1
|
Villalba-Riquelme E, de la Torre-Martínez R, Fernández-Carvajal A, Ferrer-Montiel A. Oxaliplatin reversibly and differentially affects electrogenic activity of small IB4(+) of male and female rat sensory neurons. Biomed Pharmacother 2025; 183:117849. [PMID: 39914991 DOI: 10.1016/j.biopha.2025.117849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 05/07/2025] Open
Abstract
Oxaliplatin-induced peripheral neuropathy (OIPN) is a prevalent drug side effect with unclear molecular mechanism and sex differences. We report that a 48-hour exposure of 1 µg·mL-1 oxaliplatin potently and reversibly increased the excitability of IB4(+) male and female nociceptors. In females, oxaliplatin augmented spontaneous and tonic activity, increased the overshoot and amplitude of action potentials, depolarized the membrane potential, reduced the rheobase, and raised the firing frequency, primarily due to the depolarization of KV currents and a mild up-regulation of TTX-resistant (TTX-r) NaV currents. In males, oxaliplatin reduced the rheobase, augmented the firing frequency, and prolonged the action potential peak time, mediated by increased TTX-r NaV currents, hyperpolarization of their activation curve, and a faster recovery from inactivation. Oxaliplatin also up-regulated TRPV1 currents in male and female IB4(+) nociceptors and augmented TRPA1 responses in male IB4(+) neurons. These findings highlight NaV1.8, TRPV1, and TRPA1 as therapeutic targets to mitigate OIPN and validate the use of long-term nociceptor cultures as preclinical models for studying peripheral neuropathies.
Collapse
Affiliation(s)
- Eva Villalba-Riquelme
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE). Universitas Miguel Hernández, Elche 03202, Spain.
| | | | - Asia Fernández-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE). Universitas Miguel Hernández, Elche 03202, Spain
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE). Universitas Miguel Hernández, Elche 03202, Spain.
| |
Collapse
|
2
|
Kim H, Roh D, Oh SB. EGFR Tyrosine Kinase Inhibitor Lazertinib Activates a Subset of Mouse Sensory Neurons Via TRPA1. THE JOURNAL OF PAIN 2024; 25:104435. [PMID: 38008390 DOI: 10.1016/j.jpain.2023.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/21/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Lazertinib (JNJ-73841937, YH25448) is a mutant-selective irreversible epidermal growth factor receptor tyrosine kinase inhibitor targeting both the T790M and activating mutation while sparing wild-type epidermal growth factor receptor. Paresthesia is one of the most common adverse events seen with lazertinib treatment, suggesting that lazertinib could affect the sensory nervous system. However, the mechanism of action for this paresthesia remains unclear. In this study, we investigated whether and how lazertinib affects peripheral sensory neurons. Through Fura-2-based calcium imaging and whole-cell patch clamp recording in primary-cultured dorsal root ganglion (DRG) neurons from adult mice, we found that application of lazertinib elicits spontaneous calcium responses in a subset of small-to-medium-sized neurons. Moreover, lazertinib induced spontaneous firings and hyperexcitability in a subset of transient receptor potential vanilloid 1-lineage DRG neurons and sensitized transient receptor potential ankyrin 1 (TRPA1) response, while sparing transient receptor potential vanilloid 1 response. Lazertinib-responsive neurons were also responsive to capsaicin, further supporting that lazertinib selectively activates nociceptive neurons. Lazertinib-induced calcium responses were pharmacologically blocked with HC-030031 (TRPA1 antagonist) and MDL-12330A (adenylyl cyclase inhibitor), suggesting that lazertinib activates sensory neurons through indirect activation of TRPA1. However, unlike vincristine which produces peripheral neuropathy by axonal degeneration, lazertinib did not cause neurite fragmentation in cultured DRG neurons. Finally, intraplantar injection of lazertinib induced TRPA1-dependent pain-like behaviors in vivo. Collectively, our data suggest a direct effect of lazertinib on nociceptive sensory neurons via TRPA1 selective mechanisms, which could be a putative mechanism of lazertinib-induced sensory abnormalities in clinical patients. PERSPECTIVE: This article presents a TRPA1-dependent, lazertinib-induced activation of mouse sensory neurons in vitro and lazertinib-induced pain-like behaviors in vivo. The same mechanisms may underlie the clinical condition, suggesting that TRPA1 could be a potential therapeutic target to manage lazertinib-induced paresthesia.
Collapse
Affiliation(s)
- Hayun Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea
| | - Dahee Roh
- Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Seog Bae Oh
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul 08826, Republic of Korea; Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
3
|
Gehr NL, Karlsson P, Timm S, Christensen S, Hvid CA, Peric J, Hansen TF, Lauritzen L, Finnerup NB, Ventzel L. Study protocol: fish oil supplement in prevention of oxaliplatin-induced peripheral neuropathy in adjuvant colorectal cancer patients - a randomized controlled trial. (OxaNeuro). BMC Cancer 2024; 24:168. [PMID: 38308227 PMCID: PMC10837958 DOI: 10.1186/s12885-024-11856-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/08/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Oxaliplatin-induced peripheral neuropathy (OIPN) in general and painful OIPN in particular is a debilitating late effect that severely affects cancer survivors' quality of life and causes premature cessation of potentially lifesaving treatment. No preventive treatments and no effective treatment for chronic OIPN exist despite many attempts. One of several suggested mechanisms includes neuroinflammation as a contributing factor to OIPN. Fish oil containing long-chain n-3 polyunsaturated fatty acids (n-3 LCPUFAs) are precursors to specialized proresolving mediators that mediate the resolution of inflammation. Our primary hypothesis is that a high supplementation of n-3 LCPUFAs will lower the prevalence and severity of OIPN. METHODS The OxaNeuro project is an investigator-initiated, multicenter, double-blinded, randomized, placebo-controlled clinical study. We will include 120 patients eligible to receive adjuvant oxaliplatin after colorectal cancer surgery. Patients will receive fish oil capsules containing n-3 LCPUFAs or corn oil daily for 8 months. The primary endpoint is the prevalence of OIPN at 8 months defined as relevant symptoms, including one of the following: abnormal nerve conduction screening, abnormal vibration threshold test, abnormal skin biopsy, or abnormal pinprick test. Additional endpoints include the intensity and severity of OIPN-related neuropathic pain, patient-reported OIPN symptoms, quality of life, mental health symptoms, body composition, and cognitive evaluation. Furthermore, we will evaluate inflammatory biomarkers in blood samples and skin biopsies, including the potential OIPN biomarker neurofilament light protein (NfL) which will be measured before each cycle of chemotherapy. DISCUSSION If readily available fish oil supplementation alleviates OIPN prevalence and severity, it will significantly improve the lives of both cancer survivors and palliative cancer patients receiving oxaliplatin; it will improve their quality of life, optimize chemotherapeutic treatment plans by lowering the need for dose reduction or premature cessation, and potentially increase survival. TRIAL REGISTRATION ClinicalTrial.gov identifier: NCT05404230 Protocol version: 1.2, April 25th. 2023.
Collapse
Affiliation(s)
- Nina Lykkegaard Gehr
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark.
| | - Páll Karlsson
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Signe Timm
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Signe Christensen
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Jana Peric
- Department of Oncology, Soenderborg Hospital, University Hospital of Southern Denmark, Soenderborg, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Lotte Lauritzen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Brix Finnerup
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Lise Ventzel
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| |
Collapse
|
4
|
Matsuda S, Igawa Y, Uchisawa H, Iki S, Osumi M. Thermal Grill Illusion in Post-Stroke Patients: Analysis of Clinical Features and Lesion Areas. J Pain Res 2023; 16:3895-3904. [PMID: 38026455 PMCID: PMC10656876 DOI: 10.2147/jpr.s433309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose In the thermal grill illusion, participants experience a feeling similar to burning pain. The illusion is induced by simultaneously touching warm and cool stimuli in alternating positions. In post-stroke pain, central sensitization is caused by a variety of factors, including damage to the spinothalamic tract and shoulder pain. Because the thermal grill illusion depends on central mechanisms, it has recently been suggested that it may be a useful indicator of central sensitization. Therefore, we hypothesized that post-stroke patients who are more likely to experience central sensitization may also be more likely to experience a thermal grill sensation of pain and discomfort than the likelihood among those who are less likely to experience central sensitization. However, the effects of the thermal grill illusion in post-stroke patients have not yet been reported. In this pilot study, we conducted the thermal grill illusion procedure in post-stroke patients and analyzed the relationship between clinical somatosensory functions and thermal grill sensations. We also conducted brain imaging analysis to identify brain lesion areas that were associated with thermal grill sensations. Patients and Methods Twenty patients (65.7 ± 11.9 years old) with post-stroke patients participated in this study. The thermal grill illusion procedure was performed as follows: patients simultaneously touched eight water-filled copper bars, with the water temperature adjusted to provide alternate warm (40°C) and cold (20°C) stimuli. Results Thermal grill sensation of pain and discomfort tended to be associated with the wind-up phenomenon in bedside quantitative sensory testing and thermal grill sensation of discomfort was also related to damage to the thalamic lateral nucleus. Conclusion These findings suggest that the thermal grill illusion might measure central sensitization, and that secondary brain hyperactivity might lead to increased thermal grill sensations.
Collapse
Affiliation(s)
| | - Yuki Igawa
- Graduate School of Health Sciences, Kio University, Nara, Japan
- Department of Rehabilitation, Nishiyamato Rehabilitation Hospital, Nara, Japan
| | - Hidekazu Uchisawa
- Graduate School of Health Sciences, Kio University, Nara, Japan
- Department of Rehabilitation, Nishiyamato Rehabilitation Hospital, Nara, Japan
| | - Shinya Iki
- Department of Rehabilitation, Kawaguchi Neurosurgery Rehabilitation Clinic, Osaka, Japan
| | - Michihiro Osumi
- Graduate School of Health Sciences, Kio University, Nara, Japan
- Neurorehabilitation Research Center, Kio University, Nara, Japan
| |
Collapse
|
5
|
Adam F, Jouët P, Sabaté JM, Perrot S, Franchisseur C, Attal N, Bouhassira D. Thermal grill illusion of pain in patients with chronic pain: a clinical marker of central sensitization? Pain 2023; 164:638-644. [PMID: 35972466 DOI: 10.1097/j.pain.0000000000002749] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT The thermal grill illusion of pain (TGIP) is a paradoxical burning pain sensation elicited by the simultaneous application of innocuous cutaneous warm and cold stimuli with a thermode ("thermal grill") consisting of interlaced heated and cooled bars. Its neurophysiological mechanisms are unclear, but TGIP may have some mechanisms in common with pathological pain, including central sensitization in particular, through the involvement of N-methyl- d -aspartate receptors. However, few studies have investigated TGIP in patients with chronic pain and its clinical relevance is uncertain. We hypothesized that the TGIP would be increased in comparison with controls in patients with fibromyalgia or irritable bowel syndrome, which are regarded as typical "nociplastic" primary pain syndromes related to changes in central pain processing. We compared the sensations elicited by a large range of combinations of temperature differentials between the warm and cold bars of a thermal grill applied to the hand between patients with fibromyalgia (n = 30) or irritable bowel syndrome (n= 30) and controls (n = 30). The percentage of TGIP responses and the intensity and unpleasantness of TGIP were significantly greater in patients than controls. Furthermore, positive correlations were found between TGIP intensity and clinical pain intensity and between TGIP intensity and the cold pain threshold measured on the hand. These results are consistent with our working hypothesis of shared mechanisms between TGIP and clinical pain mechanisms in patients with nociplastic chronic pain syndromes and suggest that TGIP might represent a clinical marker of central sensitization in these patients.
Collapse
Affiliation(s)
- Frédéric Adam
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
- Department of Anesthesiology, Saint Joseph Hospital, Paris, France
| | - Pauline Jouët
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
- Gastroenterology and Digestive Oncology Department, Avicenne Hospital, AP-HP, Bobigny, France
| | - Jean-Marc Sabaté
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
- Gastroenterology and Digestive Oncology Department, Avicenne Hospital, AP-HP, Bobigny, France
| | - Serge Perrot
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
- Pain Clinic, Cochin Hospital, University of Paris, Paris, France
| | - Claire Franchisseur
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Nadine Attal
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
| | - Didier Bouhassira
- Inserm U987, APHP, UVSQ, Paris-Saclay University, Ambroise Pare Hospital, Boulogne-Billancourt, France
| |
Collapse
|
6
|
Cold evoked potentials elicited by rapid cooling of the skin in young and elderly healthy individuals. Sci Rep 2022; 12:4137. [PMID: 35264694 PMCID: PMC8907280 DOI: 10.1038/s41598-022-07967-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/21/2022] [Indexed: 11/08/2022] Open
Abstract
Cold-evoked potentials (CEPs) constitute a novel electrophysiological tool to assess cold-specific alterations in somatosensory function. As an important step towards the clinical implementation of CEPs as a diagnostic tool, we evaluated the feasibility and reliability of CEPs in response to rapid cooling of the skin (-300 °C/s) and different stimulation sites in young and elderly healthy individuals. Time-locked electroencephalographic responses were recorded from at vertex in fifteen young (20-40 years) and sixteen elderly (50-70 years), individuals in response to 15 rapid cold stimuli (-300 °C/s) applied to the skin of the hand dorsum, palm, and foot dorsum. High CEP proportions were shown for young individuals at all sites (hand dorsum/palm: 100% and foot: 79%) and elderly individuals after stimulation of the hand dorsum (81%) and palm (63%), but not the foot (44%). Depending on the age group and stimulation site, test-retest reliability was "poor" to "substantial" for N2P2 amplitudes and N2 latencies. Rapid cooling of the skin enables the recording of reliable CEPs in young individuals. In elderly individuals, CEP recordings were only robust after stimulation of the hand, but particularly challenging after stimulation of the foot. Further improvements in stimulation paradigms are warranted to introduce CEPs for clinical diagnostics.
Collapse
|
7
|
New diagnostic measures of oxaliplatin-induced peripheral sensory neuropathy. Cancer Treat Res Commun 2022; 31:100543. [PMID: 35255440 DOI: 10.1016/j.ctarc.2022.100543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Oxaliplatin-induced peripheral neuropathy (OIPN) is an unwanted side effect of oxaliplatin chemotherapy treatment. OIPN manifests in an acute phase that lasts a few days after injection and a persistent phase that may become chronic. Currently, there is no consensus about a clinically applicable, quantitative, and objective measure of OIPN. METHODS Seventeen patients treated with oxaliplatin containing adjuvant chemotherapy for stage III colon cancer, but otherwise healthy, were tested with six quantitative sensory tests (QST) and five large fibre perception threshold tracking (PTT) measures (quantified by, e.g., rheobase and electrotonus threshold) one hour before each of the 12 chemotherapy cycles given at two weeks' intervals. These measures were repeated at 3, 6, and 12-month follow-ups. The temporal development of OIPN assessed by the Common Terminology Criteria for Adverse Events (CTCAE) scale, QST, and PTT measures was calculated by linear regression. RESULTS The CTCAE score showed a tri-phasic increase during the treatment and remained increased during the follow-up. The vibration threshold (R = 0.25, p<0.001), the cold pain threshold (R = 0.17, p = 0.02), and the rheobase (R = 0.28, p < 0.001) increased during treatment, whereas the cold detection threshold (R=-0.16, p = 0.002) decreased. The cold pain threshold and the rheobase remained increased, and the cold detection and heat pain threshold remained decreased during follow-up. CONCLUSIONS Increased cold pain sensitivity and decreased large fibre sensitivity (increased rheobase) correlate to the persistent OIPN, whereas the CTCAE score assesses both acute and persistent OIPN. Furthermore, the novel PTT method assessed the nerve excitability changes caused by the oxaliplatin.
Collapse
|
8
|
Dai R, Lu C, Yun L, Lenze E, Avidan M, Kannampallil T. Comparing stress prediction models using smartwatch physiological signals and participant self-reports. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 208:106207. [PMID: 34161847 DOI: 10.1016/j.cmpb.2021.106207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/24/2021] [Indexed: 06/13/2023]
Abstract
Recent advances in wearable technology have facilitated the non-obtrusive monitoring of physiological signals, creating opportunities to monitor and predict stress. Researchers have utilized machine learning methods using these physiological signals to develop stress prediction models. Many of these prediction models have utilized objective stressor tasks (e.g., a public speaking task or solving math problems). Alternatively, the subjective user responses with self-reports have also been used for measuring stress. In this paper, we describe a methodological approach (a) to compare the prediction performance of models developed using objective markers of stress using participant-reported subjective markers of stress from self-reports; and (b) to develop personalized stress models by accounting for inter-individual differences. Towards this end, we conducted a laboratory-based study with 32 healthy volunteers. Participants completed a series of stressor tasks-social, cognitive and physical-wearing an instrumented commercial smartwatch that collected physiological signals and participant responses using timed self-reports. After extensive data preprocessing using a combination of signal processing techniques, we developed two types of models: objective stress models using the stressor tasks as labels; and subjective stress models using participant responses to each task as the label for that stress task. We trained and tested several machine learning algorithms-support vector machine (SVM), random forest (RF), gradient boosted trees (GBT), AdaBoost, and Logistic Regression (LR)-and evaluated their performance. SVM had the best performance for the models using the objective stressor (i.e., stressor tasks) with an AUROC of 0.790 and an F-1 score of 0.623. SVM also had the highest performance for the models using the subjective stress (i.e., participant self-reports) with an AUROC of 0.719 and an F-1 score of 0.520. Model performance improved with a personalized threshold model to an AUROC of 0.751 and an F-1 score of 0.599. The performance of the stress models using an instrumented commercial smartwatch was comparable to similar models from other state-of-the-art laboratory-based studies. However, the subjective stress models had a lower performance, indicating the need for further research on the use of self-reports for stress-related studies. The improvement in performance with the personalized threshold-based models provide new directions for building stress prediction models.
Collapse
Affiliation(s)
- Ruixuan Dai
- Department of Computer Science, McKelvey School of Engineering, USA
| | - Chenyang Lu
- Department of Computer Science, McKelvey School of Engineering, USA
| | | | | | | | - Thomas Kannampallil
- Department of Anesthesiology, USA; Institute for Informatics, School of Medicine, Washington University in St. Louis, St Louis, MO, USA.
| |
Collapse
|
9
|
Doshi TL, Dworkin RH, Polomano RC, Carr DB, Edwards RR, Finnerup NB, Freeman RL, Paice JA, Weisman SJ, Raja SN. AAAPT Diagnostic Criteria for Acute Neuropathic Pain. PAIN MEDICINE 2021; 22:616-636. [PMID: 33575803 DOI: 10.1093/pm/pnaa407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Acute neuropathic pain is a significant diagnostic challenge, and it is closely related to our understanding of both acute pain and neuropathic pain. Diagnostic criteria for acute neuropathic pain should reflect our mechanistic understanding and provide a framework for research on and treatment of these complex pain conditions. METHODS The Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership with the U.S. Food and Drug Administration (FDA), the American Pain Society (APS), and the American Academy of Pain Medicine (AAPM) collaborated to develop the ACTTION-APS-AAPM Pain Taxonomy (AAAPT) for acute pain. A working group of experts in research and clinical management of neuropathic pain was convened. Group members used literature review and expert opinion to develop diagnostic criteria for acute neuropathic pain, as well as three specific examples of acute neuropathic pain conditions, using the five dimensions of the AAAPT classification of acute pain. RESULTS AAAPT diagnostic criteria for acute neuropathic pain are presented. Application of these criteria to three specific conditions (pain related to herpes zoster, chemotherapy, and limb amputation) illustrates the spectrum of acute neuropathic pain and highlights unique features of each condition. CONCLUSIONS The proposed AAAPT diagnostic criteria for acute neuropathic pain can be applied to various acute neuropathic pain conditions. Both the general and condition-specific criteria may guide future research, assessment, and management of acute neuropathic pain.
Collapse
Affiliation(s)
- Tina L Doshi
- Department of Anesthesiology and Critical Care Medicine, Division of Pain Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Robert H Dworkin
- Department of Anesthesiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, and Department of Neurology, Center for Human Experimental Therapeutics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Rosemary C Polomano
- Division of Biobehavioral Health Sciences, University of Pennsylvania-School of Nursing, Philadelphia, Pennsylvania, USA
| | - Daniel B Carr
- Public Health and Community Medicine Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Robert R Edwards
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, and Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Roy L Freeman
- Center for Autonomic and Peripheral Nerve Disorders, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Judith A Paice
- Cancer Pain Program, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Steven J Weisman
- Jane B. Pettit Pain and Headache Center, Children's Wisconsin, Departments of Anesthesiology and Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, Division of Pain Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Efficacy assessment of salicylidene salicylhydrazide in chemotherapy associated peripheral neuropathy. Eur J Pharmacol 2020; 888:173481. [DOI: 10.1016/j.ejphar.2020.173481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
|
11
|
Forstenpointner J, Berry D, Baron R, Borsook D. The cornucopia of central disinhibition pain - An evaluation of past and novel concepts. Neurobiol Dis 2020; 145:105041. [PMID: 32800994 DOI: 10.1016/j.nbd.2020.105041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/18/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
Abstract
Central disinhibition (CD), as applied to pain, decreases thresholds of endogenous systems. This provokes onset of spontaneous or evoked pain in an individual beyond the ability of the nervous system to inhibit pain resulting from a disease or tissue damage. The original CD concept as proposed by Craig entails a shift from the lateral pain pathway (i.e. discriminative pain processing) towards the medial pain pathway (i.e. emotional pain processing), within an otherwise neurophysiological intact environment. In this review, the original CD concept as proposed by Craig is extended by the primary "nociceptive pathway damage - CD" concept and the secondary "central pathway set point - CD". Thereby, the original concept may be transferred into anatomical and psychological non-functional conditions. We provide examples for either primary or secondary CD concepts within different clinical etiologies as well as present surrogate models, which directly mimic the underlying pathophysiology (A-fiber block) or modulate the CD pathway excitability (thermal grill). The thermal grill has especially shown promising advancements, which may be useful to examine CD pathway activation in the future. Therefore, within this topical review, a systematic review on the thermal grill illusion is intended to stimulate future research. Finally, the authors review different mechanism-based treatment approaches to combat CD pain.
Collapse
Affiliation(s)
- Julia Forstenpointner
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany; Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA.
| | - Delany Berry
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
| | - David Borsook
- Center for Pain and the Brain, Boston Children's Hospital, Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
13
|
Bennedsgaard K, Ventzel L, Grafe P, Tigerholm J, Themistocleous AC, Bennett DL, Tankisi H, Finnerup NB. Cold aggravates abnormal excitability of motor axons in oxaliplatin-treated patients. Muscle Nerve 2020; 61:796-800. [PMID: 32133655 PMCID: PMC7318596 DOI: 10.1002/mus.26852] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Cold allodynia is often seen in the acute phase of oxaliplatin treatment, but the underlying pathophysiology remains unclear. METHODS Patients scheduled for adjuvant oxaliplatin for colorectal cancer were examined with quantitative sensory testing and nerve excitability tests at baseline and after the second or third oxaliplatin cycle at different skin temperatures. RESULTS Seven patients were eligible for examination. All patients felt evoked pain and tingling when touching something cold after oxaliplatin infusion. Oxaliplatin decreased motor nerve superexcitability (P < .001), increased relative refractory period (P = .011), and caused neuromyotonia-like after-activity. Cooling exacerbated these changes and prolonged the accommodation half-time. DISCUSSION The findings suggest that a combined effect of oxaliplatin and cooling facilitates nerve excitability changes and neuromyotonia-like after-activity in peripheral nerve axons. A possible mechanism is the slowing in gating of voltage-dependent fast sodium and slow potassium channels, which results in symptoms of cold allodynia.
Collapse
Affiliation(s)
- Kristine Bennedsgaard
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lise Ventzel
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Peter Grafe
- Institute of Physiology, Ludwig-Maximilians University, Munich, Germany
| | - Jenny Tigerholm
- Center of Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Hatice Tankisi
- Department of Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Rosner J, Rinert J, Ernst M, Curt A, Hubli M. Cold evoked potentials: Acquisition from cervical dermatomes. Neurophysiol Clin 2019; 49:49-57. [DOI: 10.1016/j.neucli.2018.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/06/2023] Open
|
15
|
Fardo F, Finnerup NB, Haggard P. Organization of the Thermal Grill Illusion by Spinal Segments. Ann Neurol 2018; 84:463-472. [PMID: 30063258 PMCID: PMC6175302 DOI: 10.1002/ana.25307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Objective A common symptom of neuropathy is the misperception of heat and pain from cold stimuli. Similar cold allodynic sensations can be experimentally induced using the thermal grill illusion (TGI) in humans. It is currently unclear whether this interaction between thermosensory and nociceptive signals depends on spinal or supraspinal integration mechanisms. To address this issue, we developed a noninvasive protocol to assess thermosensory integration across spinal segments. Methods We leveraged anatomical knowledge regarding dermatomes and their spinal projections to investigate potential contributions of spinal integration to the TGI. We simultaneously stimulated a pair of skin locations on the arm or lower back using 1 cold (∼20°C) and 1 warm thermode (∼40°C). The 2 thermodes were always separated by a fixed physical distance on the skin, but elicited neural activity across a varying number of spinal segments, depending on which dermatomal boundaries the 2 stimuli spanned. Results Participants consistently overestimated the actual cold temperature on the skin during combined cold and warm stimulation, confirming the TGI effect. The TGI was present when cold and warm stimuli were delivered within the same dermatome, or across dermatomes corresponding to adjacent spinal segments. In striking contrast, no TGI effect was found when cold and warm stimuli projected to nonadjacent spinal segments. Interpretation These results demonstrate that the strength of the illusion is modulated by the segmental distance between cold and warm afferents. This suggests that both temperature perception and thermal–nociceptive interactions depend upon low‐level convergence mechanisms operating within a single spinal segment and its immediate neighbors. Ann Neurol 2018;84:463–472
Collapse
Affiliation(s)
- Francesca Fardo
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom.,Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Interacting Minds Center, Aarhus University, Aarhus, Denmark
| | - Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Patrick Haggard
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom
| |
Collapse
|
16
|
Selective cold pain inhibition by targeted block of TRPM8-expressing neurons with quaternary lidocaine derivative QX-314. Commun Biol 2018; 1:53. [PMID: 30271936 PMCID: PMC6123689 DOI: 10.1038/s42003-018-0062-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/02/2018] [Indexed: 01/04/2023] Open
Abstract
Treatment of pain with local anesthetics leads to an unfavorable decrease in general sensory acuity due to their indiscriminate block of both pain sensing (nociceptors) and non-pain sensing nerves. However, the cell impermeant lidocaine derivative QX-314 can be selectively targeted to only nociceptors by permeation through ligand-gated cation channels. Here we show that localized injection of QX-314 with agonists for the menthol receptor TRPM8 specifically blocks cold-evoked behaviors in mice, including cold allodynia and hyperalgesia. Remarkably, cooling stimuli also promotes QX-314-mediated inhibition of cold behaviors, and can be used to block cold allodynia, while retaining relatively normal cold sensation. The effects of both agonist and thermally evoked uptake of QX-314 are TRPM8-dependent, results demonstrating an effective approach to treat localized cold pain without altering general somatosensation. Serra Ongun, Angela Sarkisian and David McKemy show that localized co-injection of lidocaine derivative QX-314 and receptor agonists is able to block cold sensitivity in mice in a targeted way, with implications for treating cold pain associated with injury and disease.
Collapse
|
17
|
Glimelius B, Manojlovic N, Pfeiffer P, Mosidze B, Kurteva G, Karlberg M, Mahalingam D, Buhl Jensen P, Kowalski J, Bengtson M, Nittve M, Näsström J. Persistent prevention of oxaliplatin-induced peripheral neuropathy using calmangafodipir (PledOx ®): a placebo-controlled randomised phase II study (PLIANT). Acta Oncol 2018; 57:393-402. [PMID: 29140155 DOI: 10.1080/0284186x.2017.1398836] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Oxaliplatin causes disabling acute and chronic peripheral neuropathy. We explored the preventive effects of calmangafodipir, mimicking the mitochondrial enzyme manganese superoxide dismutase, thereby protecting cells from oxidative stress, in a placebo-controlled, double-blinded randomised phase II study (ClinicalTrials.gov.NCT01619423) in patients with metastatic colorectal cancer (mCRC). PATIENT AND METHODS mCRC patients treated with modified FOLFOX-6 (folinic acid 200 mg/m2, 5-fluorouracil bolus 400 mg/m2, oxaliplatin 85 mg/m2 and 5-fluorouracil 2400 mg/m2 continuous infusion for 46 h) every fortnight for 8 cycles in first or second line were eligible. Calmangafodipir was given in a phase I dose-finding and in a phase II placebo-controlled study, as a 5-min infusion 10 min prior to oxaliplatin. Neurotoxicity was evaluated by the physician using the Oxaliplatin Sanofi Specific Scale and by the patient using the cold allodynia test and the Leonard scale. RESULTS Eleven patients were included in phase I without any detectable toxicity to calmangafodipir. In the phase II study, 173 patients were randomised to placebo (n = 60), calmangafodipir 2 µmol/kg (n = 57) and calmangafodipir 5 µmol/kg (n = 45, initially 10 µmol/kg, n = 11). Calmangafodipir-treated patients (all three doses pooled) had less physician graded neurotoxicity (odds ratio (90% confidence interval one-sided upper level) 0.62(1.15), p = .16), significantly less problems with cold allodynia (mean 1.6 versus 2.3, p < .05) and significantly fewer sensory symptoms in the Leonard scale (cycle 1-8 mean 1.9 versus 3.0, p < .05 and during follow-up after 3 and 6 months, mean 3.5 versus 7.3, p < .01). Response rate, progression-free and overall survival did not differ among groups. CONCLUSIONS Calmangafodipir at a dose of 5 µmol/kg appears to prevent the development of oxaliplatin-induced acute and delayed CIPN without apparent influence on tumour outcomes.
Collapse
Affiliation(s)
- Bengt Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nebojsa Manojlovic
- Clinic for Gastroenterology and Hepatology of Military Medical Academy of Serbia, Belgrade, Serbia
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Baadur Mosidze
- LTD High Technology Medical Center, University Clinic, Tbilisi, Georgia
| | | | - Mia Karlberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Devalingam Mahalingam
- Cancer Therapy and Research Center, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | | | | | | | | | | |
Collapse
|
18
|
Heide R, Bostock H, Ventzel L, Grafe P, Bergmans J, Fuglsang-Frederiksen A, Finnerup NB, Tankisi H. Axonal excitability changes and acute symptoms of oxaliplatin treatment: In vivo evidence for slowed sodium channel inactivation. Clin Neurophysiol 2017; 129:694-706. [PMID: 29233604 DOI: 10.1016/j.clinph.2017.11.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 10/16/2017] [Accepted: 11/05/2017] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Neurotoxicity is the most frequent dose-limiting side effect of the anti-cancer agent oxaliplatin, but the mechanisms are not well understood. This study used nerve excitability testing to investigate the pathophysiology of the acute neurotoxicity. METHODS Questionnaires, quantitative sensory tests, nerve conduction studies and nerve excitability testing were undertaken in 12 patients with high-risk colorectal cancer treated with adjuvant oxaliplatin and in 16 sex- and age-matched healthy controls. Examinations were performed twice for patients: once within 3 days after oxaliplatin treatment (post-infusion examination) and once shortly before the following treatment (recovery examination). RESULTS The most frequent post-infusion symptoms were tingling paresthesias and cold allodynia. The most prominent nerve excitability change was decreased superexcitability of motor axons which correlated with the average intensity of abnormal sensations (Spearman Rho = 0.80, p < .01). The motor nerve excitability changes were well modeled by a slowing of sodium channel inactivation, and were proportional to dose/m2 with a half-life of about 10d. CONCLUSIONS Oxaliplatin induces reversible slowing of sodium channel inactivation in motor axons, and these changes are closely related to the reversible cold allodynia. However, further studies are required due to small sample size in this study. SIGNIFICANCE Nerve excitability data provide an index of sodium channel dysfunction: an objective biomarker of acute oxaliplatin neurotoxicity.
Collapse
Affiliation(s)
- Rikke Heide
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark; Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hugh Bostock
- Institute of Neurology, Queen Square House, London, United Kingdom
| | - Lise Ventzel
- Department of Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Grafe
- Institute of Physiology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Joseph Bergmans
- Laboratory of Clinical Neurophysiology, Faculty of Medicine, University of Louvain, Brussels, Belgium
| | | | - Nanna B Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
19
|
Vale TA, Symmonds M, Polydefkis M, Byrnes K, Rice ASC, Themistocleous AC, Bennett DLH. Chronic non-freezing cold injury results in neuropathic pain due to a sensory neuropathy. Brain 2017; 140:2557-2569. [PMID: 28969380 PMCID: PMC5841153 DOI: 10.1093/brain/awx215] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 01/03/2023] Open
Abstract
Non-freezing cold injury develops after sustained exposure to cold temperatures, resulting in tissue cooling but not freezing. This can result in persistent sensory disturbance of the hands and feet including numbness, paraesthesia and chronic pain. Both vascular and neurological aetiologies of this pain have been suggested but remain unproven. We prospectively approached patients referred for clinical assessment of chronic pain following non-freezing cold injury between 12 February 2014 and 30 November 2016. Of 47 patients approached, 42 consented to undergo detailed neurological evaluations including: questionnaires to detail pain location and characteristics, structured neurological examination, quantitative sensory testing, nerve conduction studies and skin biopsy for intraepidermal nerve fibre assessment. Of the 42 study participants, all had experienced non-freezing cold injury while serving in the UK armed services and the majority were of African descent (76.2%) and male (95.2%). Many participants reported multiple exposures to cold. The median time between initial injury and referral was 3.72 years. Pain was principally localized to the hands and the feet, neuropathic in nature and in all study participants associated with cold hypersensitivity. Clinical examination and quantitative sensory testing were consistent with a sensory neuropathy. In all cases, large fibre nerve conduction studies were normal. The intraepidermal nerve fibre density was markedly reduced with 90.5% of participants having a count at or below the 0.05 centile of published normative controls. Using the Neuropathic Pain Special Interest Group of the International Association for the Study of Pain grading for neuropathic pain, 100% had probable and 95.2% definite neuropathic pain. Chronic non-freezing cold injury is a disabling neuropathic pain disorder due to a sensory neuropathy. Why some individuals develop an acute painful sensory neuropathy on sustained cold exposure is not yet known, but individuals of African descent appear vulnerable. Screening tools, such as the DN4 questionnaire, and treatment algorithms for neuropathic pain should now be used in the management of these patients.
Collapse
Affiliation(s)
- Tom A Vale
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Mkael Symmonds
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michael Polydefkis
- Cutaneous Nerve Laboratory, Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kelly Byrnes
- Cutaneous Nerve Laboratory, Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Andrew S C Rice
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital Campus, London, UK
- Pain Medicine, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | | | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
20
|
Inflammatory and neuropathic cold allodynia are selectively mediated by the neurotrophic factor receptor GFRα3. Proc Natl Acad Sci U S A 2016; 113:4506-11. [PMID: 27051069 DOI: 10.1073/pnas.1603294113] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tissue injury prompts the release of a number of proalgesic molecules that induce acute and chronic pain by sensitizing pain-sensing neurons (nociceptors) to heat and mechanical stimuli. In contrast, many proalgesics have no effect on cold sensitivity or can inhibit cold-sensitive neurons and diminish cooling-mediated pain relief (analgesia). Nonetheless, cold pain (allodynia) is prevalent in many inflammatory and neuropathic pain settings, with little known of the mechanisms promoting pain vs. those dampening analgesia. Here, we show that cold allodynia induced by inflammation, nerve injury, and chemotherapeutics is abolished in mice lacking the neurotrophic factor receptor glial cell line-derived neurotrophic factor family of receptors-α3 (GFRα3). Furthermore, established cold allodynia is blocked in animals treated with neutralizing antibodies against the GFRα3 ligand, artemin. In contrast, heat and mechanical pain are unchanged, and results show that, in striking contrast to the redundant mechanisms sensitizing other modalities after an insult, cold allodynia is mediated exclusively by a single molecular pathway, suggesting that artemin-GFRα3 signaling can be targeted to selectively treat cold pain.
Collapse
|