Abstract
Background:
Chronic Prostatitis/Chronic Pelvic Pain Syndrome (CP/CPPS) is a disorder that is characterized by persistent pelvic pain in men of any age. Although several studies suggest that the transient receptor potential vanilloid 1 (TRPV1) channel is involved in various pathways of chronic pain, the TRPV1 channel has not been implicated in chronic pelvic pain associated with CP/CPPS.
Methods:
Male C57BL/6J (B6) and TRPV1 knockout (TRPV1 KO) mice (5-7 weeks old) were used to study the development of pelvic allodynia in a murine model of CP/CPPS called experimental autoimmune prostatitis (EAP). The prostate lobes, dorsal root ganglia (DRG), and spinal cord were excised at day 20. The prostate lobes were assessed for inflammation, TRPV1 expression, and mast cell activity. DRG and spinal cord, between the L6-S4 regions, were analyzed to determine levels of phosphorylated ERK1/2 (p-ERK 1/2). To examine the therapeutic potential of TRPV1, B6 mice with EAP received intraurethral infusion of a TRPV1 antagonist at day 20 (repeated every two days) and pelvic pain was evaluated at days 20, 25, 30, and 35.
Results:
Our data showed that B6 mice with EAP developed pelvic tactile allodynia at days 7, 14, and 20. In contrast, TRPV1 KO mice with EAP do not develop pelvic tactile allodynia at any time point. Although we observed no change in levels of TRPV1 protein expression in the prostate from B6 mice with EAP, there was evidence of significant inflammation and elevated mast cell activation. Interestingly, the prostate from TRPV1 KO mice with EAP showed a lack of mast cell activation despite evidence of prostate inflammation. Next, we observed a significant increase of p-ERK1/2 in the DRG and spinal cord from B6 mice with EAP; however, p-ERK1/2 expression was unaltered in TRPV1 KO mice with EAP. Finally, we confirmed that intraurethral administration of a TRPV1 antagonist peptide reduced pelvic tactile allodynia in B6 mice with EAP after day 20.
Conclusions:
We demonstrated that in a murine model of CP/CPPS, the TRPV1 channel is key to persistent pelvic tactile allodynia and blocking TRPV1 in the prostate may be a promising strategy to quell chronic pelvic pain.
Keywords: Vanilloid 1, Pelvic pain, CPPS, Mast cells, Inflammation
1. INTRODUCTION
Men diagnosed with chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) experience chronic pain in the prostate, urethra, and genitals with urinary symptoms.1,2 The lack of effective therapies in CP/CPPS patients severely impacts the patient’s quality-of-life.3 We have previously used multiple mouse models of CP/CPPS to study the initiation and maintenance of CP/CPPS symptoms.4–6 Experimental autoimmune prostatitis (EAP) is the most common murine model that recapitulates several pathological features reported in CP/CPPS patients7–10 and has been used to discover novel targets and improved treatment strategies.9
Since chronic pain is predominantly treated with opioids that generate undesired side effects (e.g., addiction), the research community is desperately seeking non-opioid drugs to treat chronic pain.11,12 There is a large body of literature indicating that the non-selective cation channel known as transient receptor potential vanilloid 1 (TRPV1) mediates the effects of some stimuli that elicit pain, hyperalgesia, and inflammation.13–15 The TRPV1 channel is found in neurons, epithelial cells, mast cells, and several other immune cells.16,17 Since prostate biopsies are not required to diagnose CP/CPPS patients, the expression and role of TRPV1 in the prostate is understudied. Nevertheless, human prostate biopsies obtained from prostate cancer patients showed that the TRPV1 protein is located predominantly in epithelial cells.18 Despite evidence that TRPV1 is involved in multiple chronic pain conditions, CP/CPPS patients have not been evaluated as potential candidates to benefit from targeted TRPV1 treatments.19
The administration of TRPV1 agonists or antagonists in animal pain models and in clinical trials have generated mixed results based on the delivery site of the compound.20–23 For example, intra-articular injection of the TRPV1 agonist, resiniferatoxin (RTX), in canines with osteoarthritis provided long-term pain relief and intrathecal injections in mice blunts mechanical allodynia.21,24 However, clinical trials conducted in patients with interstitial cystitis (IC)/painful bladder syndrome (PBS) that received intravesicular administration of RTX showed no benefit.22 In a mouse model of neuropathic pain, a TRPV1 antagonist administered intrathecally reduced mechanical allodynia.25 Moreover, clinical trials performed in healthy volunteers showed that a TRPV1 antagonist applied intradermally can mitigate heat evoked pain.23,26 The viability of TRPV1 targeting for chronic pelvic pain and the optimal route of administration therefore require further exploration.
Despite the mixed research results on TRPV1 compounds, studies have demonstrated that TRPV1 deficient mice show decreased pain in models of arthritis, bone cancer, and inflammatory bowel disease.27–29 However, it is unclear whether TRPV1 plays a role in the mouse model of CP/CPPS. Therefore, this study will (i) explore the role of TRPV1 in the maintenance of pelvic tactile allodynia, (ii) establish the relevance of TRPV1 to prostate inflammation and mast cell activation, and (iii) determine whether blocking the TRPV1 channel reduces pelvic tactile allodynia in mice with EAP.
2. MATERIALS AND METHODS
2.1. Animals and Treatments
All animal studies were approved by the Northwestern University Animal Care and Use Committee. Animal experiments were conducted with following strains of mice: C57BL/6J (B6; 000664) and TRPV1 deficient mice (TRPV KO; B6.129X1-Trpv1tm1Jul/J). Mice were obtained from Jackson Laboratory (Bar Harbor, ME) at 5-7 weeks old. All experiments were performed on adult male mice that were age matched littermates. Rat prostate antigen was diluted (10mg/ml) with adjuvant and subcutaneously injected into the shoulder of mice to induce EAP as previously described by our laboratory.5 In therapeutic experiments, a lubricated (sterile) polyethylene (PE-10) (BD Medical; cat.63019-004) catheter tube was inserted intraurethrally and the end of the tube was placed in an area adjacent to the prostate. The length (~1.5cm) and placement of the catheter was determined with Evan’s blue dye applied intraurethrally to littermate mice not used for data analysis. The catheter was attached to a 30-gauge needle mounted on a gastight micro syringe (Hamilton; No.80301) and 10µl of either 0.9% saline (sterile) or TRPV1 antagonist peptide (L-R4W2) (1mM; Tocris; No.1577) was infused into mice with EAP and control cohorts at day 20. The delivery of the peptide or control was repeated every two days for 15 days.
2.2. Behavioral assay
We examined the development of pelvic tactile allodynia in mice with EAP using methodology described in detail with video recordings at: (https://www.jove.com/video/50158/measurement-tactile-allodynia-murine-model-bacterial).4,30 Briefly, mice were habituated for one hour by placing them in individual plexiglass chambers (6×10×12cm) on top of a wire mesh floor and elevated 2 feet via a metal platform rig. Then, von Frey filaments with calibrated forces of 0.04, 0.16, 0.4, 1, and 4g were used to determine the emergence and maintenance of pelvic tactile allodynia. The filaments were applied in increasing force order to the pelvic area considered in close proximity to the prostate with a brief resting period between filaments (approximately 5 seconds). Each filament was applied 10 times by an observer blinded to all groups. The behavioral response(s) to the filament were established as follows: 1) licking or scratching of the pelvic area, 2) retraction of the abdomen, and 3) avoidance (jumping). Behavioral assays were conducted at day 0 (baseline), 7, 14, and 20 after inducing EAP. However, mice treated with either saline or the TRPV1 antagonistic peptide intraurethrally at day 20 were examined an additional 15 days. Data were analyzed as the mean percentage of response frequency ± SEM.
2.3. Inflammation scoring and mast cell counts
Mice used to assess prostatic inflammation and mast cell counts were euthanized on day 20 via cervical dislocation and the following prostate lobes were excised and placed in 10% formalin: ventral prostate (VP), anterior prostate (AP), dorsal and lateral prostate (DLP). Prostate lobes were sectioned and processed at the Northwestern pathology. Prostates were embedded in paraffin, sectioned (5µm thickness), and stained with hematoxylin & eosin (H&E). Prostate sections were scored for inflammation using criteria established by Nickel and colleagues.31 Briefly, the tissue-section score can range from 0 to 3, a score of 0 indicated no inflammation and a score of 3 signaled severe inflammation with a high number of infiltrating cells (e.g., leukocytes) and visible destruction of either the stroma or duct/gland epithelium or both. In a separate set of prostate sections, we stained with toluidine blue (0.02%) and counted the number of mast cells under a 20X objective. The selected prostate area for mast cell counts was random. The dispersal of the toluidine blue stained granules in and around the cell membrane was used to determine whether the mast cells were resting or activated, as previously described.30 Slides were covered with mounting medium (VectaMount; cat.H-5000) and sealed with a coverslip. Images were viewed and collected with a Leica DMI 6000B inverted microscope. Experiments were conducted on consecutive sections (4/animal; n=6-7) and prostate areas were imaged randomly by observers blinded to experimental groups. Percentage differences were determined relative to respective controls.
2.4. Tissue procedures and Western blots
After mice were euthanized as described above, the lumbosacral region (L6-S4) of the spinal cord and corresponding dorsal root ganglia (DRG) were excised, lysed with RIPA buffer (Sigma; cat.R0278), and homogenized. A Bradford protein kit assay (Thermo Scientific; cat.23225) was used to determine total protein and 20µg of protein were loaded onto a 10% SDS-PAGE gel (Bio-Rad; cat.3450010). Proteins were then transferred to PVDF membranes (Bio-Rad; cat.1620177). Membranes were blocked with 1% milk diluent (SeraCare; cat.50-82-01) for 1 hour and incubated with primary rabbit polyclonal anti-phosphorylated-ERK1/2 (p-ERK1/2) (1:1,000; Cell Signaling; cat.9101) antibody overnight at 4○C. In a separate experiment, prostate proteins (20µg) were transferred to PVDF membranes, blocked, and incubated with rabbit polyclonal anti-TRPV1 (1:500; Novus Biologicals; cat.NBP1-97417) antibody overnight at 4○C. The next day the membranes were washed with 1X Tris-Buffered Saline mixed with 0.1% Tween® 20 (TBST) and placed in goat anti-rabbit IgG attached to horseradish peroxidase (HRP) (1:3,000; Bio-Rad; cat.170-6515) secondary antibody and proteins were detected using chemiluminescent HRP substrate (ThermoFisher; cat.34580). Then membranes were rinsed with stripping buffer for 5 minutes, washed, and placed in blocking buffer at room temperature for 1 hour. The membranes were relabeled with either rabbit polyclonal anti-total-ERK1/2 (T-ERK1/2) (1:1,000; Cell Signaling; cat.9102) or rabbit monoclonal anti-β-actin (1:1,000; Cell Signaling; cat.8457) antibody and incubated with goat anti-rabbit IgG bound to HRP. Again, proteins were detected with ECL substrate. Image J software was used to determine optical density of protein bands in the immunoblot.
2.5. Statistical analysis
GraphPad Prism software (La Jolla, CA, version 7) was used to apply two tailed (Welch’s) t-tests to experiments comparing two groups. Behavior assays and mast cell studies were analyzed using a two-way ANOVA test followed by either Bonferroni’s or LSD Fisher’s multiple comparison post-hoc test, respectively. Quantitative data were expressed as mean ± SEM and considered statistically significant different if P<0.05.
3. RESULTS
3.1. TRPV1 deficient mice with EAP do not develop pelvic tactile allodynia
Several studies conducted on TRPV1 KO mice demonstrated changes to synaptic plasticity and a higher tolerance for pain.32–34 Hence, we explored whether TRPV1 KO mice with EAP develop pelvic tactile allodynia at days 0, 7, 14, and 20. Our study revealed that TRPV1 KO mice with EAP (TRPV1 KO-EAP) do not show evidence of sensitization or changes in response frequency at any time point compared to naive TRPV1 KO (TRPV1 KO-Naive) mice (Figure 1A). In contrast, B6 mice with EAP have an increased response frequency compared to naive cohorts at days 7, 14, and 20 (Figure 1B) with this response known to be chronic beyond day 20 (an additional 17 days).35 The present data suggests that TRPV1 channels play an essential role in the development of pelvic tactile allodynia in our mouse model of CP/CPPS.
FIGURE 1.

TRPV1 is required for pelvic pain. (A-B) Graphs show response frequencies measured with von Frey filaments applied to C57BL/6J (B6) mice and TRPV1 KO mice at days 0, 7, 14, and 20. (A) B6 mice with EAP (n=10) develop increased pelvic tactile allodynia compared to naive (n=10) mice at days 7, 14, and 20; (B) however, mice lacking the TRPV1 channel with EAP (TRPV1 KO-EAP; n=7) do not show a change in pelvic tactile allodynia at any time point, compared to respective control (TRPV1 KO-Naive; n=8). Data represents the mean ± SEM and was generated from three independent experiments. (*) denotes P<0.05.
3.2. TRPV1 KO mice with EAP experience prostate inflammation but mast cell activity is suppressed during inflammation
Although TRPV1 KO mice have been studied in various models of inflammation and mast cell activation, the results are not clear due to conflicting data from different tissues.36–40 Thus, we examined whether TRPV1 KO mice or the B6 background strain develop inflammation in the prostate after EAP induction. Prostate sections stained with H&E and scored for inflammation showed that B6 mice with EAP developed significantly higher levels of inflammation (72%) in the DLP lobes compared to control cohorts at day 20 (Figures 2Aa,b–2B). Moreover, we observed abundant leukocyte infiltration in the stroma and near epithelial cells (red arrows) in the DLP lobes from B6 mice with EAP (Figure 2Ab). Interestingly, our TRPV1 KO mouse data was consistent with studies that suggest that the absence of the TRPV1 channel does not diminish tissue inflammation,41–43 since we observed that the DLP lobes from TRPV1 KO mice with EAP showed a significantly higher level of inflammation (69%) and leukocyte infiltration (red arrow) compared to respective controls (Figures 2Ca,b–2D). In addition, we confirmed that the ventral (Figures 2Ac,d; 2Cc,d) and anterior (Figures 2Ae,f; 2Ce,f) prostates lobes obtained from B6 and TRPV1 KO mice with EAP do not show significant inflammation at day 20 (Figures 2B,2D; respectively). Taken together, our data shows that leukocyte infiltration of the prostate is not dependent on TRPV1 expression in the EAP model of chronic prostatitis.
FIGURE 2.

Absence of TRPV1 does not change prostate inflammation. (A,C) Representative images and (B,D) relative quantification of prostate inflammation in the dorsal and lateral prostate (DLP), ventral prostate (VP), and anterior prostate (AP) lobes stained with H&E. (A-B) The DLP lobes excised from mice with (Ab) EAP showed a significant increase in inflammation compared to (Ac) control cohorts. (C-D) Likewise, (Cb) the DLP lobes excised from TRPV1 KO mice with EAP (TRPV1 KO-EAP) demonstrated robust inflammation and leukocyte infiltration (red arrows), compared to respective (Ca) controls (TRPV1 KO-Naive). (A-D) In contrast, the (Ac,d; Cc,d) VP and (Ae,f; Ce,f) AP lobes lacked significant inflammation in all mice. Scale bars indicate 100 microns. Data represents the mean ± SEM. (*) denotes P<0.05.
Since inflammation was predominantly observed in the DLP lobes at day 20, we asked whether TRPV1 expression was altered in the DLP lobes from B6 mice with EAP. Our results revealed that the level of TRPV1 expression in the DLP lobes was unchanged between B6 mice with EAP and respective controls (Figures 3A–B). Next, we investigated changes in mast cells located in the prostate via toluidine blue staining and we observed no difference in total number of mast cells (8% difference) between B6 mice with EAP and control mice (Figures 4Aa,b–4B). However, B6 mice with EAP showed a significant increase (103%) in the number of activated mast cell (red arrows) and a significant decrease (62%) in the number of resting mast cells compared to control cohorts at day 20 (Figures 4Aa,b–4B). We then assessed the status of mast cells in TRPV1 KO mice with and without EAP. At day 20, the total number of mast cells in the prostates obtained from TRPV1 KO mice with EAP was not significantly different from naive TRPV1 KO mice. In contrast to B6 mice with EAP, TRPV1 KO mice with EAP showed no significant difference in resting or activated mast cell numbers at day 20 compared to naive TRPV1 KO mice (Figures 4Ca,b–4D). These results suggest that the lack of TRPV1 diminishes the capability of mast cells in the prostate to undergo activation in response to EAP.
FIGURE 3.

TRPV1 expression is unaltered in the prostate of mice with EAP. (A-B) Protein analysis and immunohistochemistry of the DLP lobes showed no differences in expression of TRPV1 between B6 mice with EAP and respective controls. (A) Representative western blots and (B) densitometry data comparing TRPV1 expression in B6 mice with EAP (n=3) to control (n=3) cohorts. Data represents the mean ± SEM.
FIGURE 4.

Active mast cells increase after prostate inflammation and the absence of TRPV1 hinders activity. (A-D) Panels show representative images of the prostate stained with toluidine blue and excised from B6 and TRPV1 KO mice with or without EAP; graphs depict quantification of mast cell numbers in the prostate for each group. (A) Although B6 with EAP showed no significant changes in total number of mast cells (white arrows) compared to control at day 20, (B) B6 mice with EAP showed an increased number of active mast cells (red arrows) compared to controls. (C-D) Prostates from mice with EAP that lack TRPV1 channels (TRPV1 KO-EAP) showed no changes in mast cell numbers or activity, compared to respective controls (TRPV1 KO-Naive). Scale bars indicate 100 microns. Data represents the mean ± SEM. (*) denotes P<0.05.
3.3. TRPV1 mediates ERK1/2 phosphorylation in DRG and spinal cord
A previous study conducted by our laboratory showed that the DRG from B6 mice with EAP had increased p-ERK1/2 compared to control mice.8 Therefore, we sought to determine whether the absence of the TRPV1 channel changed the expression of p-ERK1/2 in the DRG and spinal cord (L6-S4 regions) excised from mice with EAP. The data showed that B6 mice with EAP had a significantly (97%) higher level of p-ERK1/2 expression in the lumbosacral region of the DRG (Figures 5A–5B) and spinal cord (Figures 5D–5E) compared to control cohorts at day 20. However, TRPV1 KO mice with EAP showed a similar level (<8% difference) of p-ERK1/2 expression in the lumbosacral region of the DRG (Figures 5A,5C) and spinal cord (Figures 5D,5F), compared to control TRPV1 KO mice. Collectively, the data indicates that TRPV1 expression is important for initiating the MAP kinase signaling pathways via the DRG in mice with EAP.
FIGURE 5.

ERK1/2 phosphorylation is linked to TRPV1 expression. (A) Western blot analysis and quantification of ERK1/2 phosphorylation in the lumbosacral region of the DRG (n=3) and (B) spinal cord (n=3) from mice with EAP revealed a significant increase in p-ERK1/2 expression compared to control littermates at day 20. However, (C) DRG and (D) spinal cords obtained from TRPV1 deficient mice with EAP showed no changed in ERK1/2 phosphorylation compared to control cohorts. Densitometry data represents the mean ± SEM. (*) denotes P<0.05.
3.4. TRPV1 antagonist blunts persistent pelvic tactile allodynia
Although several TRPV1 antagonists have been developed and tested on animal models with chronic pain, the efficacy of TRPV1 antagonists targeted to a particular visceral organ (e.g., prostate) have not been studied.44–46 Thus, we considered whether an arginine-rich hexapeptide (RRRRWW-NH2; L-R4W2), a non-competitive TRPV1 antagonist, administered via intraurethral instillation to the prostate inhibits the maintenance of pelvic tactile allodynia in mice with EAP. Our results showed that mice with EAP that received L-R4W2 (every two days) at days 20, 22, 24, 26, 28, 30, 32, and 34 (Figure 6A) developed a pain response similar to B6 mice without EAP that received either saline or L-R4W2 treatment (Figure 6B). In contrast, mice with EAP that received intraurethral infusion of saline developed tactile allodynia from day 7 to 35 (Figure 6B). In line with the role of TRPV1 as a pain receptor, the data highlights the importance of TRPV1 activation in sustained pelvic tactile allodynia and indicates that TRPV1 antagonists may have a role in the treatment of chronic pelvic pain.
FIGURE 6.

Pelvic tactile allodynia is diminished by TRPV1 antagonist peptide. (A) Schematic shows mice treatment regime. Mice (B6) with or without EAP received intraurethral (IU) instillation (10µl) of either sterile 0.9% saline or a TRPV1 antagonist peptide (L-R4W2; 1mM) from day 20 (indicated by black arrows) to 35. (B) After initial treatment at day 20, mice with EAP that received L-R4W2 (EAP+Antagonist; n=3) treatment showed a similar response frequency to mice without EAP that received either saline (Naive+Saline; n=3) or L-R4W2 (Naive+Antagonist; n=3) at days 25, 30, and 35. However, mice with EAP that received intraurethral saline (EAP+Saline; n=3) showed increased response frequency between days 7-35. Data represents the mean ± SEM from 3 independent experiments. (*) denotes P<0.05.
4. DISCUSSION
Within the transient receptor potential vanilloid channel subfamily, TRPV1 is the most well-known receptor and is linked to thermosensation, vasodilation, inflammation, and nociception.47–50 However, studies that examined the functional relevance of TRPV1 during inflammation seem to yield conflicting results due to different outcomes observed in various models of inflammation targeting specific organs. Since the role of TRPV1 is not fully explored in a validated murine model of CP/CPPS (e.g., EAP) that elicits chronic pelvic pain and inflammation of the prostate via autoimmune mechanisms, we investigated whether TRPV1 is required for the development of pain and inflammation and whether targeted inhibition of the TRPV1 channel might be a useful therapy to mitigate symptoms associated with CP/CPPS.
Previously, our laboratory published data indicating that B6 mice with EAP develop sustained pelvic tactile allodynia and prostate inflammation by day 30.8 Here, we scrutinized our CP/CPPS mouse model at day 20 and demonstrated that the absence of TRPV1 disrupts the development of pelvic tactile allodynia in mice with EAP (Figures 1A–1B). Hence, our results align with other pain studies that showed a blunted pain response in TRPV1 KO mice.27,51,52 Then, we assessed whether inflammation is diminished in TRPV1 deficient tissue; as reported by several studies.53,54 Our data revealed that global deletion of the TRPV1 channel did not quench prostate inflammation after the induction of EAP at day 20 (Figures 2C–2D). Thus, our results were consistent with studies that observed inflammation of visceral tissue in TRPV1 KO mice.41,55,56 Although the mechanisms that cause differences in inflammation between tissues lacking TRPV1 is not known, a study reported that TRPV1 KO mice may have a compensatory mechanism to elicit inflammation.57
To add to the complexity, the level of TRPV1 expression in inflamed tissue remains a controversial issue; some studies suggest that TRPV1 expression in human visceral tissue with inflammation is upregulated, whereas others maintained that TRPV1 levels remain the same.58 Biopsies from prostate cancer patients showed increased expression of TRPV1 in the epithelium; however, a direct link between CP/CPPS and subsequent development of prostate cancer is not currently supported by the literature.18 As a result, the level of TRPV1 expression in the prostate is not well defined in CP/CPPS patients or relevant murine models. In the present study, we confirmed that the level of TRPV1 expression is stable in the prostate of mice with EAP (Figures 3A–3B). Although we attempted to identify the cellular location of TRPV1 expression in the DLP lobes via immunohistochemistry, we were unsuccessful due to nonspecific labeling by the TRPV1 antibody (unpublished).
Prior work from our group demonstrated that mast cell activity was enhanced in the prostates from mice with EAP and we confirmed that intraurethral instillation of an ortholog of tryptase-β (predominantly released by mast cells) in mice induced pelvic tactile allodynia.8,30 In the same study, we reported high levels of tryptase-β in the expressed prostatic section (EPS) fluid collected from CP/CPPS patients.8 Therefore, in our present study, we explored whether deletion of the TRPV1 channel from prostate tissue altered mast cell activity in mice with EAP. The data revealed that the absence of the TRPV1 channel restricted degranulation of mast cells in the prostates collected from mice with EAP at day 20 (Figures 4Cb–4D). Differences between our data and other inflammation models can be explained by studies that suggest mast cells can express different cytokines, proteases, and receptors (subtypes) depending on the tissue that they reside.59–61 Here we determined that in a murine autoimmune model of CP/CPPS, the TRPV1 channel influences the activity of mast cells found in the prostate while not having any effect on overall prostate inflammation.
Studies on pain mechanism(s) show that TRPV1 activation in neuronal cells can influence the MAP/ERK1/2 signaling pathway.27,62 In previous studies, we established that mice with EAP have increased ERK1/2 phosphorylation in the DRG.8 Thus, we investigated whether p-ERK1/2 expression changed in the lumbosacral region of the DRG and spinal cord excised from mice with EAP that lack TRPV1. Of note in our present research, we demonstrated that TRPV1 deficient mice with EAP do not have altered expression levels of p-ERK1/2 in either the DRG or spinal cord lumbosacral region (Figure 5A), which correlated with the absence of pelvic tactile allodynia (Figure 1B) and suppressed mast cell activity at day 20 (Figures 4C–4D). These results confirmed that TRPV1 is important for ERK1/2 signaling in regions of the DRG and spinal cord that receive sensory information from the prostate; hence, TRPV1 is required to induce chronic pelvic pain in mice with EAP.
Although there has been controversy regarding the role of TRPV1 agonists and antagonist to treat chronic pain due to conflicting results and undesired secondary effects (e.g., hyperthermia or hypothermia), the TRPV1 channel remains a promising target due to its crucial function in neuronal sensitization and pain.63 Moreover, advances in drug development showed that a new generation of TRPV1 antagonists will be more selective with fewer side effects and may offer better outcomes if applied directly to the affected organ.19 Since the newer TRPV1 antagonist(s) were not commercially available during the execution of our experiments, we used an older (less specific) TRPV1 antagonist peptide called L-R4W2 to determine the therapeutic potential of TRPV1 antagonism in our model. To diminish any potential systemic side effects of L-R4W2, the TRPV1 antagonist was delivered intraurethrally into the prostate of mice with established pelvic tactile allodynia at days 20, 22, 24, 26, 28, 30, 32, and 34 (Figure 6A). Notably, our data confirmed that treatment with L-R4W2 reversed the level of pelvic tactile allodynia in mice with EAP after initial infusion at day 20 (Figure 6B). Our results therefore parallel data that supports TRPV1 antagonists as a viable analgesic to treat chronic pain.20,64 Future studies will address whether L-R4W2 or similar TRPV1 antagonists mitigate chronic visceral pain via other delivery methods (e.g., intrathecal, intra-articular, or intra-cisternal infusion).
5. CONCLUSIONS
In this study, our findings provide a framework for understanding the role of the TRPV1 channel during the development of chronic pelvic pain in an animal model of CP/CPPS called EAP. We demonstrated that 1) the absence of the TRPV1 channel disrupts the progression of pelvic tactile allodynia, 2) inflammation of the prostate in mice with EAP is not dependent on TRPV1 activation and TRPV1 expression levels do not change, 3) global TRPV1 deletion reduced mast cell activation in mice with EAP, 4) the lumbosacral region of the DRG and spinal cord tissue excised from TRPV1 deficient mice with EAP showed normal levels p-ERK1/2, and 5) pharmacological targeting of TRPV1 with an antagonist peptide alleviated pelvic tactile allodynia in mice with EAP. Altogether, our study suggests that blocking the activation of the TRPV1 channel in conjunction with anti-inflammatory compounds might be a suitable treatment option to lessen chronic pelvic pain associated with CP/CPPS.
Synopsis.
The TRPV1 channel is a key mediator in persistent pelvic pain in EAP and targeted therapy with a TRPV1 antagonist may alleviate CP/CPPS symptoms.
Acknowledgements
This project was supported by award numbers F32 DK104544 (KR) and R01 DK083609 (PT) from the National Institute Of Diabetes And Digestive And Kidney Diseases.
Funding Information: National Institute of Diabetes and Digestive and Kidney Diseases, Grant numbers: F32 DK104544 and R01 DK083609.
Footnotes
Disclosure Statement: The authors have no financial interests to disclose.
Conflict of interest statement
The authors do not have any conflict of interest to report with regard to the study reported in this manuscript.
References
- 1.Clemens JQ, Brown SO, Calhoun EA. Mental health diagnoses in patients with interstitial cystitis/painful bladder syndrome and chronic prostatitis/chronic pelvic pain syndrome: a case/control study. J Urol. 2008;180(4):1378–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Schaeffer AJ, Datta NS, Fowler JE Jr., et al. Overview summary statement. Diagnosis and management of chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS). Urology. 2002;60(6 Suppl):1–4. [DOI] [PubMed] [Google Scholar]
- 3.Tripp DA, Nickel JC, Shoskes D, Koljuskov A. A 2-year follow-up of quality of life, pain, and psychosocial factors in patients with chronic prostatitis/chronic pelvic pain syndrome and their spouses. World J Urol. 2013;31(4):733–739. [DOI] [PubMed] [Google Scholar]
- 4.Quick ML, Done JD, Thumbikat P. Measurement of tactile allodynia in a murine model of bacterial prostatitis. J Vis Exp. 2013(71):e50158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rudick CN, Schaeffer AJ, Thumbikat P. Experimental autoimmune prostatitis induces chronic pelvic pain. Am J Physiol Regul Integr Comp Physiol. 2008;294(4):R1268–1275. [DOI] [PubMed] [Google Scholar]
- 6.Vykhovanets EV, Resnick MI, MacLennan GT, Gupta S. Experimental rodent models of prostatitis: limitations and potential. Prostate Cancer Prostatic Dis. 2007;10(1):15–29. [DOI] [PubMed] [Google Scholar]
- 7.Murphy SF, Schaeffer AJ, Done J, et al. IL17 Mediates Pelvic Pain in Experimental Autoimmune Prostatitis (EAP). PLoS One. 2015;10(5):e0125623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Roman K, Done JD, Schaeffer AJ, Murphy SF, Thumbikat P. Tryptase-PAR2 axis in experimental autoimmune prostatitis, a model for chronic pelvic pain syndrome. Pain. 2014;155(7):1328–1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Breser ML, Motrich RD, Sanchez LR, Rivero VE. Chronic Pelvic Pain Development and Prostate Inflammation in Strains of Mice With Different Susceptibility to Experimental Autoimmune Prostatitis. Prostate. 2017;77(1):94–104. [DOI] [PubMed] [Google Scholar]
- 10.Motrich RD, van Etten E, Depovere J, Riera CM, Rivero VE, Mathieu C. Impact of vitamin D receptor activity on experimental autoimmune prostatitis. J Autoimmun. 2009;32(2):140–148. [DOI] [PubMed] [Google Scholar]
- 11.Zaman T, Rife TL, Batki SL, Pennington DL. An Electronic Intervention to Improve Safety for Pain Patients Co-Prescribed Chronic Opioids and Benzodiazepines. Subst Abus. 2018:1–31. [DOI] [PubMed] [Google Scholar]
- 12.Vadivelu N, Kai AM, Kodumudi V, Sramcik J, Kaye AD. The Opioid Crisis: a Comprehensive Overview. Curr Pain Headache Rep. 2018;22(3):16. [DOI] [PubMed] [Google Scholar]
- 13.Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389(6653):816–824. [DOI] [PubMed] [Google Scholar]
- 14.Tominaga M, Caterina MJ, Malmberg AB, et al. The cloned capsaicin receptor integrates multiple pain-producing stimuli. Neuron. 1998;21(3):531–543. [DOI] [PubMed] [Google Scholar]
- 15.Davis JB, Gray J, Gunthorpe MJ, et al. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature. 2000;405(6783):183–187. [DOI] [PubMed] [Google Scholar]
- 16.Lazzeri M, Vannucchi MG, Zardo C, et al. Immunohistochemical evidence of vanilloid receptor 1 in normal human urinary bladder. Eur Urol. 2004;46(6):792–798. [DOI] [PubMed] [Google Scholar]
- 17.Stander S, Moormann C, Schumacher M, et al. Expression of vanilloid receptor subtype 1 in cutaneous sensory nerve fibers, mast cells, and epithelial cells of appendage structures. Exp Dermatol. 2004;13(3):129–139. [DOI] [PubMed] [Google Scholar]
- 18.Pecze L, Josvay K, Blum W, et al. Activation of endogenous TRPV1 fails to induce overstimulation-based cytotoxicity in breast and prostate cancer cells but not in pain-sensing neurons. Biochim Biophys Acta. 2016;1863(8):2054–2064. [DOI] [PubMed] [Google Scholar]
- 19.Islas L, Szallasi A. Manipulating TRPV1 antagonists: how to cool down a hot molecule? Acta Physiol (Oxf). 2018:e13088. [DOI] [PubMed] [Google Scholar]
- 20.Mayorga AJ, Flores CM, Trudeau JJ, et al. A randomized study to evaluate the analgesic efficacy of a single dose of the TRPV1 antagonist mavatrep in patients with osteoarthritis. Scand J Pain. 2017;17:134–143. [DOI] [PubMed] [Google Scholar]
- 21.Iadarola MJ, Sapio MR, Raithel SJ, Mannes AJ, Brown DC. Long-term pain relief in canine osteoarthritis by a single intra-articular injection of resiniferatoxin, a potent TRPV1 agonist. Pain. 2018;159(10):2105–2114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Payne CK, Mosbaugh PG, Forrest JB, et al. Intravesical resiniferatoxin for the treatment of interstitial cystitis: a randomized, double-blind, placebo controlled trial. J Urol. 2005;173(5):1590–1594. [DOI] [PubMed] [Google Scholar]
- 23.Sjogren E, Kullenberg T, Jonzon B, et al. Clinical testing of three novel transient receptor potential cation channel subfamily V member 1 (TRPV1) antagonists in a pharmacodynamic intradermal capsaicin model. Eur J Pain. 2018. [DOI] [PubMed] [Google Scholar]
- 24.Leo M, Schulte M, Schmitt LI, Schafers M, Kleinschnitz C, Hagenacker T. Intrathecal Resiniferatoxin Modulates TRPV1 in DRG Neurons and Reduces TNF-Induced Pain-Related Behavior. Mediators Inflamm. 2017;2017:2786427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Watabiki T, Kiso T, Tsukamoto M, Aoki T, Matsuoka N. Intrathecal administration of AS1928370, a transient receptor potential vanilloid 1 antagonist, attenuates mechanical allodynia in a mouse model of neuropathic pain. Biol Pharm Bull. 2011;34(7):1105–1108. [DOI] [PubMed] [Google Scholar]
- 26.Sjogren E, Stahle L, Quiding H, Jonzon B, Halldin MM, Sundgren AK. The effect of intradermal microdosing of a transient receptor potential cation channel subfamily V member 1 antagonist on heat evoked pain and thermal thresholds in normal and ultraviolet-C exposed skin in healthy volunteers. Eur J Pain. 2019. [DOI] [PubMed] [Google Scholar]
- 27.Chen Y, Willcockson HH, Valtschanoff JG. Vanilloid receptor TRPV1-mediated phosphorylation of ERK in murine adjuvant arthritis. Osteoarthritis Cartilage. 2009;17(2):244–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wakabayashi H, Wakisaka S, Hiraga T, et al. Decreased sensory nerve excitation and bone pain associated with mouse Lewis lung cancer in TRPV1-deficient mice. J Bone Miner Metab. 2017. [DOI] [PubMed] [Google Scholar]
- 29.Szitter I, Pozsgai G, Sandor K, et al. The role of transient receptor potential vanilloid 1 (TRPV1) receptors in dextran sulfate-induced colitis in mice. J Mol Neurosci. 2010;42(1):80–88. [DOI] [PubMed] [Google Scholar]
- 30.Done JD, Rudick CN, Quick ML, Schaeffer AJ, Thumbikat P. Role of mast cells in male chronic pelvic pain. J Urol. 2012;187(4):1473–1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Nickel JC, True LD, Krieger JN, Berger RE, Boag AH, Young ID. Consensus development of a histopathological classification system for chronic prostatic inflammation. BJU Int. 2001;87(9):797–805. [DOI] [PubMed] [Google Scholar]
- 32.Caterina MJ, Leffler A, Malmberg AB, et al. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science. 2000;288(5464):306–313. [DOI] [PubMed] [Google Scholar]
- 33.Marsch R, Foeller E, Rammes G, et al. Reduced anxiety, conditioned fear, and hippocampal long-term potentiation in transient receptor potential vanilloid type 1 receptor-deficient mice. J Neurosci. 2007;27(4):832–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wei NN, Lv HN, Wu Y, et al. Selective Activation of Nociceptor TRPV1 Channel and Reversal of Inflammatory Pain in Mice by a Novel Coumarin Derivative Muralatin L from Murraya alata. J Biol Chem. 2016;291(2):640–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wong L, Done JD, Schaeffer AJ, Thumbikat P. Experimental autoimmune prostatitis induces microglial activation in the spinal cord. Prostate. 2015;75(1):50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Fernandes ES, Liang L, Smillie SJ, et al. TRPV1 deletion enhances local inflammation and accelerates the onset of systemic inflammatory response syndrome. J Immunol. 2012;188(11):5741–5751. [DOI] [PubMed] [Google Scholar]
- 37.Solis-Lopez A, Kriebs U, Marx A, et al. Analysis of TRPV channel activation by stimulation of FCepsilonRI and MRGPR receptors in mouse peritoneal mast cells. PLoS One. 2017;12(2):e0171366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Devos FC, Boonen B, Alpizar YA, et al. Neuro-immune interactions in chemical-induced airway hyperreactivity. Eur Respir J. 2016;48(2):380–392. [DOI] [PubMed] [Google Scholar]
- 39.Samivel R, Kim DW, Son HR, et al. The role of TRPV1 in the CD4+ T cell-mediated inflammatory response of allergic rhinitis. Oncotarget. 2016;7(1):148–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhou Y, Follansbee T, Wu X, et al. TRPV1 mediates inflammation and hyperplasia in imiquimod (IMQ)-induced psoriasiform dermatitis (PsD) in mice. J Dermatol Sci. 2018;92(3):264–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Caceres AI, Brackmann M, Elia MD, et al. A sensory neuronal ion channel essential for airway inflammation and hyperreactivity in asthma. Proc Natl Acad Sci U S A. 2009;106(22):9099–9104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang Y, Wang DH. TRPV1 ablation aggravates inflammatory responses and organ damage during endotoxic shock. Clin Vaccine Immunol. 2013;20(7):1008–1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lei Q, Pan XQ, Villamor AN, et al. Lack of transient receptor potential vanilloid 1 channel modulates the development of neurogenic bladder dysfunction induced by cross-sensitization in afferent pathways. J Neuroinflammation. 2013;10:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chiba T, Oka Y, Sashida H, et al. Vincristine-induced peripheral neuropathic pain and expression of transient receptor potential vanilloid 1 in rat. J Pharmacol Sci. 2017;133(4):254–260. [DOI] [PubMed] [Google Scholar]
- 45.Rossato MF, Rigo FK, Oliveira SM, et al. Participation of transient receptor potential vanilloid 1 in paclitaxel-induced acute visceral and peripheral nociception in rodents. Eur J Pharmacol. 2018;828:42–51. [DOI] [PubMed] [Google Scholar]
- 46.Labuz D, Spahn V, Celik MO, Machelska H. Opioids and TRPV1 in the peripheral control of neuropathic pain--Defining a target site in the injured nerve. Neuropharmacology. 2016;101:330–340. [DOI] [PubMed] [Google Scholar]
- 47.Laursen WJ, Schneider ER, Merriman DK, Bagriantsev SN, Gracheva EO. Low-cost functional plasticity of TRPV1 supports heat tolerance in squirrels and camels. Proc Natl Acad Sci U S A. 2016;113(40):11342–11347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Marche P, Dubois S, Abraham P, et al. Neurovascular microcirculatory vasodilation mediated by C-fibers and Transient receptor potential vanilloid-type-1 channels (TRPV 1) is impaired in type 1 diabetes. Sci Rep. 2017;7:44322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Schwartz ES, La JH, Scheff NN, Davis BM, Albers KM, Gebhart GF. TRPV1 and TRPA1 antagonists prevent the transition of acute to chronic inflammation and pain in chronic pancreatitis. J Neurosci. 2013;33(13):5603–5611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chen Y, Yang C, Wang ZJ. Proteinase-activated receptor 2 sensitizes transient receptor potential vanilloid 1, transient receptor potential vanilloid 4, and transient receptor potential ankyrin 1 in paclitaxel-induced neuropathic pain. Neuroscience. 2011;193:440–451. [DOI] [PubMed] [Google Scholar]
- 51.Yang J, Hsieh CL, Lin YW. Role of Transient Receptor Potential Vanilloid 1 in Electroacupuncture Analgesia on Chronic Inflammatory Pain in Mice. Biomed Res Int. 2017;2017:5068347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lapointe TK, Basso L, Iftinca MC, et al. TRPV1 sensitization mediates postinflammatory visceral pain following acute colitis. Am J Physiol Gastrointest Liver Physiol. 2015;309(2):G87–99. [DOI] [PubMed] [Google Scholar]
- 53.Choi JY, Lee HY, Hur J, et al. TRPV1 Blocking Alleviates Airway Inflammation and Remodeling in a Chronic Asthma Murine Model. Allergy Asthma Immunol Res. 2018;10(3):216–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Keeble J, Russell F, Curtis B, Starr A, Pinter E, Brain SD. Involvement of transient receptor potential vanilloid 1 in the vascular and hyperalgesic components of joint inflammation. Arthritis Rheum. 2005;52(10):3248–3256. [DOI] [PubMed] [Google Scholar]
- 55.Mori T, Saito K, Ohki Y, Arakawa H, Tominaga M, Tokuyama K. Lack of transient receptor potential vanilloid-1 enhances Th2-biased immune response of the airways in mice receiving intranasal, but not intraperitoneal, sensitization. Int Arch Allergy Immunol. 2011;156(3):305–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhong B, Rubinstein J, Ma S, Wang DH. Genetic ablation of TRPV1 exacerbates pressure overload-induced cardiac hypertrophy. Biomed Pharmacother. 2018;99:261–270. [DOI] [PubMed] [Google Scholar]
- 57.Romac JM, McCall SJ, Humphrey JE, Heo J, Liddle RA. Pharmacologic disruption of TRPV1-expressing primary sensory neurons but not genetic deletion of TRPV1 protects mice against pancreatitis. Pancreas. 2008;36(4):394–401. [DOI] [PubMed] [Google Scholar]
- 58.van Wanrooij SJ, Wouters MM, Van Oudenhove L, et al. Sensitivity testing in irritable bowel syndrome with rectal capsaicin stimulations: role of TRPV1 upregulation and sensitization in visceral hypersensitivity? Am J Gastroenterol. 2014;109(1):99–109. [DOI] [PubMed] [Google Scholar]
- 59.Weidner N, Austen KF. Heterogeneity of mast cells at multiple body sites. Fluorescent determination of avidin binding and immunofluorescent determination of chymase, tryptase, and carboxypeptidase content. Pathol Res Pract. 1993;189(2):156–162. [DOI] [PubMed] [Google Scholar]
- 60.Bradding P, Okayama Y, Howarth PH, Church MK, Holgate ST. Heterogeneity of human mast cells based on cytokine content. J Immunol. 1995;155(1):297–307. [PubMed] [Google Scholar]
- 61.Dwyer DF, Barrett NA, Austen KF, Immunological Genome Project C. Expression profiling of constitutive mast cells reveals a unique identity within the immune system. Nat Immunol. 2016;17(7):878–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hensellek S, Brell P, Schaible HG, Brauer R, Segond von Banchet G. The cytokine TNFalpha increases the proportion of DRG neurones expressing the TRPV1 receptor via the TNFR1 receptor and ERK activation. Mol Cell Neurosci. 2007;36(3):381–391. [DOI] [PubMed] [Google Scholar]
- 63.Tsuji F, Aono H. Role of transient receptor potential vanilloid 1 in inflammation and autoimmune diseases. Pharmaceuticals (Basel). 2012;5(8):837–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Horvath A, Tekus V, Bencze N, et al. Analgesic effects of the novel semicarbazide-sensitive amine oxidase inhibitor SZV 1287 in mouse pain models with neuropathic mechanisms: Involvement of transient receptor potential vanilloid 1 and ankyrin 1 receptors. Pharmacol Res. 2018;131:231–243. [DOI] [PubMed] [Google Scholar]
