Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Zhen Ci Yan Jiu ; 48(3): 217-25, 2023 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-36951072

ABSTRACT

OBJECTIVE: To investigate the relationship between acupoint sensitization on the body surface and neuronal intrinsic excitability of the medium- and small-size dorsal root ganglion (DRG) neurons from the perspective of ion channel kinetics in mice with gastric ulcer. METHODS: Male C57BL/6J mice were randomly divided into control (n=32) and model groups (n=34). The gastric ulcer model was established by injection of 60% glacial acetic acid (0.2 mL/100 g) into the gastric wall muscle layer and submucosa near the pylorus in the minor curvature of the stomach. In contrast, the same dose of normal saline was injected in the same way in the control group. Six days after modeling, Evans blue (EB) solution was injected into the mouse's tail vein for observing the number and distribution of the exudation blue spots on the body surface. Histopathological changes of the gastric tissue were observed by H.E. staining. Then, whole-cell membrane currents and intrinsic excitability of medium- and small-size neurons in the spinal T9-T11 DRGs were measured by in vitro electrophysiology combining with biocytin-ABC method. RESULTS: In the control group, EB exudation blue spots were not obvious, while in the model group, the blue spots on the body surface were densely distributed in the area of spinal T9-T11 segments, the epigastric region, and the skin around "Zhongwan" (CV12) and "Huaroumen" (ST24) regions, and near the surgical incision region. Compared with the control group, the model group had a high level of eosinophilic infiltrates in the submucosa of gastric tissues, severe gastric fossa structure damage, gastric fundus gland dilation and other pathological manifestations. The number of exudation blue spots was proportional to the degree of inflammatory reaction in the stomach. In comparison with the control group, the spike discharges of type II of medium-size DRG neurons in T9-T11 segments were decreased, and the current of whole-cell membrane was increased, basic intensity was decreased (P<0.05), discharge frequency and discharge number were increased (P<0.01,P<0.000 1); while the discharges of type I small-size DRG neurons were decreased, those of type II neurons increased, the whole-cell membrane current was decreased, and discharge frequency and discharge number were decreased (P<0.01, P<0.000 1). CONCLUSION: Both the medium- and small-size DRG neurons from the spinal T9-T11 segments involve in gastric ulcer-induced acupoint sensitization via their different spike discharge activities. And intrinsic excitability of these DRG neurons can not only dynamically encode the plasticity of acupoint sensitization, but also can help us understand the neural mechanism of acupoint sensitization induced by visceral injury.


Subject(s)
Ganglia, Spinal , Stomach Ulcer , Rats , Mice , Male , Animals , Ganglia, Spinal/physiology , Stomach Ulcer/genetics , Stomach Ulcer/therapy , Rats, Sprague-Dawley , Acupuncture Points , Mice, Inbred C57BL , Neurons
2.
Neuromodulation ; 25(7): 1024-1032, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35760751

ABSTRACT

INTRODUCTION: Spinal cord stimulation (SCS) can provide long-term pain relief for various chronic pain conditions, but some patients have no relief with trial stimulation or lose efficacy over time. To "salvage" relief in patients who do not respond or have lost efficacy, alternative stimulation paradigms or anatomical targets can be considered. Dorsal root ganglion stimulation (DRG-S) has a different mechanism of action and anatomical target than SCS. OBJECTIVES: We assessed DRG-S salvage therapy outcomes in patients who did not respond to SCS or had lost SCS efficacy. MATERIALS AND METHODS: We retrospectively included consecutive patients from 2016 to 2020 who were salvaged with DRG-S after failed SCS trials (<50% pain reduction) or who had lost efficacy after permanent SCS. We compared numerical rating scale (NRS) pain, Oswestry disability index (ODI), health-related quality of life (EuroQol five-dimensions five-level), and oral morphine equivalent (OME) opioid requirements before DRG-S salvage and at patients' last follow-up. RESULTS: A total of 60 patients who had failed SCS were salvaged with DRG-S. The mean age was 56 ± 12 years, and the most common diagnoses were complex regional pain syndrome (n = 24) and failed back surgery syndrome (n = 24). The most common failed modalities included tonic (n = 32), Burst (n = 18), and high-frequency (n = 10) SCS. The median follow-up duration of salvage DRG-S was 34 months. With DRG-S, NRS decreased (8.7 ± 1.2 to 3.8 ± 2.1), and OME declined (median 23 mg to median 15 mg), whereas EuroQol 5D scores increased (0.40 ± 0.15 to 0.71 ± 0.15), and ODI improved (64 ± 14% to 31 ± 18%) (all p < 0.05). CONCLUSIONS: DRG-S can be used in patients with chronic pain who have previously failed to receive persistent benefit from SCS.


Subject(s)
Chronic Pain , Spinal Cord Stimulation , Adult , Aged , Analgesics, Opioid , Chronic Pain/therapy , Ganglia, Spinal/physiology , Humans , Middle Aged , Morphine Derivatives , Quality of Life , Retrospective Studies , Salvage Therapy , Spinal Cord , Spinal Cord Stimulation/methods , Treatment Outcome
3.
A A Pract ; 16(5): e01589, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35587521

ABSTRACT

Dorsal root ganglion stimulation (DRG-S) is widely accepted for treating focal pain syndromes. We present the case of a 46-year-old woman with severe lumbar radiculopathy with an implanted spinal cord stimulator (SCS) that had lost efficacy. She developed an incisional hernia after undergoing a minimally invasive, extreme lateral interbody fusion and SCS explant. After herniorrhaphy, she presented with severe pain at the T10-T11 dermatomes, which we treated with DRG-S. One-year after lumbar fusion, her refractory lumbar and radicular pain returned, which we ultimately treated with bilateral T12+S1 DRG-S. DRG-S was thus used to successfully treat focal postsurgical and diffuse chronic pain.


Subject(s)
Chronic Pain , Electric Stimulation Therapy , Chronic Pain/therapy , Female , Ganglia, Spinal/physiology , Humans , Lumbosacral Region , Middle Aged , Pain Management
4.
Pain Pract ; 22(5): 541-546, 2022 06.
Article in English | MEDLINE | ID: mdl-35485298

ABSTRACT

BACKGROUND: Radiofrequency (RF) treatment of the genicular nerves offers pain relief in patients suffering from chronic knee pain including persistent post-surgical knee pain (PPSP). We present the first case report of the development of complex regional pain syndrome (CRPS) in a chronic knee pain patient after an RF ablation of the genicular nerves that was successfully treated with dorsal root ganglion (DRG) stimulation. CASE PRESENTATION: The patient developed increased pain, sympathetic and dysmorphic changes of the index knee 10 weeks after RF treatment for PPSP. Diagnosis of CRPS type II was made using positive clinical findings and the Budapest diagnostic tool. Laboratory workup and PET-CT were negative. The patient was refractory to usual care and she was treated successfully with dorsal ganglion root stimulation. CONCLUSIONS: Complex regional pain syndrome is a possible complication of RF ablation of the genicular nerves in patients with chronic knee pain, and DRG stimulation may be a treatment option. Physicians should be aware of this complication, especially when patients have a medical history of CRPS.


Subject(s)
Catheter Ablation/adverse effects , Complex Regional Pain Syndromes/therapy , Ganglia, Spinal/physiology , Osteoarthritis, Knee/complications , Transcutaneous Electric Nerve Stimulation , Arthralgia/etiology , Arthralgia/therapy , Complex Regional Pain Syndromes/etiology , Female , Ganglia, Spinal/diagnostic imaging , Humans , Pain/surgery , Positron Emission Tomography Computed Tomography
5.
Med Biol Eng Comput ; 60(5): 1527-1540, 2022 May.
Article in English | MEDLINE | ID: mdl-35349032

ABSTRACT

Overactive bladder patients suffer from a frequent, uncontrollable urge to urinate, which can lead to a poor quality of life. We aim to improve open-loop sacral neuromodulation therapy by developing a conditional stimulation paradigm using neural recordings from dorsal root ganglia (DRG) as sensory feedback. Experiments were performed in 5 anesthetized felines. We implemented a Kalman filter-based algorithm to estimate the bladder pressure in real-time using sacral-level DRG neural recordings and initiated sacral root electrical stimulation when the algorithm detected an increase in bladder pressure. Closed-loop neuromodulation was performed during continuous cystometry and compared to bladder fills with continuous and no stimulation. Overall, closed-loop stimulation increased bladder capacity by 13.8% over no stimulation (p < 0.001) and reduced stimulation time versus continuous stimulation by 57.7%. High-confidence bladder single units had a reduced sensitivity during stimulation, with lower linear trendline fits and higher pressure thresholds for firing observed during stimulation trials. This study demonstrates the utility of decoding bladder pressure from neural activity for closed-loop control of sacral neuromodulation. An underlying mechanism for sacral neuromodulation may be a reduction in bladder sensory neuron activity during stimulation. Real-time validation during behavioral studies is necessary prior to clinical translation of closed-loop sacral neuromodulation.


Subject(s)
Electric Stimulation Therapy , Ganglia, Spinal , Animals , Cats , Feedback, Sensory , Ganglia, Spinal/physiology , Humans , Quality of Life , Urinary Bladder/physiology
6.
Physiol Rep ; 9(16): e14975, 2021 08.
Article in English | MEDLINE | ID: mdl-34405579

ABSTRACT

Voltage-gated ion channels play a key role in the action potential (AP) initiation and its propagation in sensory neurons. Modulation of their activity during chronic inflammation creates a persistent pain state. In this study, we sought to determine how peripheral inflammation caused by complete Freund's adjuvant (CFA) alters the fast sodium (INa ), L-type calcium (ICaL ), and potassium (IK ) currents in primary afferent fibers to increase nociception. In our model, intraplantar administration of CFA induced mechanical allodynia and thermal hyperalgesia at day 14 post-injection. Using whole-cell patch-clamp recording in dissociated small (C), medium (Aδ), and large-sized (Aß) rat dorsal root ganglion (DRG) neurons, we found that CFA prolonged the AP duration and increased the amplitude of the tetrodotoxin-resistant (TTX-r) INa in Aß fibers. In addition, CFA accelerated the recovery of INa from inactivation in C and Aδ nociceptive fibers but enhanced the late sodium current (INaL ) only in Aδ and Aß neurons. Inflammation similarly reduced the amplitude of ICaL in each neuronal cell type. Fourteen days after injection, CFA reduced both components of IK (IKdr and IA ) in Aδ fibers. We also found that IA was significantly larger in C and Aδ neurons in normal conditions and during chronic inflammation. Our data, therefore, suggest that targeting the transient potassium current IA represents an efficient way to shift the balance toward antinociception during inflammation, since its activation will selectively decrease the AP duration in nociceptive fibers. Altogether, our data indicate that complex interactions between IK , INa , and ICaL reduce pain threshold by concomitantly enhancing the activity of nociceptive neurons and reducing the inhibitory action of Aß fibers during chronic inflammation.


Subject(s)
Action Potentials , Neurons, Afferent/metabolism , Nociceptive Pain/metabolism , Potassium Channels, Voltage-Gated/metabolism , Animals , Calcium Channels, L-Type/metabolism , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Male , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Nociception , Nociceptive Pain/physiopathology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Tetrodotoxin/pharmacology
7.
J Ethnopharmacol ; 281: 114495, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34364968

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Allium macrostemon Bunge. is an edible Chinese herb traditionally used for the treatment of thoracic pain, stenocardia, heart asthma and diarrhea. Although its biological potential has been extensively proven such as antioxidant activity, antiplatelet aggregation, vasodilation and antidepressant-like activity, there are no reports in the literature regarding its pharmacological analgesic activity. AIM OF THE STUDY: The study was carried out to examine the anti-nociceptive activity of the crude extract of A. macrostemon bulbs and interpret its likely molecular target. MATERIALS AND METHODS: The bulbs of A. macrostemon were gathered, dried-up, and extracted with water (AMWD). AMWD was subjected to activity testing, using chemical-induced (acetic acid and formalin test) and heat-induced (hot plate) pain models. To evaluate the likely mechanistic strategy involved in the analgesic effect of AMWD, whole-cell patch clamp recordings were conducted in acutely dissociated dorsal root ganglion (DRG) neurons and human embryonic kidney 293T (HEK293T) cells expressing pain-related receptors. Electrophysiological methods were employed to detect the action potentials of DRG neurons and potential targets of A. macrostemon. RESULTS: AMWD showed significant palliative effect in all heat and chemical induced pain assays. Moreover, AMWD significantly reduces the excitability of dorsal root ganglion neurons by reducing the firing frequency of action potentials. Further analysis revealed that voltage-gated sodium channel Nav1.7 is the potential target of A. macrostemon for its analgesic activity. CONCLUSION: This study has brought new scientific evidence of preclinical efficacy of A. macrostemon as an anti-nociceptive agent. Apparently, these effects are involved with the inhibition of the voltage-sensitive Nav1.7 channel contributing to the reduction of peripheral neuronal excitability. Our present study justifies the folkloric usage of A. macrostemon as a remedy for several pain states. Furthermore, A. macrostemon is a good resource for the development of analgesic drugs targeting Nav1.7 channel.


Subject(s)
Analgesics/therapeutic use , Chive , Pain/drug therapy , Plant Extracts/therapeutic use , Voltage-Gated Sodium Channel Blockers/therapeutic use , Acetic Acid , Analgesics/pharmacology , Animals , Cell Survival/drug effects , Formaldehyde , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , HEK293 Cells , Hot Temperature , Humans , Locomotion/drug effects , Male , Mice, Inbred C57BL , NAV1.7 Voltage-Gated Sodium Channel/physiology , Nociceptors/physiology , Pain/etiology , Plant Extracts/pharmacology , Plant Roots , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Voltage-Gated Sodium Channel Blockers/pharmacology
8.
PLoS One ; 16(3): e0248139, 2021.
Article in English | MEDLINE | ID: mdl-33690613

ABSTRACT

This work tries to help overcome the lack of relevant translational screening assays, as a limitation for the identification of novel analgesics for neuropathic pain. Hyperexcitability and neurite shortening are common adverse effects of antiviral and antitumor drugs, leading to neuropathic pain. Now, as seen in the drug screening that we developed here, a high-content microscopy-based assay with immortalized dorsal root ganglia (DRG) neurons (differentiated F11 cells) allowed to identify drugs able to protect against the iatrogenic neurite shortening induced by the antitumor drug vincristine and the antiviral drug rilpivirine. We observed that vincristine and rilpivirine induced a significant reduction in the neurite length, which was reverted by α-lipoic acid. We had also evidenced protective effects of pregabalin and melatonin, acting through the α2δ-2 subunit of the voltage-dependent calcium channels and the MT1 receptor, respectively. Additionally, two hits originated from a previous primary screening aimed to detect inhibitors of hyperexcitability to inflammatory mediators in DRG neurons (nitrendipine and felodipine) also prevented neurite shortening in our model. In summary, in this work we developed a novel secondary assay for identifying hits with neuroprotective effect against iatrogenic neurite shortening, consistent with the anti-hyperexcitability action previously tested: highlighting nitrendipine and felodipine against iatrogenic damage in DRG neurons.


Subject(s)
Drug Evaluation, Preclinical/methods , Neurites/drug effects , Analgesics/pharmacology , Cell Line , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Humans , Iatrogenic Disease , Melatonin/pharmacology , Neuralgia/drug therapy , Neurites/metabolism , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/pharmacology , Pregabalin/pharmacology , Rilpivirine/adverse effects , Rilpivirine/pharmacology , Thioctic Acid/pharmacology , Vincristine/adverse effects , Vincristine/pharmacology
9.
J Mol Neurosci ; 71(6): 1290-1300, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33417168

ABSTRACT

To study the effect of photobiomodulation (PBM) on axon regeneration and secretion change of dorsal root ganglion (DRG) under oxidative stress after spinal cord injury (SCI), and further explore the effect of changes in DRG secretion caused by PBM on the polarization of macrophages. The PBM-DRG model was constructed to perform PBM on neurons under oxidative stress simulated in vitro. And the irradiation conditions were as follows: wavelength, 810 nm; power density, 2 mW/cm2; irradiation area, 4.5 cm2; and irradiation time, 440 s. Then resulted in an energy of 4 J (2 mW/cm2 × 4.5 cm2 × 440 s). About 100 µM H202 was added to the culture medium to simulate oxidative stress after SCI. An ROS (reactive oxygen species) assay kit was used to measure ROS contend in the DRG. The survival level of the neurons was measured using the CCK-8 method, and the axon regeneration of neurons was observed by using immunofluorescence. The secretion level of CCL2 from DRG was determined by RT-qPCR and ELISA. Further culturing macrophages of DRG-conditioned medium culture, the expression level of iNOS and Arg-1 in macrophages was assessed using Western blot analysis. The expression level of TNF-α and IL-1ß was determined by ELISA. After adding the neutralizing antibody of CCL2 to the DRG neuron-conditioned medium following PBM irradiation to culture macrophages to observe the effects on macrophage polarization and secretion. PBM could reduce ROS levels in neurons, increase neuronal survival under oxidative stress, and promote neuronal axon regeneration. In addition, PBM could also promote CCL2 secretion by DRG under oxidative stress. By constructing a DRG supernatant-M1 macrophage adoptive culture model, we found that the supernatant of DRG after PBM intervention could reduce the expression level of iNOS and the secretion of TNF-α and IL-1ß in M1 macrophages; at the same time, it could also up-regulate the expression of Arg-1, one of the markers of M2 macrophages. Furthermore, these effects could be prevented by the addition of neutralizing antibodies of CCL2. PBM could promote survival and axonal regeneration of DRG under SCI oxidative stress, increase the secretion level of CCL2 by DRG, and this change can reduce the polarization of macrophages to M1, further indicating that PBM could promote spinal cord injury repair.


Subject(s)
Axons/metabolism , Chemokine CCL2/metabolism , Macrophages/cytology , Oxidative Stress , Phototherapy/methods , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Animals , Axons/radiation effects , Cell Differentiation , Cells, Cultured , Chemokine CCL2/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Interleukin-1beta/metabolism , Light , Macrophages/immunology , Macrophages/radiation effects , Male , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/metabolism
10.
J Pharmacol Exp Ther ; 377(1): 20-28, 2021 04.
Article in English | MEDLINE | ID: mdl-33431609

ABSTRACT

Treatment of chronic pain remains an unmet medical need. The neuronal voltage-gated potassium Kv7/KCNQ/M channel has been implicated as a therapeutic target for chronic pain. However, whether pharmacological activation of the Kv7 channel can alleviate pain remains elusive. In this study, we show that selective activation of native M-currents by a novel channel opener SCR2682 reduces repetitive firings of dorsal root ganglia (DRG) sensory neurons. Intraperitoneal administration of SCR2682 relieves mechanical allodynia and thermal hyperalgesia in rat models of pain induced by complete Freund's adjuvant (CFA) or spared nerve injury (SNI) in a dose-dependent manner without affecting locomotor activity. The antinociceptive efficacy of SCR2682 can be reversed by the channel-specific blocker XE991. Furthermore, SCR2682 increases Kv7.2/KCNQ2 mRNA and protein expression in DRG neurons from rats in the SNI model of neuropathic pain. Taken together, pharmacological activation of neuronal Kv7 channels by opener SCR2682 can alleviate pain in rats, thus possessing therapeutic potential for chronic pain or hyperexcitability-related neurologic disorders. SIGNIFICANCE STATEMENT: A novel voltage-gated potassium Kv7 channel opener SCR2682 inhibits action potential firings in dorsal root ganglia sensory neurons and exhibits efficacy in antinociception, thus possessing a developmental potential for treatment of chronic pain or epilepsy.


Subject(s)
Analgesics/therapeutic use , Chronic Pain/drug therapy , KCNQ2 Potassium Channel/metabolism , Membrane Transport Modulators/therapeutic use , Pyridines/therapeutic use , Action Potentials , Analgesics/pharmacology , Animals , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , KCNQ2 Potassium Channel/agonists , Male , Membrane Transport Modulators/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
11.
Mol Pain ; 16: 1744806920955103, 2020.
Article in English | MEDLINE | ID: mdl-32880221

ABSTRACT

Neuropathic pain is a chronic disease state resulting from injury to the nervous system. This type of pain often responds poorly to standard treatments and occasionally may get worse instead of better over time. Patients who experience neuropathic pain report sensitivity to cold and mechanical stimuli. Since the nociceptive system of African naked mole-rats contains unique adaptations that result in insensitivity to some pain types, we investigated whether naked mole-rats may be resilient to sensitivity following nerve injury. Using the spared nerve injury model of neuropathic pain, we showed that sensitivity to mechanical stimuli developed similarly in mice and naked mole-rats. However, naked mole-rats lacked sensitivity to mild cold stimulation after nerve injury, while mice developed robust cold sensitivity. We pursued this response deficit by testing behavior to activators of transient receptor potential (TRP) receptors involved in detecting cold in naïve animals. Following mustard oil, a TRPA1 activator, naked mole-rats responded similarly to mice. Conversely, icilin, a TRPM8 agonist, did not evoke pain behavior in naked mole-rats when compared with mice. Finally, we used RNAscope to probe for TRPA1 and TRPM8 messenger RNA expression in dorsal root ganglia of both species. We found increased TRPA1 messenger RNA, but decreased TRPM8 punctae in naked mole-rats when compared with mice. Our findings likely reflect species differences due to evolutionary environmental responses that are not easily explained by differences in receptor expression between the species.


Subject(s)
Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Neuralgia/metabolism , TRPA1 Cation Channel/metabolism , TRPM Cation Channels/metabolism , Animals , Cold Temperature , Disease Models, Animal , Female , Ganglia, Spinal/injuries , Male , Mice , Mole Rats , Mustard Plant , Neurons/metabolism , Neurons/physiology , Nociception , Pain Measurement , Plant Oils/pharmacology , Pyrimidinones/pharmacology , TRPA1 Cation Channel/genetics , TRPM Cation Channels/agonists , TRPM Cation Channels/genetics
12.
Mol Brain ; 13(1): 73, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32393368

ABSTRACT

Chronic pain can be the result of an underlying disease or condition, medical treatment, inflammation, or injury. The number of persons experiencing this type of pain is substantial, affecting upwards of 50 million adults in the United States. Pharmacotherapy of most of the severe chronic pain patients includes drugs such as gabapentinoids, re-uptake blockers and opioids. Unfortunately, gabapentinoids are not effective in up to two-thirds of this population and although opioids can be initially effective, their long-term use is associated with multiple side effects. Therefore, there is a great need to develop novel non-opioid alternative therapies to relieve chronic pain. For this purpose, we screened a small library of natural products and their derivatives in the search for pharmacological inhibitors of voltage-gated calcium and sodium channels, which are outstanding molecular targets due to their important roles in nociceptive pathways. We discovered that the acetylated derivative of the ent-kaurane diterpenoid, geopyxin A, 1-O-acetylgeopyxin A, blocks voltage-gated calcium and tetrodotoxin-sensitive voltage-gated sodium channels but not tetrodotoxin-resistant sodium channels in dorsal root ganglion (DRG) neurons. Consistent with inhibition of voltage-gated sodium and calcium channels, 1-O-acetylgeopyxin A reduced reduce action potential firing frequency and increased firing threshold (rheobase) in DRG neurons. Finally, we identified the potential of 1-O-acetylgeopyxin A to reverse mechanical allodynia in a preclinical rat model of HIV-induced sensory neuropathy. Dual targeting of both sodium and calcium channels may permit block of nociceptor excitability and of release of pro-nociceptive transmitters. Future studies will harness the core structure of geopyxins for the generation of antinociceptive drugs.


Subject(s)
Calcium Channel Blockers/pharmacology , Ganglia, Spinal/drug effects , Limonins/pharmacology , Neuralgia/drug therapy , Pharmaceutical Preparations/administration & dosage , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium Channels/drug effects , Calcium Channels/physiology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , HIV Infections/drug therapy , HIV Infections/physiopathology , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Hyperalgesia/virology , Limonins/administration & dosage , Limonins/chemistry , Neuralgia/metabolism , Neuralgia/virology , Nociceptors/drug effects , Pharmaceutical Preparations/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channels/drug effects , Sodium Channels/physiology , Tetrodotoxin/pharmacology
13.
J Pain ; 21(3-4): 399-408, 2020.
Article in English | MEDLINE | ID: mdl-31494275

ABSTRACT

The ACCURATE randomized, controlled trial compared outcomes of dorsal root ganglion (DRG) stimulation versus tonic spinal cord stimulation (SCS) in 152 subjects with chronic lower extremity pain due to complex regional pain syndrome (CRPS) type I or II. This ACCURATE substudy was designed to evaluate whether therapy habituation occurs with DRG stimulation as compared to SCS through 12-months. A modified intention-to-treat analysis was performed to assess percentage pain relief (PPR) and responder rates at follow-up visits (end-of-trial, 1, 3, 6, 9, 12-months postpermanent implant) for all subjects that completed trial stimulation (DRG:N = 73, SCS:N = 72). For both groups, mean PPR was significantly greater at end-of-trial (DRG = 82.2%, SCS =0 77.0%) than all other follow-ups. Following permanent DRG system implantation, none of the time points were significantly different from one another in PPR (range = 69.3-73.9%). For the SCS group, PPR at 9-months (58.3%) and 12-months (57.9%) was significantly less than at 1-month (66.9%). The responder rate also decreased for the SCS group from 1-month (68.1%) to 12-months (61.1%). After stratifying by diagnosis, it was found that only the CRPS-I population had diminishing pain relief with SCS. DRG stimulation resulted in more stable pain relief through 12-months, while tonic SCS demonstrated therapy habituation at 9- and 12-months. Trial Registration: The ACCURATE study was registered at ClinicalTrials.gov with Identifier NCT01923285. PERSPECTIVE: This article reports on an ACCURATE substudy, which found that long-term therapy habituation occurred at 12-months with SCS, but not DRG stimulation, in patients with CRPS. The underlying mechanisms of action for these results remain unclear, although several lines of inquiry are proposed.


Subject(s)
Causalgia/therapy , Electric Stimulation Therapy , Ganglia, Spinal , Habituation, Psychophysiologic , Outcome Assessment, Health Care , Reflex Sympathetic Dystrophy/therapy , Spinal Cord Stimulation , Adult , Aged , Female , Follow-Up Studies , Ganglia, Spinal/physiology , Habituation, Psychophysiologic/physiology , Humans , Implantable Neurostimulators , Male , Middle Aged , Time Factors
14.
CNS Neurosci Ther ; 26(1): 136-143, 2020 01.
Article in English | MEDLINE | ID: mdl-31334605

ABSTRACT

AIMS: It is hypothesized that dorsal root ganglion stimulation (DRGS), sharing some of the mechanisms of traditional spinal cord stimulation (SCS) of the dorsal columns, induces γ-aminobutyric acid (GABA) release from interneurons in the spinal dorsal horn. METHODS: We used quantitative immunohistochemical analysis in order to investigate the effect of DRGS on intensity of intracellular GABA-staining levels in the L4-L6 spinal dorsal horn of painful diabetic polyneuropathy (PDPN) animals. To establish the maximal pain relieving effect, we tested for mechanical hypersensitivity to von Frey filaments and animals received 30 minutes of DRGS at day 3 after implantation of the electrode. One day later, 4 Sham-DRGS animals and four responders-to-DRGS received again 30 minutes of DRGS and were perfused at the peak of DRGS-induced pain relief. RESULTS: No significant difference in GABA-immunoreactivity was observed between DRGS and Sham-DRGS in lamina 1-3 of the spinal levels L4-6 neither ipsilaterally nor contralaterally. CONCLUSIONS: Dorsal root ganglion stimulation does not induce GABA release from the spinal dorsal horn cells, suggesting that the mechanisms underlying DRGS in pain relief are different from those of conventional SCS. The modulation of a GABA-mediated "Gate Control" in the DRG itself, functioning as a prime Gate of nociception, is suggested and discussed.


Subject(s)
Diabetic Neuropathies/complications , Electric Stimulation Therapy/methods , Ganglia, Spinal/physiology , Pain Management/methods , Spinal Cord Dorsal Horn/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Electrodes, Implanted , Female , Hyperalgesia , Pain/etiology , Pain Measurement , Rats , Rats, Sprague-Dawley
16.
Pain Med ; 20(Suppl 1): S41-S46, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31152174

ABSTRACT

OBJECTIVE: The objective of this study is to review the available evidence for dorsal root ganglion (DRG) stimulation for the treatment of complex regional pain syndrome type II (CRPS II; peripheral causalgia) associated with chronic neuropathic postsurgical pain (NPP). DESIGN: Available literature was identified through a search of the US National Library of Medicine's Medline database, PubMed.gov. References from published articles also were reviewed for relevant citations. RESULTS: The data published to date support the use of DRG stimulation to treat chronic NPP of the groin, knee, and foot. NPP following procedures such as thoracotomy, hernia surgery, and knee replacement surgery were identified as some of the conditions for which DRG stimulation is likely to be effective. CONCLUSION: DRG stimulation is known to be an effective treatment for focal neuropathic pain. Currently, NPP of the foot, groin, and knee all appear to be the conditions with the most clinical experience, backed by a limited but growing body of evidence. However, prospective studies lag behind real-world clinical experience and are needed to confirm these findings.


Subject(s)
Causalgia/therapy , Electric Stimulation Therapy/methods , Ganglia, Spinal/physiology , Pain Management/methods , Pain, Postoperative/therapy , Chronic Pain/therapy , Humans , Neuralgia/therapy
17.
Pain Med ; 20(Suppl 1): S23-S30, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31152179

ABSTRACT

OBJECTIVE: The dorsal root ganglion (DRG) is a novel target for neuromodulation, and DRG stimulation is proving to be a viable option in the treatment of chronic intractable neuropathic pain. Although the overall principle of conventional spinal cord stimulation (SCS) and DRG stimulation-in which an electric field is applied to a neural target with the intent of affecting neural pathways to decrease pain perception-is similar, there are significant differences in the anatomy and physiology of the DRG that make it an ideal target for neuromodulation and may account for the superior outcomes observed in the treatment of certain chronic neuropathic pain states. This review highlights the anatomy of the DRG, its function in maintaining homeostasis and its role in neuropathic pain, and the unique value of DRG as a target in neuromodulation for pain. METHODS: A narrative literature review was performed. RESULTS: Overall, the DRG is a critical structure in sensory transduction and modulation, including pain transmission and the maintenance of persistent neuropathic pain states. Unique characteristics including selective somatic organization, specialized membrane characteristics, and accessible and consistent location make the DRG an ideal target for neuromodulation. Because DRG stimulation directly recruits the somata of primary sensory neurons and harnesses the filtering capacity of the pseudounipolar neural architecture, it is differentiated from SCS, peripheral nerve stimulation, and other neuromodulation options. CONCLUSIONS: There are several advantages to targeting the DRG, including lower energy usage, more focused and posture-independent stimulation, reduced paresthesia, and improved clinical outcomes.


Subject(s)
Electric Stimulation Therapy/methods , Ganglia, Spinal/physiology , Neuralgia/therapy , Chronic Pain/therapy , Ganglia, Spinal/anatomy & histology , Humans
18.
J Transl Med ; 17(1): 205, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217010

ABSTRACT

BACKGROUND: In our recent clinical trial, increased peripheral concentrations of pro-inflammatory molecular mediators were determined in complex regional pain syndrome (CRPS) patients. After 3 months adjunctive unilateral, selective L4 dorsal root ganglion stimulation (L4-DRGSTIM), significantly decreased serum IL-10 and increased saliva oxytocin levels were assessed along with an improved pain and functional state. The current study extended molecular profiling towards gene expression analysis of genes known to be involved in the gonadotropin releasing hormone receptor and neuroinflammatory (cytokines/chemokines) signaling pathways. METHODS: Blood samples were collected from 12 CRPS patients for whole-transcriptome profiling in order to assay 18,845 inflammation-associated genes from frozen blood at baseline and after 3 months L4-DRGSTIM using PANTHER™ pathway enrichment analysis tool. RESULTS: Pathway enrichment analyses tools (GOrilla™ and PANTHER™) showed predominant involvement of inflammation mediated by chemokines/cytokines and gonadotropin releasing hormone receptor pathways. Further, screening of differentially regulated genes showed changes in innate immune response related genes. Transcriptomic analysis showed that 21 genes (predominantly immunoinflammatory) were significantly changed after L4-DRGSTIM. Seven genes including TLR1, FFAR2, IL1RAP, ILRN, C5, PKB and IL18 were down regulated and fourteen genes including CXCL2, CCL11, IL36G, CRP, SCGB1A1, IL-17F, TNFRSF4, PLA2G2A, CREB3L3, ADAMTS12, IL1F10, NOX1, CHIA and BDKRB1 were upregulated. CONCLUSIONS: In our sub-group analysis of L4-DRGSTIM treated CRPS patients, we found either upregulated or downregulated genes involved in immunoinflammatory circuits relevant for the pathophysiology of CRPS indicating a possible relation. However, large biobank-based approaches are recommended to establish genetic phenotyping as a quantitative outcome measure in CRPS patients. Trial registration The study protocol was registered at the 15.11.2016 on German Register for Clinical Trials (DRKS ID 00011267). https://www.drks.de/drks_web/navigate.do?navigationId=trial.HTML&TRIAL_ID=DRKS00011267.


Subject(s)
Chronic Pain/therapy , Complex Regional Pain Syndromes/therapy , Inflammation/blood , Inflammation/genetics , Neuralgia/therapy , Pain Management/methods , Transcutaneous Electric Nerve Stimulation/methods , Aged , Biomarkers/blood , Biomarkers/metabolism , Chronic Pain/blood , Complex Regional Pain Syndromes/blood , Complex Regional Pain Syndromes/genetics , Complex Regional Pain Syndromes/metabolism , Cytokines/blood , Cytokines/genetics , Female , Ganglia, Spinal/physiology , Gene Expression Profiling , Humans , Inflammation/etiology , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Knee/pathology , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Neuralgia/blood , Pain, Postoperative/blood , Pain, Postoperative/etiology , Pain, Postoperative/therapy , Saliva/chemistry , Saliva/metabolism
19.
J Ethnopharmacol ; 233: 56-63, 2019 Apr 06.
Article in English | MEDLINE | ID: mdl-30599222

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Strychnos nux-vomica L. (Loganiaceae) is grown extensively in South Asian. The dried seed of this plant, nux vomica, has been clinically used in Chinese medicine for relieving rheumatic pain, reducing swelling and treating cancer. Brucine, the second abundant alkaloid constituent of nux vomica, shows excellent clinical therapeutic effect, especially in relieving pain, but mechanism of brucine in relieving pain is still unclear. AIM OF THE STUDY: Explore the analgesic effect of brucine, reveal the molecular mechanism of brucine analgesia. MATERIALS AND METHODS: Antinociceptive effects of brucine were assessed in acute and chronic pain mice model. Electrophysiological experiments were used to evaluate the effects of brucine on neuronal activity and sodium channel function. RESULTS: In acute pain models, brucine significantly inhibits response induced by nociceptive heat and mechanical stimulation. Furthermore, thermal hypersensitivity and mechanical allodynia were also alleviated by brucine treatment in a chronic constriction injury (CCI) mouse model. Sodium channel plays a crucial role in neuropathic pain. Electrophysiological results show that brucine inhibits the excitability of DRG neurons directly, the number of action potential (AP) was significantly reduced after brucine treatment, and this kind of inhibition is due to brucine inhibits both tetrodotoxin-sensitive (TTXs) and tetrodotoxin-resistant (TTXr) sodium channel. CONCLUSIONS: Taken together, brucine is a novel drug candidate in treating acute and chronic pain diseases, which might be attributed to inhibition the excitability of sodium channel directly.


Subject(s)
Analgesics/pharmacology , Analgesics/therapeutic use , Neuralgia/drug therapy , Sodium Channels/physiology , Strychnine/analogs & derivatives , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Male , Mice, Inbred C57BL , Neuralgia/physiopathology , Neurons/drug effects , Neurons/physiology , Strychnine/pharmacology , Strychnine/therapeutic use
20.
ACS Appl Mater Interfaces ; 11(1): 356-372, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30516370

ABSTRACT

Magnetic electrospun fibers are of interest for minimally invasive biomaterial applications that also strive to provide cell guidance. Magnetic electrospun fibers can be injected and then magnetically positioned in situ, and the aligned fiber scaffolds provide consistent topographical guidance to cells. In this study, magnetically responsive aligned poly-l-lactic acid electrospun fiber scaffolds were developed and tested for neural applications. Incorporating oleic acid-coated iron oxide nanoparticles significantly increased neurite outgrowth, reduced the fiber alignment, and increased the surface nanotopography of the electrospun fibers. After verifying neuron viability on two-dimensional scaffolds, the system was tested as an injectable three-dimensional scaffold. Small conduits of aligned magnetic fibers were easily injected in a collagen or fibrinogen hydrogel solution and repositioned using an external magnetic field. The aligned magnetic fibers provided internal directional guidance to neurites within a three-dimensional collagen or fibrin model hydrogel, supplemented with Matrigel. Neurites growing from dorsal root ganglion explants extended 1.4-3× farther on the aligned fibers compared with neurites extending in the hydrogel alone. Overall, these results show that magnetic electrospun fiber scaffolds can be injected and manipulated with a magnetic field in situ to provide directional guidance to neurons inside an injectable hydrogel. Most importantly, this injectable guidance system increased both neurite alignment and neurite length within the hydrogel scaffold.


Subject(s)
Ganglia, Spinal/physiology , Hydrogels/chemistry , Nerve Regeneration , Neurites/metabolism , Tissue Scaffolds/chemistry , Animals , Ganglia, Spinal/cytology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL