Abstract

(−)-Naringenin 4′,7-dimethyl ether ((−)-NRG-DM) was isolated for the first time by our lab from Nardostachys jatamansi DC, a traditional medicinal plant frequently used to attenuate pain in Asia. As a natural derivative of analgesic, the current study was designed to test the potential analgesic activity of (−)-NRG-DM and its implicated mechanism. The analgesic activity of (−)-NRG-DM was assessed in a formalin-induced mouse inflammatory pain model and mustard oil-induced mouse colorectal pain model, in which the mice were intraperitoneally administrated with vehicle or (−)-NRG-DM (30 or 50 mg/kg) (n = 10 for each group). Our data showed that (−)-NRG-DM can dose dependently (30~50 mg/kg) relieve the pain behaviors. Notably, (−)-NRG-DM did not affect motor coordination in mice evaluated by the rotarod test, in which the animals were intraperitoneally injected with vehicle or (−)-NRG-DM (100, 200, or 400 mg/kg) (n = 10 for each group). In acutely isolated mouse dorsal root ganglion neurons, (−)-NRG-DM (1~30 μM) potently dampened the stimulated firing, reduced the action potential threshold and amplitude. In addition, the neuronal delayed rectifier potassium currents (IK) and voltage-gated sodium currents (INa) were significantly suppressed. Consistently, (−)-NRG-DM dramatically inhibited heterologously expressed Kv2.1 and Nav1.8 channels which represent the major components of the endogenous IK and INa. A pharmacokinetic study revealed the plasma concentration of (−)-NRG-DM is around 7 µM, which was higher than the effective concentrations for the IK and INa. Taken together, our study showed that (−)-NRG-DM is a potential analgesic candidate with inhibition of multiple neuronal channels (mediating IK and INa).

Highlights

  • Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage [1]. It can be subdivided into somatic pain and visceral pain according to the originating tissue, while it can be categorized into acute and chronic pain based on the ongoing time [2]

  • RWGh-iDleMat stihgendifoicseanotfly30amttegn/ukagt,e(d−p)-aNinRfGu-lDbMehaviors sihnoartednoesde-tdheepleicnkdinegnttimmeanannedrpdauinrisncgorpehoafsteheI tawnod bpehhaasveioIrIal(Fsitgaguerse, 2b)u.tAotnltyhehaddoase of 50 smiggn/ikfigca, n(−t)i-nNflRueGn-cDe Min spihgansiefiIc.antly attenuated painful behaviors, including the licking time and Tooveexracllul dpeaipnosscsiobrlee innonb-ostphecpihfiacsmesusincleforremlaaxlainnt-ionrjescetdeadtimveiceef.feWctsh,ilteheateftfheectsdoosfe of 30 (−)-NRG-DM on motor performance were evaluated in the rotarod test. (−)-NRG-DM wmags/wkge,ll(t−o)l-eNraRteGd-DinMtheshrootratreondedtestth, ewiltihckninogsigtinmifiecaanntdefpfeacitns osncotrhee aobfiltihtye ttowroembaeihnavioral osntatghees,robtuattionnglyrohdaadftaersiingtnriafpicearnittoinnefalluaednmceininistprhataiosen Io. f (−)-NRG-DM at 100 mg/kg, 200 mg/kg, or even 400 mg/kg (Table 1)

  • The current study showed that the analgesic (−)-NRG-DM directly dampens neuron excitability in acutely isolated mouse small-diameter dorsal root ganglia (DRG) neurons with a reduced threshold and amplitude of action potential firing (Figure 4A)

Read more

Summary

Introduction

Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage [1]. The Kv2.1 channels are thought to represent a major component of IK, which helps to set the resting membrane potential and shape the action potentials [18–20]. Modulation of these channels underlies multiple analgesics. Flavonoids are polyphenolic structures naturally distributed in most plants and consumed daily They have been widely used for analgesic, anti-inflammatory, and antioxidant effects along with safe preclinical and clinical profiles [28,29]. As a flavonoid originating from a widely used folk medicine for pain relief, we hypothesized that (−)-NRG-DM might have analgesic activity similar to its naringenin prototype which has been reported to alleviate pain in multiple models [32].

Results
Analgesic Effects of (−)-NRG-DM in Mustard Oil-Induced Mouse Colorectal Pain Model
Inhibitory Effects of (−)-NRG-DM on Neuronal
Materials and Methods
Animals
Formalin-Induced Inflammatory Model
Mustard Oil-Induced Mouse Colorectal Pain Model
Rotarod Test
Pharmacokinetic Study
Preparation of Dorsal Root Ganglion Neurons
Cell Culture and Transfection
Electrophysiological Recordings
4.10. Statistics
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call