Society (ICS) as contraction of reduced strength and/or duration, resulting in prolonged bladder emptying and/or a failure to achieve complete bladder emptying within a normal timespan [5,7,8,9]. The underactive bladder comprises mostly voiding phase symptoms such as slow stream, intermittency, hesitancy, feeling of incomplete emptying of the bladder, and urinary retention [10]. Increased capacity and decreased sensation, together with recurrent urinary tract infections, may also be present in DBD [11,12]. Preclinical models of type 1 (streptozotocin, Akita mice) and type 2 diabetes (high-fat diets, ob/ob and db/db mice) have provided further evidence confirming the relationship between diabetes and obesity with voiding dysfunction. However, little is known about the importance of MGO generation and, hence, AGEs–RAGE activation in the pathophysiology of diabetic-associated bladder dysfunction [13]. Interestingly, in mice treated orally with MGO for prolonged periods, voiding spot assays in conscious mice and urodynamic evaluation in anesthetized mice revealed significant increases in total void volume, volume per void, micturition frequency, and nonvoiding contractions number, along with enhanced in vitro bladder contractility [14]. In addition, elevated levels of MGO, AGEs, RAGE, and ROS were found in bladder tissues from mice chronically treated with MGO, pointing out that they could be important markers of DBD pathophysiology [15]. Similar data were obtained in bladder tissues of diabetic obese ob/ob mice [16]. The antihyperglycemic drug metformin [17,18,19] and polyphenols like resveratrol and epigallocatechin-3-gallate [20] can directly scavenge MGO, explaining, at least in part, their capacity to ameliorate diabetes-associated bladder dysfunction. However, no clinical trials exist aiming to test inhibitors of the MGO–AGEs–RAGE signaling as potential drugs to prevent and treat manifestations of diabetes-associated bladder dysfunction. Therefore, the design and development of new drugs that inhibit the MGO–AGEs–RAGE axis may become an interesting approach for the prevention and treatment of bladder dysfunction in diabetic conditions. The present review aimed to provide an updated overview on bladder dysfunction in diabetic and obesity conditions in animals and humans, emphasizing the MGO–AGEs–RAGE signaling pathway as a potential mechanism implicated in the pathophysiology of this disorder, focusing on bladder overactivity. Drugs that inactivate MGO or inhibit AGEs formation in parallel to reducing diabetic-associated bladder dysfunction are also reviewed here.

2. Lower Urinary Tract Symptoms (LUTS) and Overactive Bladder (OAB) Syndrome

Urinary bladder function is regulated by a complex interaction of efferent and afferent fibers from the autonomic nervous system and somatic innervation [21]. An imbalance between these systems leads to lower urinary tract symptoms (LUTS), which comprise storage, voiding, and post-micturition symptoms [5]. Storage symptoms consist of altered bladder sensation, increased daytime frequency, nocturia, and urgency incontinence, whereas voiding symptoms consists of hesitancy, intermittency, weak or irregular stream, straining, and terminal dribble. Post-micturition symptoms include dribbling and sensation of incomplete voiding. The storage symptoms are generally more bothersome than voiding or post-micturition symptoms, as observed in both men and women. Overactive bladder (OAB) syndrome is a subgroup of storage symptoms consisting mainly of urinary urgency. In men, LUTS typically occur in association with bladder outlet obstruction (BOO) secondary to benign prostatic hyperplasia (BPH), despite that it may occur independently of BOO or prostatic diseases, whereas in women, the most frequent LUTS is stress urinary incontinence [22]. Epidemiological studies have shown OAB to be a widely prevalent condition in men and women [5,23], with an incidence of 16.6% in a sample from Europe [24], 16.9% in women and 16.0% in men in a sample from the USA [25], and an overall prevalence of OAB of 18.9% in a South American population [26,27]. Epidemiological studies applying the ICS definition of OAB across multiple countries found a prevalence of 11–13% [5]. LUTS negatively impacts the social quality of life and sexual health of patients [28,29].

3. Association of Metabolic Syndrome and Diabetes with Urinary Bladder Dysfunction

Metabolic syndrome, the medical term for a combination of cardiometabolic risk factors such as central obesity, hyperglycemia, hypertension, and dyslipidemia, is critically involved in the onset of many cardiovascular diseases, being the leading cause of death worldwide [30]. Outside the cardiovascular system, metabolic syndrome associated with increased body mass index (BMI) represents an important risk factor for LUTS/OAB and urinary incontinence [31,32,33,34,35,36,37,38,39,40,41,42,43,44], despite some studies showing no positive association between metabolic syndrome and LUTS in men and women [45,46]. An association between metabolic syndrome and interstitial cystitis/bladder pain syndrome (IC/BPS) has been reported in women [47]. Elevated body mass index and diabetes also increase the risk of urinary tract infections and pyelonephritis [48,49]. Metabolic syndrome is also associated with LUTS secondary to benign prostatic hyperplasia [50,51,52]. Surgical and nonsurgical weight loss leads to improvements in stress urinary incontinence [53], even though a definite conclusion has not been achieved [54].

4. Bladder Dysfunction in Type 1 and Type 2 Diabetes in Patients and Animals

Diabetes mellitus is a chronic metabolic disease characterized by high blood sugar levels (hyperglycemia) as a result of abnormal insulin production and/or insulin function. The most common and bothersome urological complication of diabetes mellitus is DBD (or diabetic cystopathy), which affects more than 80% of individuals diagnosed with diabetes [55,56,57,58,59]. The pathophysiology of DBD is multifactorial and may involve alterations at all levels of the urinary tract, including the detrusor, urethra, urothelium, and innervation [60]. Clinical DBD manifestations consist of storage bladder problems such as OAB and urge incontinence, and voiding problems like poor emptying with resultant elevated post-void residual urine [7,12]. Increased capacity and decreased sensation together with recurrent urinary tract infections may also be present in DBD [11]. Preclinical models of type 1 (T1DM) and type 2 diabetes (T2DM) have provided further evidence confirming the relationship between metabolic diseases and bladder dysfunction [61,62,63,64,65,66,67].
T1DM can be mimicked by injection of streptozotocin (STZ) in rodents, a cytotoxic glucose analogue that destroys pancreatic β-cells due to its high affinity for glucose transporter 2 (GLUT2) [68,69]. Analysis of bladders from STZ-induced diabetes in male and female rodents (rats and mice) revealed increased bladder mass [70,71,72,73,74,75], which is suggested to represent a physical adaptation to increased urine production [76,77,78]. Despite that high glucose levels and diabetic polyuria have been proposed as pathophysiological mechanisms explaining bladder enlargement in the STZ model [79], a recent study comprising different models of diabetes in rodents, including T1DM, did not confirm such a proposition [80]. Insulin administration can prevent, or even reverse, most of the morphological, functional, and molecular bladder alterations in the STZ model [79,81,82,83]. Moreover, increases in both volume and frequency of micturition [70,73], as well as in urinary frequency, capacity, and number of nonvoiding contraction (NVCs) [66,84], have been reported in STZ-induced diabetes, as revealed by urodynamic studies. Spontaneous voiding spot assays [85,86] also revealed significant increases in voiding frequency, total voided volume, and mean volume per micturition in STZ-injected mice [87], which are paralleled by in vitro detrusor overactivity [66,88]. However, after prolonged hyperglycemia and insulin resistance in response to STZ, bladders may progress to an underactive detrusor and an inability to produce an effective voiding [64] through mechanisms mediated by the activation of NLRP-3 inflammasome [89]. Therefore, in STZ-induced diabetes, a temporal effect of diabetes on bladder activity has been established, that is, an early phase of compensatory followed by a later phase of decompensated bladder function [7,63,77,78]. In female Akita mice (T1DM model), diabetic bladder dysfunction also progresses from overactivity to underactivity [90]. At the molecular level, the impairment of the nitric oxide—soluble guanylate cyclase (sGC)—cyclic GMP signaling [82,91] and NLRP3 inflammasome activation in urothelial cells [89] have been proposed as a critical mechanism contributing to bladder dysfunction. Nevertheless, conflicting data on different parameters of bladder activity in animal models of STZ-induced diabetes models have been obtained, which may possibly rely on both animal species and strain used, in addition to the disease time course [77,78,92,93]. Experimental T1DM in rats and rabbits can also be induced using alloxan, a hydrophilic unstable compound that shares a structure similar to glucose [94]. Increases in bladder weight, detrusor smooth muscle cells, capacity, and urinary output, along with irregular bladder contractions, were observed in alloxan-induced diabetic rats [95,96,97,98]. In rabbits made diabetic by alloxan, an increase in bladder weight [99] and a reduction in the vitro bladder contractions to carbachol were reported [100].
The main T2DM models that result in hyperinsulinemia and insulin resistance rely on allowing animals free access to diets highly enriched in fats [101,102]. In addition to producing the classical obesity-associated vascular dysfunction, male mice fed high-fat diets progress to an overactive bladder phenotype, as evidenced mainly by filling cystometry in anesthetized and awake rats and mice [103,104,105]. The resulting increased body weight, hyperglycemia, and insulin resistance by prolonged high-fat diet intake in mice is also accompanied by in vitro bladder overactivity as a consequence of high extracellular calcium influx through L-type voltage-operated calcium [106,107,108,109]. The importance of calcium channels to bladder dysfunction has also been confirmed in diabetic db/db mice [110]. High-fat diet-fed obese mice also display impaired urethral smooth muscle relaxations [111,112] and prostate hypercontractility [105,113], which are suggested to contribute to the resulting bladder overactivity. Impaired striated urethral muscle contractions were reported in Zucker obese rats [114]. Contrasting to these studies, no evidence of bladder dysfunction was observed in obese mice fed a high-fat diet for 16 weeks, as assessed by void spot assays [115]. The temporal effects (up to 42 weeks) of different diets consisting of fructose, cholesterol, and lard, at varying proportions and combinations, on 24 h urinary behavior and conscious cystometry were investigated in rats [116]. Compared with the control group, the total voided volume was lower in all experimental diets, and animals receiving 32.5% lard diet alone exhibited decreases in bladder capacity, mean voided volume, and inter-micturition intervals that were indicative of an overactive bladder phenotype [116].
Leptin is a satiety hormone that is synthesized by adipocytes, the levels of which increase with the adipose tissue mass [117]. Mice genetically deficient in leptin (ob/ob) or in the leptin receptor (db/db) are hyperphagic, obese, hyperinsulinemic, and hyperglycemic, and have been widely used as a T2DM model [118]. Similarly, in the STZ- or diet-induced obesity models, ob/ob male mice exhibit bladder dysfunction characterized by increases in urine volume and in vitro bladder smooth muscle contractions [119]. Increases in total void volume and volume per void with no alterations of spot number were observed in five-week-old male and female ob/ob mice, as evaluated by void spot assays [16]. Four- and six-month-old ob/ob mice exhibited some degree of bladder dysfunction such as increases in total urine volume and number of primary void spots, although that depended on animal sex and animal age [115]. In male db/db mice, increases in bladder weight, voiding frequency, and capacity together with elevated in vitro contractions were described [110]. Increases in detrusor smooth muscle area, urothelium area, and collagen content were also reported in male and female db/db mice at 12, 24, or 52 weeks of age, which was suggested to reflect a compensatory response to the increased urine output [120]. Double-knockout hepatic-specific insulin receptor substrate 1 and 2 (IRS1 and IRS2) female mice that develop T2DM exhibit bladder overactivity, high frequency of nonvoiding contractions, decreased voided volume, and dispersed urine spot patterns [121].

5. Methylglyoxal–Advanced Glycation End Products (AGEs)–RAGE Signaling Pathway

The abnormal accumulation of highly reactive dicarbonyl compounds as a consequence of glycolytic overload has been referred to as dicarbonyl stress [1,3]. 1,2-Dicarbonyl compounds include glucosone, 3-deoxyglucosone, methylglyoxal (MGO), and glyoxal, but MGO is one of the most studied, given that it exerts a critical role in diabetes-associated cardiovascular complications, such as diabetic nephropathy, endothelial dysfunction, postinfarct remodeling, and impairment of insulin signaling [122,123,124,125,126]. Methylglyoxal, chemically referred as acetylformaldehyde, 2-ketopropionaldehyde, pyruvaldehyde, or 2-oxo-propanal, is a highly reactive dicarbonyl compound formed endogenously from 3-carbon glycolytic intermediates of glycolysis (dihydroxyacetone phosphate and glyceraldehyde-3-phosphate), although it can also be generated as a byproduct of protein, lipid, and ketones [127,128]. In addition to the endogenous production in mammalian cells, MGO may be present at marked levels in many food products and beverages, as well as in microorganisms [129]. In healthy conditions, glyoxalases (Glo) are the most important enzymatic detoxification system that converts MGO into its end-product D-lactate [1]. Glyoxalases comprise two major enzymes, namely, Glo1 (lactoylglutathione methylglyoxal lyase) and Glo2 (hydroxyacylglutathione hydrolase), with Glo1 described as a rate-limiting enzyme [130,131,132]. Interestingly, the increased levels of glucose and MGO are normalized in Glo-1 transgenic rats after induction of diabetes by intravenous injection of STZ [123,133].
The endogenous process by which endogenous MGO promotes post-translational modification of peptides or proteins, ultimately leading to generation of AGEs, is referred to as glycation [2]. The main MGO-derived AGEs in mammalian metabolism are arginine-derived hydroimidazolone (MG-H1) and carboxyethyl-lysine (CEL) [134], but other AGEs may be generated depending on the dicarbonyl species formed [135]. The mechanism of MG-H1 generation involves the replacement of the hydrophilic positively charged arginine residue by an uncharged, hydrophobic MG-H1 residue, producing misfolding and activation of the unfolded protein response [1,136]. Incubation of human plasma from heathy donors with different concentrations of MGO (10 and 100 µM) for 24 h induced a time- and dose-dependent increase in MG-H1 levels, as detected within the first 6 h [137].
Once generated, AGEs bind their cell membrane-anchored ligand receptor (termed RAGE), which is a member of the immunoglobulin superfamily of cell surface receptors able to recognize endogenous ligands [4]. Structurally, RAGE consists of three immunoglobulin domains, that is, (i) an extracellular part consisting of one V type and two C types (C1 and C2), (ii) a transmembrane spanning helix, and (iii) a short, highly charged intracellular cytoplasmic “C” terminal tail that is primarily associated with the downstream signaling pathways [138]. The extracellular domain devoid of cytoplasmic and transmembrane domains is called soluble RAGE (sRAGE), which comprise two forms, namely, cleaved RAGE (cRAGE) and endogenous secretory RAGE (esRAGE or RAGEv1) [139]. cRAGE is generated at the cell surface by the proteolytic cleavage of RAGE at the boundary between its extracellular and transmembrane portions, whereas esRAGE results from alternative splicing of RAGE pre-mRNA. In advanced chronic kidney disease (CKD) patients, an inverse association between risk of mortality and cRAGE/esRAGE ratio was reported [140]. RAGE plays an important role in the innate immune response and as a mediator of proinflammatory processes, triggering multiple intracellular signaling pathways, including the generation of proinflammatory mediators such as IL-1β, VCAM-1, and TNF-α via the transcription factor NF-κB [138] and phosphorylation of JNK and p38MAPK [141]. Furthermore, many of the RAGE actions have been attributed to the activation of NADPH oxidase [142], which leads to excess formation of ROS, thus contributing to generate a pro-oxidant environment [143,144,145]. Figure 1 illustrates the MGO–AGEs–RAGE signaling and glyoxalase system (Glo1 and Glo2 enzymes).
Plasma levels of MGO are markedly elevated in conditions of hyperglycemia associated with diabetes mellitus in men and women, as usually detected by liquid chromatography–mass spectrometry, enzyme-linked immunosorbent, or electrochemical biosensor assays [146,147,148,149,150,151,152,153]. The patients included in these studies comprised men and women aged 54–61 years old, insulin- and noninsulin users, with accompanying diseases such as chronic renal failure, diabetic nephropathy, and coronary heart disease. Obese patients also have increased MGO levels in plasma, which can be even higher if the obese patients have diabetes [125,154]. The urine levels of MGO in diabetic patients are also higher than those in nondiabetic individuals [155]. In addition, in healthy volunteers, a rapid increase (49 min) in plasma levels of MGO was observed after oral glucose tolerance test (OGTT) [152]. Likewise, fasted healthy mice intraperitoneally injected with a glucose solution displayed a rapid elevation in plasma levels of MGO, as detected at 30 min after glucose administration [137]. Interestingly, lower levels of Glo1 activity in red blood cells paralleled the increased plasma MGO levels in T2DM patients displaying acute coronary syndrome [156].

6. MGO–AGEs–RAGE Axis as a Key Player of Bladder Dysfunction in Animals and Humans

There is a large amount of data on MGO in different organs at physiological and pathological conditions [2], but surprisingly few studies have explored the role of the MGO–AGEs–RAGE signaling pathway in the pathophysiology of the lower urinary tract system. The existing literature in this field has been restricted to bladder pain via the release of high mobility group box 1 protein (HMGB1) [157,158] and bladder cancer [159,160], which are not the focus of the present review.
In T2DM patients diagnosed with moderate/severe LUTS, serum levels of AGEs are positively correlated with symptoms and overactive bladder, suggesting that levels of AGEs may be early markers of diabetes-associated LUTS [13]. In addition, an immunohistochemical study in human bladders showed positive sites for carboxymethyl-lysine and pentosidine in the connective tissue between muscle bundles and muscle fibers, suggesting that extracellular matrix is the main site of action for AGE accumulation [161]. The MG-H1 free adduct has been described as the most responsive AGE associated with chronic kidney disease status, with higher levels in diabetic compared with nondiabetic individuals [162]. The MG-H1 residue contents of plasma protein are also elevated in male spontaneously diabetic Torii (SDT) rats at the age of 16 weeks [163].
The model of chronic overload intake of MGO at doses of 50 to 75 mg/kg for 6 to 12 weeks, as supplemented in the drinking water of the animals or injected intraperitoneally in rats and mice, has been shown to mimic some cardiovascular complications of diabetes in the absence of hyperglycemia such as endothelial dysfunction, microvascular damage, atherogenesis [164,165,166,167], cardiac dysfunction [168,169], and renal damage [170,171,172] (Table 1). However, the direct contribution of MGO to bladder dysfunction remains poorly investigated. This model of exogenous animal supplementation with MGO clearly differs from the classical diabetic animals in that MGO is not generated from the endogenous glucose metabolism, and, consequently, does not itself affect the glucose levels and insulin sensitivity [164,173,174,175]. Intake of MGO to healthy mice for 7 days (500 to 2000 mg/kg) significantly increased the urine levels of this dicarbonyl molecule [176]. Serum levels achieved by a 12-week intake of 0.5% MGO to healthy mice [174,175,177] reached comparable levels to those found in plasma of diabetic/obese individuals [125,154]. Levels of MGO levels were also increased in both plasma and urine after a 6-month MGO administration to mice at the doses of 200 mg/kg [176] and 500 mg/kg [178]. Likewise, in high-fat fed mice, levels of plasma and urine levels of MGO were significantly higher than animals kept on low-fat diet [179]. Diabetic obese ob/ob mice also displayed elevated serum MGO compared with normoglycemic animals [16] (Table 1).
Bladders from male mice treated orally with MGO for 4 weeks revealed tissue disorganization, partial loss of the urothelium, and mucosal edema along with marked cell infiltration [14]. Urodynamic evaluation (cystometric assays) in these male animals showed marked increases in micturition frequency and number of nonvoiding contractions (NVCs) with no alterations in bladder capacity [14]. Cystometric assays in male mice treated orally with MGO for an extended period (12 weeks) showed significant increases in the frequency of NVCs, bladder capacity, inter-micturition pressure, and residual volume [177]. In female mice treated with MGO for 12 weeks, cystometric assays confirmed urodynamic alterations such as increases in NVCs frequency, bladder capacity, inter-micturition pressure, and residual volume [15]. Using the model of spontaneous void spot assay (VSA) on filter paper, male mice treated with MGO for 12 weeks revealed an increased volume per void with no changes in the spot number as compared with the untreated group. In the female group, this treatment increased the spot number (mainly the number of microvolume spots) but, rather, reduced the volume per void [177,180]. During the MGO treatment, no alterations in the water consumption are observed in any group [177]. In this VSA assay, the term thigmotaxis refers to the wall-seeking behavior, that is, the tendency of mice to urinate next to the walls of the cage, which is interpreted as a rational response related to the fear of predation [181,182]. In healthy conditions, mice of both sexes will urinate at the corner of the filter paper, and when the animal loses this outlet control, urinating in the center of the filter, this may indicate bladder dysfunction. In the 12-week MGO treatment, whist the male mice had 95% of the voided spots in the corners of the filter paper, the voided spots in the female group were also detected in the center of the filter, indicating an altered outlet behavior in favor of an overactive bladder phenotype. The in vitro contractions to electrical-field stimulation (EFS; neurogenic contractions) as well as those induced by selective muscarinic and purinergic P2X1 receptor activation (using carbachol and α, β-methylene ATP as receptor agonists, respectively) were also evaluated in bladders of male and female mice treated with MGO for 12 weeks (Table 2). In intact bladder preparations of male mice, higher contractions to EFS, carbachol, and α,β-methylene ATP were observed after MGO treatment [177]. In the female group, higher contractile responses to EFS and α,β-methylene ATP (but not to carbachol) were also observed in intact bladder preparations from animals treated with MGO for 12 weeks [15]. An increased carbachol-induce response by MGO treatment in the female mice is solely observed when the urothelium is removed from the preparations. Moreover, the higher EFS-induced contractions in the MGO group were normalized by prior tissue incubation with the selective TRPA1 blocker HC-030031, suggesting that MGO exposure via TRPA1 activation leads to enhancement of purinergic over cholinergic neurotransmission in the bladder [180] (Figure 2). Table 2 summarizes the main in vivo and in vitro bladder alterations observed in male and female mice treated with MGO for 12 weeks.
In MGO-treated mice, elevated levels of AGEs and RAGE in bladder tissues were also observed [15,177]. Likewise, hyperglycemic diabetic leptin-deficient male and female mice (ob/ob) exhibit bladder dysfunction, as evidenced by the increases in total void volume and volume per void (void spot assay) in addition to high collagen content in the bladders [16]. These bladder alterations were associated with high levels of total AGEs, MG-H1 and RAGE found in bladder tissues, which is consistent with the findings that the AGE breaker alagebrium (ALT-711) at 1 mg/kg during 8 weeks in the drinking water nearly reversed all the molecular and functional alterations in ob/ob mice [16] (Table 3).

7. Drugs Presenting Potential to Downregulate AGEs Formation and Oxidative Stress in Bladder Tissues

It is well established that NADPH oxidase and increased levels of superoxide anion (O2) and hydrogen peroxide (H2O2) play a critical role in diabetic complications [55,183,184,185,186,187,188,189,190,191,192]. Oxidative stress at excessive levels also plays an important role in pathophysiology of bladder outlet obstruction [193], cyclophosphamide-induced cystitis [194], benign prostatic hyperplasia [195] and STZ-induced bladder dysfunction [196]. Hydrogen peroxide (H2O2) is reported to activate bladder afferent signaling inducing detrusor overactivity [197]. In human and dog bladders in vitro, H2O2 itself produced contractions and potentiated the contractions induced by electrical-field stimulation, an effect attenuated by the natural NADPH oxidase inhibitor apocynin [198]. Given obesity-associated bladder dysfunction correlates with increased oxidative stress and that MGO treatment leads to excess ROS production, it is plausible that drugs that inactivate MGO [20,199] or that protect bladder cells from the oxidative insult [200,201] offer an interesting approach to reduce the deleterious effects of AGEs in the bladder (Figure 3). Therefore, we summarized below some drugs reported to ameliorate bladder dysfunction in animals including some polyphenols and metformin whose protective mechanisms may be related to their ability to downregulate AGEs formation and oxidative stress in bladder tissues.

7.1. Polyphenols: Resveratrol and Epigallocatechin-3-Gallate

Resveratrol is a polyphenol present in numerous plant-based foods that increases lipolysis and reduces lipogenesis in adipocytes, being suggested as a therapeutic alternative to treat obesity-related diseases [202,203]. Two-week therapy with resveratrol (100 mg/kg/day, given by gavage) in high-fat-diet-fed obese mice reduced the in vivo urodynamic changes, the in vitro bladder overactivity, and the ROS production in bladder tissues [104]. Resveratrol treatment also increased the nitric oxide levels and restored the impaired urethral relaxations in obese mice, an effect mimicked by the antioxidant enzyme SOD [112]. Likewise, the in vitro urethral hyperactivity was restored by resveratrol in obese mice [112]. In the bladders of STZ-induced diabetic rats, daily oral treatment with resveratrol (10 mg/kg) reduced the histological abnormalities and inhibited the expression and localization of markers of oxidative stress and DNA oxidative damage [204]. Intragastric administration of resveratrol (20 mg/kg/day) reduced bladder hypertrophy, tissue damage, inflammatory cell infiltration, and levels of inflammatory cytokines in the bladders of STZ-induced diabetic rats [205]. In the chronic prostatitis model in rats, oral administration of resveratrol (10 mg/kg) for 10 days reduced the resulting overactive bladder and fibrosis by reducing the protein expressions SCF, c-Kit, and p-AKT [206,207]. At the molecular level, resveratrol exhibited a high inhibition rate on the fluorescent formation of AGEs mainly due to scavenging free radicals and capturing MGO [20]. Epigallocatechin-3-gallate is another polyphenol compound present in green tea that has also favorable effects on bladder overactivity, as evidenced in ovariectomized rats fed standard chow [201] and high-fat, high-sugar diet [208]. Treatment with epigallocatechin-3-gallate reduced the expressions of transforming growth factor-β (TGF-β) and type I collagen, as well as the apoptosis and oxidative stress in the bladders [208,209]. In a bladder outlet obstruction (BOO) model in rats, intraperitoneal injection of epigallocatechin-3-gallate (4.5 mg/kg/day) reduced the histologic changes and submucosal endoplasmic reticulum (ER) stress-related apoptosis, recovering the bladder compliance and inter-contractile intervals [210]. At 6 mM, epigallocatechin-3-gallate was also shown to exert anti-AGEs activity through its capacity to strongly trap and inactivate MGO [211]. In diabetic db/db mice, 16-week oral administration of (+)-catechin (15, 30, and 60 mg/kg) directly trapped MGO, hence downregulating the downstream signal transduction and inflammatory response induced by AGE–RAGE interaction in the kidney [212]. Therefore, although the mechanisms behind the uroprotective actions of polyphenols on diabetes-associated bladder dysfunction deserve further investigation, they could involve the capacity of these molecules to directly trap MGO, further inhibiting MGO-induced glycation, AGEs formation, and RAGE activation.

7.2. Metformin

Metformin is a first-line pharmacological treatment for T2DM patients as monotherapy or in combination with sulfonylureas or dipeptidyl peptidase 4 inhibitors [213]. The orally administered doses of metformin (as immediate-release or extended-release formulations) usually vary from 0.5 to 2.5 g daily, being safety and effective for long-term glycemic control. Metformin is associated with low risk of hypoglycemia and documented cardiovascular benefits [214]. Metformin increases tissue sensitivity to insulin and decreases the levels of glycated hemoglobin by mechanisms involving the activation of adenosine monophosphate-activated protein kinase (AMPK) and non-AMPK pathways [215], but its exact mechanism of action remains largely incomplete [213]. Recently, metformin was shown to increase intestinal glucose uptake, influencing hepatic glucose production through a gut–liver crosstalk [216]. Metformin is among the molecules reported to strongly react with MGO [17], forming an imidazolinone metabolite [19]. In addition, in the plasma of T2DM patients, metformin, through its guanidine group, was shown to bind to MGO, reducing this dicarbonyl concentration [18], hence reducing AGEs formation, which paralleled a significant increase in Glo1 activity [217]. A two-week treatment of high-fat-diet-fed mice with metformin (300 mg/kg) reversed the bladder overactivity, as evidenced by in vivo and in vitro studies [106]. Metformin also normalized the enhanced serum levels of MGO and fluorescent AGEs in mice treated chronically with MGO [177]. In bladders of MGO-treated mice, metformin treatment reduced Glo1 expression and activity, urothelium thickness, and collagen content, as well as the in vitro and in vivo micturition dysfunction [177]. It is, therefore, plausible to suggest that the beneficial effects of metformin in obesity-associated bladder dysfunction rely at least in part on its MGO capturing property. Of interest, oral administration of metformin (150 mg/kg, gavage) reduced both bladder remodeling and dysfunction in models of partial bladder outlet obstruction in rats [218], erectile dysfunction in obese mice [219], and diabetic nephropathy in STZ-induced diabetes [220].

8. Concluding Remarks and Future Therapeutics

Diabetic bladder dysfunction is a highly prevalent condition manifesting as storage (such as OAB and urge incontinence) and voiding problems (poor emptying with resultant elevated capacity). Increased capacity and decreased sensation together with recurrent urinary tract infections may also be present in DBD. Preclinical models of T1DM and T2DM in rodents have provided further evidence confirming the relationship between diabetes and bladder dysfunction. Hyperglycemia in diabetic/obese patients significantly elevates the levels of α-dicarbonyl compounds, including MGO, in plasma and urine as a consequence of the glycolytic overload. MGO promotes post-translational modification of peptides and proteins, ultimately leading to the formation of AGEs such as MG-H1. AGEs bind their cell membrane-anchored ligand receptor RAGE, triggering multiple intracellular signaling pathways, among which ROS production at excessive levels plays a critical role. However, little is known about the importance of MGO generation and AGEs–RAGE activation in the pathophysiology of diabetic-associated bladder dysfunction. Voiding spot assays and cystometrical evaluation in mice treated chronically with MGO have revealed significant increases in total void volume, volume per void, micturition frequency, and nonvoiding contractions number, along with enhanced in vitro bladder contractility. Moreover, levels of MGO, AGEs, RAGE, and ROS are all elevated in the bladder tissues obtained from MGO-treated animals and diabetic ob/ob mice. The antihyperglycemic drug metformin and the polyphenols resveratrol and epigallocatechin-3-gallate can directly scavenger MGO, exerting uroprotective actions. Therefore, we propose here that evaluation of MGO, AGEs, and RAGE levels may constitute important biomarkers of DBD pathophysiology. The design and development of new drugs that inhibit the MGO–AGEs–RAGE axis may become an interesting approach for the prevention and treatment of bladder dysfunction in diabetic conditions.

Author Contributions

Conceptualization and original draft preparation, E.A.; review and editing, A.L.O., M.G.d.O., F.Z.M. and E.A.; supervision, E.A.; funding acquisition, F.Z.M. and E.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by “Fundação de Amparo à Pesquisa do Estado de São Paulo” (FAPESP), grant number 2017/15175-1.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Rabbani, N.; Xue, M.; Thornalley, P.J. Dicarbonyl stress, protein glycation and the unfolded protein response. Glycoconj. J. 2021, 38, 331–340. [Google Scholar] [CrossRef] [PubMed]
  2. Schalkwijk, C.G.; Stehouwer, C.D.A. Methylglyoxal, a Highly Reactive Dicarbonyl Compound, in Diabetes, Its Vascular Complications, and Other Age-Related Diseases. Physiol. Rev. 2020, 100, 407–461. [Google Scholar] [CrossRef]
  3. Stratmann, B. Dicarbonyl Stress in Diabetic Vascular Disease. Int. J. Mol. Sci. 2022, 23, 6186. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, H.J.; Jeong, M.S.; Jang, S.B. Molecular characteristics of RAGE and advances in small-molecule inhibitors. Int. J. Mol. Sci. 2021, 22, 6904. [Google Scholar] [CrossRef]
  5. Abrams, P.; Cardozo, L.; Fall, M.; Griffiths, D.; Rosier, P.; Ulmsten, U.; Van Kerrebroeck, P.; Victor, A.; Wein, A.; Standardisation Sub-Committee of the International Continence Society. The standardisation of terminology in lower urinary tract function: Report from the standardisation sub-committee of the International Continence Society. Urology 2003, 61, 37–49. [Google Scholar] [CrossRef] [PubMed]
  6. D’Ancona, C.; Haylen, B.; Oelke, M.; Abranches-Monteiro, L.; Arnold, E.; Goldman, H.; Hamid, R.; Homma, Y.; Marcelissen, T.; Rademakers, K.; et al. Standardisation Steering Committee ICS and the ICS Working Group on Terminology for Male Lower Urinary Tract & Pelvic Floor Symptoms and Dysfunction. The International Continence Society (ICS) report on the terminology for adult male lower urinary tract and pelvic floor symptoms and dysfunction. Neurourol. Urodyn. 2019, 38, 433–477. [Google Scholar] [CrossRef] [PubMed]
  7. Liu, G.; Daleeneshgari, F. Diabetic bladder dysfunction. Chin. Med. J. 2014, 127, 1357–1364. [Google Scholar] [PubMed]
  8. Chapple, C.R.; Osman, N.I.; Birder, L.; van Koeveringe, G.A.; Oelke, M.; Nitti, V.W.; Drake, M.J.; Yamaguchi, O.; Abrams, P.; Smith, P.P. The underactive bladder: A new clinical concept? Eur. Urol. 2015, 68, 351–353. [Google Scholar] [CrossRef]
  9. Cohn, J.A.; Brown, E.T.; Kaufman, M.R.; Dmochowski, R.R.; Reynolds, W.S. Underactive bladder in women: Is there any evidence? Curr. Opin. Urol. 2016, 26, 309–314. [Google Scholar] [CrossRef]
  10. Wang, J.; Ren, L.; Liu, X.; Liu, J.; Ling, Q. Underactive bladder and detrusor underactivity: New advances and prospectives. Int. J. Mol. Sci. 2023, 24, 15517. [Google Scholar] [CrossRef]
  11. Yoshimura, N.; Chancellor, M.B.; Andersson, K.E.; Christ, G.J. Recent advances in understanding the biology of diabetes-associated bladder complications and novel therapy. BJU Int. 2005, 95, 733–738. [Google Scholar] [CrossRef] [PubMed]
  12. Daneshgari, F.; Liu, G.; Hanna-Mitchell, A.T. Path of translational discovery of urological complications of obesity and diabetes. Am. J. Physiol. Renal Physiol. 2017, 312, F887–F896. [Google Scholar] [CrossRef] [PubMed]
  13. Galì, A.; Mucciardi, G.; Butticè, S.; Subba, E.; D’Amico, C.; Lembo, F.; Magno, C. Correlation between advanced glycation end-products, lower urinary tract symptoms and bladder dysfunctions in patients with type 2 diabetes mellitus. Low. Urin. Tract. Symptoms 2017, 9, 15–20. [Google Scholar] [CrossRef] [PubMed]
  14. De Oliveira, M.G.; Medeiros, M.L.; Tavares, E.B.G.; Mónica, F.Z.; Antunes, E. Methylglyoxal, a reactive glucose metabolite, induces bladder overactivity in addition to inflammation in mice. Front. Physiol. 2020, 11, 290. [Google Scholar] [CrossRef] [PubMed]
  15. Oliveira, A.L.; Medeiros, M.L.; de Oliveira, M.G.; Teixeira, C.J.; Mónica, F.Z.; Antunes, E. Enhanced RAGE expression and excess reactive-oxygen species production mediates Rho Kinase-dependent detrusor overactivity after methylglyoxal exposure. Front. Physiol. 2022, 13, 860342. [Google Scholar] [CrossRef] [PubMed]
  16. Oliveira, A.L.; Medeiros, M.L.; Ghezzi, A.C.; Dos Santos, G.A.; Mello, G.C.; Mónica, F.Z.; Antunes, E. Evidence that methylglyoxal and receptor for advanced glycation end products are implicated in bladder dysfunction of obese diabetic ob/ob mice. Am. J. Physiol. Renal Physiol. 2023, 325, F436–F447. [Google Scholar] [CrossRef] [PubMed]
  17. Ruggiero-Lopez, D.; Lecomte, M.; Moinet, G.; Patereau, G.; Lagarde, M.; Wiernsperger, N. Reaction of metformin with dicarbonyl compounds. Possible implication in the inhibition of advanced glycation end product formation. Biochem. Pharmacol. 1999, 58, 1765–1773. [Google Scholar] [CrossRef] [PubMed]
  18. Beisswenger, P.J.; Howell, S.K.; Touchette, A.D.; Lal, S.; Szwergold, B.S. Metformin reduces systemic methylglyoxal levels in type 2 diabetes. Diabetes 1999, 48, 198–202. [Google Scholar] [CrossRef] [PubMed]
  19. Kinsky, O.R.; Hargraves, T.L.; Anumol, T.; Jacobsen, N.E.; Dai, J.; Snyder, S.A.; Monks, T.J.; Lau, S.S. Metformin scavenges methylglyoxal to form a novel imidazolinone metabolite in humans. Chem. Res. Toxicol. 2016, 29, 227–234. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, H.; Huo, X.; Wang, S.; Yin, Z. The inhibitory effects of natural antioxidants on protein glycation as well as aggregation induced by methylglyoxal and underlying mechanisms. Colloids Surf. B Biointerfaces 2022, 212, 112360. [Google Scholar] [CrossRef]
  21. Roy, H.A.; Green, A.L. The central autonomic network and regulation of bladder function. Front. Neurosci. 2019, 13, 535. [Google Scholar] [CrossRef] [PubMed]
  22. Cox, L.; Rovner, E.S. Lower urinary tract symptoms in women: Epidemiology, diagnosis, and management. Curr. Opin. Urol. 2016, 26, 328–333. [Google Scholar] [CrossRef] [PubMed]
  23. Irwin, D.E.; Milsom, I.; Hunskaar, S.; Reilly, K.; Kopp, Z.; Herschorn, S.; Coyne, K.; Kelleher, C.; Hampel, C.; Artibani, W.; et al. Population-based survey of urinary incontinence, overactive bladder, and other lower urinary tract symptoms in five countries: Results of the EPIC study. Eur. Urol. 2006, 50, 1306–1315. [Google Scholar] [CrossRef] [PubMed]
  24. Milsom, I.; Abrams, P.; Cardozo, L.; Roberts, R.G.; Thüroff, J.; Wein, A.J. How widespread are the symptoms of an overactive bladder and how are they managed? A population-based prevalence study. BJU Int. 2001, 87, 760–766. [Google Scholar] [CrossRef] [PubMed]
  25. Stewart, W.F.; Van Rooyen, J.B.; Cundiff, G.W.; Abrams, P.; Herzog, A.R.; Corey, R.; Hunt, T.L.; Wein, A.J. Prevalence and burden of overactive bladder in the United States. World J. Urol. 2003, 20, 327–336. [Google Scholar] [CrossRef]
  26. Teloken, C.; Caraver, F.; Weber, F.A.; Teloken, P.E.; Moraes, J.F.; Sogari, P.R.; Graziottin, T.M. Overactive bladder: Prevalence and implications in Brazil. Eur. Urol. 2006, 49, 1087–1092. [Google Scholar] [CrossRef] [PubMed]
  27. Moreira, E.D., Jr.; Neves, R.C.; Neto, A.F.; Duarte, F.G.; Moreira, T.L.; Lobo, C.F.; Glasser, D.B. A population-based survey of lower urinary tract symptoms (LUTS) and symptom-specific bother: Results from the Brazilian LUTS epidemiology study (BLUES). World J. Urol. 2013, 31, 1451–1458. [Google Scholar] [CrossRef]
  28. Coyne, K.S.; Kvasz, M.; Ireland, A.M.; Milsom, I.; Kopp, Z.S.; Chapple, C.R. Urinary incontinence and its relationship to mental health and health-related quality of life in men and women in Sweden, the United Kingdom, and the United States. Eur. Urol. 2012, 61, 88–95. [Google Scholar] [CrossRef] [PubMed]
  29. Gomes, C.M.; Averbeck, M.A.; Koyama, M.; Soler, R. Impact of OAB symptoms on work, quality of life and treatment-seeking behavior in Brazil. Curr. Med. Res. Opin. 2020, 36, 1403–1415. [Google Scholar] [CrossRef] [PubMed]
  30. Jia, G.; Bai, H.; Mather, B.; Hill, M.A.; Jia, G.; Sowers, J.R. Diabetic vasculopathy: Molecular mechanisms and clinical insights. Int. J. Mol. Sci. 2024, 25, 804. [Google Scholar] [CrossRef] [PubMed]
  31. Rohrmann, S.; Smit, E.; Giovannucci, E.; Platz, E.A. Association between markers of the metabolic syndrome and lower urinary tract symptoms in the Third National Health and Nutrition Examination Survey (NHANES III). Int. J. Obes. 2005, 29, 310–316. [Google Scholar] [CrossRef] [PubMed]
  32. Hunskaar, S. A systematic review of overweight and obesity as risk factors and targets for clinical intervention for urinary incontinence in women. Neurourol. Urodyn. 2008, 27, 749–757. [Google Scholar] [CrossRef] [PubMed]
  33. Tai, H.C.; Chung, S.D.; Ho, C.H.; Tai, T.Y.; Yang, W.S.; Tseng, C.H.; Wu, H.P.; Yu, H.J. Metabolic syndrome components worsen lower urinary tract symptoms in women with type 2 diabetes. J. Clin. Endocrinol. Metab. 2010, 95, 1143–1150. [Google Scholar] [CrossRef] [PubMed]
  34. Uzun, H.; Zorba, O.U. Metabolic syndrome in female patients with overactive bladder. Urology 2012, 79, 72–75. [Google Scholar] [CrossRef] [PubMed]
  35. Bang, W.J.; Lee, J.Y.; Koo, K.C.; Hah, Y.S.; Lee, D.H.; Cho, K.S. Is type-2 diabetes mellitus associated with overactive bladder symptoms in men with lower urinary tract symptoms? Urology 2014, 84, 670–674. [Google Scholar] [CrossRef] [PubMed]
  36. Bunn, F.; Kirby, M.; Pinkney, E.; Cardozo, L.; Chapple, C.; Chester, K.; Cruz, F.; Haab, F.; Kelleher, C.; Milsom, I.; et al. Is there a link between overactive bladder and the metabolic syndrome in women? A systematic review of observational studies. Int. J. Clin. Pract. 2015, 69, 199–217. [Google Scholar] [CrossRef] [PubMed]
  37. Zacche, M.M.; Giarenis, I.; Thiagamoorthy, G.; Robinson, D.; Cardozo, L. Is there an association between aspects of the metabolic syndrome and overactive bladder? A prospective cohort study in women with lower urinary tract symptoms. Eur. J. Obstet. Gynecol. Reprod. Biol. 2017, 217, 1–5. [Google Scholar] [CrossRef] [PubMed]
  38. Lai, H.H.; Helmuth, M.E.; Smith, A.R.; Wiseman, J.B.; Gillespie, B.W.; Kirkali, Z. Symptoms of lower urinary tract dysfunction research network (LURN). relationship between central Obesity, general obesity, overactive bladder syndrome and urinary incontinence among male and female patients seeking care for their lower urinary tract symptoms. Urology 2019, 123, 34–43. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, J.K.; Lee, Y.G.; Han, K.; Han, J.H. Obesity, metabolic health, and urological disorders in adults: A nationwide population-based study. Sci. Rep. 2021, 11, 8687. [Google Scholar] [CrossRef] [PubMed]
  40. Pereira, T.A.; D’ancona, C.A.L.; Cândido, E.C.; Achermann, A.P.P.; Chaim, E.A. Prevalence of LUTS and urodynamics results in obese women. Neurourol. Urodyn. 2022, 41, 468–474. [Google Scholar] [CrossRef] [PubMed]
  41. Heo, J.E.; Kim, D.G.; Yoo, J.W.; Lee, K.S. Metabolic syndrome-related factors as possible targets for lower urinary tract symptoms in Korean males. Aging Male 2023, 26, 6–12. [Google Scholar] [CrossRef] [PubMed]
  42. Chilaka, C.; Toozs-Hobson, P.; Chilaka, V. Pelvic floor dysfunction and obesity. Best Pract. Res. Clin. Obstet. Gynaecol. 2023, 90, 102389. [Google Scholar] [CrossRef] [PubMed]
  43. Li, S.; Zou, J.; Wang, Z.; Wang, M.; Yuan, Y.; Lv, H. Correlation Between Insulin resistance and urinary incontinence in female patients with type 2 Diabetes Mellitus. Int. Urogynecol. J. 2024, 35, 431–440. [Google Scholar] [CrossRef] [PubMed]
  44. Alsannan, B.; Laganà, A.S.; Alhermi, J.; Almansoor, S.; Ayed, A.; Venezia, R.; Etrusco, A. Prevalence of overactive bladder among overweight and obese women: A prospective cross-sectional cohort study. Eur. J. Obstet. Gynecol. Reprod. Biol. 2024, 295, 59–64. [Google Scholar] [CrossRef] [PubMed]
  45. Park, H.K.; Lee, H.W.; Lee, K.S.; Byun, S.S.; Jeong, S.J.; Hong, S.K.; Lee, S.E.; Park, J.H.; Lee, S.B.; Kim, K.W.; et al. Relationship between lower urinary tract symptoms and metabolic syndrome in a community-based elderly population. Urology 2008, 72, 556. [Google Scholar] [CrossRef] [PubMed]
  46. Eom, C.S.; Park, J.H.; Cho, B.L.; Choi, H.C.; Oh, M.J.; Kwon, H.T. Metabolic syndrome and accompanying hyperinsulinemia have favorable effects on lower urinary tract symptoms in a generally healthy screened population. J. Urol. 2011, 186, 175–179. [Google Scholar] [CrossRef] [PubMed]
  47. Peng, L.; Di, X.P.; He, S.X.; Zeng, X.; Shen, H.; Zhu, H.L.; Luo, D.Y. Metabolic syndrome in women with and without interstitial cystitis/bladder pain syndrome. Int. Urogynecol. J. 2021, 32, 1299–1306. [Google Scholar] [CrossRef] [PubMed]
  48. Pugliese, G.; Liccardi, A.; Graziadio, C.; Barrea, L.; Muscogiuri, G.; Colao, A. Obesity and infectious diseases: Pathophysiology and epidemiology of a double pandemic condition. Int. J. Obes. 2022, 46, 449–465. [Google Scholar] [CrossRef]
  49. Pari, B.; Gallucci, M.; Ghigo, A.; Brizzi, M.F. Insight on Infections in diabetic setting. Biomedicines 2023, 11, 971. [Google Scholar] [CrossRef] [PubMed]
  50. Kirby, M.G.; Wagg, A.; Cardozo, L.; Chapple, C.; Castro-Diaz, D.; de Ridder, D.; Espuna-Pons, M.; Haab, F.; Kelleher, C.; Kölbl, H.; et al. Overactive bladder: Is there a link to the metabolic syndrome in men? Neurourol. Urodyn. 2010, 29, 1360–1364. [Google Scholar] [CrossRef] [PubMed]
  51. Calogero, A.E.; Burgio, G.; Condorelli, R.A.; Cannarella, R.; La Vignera, S. Epidemiology and risk factors of lower urinary tract symptoms/benign prostatic hyperplasia and erectile dysfunction. Aging Male 2019, 22, 12–19. [Google Scholar] [CrossRef] [PubMed]
  52. Omran, A.; Leca, B.M.; Oštarijaš, E.; Graham, N.; Da Silva, A.S.; Zaïr, Z.M.; Miras, A.D.; le Roux, C.W.; Vincent, R.P.; Cardozo, L.; et al. Metabolic syndrome is associated with prostate enlargement: A systematic review, meta-analysis, and meta-regression on patients with lower urinary tract symptom factors. Ther. Adv. Endocrinol. Metab. 2021, 12, 20420188211066210. [Google Scholar] [CrossRef] [PubMed]
  53. Subak, L.L.; Richter, H.E.; Hunskaar, S. Obesity and urinary incontinence: Epidemiology and clinical research update. J. Urol. 2009, 182, S2–S7. [Google Scholar] [CrossRef]
  54. Bauer, S.R.; Harrison, S.L.; Cawthon, P.M.; Senders, A.; Kenfield, S.A.; Suskind, A.M.; McCulloch, C.E.; Covinsky, K.; Marshall, L.M. Longitudinal changes in adiposity and lower urinary tract symptoms among older men. J. Gerontol. Ser. A 2022, 77, 2102–2109. [Google Scholar] [CrossRef] [PubMed]
  55. Daneshgari, F.; Liu, G.; Birder, L.; Hanna-Mitchell, A.T.; Chacko, S. Diabetic bladder dysfunction: Current translational knowledge. J. Urol. 2009, 182, S18–S26. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, C.C.; Chancellor, M.B.; Lin, J.M.; Hsieh, J.H.; Yu, H.J. Type 2 diabetes but not metabolic syndrome is associated with an increased risk of lower urinary tract symptoms and erectile dysfunction in men aged <45 years. BJU Int. 2010, 105, 1136–1140. [Google Scholar] [CrossRef] [PubMed]
  57. Moul, S.; McVary, K.T. Lower urinary tract symptoms, obesity and the metabolic syndrome. Curr. Opin. Urol. 2010, 20, 7–12. [Google Scholar] [CrossRef] [PubMed]
  58. Arrellano-Valdez, F.; Urrutia-Osorio, M.; Arroyo, C.; Soto-Vega, E. A comprehensive review of urologic complications in patients with diabetes. Springerplus 2014, 3, 549. [Google Scholar] [CrossRef] [PubMed]
  59. Blair, Y.; Wessells, H.; Pop-Busui, R.; Ang, L.; Sarma, A.V. Urologic complications in diabetes. J. Diabetes Complicat. 2022, 36, 108288. [Google Scholar] [CrossRef] [PubMed]
  60. Yuan, Z.; Tang, Z.; He, C.; Tang, W. Diabetic cystopathy: A review. J. Diabetes 2015, 7, 442–447. [Google Scholar] [CrossRef] [PubMed]
  61. Gasbarro, G.; Lin, D.L.; Vurbic, D.; Quisno, A.; Kinley, B.; Daneshgari, F.; Damaser, M.S. Voiding function in obese and type 2 diabetic female rats. Am. J. Physiol. Renal Physiol. 2010, 298, F72–F77. [Google Scholar] [CrossRef] [PubMed]
  62. Fry, C.H.; Daneshgari, F.; Thor, K.; Drake, M.; Eccles, R.; Kanai, A.J.; Birder, L.A. Animal models and their use in understanding lower urinary tract dysfunction. Neurourol. Urodyn. 2010, 29, 603–608. [Google Scholar] [CrossRef]
  63. Liu, G.; Li, M.; Vasanji, A.; Daneshgari, F. Temporal diabetes and diuresis-induced alteration of nerves and vasculature of the urinary bladder in the rat. BJU Int. 2011, 107, 1988–1993. [Google Scholar] [CrossRef] [PubMed]
  64. Nirmal, J.; Tyagi, P.; Chuang, Y.C.; Lee, W.C.; Yoshimura, N.; Huang, C.C.; Rajaganapathy, B.; Chancellor, M.B. Functional and molecular characterization of hyposensitive underactive bladder tissue and urine in streptozotocin-induced diabetic rat. PLoS ONE 2014, 9, e102644. [Google Scholar] [CrossRef] [PubMed]
  65. He, Q.; Babcook, M.A.; Shukla, S.; Shankar, E.; Wang, Z.; Liu, G.; Erokwu, B.O.; Flask, C.A.; Lu, L.; Daneshgari, F.; et al. Obesity-initiated metabolic syndrome promotes urinary voiding dysfunction in a mouse model. Prostate 2016, 76, 964–976. [Google Scholar] [CrossRef] [PubMed]
  66. De Oliveira, M.G.; Nascimento, D.M.; Alexandre, E.C.; Bonilla-Becerra, S.M.; Zapparoli, A.; Mónica, F.Z.; Antunes, E. Menthol ameliorates voiding dysfunction in types I and II diabetic mouse model. Neurourol. Urodyn. 2018, 37, 2510–2518. [Google Scholar] [CrossRef] [PubMed]
  67. Blaha, I.; López-Oliva, M.E.; Martínez, M.P.; Recio, P.; Agis-Torres, Á.; Martínez, A.C.; Benedito, S.; García-Sacristán, A.; Prieto, D.; Fernandes, V.S.; et al. Bladder dysfunction in an obese Zucker rat: The role of TRPA1 channels, oxidative Stress, and hydrogen sulfide. Oxid. Med. Cell Longev. 2019, 2019, 5641645. [Google Scholar] [CrossRef] [PubMed]
  68. Eleazu, C.O.; Eleazu, K.C.; Chukwuma, S.; Essien, U.N. Review of the mechanism of cell death resulting from streptozotocin challenge in experimental animals, its practical use and potential risk to humans. J. Diabetes Metab. Disord. 2013, 12, 60. [Google Scholar] [CrossRef] [PubMed]
  69. Pandey, S.; Chmelir, T.; Chottova, D.M. Animal models in diabetic research-history, presence, and future perspectives. Biomedicines 2023, 11, 2852. [Google Scholar] [CrossRef] [PubMed]
  70. Andersson, P.O.; Malmgren, A.; Uvelius, B. Cystometrical and in vitro evaluation of urinary bladder function in rats with STZ-induced diabetes. J. Urol. 1988, 139, 1359–1362. [Google Scholar] [CrossRef]
  71. Eika, B.; Levin, R.M.; Longhurst, P.A. Comparison of urinary bladder function in rats with hereditary diabetes insipidus, streptozotocin-induced diabetes mellitus, and nondiabetic osmotic diuresis. J. Urol. 1994, 151, 496–502. [Google Scholar] [CrossRef] [PubMed]
  72. Tammela, T.L.; Leggett, R.E.; Levin, R.M.; Longhurst, P.A. Temporal changes in micturition and bladder contractility after sucrose diuresis and streptozotocin-induced diabetes mellitus in rats. J. Urol. 1995, 153, 2014–2021. [Google Scholar] [CrossRef] [PubMed]
  73. Turner, W.H.; Brading, A.F. Smooth muscle of the bladder in the normal and the diseased state: Pathophysiology, diagnosis and treatment. Pharmacol. Ther. 1997, 75, 77–110. [Google Scholar] [CrossRef] [PubMed]
  74. Pitre, D.A.; Ma, T.; Wallace, L.J.; Bauer, J.A. Time-dependent urinary bladder remodeling in the streptozotocin-induced diabetic rat model. Acta Diabetol. 2002, 39, 23–27. [Google Scholar] [CrossRef] [PubMed]
  75. Arioglu, I.E.; Ellenbroek, J.H.; Michel, M.C. A systematic review of urinary bladder hypertrophy in experimental diabetes: Part I. Streptozotocin-induced rat models. Neurourol. Urodyn. 2018, 37, 1212–1219. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, G.; Daneshgari, F. Alterations in neurogenically mediated contractile responses of urinary bladder in rats with diabetes. Am. J. Physiol. Renal Physiol. 2005, 288, F1220–F1226. [Google Scholar] [CrossRef] [PubMed]
  77. Daneshgari, F.; Huang, X.; Liu, G.; Bena, J.; Saffore, L.; Powell, C.T. Temporal differences in bladder dysfunction caused by diabetes, diuresis, and treated diabetes in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2006, 290, R1728–R1735. [Google Scholar] [CrossRef] [PubMed]
  78. Daneshgari, F.; Liu, G.; Imrey, P.B. Time dependent changes in diabetic cystopathy in rats include compensated and decompensated bladder function. J. Urol. 2006, 176, 380–386. [Google Scholar] [CrossRef] [PubMed]
  79. Xiao, N.; Huang, Y.; Kavran, M.; Elrashidy, R.A.; Liu, G. Short-term diabetes- and diuresis-induced alterations of the bladder are mostly reversible in rats. Int. J. Urol. 2015, 22, 410–415. [Google Scholar] [CrossRef] [PubMed]
  80. Yesilyurt, Z.E.; Matthes, J.; Hintermann, E.; Castañeda, T.R.; Elvert, R.; Beltran-Ornelas, J.H.; Silva-Velasco, D.L.; Xia, N.; Kannt, A.; Christen, U.; et al. Analysis of 16 studies in nine rodent models does not support the hypothesis that diabetic polyuria is a main reason of urinary bladder enlargement. Front. Physiol. 2022, 13, 923555. [Google Scholar] [CrossRef] [PubMed]
  81. Cao, N.; Alexandre, E.C.; Gotoh, D.; Kurobe, M.; Mizoguchi, S.; Gu, B.; Yoshimura, N. Urethral dysfunction and alterations of nitric oxide mechanisms in streptozotocin-induced diabetic rats with or without low-dose insulin treatment. Life Sci. 2020, 249, 117537. [Google Scholar] [CrossRef] [PubMed]
  82. Gotoh, D.; Cao, N.; Alexandre, E.C.; Saito, T.; Morizawa, Y.; Hori, S.; Miyake, M.; Torimoto, K.; Fujimoto, K.; Yoshimura, N. Effects of low-dose insulin or a soluble guanylate cyclase activator on lower urinary tract dysfunction in streptozotocin-induced diabetic rats. Life Sci. 2021, 286, 120001. [Google Scholar] [CrossRef] [PubMed]
  83. Erdogan, B.R.; Liu, G.; Arioglu-Inan, E.; Michel, M.C. Established and emerging treatments for diabetes-associated lower urinary tract dysfunction. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2022, 395, 887–906. [Google Scholar] [CrossRef] [PubMed]
  84. Leiria, L.O.; Mónica, F.Z.; Carvalho, F.D.; Claudino, M.A.; Franco-Penteado, C.F.; Schenka, A.; Grant, A.D.; De Nucci, G.; Antunes, E. Functional, morphological and molecular characterization of bladder dysfunction in streptozotocin-induced diabetic mice: Evidence of a role for L-type voltage-operated Ca2+ channels. Br. J. Pharmacol. 2011, 163, 1276–1288. [Google Scholar] [CrossRef]
  85. Yu, W.; Ackert-Bicknell, C.; Larigakis, J.D.; MacIver, B.; Steers, W.D.; Churchill, G.A.; Hill, W.G.; Zeidel, M.L. Spontaneous voiding by mice reveals strain-specific lower urinary tract function to be a quantitative genetic trait. Am. J. Physiol. Renal Physiol. 2014, 306, F1296–F1307. [Google Scholar] [CrossRef] [PubMed]
  86. Bjorling, D.E.; Wang, Z.; Vezina, C.M.; Ricke, W.A.; Keil, K.P.; Yu, W.; Guo, L.; Zeidel, M.L.; Hill, W.G. Evaluation of voiding assays in mice: Impact of genetic strains and sex. Am. J. Physiol. Renal Physiol. 2015, 308, F1369–F1378. [Google Scholar] [CrossRef] [PubMed]
  87. Mossa, A.H.; Galan, A.; Cammisotto, P.G.; Velasquez Flores, M.; Shamout, S.; Barcelona, P.; Saragovi, H.U.; Campeau, L. Antagonism of proNGF or its receptor p75NTR reverses remodelling and improves bladder function in a mouse model of diabetic voiding dysfunction. Diabetologia 2020, 63, 1932–1946. [Google Scholar] [CrossRef] [PubMed]
  88. Yoshizawa, T.; Hayashi, Y.; Yoshida, A.; Yoshida, S.; Ito, Y.; Yamaguchi, K.; Yamada, S.; Takahashi, S. Concomitant alteration in number and affinity of P2X and muscarinic receptors are associated with bladder dysfunction in early stage of diabetic rats. Int. Urol. Nephrol. 2018, 50, 451–458. [Google Scholar] [CrossRef] [PubMed]
  89. Hughes, F.M., Jr.; Allkanjari, A.; Odom, M.R.; Jin, H.; Purves, J.T. 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. [Google Scholar] [CrossRef] [PubMed]
  90. Hughes, F.M., Jr.; Allkanjari, A.; Odom, M.R.; Mulcrone, J.E.; Jin, H.; Purves, J.T. Male Akita mice develop signs of bladder underactivity independent of NLRP3 as a result of a decrease in neurotransmitter release from efferent neurons. Am. J. Physiol. Renal Physiol. 2023, 325, F61–F72. [Google Scholar] [CrossRef]
  91. Mónica, F.Z.; Antunes, E. Stimulators and activators of soluble guanylate cyclase for urogenital disorders. Nat. Rev. Urol. 2018, 15, 42–54. [Google Scholar] [CrossRef] [PubMed]
  92. Torimoto, K.; Fraser, M.O.; Hirao, Y.; De Groat, W.C.; Chancellor, M.B.; Yoshimura, N. Urethral dysfunction in diabetic rats. J. Urol. 2004, 171, 1959–1964. [Google Scholar] [CrossRef] [PubMed]
  93. Melman, A.; Zotova, E.; Kim, M.; Arezzo, J.; Davies, K.; DiSanto, M.; Tar, M. Longitudinal studies of time-dependent changes in both bladder and erectile function after streptozotocin-induced diabetes in Fischer 344 male rats. BJU Int. 2009, 104, 1292–1300. [Google Scholar] [CrossRef] [PubMed]
  94. Radenković, M.; Stojanović, M.; Prostran, M. Experimental diabetes induced by alloxan and streptozotocin: The current state of the art. J. Pharmacol. Toxicol. Methods 2016, 78, 13–31. [Google Scholar] [CrossRef] [PubMed]
  95. Uvelius, B. Detrusor smooth muscle in rats with alloxan-induced diabetes. J. Urol. 1986, 136, 949–952. [Google Scholar] [CrossRef] [PubMed]
  96. Paro, M.; Italiano, G.; Travagli, R.A.; Petrelli, L.; Zanoni, R.; Prosdocimi, M.; Fiori, M.G. Cystometric changes in alloxan diabetic rats: Evidence for functional and structural correlates of diabetic autonomic neuropathy. J. Auton. Nerv. Syst. 1990, 30, 1–11. [Google Scholar] [CrossRef]
  97. Rodrigues, A.A., Jr.; Suaid, H.J.; Tucci, S., Jr.; Fazan, V.P.; Foss, M.C.; Cologna, A.J.; Martins, A.C. Long term evaluation of functional and morphological bladder alterations on alloxan-induced diabetes and aging: Experimental study in rats. Acta Cir. Bras. 2008, 23 (Suppl. S1), 53–58; discussion 58. [Google Scholar] [CrossRef] [PubMed]
  98. Rocha, J.N. Functional and biochemical characteristics of urinary bladder muscarinic receptors in long-term alloxan diabetic rats. Einstein 2015, 13, 404–409. [Google Scholar] [CrossRef] [PubMed]
  99. Chang, S.; Hypolite, J.A.; DiSanto, M.E.; Changolkar, A.; Wein, A.J.; Chacko, S. Increased basal phosphorylation of detrusor smooth muscle myosin in alloxan-induced diabetic rabbit is mediated by upregulation of Rho-kinase beta and CPI-17. Am. J. Physiol. Renal Physiol. 2006, 290, F650–F656. [Google Scholar] [CrossRef]
  100. Ichiyanagi, N.; Tsujii, T.; Masuda, H.; Kihara, K.; Goto, M.; Azuma, H. Changed responsiveness of the detrusor in rabbits with alloxan induced hyperglycemia: Possible role of 5-hydroxytryptamine for diabetic bladder dysfunction. J. Urol. 2002, 168, 303–307. [Google Scholar] [CrossRef] [PubMed]
  101. Aravani, D.; Kassi, E.; Chatzigeorgiou, A.; Vakrou, S. Cardiometabolic syndrome: An update on available mouse models. Thromb. Haemost. 2021, 121, 703–715. [Google Scholar] [CrossRef] [PubMed]
  102. Winzell, M.S.; Ahrén, B. The high-fat diet-fed mouse: A model for studying mechanisms and treatment of impaired glucose tolerance and type 2 diabetes. Diabetes 2004, 53 (Suppl. S3), S215–S219. [Google Scholar] [CrossRef] [PubMed]
  103. Rahman, N.U.; Phonsombat, S.; Bochinski, D.; Carrion, R.E.; Nunes, L.; Lue, T.F. An animal model to study lower urinary tract symptoms and erectile dysfunction: The hyperlipidaemic rat. BJU Int. 2007, 100, 658–663. [Google Scholar] [CrossRef] [PubMed]
  104. Alexandre, E.C.; Calmasini, F.B.; de Oliveira, M.G.; Silva, F.H.; da Silva, C.P.V.; André, D.M.; Leonardo, F.C.; Delbin, M.A.; Antunes, E. Chronic treatment with resveratrol improves overactive bladder in obese mice via antioxidant activity. Eur. J. Pharmacol. 2016, 788, 29–36. [Google Scholar] [CrossRef] [PubMed]
  105. Calmasini, F.B.; de Oliveira, M.G.; Alexandre, E.C.; da Silva, F.H.; da Silva, C.P.V.; Candido, T.Z.; Antunes, E.; Mónica, F.Z. Long-term treatment with the beta-3 adrenoceptor agonist, mirabegron ameliorates detrusor overactivity and restores cyclic adenosine monophosphate (cAMP) levels in obese mice. Neurourol. Urodyn. 2017, 36, 1511–1518. [Google Scholar] [CrossRef] [PubMed]
  106. Leiria, L.O.; Sollon, C.; Calixto, M.C.; Lintomen, L.; Mónica, F.Z.; Anhê, G.F.; De Nucci, G.; Zanesco, A.; Grant, A.D.; Antunes, E. Role of PKC and CaV1.2 in detrusor overactivity in a model of obesity associated with insulin resistance in mice. PLoS ONE 2012, 7, e48507. [Google Scholar] [CrossRef] [PubMed]
  107. Leiria, L.O.; Sollon, C.; Báu, F.R.; Mónica, F.Z.; D’Ancona, C.L.; De Nucci, G.; Grant, A.D.; Anhê, G.F.; Antunes, E. Insulin relaxes bladder via PI3K/AKT/eNOS pathway activation in mucosa: Unfolded protein response-dependent insulin resistance as a cause of obesity-associated overactive bladder. J. Physiol. 2013, 591, 2259–2273. [Google Scholar] [CrossRef] [PubMed]
  108. Leiria, L.O.; Silva, F.H.; Davel, A.P.; Alexandre, E.C.; Calixto, M.C.; De Nucci, G.; Mónica, F.Z.; Antunes, E. The soluble guanylyl cyclase activator BAY 60-2770 ameliorates overactive bladder in obese mice. J. Urol. 2014, 191, 539–547. [Google Scholar] [CrossRef] [PubMed]
  109. Malysz, J.; Petkov, G.V. Urinary bladder smooth muscle ion channels: Expression, function, and regulation in health and disease. Am. J. Physiol. Renal Physiol. 2020, 319, F257–F283. [Google Scholar] [CrossRef] [PubMed]
  110. Jiang, X.; Luttrell, I.; Chitaley, K.; Yang, C.C. T- and L-type voltage-gated calcium channels: Their role in diabetic bladder dysfunction. Neurourol. Urodyn. 2014, 33, 147–152. [Google Scholar] [CrossRef] [PubMed]
  111. Alexandre, E.C.; Leiria, L.O.; Silva, F.H.; Mendes-Silvério, C.B.; Calmasini, F.B.; Davel, A.P.; Mónica, F.Z.; De Nucci, G.; Antunes, E. Soluble guanylyl cyclase (sGC) degradation and impairment of nitric oxide-mediated responses in urethra from obese mice: Reversal by the sGC activator BAY 60-2770. J. Pharmacol. Exp. Ther. 2014, 349, 2–9. [Google Scholar] [CrossRef] [PubMed]
  112. Alexandre, E.C.; Calmasini, F.B.; Sponton, A.C.D.S.; de Oliveira, M.G.; André, D.M.; Silva, F.H.; Delbin, M.A.; Mónica, F.Z.; Antunes, E. Influence of the periprostatic adipose tissue in obesity-associated mouse urethral dysfunction and oxidative stress: Effect of resveratrol treatment. Eur. J. Pharmacol. 2018, 836, 25–33. [Google Scholar] [CrossRef] [PubMed]
  113. Calmasini, F.B.; McCarthy, C.G.; Wenceslau, C.F.; Priviero, F.B.M.; Antunes, E.; Webb, R.C. Toll-like receptor 9 regulates metabolic profile and contributes to obesity-induced benign prostatic hyperplasia in mice. Pharmacol. Rep. 2020, 72, 179–187. [Google Scholar] [CrossRef] [PubMed]
  114. Lee, Y.C.; Lin, G.; Wang, G.; Reed, A.; Lu, Z.; Wang, L.; Banie, L.; Lue, T.F. Impaired contractility of the circular striated urethral sphincter muscle may contribute to stress urinary incontinence in female zucker fatty rats. Neurourol. Urodyn. 2017, 36, 1503–1510. [Google Scholar] [CrossRef] [PubMed]
  115. Kim, A.K.; Hamadani, C.; Zeidel, M.L.; Hill, W.G. Urological complications of obesity and diabetes in males and females of three mouse models: Temporal manifestations. Am. J. Physiol. Renal Physiol. 2020, 318, F160–F174. [Google Scholar] [CrossRef] [PubMed]
  116. Wu, L.; Wang, M.; Maher, S.; Fu, P.; Cai, D.; Wang, B.; Gupta, S.; Hijaz, A.; Daneshgari, F.; Liu, G. Effects of different diets used to induce obesity/metabolic syndrome on bladder function in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2023, 324, R70–R81. [Google Scholar] [CrossRef] [PubMed]
  117. Oswal, A.; Yeo, G. Leptin and the control of body weight: A review of its diverse central targets, signaling mechanisms, and role in the pathogenesis of obesity. Obesity 2010, 18, 221–229. [Google Scholar] [CrossRef] [PubMed]
  118. Ellenbroek, J.H.; Arioglu, E.; Michel, M.C. A systematic review of urinary bladder hypertrophy in experimental diabetes: Part 2. Comparison of animal models and functional consequences. Neurourol. Urodyn. 2018, 37, 2346–2360. [Google Scholar] [CrossRef] [PubMed]
  119. Nobe, K.; Yamazak, T.; Tsumita, N.; Hashimoto, T.; Honda, K. Glucose-dependent enhancement of diabetic bladder contraction is associated with a rho kinase-regulated protein kinase C pathway. J. Pharmacol. Exp. Ther. 2009, 328, 940–950. [Google Scholar] [CrossRef] [PubMed]
  120. Wu, L.; Zhang, X.; Xiao, N.; Huang, Y.; Kavran, M.; Elrashidy, R.A.; Wang, M.; Daneshgari, F.; Liu, G. Functional and morphological alterations of the urinary bladder in type 2 diabetic FVB (db/db) mice. J. Diabetes Complicat. 2016, 30, 778–785. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, Z.; Cheng, Z.; Cristofaro, V.; Li, J.; Xiao, X.; Gomez, P.; Ge, R.; Gong, E.; Strle, K.; Sullivan, M.P.; et al. Inhibition of TNF-α improves the bladder dysfunction that is associated with type 2 diabetes. Diabetes 2012, 61, 2134–2145. [Google Scholar] [CrossRef] [PubMed]
  122. Riboulet, C.A.; Pierron, I.; Durand, J.; Murdaca, J.; Giudicelli, E.; Van, O. Methylglyoxal impairs the insulin signaling pathways independently of the formation of intracellular reactive oxygen species. Diabetes 2006, 55, 1289–1299. [Google Scholar] [CrossRef] [PubMed]
  123. Brouwers, O.; Niessen, P.M.; Ferreira, I.; Miyata, T.; Scheffer, P.G.; Teerlink, T.; Schrauwen, P.; Brownlee, M.; Stehouwer, C.D.; Schalkwijk, C.G. Overexpression of glyoxalase-I reduces hyperglycemia-induced levels of advanced glycation end products and oxidative stress in diabetic rats. J. Biol. Chem. 2011, 286, 1374–1380. [Google Scholar] [CrossRef] [PubMed]
  124. Shamsaldeen, Y.A.; Mackenzie, L.S.; Lione, L.A.; Benham, C.D. Methylglyoxal, a metabolite increased in diabetes is associated with insulin resistance, vascular dysfunction and neuropathies. Curr. Drug Metab. 2016, 17, 359–367. [Google Scholar] [CrossRef] [PubMed]
  125. De la Cruz-Ares, S.; Cardelo, M.P.; Gutiérrez-Mariscal, F.M.; Torres-Peña, J.D.; García-Rios, A.; Katsiki, N.; Malagón, M.M.; López-Miranda, J.; Pérez-Martínez, P.; Yubero-Serrano, E.M. Endothelial dysfunction and advanced glycation end products in patients with newly diagnosed versus established diabetes: From the CORDIOPREV Study. Nutrients 2020, 12, 238. [Google Scholar] [CrossRef] [PubMed]
  126. Heber, S.; Haller, P.M.; Kiss, A.; Jäger, B.; Huber, K.; Fischer, M.J.M. Association of plasma methylglyoxal increase after myocardial infarction and the left ventricular ejection fraction. Biomedicines 2024, 10, 605. [Google Scholar] [CrossRef] [PubMed]
  127. Thornalley, P.J.; Langborg, A.; Minhas, H.S. Formation of glyoxal, methylglyoxal and 3-deoxyglucosone in the glycation of proteins by glucose. Biochem. J. 1999, 344 Pt 1, 109–116. [Google Scholar] [CrossRef] [PubMed]
  128. Kalapos, M.P.; de Bari, L. Hidden biochemical fossils reveal an evolutionary trajectory for glycolysis in the prebiotic era. FEBS Lett. 2022, 596, 1955–1968. [Google Scholar] [CrossRef] [PubMed]
  129. Lai, S.W.T.; Lopez Gonzalez, E.J.; Zoukari, T.; Ki, P.; Shuck, S.C. Methylglyoxal and Its Adducts: Induction, Repair, and Association with Disease. Chem. Res. Toxicol. 2022, 35, 1720–1746. [Google Scholar] [CrossRef] [PubMed]
  130. Maessen, D.E.; Stehouwer, C.D.; Schalkwijk, C.G. The role of methylglyoxal and the glyoxalase system in diabetes and other age-related diseases. Clin. Sci. 2015, 28, 839–861. [Google Scholar] [CrossRef] [PubMed]
  131. Hanssen, N.M.J.; Stehouwer, C.D.A.; Schalkwijk, C.G. Methylglyoxal stress, the glyoxalase system, and diabetic chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 2019, 28, 26–33. [Google Scholar] [CrossRef] [PubMed]
  132. Miranda, E.R.; Haus, J.M. Glyoxalase I is a novel target for the prevention of metabolic derangement. Pharmacol. Ther. 2023, 250, 108524. [Google Scholar] [CrossRef] [PubMed]
  133. Brouwers, O.; Niessen, P.M.; Haenen, G.; Miyata, T.; Brownlee, M.; Stehouwer, C.D.; De Mey, J.G.; Schalkwijk, C.G. Hyperglycaemia-induced impairment of endothelium-dependent vasorelaxation in rat mesenteric arteries is mediated by intracellular methylglyoxal levels in a pathway dependent on oxidative stress. Diabetologia 2010, 53, 989–1000. [Google Scholar] [CrossRef] [PubMed]
  134. Thornalley, P.J.; Waris, S.; Fleming, T.; Santarius, T.; Larkin, S.J.; Winklhofer-Roob, B.M.; Stratton, M.R.; Rabbani, N. Imidazopurinones are markers of physiological genomic damage linked to DNA instability and glyoxalase 1-associated tumour multidrug resistance. Nucleic Acids Res. 2010, 38, 5432–5442. [Google Scholar] [CrossRef] [PubMed]
  135. Ban, I.; Sugawa, H.; Nagai, R. Protein Modification with Ribose Generates Nδ-(5-hydro-5-methyl-4-imidazolone-2-yl)-ornithine. Int. J. Mol. Sci. 2022, 23, 1224. [Google Scholar] [CrossRef] [PubMed]
  136. Irshad, Z.; Xue, M.; Ashour, A.; Larkin, J.R.; Thornalley, P.J.; Rabbani, N. Activation of the unfolded protein response in high glucose treated endothelial cells is mediated by methylglyoxal. Sci. Rep. 2019, 9, 7889. [Google Scholar] [CrossRef]
  137. Zhang, X.; Scheijen, J.L.; Stehouwer, C.D.A.; Wouters, K.; Schalkwijk, C. Increased methylglyoxal formation in plasma and tissues during a glucose tolerance test is derived from exogenous glucose. Clin. Sci. 2023, 20, CS20220753. [Google Scholar] [CrossRef] [PubMed]
  138. Erusalimsky, J.D. The use of the soluble receptor for advanced glycation-end products (sRAGE) as a potential biomarker of disease risk and adverse outcomes. Redox Biol. 2021, 42, 101958. [Google Scholar] [CrossRef]
  139. Jangde, N.; Ray, R.; Rai, V. RAGE and its ligands: From pathogenesis to therapeutics. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 555–575. [Google Scholar] [CrossRef] [PubMed]
  140. Dozio, E.; Vettoretti, S.; Caldiroli, L.; Nerini-Molteni, S.; Tacchini, L.; Ambrogi, F.; Messa, P.; Corsi Romanelli, M.M. Advanced glycation end products (AGE) and soluble forms of AGE receptor: Emerging role as mortality risk factors in CKD. Biomedicines 2020, 8, 638. [Google Scholar] [CrossRef]
  141. Ganbaatar, B.; Fukuda, D.; Shinohara, M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Hirata, K.I.; Sata, M. Empagliflozin ameliorates endothelial dysfunction and suppresses atherogenesis in diabetic apolipoprotein E-deficient mice. Eur. J. Pharmacol. 2020, 875, 173040. [Google Scholar] [CrossRef] [PubMed]
  142. Korac, B.; Kalezic, A.; Pekovic-Vaughan, V.; Korac, A.; Jankovic, A. Redox changes in obesity, metabolic syndrome, and diabetes. Redox Biol. 2021, 42, 101887. [Google Scholar] [CrossRef] [PubMed]
  143. Seryogina, E.S.; Kamynina, A.V.; Koroev, D.O.; Volpina, O.M.; Vinokurov, A.Y.; Abramov, A.Y. RAGE induces physiological activation of NADPH oxidase in neurons and astrocytes and neuroprotection. FEBS J. 2024. [Google Scholar] [CrossRef]
  144. Dobrucki, I.T.; Miskalis, A.; Nelappana, M.; Applegate, C.; Wozniak, M.; Czerwinski, A.; Kalinowski, L.; Dobrucki, L.W. Receptor for advanced glycation end-products: Biological significance and imaging applications. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2023, 5, e1935. [Google Scholar] [CrossRef] [PubMed]
  145. Reddy, V.P. Oxidative Stress in Health and Disease. Biomedicines 2023, 11, 2925. [Google Scholar] [CrossRef] [PubMed]
  146. Odani, H.; Shinzato, T.; Matsumoto, Y.; Usami, J.; Maeda, K. Increase in three alpha, beta-dicarbonyl compound levels in human uremic plasma: Specific in vivo determination of intermediates in advanced Maillard reaction. Biochem. Biophys. Res. Commun. 1999, 256, 89–93. [Google Scholar] [CrossRef] [PubMed]
  147. Lapolla, A.; Flamini, R.; Dalla, V.A.; Senesi, A.; Reitano, R.; Fedele, D.; Basso, E.; Seraglia, R.; Traldi, P. Glyoxal and methylglyoxal levels in diabetic patients: Quantitative determination by a new GC/MS method. Clin. Chem. Lab. Med. 2003, 41, 1166–1173. [Google Scholar] [CrossRef] [PubMed]
  148. Ogawa, S.; Nakayama, K.; Nakayama, M.; Mori, T.; Matsushima, M.; Okamura, M.; Senda, M.; Nako, K.; Miyata, T.; Ito, S. Methylglyoxal is a predictor in type 2 diabetic patients of intima-media thickening and elevation of blood pressure. Hypertension 2010, 56, 471–476. [Google Scholar] [CrossRef] [PubMed]
  149. Lu, J.; Randell, E.; Han, Y.; Adeli, K.; Krahn, J.; Meng, Q.H. Increased plasma methylglyoxal level, inflammation, and vascular endothelial dysfunction in diabetic nephropathy. Clin. Biochem. 2011, 44, 307–311. [Google Scholar] [CrossRef] [PubMed]
  150. Terawaki, H.; Nakao, M.; Nakayama, K.; Nakayama, M.; Kimura, A.; Takane, K.; Mitome, J.; Hamaguchi, A.; Ogura, M.; Yokoyama, K.; et al. Peritoneal clearance and transport of methylglyoxal. Nephrol. Dial. Transplant. 2011, 26, 753–754. [Google Scholar] [CrossRef] [PubMed]
  151. Senda, M.; Ogawa, S.; Nako, K.; Okamura, M.; Sakamoto, T.; Ito, S. The strong relation between post-hemodialysis blood methylglyoxal levels and post-hemodialysis blood glucose concentration rise. Clin. Exp. Nephrol. 2015, 9, 527–533. [Google Scholar] [CrossRef] [PubMed]
  152. Ramachandra, B.L.; Vedantham, S.; Krishnan, U.M.; Rayappan, J.B.B. Methylglyoxal—An emerging biomarker for diabetes mellitus diagnosis and its detection methods. Biosens. Bioelectron. 2019, 133, 107–124. [Google Scholar] [CrossRef] [PubMed]
  153. Gutierrez-Mariscal, F.M.; Cardelo, M.P.; de la Cruz, S.; Alcala-Diaz, J.F.; Roncero-Ramos, I.; Guler, I.; Vals-Delgado, C.; López-Moreno, A.; Luque, R.M.; Delgado-Lista, J.; et al. Reduction in circulating advanced glycation end products by mediterranean diet is associated with increased likelihood of yype 2 diabetes remission in patients with coronary heart disease: From the Cordioprev Study. Mol. Nutr. Food Res. 2021, 65, e1901290. [Google Scholar] [CrossRef] [PubMed]
  154. Maessen, D.E.; Hanssen, N.M.; Lips, M.A.; Scheijen, J.L.; Willems, D.K.; Pijl, H.; Stehouwer, C.D.; Schalkwijk, C.G. Energy restriction and Roux-en-Y gastric bypass reduce postprandial α-dicarbonyl stress in obese women with type 2 diabetes. Diabetologia 2016, 59, 2013–2017. [Google Scholar] [CrossRef] [PubMed]
  155. Pastor-Belda, M.; Fernández-García, A.J.; Campillo, N.; Pérez-Cárceles, M.D.; Motas, M.; Hernández-Córdoba, M.; Viñas, P. Glyoxal and methylglyoxal as urinary markers of diabetes. Determination using a dispersive liquid-liquid microextraction procedure combined with gas chromatography-mass spectrometry. J. Chromatogr. A 2017, 1509, 43–49. [Google Scholar] [CrossRef] [PubMed]
  156. Bora, S.; Shankarrao Adole, P.; Vishwanath Vinod, K.; Ananthkrishna Pillai, A.; Ahmed, S. The genetic polymorphisms and activity of glyoxalase 1 as a risk factor for acute coronary syndrome in South Indians with type 2 diabetes mellitus. Gene 2023, 10, 147701. [Google Scholar] [CrossRef] [PubMed]
  157. Tanaka, J.; Yamaguchi, K.; Ishikura, H.; Tsubota, M.; Sekiguchi, F.; Seki, Y.; Tsujiuchi, T.; Murai, A.; Umemura, T.; Kawabata, A. Bladder pain relief by HMGB1 neutralization and soluble thrombomodulin in mice with cyclophosphamide-induced cystitis. Neuropharmacology 2014, 79, 112–118. [Google Scholar] [CrossRef]
  158. Hiramoto, S.; Tsubota, M.; Yamaguchi, K.; Okazaki, K.; Sakaegi, A.; Toriyama, Y.; Tanaka, J.; Sekiguchi, F.; Ishikura, H.; Wake, H.; et al. Cystitis-related bladder pain involves ATP-dependent HMGB1 release from macrophages and its downstream H2S/Cav3.2 signaling in mice. Cells 2020, 9, 1748. [Google Scholar] [CrossRef]
  159. Khorramdelazad, H.; Bagheri, V.; Hassanshahi, G.; Karami, H.; Moogooei, M.; Zeinali, M.; Abedinzadeh, M. S100A12 and RAGE expression in human bladder transitional cell carcinoma: A role for the ligand/RAGE axis in tumor progression? Asian Pac. J. Cancer Prev. 2015, 16, 2725–2729. [Google Scholar] [CrossRef] [PubMed]
  160. Karkin, K.; İzol, V.; Kaplan, M.; Değer, M.; Akdoğan, N.; Tansuğ, M.Z. Demonstration of advanced glycation end product (AGE) expression in bladder cancer tissue in type-2 diabetic and non-diabetic patients and the relationship between AGE accumulation and endoplasmic reticulum stress with bladder cancer. Int. J. Clin. Pract. 2021, 75, e14526. [Google Scholar] [CrossRef] [PubMed]
  161. Matsumoto, K.; Fujiwara, Y.; Nagai, R.; Yoshida, M. Immunohistochemical detection of advanced glycation end products in human bladder with specific monoclonal antibody. Int. J. Urol. 2009, 16, 402–405. [Google Scholar] [CrossRef] [PubMed]
  162. Rabbani, N.; Thornalley, P.J. Emerging glycation-based therapeutics-glyoxalase 1 inducers and glyoxalase 1 inhibitors. Int. J. Mol. Sci. 2022, 23, 2453. [Google Scholar] [CrossRef] [PubMed]
  163. Chen, S.J.; Aikawa, C.; Yoshida, R.; Matsui, T. Methylglyoxal-derived hydroimidazolone residue of plasma protein can behave as a predictor of prediabetes in Spontaneously Diabetic Torii rats. Physiol. Rep. 2015, 3, e12477. [Google Scholar] [CrossRef] [PubMed]
  164. Berlanga, J.; Cibrian, D.; Guillén, I.; Freyre, F.; Alba, J.S.; Lopez-Saura, P.; Merino, N.; Aldama, A.; Quintela, A.M.; Triana, M.E.; et al. Methylglyoxal administration induces diabetes-like microvascular changes and perturbs the healing process of cutaneous wounds. Clin. Sci. 2005, 109, 83–95. [Google Scholar] [CrossRef]
  165. Sena, C.M.; Matafome, P.; Crisóstomo, J.; Rodrigues, L.; Fernandes, R.; Pereira, P.; Seiça, R.M. Methylglyoxal promotes oxidative stress and endothelial dysfunction. Pharmacol. Res. 2012, 65, 497–506. [Google Scholar] [CrossRef] [PubMed]
  166. Tikellis, C.; Pickering, R.J.; Tsorotes, D.; Huet, O.; Cooper, M.E.; Jandeleit-Dahm, K.; Thomas, M.C. Dicarbonyl stress in the absence of hyperglycemia increases endothelial inflammation and atherogenesis similar to that observed in diabetes. Diabetes 2014, 63, 3915–3925. [Google Scholar] [CrossRef] [PubMed]
  167. Hanssen, N.M.J.; Tikellis, C.; Pickering, R.J.; Dragoljevic, D.; Lee, M.K.S.; Block, T.; Scheijen, J.L.; Wouters, K.; Miyata, T.; Cooper, M.E.; et al. Pyridoxamine prevents increased atherosclerosis by intermittent methylglyoxal spikes in the aortic arches of ApoE-/- mice. Biomed. Pharmacother. 2023, 158, 114211. [Google Scholar] [CrossRef] [PubMed]
  168. Crisóstomo, J.; Matafome, P.; Santos-Silva, D.; Rodrigues, L.; Sena, C.M.; Pereira, P.; Seiça, R. Methylglyoxal chronic administration promotes diabetes-like cardiac ischaemia disease in Wistar normal rats. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 1223–1230. [Google Scholar] [CrossRef]
  169. Peyret, H.; Konecki, C.; Terryn, C.; Dubuisson, F.; Millart, H.; Feliu, C.; Djerada, Z. Methylglyoxal induces cardiac dysfunction through mechanisms involving altered intracellular calcium handling in the rat heart. Chem. Biol. Interact. 2024, 394, 110949. [Google Scholar] [CrossRef] [PubMed]
  170. Rodrigues, L.; Matafome, P.; Crisóstomo, J.; Santos-Silva, D.; Sena, C.; Pereira, P.; Seiça, R. Advanced glycation end products and diabetic nephropathy: A comparative study using diabetic and normal rats with methylglyoxal-induced glycation. J. Physiol. Biochem. 2014, 70, 173–184. [Google Scholar] [CrossRef] [PubMed]
  171. Jung, E.; Kang, W.S.; Jo, K.; Kim, J. Ethyl pyruvate prevents renal damage induced by methylglyoxal-derived advanced glycation end products. J. Diabetes Res. 2019, 2019, 4058280. [Google Scholar] [CrossRef]
  172. Mojadami, S.; Ahangarpour, A.; Mard, S.A.; Khorsandi, L. Diabetic nephropathy induced by methylglyoxal: Gallic acid regulates kidney microRNAs and glyoxalase1-Nrf2 in male mice. Arch. Physiol. Biochem. 2023, 29, 655–662. [Google Scholar] [CrossRef]
  173. Medeiros, M.L.; de Oliveira, M.G.; Tavares, E.G.; Mello, G.C.; Anhê, G.F.; Mónica, F.Z.; Antunes, E. Long-term methylglyoxal intake aggravates murine Th2-mediated airway eosinophil infiltration. Int. Immunopharmacol. 2020, 81, 106254. [Google Scholar] [CrossRef] [PubMed]
  174. Medeiros, M.L.; Oliveira, A.L.; de Oliveira, M.G.; Mónica, F.Z.; Antunes, E. Methylglyoxal exacerbates lipopolysaccharide-induced acute lung injury via RAGE-induced ROS generation: Protective effects of metformin. J. Inflamm. Res. 2021, 14, 6477–6489. [Google Scholar] [CrossRef] [PubMed]
  175. Medeiros, M.L.; Oliveira, A.L.; Mello, G.C.; Antunes, E. Metformin counteracts the deleterious effects of methylglyoxal on ovalbumin-induced airway eosinophilic inflammation and remodeling. Int. J. Mol. Sci. 2023, 24, 9549. [Google Scholar] [CrossRef] [PubMed]
  176. Zunkel, K.; Simm, A.; Bartling, B. Long-term intake of the reactive metabolite methylglyoxal is not toxic in mice. Food Chem. Toxicol. 2020, 141, 111333. [Google Scholar] [CrossRef] [PubMed]
  177. Oliveira, A.L.; de Oliveira, M.G.; Medeiros, M.L.; Mónica, F.; Antunes, E. Metformin abrogates the voiding dysfunction induced by prolonged methylglyoxal intake. Eur. J. Pharmacol. 2021, 910, 174502. [Google Scholar] [CrossRef] [PubMed]
  178. Al-Robaiy, S.; Navarrete, S.A.; Simm, A. RAGE-dependent effect of exogenous methylglyoxal intake on lung biomechanics in mice. Nutrients 2022, 15, 23. [Google Scholar] [CrossRef] [PubMed]
  179. Tang, Y.; Zhao, Y.; Wang, P.; Sang, S. Simultaneous determination of multiple reactive carbonyl species in high fat diet-induced metabolic disordered mice and the inhibitory effects of rosemary on carbonyl stress. J. Agric. Food Chem. 2021, 69, 1123–1131. [Google Scholar] [CrossRef]
  180. Oliveira, A.L.; Medeiros, M.L.; Gomes, E.T.; Mello, G.C.; Costa, S.K.; Mónica, F.Z.; Antunes, E. TRPA1 channel mediates methylglyoxal-induced mouse bladder dysfunction. Front. Physiol. 2023, 14, 1308077. [Google Scholar] [CrossRef] [PubMed]
  181. Hill, W.G.; Zeidel, M.L.; Bjorling, D.E.; Vezina, C.M. Void spot assay: Recommendations on the use of a simple micturition assay for mice. Am. J. Physiol. Renal Physiol. 2018, 315, F1422–F1429. [Google Scholar] [CrossRef] [PubMed]
  182. Wegner, K.A.; Abler, L.L.; Oakes, S.R.; Mehta, G.S.; Ritter, K.E.; Hill, W.G.; Zwaans, B.M.; Lamb, L.E.; Wang, Z.; Bjorling, D.E.; et al. Void spot assay procedural optimization and software for rapid and objective quantification of rodent voiding function, including overlapping urine spots. Am. J. Physiol. Renal Physiol. 2018, 315, F1067–F1080. [Google Scholar] [CrossRef] [PubMed]
  183. Laddha, A.P.; Kulkarni, Y.A. NADPH oxidase: A membrane-bound enzyme and its inhibitors in diabetic complications. Eur. J. Pharmacol. 2020, 881, 173206. [Google Scholar] [CrossRef] [PubMed]
  184. Poladia, D.P.; Bauer, J.A. Early cell-specific changes in nitric oxide synthases, reactive nitrogen species formation, and ubiquitinylation during diabetes-related bladder remodeling. Diabetes Metab. Res. Rev. 2003, 19, 313–319. [Google Scholar] [CrossRef] [PubMed]
  185. Beshay, E.; Carrier, S. Oxidative stress plays a role in diabetes-induced bladder dysfunction in a rat model. Urology 2004, 64, 1062–1067. [Google Scholar] [CrossRef] [PubMed]
  186. Kanika, N.D.; Chang, J.; Tong, Y.; Tiplitsky, S.; Li, J.; Yohannes, E.; Tar, M.; Chance, M.; Christ, G.J.; Melman, A.; et al. Oxidative stress status accompanying diabetic bladder cystopathy results in the activation of protein degradation pathways. BJU Int. 2011, 107, 1676–1684. [Google Scholar] [CrossRef]
  187. Tomechko, S.E.; Liu, G.; Tao, M.; Schlatzer, D.; Powell, C.T.; Gupta, S.; Chance, M.R.; Daneshgari, F. Tissue specific dysregulated protein subnetworks in type 2 diabetic bladder urothelium and detrusor muscle. Mol. Cell Proteom. 2015, 14, 635–645. [Google Scholar] [CrossRef] [PubMed]
  188. Wang, Y.; Deng, G.G.; Davies, K.P. Novel insights into development of diabetic bladder disorder provided by metabolomic analysis of the rat nondiabetic and diabetic detrusor and urothelial layer. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E471–E479. [Google Scholar] [CrossRef] [PubMed]
  189. Andersson, K.E. Oxidative stress and its possible relation to lower urinary tract functional pathology. BJU Int. 2018, 121, 527–533. [Google Scholar] [CrossRef] [PubMed]
  190. Andersson, K.E. Oxidative stress and its relation to lower urinary tract symptoms. Int. Neurourol. J. 2022, 26, 261–267. [Google Scholar] [CrossRef]
  191. Song, Q.X.; Sun, Y.; Deng, K.; Mei, J.Y.; Chermansky, C.J.; Damaser, M.S. Potential role of oxidative stress in the pathogenesis of diabetic bladder dysfunction. Nat. Rev. Urol. 2022, 19, 581–596. [Google Scholar] [CrossRef] [PubMed]
  192. Khosla, L.; Gong, S.; Weiss, J.P.; Birder, L.A. Oxidative stress biomarkers in age-related lower urinary tract disorders: A systematic review. Int. Neurourol. J. 2022, 26, 3–19. [Google Scholar] [CrossRef]
  193. Miyata, Y.; Matsuo, T.; Mitsunari, K.; Asai, A.; Ohba, K.; Sakai, H. A review of oxidative stress and urinary dysfunction caused by bladder outlet obstruction and treatments using antioxidants. Antioxidants 2019, 8, 132. [Google Scholar] [CrossRef] [PubMed]
  194. De Oliveira, M.; Monica, F.Z.; Passos, G.R.; Victorio, J.A.; Davel, A.P.; Oliveira, A.L.L.; Parada, C.A.; D’Ancona, C.A.L.; Hill, W.G.; Antunes, E. Selective pharmacological inhibition of NOX2 by GSK2795039 improves bladder dysfunction in cyclophosphamide-induced cystitis in mice. Antioxidants 2022, 12, 92. [Google Scholar] [CrossRef] [PubMed]
  195. Calmasini, F.B.; de Oliveira, M.G.; Alexandre, E.C.; Silva, F.H.; Tavares, E.B.G.; André, D.M.; Zapparoli, A.; Antunes, E. Obesity-induced mouse benign prostatic hyperplasia (BPH) is improved by treatment with resveratrol: Implication of oxidative stress, insulin sensitivity and neuronal growth factor. J. Nutr. Biochem. 2018, 55, 53–58. [Google Scholar] [CrossRef] [PubMed]
  196. Elrashidy, R.A.; Kavran, M.; Asker, M.E.; Mohamed, H.E.; Daneshgari, F.; Liu, G. Smooth muscle-specific deletion of MnSOD exacerbates diabetes-induced bladder dysfunction in mice. Am. J. Physiol. Renal Physiol. 2019, 317, F906–F912. [Google Scholar] [CrossRef] [PubMed]
  197. Masuda, H.; Kihara, K.; Saito, K.; Matsuoka, Y.; Yoshida, S.; Chancellor, M.B.; de Groat, W.C.; Yoshimura, N. Reactive oxygen species mediate detrusor overactivity via sensitization of afferent pathway in the bladder of anaesthetized rats. BJU Int. 2008, 101, 775–780. [Google Scholar] [CrossRef]
  198. Frara, N.; Giaddui, D.; Braverman, A.S.; Jawawdeh, K.; Wu, C.; Ruggieri, M.R.; Barbe, M.F. Mechanisms involved in nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox)-derived reactive oxygen species (ROS) modulation of muscle function in human and dog bladders. PLoS ONE 2023, 18, e0287212. [Google Scholar] [CrossRef] [PubMed]
  199. Rowan, S.; Bejarano, E.; Taylor, A. Mechanistic targeting of advanced glycation end-products in age-related diseases. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3631–3643. [Google Scholar] [CrossRef] [PubMed]
  200. Coyle, C.H.; Philips, B.J.; Morrisroe, S.N.; Chancellor, M.B.; Yoshimura, N. Antioxidant effects of green tea and its polyphenols on bladder cells. Life Sci. 2008, 83, 12–18. [Google Scholar] [CrossRef] [PubMed]
  201. Nomiya, M.; Andersson, K.E.; Yamaguchi, O. Chronic bladder ischemia and oxidative stress: New pharmacotherapeutic targets for lower urinary tract symptoms. Int. J. Urol. 2015, 22, 40–46. [Google Scholar] [CrossRef] [PubMed]
  202. Szkudelska, K.; Szkudelski, T. Resveratrol, obesity and diabetes. Eur. J. Pharmacol. 2010, 635, 1–8. [Google Scholar] [CrossRef] [PubMed]
  203. Tabatabaei-Malazy, O.; Lavari, N.; Abdollahi, M. Natural products in the clinical management of metabolic syndrome. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2024. [Google Scholar] [CrossRef]
  204. Tsounapi, P.; Honda, M.; Hikita, K.; Sofikitis, N.; Takenaka, A. Oxidative stress alterations in the bladder of a short-period type 2 diabetes rat model: Antioxidant treatment can be beneficial for the bladder. In Vivo 2019, 33, 1819–1826. [Google Scholar] [CrossRef] [PubMed]
  205. Xu, F.; Du, H.; Hou, J.; Li, N. Anti-inflammation properties of resveratrol in the detrusor smooth muscle of the diabetic rat. Int. Urol. Nephrol. 2022, 54, 2833–2843. [Google Scholar] [CrossRef] [PubMed]
  206. He, Y.; Zeng, H.; Yu, Y.; Zhang, J.; Liu, Q.; Yang, B. Resveratrol improved detrusor fibrosis induced by mast cells during progression of chronic prostatitis in rats. Eur. J. Pharmacol. 2017, 815, 495–500. [Google Scholar] [CrossRef]
  207. Yu, Y.; Jiang, J.; He, Y.; Wang, W.; Shen, C.; Yang, B. Resveratrol improves urinary dysfunction in rats with chronic prostatitis and suppresses the activity of the stem cell factor/c-Kit signaling pathway. Mol. Med. Rep. 2017, 16, 1395–1400. [Google Scholar] [CrossRef] [PubMed]
  208. Lee, Y.L.; Lin, K.L.; Wu, B.N.; Chuang, S.M.; Wu, W.J.; Lee, Y.C.; Ho, W.T.; Juan, Y.S. Epigallocatechin-3-gallate alleviates bladder overactivity in a rat model with metabolic syndrome and ovarian hormone deficiency through mitochondria apoptosis pathways. Sci. Rep. 2018, 8, 5358. [Google Scholar] [CrossRef] [PubMed]
  209. Juan, Y.S.; Chuang, S.M.; Lee, Y.L.; Long, C.Y.; Wu, T.H.; Chang, W.C.; Levin, R.M.; Liu, K.M.; Huang, C.H. Green tea catechins decrease oxidative stress in surgical menopause-induced overactive bladder in a rat model. BJU Int. 2012, 110, E236–E244. [Google Scholar] [CrossRef] [PubMed]
  210. Hsieh, J.T.; Kuo, K.L.; Liu, S.H.; Shi, C.S.; Chang, H.C.; Lin, W.C.; Chou, C.T.; Hsu, C.H.; Liao, S.M.; Wang, Z.H.; et al. Epigallocatechin Gallate Attenuates Partial Bladder Outlet obstruction-induced bladder injury via suppression of endoplasmic reticulum stress-related apoptosis-in vivo study. Urology 2016, 91, 242.e1–242.e9. [Google Scholar] [CrossRef] [PubMed]
  211. Sang, S.; Shao, X.; Bai, N.; Lo, C.Y.; Yang, C.S.; Ho, C.T. Tea polyphenol (-)-epigallocatechin-3-gallate: A new trapping agent of reactive dicarbonyl species. Chem. Res. Toxicol. 2007, 20, 1862–1870. [Google Scholar] [CrossRef]
  212. Zhu, D.; Wang, L.; Zhou, Q.; Yan, S.; Li, Z.; Sheng, J.; Zhang, W. (+)-Catechin ameliorates diabetic nephropathy by trapping methylglyoxal in type 2 diabetic mice. Mol. Nutr. Food Res. 2014, 58, 2249–2260. [Google Scholar] [CrossRef] [PubMed]
  213. Foretz, M.; Guigas, B.; Viollet, B. Metformin: Update on mechanisms of action and repurposing potential. Nat. Rev. Endocrinol. 2023, 19, 460–476. [Google Scholar] [CrossRef] [PubMed]
  214. Schernthaner, G.; Schernthaner, G.H. The right place for metformin today. Diabetes Res. Clin. Pract. 2020, 159, 107946. [Google Scholar] [CrossRef] [PubMed]
  215. Mbara, K.C.; Mofo, P.E.; Driver, C.; Nzuza, S.; Mkhombo, N.T.; Gcwensa, S.K.; Mcobothi, E.N.; Owira, P.M. Metformin turns 62 in pharmacotherapy: Emergence of non-glycaemic effects and potential novel therapeutic applications. Eur. J. Pharmacol. 2021, 898, 173934. [Google Scholar] [CrossRef]
  216. Tobar, N.; Rocha, G.Z.; Santos, A.; Guadagnini, D.; Assalin, H.B.; Camargo, J.A.; Gonçalves, A.E.S.S.; Pallis, F.R.; Oliveira, A.G.; Rocco, S.A.; et al. Metformin acts in the gut and induces gut-liver crosstalk. Proc. Natl. Acad. Sci. USA 2023, 120, e2211933120. [Google Scholar] [CrossRef] [PubMed]
  217. Kender, Z.; Fleming, T.; Kopf, S.; Torzsa, P.; Grolmusz, V.; Herzig, S.; Schleicher, E.; Rácz, K.; Reismann, P.; Nawroth, P.P. Effect of metformin on methylglyoxal metabolism in patients with type 2 diabetes. Exp. Clin. Endocrinol. Diabetes 2014, 122, 316–319. [Google Scholar] [CrossRef] [PubMed]
  218. Chen, L.; Lv, L.; Zhang, L.; Gao, Z.; Liu, Y.; Wang, S.; Zhou, N.; Xia, Y.; Cui, J.; Jiang, X.; et al. Metformin ameliorates bladder dysfunction in a rat model of partial bladder outlet obstruction. Am. J. Physiol. Renal Physiol. 2021, 320, F838–F858. [Google Scholar] [CrossRef] [PubMed]
  219. Silva, F.H.; Alexandre, E.C.; Calmasini, F.B.; Calixto, M.C.; Antunes, E. Treatment with metformin improves erectile dysfunction in a murine model of obesity associated with insulin resistance. Urology 2015, 86, 423.e1–423.e6. [Google Scholar] [CrossRef] [PubMed]
  220. Zhang, S.; Xu, H.; Yu, X.; Wu, Y.; Sui, D. Metformin ameliorates diabetic nephropathy in a rat model of low-dose streptozotocin-induced diabetes. Exp. Ther. Med. 2017, 14, 383–390. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Methylglyoxal (MGO) is a highly reactive α-dicarbonyl compound generated endogenously during the glycolytic pathway. Hyperglycemia in type 1 (T1DM) and type 2 diabetic (T2DM) individuals markedly elevates plasma and urinary levels of MGO as a consequence of glycolytic overload. The abnormal accumulation of MGO (dicarbonyl stress) has been implicated in many diseases. Methylglyoxal promotes post-translational modification of peptides or proteins, leading to the formation of advanced glycation end products (AGEs), including hydroimidazolone derived from arginine (MG-H1) and carboxyethyl-lysine (CEL). AGEs bind to their receptor ligand (termed RAGE) anchored in cell membranes, triggering multiple intracellular signaling pathways, leading to increased reactive oxygen species (ROS) production. Under healthy conditions, glyoxalases (Glo) are the most important enzymatic detoxification system converting MGO into its final product D-lactate. Glyoxalases comprise two main enzymes, namely, Glo1 (lactoylglutathione methylglyoxal lyase) and Glo2 (hydroxyacylglutathione hydrolase), with Glo1 described as a rate-limiting enzyme in detoxification. This image was produced with the assistance of Servier Medical Art (Servier; https://smart.servier.com/ accessed on 18 March 2024), licensed under a Creative Commons Attribution 4.0 Unported License.
Figure 1. Methylglyoxal (MGO) is a highly reactive α-dicarbonyl compound generated endogenously during the glycolytic pathway. Hyperglycemia in type 1 (T1DM) and type 2 diabetic (T2DM) individuals markedly elevates plasma and urinary levels of MGO as a consequence of glycolytic overload. The abnormal accumulation of MGO (dicarbonyl stress) has been implicated in many diseases. Methylglyoxal promotes post-translational modification of peptides or proteins, leading to the formation of advanced glycation end products (AGEs), including hydroimidazolone derived from arginine (MG-H1) and carboxyethyl-lysine (CEL). AGEs bind to their receptor ligand (termed RAGE) anchored in cell membranes, triggering multiple intracellular signaling pathways, leading to increased reactive oxygen species (ROS) production. Under healthy conditions, glyoxalases (Glo) are the most important enzymatic detoxification system converting MGO into its final product D-lactate. Glyoxalases comprise two main enzymes, namely, Glo1 (lactoylglutathione methylglyoxal lyase) and Glo2 (hydroxyacylglutathione hydrolase), with Glo1 described as a rate-limiting enzyme in detoxification. This image was produced with the assistance of Servier Medical Art (Servier; https://smart.servier.com/ accessed on 18 March 2024), licensed under a Creative Commons Attribution 4.0 Unported License.
Biomedicines 12 00939 g001
Figure 2. Bladder alterations at the level of urothelium, lamina propria, and detrusor smooth muscle in mice treated with methylglyoxal (MGO) for 4 and 12 weeks. Activation of the MGO–AGEs–RAGE axis leads to urothelial damage, tissue disorganization, edema, and inflammatory cellular infiltration, along with sensitivity alterations due to TRPA1 channel activation. The in vitro detrusor contractile responses to electrical-field stimulation (EFS), α,β-methylene ATP (purinergic P2X1 receptor agonist), and carbachol (nonselective muscarinic agonist) due to increased Ca2+ sensitization machinery are higher in MGO-treated mice. Higher collagen deposition is seen in bladders of MGO-treated mice. Urodynamic changes, including increases in nonvoiding contractions (NVCs), frequency, bladder capacity, inter-micturition pressure, and residual volume, may also be observed in MGO groups. Drugs capable of scavenging MGO and protecting bladder cells from oxidative insult, such as the polyphenols resveratrol and epigallocatechin-3-gallate, and the antihyperglycemic metformin exert reduce AGEs levels and oxidative stress in bladder tissues. This image was produced with the assistance of Servier Medical Art (Servier; https://smart.servier.com/ accessed on 18 March 2024), licensed under a Creative Commons Attribution 4.0 Unported License.
Figure 2. Bladder alterations at the level of urothelium, lamina propria, and detrusor smooth muscle in mice treated with methylglyoxal (MGO) for 4 and 12 weeks. Activation of the MGO–AGEs–RAGE axis leads to urothelial damage, tissue disorganization, edema, and inflammatory cellular infiltration, along with sensitivity alterations due to TRPA1 channel activation. The in vitro detrusor contractile responses to electrical-field stimulation (EFS), α,β-methylene ATP (purinergic P2X1 receptor agonist), and carbachol (nonselective muscarinic agonist) due to increased Ca2+ sensitization machinery are higher in MGO-treated mice. Higher collagen deposition is seen in bladders of MGO-treated mice. Urodynamic changes, including increases in nonvoiding contractions (NVCs), frequency, bladder capacity, inter-micturition pressure, and residual volume, may also be observed in MGO groups. Drugs capable of scavenging MGO and protecting bladder cells from oxidative insult, such as the polyphenols resveratrol and epigallocatechin-3-gallate, and the antihyperglycemic metformin exert reduce AGEs levels and oxidative stress in bladder tissues. This image was produced with the assistance of Servier Medical Art (Servier; https://smart.servier.com/ accessed on 18 March 2024), licensed under a Creative Commons Attribution 4.0 Unported License.
Biomedicines 12 00939 g002
Figure 3. The abnormal accumulation of methylglyoxal (MGO) enhances the dicarbonyl stress leading to protein glycation and excessive RAGE-mediated ROS production in the urinary bladder. This image was produced with the assistance of Servier Medical Art (Servier; https://smart.servier.com/ accessed on 18 March 2024), licensed under a Creative Commons Attribution 4.0 Unported License.
Figure 3. The abnormal accumulation of methylglyoxal (MGO) enhances the dicarbonyl stress leading to protein glycation and excessive RAGE-mediated ROS production in the urinary bladder. This image was produced with the assistance of Servier Medical Art (Servier; https://smart.servier.com/ accessed on 18 March 2024), licensed under a Creative Commons Attribution 4.0 Unported License.
Biomedicines 12 00939 g003
Table 1. Main findings produced by methylglyoxal (MGO) treatment in rodents.
Table 1. Main findings produced by methylglyoxal (MGO) treatment in rodents.
Reference Number Dose Route of Administration Animal and Strain Sex Treatment with MGO
[164]50–75 mg/kgIntraperitonealWistar ratMaleMicrovascular damage
Microvessel degeneration
[165]50–75 mg/KgDrinking waterSpontaneously diabetic (GK) ratsMaleEndothelial dysfunction
[166]50 mmol/LDrinking waterC57Bl6 ApoE-/-MaleAtherosclerosis
[170]50–75 mg/kgDrinking waterGoto-Kakizaki (GK), nonobese type 2 diabetic ratsNDRenal disease
[171]17.25 mg/kgIntraperitonealSprague Dawley (SD) ratsNDRenal disease
[172]600 mg/kg/dayOralNMRI miceMaleDiabetic nephropathy
[176]500–2000 mg/kgDrinking waterRAGE−/Glo1 ++ miceMale
Female
Renal dysfunction
[178]500 mg/kgDrinking waterRAGE-KO miceMale
Female
Increased airway resistance/decreased maximal inspiratory flow
[173,175]0.5%Drinking waterC57BL/6Junib miceMaleAggravation of allergic airway disease and acute lung injury
ND, nondetermined; T2DM, type 2 diabetes mellitus; KO, knockout.
Table 2. In vivo and in vitro bladder alterations in male and female mice treated with methylglyoxal (MGO) 12 weeks [15,177,180].
Table 2. In vivo and in vitro bladder alterations in male and female mice treated with methylglyoxal (MGO) 12 weeks [15,177,180].
ParameterMaleFemale
Urodynamic evaluationNumber of nonvoiding contractions (NVCs)Biomedicines 12 00939 i001Biomedicines 12 00939 i003
Frequency of voiding Biomedicines 12 00939 i002Biomedicines 12 00939 i001
Bladder capacityBiomedicines 12 00939 i001Biomedicines 12 00939 i002
Bladder smooth Muscle contractility in vitro
(presence of urothelium)
Neurogenic contractions (electrical-Field Stimulation, EFS)Biomedicines 12 00939 i001Biomedicines 12 00939 i001
Muscarinic-mediated contractions (carbachol)Biomedicines 12 00939 i001Biomedicines 12 00939 i003
Purinergic-mediated contractions (α,β-methylene ATP)Biomedicines 12 00939 i001Biomedicines 12 00939 i001
Void spot analysisTotal void volume Biomedicines 12 00939 i001Biomedicines 12 00939 i003
Volume per voidBiomedicines 12 00939 i001Biomedicines 12 00939 i002
Urine spot number Biomedicines 12 00939 i003Biomedicines 12 00939 i001
Urine spot in centerBiomedicines 12 00939 i003Biomedicines 12 00939 i001
Urine spot in cornerBiomedicines 12 00939 i003Biomedicines 12 00939 i003
HistologyCollagen contentBiomedicines 12 00939 i001Biomedicines 12 00939 i001
Arrows indicate Biomedicines 12 00939 i001 increased; Biomedicines 12 00939 i002 decreased or Biomedicines 12 00939 i003 unaltered parameters.
Table 3. Protective effects of alagebrium (ALT-711) on the levels of total AGEs, MG-H1, RAGE and collagen in bladder tissues of obese diabetic ob/ob mice [16].
Table 3. Protective effects of alagebrium (ALT-711) on the levels of total AGEs, MG-H1, RAGE and collagen in bladder tissues of obese diabetic ob/ob mice [16].
Parameterob/obob/ob + ALT-711
Blood glucoseBiomedicines 12 00939 i001Biomedicines 12 00939 i003
AGEs in bladderBiomedicines 12 00939 i001Biomedicines 12 00939 i002
MG-H1 content in bladderBiomedicines 12 00939 i001Biomedicines 12 00939 i002
RAGE content in bladderBiomedicines 12 00939 i001Biomedicines 12 00939 i002
Collagen content in the bladdersBiomedicines 12 00939 i001Biomedicines 12 00939 i002
Volume per voidBiomedicines 12 00939 i001Biomedicines 12 00939 i002
Number of voidsBiomedicines 12 00939 i002Biomedicines 12 00939 i001
Void sizeBiomedicines 12 00939 i001Biomedicines 12 00939 i002
Arrows indicate Biomedicines 12 00939 i001 increased; Biomedicines 12 00939 i002 decreased or Biomedicines 12 00939 i003 unaltered parameters.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oliveira, A.L.; de Oliveira, M.G.; Mónica, F.Z.; Antunes, E. Methylglyoxal and Advanced Glycation End Products (AGEs): Targets for the Prevention and Treatment of Diabetes-Associated Bladder Dysfunction? Biomedicines 2024, 12, 939. https://doi.org/10.3390/biomedicines12050939

AMA Style

Oliveira AL, de Oliveira MG, Mónica FZ, Antunes E. Methylglyoxal and Advanced Glycation End Products (AGEs): Targets for the Prevention and Treatment of Diabetes-Associated Bladder Dysfunction? Biomedicines. 2024; 12(5):939. https://doi.org/10.3390/biomedicines12050939

Chicago/Turabian Style

Oliveira, Akila Lara, Mariana Gonçalves de Oliveira, Fabíola Zakia Mónica, and Edson Antunes. 2024. "Methylglyoxal and Advanced Glycation End Products (AGEs): Targets for the Prevention and Treatment of Diabetes-Associated Bladder Dysfunction?" Biomedicines 12, no. 5: 939. https://doi.org/10.3390/biomedicines12050939

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop