Open Access

Ketamine‑induced bladder dysfunction is associated with extracellular matrix accumulation and impairment of calcium signaling in a mouse model

  • Authors:
    • Cheng‑Huang Shen
    • Shou‑Tsung Wang
    • Shou‑Chieh Wang
    • Shu‑Mei Lin
    • Lei‑Chen Lin
    • Yuan‑Chang Dai
    • Yi‑Wen Liu
  • View Affiliations

  • Published online on: January 29, 2019     https://doi.org/10.3892/mmr.2019.9907
  • Pages: 2716-2728
  • Copyright: © Shen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Due to the rising abuse of ketamine usage in recent years, ketamine‑induced urinary tract syndrome has received increasing attention. The present study aimed to investigate the molecular mechanism underlying ketamine‑associated cystitis in a mouse model. Female C57BL/6 mice were randomly divided into two groups: One group was treated with ketamine (100 mg/kg/day of ketamine for 20 weeks), whereas, the control group was treated with saline solution. In each group, micturition frequency and urine volume were examined to assess urinary voiding functions. Mouse bladders were extracted and samples were examined for pathological and morphological alterations using hematoxylin and eosin staining, Masson's trichrome staining and scanning electron microscopy. A cDNA microarray was conducted to investigate the differentially expressed genes following treatment with ketamine. The results suggested that bladder hyperactivity increased in the mice treated with ketamine. Furthermore, treatment with ketamine resulted in a smooth apical epithelial surface, subepithelial vascular congestion and lymphoplasmacytic aggregation. Microarray analysis identified a number of genes involved in extracellular matrix accumulation, which is associated with connective tissue fibrosis progression, and in calcium signaling regulation, that was associated with urinary bladder smooth muscle contraction. Collectively, the present results suggested that these differentially expressed genes may serve critical roles in ketamine‑induced alterations of micturition patterns and urothelial pathogenesis. Furthermore, the present findings may provide a theoretical basis for the development of effective therapies to treat ketamine‑induced urinary tract syndrome.

Introduction

Ketamine, a non-competitive N-methyl-D-aspartic acid receptor antagonist, was discovered in 1961 and used as a clinical anesthetic (1). Ketamine-induced urinary tract syndrome has received increasing attention due to the rising illicit use of ketamine as a recreational drug among young adults. Previous studies investigated the ketamine-induced urological sequelae (2,3), and it was demonstrated that long-term ketamine abuse may cause lower urinary tract syndromes (LUTS) that resemble interstitial cystitis (4). LUTS are associated with decreased bladder capacity, incontinence, hematuria and suprapubic pain resulting from neurological disorders (4). A number of possible factors responsible for the association between ketamine and LUTS have been proposed on the basis of clinical observations (5), including direct toxic damage on the urothelium causing bladder barrier dysfunction, chronic neurogenic inflammation and immunoglobulin E-mediated hypersensitivity. However, the causative mechanism underlying the association between ketamine abuse and ketamine-associated cystitis (KC) remains unclear.

An increasing number of studies investigating the causes of KC have used in vivo rodent models. Accumulating evidence demonstrated that certain specific molecular factors, including cyclooxygenase-2 and inducible nitric oxide synthase (6,7) may interact with the submucosa inflammatory environment caused by ketamine abuse in clinical cases. In addition, oxidative stress mediated by mitochondria- and endoplasmic reticulum-dependent pathways was identified to be involved in the apoptosis of bladder cells and urothelial defects (8). A previous study demonstrated that long-term treatment with ketamine may lead to submucosa fibrosis, which is associated with epithelial-mesenchymal transition (EMT) mediated by transforming growth factor-β1 (TGF-β1) (9). LUTS-associated voiding dysfunctions were identified to be associated with an increased protein expression level of P2X purinergic receptor 1 (P2RX1) (10), and the downregulation of phosphorylated (p-) transgelin (TAGLN) was identified to be associated with the detrusor overactivity occurring in KC (11).

Due to the unclear etiology and the unavailability of clinical treatments for KC, a genomic approach, using microarray analyses to identify the molecular factors underlying KC, represents an effective strategy for the development of adequate therapies. Our previous studies based on microarray analysis examined a Balb/c mouse model (12,13). Administration of low-dosage and short-term ketamine (30 mg/kg/day for 8 weeks) induced a significant decrease in keratin 14 expression in urothelial tissue, suggesting that ketamine was able to damage the urothelial cell structure (12). Furthermore, using high-dosage, long-term ketamine injections (100 mg/kg/day for 20 weeks), the expression levels of numerous genes involved in the chronic wound healing response were altered, suggesting the presence of an initial stage of fibrosis from ketamine abuse (13). However, no obvious inflammatory and histological anomalies were identified in the bladders of control mice and mice treated with ketamine. The C57BL/6 mouse strain was demonstrated to exhibit increased Type 1 helper T cell-dominant responses compared with Balb/c mice, which presents the Type 2 helper T cell phenotype (14). Due to different inflammatory responses between these two strains, C57BL/6 mice were used in the present study to investigate the effect of ketamine on inflammation. The present study aimed to identify potential disease-associated genes that may help in characterizing the pathogenesis of KC.

Materials and methods

Animals and drug treatment

A total of 40 6-week-old female C57BL/6 mice (weighing 16.9±0.95 g) were purchased from The National Laboratory Animal Center (Taipei, Taiwan). The animals were maintained in an animal care facility in The Biotechnology and Health Hall of National Chiayi University (Chiayi, Taiwan) for 1 week. A total of 20 7-week-old mice received a daily 100 mg/kg intraperitoneal injection of ketamine (Imalgene 1000; Merial; Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany) for 20 consecutive weeks to model the effects of ketamine abuse. A control group consisting of 20 mice was injected with saline solution. The mice were housed in polycarbonate cages, provided with food and water ad libitum, and maintained in a 12-h light-dark cycle at 22±2°C with 60±5% humidity. The health conditions of the mice were monitored daily for 20 weeks by observing specific behaviors, including squeals, decreased locomotor activity and abnormal posture. The mice were weighed weekly. Each mouse was placed in a transparent polycarbonate cage and sacrificed by gradual-fill CO2 exposure with a displacement rate of 20% chamber volume/min. When impaired spontaneous breathing and unconsciousness were observed, the CO2 flow was maintained for >1 min. Cessation of cardiac activity was confirmed via palpation. All animal experiments were approved by The Institutional Animal Care and Use Committee of National Chiayi University (approval no. 104047) and according to the guidelines of The Animal Research: Reporting in vivo Experiments, recommended by The National Centre for the Replacement, Refinement and Reduction of Animals in Research (15).

Mouse voiding behavior analysis

At the end of the 20th week (n=16/group; 4/group mice were excluded due a lack of voiding), urine-voiding characteristics were investigated using a modified voided stain on paper (VSOP) method (16). A total of 20 µl/g distilled water was supplied to each mouse. Following 30 min, a mouse was placed in a standard polycarbonate mouse cage with filter paper (185 mm; Advantec Toyo Kaisha, Ltd., Tokyo, Japan) under the bottom grid. The voiding area and the number of urine spots >0.5 cm in diameter were recorded for 2 h. The urine stains on the filter papers were directly scanned into image files using a scanner, and the stained areas were calculated using Image J (version 1.47; National Institutes of Health, Bethesda, MD, USA) software.

Total RNA extraction

The mouse urinary bladders in each group (n=5/group) were incubated overnight in RNAlater Stabilization Solution (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and subsequently homogenized in TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), according to the manufacturer's protocol. RNA concentration and purity were measured using the NanoDrop 1,000 spectrophotometer (Thermo Fisher Scientific, Inc., Wilmington, DE, USA), and purity was checked using ratios of absorbance values at 260/280 and 260/230 nm. The quality of RNA was assessed using the Bioanalyzer 2,100 (Agilent Technologies, Inc., Santa Clara, CA, USA), and three high-quality samples from each group were analyzed using an oligonucleotide DNA microarray.

Scanning electron microscopy (SEM) and histological examination

A total of seven bladders from sacrificed mice from each group (n=7/group) were inflated in situ with Bouin's fixative, as described in a previous study (17). Following removal, the bladders were incubated in Bouin's fixative at room temperature for 2 h. Each bladder was subsequently cut longitudinally into strips, rinsed with PBS, and processed by dehydration in an ascending series of ethanol solutions, followed by overnight immersion in 100% acetone at 4°C. Subsequently, the samples were critical point dried using liquid CO2 (HCP-2 critical point dryer; Hitachi, Ltd., Tokyo, Japan). The dried samples were subsequently mounted on an aluminum stub, sputter-coated with gold, and imaged using a S-3500N scanning electron microscope (magnification, ×500 or ×900) (Hitachi, Ltd.). A portion of mouse bladders (n=8/group) was fixed in 10% phosphate-buffered formalin for 24 h at room temperature and subsequently embedded in paraffin and cut into 3–4 µm sections for subsequent histopathological observation. Hematoxylin and eosin (HE), and Masson's trichrome stainings were performed at room temperature to examine the morphology of the tissues by light microscope (magnification, ×200 or ×400).

Analysis of mouse oligonucleotide DNA microarray

The Mouse Whole Genome OneArray® (version 2; Phalanx Biotech Group, Hsinchu, Taiwan) contained 27,307 DNA oligonucleotide probes. Each probe consisted of 60 nucleotides and was designed in the sense direction. Among these probes, 26,423 corresponded to the genes annotated in the RefSeq v51 (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/) and Ensembl v65 (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.18/) databases. A total of 884 control probes were additionally included. Fluorescent antisense RNA (aRNA) targets were prepared from 1 µg total RNA samples using the OneArray® Amino Allyl aRNA Amplification kit (Phalanx Biotech Group) and Cy5 dye (GE Healthcare, Chicago, IL, USA). The amplification was performed as follows. Firstly, RNA samples were mixed with first strand cDNA synthesized (FSS) in a PCR tube, followed by incubating for 10 min at 70°C in a thermal cycler. FSS enzyme was added and incubated for a further 1 h at 42°C. Next, Second Strand Master Mix was added to each sample and incubated in the following conditions: 16°C for 15 min, 37°C for 45 min, 65°C for 15 min and 4°C for 5 min. Finally, In Vitro Transcription Master Mix was added to each sample and incubated for a further 16 h at 37°C. Fluorescent targets were hybridized to the Mouse Whole Genome OneArray® with a hybridization buffer (Phalanx Biotech Group) using a hybridization system (Phalanx Biotech Group). Following 16 h of hybridization at 50°C, non-specific binding targets were washed away. The slides were scanned using a DNA microarray scanner (G2505C; Agilent Technologies, Inc.), and the Cy5 fluorescent intensity of each spot was analyzed using GenePix software (version 4.1; Molecular Devices, LLC, Sunnyvale, CA, USA). Each sample was analyzed using technical and biological duplicates, with a reproducibility >0.975 (https://www.excelfunctions.net/excel-correl-function.html). The signal intensity data were analyzed using the Rosetta Resolver® system (Rosetta Biosoftware, Seattle, WA, USA) for data preprocessing and 75 percentile centering normalization. The sample errors were additionally estimated using an error-weighted method. The fold change and the P-value for pairwise sample comparison were calculated for evaluation of differentially expressed genes (DEGs). The DEGs meeting the criteria of log2|fold change| ≥0.5 and P<0.05 were considered for further analysis.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

A total of 2 µg total RNA was used for RT using the High-Capacity cDNA RT kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). Each reaction included 20 ng cDNA, forward and reverse primers at a concentration of 500 nM and 2× Fast SYBR Green PCR master mix (Applied Biosystems; Thermo Fisher Scientific, Inc.). A total of 10 µl reaction volume was used for RT-qPCR analysis with the appropriate primers. Each sample was tested in triplicate. A Bio-Rad CFX Connect real-time PCR machine (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and Bio-Rad CFX Manager software (version 3.0; Bio-Rad Laboratories, Inc.) were used for the experimental setup and data analysis. The thermocycling conditions were as follows: 95°C for 5 min, followed by 39 cycles of 95°C for 15 sec and 60°C for 30 sec, and finally 95°C for 10 sec. The relative quantification was performed using the 2−ΔΔCq method (18). The RT-qPCR data of target genes were normalized to the reference gene GAPDH. The gene analyzed with RT-qPCR and the respective primers were the following: Fibronectin (FN1)-Forward (F), 5′-CCTCGGGAATGGAAAGGGAG-3′ and FN1-Reverse (R), 5′-ACGTGCAGGAGCAAATGGC-3′; versican (VCAN)-F, 5′-AGGAAGAGCAGACTGGAGTTGG-3′ and VCAN-R, 5′-GCTGAGACCCAGGACCATGT-3′; potassium calcium-activated channel subfamily M α1 (KCNMA1)-F, 5′-CCATCCCGTCCACAGCAAAT-3′ and KCNMA1-R, 5′-AGGCATTATCCGGCTCATCTG-3′; potassium calcium-activated channel subfamily M regulatory β subunit 4 (KCNMB4)-F, 5′-CTTCACCTGTGGCACCGACT-3′ and KCNMB4-R, 5′-AGGGCGGGATATAGGAGCAC-3′; ryanodine receptor 1 (RYR1)-F, 5′-TCCGCAAGCAGTATGAGGACC-3′ and RYR1-R, 5′-AGTTCAGGATCATCTGTCCCCAG-3′; protein kinase C β (PRKCB)-F, 5′-TGCCCACCGATAGAGGTACA-3′ and PRKCB-R, 5′-TGACCAGGAACATCAGCATCT-3′; GAPDH-F, 5′-AAGGTCGGTGTGAACGGATT-3′ and GAPDH-R, 5′-GTGAGTGGAGTCATACTGGAACAT-3′.

Gene pathway analysis

To investigate the biological function of the genes differentially expressed between the control mice and the mice treated with ketamine, the Kyoto Encyclopedia of Genes and Genomes (KEGG) database and NABA database (19) were used to analyze the DEGs and identify the enriched pathways. The 3,000 most significant DEGs were analyzed. Gene set enrichment analysis (GSEA; Molecular Signatures Database v6.2; http://software.broadinstitute.org/gsea/msigdb/collections.jsp) was used to perform data enrichment analysis based on the Canonical Pathway database. The pathways meeting the criterion of P<0.0005 were considered for further analysis. Pathview (v1.4.2; http://www.bioconductor.org/packages/release/bioc/html/pathview.html), KEGGREST (v1.4.1; http://www.bioconductor.org/packages/release/bioc/html/KEGGREST.html) and KEGGgraph (v1.22.1; http://www.bioconductor.org/packages/release/bioc/html/KEGGgraph.html) R packages (v3.1.1; http://cran.r-project.org/bin/windows/base/old/3.1.1/NEWS.R-3.1.1.html) were used for pathway visualization.

Statistical analysis

SigmaPlot (version 12.5; Systat Software, Inc., San Jose, CA, USA) was used for the statistical analyses. Data are presented as the mean ± standard deviation or as the mean ± standard error. The RT-qPCR results were calculated from experiments repeated three times. Statistical differences were assessed using Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Alteration of body weight in mice

All mice (n=40) survived the 20-week treatments. Abnormal behaviors and body weight loss were not identified. However, the rate of weight gain in mice treated with ketamine significantly decreased from the first week, compared with control mice, and this trend continued for the remaining 19 weeks (Fig. 1). The weight of mice belonging to the control group increased by an average of 6.41 g, whereas, the weight of the mice treated with ketamine increased by an average of 3.05 g. The phenotype was consistent with the weight loss observed in cases of ketamine abuse in humans (20) and in previous animal studies (10,13), and may be caused by reduced food consumption due to the side effects of ketamine, including dizziness, loss of appetite or nausea (21).

Effect of treatment with ketamine on voiding behavior of mice

To evaluate the effect of ketamine on bladder voiding function, the voiding area and urine spot numbers were recorded for 2 h using a modified VSOP method at the end of the 20th week. The spot areas of mice treated with ketamine were significantly decreased compared with control mice, whereas the spot numbers representing voiding frequency were increased compared with the controls (Fig. 2), thus suggesting that altered voiding function was induced by long-term treatment with ketamine.

Bladder histological examination

The urinary bladders of mice treated with ketamine exhibited significant histopathological alterations compared with the control group. At the end of the 20th week, mice treated with ketamine exhibited a smooth apical epithelial surface and subepithelial vascular congestion. In contrast, the apical side of the epithelium in the control group exhibited a rough surface (Fig. 3A). In addition, the mice treated with ketamine exhibited protrusive and enlarged mucosal folds resembling fibrous expansion of connective tissue stroma (Fig. 3B and C). However, signs of lymphoplasmacytic aggregation were detected in two bladder samples (Fig. 3A), suggesting that treatment with ketamine was not sufficient to induce inflammation in the majority of C57BL/6 mice.

SEM examination of the mice bladder epithelium

The apical surface of the mouse urinary epithelium is covered in scallop-shaped membrane plaques consisting of uroplakins and other integral membrane proteins (22). The SEM results suggested that the urothelial plaques of the control mice were intact, exhibiting a rough surface (Fig. 4A). In contrast, the plaques on the bladders of mice treated with ketamine were thinner and smoother, and tight junctions were identified around hexagon-shaped umbrella cells (Fig. 4B). These morphological alterations were consistent with the results of the HE staining, suggesting a smooth apical epithelial surface in bladders from mice treated with ketamine. However, no additional abnormalities, including denuded or detached urothelium, were identified, suggesting that the urothelial barrier function was not severely impaired following treatment with ketamine.

DEGs and pathways identified via microarray analysis

To analyze the gene network underlying KC development, a Mouse Whole Genome OneArray® was performed to determine the genes regulated by treatment with ketamine. Mice treated with ketamine presented 284 upregulated and 527 downregulated genes compared with control mice. Subsequently, to obtain a comprehensive overview of the identified DEGs and investigate their biological functions, GSEA was performed to examine the involved pathways, based on the KEGG and NABA database. The nine most significant pathways were identified (Fig. 5), including ‘matrisome (ECM glycoproteins, matrisome and matrisome associated)’, ‘calcium signaling pathway’, ‘small cell lung cancer’, ‘MAPK signaling’, ‘regulation of actin cytoskeleton’, ‘neuroactive ligand receptor interaction’ and ‘complement and coagulation cascades’. The DEGs involved in these pathways are presented in Table I. Extracellular matrix (ECM)- and calcium signaling-associated genes were considered for further analysis due to the role they serve in fibrosis and dysregulated micturition (23,24).

Table I.

List of differentially expressed genes and significantly enriched pathways following treatment with ketamine.

Table I.

List of differentially expressed genes and significantly enriched pathways following treatment with ketamine.

A, Matrisome (systematic name: M5889)

Gene symbolNCBI gene IDGene nameFold changeP-value
CILP214425Cartilage intermediate layer protein1.12 3.44×10−2
CLEC4D17474C-type lectin domain family 4 member D1.08 1.44×10−2
LAMC216782Laminin γ 20.95 2.24×10−2
VCAN13003Versican0.88 1.30×10−3
AGT11606 Angiotensinogen0.82 4.10×10−3
FN114268Fibronectin 10.79 8.25×10−5
FGL214190Fibrinogen-like 20.64 1.17×10−3
COL1A212843Collagen type 1 α 20.61 4.46×10−2
FBLN214115Fibulin 20.60 2.46×10−5
S100A2628324S100 calcium binding protein A20.59 3.51×10−2

B, Calcium signaling pathway (systematic name: M2890)

Gene symbolNCBI gene IDGene nameFold changeP-value

CHRNA711441Cholinergic receptor nicotinic α polypeptide 7−1.24 5.74×10−5
CAMK2A12322 Calcium/calmodulin-dependent protein kinase 2 α−1.07 4.34×10−3
RYR120190Ryanodine receptor 1−0.90 9.62×10−7
CACNA1B12287Calcium channel voltage-dependent N type α1 B subunit−0.77 5.64×10−3
BDKRB212062Bradykinin receptor, β 2−0.62 3.49×10−3
NOS218126Nitric oxide synthase 2−0.61 1.17×10−3
KCNMB458802Potassium calcium-activated channel subfamily M regulatory β subunit 4−0.46 9.25×10−2
KCNMA116531Potassium calcium-activated channel subfamily M α 1−0.35 1.29×10−3
EDNRB13618Endothelin receptor type B0.53 9.31×10−3
PLN18821Phospholamban0.53 2.91×10−2
PRKCD18753Protein kinase C δ0.53 2.84×10−2
PRKCB18751Protein kinase C β0.46 2.45×10−2
EDN113614Endothelin 10.40 2.50×10−2

C, Small cell lung cancer (systematic name: M3228)

Gene symbolNCBI gene IDGene nameFold changeP-value

E2F113555E2F transcription factor 1−0.69 3.64×10−4
NFKB118033Nuclear factor of κ light polypeptide gene enhancer in B cells 1−0.63 1.23×10−5
NOS218126Nitric oxide synthase 2−0.61 1.17×10−3
LAMC216782Laminin γ 20.95 2.24×10−2
FN114268Fibronectin 10.79 8.25×10−5
DUSP818218Dual specificity phosphatase 8−1.26 1.60×10−4
HSPA1B15511Heat shock 70 kDa protein 1B−0.79 3.52×10−11
CACNA1B12287Calcium channel voltage-dependent N type α1 B subunit−0.77 5.64×10−3
NR4A115370Nuclear receptor subfamily 4 group A member 1−0.70 1.04×10−5
RRAS266922Related ras viral oncogene homolog 2−0.68 1.20×10−9
FGF1414169Fibroblast growth factor 14−0.65 3.66×10−2
DUSP119252Dual specificity phosphatase 1−0.63 3.99×10−5
NFKB118033Nuclear factor of κ light polypeptide gene enhancer in B-cells 1−0.63 1.23×10−5

E, Regulation of actin cytoskeleton (systematic name: M18306)

Gene symbolNCBI gene IDGene nameFold changeP-value

INSRR23920Insulin receptor-related receptor−0.90 7.72×10−4
F214061Coagulation factor 2−0.74 4.86×10−4
RRAS266922Related ras viral oncogene homolog 2−0.68 1.20×10−9
FGF1414169Fibroblast growth factor 14−0.65 3.66×10−2
BDKRB212062Bradykinin receptor B2−0.62 3.49×10−3
FN114268Fibronectin 10.79 8.25×10−5
CFL212632Cofilin 20.68 2.07×10−4

F, Neuroactive ligand receptor interaction (systematic name: M13380)

Gene symbolNCBI gene IDGene nameFold changeP-value

OPRD118386Opioid receptor δ 1−1.25 7.31×10−6
CHRNA711441Cholinergic receptor nicotinic α 7−1.24 5.74×10−5
GHRHR14602Growth hormone releasing hormone receptor−1.19 2.09×10−3
F214061Coagulation factor 2−0.74 4.86×10−4
GRIA214800Glutamate receptor ionotropic ampa 2−0.78 2.92×10−2
BDKRB212062Bradykinin receptor B2−0.62 3.49×10−3
PPYR119065Pancreatic polypeptide receptor 10.69 2.43×10−3

G, Complement and coagulation cascades (systematic name: M16894)

Gene symbolNCBI gene IDGene nameFold changeP-value

F214061Coagulation factor 2−0.74 4.86×10−4
MASP117174Mannan-binding lectin serine peptidase 1−0.68 3.67×10−2
C612274Complement component 6−0.67 1.82×10−2
BDKRB212062Bradykinin receptor B2−0.62 3.49×10−3

[i] Fold change values are expressed in log2 scale. NCBI, National Center for Biotechnology Information.

Molecular components associated with ECM and calcium signaling are regulated by treatment with ketamine

Genes with a role in ECM, including FN1, fibulin 2, fibrinogen-like 2, laminin γ 2 (LAMC2) and collagen type 1 α 2 (COL1A2) were significantly upregulated (Table I), which was consistent with the histopathological stain results, as the bladders presented connective tissue expansion in the submucosa region (Fig. 3C). Additional ECM-associated genes VCAN, angiotensinogen (AGT) and C-type lectin domain family 4 member D were upregulated, suggesting a role for these genes in modulating the interactions between the ECM and neighboring cells, promoting a profibrotic milieu. Furthermore, genes with a role in the calcium signaling pathway were differentially expressed. In particular, genes associated with calcium transport and intracellular calcium storage were identified, including calcium/calmodulin-dependent protein kinase 2 α (CAMK2A), RYR1, PRKCB, endothelin receptor type B (EDNRB) and phospholamban. Notably, calcium signaling may influence urinary bladder smooth muscle (UBSM) contraction (23). In addition, the large-conductance voltage- and Ca2+-activated (BK) channel is the principal K+ channel regulating local Ca2+ release by the RYRs, located in the sarcoplasmic reticulum membrane (25). Therefore, BK channels are able to regulate the frequency of Ca2+ sparks and subsequent UBSM excitability (25). Notably, two genes, KCNMA1 and KCNMB4, that constitute the α and β subunits of the BK channels (23), were downregulated in mice treated with ketamine. To understand the role of these genes in the calcium signaling pathway, Pathview software was used to visualize the DEGs involved in this pathway (Fig. 6).

Furthermore, the expressions of six genes associated with calcium regulation were validated by RT-qPCR (Fig. 7). The RT-qPCR results suggested that KCNMB4 was significantly downregulated in mice treated with ketamine, and three genes, VCAN, FN1 and PRKCB, were upregulated in the ketamine group, although the difference was not significant. However, the expression levels of KCNMA1 and RYR1 was not affected, according to the RT-qPCR data. Therefore, the microarray analysis performed represents a strategy to investigate the effects of ketamine on the transcriptome profile.

The role of calcium signaling in ketamine-induced UBSM contraction is presented in Fig. 8. Calcium signaling represents one of the principal causes involved in the abnormal voiding behavior identified in mice treated with ketamine (23). The present results provided the basis for further studies aimed to investigate whether ketamine abuse may cause bladder dysfunction in humans.

Discussion

Following a 20-week period, the body weights of mice treated with ketamine were significantly lower compared with the control mice, due to a reduced food intake and the side effects of ketamine (21). Additionally, the phenotype observed may be due to physiological stress caused by chronic ketamine consumption (26). Nevertheless, mice treated with ketamine exhibited increased micturition frequency and decreased voiding volume compared with the control group, suggesting that long-term treatment with ketamine may lead to urinary bladder overactivity, a symptom previously identified to be associated with patients with KC (27). The present finding is consistent with a number of previous studies using rodent models, including mice and rats (8,10). Although the molecular mechanism of ketamine-induced bladder hyperactivity is not fully understood, certain molecular factors, including p-TAGLN (7) and P2RX1 (10), have been proposed to cause detrusor dysfunction.

In the present study, the microarray data suggested that long-term treatment with ketamine may influence detrusor contraction, by influencing the expression of components associated with the calcium signaling pathway. The BK channel is one of the principal potassium channels regulating UBSM function (28). BK channels are expressed in UBSM cells and contain four pore-forming α-subunits, KCNMA1, and four regulatory β1-subunits, KCNMB1, or β4-subunits, KCNMB4 (29). The BK channel has been identified to provide a negative-feedback mechanism to limit the amplitude and duration of UBSM action potentials and associated phasic contractions in response to intracellular Ca2+ spark (30). Decreased gene expression or gene knockout of KCNMB4 was identified to be associated with increased action potential frequency and UBSM excitability (31,32). Therefore, the downregulation of the expression level of KCNMB4, as detected by the present microarray analysis, may represent a cause of the ketamine-associated detrusor overactivity. Furthermore, previous studies suggested that endothelin-1 (EDN1) and its downstream signaling regulated by protein kinase C (PKC) are able to induce smooth muscle contraction by reducing the frequency of Ca2+ sparks (33,34). PKC is additionally involved in maintaining contractility by inhibiting the myosin light chain (MYL) phosphatase complex activity and subsequently increasing the protein expression level of p-MYL (35). Consequently, the increased expression levels of components of the endothelin pathway, including EDNRB and EDN1, in addition to the increased expression levels of proteins belonging to the PKC family, including protein kinase C δ and PRKCB (36), may promote detrusor contraction, and these genes were identified to be differentially expressed in the present study. Furthermore, the downregulation of the expression level of CAMK2 may represent an additional cause of detrusor contraction, since decreased CAMK2 activity was previously identified to be associated with insufficient local Ca2+ spark and Ca2+ reuptake into the sarcoplasmic reticulum (37). Therefore, the increased cytosolic calcium concentration may promote Ca2+-dependent smooth muscle contraction (38). Collectively, the present study suggested that calcium signaling may serve a role in ketamine-induced bladder hyperactivity.

A VSOP test suggested that ketamine was able to significantly affect the voiding behavior of mice following a 20-week treatment. Therefore, to avoid additional secondary effects, the treatment was not prolonged beyond the 20th week. The present results suggested that high-dosage long-term ketamine administration promoted inflammatory cell infiltration and vascular congestion in the submucosal layer of the bladder, which were observed in patients with long-term ketamine abuse (39,40). Although the induced inflammatory response was not present in all mice, the pathological outcome was consistent with previous studies using an identical mouse strain, C57BL/6, and a similar ketamine regimen, 100 mg/kg/day (10,41). However, previous studies performed on rat models identified a pronounced and consistent inflammatory response (7,42). This inconsistency in the inflammatory response between rats and mice may be due to the increased metabolic rate of mice (43). Mice may present a decreased retention time of ketamine and its inflammation-associated metabolites, resulting in a decreased induction of inflammation (44). Furthermore, although urothelial thickness was identified to be unaffected in the present study, SEM analysis suggested that the surface of the bladders of mice treated with ketamine was flat and smooth compared with control mice, which exhibited a rough surface with ridge-like structures, suggesting a decreased accumulation of uroplakins in the urothelium. The present microarray data identified a normal expression level of genes encoding for tight junction proteins, including claudin and occludin, suggesting that the function of the urothelium barrier was not affected. The present results are similar to a previous study published by Rajandram et al (44), demonstrating that a low-dosage ketamine regimen (30 mg/kg), performed for 12 weeks, did not impair the bladder mucosal surface, which exhibited defined tight junctions. However, the present study used a high-dosage, long-term treatment with ketamine (100 mg/kg/day for 20 weeks), and it was hypothesized that increased ketamine consumption may affect the morphology of the bladder apical membrane.

Ketamine is able to cause bladder interstitial fibrosis (4), which induces an irreversible decline in bladder function. Our previous study using Balb/c mice suggested that certain molecular events associated with abnormal wound healing processes may contribute to early-stage fibrosis (13). In the present study, using a C57BL/6 mouse model, submucosal extruding folds were observed in the bladders of mice treated with ketamine. Notably, the expression levels of genes involved in ECM deposition, including FN1, LAMC2 and COL1A2, were upregulated, suggesting that long-term treatment with ketamine may promote bladder fibrosis by regulating the ECM. Furthermore, the expression levels of two genes encoding for ECM components associated with fibrosis, VCAN (45) and AGT (46,47), were upregulated. The present results suggested that active fibroblasts, following treatment with ketamine, may secrete these ECM modulators, subsequently inducing fibrogenic response. Previous studies using Sprague Dawley (SD) rats have demonstrated that TGF-β1 signaling (48) and EMT (9) are associated with ketamine-induced fibrosis. Furthermore, injection of human umbilical cord blood-derived mesenchymal stem cells or antifibrotic compounds, including N-acetylcysteine (49) and hyaluronic acid (50), in SD rat bladder tissue have been demonstrated to decrease fibrosis by inactivating TGF-β signaling. However, TGF-β-associated genes were unaltered in the present microarray analysis. The inconsistent role of TGF-β may be due to development- and species-specific responses. In addition, ketamine-induced fibrosis may be mediated by TGF-β-independent pathways (42). Therefore, further investigation is required to elucidate the discrepancies between rats and mice in response to treatment with ketamine.

In the present study, C57BL/6 mice were treated with ketamine for 20 weeks. To examine the effects of long-term ketamine abuse on urinary bladders, the transcriptional profiles were investigated. The calcium signaling pathway was identified as a possible mechanism underlying detrusor contractility and urinary bladder overactivity. The present findings may help in understanding the molecular mechanism underlying KC and in identifying novel molecular targets for the development of effective therapies to treat ketamine-induced urinary tract syndrome.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from The Ministry of Science and Technology of the Republic of China, Taiwan (grant no. MOST 104-2320-B-415-001-MY3; Taiwan) and from The Ditmanson Medical Foundation Chiayi Christian Hospital Research Program (grant no. R105-008; Taiwan).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

CHS, STW, and YWL were involved in the design of the study, acquisition and interpretation of data, and drafting the article. STW and SCW performed the experiments. SML and LCL assisted with the SEM experiment. YCD assisted with the histopathological analysis and interpretation. CHS, STW and YWL analyzed and discussed the data.

Ethics approval and consent to participate

All animal experiments were approved by The Institutional Animal Care and Use Committee of National Chiayi University (approval no. 104047) and according to the guidelines of The Animal Research: Reporting in vivo Experiments, recommended by The National Centre for the Replacement, Refinement and Reduction of Animals in Research.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Domino EF, Chodoff P and Corssen G: Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin Pharmacol Ther. 6:279–291. 1965. View Article : Google Scholar : PubMed/NCBI

2 

Shahani R, Streutker C, Dickson B and Stewart RJ: Ketamine-associated ulcerative cystitis: A new clinical entity. Urology. 69:810–812. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Chu PS, Kwok SC, Lam KM, Chu TY, Chan SW, Man CW, Ma WK, Chui KL, Yiu MK, Chan YC, et al: 'Street ketamine'-associated bladder dysfunction: A report of ten cases. Hong Kong Med J. 13:311–313. 2007.PubMed/NCBI

4 

Chu PS, Ma WK, Wong SC, Chu RW, Cheng CH, Wong S, Tse JM, Lau FL, Yiu MK and Man CW: The destruction of the lower urinary tract by ketamine abuse: A new syndrome? BJU Int. 102:1616–1622. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Jhang JF, Hsu YH and Kuo HC: Possible pathophysiology of ketamine-related cystitis and associated treatment strategies. Int J Urol. 22:816–825. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Chuang SM, Liu KM, Li YL, Jang MY, Lee HH, Wu WJ, Chang WC, Levin RM and Juan YS: Dual involvements of cyclooxygenase and nitric oxide synthase expressions in ketamine-induced ulcerative cystitis in rat bladder. Neurourol Urodyn. 32:1137–1143. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Gu D, Huang J, Yin Y, Shan Z, Zheng S and Wu P: Long-term ketamine abuse induces cystitis in rats by impairing the bladder epithelial barrier. Mol Biol Rep. 41:7313–7322. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Liu KM, Chuang SM, Long CY, Lee YL, Wang CC, Lu MC, Lin RJ, Lu JH, Jang MY, Wu WJ, et al: Ketamine-induced ulcerative cystitis and bladder apoptosis involve oxidative stress mediated by mitochondria and the endoplasmic reticulum. Am J Physiol Renal Physiol. 309:F318–F331. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Wang J, Chen Y, Gu D, Zhang G, Chen J, Zhao J and Wu P: Ketamine-induced bladder fibrosis involves epithelial-to-mesenchymal transition mediated by transforming growth factor-β1. Am J Physiol Renal Physiol. 313:F961–F972. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Meng E, Chang HY, Chang SY, Sun GH, Yu DS and Cha TL: Involvement of purinergic neurotransmission in ketamine induced bladder dysfunction. J Urol. 186:1134–1141. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Gu D, Huang J, Shan Z, Yin Y, Zheng S and Wu P: Effects of long-term ketamine administration on rat bladder protein levels: A proteomic investigation using two-dimensional difference gel electrophoresis system. Int J Urol. 20:1024–1031. 2013.PubMed/NCBI

12 

Shen CH, Wang ST, Lee YR, Liu SY, Li YZ, Wu JD, Chen YJ and Liu YW: Biological effect of ketamine in urothelial cell lines and global gene expression analysis in the bladders of ketamine-injected mice. Mol Med Rep. 11:887–895. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Shen CH, Wang SC, Wang ST, Lin SM, Wu JD, Lin CT and Liu YW: Evaluation of urinary bladder fibrogenesis in a mouse model of long-term ketamine injection. Mol Med Rep. 14:1880–1890. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Watanabe H, Numata K, Ito T, Takagi K and Matsukawa A: Innate immune response in Th1- and Th2-dominant mouse strains. Shock. 22:460–466. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Kilkenny C, Browne W, Cuthill IC, Emerson M and Altman DG; National Centre for the Replacement and Refinement Reduction of Amimals in Research, : Animal research: Reporting in vivo experiments-the ARRIVE guidelines. J Cereb Blood Flow Metab. 31:991–993. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Sugino Y, Kanematsu A, Hayashi Y, Haga H, Yoshimura N, Yoshimura K and Ogawa O: Voided stain on paper method for analysis of mouse urination. Neurourol Urodyn. 27:548–552. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Cohen SM, Ohnishi T, Clark NM, He J and Arnold LL: Investigations of rodent urinary bladder carcinogens: Collection, processing, and evaluation of urine and bladders. Toxicol Pathol. 35:337–347. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Naba A, Clauser KR, Ding H, Whittaker CA, Carr SA and Hynes RO: The extracellular matrix: Tools and insights for the ‘omics’ era. Matrix Biol. 49:10–24. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Corazza O, Assi S and Schifano F: From ‘Special K’ to ‘Special M’: The evolution of the recreational use of ketamine and methoxetamine. CNS Neurosci Ther. 19:454–460. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Cvrcek P: Side effects of ketamine in the long-term treatment of neuropathic pain. Pain Med. 9:253–257. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Lee G: Uroplakins in the lower urinary tract. Int Neurourol J. 15:4–12. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Petkov GV: Central role of the BK channel in urinary bladder smooth muscle physiology and pathophysiology. Am J Physiol Regul Integr Comp Physiol. 307:R571–R584. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Wynn TA: Cellular and molecular mechanisms of fibrosis. J Pathol. 214:199–210. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Petkov GV: Role of potassium ion channels in detrusor smooth muscle function and dysfunction. Nat Rev Urol. 9:30–40. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Everds NE, Snyder PW, Bailey KL, Bolon B, Creasy DM, Foley GL, Rosol TJ and Sellers T: Interpreting stress responses during routine toxicity studies: A review of the biology, impact, and assessment. Toxicol Pathol. 41:560–614. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Tsai TH, Cha TL, Lin CM, Tsao CW, Tang SH, Chuang FP, Wu ST, Sun GH, Yu DS and Chang SY: Ketamine-associated bladder dysfunction. Int J Urol. 16:826–829. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Heppner TJ, Bonev AD and Nelson MT: Ca(2+)-activated K+ channels regulate action potential repolarization in urinary bladder smooth muscle. Am J Physiol. 273:C110–C117. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Hristov KL, Chen M, Kellett WF, Rovner ES and Petkov GV: Large-conductance voltage- and Ca2+-activated K+ channels regulate human detrusor smooth muscle function. Am J Physiol Cell Physiol. 301:C903–C912. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Hayase M, Hashitani H, Kohri K and Suzuki H: Role of K+ channels in regulating spontaneous activity in detrusor smooth muscle in situ in the mouse bladder. J Urol. 181:2355–2365. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Kita M, Yunoki T, Takimoto K, Miyazato M, Kita K, de Groat WC, Kakizaki H and Yoshimura N: Effects of bladder outlet obstruction on properties of Ca2+-activated K+ channels in rat bladder. Am J Physiol Regul Integr Comp Physiol. 298:R1310–R1319. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Brenner R, Chen QH, Vilaythong A, Toney GM, Noebels JL and Aldrich RW: BK channel beta4 subunit reduces dentate gyrus excitability and protects against temporal lobe seizures. Nat Neurosci. 8:1752–1759. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Khimji AK and Rockey DC: Endothelin-biology and disease. Cell Signal. 22:1615–1625. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Jaggar JH, Porter VA, Lederer WJ and Nelson MT: Calcium sparks in smooth muscle. Am J Physiol Cell Physiol. 278:C235–C256. 2000. View Article : Google Scholar : PubMed/NCBI

35 

Wang T, Kendig DM, Smolock EM and Moreland RS: Carbachol-induced rabbit bladder smooth muscle contraction: Roles of protein kinase C and Rho kinase. Am J Physiol Renal Physiol. 297:F1534–F1542. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Wang T, Kendig DM, Trappanese DM, Smolock EM and Moreland RS: Phorbol 12,13-dibutyrate-induced, protein kinase C-mediated contraction of rabbit bladder smooth muscle. Front Pharmacol. 2:832012. View Article : Google Scholar : PubMed/NCBI

37 

Picht E, DeSantiago J, Huke S, Kaetzel MA, Dedman JR and Bers DM: CaMKII inhibition targeted to the sarcoplasmic reticulum inhibits frequency-dependent acceleration of relaxation and Ca2+ current facilitation. J Mol Cell Cardiol. 42:196–205. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Kuo IY and Ehrlich BE: Signaling in muscle contraction. Cold Spring Harb Perspect Biol. 7:a0060232015. View Article : Google Scholar : PubMed/NCBI

39 

Lin HC, Lee HS, Chiueh TS, Lin YC, Lin HA, Lin YC, Cha TL and Meng E: Histopathological assessment of inflammation and expression of inflammatory markers in patients with ketamine-induced cystitis. Mol Med Rep. 11:2421–2428. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Jhang JF, Hsu YH, Jiang YH, Lee CL and Kuo HC: Histopathological characteristics of ketamine-associated uropathy and their clinical association. Neurourol Urodyn. 37:1764–1772. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Duan Q, Wu T, Yi X, Liu L, Yan J and Lu Z: Changes to the bladder epithelial barrier are associated with ketamine-induced cystitis. Exp Ther Med. 14:2757–2762. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Hills CE, Jin T, Siamantouras E, Liu IK, Jefferson KP and Squires PE: ‘Special k’ and a loss of cell-to-cell adhesion in proximal tubule-derived epithelial cells: modulation of the adherens junction complex by ketamine. PLoS One. 8:e718192013. View Article : Google Scholar : PubMed/NCBI

43 

Martignoni M, Groothuis G and de Kanter R: Comparison of mouse and rat cytochrome P450-mediated metabolism in liver and intestine. Drug Metab Dispos. 34:1047–1054. 2006.PubMed/NCBI

44 

Rajandram R, Ong TA, Razack AH, MacIver B, Zeidel M and Yu W: Intact urothelial barrier function in a mouse model of ketamine-induced voiding dysfunction. Am J Physiol Renal Physiol. 310:F885–F894. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Wight TN: Provisional matrix: A role for versican and hyaluronan. Matrix Biol. 60-61:38–56. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Thanigaimani S, Lau DH, Agbaedeng T, Elliott AD, Mahajan R and Sanders P: Molecular mechanisms of atrial fibrosis: Implications for the clinic. Expert Rev Cardiovasc Ther. 15:247–256. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Nogueira A, Pires MJ and Oliveira PA: Pathophysiological mechanisms of renal fibrosis: A review of animal models and therapeutic strategies. In Vivo. 31:1–22. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Juan YS, Lee YL, Long CY, Wong JH, Jang MY, Lu JH, Wu WJ, Huang YS, Chang WC and Chuang SM: Translocation of NF-κB and expression of cyclooxygenase-2 are enhanced by ketamine-induced ulcerative cystitis in rat bladder. Am J Pathol. 185:2269–2285. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Kim A, Yu HY, Heo J, Song M, Shin JH, Lim J, Yoon SJ, Kim Y, Lee S, Kim SW, et al: Mesenchymal stem cells protect against the tissue fibrosis of ketamine-induced cystitis in rat bladder. Sci Rep. 6:308812016. View Article : Google Scholar : PubMed/NCBI

50 

Lee YL, Lin KL, Chuang SM, Lee YC, Lu MC, Wu BN, Wu WJ, Yuan SF, Ho WT and Juan YS: Elucidating mechanisms of bladder repair after hyaluronan instillation in ketamine-induced ulcerative cystitis in animal model. Am J Pathol. 187:1945–1959. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2019
Volume 19 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen CH, Wang ST, Wang SC, Lin SM, Lin LC, Dai YC and Liu YW: Ketamine‑induced bladder dysfunction is associated with extracellular matrix accumulation and impairment of calcium signaling in a mouse model. Mol Med Rep 19: 2716-2728, 2019
APA
Shen, C., Wang, S., Wang, S., Lin, S., Lin, L., Dai, Y., & Liu, Y. (2019). Ketamine‑induced bladder dysfunction is associated with extracellular matrix accumulation and impairment of calcium signaling in a mouse model. Molecular Medicine Reports, 19, 2716-2728. https://doi.org/10.3892/mmr.2019.9907
MLA
Shen, C., Wang, S., Wang, S., Lin, S., Lin, L., Dai, Y., Liu, Y."Ketamine‑induced bladder dysfunction is associated with extracellular matrix accumulation and impairment of calcium signaling in a mouse model". Molecular Medicine Reports 19.4 (2019): 2716-2728.
Chicago
Shen, C., Wang, S., Wang, S., Lin, S., Lin, L., Dai, Y., Liu, Y."Ketamine‑induced bladder dysfunction is associated with extracellular matrix accumulation and impairment of calcium signaling in a mouse model". Molecular Medicine Reports 19, no. 4 (2019): 2716-2728. https://doi.org/10.3892/mmr.2019.9907