Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Proc Natl Acad Sci U S A ; 114(28): E5683-E5692, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652355

ABSTRACT

Migraineurs avoid light because it intensifies their headache. However, this is not the only reason for their aversion to light. Studying migraineurs and control subjects, we found that lights triggered more changes in autonomic functions and negative emotions during, rather than in the absence of, migraine or in control subjects, and that the association between light and positive emotions was stronger in control subjects than migraineurs. Seeking to define a neuroanatomical substrate for these findings, we showed that, in rats, axons of retinal ganglion cells converge on hypothalamic neurons that project directly to nuclei in the brainstem and spinal cord that regulate parasympathetic and sympathetic functions and contain dopamine, histamine, orexin, melanin-concentrating hormone, oxytocin, and vasopressin. Although the rat studies define frameworks for conceptualizing how light triggers the symptoms described by patients, the human studies suggest that the aversive nature of light is more complex than its association with headache intensification.


Subject(s)
Hypothalamus/physiology , Light , Migraine Disorders/physiopathology , Neurons/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Autonomic Nervous System/physiology , Case-Control Studies , Color , Emotions , Female , Humans , Male , Middle Aged , Models, Neurological , Photophobia , Rats , Rats, Sprague-Dawley , Retina/physiology , Sympathetic Nervous System/physiology , Young Adult
2.
Ann Neurol ; 73(6): 741-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23447360

ABSTRACT

OBJECTIVE: To examine changes in the response properties of meningeal nociceptors that might lead to migraine pain and examine endogenous processes that could play a role in mediating them using a clinically relevant model of migraine triggering, namely infusion of the nitric oxide (NO) donor nitroglycerin (NTG). METHODS: Single-unit recordings made in the trigeminal ganglion of rats were used to test changes in the activity and mechanosensitivity of meningeal nociceptors in response to administration of the migraine trigger NTG or another NO donor S-nitroso-N-acetyl-DL-penicillamine (SNAP) at doses relevant to the human model of migraine headache. Immunohistochemistry and pharmacological manipulations were used to investigate the possible role of meningeal vascular signaling in mediating the responses of meningeal nociceptors to NO. RESULTS: Infusion of NTG promoted a delayed and robust increase in the mechanosensitivity of meningeal nociceptors, with a time course resembling the development of the delayed migraine headache. A similar sensitization was elicited by dural application of NTG and SNAP. NTG-evoked delayed meningeal nociceptor sensitization was associated with a robust extracellular signal-regulated kinase (ERK) phosphorylation in meningeal arteries. Pharmacological blockade of meningeal ERK phosphorylation inhibited the development of NTG-evoked delayed meningeal nociceptor sensitization. INTERPRETATION: The development of delayed mechanical sensitization evoked by the migraine trigger NTG is potentially of great importance as the first finding of a neurophysiological correlate of migraine headache in meningeal nociceptors. The arterial ERK phosphorylation and its involvement in mediating the NTG-evoked delayed sensitization points to an important, yet unappreciated, role of the meningeal vasculature in the genesis of migraine pain.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Meninges/blood supply , Meninges/enzymology , Migraine Disorders/enzymology , Nociceptors/enzymology , Animals , Male , Meninges/drug effects , Migraine Disorders/chemically induced , Nitric Oxide Donors/toxicity , Nociceptors/drug effects , Rats , Rats, Sprague-Dawley , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/enzymology
3.
Epilepsy Behav ; 28(2): 243-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23562239

ABSTRACT

We proposed recently that induction of delayed activation of trigeminovascular neurons by cortical spreading depression (CSD) can explain the delayed onset of headache after the migraine aura ("aura"). This prompted us to search for ways to block the neuronal activation by CSD - a preclinical correlate of an attempt to find a drug that can block the initiation of headache when administered shortly after onset of aura (i.e., preemptively). Because migraine headache and epileptic seizures are comorbid chronic neurological disorders characterized by hyperexcitable brain networks, we began the search for such goal with an M-type potassium channel opener. We opted to use ezogabine, recently approved by the FDA as adjunctive treatment of partial onset seizures in adults, because it is a selective KCNQ2/3 channel opener. When CSD was induced before ezogabine injection (8.25 mg/kg, i.p.), 40% (6/15) of the units doubled their firing rate about 45 min later for about 95 min. Similarly, when CSD was induced before vehicle was injected (4% DMSO, 0.5% methylcellulose), 50% (3/6) of the units doubled their firing rate about 30 min later for about 120 min. When CSD was triggered 1h after ezogabine injection, it activated only 8% of the units. By itself, ezogabine injection resulted in a 30% attenuation of ongoing firing in all 10 control units. Thus, activation of KCNQ2/3 channels during the aura is unlikely to preempt the onset of headache but may reduce the incidence of migraine if given during prodromes that precede the headache by hours. Given the mechanistic similarities between migraine aura and epileptic seizures, it may be worthwhile to determine whether preemptive administration of ezogabine can prevent oncoming seizures in patients whose warning signs precede their seizures by more than an hour.


Subject(s)
Anticonvulsants/administration & dosage , Carbamates/administration & dosage , Cortical Spreading Depression/drug effects , Nociceptors/drug effects , Phenylenediamines/administration & dosage , Trigeminal Ganglion/cytology , Action Potentials/drug effects , Animals , Cortical Spreading Depression/physiology , Drug Administration Schedule , Male , Pain Measurement , Physical Stimulation , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Time Factors
4.
J Neurosci ; 31(40): 14204-17, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976505

ABSTRACT

This study identifies massive axonal arbors of trigeminovascular (dura-sensitive) thalamic neurons in multiple cortical areas and proposes a novel framework for conceptualizing migraine headache and its associated symptoms. Individual dura-sensitive neurons identified and characterized electrophysiologically in first-order and higher-order relay thalamic nuclei were juxtacellularly filled with an anterograde tracer that labeled their cell bodies and processes. First-order neurons located in the ventral posteromedial nucleus projected mainly to trigeminal areas of primary (S1) as well as secondary (S2) somatosensory and insular cortices. Higher-order neurons located in the posterior (Po), lateral posterior (LP), and lateral dorsal (LD) nuclei projected to trigeminal and extra-trigeminal areas of S1 and S2, as well as parietal association, retrosplenial, auditory, ectorhinal, motor, and visual cortices. Axonal arbors spread at various densities across most layers of the different cortical areas. Such parallel network of thalamocortical projections may play different roles in the transmission of nociceptive signals from the meninges to the cortex. The findings that individual dura-sensitive Po, LP, and LD neurons project to many functionally distinct and anatomically remote cortical areas extend current thinking on projection patterns of high-order thalamic neurons and position them to relay nociceptive information directly rather than indirectly from one cortical area to another. Such extensive input to diverse cortical areas that are involved in regulation of affect, motor function, visual and auditory perception, spatial orientation, memory retrieval, and olfaction may explain some of the common disturbances in neurological functions during migraine.


Subject(s)
Cerebral Cortex/physiology , Migraine Disorders/pathology , Migraine Disorders/physiopathology , Neurons/physiology , Thalamic Nuclei/physiology , Trigeminal Nerve/physiology , Animals , Cerebral Cortex/cytology , Male , Migraine Disorders/diagnosis , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Thalamic Nuclei/cytology
5.
Ann Neurol ; 69(5): 855-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21416489

ABSTRACT

OBJECTIVE: Cortical spreading depression (CSD) has long been implicated in migraine attacks that begin with visual aura. Having shown that a wave of CSD can trigger long-lasting activation of meningeal nociceptors--the first-order neurons of the trigeminovascular pathway thought to underlie migraine headache--we now report that CSD can activate central trigeminovascular neurons in the spinal trigeminal nucleus (C1-2). METHODS: Stimulation of the cortex with pinprick or KCl granule was used to induce CSD in anesthetized rats. Neuronal activity was monitored in C1-2 using single-unit recording. RESULTS: In 25 trigeminovascular neurons activated by CSD, mean firing rate (spikes/s) increased from 3.6 ± 1.2 before CSD (baseline) to 6.1 ± 1.8 after CSD (p < 0.0001) for a period >13 minutes. Neuronal activity returned to baseline level after 30.0 ± 3.1 minutes in 14 units, and remained elevated for 66.0 ± 8.3 (22-108) minutes through the entire recording period in the other 11 units. Neuronal activation began within 0.9 ± 0.4 (0-2.5) minutes after CSD in 7 neurons located in laminae I-II, or after a latency of 25.1 ± 4.0 (7-75) minutes in 9 neurons located in laminae I-II, and 9 neurons located in laminae III-V. In 27 trigeminovascular neurons not activated by CSD, mean firing rate was 2.0 ± 0.7 at baseline and 1.8 ± 0.7 after CSD. INTERPRETATION: We propose that CSD constitutes a nociceptive stimulus capable of activating peripheral and central trigeminovascular neurons that underlie the headache of migraine with aura.


Subject(s)
Action Potentials/physiology , Cortical Spreading Depression/physiology , Neurons/physiology , Trigeminal Nerve/physiology , Trigeminal Nucleus, Spinal/cytology , Animals , Brain Mapping , Male , Neurons/classification , Nociceptors/physiology , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Stimulation, Chemical
6.
Brain Behav Immun ; 26(2): 311-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22019552

ABSTRACT

Mast cells (MCs) are tissue resident immune cells that participate in a variety of allergic and other inflammatory conditions. In most tissues, MCs are found in close proximity to nerve endings of primary afferent neurons that signal pain (i.e. nociceptors). Activation of MCs causes the release of a plethora of mediators that can activate these nociceptors and promote pain. Although MCs are ubiquitous, conditions associated with systemic MC activation give rise primarily to two major types of pain, headache and visceral pain. In this study we therefore examined the extent to which systemic MC degranulation induced by intraperitoneal administration of the MC secretagogue compound 48/80 activates pain pathways that originate in different parts of the body and studied whether this action can lead to development of behavioral pain hypersensitivity. Using c-fos expression as a marker of central nervous system neural activation, we found that intraperitoneal administration of 48/80 leads to the activation of dorsal horn neurons at two specific levels of the spinal cord; one responsible for processing cranial pain, at the medullary/C2 level, and one that processes pelvic visceral pain, at the caudal lumbar/rostral sacral level (L6-S2). Using behavioral sensory testing, we found that this nociceptive activation is associated with development of widespread tactile pain hypersensitivity within and outside the body regions corresponding to the activated spinal levels. Our data provide a neural basis for understanding the primacy of headache and visceral pain in conditions that involve systemic MC degranulation. Our data further suggest that MC activation may lead to widespread tactile pain hypersensitivity.


Subject(s)
Low Back Pain/etiology , Mastocytosis/complications , Neck Pain/etiology , Pain/etiology , Animals , Male , Mast Cells/drug effects , Mast Cells/physiology , Mastocytosis/chemically induced , Pain/physiopathology , Pain Measurement , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/physiology , Rats , Rats, Sprague-Dawley , Touch/drug effects , Touch/physiology , p-Methoxy-N-methylphenethylamine/pharmacology
7.
J Neurosci ; 30(26): 8807-14, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20592202

ABSTRACT

Attacks of migraine with aura represent a phenomenon in which abnormal neuronal activity in the cortex produces sensory disturbances (aura) some 20-40 min before the onset of headache. The purpose of this study was to determine whether cortical spreading depression (CSD)--an event believed to underlie visual aura--can give rise to activation of nociceptors that innervate the meninges--an event believed to set off migraine headache. CSD was induced in anesthetized male rats by stimulation of the visual cortex with electrical pulses, pin prick, or KCl; single-unit activity of meningeal nociceptors was monitored in vivo in the rat before and after CSD. Regardless of the method of cortical stimulation, induction of CSD was recorded in 64 trials. In 31 of those trials, CSD induced a twofold increase in meningeal nociceptor firing rate that persisted for 37.0 +/- 4.6 min in trials in which activity returned to baseline, or >68 min in trials in which activity remained heightened at the time recording was interrupted. In two-thirds of the trials, onset of long-lasting neuronal activation began approximately 14 min after the wave of CSD. The findings demonstrates for the first time that induction of CSD by focal stimulation of the rat visual cortex can lead to long-lasting activation of nociceptors that innervate the meninges. We suggest that migraine with aura is initiated by waves of CSD that lead up to delayed activation of the trigeminovascular pathway.


Subject(s)
Cortical Spreading Depression/physiology , Meninges/physiology , Nociceptors/physiology , Visual Cortex/physiology , Action Potentials , Animals , Electric Stimulation , Male , Meninges/physiopathology , Migraine with Aura/physiopathology , Neurotoxins/toxicity , Physical Stimulation , Potassium Chloride/toxicity , Rats , Rats, Sprague-Dawley , Time Factors , Visual Cortex/drug effects , Visual Cortex/physiopathology
8.
Ann Neurol ; 68(1): 81-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20582997

ABSTRACT

OBJECTIVE: Focal somatic pain can evolve into widespread hypersensitivity to nonpainful and painful skin stimuli (allodynia and hyperalgesia, respectively). We hypothesized that transformation of headache into whole-body allodynia/hyperalgesia during a migraine attack is mediated by sensitization of thalamic neurons that process converging sensory impulses from the cranial meninges and extracephalic skin. METHODS: Extracephalic allodynia was assessed using single unit recording of thalamic trigeminovascular neurons in rats and contrast analysis of blood oxygenation level-dependent (BOLD) signals registered in functional magnetic resonance imaging (fMRI) scans of patients exhibiting extracephalic allodynia. RESULTS: Sensory neurons in the rat posterior thalamus that were activated and sensitized by chemical stimulation of the cranial dura exhibited long-lasting hyperexcitability to innocuous (brush, pressure) and noxious (pinch, heat) stimulation of the paws. Innocuous, extracephalic skin stimuli that did not produce neuronal firing at baseline (eg, brush) became as effective as noxious stimuli (eg, pinch) in eliciting large bouts of neuronal firing after sensitization was established. In migraine patients, fMRI assessment of BOLD signals showed that brush and heat stimulation at the skin of the dorsum of the hand produced larger BOLD responses in the posterior thalamus of subjects undergoing a migraine attack with extracephalic allodynia than the corresponding responses registered when the same patients were free of migraine and allodynia. INTERPRETATION: We propose that the spreading of multimodal allodynia and hyperalgesia beyond the locus of migraine headache is mediated by sensitized thalamic neurons that process nociceptive information from the cranial meninges together with sensory information from the skin of the scalp, face, body, and limbs.


Subject(s)
Migraine Disorders/physiopathology , Pain/physiopathology , Thalamus/physiopathology , Action Potentials , Adolescent , Adult , Animals , Cerebrovascular Circulation , Dura Mater/physiopathology , Hot Temperature , Humans , Magnetic Resonance Imaging , Male , Microelectrodes , Middle Aged , Oxygen/blood , Physical Stimulation , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology , Thalamus/blood supply , Young Adult
9.
Neurosci Lett ; 452(1): 33-6, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19444941

ABSTRACT

Primary headaches such as migraine can be aborted by systemic administration of non-steroidal anti-inflammatory drugs (NSAIDs), potentially through the non-selective inhibition of cyclooxygenase (COX) activity in the intracranial meninges. In this study we have used single and double labeling immunohistochemistry to examine the distribution of the COX-1 and COX-2 isoforms in the intracranial dura mater of the rat and identify cell types that express them. COX-1 immunoreactivity was found in medium and small dural blood vessels and was co-expressed with the endothelial cell markers vimentin and the endothelial isoform of nitric oxide synthase (ecNOS). COX-1 was also found to be present in most dural mast cells. COX-2 was mainly expressed in ED2-positive resident dural macrophages. Constitutive COX-2 expression was also found in some axonal profiles, many of which were co-labeled with the nociceptor peptide marker CGRP. The findings suggest that NSAIDs may abort headache, at least in part, by inhibiting either neuronal or non-neuronal COX activity in the dura mater.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Dura Mater/metabolism , Membrane Proteins/metabolism , Animals , Blood Vessels/metabolism , Calcitonin Gene-Related Peptide/metabolism , Dura Mater/cytology , Ectodysplasins/metabolism , Macrophages/metabolism , Male , Nitric Oxide Synthase Type III/metabolism , Pia Mater/metabolism , Rats , Rats, Sprague-Dawley , Vimentin/metabolism
10.
Pain ; 160(9): 2161-2171, 2019 09.
Article in English | MEDLINE | ID: mdl-31033778

ABSTRACT

Migraine is a debilitating condition; however, the pharmacological effects on central nervous system networks after successful therapy are poorly understood. Defining this neurocircuitry is critical to our understanding of the disorder and for the development of antimigraine drugs. Using an established inflammatory soup model of migraine-like pathophysiology (N = 12) compared with sham synthetic interstitial fluid migraine induction (N = 12), our aim was to evaluate changes in network-level functional connectivity after sumatriptan-naproxen infusion in awake, conscious rodents (Sprague-Dawley rats). Sumatriptan-naproxen infusion functional magnetic resonance imaging data were analyzed using an independent component analysis approach. Whole-brain analysis yielded significant between-group (inflammatory soup vs synthetic interstitial fluid) alterations in functional connectivity across the cerebellar, default mode, basal ganglia, autonomic, and salience networks. These results demonstrate the large-scale antimigraine effects of sumatriptan-naproxen co-administration after dural sensitization.


Subject(s)
Brain/diagnostic imaging , Migraine Disorders/diagnostic imaging , Migraine Disorders/prevention & control , Naproxen/administration & dosage , Nerve Net/diagnostic imaging , Sumatriptan/administration & dosage , Animals , Brain/drug effects , Drug Combinations , Infusions, Intraventricular , Magnetic Resonance Imaging/methods , Male , Nerve Net/drug effects , Rats , Rats, Sprague-Dawley
11.
Brain Res ; 1660: 36-46, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28167076

ABSTRACT

Advances in our understanding of the human pain experience have shifted much of the focus of pain research from the periphery to the brain. Current hypotheses suggest that the progression of migraine depends on abnormal functioning of neurons in multiple brain regions. Accordingly, we sought to capture functional brain changes induced by the application of an inflammatory cocktail known as inflammatory soup (IS), to the dura mater across multiple brain networks. Specifically, we aimed to determine whether IS alters additional neural networks indirectly related to the primary nociceptive pathways via the spinal cord to the thalamus and cortex. IS comprises an acidic combination of bradykinin, serotonin, histamine and prostaglandin PGE2 and was introduced to basic pain research as a tool to activate and sensitize peripheral nociceptors when studying pathological pain conditions associated with allodynia and hyperalgesia. Using this model of intracranial pain, we found that dural application of IS in awake, fully conscious, rats enhanced thalamic, hypothalamic, hippocampal and somatosensory cortex responses to mechanical stimulation of the face (compared to sham synthetic interstitial fluid administration). Furthermore, resting state MRI data revealed altered functional connectivity in a number of networks previously identified in clinical chronic pain populations. These included the default mode, sensorimotor, interoceptive (Salience) and autonomic networks. The findings suggest that activation and sensitization of meningeal nociceptors by IS can enhance the extent to which the brain processes nociceptive signaling, define new level of modulation of affective and cognitive responses to pain; set new tone for hypothalamic regulation of autonomic outflow to the cranium; and change cerebellar functions.


Subject(s)
Brain/physiopathology , Migraine Disorders/physiopathology , Touch Perception/physiology , Animals , Brain/diagnostic imaging , Brain Mapping , Disease Models, Animal , Hot Temperature , Magnetic Resonance Imaging , Male , Migraine Disorders/diagnostic imaging , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology , Nociceptive Pain/diagnostic imaging , Nociceptive Pain/physiopathology , Physical Stimulation , Rats, Sprague-Dawley , Rest
12.
PLoS One ; 9(8): e103929, 2014.
Article in English | MEDLINE | ID: mdl-25090640

ABSTRACT

Dynamic thalamic regulation of sensory signals allows the cortex to adjust better to rapidly changing behavioral, physiological and environmental demands. To fulfill this role, thalamic neurons must themselves be subjected to constantly changing modulatory inputs that originate in multiple neurochemical pathways involved in autonomic, affective and cognitive functions. Our overall goal is to define an anatomical framework for conceptualizing how a 'decision' is made on whether a trigeminovascular thalamic neuron fires, for how long, and at what frequency. To begin answering this question, we determine which neuropeptides/neurotransmitters are in a position to modulate thalamic trigeminovascular neurons. Using a combination of in-vivo single-unit recording, juxtacellular labeling with tetramethylrhodamine dextran (TMR) and in-vitro immunohistochemistry, we found that thalamic trigeminovascular neurons were surrounded by high density of axons containing biomarkers of glutamate, GABA, dopamine and serotonin; moderate density of axons containing noradrenaline and histamine; low density of axons containing orexin and melanin concentrating hormone (MCH); but not axons containing CGRP, serotonin 1D receptor, oxytocin or vasopressin. In the context of migraine, the findings suggest that the transmission of headache-related nociceptive signals from the thalamus to the cortex may be modulated by opposing forces (i.e., facilitatory, inhibitory) that are governed by continuous adjustments needed to keep physiological, behavioral, cognitive and emotional homeostasis.


Subject(s)
Anxiety/physiopathology , Migraine Disorders/physiopathology , Neurons/pathology , Neurotransmitter Agents/metabolism , Sleep , Stress, Psychological/physiopathology , Thalamus/physiopathology , Trigeminal Nerve/physiopathology , Animals , Anxiety/psychology , Biomarkers/metabolism , Brain Stem/physiopathology , Calcitonin Gene-Related Peptide/metabolism , Dopamine/metabolism , Eating , Glutamates/metabolism , Histamine/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Melanins/metabolism , Migraine Disorders/psychology , Neuropeptides/metabolism , Norepinephrine/metabolism , Orexins , Oxytocin/metabolism , Pituitary Hormones/metabolism , Rats, Sprague-Dawley , Serotonin/metabolism , Trigeminal Nerve/blood supply , Vasopressins/metabolism
13.
Pain ; 152(1): 140-149, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21036476

ABSTRACT

The proinflammatory cytokine TNF-α has been shown to promote activation and sensitization of primary afferent nociceptors. The downstream signaling processes that play a role in promoting this neuronal response remain however controversial. Increased TNF-α plasma levels during migraine attacks suggest that local interaction between this cytokine and intracranial meningeal nociceptors plays a role in promoting the headache. Here, using in vivo single unit recording in the trigeminal ganglia of anesthetized rats, we show that meningeal TNF-α action promotes a delayed mechanical sensitization of meningeal nociceptors. Using immunohistochemistry, we provide evidence for non-neuronal localization of the TNF receptors TNFR1 to dural endothelial vascular cells and TNFR2 to dural resident macrophages as well as to some CGRP-expressing dural nerve fibers. We also demonstrate that meningeal vascular TNFR1 is co-localized with COX-1 while the perivascular TNFR2 is co-expressed with COX-2. We further report here for the first time that TNF-α evoked sensitization of meningeal nociceptors is dependent upon local action of cyclooxygenase (COX). Finally, we show that local application of TNF-α to the meninges evokes activation of the p38 MAP kinase in dural blood vessels that also express TNFR1 and that pharmacological blockade of p38 activation inhibits TNF-α evoked sensitization of meningeal nociceptors. Our study suggests that meningeal action of TNF-α could play an important role in the genesis of intracranial throbbing headaches such as migraine through a mechanism that involves at least part activation of non-neuronal TNFR1 and TNFR2 and downstream activation of meningeal non-neuronal COX and the p38 MAP kinase.


Subject(s)
Cyclooxygenase 1/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Membrane Proteins/metabolism , Sensory Receptor Cells/physiology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Action Potentials/drug effects , Animals , Disease Models, Animal , Dura Mater/cytology , Dura Mater/metabolism , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Male , Physical Stimulation/adverse effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sensory Receptor Cells/drug effects , Time Factors , Trigeminal Ganglion/cytology
14.
Nat Neurosci ; 13(2): 239-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20062053

ABSTRACT

The perception of migraine headache, which is mediated by nociceptive signals transmitted from the cranial dura mater to the brain, is uniquely exacerbated by exposure to light. We found that exacerbation of migraine headache by light is prevalent among blind individuals who maintain non-image-forming photoregulation in the face of massive rod/cone degeneration. Using single-unit recording and neural tract tracing in the rat, we identified dura-sensitive neurons in the posterior thalamus whose activity was distinctly modulated by light and whose axons projected extensively across layers I-V of somatosensory, visual and associative cortices. The cell bodies and dendrites of such dura/light-sensitive neurons were apposed by axons originating from retinal ganglion cells (RGCs), predominantly from intrinsically photosensitive RGCs, the principle conduit of non-image-forming photoregulation. We propose that photoregulation of migraine headache is exerted by a non-image-forming retinal pathway that modulates the activity of dura-sensitive thalamocortical neurons.


Subject(s)
Light/adverse effects , Migraine Disorders/etiology , Migraine Disorders/physiopathology , Neurons/physiology , Retina/physiopathology , Thalamus/physiopathology , Adult , Animals , Axons/physiology , Blindness/epidemiology , Blindness/physiopathology , Cerebral Cortex/physiopathology , Dendrites/physiology , Dura Mater/physiopathology , Female , Humans , Incidence , Male , Migraine Disorders/epidemiology , Neural Pathways/physiopathology , Prevalence , Rats , Retinal Ganglion Cells/physiology
15.
J Comp Neurol ; 515(3): 331-48, 2009 Jul 20.
Article in English | MEDLINE | ID: mdl-19425099

ABSTRACT

Migraine sufferers frequently testify that their headache feels as if the calvarial bones are deformed, crushed, or broken (Jakubowski et al. [2006] Pain 125:286-295). This has lead us to postulate that the calvarial bones are supplied by sensory fibers. We studied sensory innervation of the calvaria in coronal and horizontal sections of whole-head preparations of postnatal and adult mice, via immunostaining of peripherin (a marker of thinly myelinated and unmyelinated fibers) or calcitonin gene-related peptide (CGRP; a marker more typical of unmyelinated nerve fibers). In pups, we observed nerve bundles coursing between the galea aponeurotica and the periosteum, between the periosteum and the bone, and between the bone and the meninges; as well as fibers that run inside the diploë in different orientations. Some dural fibers issued collateral branches to the pia at the frontal part of the brain. In the adult calvaria, the highest concentration of peripherin- and CGRP-labeled fibers was found in sutures, where they appeared to emerge from the dura. Labeled fibers were also observed in emissary canals, bone marrow, and periosteum. In contrast to the case in pups, no labeled fibers were found in the diploë of the adult calvaria. Meningeal nerves that infiltrate the periosteum through the calvarial sutures may be positioned to mediate migraine headache triggered by pathophysiology of extracranial tissues, such as muscle tenderness and mild trauma to the skull. In view of the concentration of sensory fibers in the sutures, it may be useful to avoid drilling the sutures in patients undergoing craniotomies for a variety of neurosurgical procedures.


Subject(s)
Afferent Pathways/cytology , Sensory Receptor Cells/cytology , Skull/innervation , Animals , Biomarkers/metabolism , Calcitonin Gene-Related Peptide/metabolism , Female , Humans , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Pain/physiopathology , Peripherins
16.
Pain ; 130(1-2): 166-76, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17459586

ABSTRACT

Intracranial headaches such as that of migraine are generally accepted to be mediated by prolonged activation of meningeal nociceptors but the mechanisms responsible for such nociceptor activation are poorly understood. In this study, we examined the hypothesis that meningeal nociceptors can be activated locally through a neuroimmune interaction with resident mast cells, granulated immune cells that densely populate the dura mater. Using in vivo electrophysiological single unit recording of meningeal nociceptors in the rat we observed that degranulation of dural mast cells using intraperitoneal administration of the basic secretagogue agent compound 48/80 (2 mg/kg) induced a prolonged state of excitation in meningeal nociceptors. Such activation was accompanied by increased expression of the phosphorylated form of the extracellular signal-regulated kinase (pERK), an anatomical marker for nociceptor activation. Mast cell-induced nociceptor interaction was also associated with downstream activation of the spinal trigeminal nucleus as indicated by an increase in c-fos expression. Our findings provide evidence linking dural mast cell degranulation to prolonged activation of the trigeminal pain pathway believed to underlie intracranial headaches such as that of migraine.


Subject(s)
Cell Degranulation/immunology , Mast Cells/immunology , Migraine Disorders/immunology , Nociceptors/immunology , Afferent Pathways/immunology , Afferent Pathways/metabolism , Animals , Cell Degranulation/drug effects , Dura Mater/immunology , Dura Mater/metabolism , Electrophysiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mast Cells/drug effects , Migraine Disorders/metabolism , Nociceptors/enzymology , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Trigeminal Nucleus, Spinal/cytology , Trigeminal Nucleus, Spinal/immunology , Trigeminal Nucleus, Spinal/metabolism , p-Methoxy-N-methylphenethylamine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL