research article

Role of Transient Receptor Potential Channels Trpv1 and Trpm8 in Diabetic Peripheral Neuropathy

Mallikarjuna R Pabbidi1, Louis S Premkumar2* 

1Department of Pharmacology University of Mississippi Medical Center, Jackson, USA

2Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA

*Corresponding author: Louis S Premkumar, Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA. Tel: +12175452179; Fax: +12175450145; Email: lpremkumar@siumed.edu 

Received Date: 21 September, 2017; Accepted Date: 12 October, 2017; Published Date: 17 October, 2017

 

Citation: Pabbidi MR, Premkumar LS (2017) Role of Transient Receptor Potential Channels Trpv1 and Trpm8 in Diabetic Peripheral Neuropathy. J Diabetes Treat: 129. DOI: 10.29011/2574-7568.000029

1.      Abstract

1.1.  Objective: Transient Receptor Potential (Vanilloid 1) TRPV1 and (Melastatin 8) TRPM8 are heat and cold sensing non-selective cation channels, respectively. We sought to correlate the modulation of TRPV1- and TRPM8-mediated membrane currents and altered thermal sensitivity in Diabetic Peripheral Neuropathy (DPN). 

1.2.  Method: Streptozotocin (STZ)-induced diabetic mice were used and thermal (heat and cold) pain sensitivities were determined using hot plate and acetone drop test, respectively. Membrane currents were recorded using patch-clamp techniques.

1.3.  Results: First, we tested thermal pain sensitivities to implicate possible role of TRPV1 and TRPM8 in DPN. Paw withdrawal latency on a hot plate test was decreased and acetone-induced cold sensitivity was enhanced in diabetic mice as compared to non-diabetic mice. Dorsal Root Ganglion (DRG) neurons dissociated from diabetic hyperalgesic mice exhibited an increase in TRPV1-mediated current and a decrease in TRPM8-mediated currents as compared to non-diabetic mice. Then, we determined the modulation of TRPV1- and TRPM8-mediated currents using HEK cells heterologously expressing TRPV1 by promoting of PKC- and PKA-mediated phosphorylation. Both Phorbol 12,13-Dibutyrate (PDBu), a PKC activator and forskolin, a PKA activator upregulated TRPV1-mediated currents, but downregulated TRPM8-mediated currents. In diabetic mice, intraplantar injection of capsaicin, a TRPV1 agonist induced nocifensive behavior but the severity of this behavior was significantly lower when co-administered with menthol, a TRPM8 agonist.

1.4.  Conclusions: These findings suggest that diabetic thermal hyperalgesia mediated by up regulation of TRPV1 function may be further aggravated by the down regulation of TRPM8 function. Targeting TRPV1 may be a useful approach to alleviate pain associated with DPN.

Keywords: Capsaicin; Diabetic Peripheral Neuropathy; Menthol; TRPV1; TRPM8

1.      Introduction 

One of the major complications of diabetes is Diabetic Peripheral Neuropathy (DPN) [1]. The symptoms of DPN range from spontaneous pain, allodynia, hyperalgesia, numbness, dysesthesia, paresthesia, and ulceration in the extremities [2-5]. Recent studies [6,7], including our own [8] suggest that specific types of neurons carry particular sensation and this distinction occurs at the level of peripheral sensory neurons. A subset of small and medium diameter sensory neurons (C and A) respond solely to noxious stimuli. It is becoming apparent that C-fibers that express TRPV1 carry inflammatory thermal hypersensitivity, whereas a population of non-TRPV1 expressing neurons that express the G-protein coupled receptor MrgprD carry mechanical sensitivity [9]. On the other hand, TRPM8 which was previously called menthol and cold receptor 1 (CMR1) was shown to play a role in cold sensation and is activated by temperatures lower than ~25-28oC, cooling agents such as menthol and icilin [10-14]. Activation of either TPRV1 or TRPM8 allows flux of Na+ and Ca2+, resulting in the membrane depolarization and tachyphylaxis. 

Previous studies using STZ-induced diabetic models indicate that there are two pain phenotypes in rodents with hyperalgesia followed by hypoalgesia [15-17]. Studies suggest that hyperalgesia phenotype is characterized by the greater release of capsaicin-evoked CGRP from the hind paw [18], increased TRPV1 activity in primary sensory neurons of the spinal cord and dorsal root ganglions [15,18]. However, the role of TRPM8 in diabetic neuropathy is not known. Both TRPV1 and M8 are modulated by cellular pH, PIP2, PKC and PKA altering threshold of activation and in turn affecting the sensitization of neuron. Menthol-induced TRPM8 sensitization in the presence of acidic pH, PIP2 and PKC was reduced whereas capsaicin-induced TRPV1 sensitization was increased [19-21]. These studies suggest that inflammatory mediators while up regulating TRPV1 activity might down regulate TRPM8 activity aggravating the pain condition resulting in hyperalgesia phenotype [21]. Localized and topical treatments are becoming recognized treatment options for painful conditions. Recently, high dose of capsaicin (8%) transdermal patch, which is a localized treatment that provides effective durable pain relief from single application in patients with peripheral neuropathic pain [22-25]. In a direct comparison, the capsaicin 8% patch has been shown to be noninferior to pregabalin in the control of neuropathic pain but has a faster onset of analgesia with fewer systemic side effects [25,27]. 

The primary aim of this study is to investigate the molecular mechanisms underlying deficiencies in temperature sensation in diabetic mice. We have used STZ-induced diabetic mouse models to identify altered heat and cold sensitivities. We have measured TRPV1 and TRPM8 channel currents and their modulation by PKC and PKA in the presence and absence of extracellular calcium from dorsal root ganglion neurons. Our results suggest that in the first week of diabetes mice exhibited increased thermal sensitivities to both heat and cold. Diabetic thermal hyperalgesia is associated with upregulation of TRPV1 but down regulation of TRPM8 activity. Furthermore, PKC- and PKA-mediated modulation of TRPV1 and TRPM8 alter thermal sensitivities was confirmed using heterologous expression system. 

2.      Materials and Methods 

2.1.  Induction of Diabetes by STZ 

The experiments were performed according to the National Institutes of Health guidelines and approved by the animal welfare committee of Southern Illinois University School of Medicine. Age-matched mice were used for induction of diabetes with STZ. STZ (Sigma, St Louis) was prepared fresh in saline adjusted to pH 4.5 with 0.1N citrate buffer [28]. A single intraperitoneal injection of STZ (200mg/kg) in mice induced diabetes in 38 of 51 mice. 0.1 N citrate buffer injected mice served as control. Glucose levels were quantitated with a One Touch Ultra blood glucose monitoring system (Life Scan, California) using whole blood obtained from the tail. Diabetes was defined as blood glucose concentrations greater than 299 mg/dl (16.7 mM) [22,28]. 

2.2.  Hot Plate Test for Thermal Sensitivity in Mice 

All the mice used in this study were housed in the barrier facility and were tested on the days the cages were not cleaned. Mice were placed individually on a Hot Plate Analgesia Meter (Harvard Apparatus, Boston, MA) maintained at a constant temperature of 52±0.3oC after observing them for 5 five minutes in the cage. The Paw Withdrawal Latency (PWL) was recorded as described previously [15]. These studies were blinded and after completing the test, the ear tags were read to place them in the appropriate groups. Since PWL of male and female mice did not have significant differences, the data were combined. 

2.3.  Acetone Test for Cold Sensation in Mice 

Acetone drop (50-70µL) was applied on the left plantar surface of the mice placed on a metal wire mesh enclosed in a chamber having a perforated roof. Acetone bubble was formed at the tip of the 1cc syringe and gently applied while the animal stood on the mesh. Foot lift frequency (flicking, licking or biting the limb) was counted in a period of 5 minutes after administering acetone on the plantar surface of the paw. This procedure was repeated for three times with a gap 10minutes. The average of the responses obtained from 3 acetone applications was taken as foot lift frequency and plotted against days of testing [29,30]. 

2.4.  Measurement of Nocifensive Behavior 

Nocifensive behavior was examined in mice after intraplantar injection of TRPV1 and TRPM8 agonists, capsaicin (2 mM), and menthol (5 mM), respectively. 20µL of the drug was injected into the hind limb paw by using Hamilton syringe (Hamilton Company, Reno, Nevada, USA). Nocifensive behavior was characterized by the latency, duration, and number of licking and/or shaking of the injected hindlimb.

2.5.  Adult DRG Neuronal Cultures 

DRG were dissected from diabetic and control mice euthanized by deep anesthesia with isoflurane followed by decapitation. Isolated DRG were collected in HBSS (calcium- and magnesium-free) on the ice and then enzymatically digested for 45 minutes at 37°C in 0.1% collagenase D, Worthington type 2 (Roche Diagnostics, Indianapolis, IN) and 0.1% trypsin, type 1 (Sigma-Aldrich, St. Louis, MO). After trituration using fire-polished Pasteur pipettes, DRG were plated on 12 mm poly D-lysine coated glass coverslips placed in 24 well plates and grown in neurobasal medium (Gibco BRL, Grand Island, NY) supplemented with L-glutamine (2 mM, Invitrogen, Grand Island, NY) and 10 l/ml B27-supplement (Gibco, Invitrogen corporation, GrandIsland, N.Y). Neurons were maintained at 37°C in a humidified atmosphere of 5% CO2 and were used within 8-10 hours. 

2.6.  HEK 293 Cells Culture 

HEK293 cells, stably expressing rat TRPM8 cDNA, were obtained from Dr D. Julius (University of California, San Francisco, San Francisco, CA) and cultured in growth medium containing 90% DMEM, 10% fetal bovine serum, 50 U/ml penicillin, 25 μg/ml streptomycin (all from Invitrogen, Grand Island, NY). Cells were grown at 37°C, in the presence of 5% CO2 and 95% ambient air. Cells were cultured in the presence of G418 sulfate, neomycin-resistant cells were expanded, and frozen stocks were kept in liquid nitrogen. 

2.7.  Whole Cell Patch Clamp Recording

Whole-cell patch-clamp recordings were performed in DRGs that were isolated from age matched diabetic and control mice. The bath and pipette were filled with sodium based solution and were prepared as described previously [15,21]. Ca2+-free solutions were used to avoid desensitization and tachyphylaxis of capsaicin-induced currents. Currents were recorded using a WPC 100 patch-clamp amplifier (E.S.F. Electronic, Goettingen, Germany) using borosilicate pipettes as described previously [15,21]. Capacitance compensation and series resistance compensation (>70%) were done before recording currents. Membrane capacitance as obtained from the compensations and also calculated by determining the to an applied square pulse (=RC). Data were filtered at 2.5kHz, digitized at 5 kHz (VR-10B; Instrutech, Great Neck, NY) and stored on the computer using a Labview interface (National Instruments, Austin, TX). Current amplitudes were measured using Channel 2 (software kindly provided by Michael Smith, Australian National University, Canberra, Australia). The traces and graphs were made using Origin (Microcal Software, Northampton, MA). 

2.8.  Data Analysis 

For behavioral experiments, mixed model analysis was performed using SAS software, which includes both fixed events (age and time after diabetes onset) and random events (number of subjects) with repeated measures of ANOVA. The comparisons were made between control group and diabetic groups. Data are shown as mean±S.E.M. (standard Error of Mean). The current amplitudes were normalized and expressed as fold change as compared to control responses. Significance was tested using unpaired Student’s t-test and the data are considered significant at p<0.05.  

All the chemicals used in this study were obtained from Sigma (St. Louis, MO). 

3.      Results

3.1.  Altered Heat and Cold Pain Sensitivity Following STZ-Induced Diabetes 

Blood glucose levels were significantly elevated by 1 week after STZ treatment and remained elevated during the course of the study (pre- STZ injection, 187±2.7 mg/dl; day 12, 532.5±23.9 mg/dl; n=10 mice, p<0.001) as compared to vehicle injected mice (pre-vehicle injection, 178.8±2.2 mg/dl; day 12, 194.6±12.5 mg/dl, n=10 mice) (Figure 1A). As disease progressed, body weights of STZ-treated diabetic mice decreased, whereas the body weights of control animals increased steadily (diabetic mice; pre- STZ injection, 22.1±0.3 gms; day 12, 20.8±0.7 gms; n=10 mice) (pre-vehicle injection, 22.7±0.6 gms; day 12, 23.7±0.4 gms, n=10 mice, p<0.001) (Figure 1B). 

Diabetic mice tested on hotplate were hyperalgesic within a week indicated by a decrease in PWL (prediabetic 9.6±0.3s; day 12, 7.1±0.4s, n=33, p<0.001 compared with control group). Vehicle injected mice did not show altered thermal pain sensitivity (pretreatment 9.4±0.3 s; day 12, 9±0.3s) (Figure 1C). Following acetone administration, the number of flickerings and lickings was significantly higher in diabetic mice (day 0, 11.7±0.6; day 12, 17.8±1.4, n=10 mice, p<0.01) as compared to non-diabetic mice (day 0, 10.7±0.4; day 12, 11.7±1.3, n=10 mice) (Figure 1D).

3.2.  Altered TRPV1 Currents in Diabetic Hyperalgesia

The peak currents in response to application of capsaicin (1 μM) were significantly larger (3.74±0.27 fold, n=5 cells from 2 mice, p<0.03) in hyperalgesic mice as compared to control mice (1±0.12 fold n=12 cells from 4 mice) (Figure 2A, B). 

In order to take the cell size into consideration, we normalized the current to the cell capacitance. The current density is found to be significantly higher in hyperalgesic (2.81±0.19 fold, n=5, p<0.05) as compared to normal non-diabetic mice (1±0.18 fold n=12) (Figure 2C).

3.3.  Altered TRPM8 Currents in Diabetic Hyperalgesia 

In order to determine whether TRPM8 channel function is altered in diabetes-induced hyperalgesia, we recorded 1mM menthol-induced TRPM8 whole cell currents from acutely dissociated DRG from diabetic hyperalgesic and non-diabetic mice. In contrast to our hypothesis, menthol-induced TRPM8-mediated peak currents were significantly smaller (0.54±0.13 fold, n=7 cells from 2 mice, p<0.03) (Figure 3A and B) in diabetic hyperalgesic mice as compared to non-diabetic mice (1±0.09 fold, n=6 cells from 4 mice). When the currents were normalized to the capacitance of the cells, we noticed a similar decrease (0.33±0.15 fold, n=7, p<0.05) as compared to normal non-diabetic mice (1±0.09 fold n=6) (Figure 2C).

3.4.  Modulation of TRPV1 by PKC 

To determine the modulation by PKC, we used HEK cells heterologously expressing TRPV1. First, whole cell currents were recorded in the presence (2mM calcium) and absence of calcium (1.5mM EGTA) as it has been shown that TRPV1 currents undergo desensitization/tachyphylaxis (–Ca2+, pre 1±0.13, n = 4 cells; post, 0.96±0.04 fold, n = 4 cells; +Ca2+, pre 1±0.1, n = 4 cells; post, 0.37±0.04 fold, n = 4 cells) (Figure 4). We recorded currents before and after activation of PKC by incubating cultured DRG neurons for 5 minutes with PKC activator PDBu (1µM). Both in the absence and presence of calcium, PKC activation potentiated capsaicin-induced (100nm) TRPV1-mediated currents (-Ca2+ pre PDBu 1±0.1, n = 8 cells; post PDBu 2.33±0.26 fold, n = 8 cells, p<0.05; +Ca2+ pre PDBu 1±0.1, n = 8 cells; post PDBu 2.2±0.3 fold, n = 8 cells, p<0.05) (Figure 5 A, B, C).

3.5.  Modulation of TRPV1 by PKA

In our studies we questioned whether TRPV1 sensitization by PKA activation plays a role in diabetic hyperalgesia. To address this question, TRPV1-mediated whole cell currents were recorded in cultured DRG neurons incubated in the presence of an adenylyl cyclase activator (FSK-forskolin, 20µM) and phosphodiesterase inhibitor (IBMX, 50µM) for 5 minutes. Forskolin activates adenylyl cyclase increasing the levels of cAMP, which in turn increases PKA activity and IBMX prevents the breakdown of cAMP by inhibiting the phosphodiesterase activity. Our results suggest that activation of PKA sensitizes TRPV1 activity both in the presence and absence of calcium (-Ca2+ , pre FSK+IBMX 1±0.13, n = 5 cells; -Ca2+ , Post FSK+IBMX, 1.89±0.43, n = 5 cells, p<0.05) (+Ca2+, pre FSK+IBMX, 1±0.13, n = 7 cells; +Ca2+ , Post FSK+IBMX, 2.45±0.5, n = 7 cells, p<0.05) (Figure 6 A, B, C).

3.6.  Modulation of TRPM8 by PKC 

In the presence of extracellular Ca2+, menthol induced currents in HEK cells heterologously expressing TRPM8 showed tachyphylaxis (-Ca2+, pre, 1±0.11, n = 3 cells; post, 0.91±0.04 fold, n = 3 cells; +Ca2+, pre 1±0.1, n = 4 cells; post, 0.67±0.09 fold, n = 4 cells) (Figure 7). To study the modulation of TRPM8-mediated currents by PKC, menthol-induced TRPM8 whole cell currents were recorded from HEK293 cells expressing TRPM8 following incubation with a PKC activator PDBu (1µM) for 5 minutes. We recorded currents both in the presence (2mM Ca2+) and absence of calcium (1.5mM EGTA), as Ca2+ induces desensitization/tachyphylaxis of TRPM8 induced currents (Figure 7). PKC activation decreases TRPM8 channel activity (-Ca2+ pre PDBu 1, n = 3; post PDBu, 0.44±0.13, n = 3, p<0.05; +Ca2+ pre PDBu 1, n = 4; post PDBu, 0.53±0.13, n = 4, p<0.05) (Figure 8A, B and C).

3.7.  Modulation of TRPM8 by PKA 

TRPM8 whole cell currents mediated by TRPM8 were recorded from HEK293 cells incubated (5 min) with an adenylyl cyclase activator (FSK-forskolin, 20µM) and phosphodiesterase inhibitor (IBMX, 50µM) (increases the levels of cAMP by preventing it from degradation by phosphodiesterases). Both in the presence and absence of calcium we noticed a decrease in TRPM8 activity (-Ca2+, pre FSK+IBMX, 1±0.04, n = 6 cells; post FSK+IBMX, 0.43±0.05, n = 6 cells, p<0.05) (+Ca2+, pre FSK+IBMX, 1±0.03, n = 3 cells; post FSK+IBMX, 0.53±0.17, n = 3 cells, p<0.05) (Figure 9 A, B and C). The current amplitude remained constant in experiments where the kinases were not activated either in the presence or absence of calcium (TRPM8; –Ca2+, pre wash 1±0.11, n = 3 cells; post wash, 0.91±0.04 fold, n = 3 cells; +Ca2+, pre wash 1±0.1, n = 4 cells; post wash, 0.67±0.09 fold, n = 4 cells).

3.8.  Capsaicin Alone or Combined with Menthol-Induced Nocifensive Behavior is Altered in Diabetic Mice 

To determine the physiological relevance of the interaction between TRPM8 and TRPV1, nocifensive behavior was examined after intraplantar injection of capsaicin or capsaicin with menthol. The latency of onset of pain, number and duration of flickerings and lickings, were recorded after intraplantar injection of 20 µl of capsaicin (2 mM) or capsaicin plus menthol (2mM and 5 mM, respectively) in non-diabetic and diabetic mice. 

In non-diabetic mice onset of pain symptoms such as licking and flickering was significantly increased from 3.07±0.29 s (n = 8 mice) after capsaicin alone to 10.7±0.8 s (n =8 mice) after capsaicin plus menthol. Similar results were obtained in diabetic mice also, although there is a reduction in onset of pain with both capsaicin (1.3±0.2 s, n = 8 mice) and capsaicin plus menthol (2.3±0.4 s, n = 8 mice) compared to non-diabetic mice (Figure 10A). The total duration of nocifensive behavior was also significantly decreased from 32.5±1.9 with capsaicin to 19.1±2.1 s with capsaicin plus menthol in non-diabetic mice. Similar decrease was noticed in diabetic mice (capsaicin, 62±7.5 s, n = 8 mice; capsaicin plus menthol, 39.5±7.8 seconds) although there was increase in duration of lickings and flickerings in diabetic mice (Figure 10B). The number of hind paw licks and flickerings was significantly decreased from 34±2.3 to 19.1±2.1 in non-diabetic mice and similar findings were noticed in diabetic mice (capsaicin, 60±7.3 s, n = 8 mice; capsaicin plus menthol, 35±4.1 sec n = 8 mice) although there were more number of lickings and flickerings in diabetic mice (Figure 10C).

Capsaicin-Induced Nocifensive Pain Behavior is Reduced by Co-administration with Menthol in Diabetic and Non-Diabetic Mice. Onset, duration, and number of lickings and/or flickerings of the injected hind paw were determined after intraplantar injection of 20µl of capsaicin (2 mM) or capsaicin plus menthol (5 mM) in diabetic and non-diabetic mice. A. In non-diabetic mice onset of pain symptoms such as licking and flickering was significantly increased from 3.07±0.29 s (n = 8 mice) after capsaicin alone to 10.7±0.8 s (n =8 mice) after capsaicin plus menthol. Similar results were obtained in diabetic mice also although onset of pain with both capsaicin (1.3±0.2 s, n = 8 mice) and capsaicin plus menthol (2.3±0.4 s, n = 8 mice) is much quicker compared to non-diabetic mice. B. The total duration of nocifensive behavior was also significantly decreased from 32.5±1.9 with capsaicin to 19.1±2.1 s with capsaicin plus menthol in non-diabetic mice. Similar decrease was noticed in diabetic mice (capsaicin, 62±7.5 s, n = 8 mice; capsaicin plus menthol, 39.5±7.8 sec) although there was increased duration of lickings and flickerings in diabetic mice. C. We also compared the total number of hind paw lickings and flickerings for the duration of nocifensive behavior in diabetic and non-diabetic mice. The number of hind paw licks and flickerings was significantly decreased from 34±2.3 to 19.1±2.1 in non-diabetic mice and similar findings were noticed in diabetic mice (capsaicin, 60±7.3 s, n = 8 mice; capsaicin plus menthol, 35±4.1 sec n = 8 mice) although there was more number of lickings and flickerings in diabetic mice. Data are represented as mean±SEM. N = 8 mice per group. *p<0.05 obtained from students unpaired t-test. 

4.      Discussion

The present study identified the role of TRPV1 and TRPM8 and their reciprocal regulation by PKC and PKA in DPN in STZ-induced diabetic mice. Our results suggest that STZ-induced mice exhibit heightened sensitivity to both heat and cold. There was an increase in TRPV1-mediated currents but a decrease in TRPM8-mediated currents in DRG isolated from diabetic hyperalgesic mice. While both PKA and PKC increased TRPV1 activity, they decreased TRPM8 channel activity. Finally, intraplantar injection of capsaicin exaggerated diabetic pain but was ameliorated when combined with menthol. 

4.1.  Diabetic Peripheral Neuropathy and the Role of TRPV1 and TRPM8 

Diabetic peripheral neuropathy, a disorder characterized by altered thermal, mechanical, and chemical sensation, is the most common chronic complication of diabetes mellitus. The symptoms of DPN can be categorized as either painful or painless [31-35]. In animal models of diabetes, hypersensitivity occurs at an earlier stage following the onset of diabetes as compared to hyposensitivity, which occurs at a later stage [15-17]. Although measurement of heat-induced pain sensitivity using hot plate analgesia meter is well established, methods to study cold-induced pain are not clear. We used acetone test to measure cold pain sensitivity in which a drop of acetone is placed on the hind paw and responses (number of lickings and flickerings) is measured [29,30,35]. Consistent with previous studies, we observed a marked increase in sensitivity to acetone application indicating cold hyperalgesia following diabetes [36-38]. However, several caveats surround the interpretation of behavioral responses evoked by acetone, namely the influence of mechanical stimulation during application, possible direct chemical effects of acetone on skin, the effect of ambient temperature on the rate of evaporation, and the prospect of olfactory cues accompanying application involvement of other cold sensitive channels [39]. In our studies influence of mechanical sensation was ruled out as saline application did not induce any nocifensive behavior. 

The ability to sense hot and cold distinctly suggests the presence of ion channels in specialized neurons within the peripheral nervous system. TRPV1 is involved in detecting noxious heat stimuli and is found in DRG neurons [40]. Much less is known about channels that sense cold. However, cloning of a menthol- and cold-activated channel, TRPM8 (CMR1), its activation at 25°C, and by menthol products suggests a discovery of cold sensing ion channel [13,14]. By using knock out strategy it has been shown that both TRPM8 and TRPA1 plays a role in inflammation or chronic constrictive injury (CCI) or acetone-induced hypersensitivity and activation of which relieves painful condition [29,30,35,37]. Although diabetic animals exhibited cold hyperalgesia, we noticed a reduction in menthol-induced TRPM8 currents and increased capsaicin-mediated TRPV1 currents as compared with non-diabetic mice. These results demonstrate that a decrease in TRPM8 and increase in TRPV1 function might aggravate the hyperalgesia observed in early stages of diabetes. Extracellular calcium has been shown to modulate TRP channels. We have demonstrated in this study that extracellular calcium reduces both TRPV1 and TRPM8 currents with repeated application a phenomenon known as tachyphylaxis. Therefore, we have compared the differences in the TRPV1 and TRPM8 currents in the presence or absence of calcium while studying the role of PKC or PKA mediated phosphorylation using HEK cells heterologously expressing TRPV1(Figsure 4-9). 

4.2.  Modulation of TRPM8 and TRPV1 Function by PKC and PKA

Our findings support that both PKC and PKA upregulate TRPV1 function suggesting that hyperalgesia noticed in early stages of diabetic neuropathy is due to enhanced activity of TRPV1 function. These results are in line with previous studies that have suggested that an elevated activity of PKC and cAMP-PKA pathway may underlie the mechanism involved in hyperalgesia noticed in early stages of diabetes [41,43]. Also, diabetic hyperalgesia is attenuated by PKC inhibitors in vivo [44]. Specific blockade of PKCe diminishes PKC-mediated enhancement of heat-induced currents in sensory neurons and epinephrine-induced hypersensitivity in vivo [45]. PKCe knockout mice exhibit reduced hyperalgesia after intracutaneous injection of epinephrine and nerve growth factor [46]. Furthermore, increased enzyme activity from hyperglycemia, specifically PKCβ, is implicated in several diabetic complications including thermal hyperalgesia [46,47]. On the other hand, potentiation of TRPV1 activity by PKC has been associated with unmasking of silent channels [48,49], increased channel gating [50,51] and the recruitment of vesicular receptors to the plasma membrane by regulated exocytosis [52].

Studies demonstrate that proinflammatory prostaglandin E2 (PGE2) induces sensitization of sensory neuron responses to heat (heat hyperalgesia) by activating Gs-coupled prostaglandin E (EP) receptor subtypes (EP3C and EP4) and subsequently the cAMP/PKA cascade [53]. Accordingly, sensitization to heat also occurs in the presence of membrane-permeant cAMP analogs activating PKA [54]. Moreover, mice carrying a null mutation for type I b PKA regulatory subunit (PKA-RI b) no longer exhibit increased heat-induced pain behavior after PGE2 administration, suggesting a crucial role of the cAMP/PKA second-messenger system in Gs-mediated hyperalgesia [55]. PKA induced TRPV1 potentiation was shown to be due to increase in translocation of catalytic PKA subunit to the plasma membrane tethered to AKAP (A-kinase anchoring protein) [56]. It was also shown that activation of PKA increases the gating of the TRPV1 channel through direct phosphorylation [57, 58]. In conclusion, sensitization of TRPV1 by PKC and PKA enhances its activity, which may result in increased sensory neuron excitability leading to the exaggerated pain sensation. 

Our findings indicate a down regulation of TRPM8 by PKC- and PKA-mediated phosphorylation, which in turn reveals that hyperalgesia noticed in early stages of diabetic neuropathy may be due to enhanced activity of TRPV1 as a result of impaired activity of TRPM8. PKC mediated phosphatase (PP1) activity was shown to involve in dephosphorylation of TRPM8 [21]. Furthermore, we suggest that inflammatory mediators while up regulating TRPV1 might down regulate TRPM8 aggravating the pain condition resulting in hyperalgesia phenotype [21]. However, mechanisms that downregulate TRPM8 activity are not clear. The events such as decreased membrane fraction, decreased phosphorylation, and decreased vesicular release of TRPM8 channels might be associated with decreased PKC-induced activity of TRPM8. PKA induced desensitization of TRPM8 might be due to either reduced gating or decreased translocation of TRPM8 and requires further investigation.

4.3.  Reciprocal Interaction of TRPV1 and TRPM8 Contributes to Diabetic Hyperalgesia 

Our studies suggest that upon activation of PKA or PKC in vitro, TRPV1-mediated currents were upregulated and TRPM8-mediated currents were down regulated. It has been shown that intraplantar injection of capsaicin induces severe pain and this pain is less severe when co-administered with menthol, suggesting a reciprocal interaction of TRPV1 and TRPM8 [21]. We noticed a similar decrease in nocifensive behavioral reflexes in diabetic mice treated with capsaicin plus menthol when compared with mice treated with capsaicin alone. In heterologous expression systems, TRPM8 is activated, whereas TRPV1 is tonically inhibited by PIP2 [20]. Acidification (pH<6.5) of neurons leads to potentiation of TRPV1 and desensitization of TRPM8. Potentiation of TRPV1 results in lowered thermal threshold activation, whereas inhibition of TRPM8 leads to an increase in thermal threshold of activation [19-21,48-61]. In normal conditions, TRPM8, which is almost exclusively expressed in a subset of neurons, may mediate a cool/soothing sensation and could function to counter the painful behavior via activation of TRPV1. In chronic pain conditions, elevated NGF could induce co-expression of TRPM8 and TRPV1 in the same neuron that may jeopardize the counter-balance mechanism we are proposing. Overall our studies support the hypothesis that diabetic hyperalgesia is associated with elevated TRPV1 activity and this hyperalgesia may be exacerbated by the decrease in TRPM8 function as the latter contributes to much needed soothing sensation. TRPV1 expressed at peripheral terminals can be targeted to treat painful peripheral neuropathies [23-27]. 

5.      Conclusions 

The present study identified the role of TRPV1 and TRPM8 and their reciprocal regulation by PKC and PKA in DPN in STZ-induced diabetic mice. Our results suggest that STZ-induced diabetic mice exhibit heightened sensitivity to both heat and cold. There was an increase in TRPV1-mediated currents but a decrease in TRPM8-mediated currents in DRG neurons isolated from diabetic hyperalgesic mice. While both PKA- and PKC-mediated phosphorylation increased TRPV1 channel activity, but they decreased TRPM8 channel activity. Finally, intraplantar injection of capsaicin exaggerated diabetic pain but was ameliorated when combined with menthol. Therefore, targeting TRPV1 or TRPM8 could be a useful approach to treat pain associated with DPN.

6.      Acknowledgements 

This work was supported with grants from NIH (DK065742, NS042296 to L.S.P).

 


Figure 1(A-D): Blood Glucose Levels, Body Weight Changes, Heat and Cold Pain Sensitivities in STZ-Induced Diabetic Mice.A. Blood glucose levels of STZ-injected diabetic mice were significantly increased (filled circles, n = 10 mice, p<0.01) in a week and remained constant throughout the course of the experiment as compared to non-diabetic mice (open circles, n = 10 mice) B. Body weights of diabetic mice decreased (filled circles, n = 10 mice) as compared to non-diabetic mice (open circles, n = 10 mice). C, D. Bar graph of the PWL (C) and number of flickerings and lickings (D) in the first week of diabetes compared to non-diabetic mice. *p < 0.05, **p < 0.001 compared with control group. 




Figure 2(A-D): Diabetic Hyperalgesic Mice Exhibit Increased TRPV1 Currents. A. Representative current traces of increased 1µM capsaicin-evoked TRPV1 currents in diabetic hyperalgesic mice as compared to control mice. B, C. Summary graph showing fold change in increased TRPV1 peak currents and current densities (non-diabetic: n=12 cells; diabetic: n=5 cells) in diabetic hyperalgesic mice as compared to non-diabetic control mice. Number in the parenthesis represents the number of DRG neurons studied and the asterisk (*) represents p<0.05 obtained from students unpaired t-test. 



Figure 3(A-C): Diabetic Hyperalgesic Mice Exhibit Reduced TRPM8 Currents. A. Representative traces showing reduced 1mM Menthol-evoked TRPM8 currents in DRG of diabetic hyperalgesic mice as compared to control mice. B, C. Summary graph showing fold change in reduced TRPM8 peak currents and current densities (diabetic n=7 cells) in diabetic hyperalgesic mice as compared to non-diabetic mice (n = 6 cells). Number in the parenthesis represents the number of DRG neutond studied and the asterisks (*, **) represent p<0.05 and p<0.004 obtained from students unpaired t-test. 



Figure 4(A-C): TRPV1-Mediated Whole Cell Currents in the Absence and Presence of Calcium. A. Representative capsaicin (1µM)-evoked TRPV1 channel currents in the absence (left trace) and presence (right trace) of calcium (2 mM) (a gap of 5 min is denoted by a break in the trace). B. Repeated capsaicin applications did not show any change in the current amplitude in the absence of calcium (open circles, n=8 cells), but in the presence of calcium, there is gradual decrease in the current amplitude (tachyphylaxis) (closed circles, n=8 cells. C. A ramp (-80-+80 mv) current voltage relationship of capsaicin induced currents showing outward rectification. Data are represented as mean±SEM. 0 represents capsaicin application at time 0 and 1 to 6 represents number of capsaicin applications.



Figure 5(A-C): Activation of PKC Potentiates TRPV1-Mediated Whole Cell Currents both in the Absence and Presence of Calcium. A. Representative capsaicin (1µM)-evoked TRPV1 channel currents before and after phorbol 12,13-dibutyrate (PDBu) incubation (1µM for 5 min denoted by a break in the trace) in the absence (left trace) and presence (right trace) of calcium. B. Changes in repeated capsaicin-induced currents after PDBu incubation in the presence (filled circles, n = 8 cells) and absence of calcium (open circles, n = 8 cells). C. Summary graph showing capsaicin-induced currents are potentiated significantly after PDBu incubation (p < 0.05). Data are represented as mean±SEM. 0 represents capsaicin application before PDBu and 1 to 6 represents number of capsaicin applications after PDBu incubation. Number in the parenthesis represents the number of cells and the asterisk (*) represents p < 0.05. PDBu denotes Phorbol 12,13-dibutyrate.




Figure 6(A-C): Activation of PKA Potentiates TRPV1-Mediated Whole Cell Current both in the Absence and Presence of Calcium. A. Representative capsaicin (1µM )-evoked TRPV1 channel currents before and after 20µM FSK+ 50µM IBMX incubation (5 min denoted by a break in the trace) in the (1µM; 5 min denoted by a break in the trace) in the absence (left trace) and presence (right trace) of calcium B. repeated capsaicin applications induces currents after 20µM FSK+ 50µM IBMX incubation in the presence (filled circles, n = 7 cells) and absence of calcium (open circles, n = 5 cells) C. Summary graph showing capsaicin-induced currents are potentiated (p < 0.05) after 20µM FSK+ 50µM IBMX incubation and no change after wash application both in the presence and absence of calcium. 0 represents capsaicin application before FSK+IBMX and 1 to 5 represents number of capsaicin applications after FSK+IBMXC. Number in the parenthesis represents the number of cells studied and the asterisk (*) represents p < 0.05. FSK: Forskolin; IBMX: 3-isobutyl-1-methyl­xanthine. 



Figure 7: TRPM8-Mediated Whole Cell Current in the Absence and Presence of Calcium. A. Representative Menthol (1 mM)-evoked TRPM8 channel currents (a 5-min gap is denoted by a break in the trace) in the absence (left trace) and presence (right trace) of calcium (2 mM). B. Repeated menthol applications did not show any change in the current amplitude in the absence of calcium (open circles, n=3 cells), but in the presence of calcium, there is gradual decrease in the current amplitude (tachyphylaxis) (closed circles, n=4 cells. C. Current-voltage relationship of menthol induced currents showing outward rectification. Data are represented as mean±SEM. 0 represents menthol application at time 0 and 1 to 6 represents number of menthol applications. 



Figure 8(A-C): Activation of PKC Down Regulates TRPM8-Mediated Whole Cell Current both in the Absence and Presence of Calcium. A. Representative Menthol (1mM)-evoked TRPM8 channel currents before and after phorbol 12,13-dibutyrate (PDBu) incubation (1µM; 5 min denoted by a break in the trace) in the absence (left trace) and presence (right trace) of calcium. B. Repeated applications of menthol-induced currents after PDBu incubation in the presence (filled circles, n = 3 cells) and absence of calcium (open circles, n =4 cells) C. Summary graph showing menthol-induced currents are inhibited significantly after PDBu incubation (p < 0.05) and no change after wash application both in the presence and absence of calcium. 0 represents capsaicin application before PDBu and 1 to 6 represents number of capsaicin applications after PDBu incubation. Number in the parenthesis represents the number of cells and the asterisk (*) represents p < 0.05. PDBu denotes Phorbol 12,13-dibutyrate.





Figure 9(A-C): Activation of PKA Down Regulates TRPM8-Mediated Whole Cell Current both in the Absence and Presence of Calcium. A. Representative Menthol (1mM)-evoked TRPM8 channel currents before and after application of FSK + IBMX (20µM and 50µM ; 5 min denoted by a break in the trace) in the absence (left trace) and presence (right trace) of calcium B. repeated applications of menthol-induced currents before and after 20µM FSK+ 50µM IBMX incubation in the presence (filled circles, n = 6 cells) and absence of calcium (open circles, n = 3 cells) and C. Summary graph showing menthol-induced currents are inhibited significantly after 20µM FSK+ 50µM IBMX incubation and no change after wash application both in the presence and absence of calcium. Number in the parenthesis represents the number of cells responses and the asterisk (*) represents p < 0.05. 0 represents pre FSK+IBMX applications and 1 to 6 represents number of menthol applications after FSK+IBMX.




Figure 10(A-C): Capsaicin-Induced Nocifensive Pain Behavior is Reduced by Co-administration with Menthol in Diabetic and Non-Diabetic Mice. Onset, duration, and number of lickings and/or flickerings of the injected hind paw were determined after intraplantar injection of 20µl of capsaicin (2 mM) or capsaicin plus menthol (5 mM) in diabetic and non-diabetic mice. A. In non-diabetic mice onset of pain symptoms such as licking and flickering was significantly increased from 3.07±0.29 s (n = 8 mice) after capsaicin alone to 10.7±0.8 s (n =8 mice) after capsaicin plus menthol. Similar results were obtained in diabetic mice also although onset of pain with both capsaicin (1.3±0.2 s, n = 8 mice) and capsaicin plus menthol (2.3±0.4 s, n = 8 mice) is much quicker compared to non-diabetic mice. B. The total duration of nocifensive behavior was also significantly decreased from 32.5±1.9 with capsaicin to 19.1±2.1 s with capsaicin plus menthol in non-diabetic mice. Similar decrease was noticed in diabetic mice (capsaicin, 62±7.5 s, n = 8 mice; capsaicin plus menthol, 39.5±7.8 sec) although there was increased duration of lickings and flickerings in diabetic mice. C. We also compared the total number of hind paw lickings and flickerings for the duration of nocifensive behavior in diabetic and non-diabetic mice. The number of hind paw licks and flickerings was significantly decreased from 34±2.3 to 19.1±2.1 in non-diabetic mice and similar findings were noticed in diabetic mice (capsaicin, 60±7.3 s, n = 8 mice; capsaicin plus menthol, 35±4.1 sec n = 8 mice) although there was more number of lickings and flickerings in diabetic mice. Data are represented as mean±SEM. N = 8 mice per group. *p<0.05 obtained from students unpaired t-test.







1.       Vinik AI (2004) Advances in diabetes for the millennium: new treatments for diabetic neuropathies. Med Gen Med 6: 13.

2.       Anand P, Terenghi G, Warner G, Kopelman P, Williams-Chestnut RE, et al. (196) The role of endogenous nerve growth factor in human diabetic neuropathy. Nature Med 2: 703-707.

3.       Sugimoto K, Murakawa Y, Sima AA (2000) Diabetic neuropathy--a continuing enigma. Diab/Metabol Res Rev 16: 408-433.

4.       Zimmermann M (2001) Pathobiology of neuropathic pain. Eur J Pharmacol 429: 23-37.

5.       Freynhagen R, Bennett MI (2009) Diagnosis and management of neuropathic pain. Brit Med J 339: b3002.

6.       Scherrer G, Imamachi N, Cao YQ, Contet C, Mennicken F, et al. (2009) Dissociation of the opioid receptor mechanisms that control mechanical and heat pain. Cell 137: 1148-1159.

7.       Cavanaugh DJ, Lee H, Lo L, Shields SD, Zylka MJ, et al. (2009) Distinct subsets of unmyelinated primary sensory fibers mediate behavioral responses to noxious thermal and mechanical stimuli. Proc Natl Acad Sci USA 106: 9075-9080.

8.       Jeffry JA, Yu SQ, Sikand P, Parihar A, Evans MS, et al. (2009) Selective targeting of TRPV1 expressing sensory nerve terminals in the spinal cord for long lasting analgesia. PloS one 4: e7021.

9.       Bader M, Alenina N, Andrade-Navarro MA, Santos RA (2014) MAS and its related G protein-coupled receptors, Mrgprs. Pharmacol Rev 66: 1080-1105.

10.    Reid G, Flonta M (2001) Cold transduction by inhibition of a background potassium conductance in rat primary sensory neurones. Neurosci Lett 297: 171-174.

11.    Reid G, Flonta ML (2001) Physiology. Cold current in thermoreceptive neurons. Nature 413: 480.

12.    Reid G, Flonta ML (2002) Ion channels activated by cold and menthol in cultured rat dorsal root ganglion neurones. Neurosci Lett 324: 164-168.

13.    McKemy DD, Neuhausser WM, Julius D (2002) Identification of a cold receptor reveals a general role for TRP channels in thermosensation. Nature 416: 52-58.

14.    Story GM, Peier AM, Reeve AJ, Eid SR, Mosbacher J, et al. (2003) ANKTM1, a TRP-like channel expressed in nociceptive neurons, is activated by cold temperatures. Cell 112: 819-829.

15.    Pabbidi RM, Yu SQ, Peng S, Khardori R, Pauza ME, et al. (2008) Influence of TRPV1 on diabetes-induced alterations in thermal pain sensitivity. Mol Pain 4: 9.

16.    Chu Q, Moreland R, Yew NS, Foley J, Ziegler R, et al. (2008) Systemic Insulin-like growth factor-1 reverses hypoalgesia and improves mobility in a mouse model of diabetic peripheral neuropathy. Mol Ther 16: 1400-1408.

17.    Vareniuk I, Pavlov IA, Obrosova IG (2008) Inducible nitric oxide synthase gene deficiency counteracts multiple manifestations of peripheral neuropathy in a streptozotocin-induced mouse model of diabetes. Diabetologia 51: 2126-2133.

18.    Ellington HC, Cotter MA, Cameron NE, Ross RA (2002) The effect of cannabinoids on capsaicin-evoked calcitonin gene-related peptide (CGRP) release from the isolated paw skin of diabetic and non-diabetic rats. Neuropharmacol 42: 966-975.

19.    Andersson DA, Chase HW, Bevan S (2004) TRPM8 activation by menthol, icilin, and cold is differentially modulated by intracellular pH. J Neurosci 24: 5364-5369.

20.    Liu B, Qin F (2005) Functional control of cold- and menthol-sensitive TRPM8 ion channels by phosphatidylinositol 4,5-bisphosphate. J Neurosci 25: 1674-1681.

21.    Premkumar LS, Raisinghani M, Pingle SC, Long C, Pimentel F (2005) Downregulation of transient receptor potential melastatin 8 by protein kinase C-mediated dephosphorylation. J Neurosci 25: 11322-11329.

22.    Kamei J, Zushida K, Morita K, Sasaki M, Tanaka S (2001) Role of vanilloid VR1 receptor in thermal allodynia and hyperalgesia in diabetic mice. Eur J Pharmacol 422: 83-86.

23.    Backonja M, Wallace MS, Blonsky ER, Cutler BJ, Malan P Jr, et al. (2008) NGX-4010 C116 Study Group. NGX-4010, a high-concentration capsaicin patch,for the treatment of postherpetic neuralgia: a randomised, double-blind study. Lancet Neurol 7: 1106-1112.

24.    Irving GA, Backonja MM, Dunteman E, Blonsky ER, Vanhove GF, et al. (2011) NGX-4010 C117 Study Group. Amulticenter, randomized, double-blind, controlled study of NGX-4010, a high-concentration capsaicin patch, for the treatment of postherpetic neuralgia. Pain Med 12: 99-109.

25.    Simpson DM, Brown S, Tobias J, NGX-4010C107 Study Group (2008) Controlled trial of high-concentration capsaicin patch for treatment of painful HIVneuropathy. Neurology 70: 2305-2313.

26.    Haanpää M,Cruccu G, Nurmikko TJ, McBride WT, Docu Axelarad A, et al. (2016) Capsaicin 8% patch versus oral pregabalin in patients with peripheral neuropathic pain. Eur J Pain 20: 316-328.

27.    Simpson DM, Robinson-PappJ, Van J, Stoker M, Jacobs H, et al. (2017) Capsaicin 8% patch in painful diabetic peripheral neuropathy: a randomised, double-blind,placebo-controlled study. J Pain 18: 42-53.

28.    Rashid MH, Inoue M, Bakoshi S, Ueda H (2003) Increased expression of vanilloid receptor 1 on myelinated primary afferent neurons contributes to the antihyperalgesic effect of capsaicin cream in diabetic neuropathic pain in mice. J Pharmacol Expt Ther 306: 709-717.

29.    Vissers K, Meert T (2005) A behavioral and pharmacological validation of the acetone spray test in gerbils with a chronic constriction injury. Anes Anal 101: 457-464.

30.    Choi Y, Yoon YW, Na HS, Kim SH, Chung JM (1994) Behavioral signs of ongoing pain and cold allodynia in a rat model of neuropathic pain. Pain 59: 369-376.

31.    Obrosova IG (2009) Diabetes and the peripheral nerve. Bioch. Biophys, Acta 1792: 931-940.

32.    Schmader KE (2002) Epidemiology and impact on quality of life of postherpetic neuralgia and painful diabetic neuropathy. Clin J Pain 18: 350-354.

33.    Zochodne DW, Ramji N, Toth C (2008) Neuronal targeting in diabetes mellitus: a story of sensory neurons and motor neurons. The Neuroscientist 14: 311-318.

34.    Schmader KE (2002) Epidemiology and impact on quality of life of postherpetic neuralgia and painful diabetic neuropathy. Clin J Pain 18: 350-354.

35.    Proudfoot CJ, Garry EM, Cottrell DF, Rosie R, Anderson H, et al. (2016) Analgesia mediated by the TRPM8 cold receptor in chronic neuropathic pain. Curr Biol 16: 1591-605.

36.    Colburn RW, Lubin ML, Stone DJ Jr, Wang Y, Lawrence D, et al. (2007) Attenuated cold sensitivity in TRPM8 null mice. Neuron 54: 379-386.

37.    Dhaka A, Murray AN, Mathur J, Earley TJ, Petrus MJ, et al. (2007) TRPM8 is required for cold sensation in mice. Neuron 54: 371-378.

38.    Amadesi S, Nie J, Vergnolle N, Cottrell GS, Grady EF, et al. (2004) Protease-activated receptor 2 sensitizes the capsaicin receptor transient receptor potential vanilloid receptor 1 to induce hyperalgesia. J Neurosci 24: 4300-4312.

39.    Lolignier S, Gkika D, Andersson D, Leipold E, Vetter I, et al. (2016) New Insight in Cold Pain: Role of Ion Channels, Modulation, and Clinical Perspectives. J Neurosci 36: 11435-11439.

40.    Julius D, Basbaum AI (2001) Molecular mechanisms of nociception. Nature 413: 203-210.

41.    Nakamura J, Kato K, Hamada Y, Nakayama M, Chaya S, et al. (1999) A protein kinase C-beta-selective inhibitor ameliorates neural dysfunction in streptozotocin-induced diabetic rats. Diabetes 48: 2090-2095.

42.    Freshwater JD, Svensson CI, Malmberg AB, Calcutt NA (2002) Elevated spinal cyclooxygenase and prostaglandin release during hyperalgesia in diabetic rats. Diabetes 51: 2249-2255.

43.    Sasase T, Yamada H, Sakoda K, Imagawa N, Abe T, et al. (2005) Novel protein kinase C-beta isoform selective inhibitor JTT-010 ameliorates both hyper- and hypoalgesia in streptozotocin- induced diabetic rats. Diab Obes Metabol 7: 586-594.

44.    Ahlgren SC, Levine JD (1994) Protein kinase C inhibitors decrease hyperalgesia and C-fiber hyperexcitability in the streptozotocin-diabetic rat. J Neurophysiol 72: 684-692.

45.    Moriyama T, Higashi T, Togashi K, Iida T, Segi E, et al. (2005) Sensitization of TRPV1 by EP1 and IP reveals peripheral nociceptive mechanism of prostaglandins. Mol Pain 1: 3.

46.    Khasar SG, Lin YH, Martin A, Dadgar J, McMahon T, et al. (1999) A novel nociceptor signaling pathway revealed in protein kinase C epsilon mutant mice. Neuron 24: 253-260.

47.    Eichberg J (2002) Protein kinase C changes in diabetes: is the concept relevant to neuropathy? Inter. Rev. Neurobiol 50: 61-82.

48.    Prescott ED, Julius D (2003) A modular PIP2 binding site as a determinant of capsaicin receptor sensitivity. Science 300: 1284-1288.

49.    Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, et al. (2001) Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition. Nature 411: 957-962.

50.    Premkumar LS, Ahern GP (2000) Induction of vanilloid receptor channel activity by protein kinase C. Nature 408: 985-990.

51.    Numazaki M, Tominaga T, Toyooka H, Tominaga M (2002) Direct phosphorylation of capsaicin receptor VR1 by protein kinase Cepsilon and identification of two target serine residues. J Biol Chem 277: 13375-13378.

52.    Bhave G, Hu HJ, Glauner KS, Zhu W, Wang H, et al. (2003) Protein kinase C phosphorylation sensitizes but does not activate the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1). Proc Natl Acad Sci USA 100: 12480-12485.

53.    Morenilla-Palao C, Planells-Cases R, Garcia-Sanz N, Ferrer-Montiel A (2004) Regulated exocytosis contributes to protein kinase C potentiation of vanilloid receptor activity. J Biol Chem 279: 25665-25672.

54.    Kumazawa T, Mizumura K, Koda H, Fukusako H (1996) EP receptor subtypes implicated in the PGE2-induced sensitization of polymodal receptors in response to bradykinin and heat. J Neurophysiol 75: 2361-2368.

55.    Southall MD, Vasko MR (2001) Prostaglandin receptor subtypes, EP3C and EP4, mediate the prostaglandin E2-induced cAMP production and sensitization of sensory neurons. J Biol. Chem 276: 16083-16091.

56.    Kress M, Rodl J, Reeh PW (1996) Stable analogues of cyclic AMP but not cyclic GMP sensitize unmyelinated primary afferents in rat skin to heat stimulation but not to inflammatory mediators, in vitro. Neuroscience 74: 609-617.

57.    Malmberg AB, Brandon EP, Idzerda RL, Liu H, McKnight GS, et al. (1997) Diminished inflammation and nociceptive pain with preservation of neuropathic pain in mice with a targeted mutation of the type I regulatory subunit of cAMP-dependent protein kinase. J Neurosci 17: 7462-7470.

58.    Rathee PK, Distler C, Obreja O, Neuhuber W, Wang GK, et al. (2002) PKA/AKAP/VR-1 module: A common link of Gs-mediated signaling to thermal hyperalgesia. J Neurosci 22: 4740-4745.

59.    Lopshire JC, Nicol GD (1998) The cAMP transduction cascade mediates the prostaglandin E2 enhancement of the capsaicin-elicited current in rat sensory neurons: whole-cell and single-channel studies. J Neurosci 18: 6081-6092.

60.    Daniels RL, Takashima Y, McKemy DD (2009) Activity of the neuronal cold sensor TRPM8 is regulated by phospholipase C via the phospholipid phosphoinositol 4,5-bisphosphate. J. Biol. Chem 284: 1570-1582.

Tominaga M, Caterina MJ, Malmberg AB, Rosen TA, Gilbert H, et al. (1998) The cloned capsaicin receptor integrates multiple pain-producing stimuli. Neuron 21: 531-543

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Diabetes and Treatment