Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.913
Filter
1.
Physiol Behav ; 281: 114575, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38692384

ABSTRACT

Fibromyalgia (FM) is characterized by chronic widespread musculoskeletal pain accompanied by fatigue and muscle atrophy. Although its etiology is not known, studies have shown that FM patients exhibit altered function of the sympathetic nervous system (SNS), which regulates nociception and muscle plasticity. Nevertheless, the precise SNS-mediated mechanisms governing hyperalgesia and skeletal muscle atrophy in FM remain unclear. Thus, we employed two distinct FM-like pain models, involving intramuscular injections of acidic saline (pH 4.0) or carrageenan in prepubertal female rats, and evaluated the catecholamine content, adrenergic signaling and overall muscle proteolysis. Subsequently, we assessed the contribution of the SNS to the development of hyperalgesia and muscle atrophy in acidic saline-injected rats treated with clenbuterol (a selective ß2-adrenergic receptor agonist) and in animals maintained under baseline conditions and subjected to epinephrine depletion through adrenodemedullation (ADM). Seven days after inducing an FM-like model with acidic saline or carrageenan, we observed widespread mechanical hyperalgesia along with loss of strength and/or muscle mass. These changes were associated with reduced catecholamine content, suggesting a common underlying mechanism. Notably, treatment with a ß2-agonist alleviated hyperalgesia and prevented muscle atrophy in acidic saline-induced FM-like pain, while epinephrine depletion induced mechanical hyperalgesia and increased muscle proteolysis in animals under baseline conditions. Together, the results suggest that reduced sympathetic activity is involved in the development of pain and muscle atrophy in the murine model of FM analyzed.


Subject(s)
Clenbuterol , Disease Models, Animal , Fibromyalgia , Hyperalgesia , Muscular Atrophy , Sympathetic Nervous System , Animals , Female , Fibromyalgia/pathology , Fibromyalgia/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Hyperalgesia/physiopathology , Hyperalgesia/pathology , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/pathology , Clenbuterol/pharmacology , Rats , Carrageenan/toxicity , Rats, Sprague-Dawley , Pain/pathology , Pain/physiopathology , Epinephrine , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Catecholamines/metabolism , Adrenergic beta-Agonists/pharmacology
2.
Pain ; 165(6): 1336-1347, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38739766

ABSTRACT

ABSTRACT: Evidence from previous studies supports the concept that spinal cord injury (SCI)-induced neuropathic pain (NP) has its neural roots in the peripheral nervous system. There is uncertainty about how and to which degree mechanoreceptors contribute. Sensorimotor activation-based interventions (eg, treadmill training) have been shown to reduce NP after experimental SCI, suggesting transmission of pain-alleviating signals through mechanoreceptors. The aim of the present study was to understand the contribution of mechanoreceptors with respect to mechanical allodynia in a moderate mouse contusion SCI model. After genetic ablation of tropomyosin receptor kinase B expressing mechanoreceptors before SCI, mechanical allodynia was reduced. The identical genetic ablation after SCI did not yield any change in pain behavior. Peptidergic nociceptor sprouting into lamina III/IV below injury level as a consequence of SCI was not altered by either mechanoreceptor ablation. However, skin-nerve preparations of contusion SCI mice 7 days after injury yielded hyperexcitability in nociceptors, not in mechanoreceptors, which makes a substantial direct contribution of mechanoreceptors to NP maintenance unlikely. Complementing animal data, quantitative sensory testing in human SCI subjects indicated reduced mechanical pain thresholds, whereas the mechanical detection threshold was not altered. Taken together, early mechanoreceptor ablation modulates pain behavior, most likely through indirect mechanisms. Hyperexcitable nociceptors seem to be the main drivers of SCI-induced NP. Future studies need to focus on injury-derived factors triggering early-onset nociceptor hyperexcitability, which could serve as targets for more effective therapeutic interventions.


Subject(s)
Disease Models, Animal , Hyperalgesia , Mechanoreceptors , Mice, Inbred C57BL , Spinal Cord Injuries , Animals , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Mice , Hyperalgesia/physiopathology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Mechanoreceptors/metabolism , Mechanoreceptors/physiology , Male , Humans , Pain Threshold/physiology , Female , Pain Measurement , Mice, Transgenic , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/physiopathology
3.
Sheng Li Xue Bao ; 76(2): 301-308, 2024 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-38658378

ABSTRACT

Delayed-onset muscle soreness (DOMS) is a common phenomenon that occurs following a sudden increase in exercise intensity or unfamiliar exercise, significantly affecting athletic performance and efficacy in athletes and fitness individuals. DOMS is characterized by allodynia and hyperalgesia, and their mechanisms remain unclear. Recent studies have reported that neurotrophic factors, such as nerve growth factor (NGF) and glial cell derived neurotrophic factor (GDNF), are involved in the development and maintenance of DOMS. This article provides a review of the research progress on the signaling pathways related to the involvement of NGF and GDNF in DOMS, hoping to provide novel insights into the mechanisms underlying allodynia and hyperalgesia in DOMS, as well as potential targeted treatment.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor , Myalgia , Nerve Growth Factor , Humans , Myalgia/physiopathology , Nerve Growth Factor/metabolism , Nerve Growth Factor/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/physiology , Signal Transduction , Animals , Hyperalgesia/physiopathology , Muscle, Skeletal/physiopathology , Muscle, Skeletal/physiology , Exercise/physiology
4.
Clin Rheumatol ; 43(6): 2047-2059, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38668988

ABSTRACT

The objective of this study is to determine whether the change in pain intensity over time differs between somatosensory functioning evolution profiles in knee osteoarthritis (KOA) patients undergoing total knee arthroplasty (TKA). This longitudinal prospective cohort study, conducted between March 2018 and July 2023, included KOA patients undergoing TKA in four hospitals in Belgium and the Netherlands. The evolution of the Knee Injury and Osteoarthritis Outcome Score (KOOS) subscale pain over time (baseline, 3 months, and 1 year post-TKA scores) was the outcome variable. The evolution scores of quantitative sensory testing (QST) and Central Sensitization Inventory (CSI) over time (baseline and 1 year post-TKA scores) were used to make subgroups. Participants were divided into separate normal, recovered, and persistent disturbed somatosensory subgroups based on the CSI, local and widespread pressure pain threshold [PPT] and heat allodynia, temporal summation [TS], and conditioned pain modulation [CPM]. Linear mixed model analyses were performed. Two hundred twenty-three participants were included. The persistent disturbed somatosensory functioning group had less pronounced pain improvement (based on CSI and local heat allodynia) and worse pain scores 1 year post-TKA (based on CSI, local PPT and heat allodynia, and TS) compared to the normal somatosensory functioning group. This persistent group also had worse pain scores 1 year post-TKA compared to the recovered group (based on CSI). The study suggests the presence of a "centrally driven central sensitization" subgroup in KOA patients awaiting TKA in four of seven grouping variables, comprising their less pain improvement or worse pain score after TKA. Future research should validate these findings further. The protocol is registered at clinicaltrials.gov (NCT05380648).


Subject(s)
Arthroplasty, Replacement, Knee , Osteoarthritis, Knee , Pain Measurement , Humans , Osteoarthritis, Knee/surgery , Osteoarthritis, Knee/physiopathology , Female , Male , Prospective Studies , Aged , Middle Aged , Longitudinal Studies , Pain Threshold , Pain, Postoperative/etiology , Belgium , Netherlands , Hyperalgesia/physiopathology
5.
J Neurotrauma ; 41(9-10): 1077-1088, 2024 May.
Article in English | MEDLINE | ID: mdl-38185845

ABSTRACT

Abstract With the recovery of motor function, some spinal cord injury (SCI) patients still suffer from severe pain-like behaviors symptoms. Whether motor function correlates with neuropathic pain-like behaviors remain unclear. In this study, a longitudinal cohort study of mice with moderate thoracic 10 contusion was performed to explore the characteristics of neuropathic pain-like behaviors and its correlation with motor function in different sexes. Pain-like behaviors data up to 42 days post-injury (dpi) were collected and compared. Mice of both sexes were divided into three groups based on their Basso Mouse Scale at 42 dpi. There was no significant difference in motor function recovery between the sexes. Female mice showed more significant mechanical allodynia than males at 14 dpi, which was sustained until 42 dpi without significant dynamic changes. However, males showed a gradually worsening state and more severe mechanical allodynia than females at 28 dpi, and then the differences disappeared. Interestingly, male mice obtained more severe cold hyperalgesia symptoms than females. Additionally, we found that there was a correlation between the occurrence of mechanical allodynia and cold and thermal hyperalgesia. Importantly, motor function recovery was positively associated with the outcomes of neuropathic pain-like behaviors after SCI, which was more obvious in female mice. Our data not only revealed the characteristics of neuropathic pain-like behaviors but also clarified the correlations between motor function recovery and neuropathic pain-like behaviors after SCI. These findings may provide new opinions and suggestions for promoting the clinical diagnosis and treatment of neuropathic pain-like behaviors after SCI.


Subject(s)
Hyperalgesia , Neuralgia , Spinal Cord Injuries , Animals , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/complications , Neuralgia/physiopathology , Neuralgia/etiology , Mice , Female , Male , Longitudinal Studies , Hyperalgesia/physiopathology , Hyperalgesia/etiology , Motor Activity/physiology , Recovery of Function/physiology , Behavior, Animal/physiology , Mice, Inbred C57BL
6.
Pain ; 165(6): 1391-1403, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38227559

ABSTRACT

ABSTRACT: Neuropathic corneal pain (NCP) is a new and ill-defined disease characterized by pain, discomfort, aching, burning sensation, irritation, dryness, and grittiness. However, the mechanism underlying NCP remain unclear. Here, we reported a novel rat model of primary NCP induced by long ciliary nerve (LCN) ligation. After sustained LCN ligation, the rats developed increased corneal mechanical and chemical sensitivity, spontaneous blinking, and photophobia, which were ameliorated by intraperitoneal injection of morphine or gabapentin. However, neither tear reduction nor corneal injury was observed in LCN-ligated rats. Furthermore, after LCN ligation, the rats displayed a significant reduction in corneal nerve density, as well as increased tortuosity and beading nerve ending. Long ciliary nerve ligation also notably elevated corneal responsiveness under resting or menthol-stimulated conditions. At a cellular level, we observed that LCN ligation increased calcitonin gene-related peptide (neuropeptide)-positive cells in the trigeminal ganglion (TG). At a molecular level, upregulated mRNA levels of ion channels Piezo2, TRPM8, and TRPV1, as well as inflammatory factors TNF-α, IL-1ß, and IL-6, were also detected in the TG after LCN ligation. Meanwhile, consecutive oral gabapentin attenuated LCN ligation-induced corneal hyperalgesia and increased levels of ion channels and inflammation factors in TG. This study provides a reliable primary NCP model induced by LCN ligation in rats using a simple, minimally invasive surgery technique, which may help shed light on the underlying cellular and molecular bases of NCP and aid in developing a new treatment for the disease.


Subject(s)
Cornea , Disease Models, Animal , Gabapentin , Neuralgia , Rats, Sprague-Dawley , Animals , Neuralgia/etiology , Male , Rats , Gabapentin/pharmacology , Gabapentin/therapeutic use , Ligation , Cornea/innervation , Trigeminal Ganglion/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , gamma-Aminobutyric Acid/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , Calcitonin Gene-Related Peptide/metabolism , Amines/pharmacology , Amines/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Eye Pain/etiology , Hyperalgesia/etiology , Hyperalgesia/physiopathology
7.
Pain ; 165(6): 1413-1424, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38231588

ABSTRACT

ABSTRACT: Women more often experience chronic pain conditions than men. Central sensitization (CS) is one key mechanism in chronic pain that can differ between the sexes. It is unknown whether CS processes are already more pronounced in healthy women than in men. In 66 subjects (33 women), a thermal CS induction protocol was applied to the dorsum of one foot and a sham protocol to the other. Spatial extent [cm 2 ] of secondary mechanical hyperalgesia (SMH) and dynamic mechanical allodynia were assessed as subjective CS proxy measures, relying on verbal feedback. Changes in nociceptive withdrawal reflex magnitude (NWR-M) and response rate (NWR-RR) recorded through surface electromyography at the biceps and rectus femoris muscles were used as objective CS proxies. The effect of the CS induction protocol on SMH was higher in women than in men (effect size 2.11 vs 1.68). Nociceptive withdrawal reflex magnitude results were statistically meaningful for women (effect size 0.31-0.36) but not for men (effect size 0.12-0.29). Differences between men and women were not meaningful. Nociceptive withdrawal reflex response rate at the rectus femoris increased in women after CS induction and was statistically different from NWR-RR in men (median differences of 13.7 and 8.4% for 120 and 140% reflex threshold current). The objective CS proxy differences indicate that dorsal horn CS processes are more pronounced in healthy women. The even larger sex differences in subjective CS proxies potentially reflect greater supraspinal influence in women. This study shows that sex differences are present in experimentally induced CS in healthy subjects, which might contribute to women's vulnerability for chronic pain.


Subject(s)
Central Nervous System Sensitization , Electromyography , Hyperalgesia , Sex Characteristics , Humans , Female , Male , Central Nervous System Sensitization/physiology , Adult , Hyperalgesia/physiopathology , Young Adult , Pain Threshold/physiology , Reflex/physiology , Pain Measurement/methods , Middle Aged
8.
Pain ; 165(6): 1304-1316, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38277178

ABSTRACT

ABSTRACT: Abnormal encoding of somatosensory modalities (ie, mechanical, cold, and heat) are a critical part of pathological pain states. Detailed phenotyping of patients' responses to these modalities have raised hopes that analgesic treatments could one day be tailored to a patient's phenotype. Such precise treatment would require a profound understanding of the underlying mechanisms of specific pain phenotypes at molecular, cellular, and circuitry levels. Although preclinical pain models have helped in that regard, the lack of a unified assay quantifying detailed mechanical, cold, and heat pain responses on the same scale precludes comparing how analgesic compounds act on different sensory phenotypes. The conflict avoidance assay is promising in that regard, but testing conditions require validation for its use with multiple modalities. In this study, we improve upon the conflict avoidance assay to provide a validated and detailed assessment of all 3 modalities within the same animal, in mice. We first optimized testing conditions to minimize the necessary amount of training and to reduce sex differences in performances. We then tested what range of stimuli produce dynamic stimulus-response relationships for different outcome measures in naive mice. We finally used this assay to show that nerve injury produces modality-specific sex differences in pain behavior. Our improved assay opens new avenues to study the basis of modality-specific abnormalities in pain behavior.


Subject(s)
Avoidance Learning , Hyperalgesia , Pain Measurement , Sex Characteristics , Animals , Female , Male , Mice , Avoidance Learning/physiology , Pain Measurement/methods , Hyperalgesia/physiopathology , Mice, Inbred C57BL , Disease Models, Animal , Pain Threshold/physiology , Physical Stimulation , Conflict, Psychological
9.
Pain ; 165(5): 1154-1168, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38147415

ABSTRACT

ABSTRACT: Painful diabetic neuropathy (PDN) is one of the most common and intractable complications of diabetes. Painful diabetic neuropathy is characterized by neuropathic pain accompanied by dorsal root ganglion (DRG) nociceptor hyperexcitability, axonal degeneration, and changes in cutaneous innervation. However, the complete molecular profile underlying the hyperexcitable cellular phenotype of DRG nociceptors in PDN has not been elucidated. This gap in our knowledge is a critical barrier to developing effective, mechanism-based, and disease-modifying therapeutic approaches that are urgently needed to relieve the symptoms of PDN. Using single-cell RNA sequencing of DRGs, we demonstrated an increased expression of the Mas-related G protein-coupled receptor d (Mrgprd) in a subpopulation of DRG neurons in the well-established high-fat diet (HFD) mouse model of PDN. Importantly, limiting Mrgprd signaling reversed mechanical allodynia in the HFD mouse model of PDN. Furthermore, in vivo calcium imaging allowed us to demonstrate that activation of Mrgprd-positive cutaneous afferents that persist in diabetic mice skin resulted in an increased intracellular calcium influx into DRG nociceptors that we assess in vivo as a readout of nociceptors hyperexcitability. Taken together, our data highlight a key role of Mrgprd-mediated DRG neuron excitability in the generation and maintenance of neuropathic pain in a mouse model of PDN. Hence, we propose Mrgprd as a promising and accessible target for developing effective therapeutics currently unavailable for treating neuropathic pain in PDN.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Hyperalgesia , Neuralgia , Receptors, G-Protein-Coupled , Animals , Mice , Calcium/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/complications , Diabetic Neuropathies/metabolism , Disease Models, Animal , Ganglia, Spinal/metabolism , Hypersensitivity/genetics , Neuralgia/metabolism , Receptors, G-Protein-Coupled/metabolism , Hyperalgesia/genetics , Hyperalgesia/physiopathology
10.
Rev. Fac. Odontol. (B.Aires) ; 38(90): 29-37, 2023. ilus, tab
Article in Spanish | LILACS | ID: biblio-1553776

ABSTRACT

Esta revisión busca proporcionar a los profesionales de la salud una mayor comprensión del dolor para su actividad clínica-asistencial. Basados en la hipóte-sis de neuroplasticidad presentada inicialmente por Ramón y Cajal y la teoría de la compuerta en la vía dolorosa presentada por Melzack y Wall, se ha ela-borado una revisión bibliográfica con el objetivo de abordar la modulación de la vía nociceptiva desde un punto de vista fisiopatológico. Asimismo, se presen-tan los principales resultados obtenidos durante los últimos años en nuestro laboratorio usando ratas Wistar hembras como modelo de dolor experimental. Finalmente, se describe un circuito original de modu-lación central a nivel del subnúcleo caudal del trigé-mino con una visión integral de los componentes del sistema nociceptivo orofacial, para ayudar al clínico a comprender situaciones de sensibilización central con perpetuación del dolor y cómo paulatinamente el sistema nervioso central pone en marcha un sistema de modulación para adaptarse y alcanzar un estado similar al basal (AU)


This review aims to provide health professionals with a better understanding of pain for their clinical-care activity. Based on the neuroplasticity hypothesis initially presented by Ramón and Cajal, and the gate theory in the pain pathway presented by Melzack and Wall, a literature review has been carried out with the aim of addressing the modulation of the nociceptive pathway from a pathophysiological point of view. The main results obtained in recent years in our laboratory using female Wistar rats as an experimental pain model are also presented. Finally, an original central modulation circuit at the level of the caudal trigeminal subnucleus is described with a comprehensive view of the components of the orofacial nociceptive system, to help the clinician to understand situations of central sensitization with perpetuation of pain and how the central nervous system gradually sets in motion a modulation system to adapt and reach a state similar to the basal one (AU)


Subject(s)
Humans , Animals , Rats , Pain/physiopathology , Central Nervous System/physiology , Nociception/physiology , Neuronal Plasticity/physiology , Astrocytes , Rats, Wistar , Hyperalgesia/physiopathology , Interneurons
11.
Phys Ther ; 102(12)2022 12 06.
Article in English | MEDLINE | ID: mdl-36124704

ABSTRACT

OBJECTIVE: The nociceptive pain processing of soft-tissue overuse conditions is under debate because no consensus currently exists. The purpose of this meta-analysis was to compare pressure pain thresholds (PPTs) in symptomatic and distant pain-free areas in 2 groups: participants with symptomatic lower extremity overuse soft-tissue conditions and controls who were pain free. METHODS: Five databases were searched from inception to December 1, 2021, for case-control studies comparing PPTs between individuals presenting with symptomatic lower extremity tendinopathy/overuse injury and controls who were pain free. Data extraction included population, diagnosis, sample size, outcome, type of algometer, and results. The methodological quality (Newcastle-Ottawa Quality Assessment Scale) and evidence level (Grading of Recommendations Assessment, Development, and Evaluation) were assessed. Meta-analyses of symptomatic, segmental related, and distant pain-free areas were compared. RESULTS: After screening 730 titles and abstracts, a total of 19 studies evaluating lower extremity overuse conditions (Achilles or patellar tendinopathy, greater trochanteric pain syndrome, plantar fasciitis, and iliotibial band syndrome) were included. The methodological quality ranged from fair (32%) to good (68%). Participants with lower extremity overuse injury had lower PPTs in both the painful and nonpainful areas, mirrored test-site, compared with controls (affected side: mean difference [MD] = -262.92 kPa, 95% CI = 323.78 to -202.05 kPa; nonaffected side: MD = -216.47 kPa, 95% CI = -304.99 to -127.95 kPa). Furthermore, people with plantar fasciitis showed reduced PPTs in the affected and nonaffected sides at segmental-related (MD = -176.39 kPa, 95% CI = -306.11 to -46.68 kPa) and distant pain-free (MD = -97.27 kPa, 95% CI = 133.21 to -61.33 kPa) areas compared with controls. CONCLUSION: Low- to moderate-quality evidence suggests a reduction of PPTs at the symptomatic area and a contralateral/mirror side in lower extremity tendinopathies and overuse conditions compared with pain-free controls, particularly in plantar fasciitis and greater trochanteric pain syndrome. Participants with plantar fasciitis showed a reduction of PPTs on the affected and non-affected sides at a segmental-related area (very low-quality evidence) and at a remote asymptomatic area (moderate-quality evidence). IMPACT: Some overuse peripheral pain conditions may be more associated with pressure pain sensitivity than others. Accordingly, examination and identification of conditions more peripherally, centrally, or mixed mediated could potentially lead to more specific and different treatment strategies.


Subject(s)
Cumulative Trauma Disorders , Hyperalgesia , Pain Threshold , Tendinopathy , Hyperalgesia/diagnosis , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Humans , Tendinopathy/complications , Cumulative Trauma Disorders/complications , Pain Measurement , Pressure , Lower Extremity
12.
Proc Natl Acad Sci U S A ; 119(30): e2114094119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858441

ABSTRACT

Clinical evidence suggests that pain hypersensitivity develops in patients with attention-deficit/hyperactivity disorder (ADHD). However, the mechanisms and neural circuits involved in these interactions remain unknown because of the paucity of studies in animal models. We previously validated a mouse model of ADHD obtained by neonatal 6-hydroxydopamine (6-OHDA) injection. Here, we have demonstrated that 6-OHDA mice exhibit a marked sensitization to thermal and mechanical stimuli, suggesting that phenotypes associated with ADHD include increased nociception. Moreover, sensitization to pathological inflammatory stimulus is amplified in 6-OHDA mice as compared to shams. In this ADHD model, spinal dorsal horn neuron hyperexcitability was observed. Furthermore, ADHD-related hyperactivity and anxiety, but not inattention and impulsivity, are worsened in persistent inflammatory conditions. By combining in vivo electrophysiology, optogenetics, and behavioral analyses, we demonstrated that anterior cingulate cortex (ACC) hyperactivity alters the ACC-posterior insula circuit and triggers changes in spinal networks that underlie nociceptive sensitization. Altogether, our results point to shared mechanisms underlying the comorbidity between ADHD and nociceptive sensitization. This interaction reinforces nociceptive sensitization and hyperactivity, suggesting that overlapping ACC circuits may be targeted to develop better treatments.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Hyperalgesia , Pain , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Disease Models, Animal , Gyrus Cinguli/physiopathology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Impulsive Behavior , Mice , Optogenetics , Oxidopamine/pharmacology , Pain/chemically induced , Pain/physiopathology , Sympatholytics/pharmacology
13.
Nature ; 606(7912): 137-145, 2022 06.
Article in English | MEDLINE | ID: mdl-35614217

ABSTRACT

Nerve injury leads to chronic pain and exaggerated sensitivity to gentle touch (allodynia) as well as a loss of sensation in the areas in which injured and non-injured nerves come together1-3. The mechanisms that disambiguate these mixed and paradoxical symptoms are unknown. Here we longitudinally and non-invasively imaged genetically labelled populations of fibres that sense noxious stimuli (nociceptors) and gentle touch (low-threshold afferents) peripherally in the skin for longer than 10 months after nerve injury, while simultaneously tracking pain-related behaviour in the same mice. Fully denervated areas of skin initially lost sensation, gradually recovered normal sensitivity and developed marked allodynia and aversion to gentle touch several months after injury. This reinnervation-induced neuropathic pain involved nociceptors that sprouted into denervated territories precisely reproducing the initial pattern of innervation, were guided by blood vessels and showed irregular terminal connectivity in the skin and lowered activation thresholds mimicking low-threshold afferents. By contrast, low-threshold afferents-which normally mediate touch sensation as well as allodynia in intact nerve territories after injury4-7-did not reinnervate, leading to an aberrant innervation of tactile end organs such as Meissner corpuscles with nociceptors alone. Genetic ablation of nociceptors fully abrogated reinnervation allodynia. Our results thus reveal the emergence of a form of chronic neuropathic pain that is driven by structural plasticity, abnormal terminal connectivity and malfunction of nociceptors during reinnervation, and provide a mechanistic framework for the paradoxical sensory manifestations that are observed clinically and can impose a heavy burden on patients.


Subject(s)
Hyperalgesia , Neuralgia , Nociceptors , Skin , Animals , Chronic Pain/physiopathology , Hyperalgesia/physiopathology , Mechanoreceptors/pathology , Mice , Neuralgia/physiopathology , Nociceptors/pathology , Skin/innervation , Skin/physiopathology
14.
Science ; 377(6601): 80-86, 2022 07.
Article in English | MEDLINE | ID: mdl-35617374

ABSTRACT

Activation of microglia in the spinal cord dorsal horn after peripheral nerve injury contributes to the development of pain hypersensitivity. How activated microglia selectively enhance the activity of spinal nociceptive circuits is not well understood. We discovered that after peripheral nerve injury, microglia degrade extracellular matrix structures, perineuronal nets (PNNs), in lamina I of the spinal cord dorsal horn. Lamina I PNNs selectively enwrap spinoparabrachial projection neurons, which integrate nociceptive information in the spinal cord and convey it to supraspinal brain regions to induce pain sensation. Degradation of PNNs by microglia enhances the activity of projection neurons and induces pain-related behaviors. Thus, nerve injury-induced degradation of PNNs is a mechanism by which microglia selectively augment the output of spinal nociceptive circuits and cause pain hypersensitivity.


Subject(s)
Hyperalgesia , Microglia , Pain , Peripheral Nerve Injuries , Spinal Cord Dorsal Horn , Animals , Extracellular Matrix/pathology , Hyperalgesia/etiology , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Microglia/pathology , Pain/pathology , Pain/physiopathology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/pathology , Spinal Cord Dorsal Horn/physiopathology
15.
Science ; 376(6588): 86-90, 2022 04.
Article in English | MEDLINE | ID: mdl-35357926

ABSTRACT

Neuropathic pain is often caused by injury and diseases that affect the somatosensory system. Although pain development has been well studied, pain recovery mechanisms remain largely unknown. Here, we found that CD11c-expressing spinal microglia appear after the development of behavioral pain hypersensitivity following nerve injury. Nerve-injured mice with spinal CD11c+ microglial depletion failed to recover spontaneously from this hypersensitivity. CD11c+ microglia expressed insulin-like growth factor-1 (IGF1), and interference with IGF1 signaling recapitulated the impairment in pain recovery. In pain-recovered mice, the depletion of CD11c+ microglia or the interruption of IGF1 signaling resulted in a relapse in pain hypersensitivity. Our findings reveal a mechanism for the remission and recurrence of neuropathic pain, providing potential targets for therapeutic strategies.


Subject(s)
Chronic Pain/physiopathology , Hyperalgesia/physiopathology , Microglia/physiology , Neuralgia/physiopathology , Peripheral Nerve Injuries/physiopathology , Spinal Cord/physiopathology , Animals , Bacterial Proteins/genetics , CD11 Antigens/genetics , CD11 Antigens/metabolism , Female , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recurrence
16.
Front Immunol ; 13: 811402, 2022.
Article in English | MEDLINE | ID: mdl-35185905

ABSTRACT

Joint pain is a complex phenomenon that involves multiple endogenous mediators and pathophysiological events. In addition to nociceptive and inflammatory pain, some patients report neuropathic-like pain symptoms. Examination of arthritic joints from humans and preclinical animal models have revealed axonal damage which is likely the source of the neuropathic pain. The mediators responsible for joint peripheral neuropathy are obscure, but lysophosphatidic acid (LPA) has emerged as a leading candidate target. In the present study, male and female Wistar rats received an intra-articular injection of LPA into the right knee and allowed to recover for 28 days. Joint pain was measured by von Frey hair algesiometry, while joint pathology was determined by scoring of histological sections. Both male and female rats showed comparable degenerative changes to the LPA-treated knee including chondrocyte death, focal bone erosion, and synovitis. Mechanical withdrawal thresholds decreased by 20-30% indicative of secondary allodynia in the affected limb; however, there was no significant difference in pain sensitivity between the sexes. Treatment of LPA animals with the neuropathic pain drug amitriptyline reduced joint pain for over 2 hours with no sex differences being observed. In summary, intra-articular injection of LPA causes joint degeneration and neuropathic pain thereby mimicking some of the characteristics of neuropathic osteoarthritis.


Subject(s)
Arthralgia/physiopathology , Arthritis, Experimental/physiopathology , Knee Joint/pathology , Lysophospholipids/administration & dosage , Neuralgia/physiopathology , Animals , Arthralgia/chemically induced , Arthralgia/pathology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Disease Models, Animal , Female , Hyperalgesia/physiopathology , Injections, Intra-Articular , Male , Neuralgia/chemically induced , Neuralgia/pathology , Pain Threshold , Rats , Rats, Wistar
17.
Nat Commun ; 13(1): 646, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115501

ABSTRACT

Efficacy of monoclonal antibodies against calcitonin gene-related peptide (CGRP) or its receptor (calcitonin receptor-like receptor/receptor activity modifying protein-1, CLR/RAMP1) implicates peripherally-released CGRP in migraine pain. However, the site and mechanism of CGRP-evoked peripheral pain remain unclear. By cell-selective RAMP1 gene deletion, we reveal that CGRP released from mouse cutaneous trigeminal fibers targets CLR/RAMP1 on surrounding Schwann cells to evoke periorbital mechanical allodynia. CLR/RAMP1 activation in human and mouse Schwann cells generates long-lasting signals from endosomes that evoke cAMP-dependent formation of NO. NO, by gating Schwann cell transient receptor potential ankyrin 1 (TRPA1), releases ROS, which in a feed-forward manner sustain allodynia via nociceptor TRPA1. When encapsulated into nanoparticles that release cargo in acidified endosomes, a CLR/RAMP1 antagonist provides superior inhibition of CGRP signaling and allodynia in mice. Our data suggest that the CGRP-mediated neuronal/Schwann cell pathway mediates allodynia associated with neurogenic inflammation, contributing to the algesic action of CGRP in mice.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Endosomes/metabolism , Hyperalgesia/physiopathology , Schwann Cells/metabolism , Signal Transduction/physiology , Animals , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Cells, Cultured , Female , HEK293 Cells , Humans , Hyperalgesia/diagnosis , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Neurons/metabolism , Nitric Oxide/metabolism , Receptor Activity-Modifying Protein 1/genetics , Receptor Activity-Modifying Protein 1/metabolism , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism
18.
Invest Ophthalmol Vis Sci ; 63(1): 7, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34989761

ABSTRACT

Purpose: Dry eye-induced chronic ocular pain is also called ocular neuropathic pain. However, details of the pathogenic mechanism remain unknown. The purpose of this study was to elucidate the pathogenic mechanism of dry eye-induced chronic pain in the anterior eye area and develop a pathophysiology-based therapeutic strategy. Methods: We used a rat dry eye model with lacrimal gland excision (LGE) to elucidate the pathogenic mechanism of ocular neuropathic pain. Corneal epithelial damage, hypersensitivity, and hyperalgesia were evaluated on the LGE side and compared with the sham surgery side. We analyzed neuronal activity, microglial and astrocytic activity, α2δ-1 subunit expression, and inhibitory interneurons in the trigeminal nucleus. We also evaluated the therapeutic effects of ophthalmic treatment and chronic pregabalin administration on dry eye-induced ocular neuropathic pain. Results: Dry eye caused hypersensitivity and hyperalgesia on the LGE side. In the trigeminal nucleus of the LGE side, neuronal hyperactivation, transient activation of microglia, persistent activation of astrocytes, α2δ-1 subunit upregulation, and reduced numbers of inhibitory interneurons were observed. Ophthalmic treatment alone did not improve hyperalgesia. In contrast, continuous treatment with pregabalin effectively ameliorated hypersensitivity and hyperalgesia and normalized neural activity, α2δ-1 subunit upregulation, and astrocyte activation. Conclusions: These results suggest that dry eye-induced hypersensitivity and hyperalgesia are caused by central sensitization in the trigeminal nucleus with upregulation of the α2δ-1 subunit. Here, we showed that pregabalin is effective for treating dry eye-induced ocular neuropathic pain even after chronic pain has been established.


Subject(s)
Analgesics/administration & dosage , Disease Models, Animal , Dry Eye Syndromes/physiopathology , Eye Pain/physiopathology , Pregabalin/administration & dosage , Administration, Ophthalmic , Animals , Astrocytes/pathology , Calcium Channels, L-Type/metabolism , Chronic Disease , Cornea/innervation , Dry Eye Syndromes/drug therapy , Eye Pain/drug therapy , Hyaluronic Acid/administration & dosage , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Microglia/pathology , Neuralgia/drug therapy , Neuralgia/physiopathology , Neurons/metabolism , Neurons/pathology , Ophthalmic Solutions , Rats , Rats, Sprague-Dawley , Trigeminal Nerve/metabolism , Trigeminal Nerve/pathology
19.
Naunyn Schmiedebergs Arch Pharmacol ; 395(3): 325-335, 2022 03.
Article in English | MEDLINE | ID: mdl-34985531

ABSTRACT

Recent studies indicate presence of a strong link between adipokines and neuropathic pain. However, the effects of asprosin, a novel adipokine, on neuropathic pain have not been studied in animal models.Mouse models were employed to investigate the antinociceptive effectiveness of asprosin in the treatment of three types of neuropathic pain, with metabolic (streptozocin/STZ), toxic (oxaliplatin/OXA), and traumatic (sciatic nerve ligation/CCI [chronic constriction nerve injury]) etiologies, respectively. Changes in nociceptive behaviors were assessed relative to controls using thermal (the hot plate and cold plate tests, at 50 °C and 4 °C respectively) and mechanical pain (von Frey test) tests after intraperitoneal (i.p.) administration of asprosin (10 µg/kg) and gabapentin (50 mg/kg) in several times intervals. Besides, possible effect of asprosin on the motor coordination of mice was assessed with a rotarod test. Serum level of asprosin was quantified by ELISA.In neuropathic pain models (STZ, OXA, and CCI), asprosin administration significantly reduced both mechanical and thermal hypersensitivity, indicating that it exhibits a clear-cut antihypersensitivity effect in the analyzed neuropathic pain models. The most effective time of asprosin on pain threshold was observed 60 min after its injection. Also, asprosin displayed no notable effect on the motor activity. Asprosin levels were significantly lower in neuropathic pain compared to healthy group (p < 0.05).The results yielded by the present study suggest that asprosin exhibits an analgesic effect in the neuropathic pain models and may have clinical utility in alleviating chronic pain associated with disease and injury originating from peripheral structures.


Subject(s)
Analgesics/pharmacology , Fibrillin-1/pharmacology , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Peptide Fragments/pharmacology , Peptide Hormones/pharmacology , Analgesics/administration & dosage , Animals , Disease Models, Animal , Fibrillin-1/administration & dosage , Gabapentin/pharmacology , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred BALB C , Neuralgia/physiopathology , Pain Threshold , Peptide Fragments/administration & dosage , Peptide Hormones/administration & dosage , Rotarod Performance Test
20.
Behav Pharmacol ; 33(1): 23-31, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35007233

ABSTRACT

The monoiodoacetate-induced rat model of osteoarthritis knee pain is widely used. However, there are between-study differences in the pain behavioural endpoints assessed and in the dose of intraarticular monoiodoacetate administered. This study evaluated the robustness of gait analysis as a pain behavioural endpoint in the chronic phase of this model, in comparison with mechanical hyperalgesia in the injected (ipsilateral) joint and development of mechanical allodynia in the ipsilateral hind paws. Groups of Sprague-Dawley rats received a single intraarticular injection of monoiodoacetate at 0.5, 1, 2 or 3 mg or vehicle (saline) into the left (ipsilateral) knee joint. An additional group of rats were not injected (naïve group). The pain behavioural methods used were gait analysis, measurement of pressure algometry thresholds in the ipsilateral knee joints, and assessment of mechanical allodynia in the ipsilateral hind paws using von Frey filaments. These pain behavioural endpoints were assessed premonoiodoacetate injection and for up to 42-days postmonoiodoacetate injection in a blinded manner. Body weights were also assessed as a measure of general health. Good general health was maintained as all rats gained weight at a similar rate for the 42-day study period. In the chronic phase of the model (days 9-42), intraarticular monoiodoacetate at 3 mg evoked robust alterations in multiple gait parameters as well as persistent mechanical allodynia in the ipsilateral hind paws. For the chronic phase of the monoiodoacetate-induced rat model of osteoarthritis knee pain, gait analysis, such as mechanical allodynia in the ipsilateral hind paws, is a robust pain behavioural measure.


Subject(s)
Arthralgia , Behavioral Symptoms , Gait Analysis/methods , Hyperalgesia , Osteoarthritis , Pain , Animals , Arthralgia/chemically induced , Arthralgia/psychology , Behavior Observation Techniques/methods , Behavior, Animal , Behavioral Symptoms/diagnosis , Behavioral Symptoms/physiopathology , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Hyperalgesia/diagnosis , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Iodoacetic Acid/administration & dosage , Osteoarthritis/physiopathology , Osteoarthritis/psychology , Pain/physiopathology , Pain/psychology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...