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










Publication year range
1.
J Headache Pain ; 24(1): 165, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38062355

ABSTRACT

BACKGROUND: Migraine is a disabling neurological disorder, characterized by recurrent headaches. During migraine attacks, individuals often experience sensory symptoms such as cutaneous allodynia which indicates the presence of central sensitization. This sensitization is prevented by oral administration of propranolol, a common first-line medication for migraine prophylaxis, that also normalized the activation of the locus coeruleus (LC), considered as the main origin of descending noradrenergic pain controls. We hypothesized that the basal modulation of trigeminal sensory processing by the locus coeruleus is shifted towards more facilitation in migraineurs and that prophylactic action of propranolol may be attributed to a direct action in LC through beta-adrenergic receptors. METHODS: We used simultaneous in vivo extracellular recordings from the trigeminocervical complex (TCC) and LC of male Sprague-Dawley rats to characterize the relationship between these two areas following repeated meningeal inflammatory soup infusions. Von Frey Hairs and air-puff were used to test periorbital mechanical allodynia. RNAscope and patch-clamp recordings allowed us to examine the action mechanism of propranolol. RESULTS: We found a strong synchronization between TCC and LC spontaneous activities, with a precession of the LC, suggesting the LC drives TCC excitability. Following repeated dural-evoked trigeminal activations, we observed a disruption in coupling of activity within LC and TCC. This suggested an involvement of the two regions' interactions in the development of sensitization. Furthermore, we showed the co-expression of alpha-2A and beta-2 adrenergic receptors within LC neurons. Finally propranolol microinjections into the LC prevented trigeminal sensitization by desynchronizing and decreasing LC neuronal activity. CONCLUSIONS: Altogether these results suggest that trigemino-coerulean coupling plays a pivotal role in migraine progression, and that propranolol's prophylactic effects involve, to some extent, the modulation of LC activity through beta-2 adrenergic receptors. This insight reveals new mechanistic aspects of LC control over sensory processing.


Subject(s)
Migraine Disorders , Propranolol , Rats , Animals , Male , Propranolol/pharmacology , Propranolol/therapeutic use , Rats, Sprague-Dawley , Locus Coeruleus , Receptors, Adrenergic, beta-2/therapeutic use , Migraine Disorders/prevention & control , Migraine Disorders/drug therapy , Hyperalgesia/drug therapy
2.
Neuroscience ; 426: 129-140, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31846750

ABSTRACT

Hyperacusis, an exaggerated, sometimes painful perception of loudness even for soft sounds, is a poorly understood distressing condition. While the involvement of modified gain of central auditory neurons and the influence of nonauditory brain regions are well-documented, the issue of where in the auditory system these abnormalities arise remains open, particularly when hyperacusis comes without sensorineural hearing loss. Here we used acute intraperitoneal administration of sodium salicylate (150 mg/kg) in rats, enough to produce > 10-dB decrease in acoustic startle threshold with mild hearing loss at low frequencies (<10 kHz). Anesthesia, necessary for middle-ear-reflex (MEMR) threshold measurements, abolished the olivocochlear efferent reflex but not the MEMR acting on frequencies < 10 kHz, and its mean threshold increased from 55 dB SPL in controls to 80 dB SPL in salicylate-treated animals 60-90 minutes after injection, with an about 3-dB increase in acoustic energy reaching the cochlea. The mean latencies of auditory brainstem-evoked responses (ABR) conspicuously decreased after salicylate, by 0.25 millisecond at 6 kHz at every level, a frequency-dependent effect absent above 12 kHz. A generic model of loudness based upon cross-frequency coincidence detection predicts that with such timing changes, a transient sound may seem as loud at <40 dB SPL as it does in controls at >60 dB SPL. Candidate circuits able to act at the same time on the startle reflex, the MEMR and ABRs may be serotoninergic, as salicylate is known to increase brain serotonin and 5-HT neurons participate in MEMR and ABR circuits.


Subject(s)
Evoked Potentials, Auditory, Brain Stem/drug effects , Hyperacusis/physiopathology , Animals , Auditory Cortex/drug effects , Auditory Threshold/drug effects , Hearing/drug effects , Hyperacusis/chemically induced , Inferior Colliculi/drug effects , Male , Rats, Sprague-Dawley , Reflex, Startle/physiology , Sodium Salicylate/pharmacology
3.
J Neurosci ; 38(49): 10489-10504, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30355630

ABSTRACT

Mechanical allodynia, a widespread pain symptom that still lacks effective therapy, is associated with the activation of a dorsally directed polysynaptic circuit within the spinal dorsal horn (SDH) or medullary dorsal horn (MDH), whereby tactile inputs into deep SDH/MDH can gain access to superficial SDH/MDH, eliciting pain. Inner lamina II (IIi) interneurons expressing the γ isoform of protein kinase C (PKCγ+) are key elements for allodynia circuits, but how they operate is still unclear. Combining behavioral, ex vivo electrophysiological, and morphological approaches in an adult rat model of facial inflammatory pain (complete Freund's adjuvant, CFA), we show that the mechanical allodynia observed 1 h after CFA injection is associated with the following (1) sensitization (using ERK1/2 phosphorylation as a marker) and (2) reduced dendritic arborizations and enhanced spine density in exclusively PKCγ+ interneurons, but (3) depolarized resting membrane potential (RMP) in all lamina IIi PKCγ+/PKCγ- interneurons. Blocking MDH 5HT2A receptors (5-HT2AR) prevents facial mechanical allodynia and associated changes in the morphology of PKCγ+ interneurons, but not depolarized RMP in lamina IIi interneurons. Finally, activation of MDH 5-HT2AR in naive animals is enough to reproduce the behavioral allodynia and morphological changes in PKCγ+ interneurons, but not the electrophysiological changes in lamina IIi interneurons, induced by facial inflammation. This suggests that inflammation-induced mechanical allodynia involves strong morphological reorganization of PKCγ+ interneurons via 5-HT2AR activation that contributes to open the gate for transmission of innocuous mechanical inputs to superficial SDH/MDH pain circuitry. Preventing 5-HT2AR-induced structural plasticity in PKCγ+ interneurons might represent new avenues for the specific treatment of inflammation-induced mechanical hypersensitivity.SIGNIFICANCE STATEMENT Inflammatory or neuropathic pain syndromes are characterized by pain hypersensitivity such as mechanical allodynia (pain induced by innocuous mechanical stimuli). It is generally assumed that mechanisms underlying mechanical allodynia, because they are rapid, must operate at only the level of functional reorganization of spinal or medullary dorsal horn (MDH) circuits. We discovered that facial inflammation-induced mechanical allodynia is associated with rapid and strong structural remodeling of specifically interneurons expressing the γ isoform of protein kinase C (PKCγ) within MDH inner lamina II. Moreover, we elucidated a 5-HT2A receptor to PKCγ/ERK1/2 pathway leading to the behavioral allodynia and correlated morphological changes in PKCγ interneurons. Therefore, descending 5-HT sensitize PKCγ interneurons, a putative "gate" in allodynia circuits, via 5-HT2A receptor-induced structural reorganization.


Subject(s)
Gene Expression Regulation, Enzymologic , Hyperalgesia/metabolism , Interneurons/metabolism , Protein Kinase C/biosynthesis , Receptor, Serotonin, 5-HT2A/metabolism , Touch/physiology , Animals , Facial Pain/metabolism , Facial Pain/pathology , Hyperalgesia/genetics , Hyperalgesia/pathology , Inflammation/metabolism , Inflammation/pathology , Interneurons/pathology , Male , Protein Kinase C/genetics , Rats , Rats, Sprague-Dawley
4.
Pain ; 158(10): 2025-2034, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28700539

ABSTRACT

Migraine is currently conceptualized as a chronic disease with episodic manifestations. In some patients, migraine attack frequency increases, leading to chronic migraine. Daily preventive therapy is initiated to decrease attack frequency. Propranolol, a first-line medication for migraine prophylaxis, reduces attack frequency in nearly 50% of patients receiving it. However, the mechanisms of its antimigraine action are unclear. We examined the effect of daily propranolol treatment (10 mg·kg per os, 8 days) in a rat model of recurrent activation of dural nociceptors (repeated infusion of an inflammatory soup (IS) on the dura through a cannula every 2-3 days). Propranolol does not abort IS-induced acute cephalic mechanical allodynia but blocks the development of a chronic cutaneous hypersensitivity upon repeated IS injections. Furthermore, propranolol prevents (1) the elevated touch-evoked Fos expression within the trigeminocervical complex, (2) enhanced both spontaneous activity, and evoked responses of second-order trigeminovascular neurons, (3) elevated touch-evoked rostral ventromedial medulla and locus coeruleus Fos expression and (4) diffuse noxious inhibitory controls impairment, induced by repeated IS injections. Our results suggest that propranolol exerts its prophylactic action, at least in part, by blocking the chronic sensitization of descending controls of pain, arising from the rostral ventromedial medulla and locus coeruleus, and in turn preventing the maintenance of a state of facilitated trigeminovascular transmission within the trigeminocervical complex. Assessing changes in these brain areas has the potential to elucidate the mechanisms for migraine transformation and to reveal novel biological and molecular targets for specific migraine-preventive therapies.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Central Nervous System Sensitization/drug effects , Dura Mater/physiology , Propranolol/pharmacology , Afferent Pathways/drug effects , Afferent Pathways/physiopathology , Animals , Chloral Hydrate/pharmacology , Electric Stimulation/adverse effects , Face/innervation , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Hypnotics and Sedatives/pharmacology , Male , Neurons/drug effects , Nociceptors/drug effects , Nociceptors/physiology , Oncogene Proteins v-fos/metabolism , Patch-Clamp Techniques , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Trigeminal Nucleus, Spinal/metabolism , Trigeminal Nucleus, Spinal/pathology
5.
Dev Neurobiol ; 77(1): 102-119, 2017 01.
Article in English | MEDLINE | ID: mdl-27346325

ABSTRACT

Protein kinase C gamma (PKCγ) interneurons, located in the superficial spinal (SDH) and medullary dorsal horns (MDH), have been shown to play a critical role in cutaneous mechanical hypersensitivity. However, a thorough characterization of their development in the MDH is lacking. Here, it is shown that the number of PKCγ-ir interneurons changes from postnatal day 3 (P3) to P60 (adult) and such developmental changes differ according to laminae. PKCγ-ir interneurons are already present at P3-5 in laminae I, IIo, and III. In lamina III, they then decrease from P11-P15 to P60. Interestingly, PKCγ-ir interneurons appear only at P6 in lamina IIi, and they conversely increase to reach adult levels at P11-15. Analysis of neurogenesis using bromodeoxyuridine (BrdU) does not detect any PKCγ-BrdU double-labeling in lamina IIi. Quantification of the neuronal marker, NeuN, reveals a sharp neuronal decline (∼50%) within all superficial MDH laminae during early development (P3-15), suggesting that developmental changes in PKCγ-ir interneurons are independent from those of other neurons. Finally, neonatal capsaicin treatment, which produces a permanent loss of most unmyelinated afferent fibers, has no effect on the development of PKCγ-ir interneurons. Together, the results show that: (i) the expression of PKCγ-ir interneurons in MDH is developmentally regulated with a critical period at P11-P15, (ii) PKCγ-ir interneurons are developmentally heterogeneous, (iii) lamina IIi PKCγ-ir interneurons appear less vulnerable to cell death, and (iv) postnatal maturation of PKCγ-ir interneurons is due to neither neurogenesis, nor neuronal migration, and is independent of C-fiber development. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 102-119, 2017.


Subject(s)
Interneurons/physiology , Medulla Oblongata/physiology , Protein Kinase C/metabolism , Spinal Cord Dorsal Horn/physiology , Age Factors , Animals , Animals, Newborn , Female , Interneurons/metabolism , Male , Medulla Oblongata/growth & development , Medulla Oblongata/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/growth & development , Spinal Cord Dorsal Horn/metabolism
6.
Pain ; 156(4): 644-655, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25790455

ABSTRACT

Descending pain-modulatory systems, either inhibitory or facilitatory, play a critical role in both acute and chronic pain. Compared with serotonin and norepinephrine, little is known about the function of dopamine (DA). We characterized the anatomical organization of descending DA pathways from hypothalamic A11 nuclei to the medullary dorsal horn (MDH) and investigated their role in trigeminal pain. Immunochemistry analysis reveals that A11 is a heterogeneous nucleus that contains at least 3 neuronal phenotypes, DA, GABA, and alpha-calcitonin gene-related peptide (α-CGRP) neurons, exhibiting different distribution patterns, with a large proportion of GABA relative to DA neurons. Using fluorogold, we show that descending pathways from A11 nuclei to MDH originate mainly from DA neurons and are bilateral. Facial nociceptive stimulation elevates Fos immunoreactivity in both ipsilateral and contralateral A11 nuclei. Fos immunoreactivity is not detected in DA or projecting neurons but, interestingly, in GABA neurons. Finally, inactivating A11, using muscimol, or partially lesioning A11 DA neurons, using the neurotoxin 6-hydroxydopamine, inhibits trigeminal pain behavior. These results show that A11 nuclei are involved in pain processing. Interestingly, however, pain seems to activate GABAergic neurons within A11 nuclei, which suggests that pain inhibits rather than activates descending DA controls. We show that such inhibition produces an antinociceptive effect. Pain-induced inhibition of descending DA controls and the resulting reduced DA concentration within the dorsal horn may inhibit the transfer of nociceptive information to higher brain centers through preferential activation of dorsal horn D2-like receptors.


Subject(s)
Dopamine/metabolism , Dorsomedial Hypothalamic Nucleus/cytology , Dorsomedial Hypothalamic Nucleus/physiology , Neurons/physiology , Trigeminal Neuralgia/therapy , gamma-Aminobutyric Acid/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Denervation , Functional Laterality , GABA-A Receptor Agonists/pharmacology , Male , Motor Activity , Muscimol/pharmacology , Neural Pathways/physiology , Neurons/classification , Neurons/drug effects , Oxidopamine/toxicity , Pain Measurement , Rats , Sympatholytics/toxicity , Trigeminal Neuralgia/physiopathology
7.
Pain ; 155(7): 1196-1205, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24631586

ABSTRACT

Migraine is a chronic disease with episodic manifestations. In a subgroup, attack frequency increases over time, leading to chronic migraine. One of the most important risk factors for migraine progression is frequency of headache attacks at baseline. Unfortunately, the actual effects of repeated activation of dural nociceptors are poorly known. We investigated the behavioral, anatomical, and electrophysiological changes induced by repeated low- and high-intensity stimulation of meningeal nociceptor by injecting an inflammatory soup in rats. Single high-intensity, but not low-intensity, stimulation produces a reversible cephalic allodynia. Upon repetition, however, low-intensity stimulation, too, induces a reversible cephalic allodynia, and high-intensity, reversible cephalic and extracephalic allodynia. Moreover, cephalic allodynia becomes, in part, persistent upon repeated high-intensity stimulation. Fos expression reveals that a single high-intensity stimulation already leads to widespread, trigeminal, and spinal central sensitization, and that such general central sensitization potentiates upon repetition. Trigeminovascular nociceptive neurons become persistently sensitized and their diffuse noxious inhibitory controls (DNIC) concomitantly impaired. Thus, compared with single stimulation, repeated dural nociceptor activation specifically leads to: 1) a gradual worsening of cutaneous hypersensitivity and general neuronal hyperexcitability and 2) spreading of cutaneous hypersensitivity superimposed on 3) persistent cephalic cutaneous hypersensitivity and trigeminal central sensitization. Such repetition-induced development of central sensitization and its consequence, cutaneous allodynia, may arise from both the general neuronal hyperexcitability that results from DNIC impairment and hyperexcitability that likely develops in trigeminal nociceptive neurons in response to their repetitive activation. These neuronal changes may in turn elevate the risk for developing chronic migraine.


Subject(s)
Central Nervous System Sensitization/physiology , Hyperalgesia/physiopathology , Migraine Disorders , Neural Inhibition/physiology , Nociceptive Pain/physiopathology , Nociceptors/physiology , Skin/innervation , Trigeminal Nerve/physiopathology , Animals , Disease Models, Animal , Disease Progression , Dura Mater , Male , Rats , Rats, Sprague-Dawley
9.
PLoS One ; 8(8): e73022, 2013.
Article in English | MEDLINE | ID: mdl-23951340

ABSTRACT

Several lines of evidence suggest that the hypothalamus is involved in trigeminal pain processing. However, the organization of descending hypothalamic projections to the spinal trigeminal nucleus caudalis (Sp5C) remains poorly understood. Microinjections of the retrograde tracer, fluorogold (FG), into the Sp5C, in rats, reveal that five hypothalamic nuclei project to the Sp5C: the paraventricular nucleus, the lateral hypothalamic area, the perifornical hypothalamic area, the A11 nucleus and the retrochiasmatic area. Descending hypothalamic projections to the Sp5C are bilateral, except those from the paraventricular nucleus which exhibit a clear ipsilateral predominance. Moreover, the density of retrogradely FG-labeled neurons in the hypothalamus varies according to the dorso-ventral localization of the Sp5C injection site. There are much more labeled neurons after injections into the ventrolateral part of the Sp5C (where ophthalmic afferents project) than after injections into its dorsomedial or intermediate parts (where mandibular and maxillary afferents, respectively, project). These results demonstrate that the organization of descending hypothalamic projections to the spinal dorsal horn and Sp5C are different. Whereas the former are ipsilateral, the latter are bilateral. Moreover, hypothalamic projections to the Sp5C display somatotopy, suggesting that these projections are preferentially involved in the processing of meningeal and cutaneous inputs from the ophthalmic branch of the trigeminal nerve in rats. Therefore, our results suggest that the control of trigeminal and spinal dorsal horn processing of nociceptive information by hypothalamic neurons is different and raise the question of the role of bilateral, rather than unilateral, hypothalamic control.


Subject(s)
Hypothalamus/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Trigeminal Caudal Nucleus/metabolism , Trigeminal Nucleus, Spinal/metabolism , Animals , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Hypothalamus/anatomy & histology , Hypothalamus/cytology , Immunohistochemistry , Male , Microinjections , Models, Anatomic , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/anatomy & histology , Paraventricular Hypothalamic Nucleus/cytology , Rats , Rats, Sprague-Dawley , Stilbamidines/administration & dosage , Stilbamidines/pharmacokinetics , Trigeminal Caudal Nucleus/anatomy & histology , Trigeminal Caudal Nucleus/cytology , Trigeminal Nucleus, Spinal/anatomy & histology , Trigeminal Nucleus, Spinal/cytology
10.
Pain ; 152(8): 1821-1831, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21514054

ABSTRACT

The transfer of nociceptive information at the level of dorsal horn is subject to extensive processing by both local segmental and supraspinal mechanisms, including descending dopaminergic controls, originating from the hypothalamic A11 nucleus. The inhibitory role of dopamine on evoked pain via activation of D2-like receptors at the level of the dorsal horn is well established. Here, by use of behavioral, electrophysiological, and anatomical techniques, we examined within the trigeminal sensory complex, first, whether descending dopaminergic controls also modulate pain behavior after an inflammatory insult, and second, under which physiological conditions these descending dopaminergic controls are actually recruited. We show that D2 receptors are mostly located within superficial medullary dorsal horn where trigeminal nociceptive fibers abut. Activating these D2-like receptors inhibits, whereas blocking them enhances, both formalin- and capsaicin-evoked pain behavior and C-fiber-evoked action potential firing of trigeminal wide dynamic range (WDR) neurons. Moreover, windup and diffuse noxious inhibitory controls (DNIC), 2 dynamic properties of C-fiber-evoked firing of WDR neurons, are inhibited by activating and blocking, respectively, these D2-like receptors. Altogether, our results are consistent with a tonic inhibition of the trigeminal nociceptive input by descending dopaminergic controls via activation of D2-like receptors at the level of superficial medullary dorsal horn. Such dopamine-dependent tonic inhibition of nociceptive information can be dynamically modulated by pain. This suggests that dysregulation of descending dopaminergic controls should translate in patients into diffuse, cephalic, and extracephalic pain symptoms--spontaneous pain, decreased pain thresholds, deficient DNIC, or some combination of these.


Subject(s)
Dopamine/metabolism , Medulla Oblongata/pathology , Pain/pathology , Posterior Horn Cells/physiopathology , Action Potentials/drug effects , Action Potentials/physiology , Afferent Pathways/physiology , Analysis of Variance , Animals , Capsaicin/adverse effects , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Electric Stimulation/methods , Male , Microinjections , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Unmyelinated/physiology , Pain/chemically induced , Pain Measurement , Protein Kinase C/metabolism , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism , Spinal Cord/pathology , Sulpiride/pharmacology
11.
Eur J Pain ; 15(7): 676-82, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21316272

ABSTRACT

Trigeminal neuropathic pain is due to lesion or dysfunction of the nervous system. Dynamic mechanical allodynia is a widespread symptom of neuropathic pain for which mechanisms are still poorly understood. Recent studies demonstrate that forebrain neurons, including neurons in the medial prefrontal cortex (mPFC) are important for the perception of acute and chronic pain. Using the phosphorylation of the extracellular-signal regulated kinase (pERK-1/2) as an anatomical marker of neuronal activation, the present study investigated how dynamic mechanical allodynia is processed in the rat ventral mPFC (prelimbic and infralimbic cortex) after chronic constriction injury to the infraorbital nerve (IoN-CCI). Two weeks after unilateral IoN-CCI, rats showed a dramatic bilateral trigeminal dynamic mechanical allodynia. Light, moving stroking of the infraorbital skin resulted in strong, bilateral upregulation of pERK-1/2 in the ventral mPFC of IoN-CCI animals. pERK-1/2 was located in neuronal cells only. Stimulus-evoked pERK-1/2 immunopositive cell bodies displayed a rostrocaudal gradient and layer-selective distribution in the ventral mPFC, being predominant in the rostral ventral mPFC and in layers II-III and V-VI of the ventral mPFC. In layers II-III, intense pERK-1/2 also extended into distal dendrites, up to layer I. These results demonstrate that trigeminal nerve injury induces a significant alteration in the ventral mPFC processing of tactile stimuli and suggest that ERK phosphorylation contributes to the mechanisms underlying abnormal pain perception under this condition.


Subject(s)
Hyperalgesia/physiopathology , Neuralgia/physiopathology , Neurons/physiology , Prefrontal Cortex/physiopathology , Trigeminal Nerve Diseases/physiopathology , Animals , Behavior, Animal/physiology , Hyperalgesia/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuralgia/metabolism , Pain Measurement , Pain Threshold/physiology , Phosphorylation , Physical Stimulation , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Trigeminal Nerve Diseases/metabolism
12.
Pain ; 142(3): 245-254, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19231081

ABSTRACT

Diffuse noxious inhibitory controls (DNIC) are very powerful long-lasting descending inhibitory controls, which are pivotal in modulating the activity of spinal and trigeminal nociceptive neurons. The principal feature of DNIC is that they are subserved by a loop that involves supraspinal structures that have not yet been identified. Using behavioral, in vivo extracellular electrophysiological and anatomical approaches, we studied the neuronal network underlying DNIC. Using a new behavioral model of DNIC, in which facial grooming produced by formalin injection into the vibrissa pad is inhibited by a conditioning noxious stimulation, formalin injection into the hindpaw, we show that blockade of NK1 receptors in the lumbar spinal cord - by intrathecal administration of the NK1 receptor antagonist, RP67580 - largely attenuates DNIC-induced facial analgesia. In a second series of experiments, WDR neurons were recorded from the trigeminal subnucleus oralis and inhibited their C-fiber-evoked responses by the conditioning noxious heat stimulation of the hindpaw. We show that inactivating the lateral parabrachial area - by microinjecting the GABA(A) agonist, muscimol - strongly attenuates DNIC-induced inhibition of C-fiber-evoked responses. Finally, our neuroanatomical tracing study demonstrates that the descending pathway for DNIC does not involve direct descending projections from the PB area. We conclude that (1) lamina I/III spinoparabrachial neurons that express the NK1 receptor and (2) parabrachial neurons are involved in the ascending part of the loop underlying DNIC and that the descending pathway for DNIC might include indirect projections to the spinal or medullary dorsal horn.


Subject(s)
Afferent Pathways/physiopathology , Nerve Net/physiopathology , Neural Inhibition , Pain/physiopathology , Pons/physiopathology , Receptors, Neurokinin-1/metabolism , Sensory Receptor Cells , Spinal Cord/physiopathology , Animals , Feedback , Male , Rats , Rats, Sprague-Dawley
13.
J Neurosci ; 26(33): 8441-50, 2006 Aug 16.
Article in English | MEDLINE | ID: mdl-16914669

ABSTRACT

Sensory maps for pain can be modified by deafferentation or injury, and such plasticity has been attributed mainly to changes in the convergence of projections in "bottom-up" mechanisms. We addressed the possible contribution of "top-down" mechanisms by investigating the functional significance of corticofugal influences from the primary somatosensory cortex (S1) to the ventroposterolateral thalamic nucleus (VPL). The strong convergence of spinal and lemniscal afferents to the VPL and the close correspondence between afferents and efferents within the VPL-S1 network suggest the existence of functionally related thalamocortical circuits that are implicated in the detection of innocuous and noxious inputs. Functional characterization of single nociceptive, wide dynamic range, and non-nociceptive VPL neurons and labeling the axons and terminal fields with the juxtacellular technique showed that all three types of cells project to a restricted area, within S1. The convergence of the terminal trees of axons from VPL neurons activated by innocuous, noxious, or both inputs suggests that their inputs are not segregated into anatomically distinct regions. Microinjections within S1 were performed for pharmacological manipulation of corticofugal modulation. Glutamatergic activation of corticofugal output enhanced noxious-evoked responses and affected in a biphasic way tactile-evoked responses of VPL cells. GABA(A)-mediated depression of corticofugal output concomitantly depressed noxious and enhanced innocuous-evoked responses of VPL neurons. Microinjections of a GABA(A) antagonist on corticofugal cells enhanced noxious-evoked responses of VPL cells. Our findings demonstrate that corticofugal influences from S1 contribute to selectively modulate somatosensory submodalities at the thalamic level.


Subject(s)
Pain/physiopathology , Somatosensory Cortex/physiopathology , Spinothalamic Tracts/physiopathology , Animals , Bicuculline/administration & dosage , Bicuculline/pharmacology , GABA Agonists/administration & dosage , GABA Agonists/pharmacology , GABA Antagonists/administration & dosage , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Hindlimb , Homocysteine/administration & dosage , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Hot Temperature , Male , Microinjections , Muscimol/administration & dosage , Muscimol/pharmacology , Neural Pathways/physiopathology , Nociceptors/physiopathology , Physical Stimulation , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Thalamic Nuclei/physiopathology
15.
Eur J Neurosci ; 21(12): 3395-402, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16026477

ABSTRACT

Neurons within the lateral ventromedial thalamic nucleus (VMl) convey selectively nociceptive information from all parts of the body. The present experiments were performed in rats and were designed to determine the organization of cortical projections from VMl neurons. In a first series of experiments, these cells were characterized electrophysiologically and individually labelled in a Golgi-like manner following juxtacellular electrophoresis of biotin-dextran. In a second experimental series, topical applications of the tracers fluorogold and tetramethylrhodamine-labelled dextran were placed into both the rostral-most and caudal areas of layer I of the dorsolateral frontal cortex, respectively. All VMl nociceptive neurons were fusiform and their full dendritic arborizations were bipolar, extending in the lateromedial axis. VMl cells are thus particularly well located to receive widespread nociceptive inputs via a brainstem link, viz. the medullary subnucleus reticularis dorsalis. VMl neurons driven by 'whole body' nociceptive receptive fields project to the rostral part of the layer I of the dorsolateral frontal cortex. These projections are widespread because double-labelling data showed a great number of VMl neurons labelled from both rostral and caudal dorsolateral cortices. The VMl comprises a homogeneous, organized subset of thalamic neurons that allow any signals of pain to modify cortical activity in a widespread manner, by interacting with the entire layer I of the dorsolateral neocortex.


Subject(s)
Afferent Pathways/anatomy & histology , Frontal Lobe/anatomy & histology , Hindlimb/innervation , Lateral Thalamic Nuclei/cytology , Neurons/physiology , Nociceptors/physiopathology , Action Potentials/physiology , Afferent Pathways/physiopathology , Animals , Brain Mapping , Electric Stimulation/methods , Frontal Lobe/physiopathology , Functional Laterality , Heterocyclic Compounds, 3-Ring/metabolism , Lateral Thalamic Nuclei/physiopathology , Male , Microscopy, Confocal/methods , Neurons/radiation effects , Rats , Rats, Sprague-Dawley , Rhodamines , Silver Staining/methods , Stilbamidines/metabolism
16.
Nat Neurosci ; 8(1): 67-71, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15592464

ABSTRACT

Oscillatory spike timing in the hippocampus is regarded as a temporal coding mechanism for space, but the underlying mechanisms are poorly understood. To contrast the predictions of the different models of phase precession, we transiently turned off neuronal discharges for up to 250 ms and reset the phase of theta oscillations by stimulating the commissural pathway in rats. After recovery from silence, phase precession continued. The phase of spikes for the first theta cycle after the perturbation was more advanced than the phase of spikes for the last theta cycle just before the perturbation. These findings indicate that phase advancement that emerges within hippocampal circuitry may be updated at the beginning of each theta cycle by extrahippocampal inputs.


Subject(s)
Hippocampus/physiology , Space Perception/physiology , Action Potentials , Animals , Electric Stimulation , Male , Maze Learning/physiology , Neural Pathways/physiology , Neurons/physiology , Rats , Rats, Long-Evans , Reaction Time , Theta Rhythm
17.
J Neurophysiol ; 92(1): 600-8, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15056678

ABSTRACT

Most neuronal interactions in the cortex occur within local circuits. Because principal cells and GABAergic interneurons contribute differently to cortical operations, their experimental identification and separation is of utmost important. We used 64-site two-dimensional silicon probes for high-density recording of local neurons in layer 5 of the somatosensory and prefrontal cortices of the rat. Multiple-site monitoring of units allowed for the determination of their two-dimensional spatial position in the brain. Of the approximately 60,000 cell pairs recorded, 0.2% showed robust short-term interactions. Units with significant, short-latency (<3 ms) peaks following their action potentials in their cross-correlograms were characterized as putative excitatory (pyramidal) cells. Units with significant suppression of spiking of their partners were regarded as putative GABAergic interneurons. A portion of the putative interneurons was reciprocally connected with pyramidal cells. Neurons physiologically identified as inhibitory and excitatory cells were used as templates for classification of all recorded neurons. Of the several parameters tested, the duration of the unfiltered (1 Hz to 5 kHz) spike provided the most reliable clustering of the population. High-density parallel recordings of neuronal activity, determination of their physical location and their classification into pyramidal and interneuron classes provide the necessary tools for local circuit analysis.


Subject(s)
Action Potentials/physiology , Extracellular Fluid/physiology , Interneurons/physiology , Neocortex/physiology , Nerve Net/physiology , Animals , Neural Inhibition/physiology , Rats , Rats, Sprague-Dawley
18.
J Comp Neurol ; 464(2): 238-56, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12898615

ABSTRACT

This study investigates, in the anesthetized rat, the dendritic extent of parabrachial (PB) neurons whose nociceptive response to noxious stimuli has been previously recorded with an extracellular micropipette. The PB neurons were then injected with biocytin through the recording micropipette, via a juxtacellular technique. The dendritic arborization of individual PB neurons was carefully compared with the projections of medullary (trigeminal) and spinal lamina I neurons. The latter projections were labeled in separate animals that received injections of Phaseolus vulgaris-leucoagglutinin restricted to the superficial layers of spinal or medullary dorsal horn. We report here that: 1) PB neurons excited chiefly by noxious stimulation of the face have their dendritic tree located primarily within the field of lamina I trigeminal projections, i.e., in the caudal portion of PB area, around the external medial and the caudal part of the external lateral subnuclei; and 2) PB neurons excited chiefly by noxious stimulation of the paw or the tail have their dendritic tree located primarily within the field of lamina I spinal projections, i.e., in PB mid-extent, around the borderline between the external lateral and both the lateral crescent and the superior lateral subnuclei. Our results suggest the presence of an extensive excitatory axodendritic link between lamina I projections and PB nociceptive neurons around the lateral crescent and the external medial subnuclei. These findings strengthen the possibility of involvement of a subgroup of PB neurons in nociceptive processes.


Subject(s)
Dendrites/ultrastructure , Neural Pathways/anatomy & histology , Nociceptors/physiology , Pons/anatomy & histology , Animals , Dendrites/physiology , Male , Medulla Oblongata/anatomy & histology , Medulla Oblongata/physiology , Pain/physiopathology , Pons/physiology , Posterior Horn Cells/anatomy & histology , Posterior Horn Cells/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/anatomy & histology , Spinal Cord/physiology , Trigeminal Nuclei/anatomy & histology , Trigeminal Nuclei/physiology
19.
Pain ; 103(1-2): 83-91, 2003 May.
Article in English | MEDLINE | ID: mdl-12749962

ABSTRACT

We have recently described a population of neurons in the lateral part of the ventromedial thalamus (VMl), that respond exclusively to noxious cutaneous stimuli, regardless of which part of the body is stimulated. The purpose of the present study was to investigate the convergence of cutaneous, muscular and visceral noxious inputs onto single, VMl neurons in anesthetized rats. VMl neurons were characterized by their responses to Adelta- and C-fiber activation as well as noxious heat applied to the hindpaw. We investigated whether they responded also to colorectal distensions. In an additional series of experiments, we tested the effects of colorectal, intraperitoneal, intramuscular and subcutaneous applications of the chemical irritant mustard oil (MO). The present study shows that a population of neurons located within the thalamic VMl nucleus, carries nociceptive somatosensory signals from the entire body. All these neurons responded to noxious cutaneous and intramuscular stimuli but not to levels of distension that could be considered innocuous or noxious, of the intact and inflammed colon and rectum. Although colorectal distension did not elicit VMl responses, convergence of visceral as well as muscle and cutaneous nociceptors was demonstrated by the increases in ongoing (background) discharges following intracolonic MO. A distinct effect is seen after MO injection into the lumen of the colon: an increase in ongoing activity for 15min but still a lack of effect of colorectal distension. Moreover, following inflammation induced by subcutaneous injections of MO VMl neurons developed responses to both thermal and mechanical innocuous skin stimulation, reminiscent of allodynia phenomena. It is suggested that the VMl contributes to attentional aspects of nociceptive processing and/or to the integration of widespread noxious events in terms of the appropriate potential motor responses.


Subject(s)
Afferent Pathways/physiology , Muscles/physiology , Neurons/physiology , Skin , Ventral Thalamic Nuclei/physiology , Viscera/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Drug Administration Routes/veterinary , Electric Stimulation , Male , Morphine/pharmacology , Muscles/innervation , Mustard Plant , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Pain/physiopathology , Physical Stimulation , Plant Extracts/pharmacology , Plant Oils , Rats , Rats, Sprague-Dawley , Skin/innervation , Stimulation, Chemical , Ventral Thalamic Nuclei/anatomy & histology , Ventral Thalamic Nuclei/drug effects , Viscera/innervation
20.
Eur J Pain ; 6(1): 81-7, 2002.
Article in English | MEDLINE | ID: mdl-11888231

ABSTRACT

The lateral part of the ventromedial thalamus (VM l) relays nociceptive inputs from the whole body surface to the dorsolateral frontal cortex. The aim of the present study was to investigate the effects of systemic morphine on nociceptive activity evoked in VM l neurones either by thermal (48 degrees C) or by supramaximal percutaneous electrical stimuli. The noxious thermal evoked responses were depressed by 10.8 +/- 10.1%, 48.3 +/- 23.0% and 67.3 +/- 10.1%, 5 min after i.v. injections of 1.0, 1.73 and 3.0 mg/kg of morphine, respectively. Moreover, strong depressive effects on the Adelta- and C-fibre responses were already present 5 min after the injection. The responses were significantly reduced by 7.2 +/- 5.9%, 32.5 +/ 11.1% and 37.2 +/- 11.8% for Adelta fibres after i.v. injections of 1.0, 1.73 and 3.0 mg/kg of morphine, respectively. The corresponding values for C-fibre evoked responses were 16.3 +/- 16.2%, 57.0 +/- 12.0% and 69.0 +/- 8.2%. The dose of morphine that reduced VM l neuronal nociceptive responses by 50% (1.73 mg/kg) was around 3.5 times lower than that necessary to inhibit the responses of its spinal or medullary relays under similar experimental conditions. These results, added to the data of the literature, suggest that supraspinal effects of morphine are primarily mediated at the thalamic level. It is tempting to speculate that morphine-induced reductions of attentional or psychomotor responses related to pain may be mediated by its action on VM l.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Nerve Fibers, Myelinated/drug effects , Nerve Fibers/drug effects , Neurons/drug effects , Pain/drug therapy , Ventral Thalamic Nuclei/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Dose-Response Relationship, Drug , Electric Stimulation , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nerve Fibers/physiology , Nerve Fibers, Myelinated/physiology , Neural Conduction/drug effects , Neural Conduction/physiology , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/physiology , Nociceptors/drug effects , Nociceptors/physiology , Pain/physiopathology , Physical Stimulation , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Rats , Rats, Sprague-Dawley , Ventral Thalamic Nuclei/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...