Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 791
Filter
1.
J Neurosci ; 39(30): 6012-6030, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31138657

ABSTRACT

Mechanical sensitization is one of the most difficult clinical pain problems to treat. However, the molecular and genetic bases of mechanical nociception are unclear. Here we develop a Drosophila model of mechanical nociception to investigate the ion channels and signaling pathways that regulate mechanical nociception. We fabricated von Frey filaments that span the subthreshold to high noxious range for Drosophila larvae. Using these, we discovered that pressure (force/area), rather than force per se, is the main determinant of aversive rolling responses to noxious mechanical stimuli. We demonstrated that the RTK PDGF/VEGF receptor (Pvr) and its ligands (Pvfs 2 and 3) are required for mechanical nociception and normal dendritic branching. Pvr is expressed and functions in class IV sensory neurons, whereas Pvf2 and Pvf3 are produced by multiple tissues. Constitutive overexpression of Pvr and its ligands or inducible overexpression of Pvr led to mechanical hypersensitivity that could be partially separated from morphological effects. Genetic analyses revealed that the Piezo and Pain ion channels are required for mechanical hypersensitivity observed upon ectopic activation of Pvr signaling. PDGF, but not VEGF, peptides caused mechanical hypersensitivity in rats. Pharmacological inhibition of VEGF receptor Type 2 (VEGFR-2) signaling attenuated mechanical nociception in rats, suggesting a conserved role for PDGF and VEGFR-2 signaling in regulating mechanical nociception. VEGFR-2 inhibition also attenuated morphine analgesic tolerance in rats. Our results reveal that a conserved RTK signaling pathway regulates baseline mechanical nociception in flies and rats.SIGNIFICANCE STATEMENT Hypersensitivity to touch is poorly understood and extremely difficult to treat. Using a refined Drosophila model of mechanical nociception, we discovered a conserved VEGF-related receptor tyrosine kinase signaling pathway that regulates mechanical nociception in flies. Importantly, pharmacological inhibition of VEGF receptor Type 2 signaling in rats causes analgesia and blocks opioid tolerance. We have thus established a robust, genetically tractable system for the rapid identification and functional analysis of conserved genes underlying mechanical pain sensitivity.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Nociception/physiology , Sensory Receptor Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Animals, Genetically Modified , Drosophila melanogaster , Intercellular Signaling Peptides and Proteins/genetics , Larva , Male , Nociception/drug effects , Physical Stimulation/adverse effects , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Species Specificity , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vertebrates
2.
Anesthesiology ; 132(4): 867-880, 2020 04.
Article in English | MEDLINE | ID: mdl-32011337

ABSTRACT

BACKGROUND: Recent emerging evidence suggests that extra-adrenal synthesis of aldosterone occurs (e.g., within the failing heart and in certain brain areas). In this study, the authors investigated evidence for a local endogenous aldosterone production through its key processing enzyme aldosterone synthase within peripheral nociceptive neurons. METHODS: In male Wistar rats (n = 5 to 8 per group) with Freund's complete adjuvant hind paw inflammation, the authors examined aldosterone, aldosterone synthase, and mineralocorticoid receptor expression in peripheral sensory neurons using quantitative reverse transcriptase-polymerase chain reaction, Western blot, immunohistochemistry, and immunoprecipitation. Moreover, the authors explored the nociceptive behavioral changes after selective mineralocorticoid receptor antagonist, canrenoate-K, or specific aldosterone synthase inhibitor application. RESULTS: In rats with Freund's complete adjuvant-induced hind paw inflammation subcutaneous and intrathecal application of mineralocorticoid receptor antagonist, canrenoate-K, rapidly and dose-dependently attenuated nociceptive behavior (94 and 48% reduction in mean paw pressure thresholds, respectively), suggesting a tonic activation of neuronal mineralocorticoid receptors by an endogenous ligand. Indeed, aldosterone immunoreactivity was abundant in peptidergic nociceptive neurons of dorsal root ganglia and colocalized predominantly with its processing enzyme aldosterone synthase and mineralocorticoid receptors. Moreover, aldosterone and its synthesizing enzyme were significantly upregulated in peripheral sensory neurons under inflammatory conditions. The membrane mineralocorticoid receptor consistently coimmunoprecipitated with endogenous aldosterone, confirming a functional link between mineralocorticoid receptors and its endogenous ligand. Importantly, inhibition of endogenous aldosterone production in peripheral sensory neurons by a specific aldosterone synthase inhibitor attenuated nociceptive behavior after hind paw inflammation (a 32% reduction in paw pressure thresholds; inflammation, 47 ± 2 [mean ± SD] vs. inflammation + aldosterone synthase inhibitor, 62 ± 2). CONCLUSIONS: Local production of aldosterone by its processing enzyme aldosterone synthase within peripheral sensory neurons contributes to ongoing mechanical hypersensitivity during local inflammation via intrinsic activation of neuronal mineralocorticoid receptors.


Subject(s)
Cytochrome P-450 CYP11B2/biosynthesis , Hyperalgesia/metabolism , Pain Measurement/methods , Sensory Receptor Cells/metabolism , Adjuvants, Immunologic/toxicity , Aldosterone/biosynthesis , Animals , Freund's Adjuvant/toxicity , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Pain Measurement/drug effects , Physical Stimulation/adverse effects , Rats , Rats, Wistar , Sensory Receptor Cells/drug effects
3.
Cephalalgia ; 40(1): 79-87, 2020 01.
Article in English | MEDLINE | ID: mdl-31382763

ABSTRACT

BACKGROUND: The role of the trigeminal autonomic reflex in headache syndromes, such as cluster headache, is undisputed but sparsely investigated. The aim of the present study was therefore, to identify neural correlates that play a role in the initiation of the trigeminal autonomic reflex. We further aimed to discriminate between components of the reflex that are involved in nociceptive compared to non-nociceptive processing. METHODS: Kinetic Oscillation Stimulation (KOS) in the left nostril was applied in order to provoke autonomic symptoms (e.g. lacrimation) via the trigeminal autonomic reflex in 26 healthy participants using functional magnetic resonance imaging. Unpleasantness and painfulness were assessed on a visual analog scale (VAS), in order to assess the quality of the stimulus (e.g. pain or no pain). RESULTS: During non-painful activation, specific regions involved in the trigeminal autonomic reflex became activated, including several brainstem nuclei but also cerebellar and bilateral insular regions. However, when the input leading to activation of the trigeminal autonomic reflex was perceived as painful, activation of the anterior hypothalamus, the locus coeruleus (LC), the ventral posteriomedial nucleus of the thalamus (VPM), as well as an activation of ipsilateral insular regions, was observed. CONCLUSION: Our results suggest the anterior hypothalamus, besides the thalamus and specific brain stem regions, play a significant role in networks that mediate autonomic output (e.g. lacrimation) following trigeminal input, but only if the trigeminal system is activated by a stimulus comprising a painful component.


Subject(s)
Discrimination Learning , Hypothalamus/diagnostic imaging , Magnetic Resonance Imaging/methods , Pain/diagnostic imaging , Reflex , Trigeminal Nerve/diagnostic imaging , Adult , Discrimination Learning/physiology , Female , Healthy Volunteers/psychology , Humans , Hypothalamus/physiology , Male , Pain/psychology , Pain Measurement/methods , Pain Measurement/psychology , Physical Stimulation/adverse effects , Proof of Concept Study , Reflex/physiology , Tears/physiology , Trigeminal Nerve/physiology
4.
Support Care Cancer ; 28(11): 5405-5410, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32144582

ABSTRACT

PURPOSE: The incidence and time of onset of acute chemotherapy-induced peripheral neuropathy (ACIPN) caused by oxaliplatin remain unclarified. Hence, we investigated the prevalence, onset time, and location of ACIPN symptoms in patients with colorectal cancer (CRC) receiving oxaliplatin without cold stimulation. METHODS: The study cohort comprised patients receiving oxaliplatin for CRC at our hospital between April 2017 and August 2018. Patients were instructed not to touch and/or drink cold things and were monitored for ACIPN symptoms in the hospital for 24 h after chemotherapy. ACIPN symptoms that appeared > 24 h after chemotherapy were recorded at the next visit. Symptom appearance time was defined as the duration from the administration of chemotherapy until the appearance of paresthesia classified as grade 1 using the Common Terminology Criteria for Adverse Events. RESULTS: Forty-five patients received chemotherapy, comprising 23 men and 22 women, aged 67 years (29-88 years). The location of ACIPN was the fingers in 55.6% of cases, pharynx in 26.7%, perioral region in 24.4%, and feet in 6.7%. The average duration from oxaliplatin administration to symptom development was 182 min (range 62-443 min) for the fingers, 291 min (176-432 min) for the pharynx, 311 min (127-494 min) for the perioral region, and 297 min (234-355 min) for the feet. Pharyngeal symptoms were more common in patients older than 65 years than in those younger than 65 years. CONCLUSIONS: The incidence and time of the onset of ACIPN caused by oxaliplatin varies between the body and regions.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Oxaliplatin/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/epidemiology , Acute Disease , Adult , Aged , Aged, 80 and over , Cold Temperature/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/epidemiology , Female , Humans , Incidence , Male , Middle Aged , Oxaliplatin/administration & dosage , Paresthesia/chemically induced , Paresthesia/epidemiology , Physical Stimulation/adverse effects , Prevalence , Time Factors
5.
Cephalalgia ; 39(13): 1720-1727, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31615269

ABSTRACT

AIM: To describe neuronal networks underlying commonly reported migraine premonitory symptoms and to discuss how these might precipitate migraine pain. BACKGROUND: Migraine headache is frequently preceded by a distinct and well characterized premonitory phase including symptoms like yawning, sleep disturbances, alterations in appetite and food intake and hypersensitivity to certain external stimuli. Recent neuroimaging studies strongly suggest the hypothalamus as the key mediator of the premonitory phase and also suggested alterations in hypothalamic networks as a mechanism of migraine attack generation. When looking at the vast evidence from basic research within the last decades, hypothalamic and thalamic networks are most likely to integrate peripheral influences with central mechanisms, facilitating the precipitation of migraine headaches. These networks include sleep, feeding and stress modulating centers within the hypothalamus, thalamic pathways and brainstem centers closely involved in trigeminal pain processing such as the spinal trigeminal nucleus and the rostral ventromedial medulla, all of which are closely interconnected. CONCLUSION: Taken together, these networks represent the pathophysiological basis for migraine premonitory symptoms as well as a possible integration site of peripheral so-called "triggers" with central attack facilitating processes.


Subject(s)
Migraine without Aura/physiopathology , Prodromal Symptoms , Affect , Appetite/physiology , Brain Stem/physiopathology , Circadian Rhythm/physiology , Craving/physiology , Eating , Homeostasis , Humans , Migraine without Aura/complications , Migraine without Aura/etiology , Migraine without Aura/psychology , Nerve Net/physiopathology , Neuroimaging , Neurotransmitter Agents/physiology , Nitric Oxide/physiology , Photophobia/etiology , Photophobia/physiopathology , Physical Stimulation/adverse effects , Sleep Stages/physiology , Suprachiasmatic Nucleus/physiopathology , Thalamus/physiopathology
6.
Cephalalgia ; 39(13): 1635-1660, 2019 11.
Article in English | MEDLINE | ID: mdl-29969040

ABSTRACT

OBJECTIVE: To review and discuss the literature relevant to the role of brainstem structure and function in headache. BACKGROUND: Primary headache disorders, such as migraine and cluster headache, are considered disorders of the brain. As well as head-related pain, these headache disorders are also associated with other neurological symptoms, such as those related to sensory, homeostatic, autonomic, cognitive and affective processing that can all occur before, during or even after headache has ceased. Many imaging studies demonstrate activation in brainstem areas that appear specifically associated with headache disorders, especially migraine, which may be related to the mechanisms of many of these symptoms. This is further supported by preclinical studies, which demonstrate that modulation of specific brainstem nuclei alters sensory processing relevant to these symptoms, including headache, cranial autonomic responses and homeostatic mechanisms. REVIEW FOCUS: This review will specifically focus on the role of brainstem structures relevant to primary headaches, including medullary, pontine, and midbrain, and describe their functional role and how they relate to mechanisms of primary headaches, especially migraine.


Subject(s)
Brain Stem/physiopathology , Headache/physiopathology , Analgesics/pharmacokinetics , Analgesics/therapeutic use , Blood-Brain Barrier , Cluster Headache/physiopathology , Headache/drug therapy , Headache Disorders, Primary/physiopathology , Homeostasis , Humans , Migraine Disorders/physiopathology , Neural Pathways/physiopathology , Parasympathetic Nervous System/physiopathology , Physical Stimulation/adverse effects , Trigeminal Nerve/physiopathology , Trigeminal Nuclei/physiopathology , Vagus Nerve/physiopathology , Vasodilation
7.
J Neurosci ; 37(49): 11806-11817, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29097595

ABSTRACT

To convert a painful stimulus into a briefly maintainable construct when the painful stimulus is no longer accessible is essential to guide human behavior and avoid dangerous situations. Because of the aversive nature of pain, this encoding process might be influenced by emotional aspects and could thus vary across individuals, but we have yet to understand both the basic underlying neural mechanisms as well as potential interindividual differences. Using fMRI in combination with a delayed-discrimination task in healthy volunteers of both sexes, we discovered that brain regions involved in this working memory encoding process were dissociable according to whether the to-be-remembered stimulus was painful or not, with the medial thalamus and the rostral anterior cingulate cortex encoding painful and the primary somatosensory cortex encoding nonpainful stimuli. Encoding of painful stimuli furthermore significantly enhanced functional connectivity between the thalamus and medial prefrontal cortex (mPFC). With regards to emotional aspects influencing encoding processes, we observed that more anxious participants showed significant performance advantages when encoding painful stimuli. Importantly, only during the encoding of pain, the interindividual differences in anxiety were associated with the strength of coupling between medial thalamus and mPFC, which was furthermore related to activity in the amygdala. These results indicate not only that there is a distinct signature for the encoding of a painful experience in humans, but also that this encoding process involves a strong affective component.SIGNIFICANCE STATEMENT To convert the sensation of pain into a briefly maintainable construct is essential to guide human behavior and avoid dangerous situations. Although this working memory encoding process is implicitly contained in the majority of studies, the underlying neural mechanisms remain unclear. Using fMRI in a delayed-discrimination task, we found that the encoding of pain engaged the activation of the medial thalamus and the functional connectivity between the thalamus and medial prefrontal cortex. These fMRI data were directly and indirectly related to participants' self-reported trait and state anxiety. Our findings indicate that the mechanisms responsible for the encoding of noxious stimuli differ from those for the encoding of innocuous stimuli, and that these mechanisms are shaped by an individual's anxiety levels.


Subject(s)
Anxiety/diagnostic imaging , Mediodorsal Thalamic Nucleus/diagnostic imaging , Memory , Pain Measurement/methods , Pain/diagnostic imaging , Prefrontal Cortex/diagnostic imaging , Adult , Anxiety/psychology , Female , Hot Temperature/adverse effects , Humans , Magnetic Resonance Imaging/methods , Male , Mediodorsal Thalamic Nucleus/physiology , Memory/physiology , Pain/psychology , Physical Stimulation/adverse effects , Prefrontal Cortex/physiology , Random Allocation , Young Adult
8.
Mol Pain ; 14: 1744806918804441, 2018.
Article in English | MEDLINE | ID: mdl-30209982

ABSTRACT

Background The amygdala plays a key role in fear learning and extinction and has emerged as an important node of emotional-affective aspects of pain and pain modulation. Impaired fear extinction learning, which involves prefrontal cortical control of amygdala processing, has been linked to neuropsychiatric disorders. Here, we tested the hypothesis that fear extinction learning ability can predict the magnitude of neuropathic pain. Results We correlated fear extinction learning in naive adult male rats with sensory and affective behavioral outcome measures (mechanical thresholds, vocalizations, and anxiety- and depression-like behaviors) before and after the induction of the spinal nerve ligation model of neuropathic pain compared to sham controls. Auditory fear conditioning, extinction learning, and extinction retention tests were conducted after baseline testing. All rats showed increased freezing responses after fear conditioning. During extinction training, the majority (75%) of rats showed a decline in freezing level to 50% in 5 min (fear extinction+), whereas 25% of the rats maintained a high freezing level (>50%, fear extinction-). Fear extinction- rats showed decreased open-arm preference in the elevated plus maze, reflecting anxiety-like behavior, but there were no significant differences in sensory thresholds, vocalizations, or depression-like behavior (forced swim test) between fear extinction+ and fear extinction- types. In the neuropathic pain model (four weeks after spinal nerve ligation), fear extinction- rats showed a greater increase in vocalizations and anxiety-like behavior than fear extinction+ rats. Fear extinction- rats, but not fear extinction+ rats, also developed depression-like behavior. Extracellular single unit recordings of amygdala (central nucleus) neurons in behaviorally tested rats (anesthetized with isoflurane) found greater increases in background activity, bursting, and evoked activity in fear extinction- rats than fear extinction+ rats in the spinal nerve ligation model compared to sham controls. Conclusion The data may suggest that fear extinction learning ability predicts the magnitude of neuropathic pain-related affective rather than sensory behaviors, which correlates with differences in amygdala activity changes.


Subject(s)
Extinction, Psychological/physiology , Fear/psychology , Learning Disabilities/etiology , Learning Disabilities/pathology , Neuralgia/complications , Acoustic Stimulation , Action Potentials/physiology , Amygdala/pathology , Analysis of Variance , Animals , Conditioning, Classical/physiology , Disease Models, Animal , Male , Mood Disorders/etiology , Neuralgia/psychology , Neurons/physiology , Pain Measurement , Pain Threshold/physiology , Physical Stimulation/adverse effects , Rats , Rats, Sprague-Dawley
9.
Mol Pain ; 14: 1744806918796763, 2018.
Article in English | MEDLINE | ID: mdl-30178698

ABSTRACT

Chronic orofacial pain is a significant health problem requiring identification of regulating processes. Involvement of epigenetic modifications that is reported for hindlimb neuropathic pain experimental models, however, is less well studied in cranial nerve pain models. Three independent observations reported here are the (1) epigenetic profile in mouse trigeminal ganglia (TG) after trigeminal inflammatory compression (TIC) nerve injury mouse model determined by gene expression microarray, (2) H3K9 acetylation pattern in TG by immunohistochemistry, and (3) efficacy of histone deacetylase (HDAC) inhibitors to attenuate development of hypersensitivity. After TIC injury, ipsilateral whisker pad mechanical sensitization develops by day 3 and persists well beyond day 21 in contrast to sham surgery. Global acetylation of H3K9 decreases at day 21 in ipsilateral TG . Thirty-four genes are significantly ( p < 0.05) overexpressed in the ipsilateral TG by at least two-fold at either 3 or 21 days post-trigeminal inflammatory compression injury. The three genes most overexpressed three days post-trigeminal inflammatory compression nerve injury are nerve regeneration-associated gene ATF3, up 6.8-fold, and two of its regeneration-associated gene effector genes, Sprr1a and Gal, up 174- and 25-fold, respectively. Although transcription levels of 25 of 32 genes significantly overexpressed three days post-trigeminal inflammatory compression return to constitutive levels by day 21, these three regeneration-associated genes remain significantly overexpressed at the later time point. On day 21, when tissues are healed, other differentially expressed genes include 39 of the top 50 upregulated and downregulated genes. Remarkably, preemptive manipulation of gene expression with two HDAC inhibitors (HDACi's), suberanilohydroxamic acid (SAHA) and MS-275, reduces the magnitude and duration of whisker pad mechanical hypersensitivity and prevents the development of a persistent pain state. These findings suggest that trigeminal nerve injury leads to epigenetic modifications favoring overexpression of genes involved in nerve regeneration and that maintaining transcriptional homeostasis with epigenetic modifying drugs could help prevent the development of persistent pain.


Subject(s)
Facial Pain/complications , Gene Expression Regulation/physiology , Histone Deacetylase Inhibitors/therapeutic use , Hyperalgesia/etiology , Hyperalgesia/prevention & control , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Benzamides/therapeutic use , Cornified Envelope Proline-Rich Proteins/genetics , Cornified Envelope Proline-Rich Proteins/metabolism , Disease Models, Animal , Facial Pain/etiology , Facial Pain/pathology , Functional Laterality , Ganglia, Spinal/pathology , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Male , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nylons , Pain Threshold/drug effects , Physical Stimulation/adverse effects , Pyridines/therapeutic use , Pyrroles/therapeutic use , Trigeminal Nerve Injuries/complications , Vibrissae/innervation
10.
Mol Pain ; 14: 1744806918783478, 2018.
Article in English | MEDLINE | ID: mdl-29956582

ABSTRACT

Background Chronic pain is a persistent unpleasant sensation that produces pathological synaptic plasticity in the central nervous system. Both human imaging study and animal studies consistently demonstrate that the anterior cingulate cortex is a critical cortical area for nociceptive and chronic pain processing. Thus far, the mechanisms of excitatory synaptic transmission and plasticity have been well characterized in the anterior cingulate cortex for various models of chronic pain. By contrast, the potential contribution of inhibitory synaptic transmission in the anterior cingulate cortex, in models of chronic pain, is not fully understood. Methods Chronic inflammation was induced by complete Freund adjuvant into the adult mice left hindpaw. We performed in vitro whole-cell patch-clamp recordings from layer II/III pyramidal neurons in two to three days after the complete Freund adjuvant injection and examined if the model could cause plastic changes, including transient and tonic type A γ-aminobutyric acid (GABAA) receptor-mediated inhibitory synaptic transmission, in the anterior cingulate cortex. We analyzed miniature/spontaneous inhibitory postsynaptic currents, GABAA receptor-mediated tonic currents, and evoked inhibitory postsynaptic currents. Finally, we studied if GABAergic transmission-related proteins in the presynapse and postsynapse of the anterior cingulate cortex were altered. Results The complete Freund adjuvant model reduced the frequency of both miniature and spontaneous inhibitory postsynaptic currents compared with control group. By contrast, the average amplitude of these currents was not changed between two groups. Additionally, the complete Freund adjuvant model did not change GABAA receptor-mediated tonic currents nor the set of evoked inhibitory postsynaptic currents when compared with control group. Importantly, protein expression of vesicular GABA transporter was reduced within the presynpase of the anterior cingulate cortex in complete Freund adjuvant model. In contrast, the complete Freund adjuvant model did not change the protein levels of GABAA receptors subunits such as α1, α5, ß2, γ2, and δ. Conclusion Our results suggest that the induction phase of inflammatory pain involves spontaneous GABAergic plasticity at presynaptic terminals of the anterior cingulate cortex.


Subject(s)
Chronic Pain/complications , Chronic Pain/pathology , Gyrus Cinguli/pathology , Inflammation/etiology , Neuronal Plasticity/physiology , Pain Threshold/physiology , gamma-Aminobutyric Acid/metabolism , Anesthetics, Local/pharmacology , Anesthetics, Local/therapeutic use , Animals , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Chronic Pain/chemically induced , Chronic Pain/drug therapy , Freund's Adjuvant/toxicity , GABA-A Receptor Antagonists/pharmacology , Gyrus Cinguli/cytology , In Vitro Techniques , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Physical Stimulation/adverse effects , Synaptic Potentials/drug effects , Synaptic Potentials/physiology , Tetrodotoxin/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
11.
Mol Pain ; 14: 1744806918795930, 2018.
Article in English | MEDLINE | ID: mdl-30146940

ABSTRACT

Objective Previous studies of neuropathic pain have suggested that the P2X4 purinoceptor (P2X4R) in spinal microglia is essential for maintaining allodynia following nerve injury. However, little is known about its role in inflammatory soup-induced trigeminal allodynia, which closely mimics chronic migraine status. Here, we determined the contributions of P2X4R and related signaling pathways in an inflammatory soup-induced trigeminal allodynia model. Methods P2X4R gene and protein levels in the trigeminal nucleus caudalis were analyzed following repeated dural inflammatory soup infusions. p38, brain-derived neurotrophic factor, excitatory amino acid transporter 3, c-Fos, and calcitonin gene-related peptide protein levels in the trigeminal nucleus caudalis, as well as trigeminal sensitivity, were assessed among the different groups. Immunofluorescence staining was used to detect protein localization and expression in the trigeminal nucleus caudalis. Results Repeated inflammatory dural stimulation induced trigeminal hyperalgesia and the upregulation of P2X4R. Immunofluorescence revealed that P2X4R was expressed in trigeminal nucleus caudalis microglial cells. Blockage of P2X4R produced an anti-nociceptive effect, which was associated with an inhibition of inflammatory soup-induced increases in p38, brain-derived neurotrophic factor, excitatory amino acid transporter 3, c-Fos, and calcitonin gene-related peptide protein levels. The tyrosine receptor kinase B antagonist ANA-12 reversed trigeminal allodynia and the upregulation of excitatory amino acid transporter 3, c-Fos, and calcitonin gene-related peptide, whereas the agonist 7,8-dihydroxyflavone exacerbated these effects. Double immunostaining indicated that p38 and brain-derived neurotrophic factor were mainly expressed in microglial cells, whereas excitatory amino acid transporter 3 was primarily expressed in trigeminal nucleus caudalis neurons. Conclusions These data indicate that microglial P2X4R is involved in the regulation of excitatory amino acid transporter 3 via brain-derived neurotrophic factor-tyrosine receptor kinase B signaling following repeated inflammatory dural stimulation. Microglial P2X4R activation and microglia-neuron interactions in the trigeminal nucleus caudalis may play a role in the pathogenesis of migraine chronicity, and the modulation of P2X4R activation might be a potential therapeutic strategy.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Hyperalgesia/etiology , Receptors, Purinergic P2X4/metabolism , Signal Transduction/physiology , Trigeminal Neuralgia/complications , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/toxicity , Analysis of Variance , Animals , Azepines/pharmacology , Benzamides/pharmacology , Disease Models, Animal , Male , Physical Stimulation/adverse effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4/genetics , Signal Transduction/drug effects , Trigeminal Neuralgia/chemically induced
12.
J Neuroinflammation ; 15(1): 245, 2018 Aug 30.
Article in English | MEDLINE | ID: mdl-30165876

ABSTRACT

BACKGROUND: The mechanism underlying migraine chronification remains unclear. Central sensitization may account for this progression. The microglia P2X4 receptor (P2X4R) plays a pivotal role in the central sensitization of inflammatory and neuropathic pain, but there is no information about P2X4R in migraine. Therefore, the aim of this study was to identify the precise role of microglia P2X4R in chronic migraine (CM). METHODS: We used an animal model with recurrent intermittent administration of nitroglycerin (NTG), which closely mimics CM. NTG-induced basal and acute mechanical hypersensitivity were evaluated using the von Frey filament test. Then, we detected Iba1 immunoreactivity (Iba1-IR) and P2X4R expression in the trigeminal nucleus caudalis (TNC). To understand the effect of microglia and P2X4R on central sensitization of CM, we examined whether minocycline, an inhibitor of microglia activation, and 5-BDBD, a P2X4R antagonist, altered NTG-induced mechanical hyperalgesia. In addition, we also evaluated the effect of 5-BDBD on c-Fos and calcitonin gene-related peptide (CGRP) expression within the TNC. RESULTS: Chronic intermittent administration of NTG resulted in acute and chronic basal mechanical hyperalgesia, accompanied with microglia activation and upregulation of P2X4R expression. Minocycline significantly decreased basal pain hypersensitivity but did not alter acute NTG-induced hyperalgesia. Minocycline also reduced microglia activation. 5-BDBD completely blocked the basal and acute hyperalgesia induced by NTG. This effect was associated with a significant inhibition of the NTG-induced increase in c-Fos protein and CGRP release in the TNC. CONCLUSIONS: Our results indicate that blocking microglia activation may have an effect on the prevention of migraine chronification. Moreover, we speculate that the P2X4R may be implicated in the microglia-neuronal signal in the TNC, which contributes to the central sensitization of CM.


Subject(s)
Microglia/metabolism , Migraine Disorders/chemically induced , Migraine Disorders/pathology , Nitroglycerin , Receptors, Purinergic P2X4/metabolism , Animals , Benzodiazepinones/pharmacology , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hyperalgesia/etiology , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Migraine Disorders/complications , Minocycline/pharmacology , Pain Threshold/drug effects , Pain Threshold/physiology , Physical Stimulation/adverse effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/genetics , Trigeminal Nucleus, Spinal/drug effects , Trigeminal Nucleus, Spinal/metabolism , Trigeminal Nucleus, Spinal/pathology
13.
J Neuroinflammation ; 15(1): 232, 2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30131066

ABSTRACT

BACKGROUND: Chemotherapy-induced peripheral neurotoxicity (CIPN) is a severe adverse effect in patients receiving antitumor agents, and no effective treatment is available. Although the mechanisms responsible for the development of CIPN are poorly understood, recent findings make neuroinflammation an attractive target to be investigated, particularly when neuropathic pain is a prominent feature such as after bortezomib administration. The aim of our study was to evaluate the effect of intravenous immunoglobulins (IVIg) delivery in chronic CIPN. The related neuro-immune aspects were investigated in a well-characterized rat model of bortezomib-induced peripheral neurotoxicity (BIPN). METHODS: After determination of a suitable schedule based on a preliminary pharmacokinetic pilot study, female Wistar rats were treated with IVIg 1 g/kg every 2 weeks. IVIg treatment was started at the beginning of bortezomib administration ("preventive" schedule), or once BIPN was already ensued after 4 weeks of treatment ("therapeutic" schedule). Neurophysiological and behavioral studies were performed to assess the extent of painful peripheral neurotoxicity induced by bortezomib, and these functional assessments were completed by pathologic examination of peripheral nerves and intraepidermal nerve fiber quantification (IENF). The role of the innate immune response in BIPN was investigated by immunochemistry characterization of macrophage infiltration in peripheral nerves. RESULTS: Both schedules of IVIg administration were able to significantly reduce bortezomib-induced heat and mechanical allodynia. Although these changes were not evidenced at the neurophysiological examination of peripheral nerves, they behavioral effects were paralleled in the animals treated with the preventive schedule by reduced axonopathy in peripheral nerves and significant protection from loss of IENF. Moreover, IVIg administration was very effective in reducing infiltration in peripheral nerves of macrophages with the M1, pro-inflammatory phenotype. CONCLUSION: Our results suggest a prominent role of neuroinflammation in BIPN and that IVIg might be considered as a possible safe and effective therapeutic option preventing M1 macrophage infiltration. However, since neuropathic pain is frequent also in other CIPN types, it also indicates the need for further investigation in other forms of CIPN.


Subject(s)
Immunoglobulins/therapeutic use , Immunologic Factors/therapeutic use , Macrophages/drug effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/pathology , Peripheral Nerves/pathology , Animals , Antineoplastic Agents/toxicity , Body Weight/drug effects , Bortezomib/toxicity , Cytokines/metabolism , Disease Models, Animal , Hot Temperature/adverse effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Macrophages/pathology , Nerve Fibers/drug effects , Nerve Fibers/pathology , Neural Conduction/drug effects , Neurotoxicity Syndromes/etiology , Neutrophil Infiltration , Physical Stimulation/adverse effects , Rats , Sensory Thresholds/drug effects , Skin/pathology
14.
Cephalalgia ; 38(6): 1049-1056, 2018 05.
Article in English | MEDLINE | ID: mdl-28708009

ABSTRACT

Introduction Although it is widely accepted that facial pain paroxysms triggered by innocuous stimuli constitute a hallmark sign of trigeminal neuralgia, very few studies to date have systematically investigated the role of the triggers involved. In the recently published diagnostic classification, triggered pain is an essential criterion for the diagnosis of trigeminal neuralgia but no study to date has been designed to address this issue directly. In this study, we set out to determine, in patients with trigeminal neuralgia, how frequently triggers are present, which manoeuvres activate them and where cutaneous and mucosal trigger zones are located. Methods Clinical characteristics focusing on trigger factors were collected from 140 patients with trigeminal neuralgia, in a cross-sectional study design. Results Provocation of paroxysmal pain by various trigger manoeuvres was reported by 136 of the 140 patients. The most frequent manoeuvres were gentle touching of the face (79%) and talking (54%). Trigger zones were predominantly reported in the perioral and nasal region. Conclusion This study confirms that in trigeminal neuralgia, paroxysmal pain is associated with triggers in virtually all patients and supports the use of triggers as an essential diagnostic feature of trigeminal neuralgia.


Subject(s)
Physical Stimulation/adverse effects , Trigeminal Neuralgia/etiology , Adult , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Trigeminal Neuralgia/physiopathology , Trigger Points/physiopathology
15.
Exp Brain Res ; 236(7): 2097-2105, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29754196

ABSTRACT

Exposure to moderate levels of ultraviolet B radiation (UVB) is painless but nevertheless induces an inflammatory response that sensitizes primary afferent nociceptors. Subsequently, heating the UVB-treated site can sensitize spinal nociceptors. We used a repeated-measures design to determine whether heating the UVB-treated site also triggers ipsilateral inhibitory controls. Specifically, a 2-cm diameter site on the forearm of 20 participants was exposed to UVB at twice the minimum erythema dose. 48 h later mechanical and thermal sensitivity had increased at the UVB-treated site, indicating primary hyperalgesia. In addition, sensitivity to blunt pressure had increased in the ipsilateral forehead, implying activation of an ipsilateral supra-spinal pro-nociceptive mechanism. Despite this, the area under the curve of the ipsilateral nociceptive blink reflex decreased when the UVB-treated site was heated to induce moderate pain. Together, these findings suggest that the UVB treatment sensitized primary nociceptive afferents and generated an ipsilateral supra-spinal pro-nociceptive mechanism. In addition, sensitization to heat induced by the UVB treatment strengthened an ipsilateral anti-nociceptive process elicited by heat-pain. Infrequent but enduring discharge of sensitized primary nociceptive afferents, driven by inflammation after UVB exposure, might initiate a lateralized supra-spinal pro-nociceptive influence that heightens awareness of impending harm on the sensitized side of the body. In addition, a lateralized anti-nociceptive response triggered by an intense barrage of nociceptive signals may help to differentiate stronger from weaker sources of pain.


Subject(s)
Functional Laterality/physiology , Nociception/physiology , Pain/physiopathology , Reflex/physiology , Adult , Female , Forehead/innervation , Humans , Hyperalgesia/physiopathology , Male , Middle Aged , Pain/etiology , Physical Stimulation/adverse effects , Psychophysics , Skin/innervation , Ultraviolet Rays/adverse effects , Young Adult
16.
Headache ; 58(1): 62-77, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29094347

ABSTRACT

OBJECTIVE: The current study examined pain and neurogenic inflammation responses to topical capsaicin during the interictal period (between headache) and their relationship with plasma oxytocin in individuals with migraine. BACKGROUND: Individuals with migraine can experience generalized (extracephalic) hyperalgesia, which can persist even between headache attacks. Elevated levels of plasma and cerebrospinal fluid oxytocin have been observed during migraine attacks, oxytocin levels being positively associated with the intensity of migraine symptoms. However, whether oxytocin plays a role in the mechanisms of generalized pain sensitization and neurogenic inflammation during the interictal period has not been studied yet. Understanding migraineurs' interictal pain phenotype and endogenous oxytocin might help identify individuals who would benefit from intranasal oxytocin treatment. METHODS: Thirty-two subjects with migraine and 26 healthy controls underwent pain testing. The current study compared capsaicin-induced pain, central sensitization (areas of secondary mechanical allodynia and hyperalgesia), and neurogenic inflammation (capsaicin-induced flare) responses on the nondominant volar forearm between migraineurs and healthy controls. Additionally, we studied plasma oxytocin levels and their relationship to migraine symptoms, experimental pain and affect. RESULTS: The results indicated a significant group effect (P = .019): Migraineurs reported greater capsaicin-induced pain unpleasantness (M = 1.2, SD = 1.4) on a 0-10 scale and showed larger areas of flare (LnM = 2.8, SD = 0.4) than healthy controls (M = 0.5, SD = 0.8; LnM = 2.6, SD = 0.4; ps < .032). In a subgroup analysis, enhanced capsaicin-induced pain unpleasantness was found in the chronic (P = .007), but not the episodic (Ps > .200), migraineurs. The oxytocin levels were elevated in migraineurs and accounted for 18% of the group difference in capsaicin-induced pain unpleasantness. Within migraineurs, interictal oxytocin levels were negatively associated with psychological distress (Ps < .030). However, during the interictal period, pain sensitivity in extracephalic regions and plasma oxytocin levels were unrelated to migraine symptom parameters (Ps > .074). Lastly, the results found no group difference in areas of secondary mechanical allodynia and hyperalgesia (Ps >.298). CONCLUSION: The current study revealed that individuals with migraine exhibit enhanced extracephalic capsaicin-induced pain unpleasantness and flare responses during interictal periods. In addition, migraineurs, especially those with chronic migraine, had slightly elevated interictal oxytocin levels compared to controls, which was associated with their affective component of experimental pain. Therefore, treatment targeting affective pain during the interictal period may help to reduce generalized pain in migraine. Furthermore, endogenous increases in oxytocin may be a compensatory mechanism that may help decrease affective distress in migraineurs. The therapeutic effects of intranasal oxytocin may benefit migraineurs by reducing their affective distress.


Subject(s)
Hyperalgesia/physiopathology , Migraine Disorders/blood , Migraine Disorders/physiopathology , Oxytocin/blood , Pain Threshold/physiology , Adolescent , Capsaicin/adverse effects , Cross-Sectional Studies , Female , Humans , Male , Pain/chemically induced , Pain/metabolism , Pain/psychology , Pain Measurement , Physical Stimulation/adverse effects , Sensory System Agents/adverse effects , Statistics, Nonparametric , Surveys and Questionnaires , Time Factors , Young Adult
17.
Cereb Cortex ; 27(7): 3752-3767, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28169393

ABSTRACT

Prenatal alcohol exposure has been linked to a broad range of developmental deficits, with eyeblink classical conditioning (EBC) among the most sensitive endpoints. This fMRI study compared EBC-related brain activity in 47 children with fetal alcohol syndrome (FAS), partial FAS (PFAS), heavily exposed (HE) non-syndromal children, and healthy controls. All of the children had previously participated in two EBC studies conducted as part of our longitudinal study of fetal alcohol spectrum disorders. Although learning-related behavioral differences were seen in all groups during the scans, controls showed more conditioned responses (CR) than the alcohol-exposed groups. Despite lower conditioning levels relative to controls, the exposed groups exhibited extensive cerebellar activations. Specifically, children with FAS/PFAS showed increased activation of cerebellar lobule VI in session 2, while HE children showed increased activation in session 1. Continuous measures of prenatal alcohol use correlated with learning-related activations in cerebellum and frontal cortices. Only controls showed significant cerebellar activation-CR correlations in the deep nuclei and lateral lobule VI, suggesting that these key regions supporting EBC may be functionally disorganized in alcohol-exposed children. These findings are the first to characterize abnormalities in brain function associated with the behavioral conditioning deficits seen in children with prenatal alcohol exposure.


Subject(s)
Blinking/physiology , Brain/diagnostic imaging , Conditioning, Classical/physiology , Fetal Alcohol Spectrum Disorders/diagnostic imaging , Fetal Alcohol Spectrum Disorders/physiopathology , Magnetic Resonance Imaging , Alcohol Drinking/physiopathology , Analysis of Variance , Child , Cohort Studies , Female , Functional Laterality , Humans , Image Processing, Computer-Assisted , Male , Maternal-Fetal Exchange , Oxygen , Physical Stimulation/adverse effects , Pregnancy , Prenatal Diagnosis
18.
Somatosens Mot Res ; 35(3-4): 192-198, 2018.
Article in English | MEDLINE | ID: mdl-30461318

ABSTRACT

BACKGROUND: Many researchers have tried to investigate pain by studying brain responses. One method used to investigate pain-related brain responses is continuous electroencephalography (EEG). The objective of the current study is to add on to our understanding of EEG responses during pain, by differentiation between EEG patterns indicative of (i) the noxious stimulus intensity and (ii) the subjective pain sensation. METHODS: EEG was recorded during the administration of tonic experimental pain, consisting of six minutes of contact heat applied to the leg via a thermode. Two stimuli above pain threshold, one at pain threshold and two non-painful stimuli were administered. Thirty-six healthy participants provided a subjective pain rating during thermal stimulation. Relative EEG power was calculated for the frequency bands alpha1, alpha2, beta1, beta2, delta, and theta. RESULTS: Whereas EEG activity could not be predicted by stimulus intensity (except in one frequency band), subjective pain sensation could significantly predict differences in EEG activity in several frequency bands. An increase in the subjective pain sensation was associated with a decrease in alpha2, beta1, beta2 as well as in theta activity across the midline electrodes. CONCLUSION: The subjective experience of pain seems to capture unique variance in EEG activity above and beyond what is captured by noxious stimulus intensity.


Subject(s)
Evoked Potentials, Somatosensory/physiology , Hyperalgesia/physiopathology , Pain Perception/physiology , Pain Threshold/physiology , Pain/physiopathology , Adult , Female , Hot Temperature/adverse effects , Humans , Male , Pain Measurement , Physical Stimulation/adverse effects , Regression Analysis , Young Adult
19.
Brain Inj ; 32(13-14): 1866-1878, 2018.
Article in English | MEDLINE | ID: mdl-30346868

ABSTRACT

Blast-induced traumatic brain injury (blast-TBI) is associated with vestibulomotor dysfunction, persistent post-traumatic headaches and post-traumatic stress disorder, requiring extensive treatments and reducing quality-of-life. Treatment and prevention of these devastating outcomes require an understanding of their underlying pathophysiology through studies that take advantage of animal models. Here, we report that cranium-directed blast-TBI in rats results in signs of pain that last at least 8 weeks after injury. These occur without significantly elevated behavioural markers of anxiety-like conditions and are not associated with glial up-regulation in sensory thalamic nuclei. These injuries also produce transient vestibulomotor abnormalities that resolve within 3 weeks of injury. Thus, blast-TBI in rats recapitulates aspects of the human condition.


Subject(s)
Brain Injuries/complications , Facial Pain/etiology , Reflex, Vestibulo-Ocular/physiology , Sensation Disorders/etiology , Analysis of Variance , Animals , Blast Injuries/complications , Brain Injuries/etiology , Dark Adaptation/physiology , Disease Models, Animal , Exploratory Behavior/physiology , Hyperalgesia/diagnosis , Hyperalgesia/etiology , Male , Maze Learning , Neuroglia/metabolism , Neuroglia/pathology , Pain Measurement , Pain Threshold/physiology , Physical Stimulation/adverse effects , Postural Balance , Rats , Rats, Long-Evans , Rotarod Performance Test , Thalamus/pathology , Time Factors
20.
Eur J Anaesthesiol ; 35(7): 505-510, 2018 07.
Article in English | MEDLINE | ID: mdl-29538008

ABSTRACT

BACKGROUND: Drugs with antagonistic actions on the Toll-like receptor 4 (Tlr4), such as naloxone at ultra low doses, have been used to inhibit opioid-induced hyperalgesia in rodents suggesting the involvement of this receptor and pathway on opioid-induced hyperalgesia. OBJECTIVE: The aim of this study was to determine whether mice without the Tlr4 gene (Tlr4) would not develop remifentanil-induced hyperalgesia. DESIGN: An experimental randomised animal study. SETTING: Experimental Unit, Complutense University of Madrid, Madrid, Spain. ANIMALS: Twelve adult female wild-type mice and 12 adult Tlr4 mice. INTERVENTIONS: Under sevoflurane anaesthesia, a 1-h, constant rate subcutaneous infusion of remifentanil (4 µg kg min) or 0.9% saline. MAIN OUTCOME MEASURES: Mechanical nociceptive thresholds were evaluated using a von Frey hair test before (baseline) and on days 5, 6 and 7 after treatment. Hyperalgesia was considered to be a decrease in the mechanical nociceptive threshold. Changes in mechanical nociceptive thresholds in the different groups were compared with one-sided paired t tests. RESULTS: Baseline mechanical nociceptive thresholds were similar in all groups (2.2 ±â€Š0.1 g). Remifentanil produced a 24% decrease in mechanical nociceptive thresholds in the wild-type mice (1.7 ±â€Š0.0 g, averaged over 3 days, P = 0.00021), whereas the nociceptive thresholds were not changed in Tlr4 mice (2.2 ±â€Š0.1 g, P = 0.857) or in mice receiving 0.9% saline (Tlr4, 2.2 ±â€Š0.1 g, P = 0.807; wild-type, 2.2 ±â€Š0.1 g, P = 0.962). CONCLUSION: Tlr4 receptor involvement is suggested in the development of remifentanil-induced hyperalgesia in mice. TRIAL REGISTRATION: CEA-UCM 107/2012.


Subject(s)
Analgesics, Opioid/toxicity , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Remifentanil/toxicity , Toll-Like Receptor 4/deficiency , Animals , Female , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Physical Stimulation/adverse effects , Random Allocation , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL