Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Brain ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916480

ABSTRACT

BACKGROUND: Pain is a non-motor symptom that impairs quality of life in Parkinson's patients. Pathological nociceptive hypersensitivity in patients could be due to changes in the processing of somatosensory information at the level of the basal ganglia, including the subthalamic nucleus (STN), but the underlying mechanisms are not yet defined. Here, we investigated the interaction between the STN and the dorsal horn of the spinal cord (DHSC), by first examining the nature of STN neurons that respond to peripheral nociceptive stimulation and the nature of their responses under normal and pathological conditions. Next, we studied the consequences of deep brain stimulation (DBS) of the STN on the electrical activity of DHSC neurons. Then, we investigated whether the therapeutic effect of STN-DBS would be mediated by the brainstem descending pathway involving the rostral ventromedial medulla (RVM). Finally, to better understand how the STN modulates allodynia, we used Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) expressed in the STN. METHODS: The study was carried out on the 6-OHDA rodent model of Parkinson's disease, obtained by stereotactic injection of the neurotoxin into the medial forebrain bundle of rats and mice. In these animals, we used motor and nociceptive behavioral tests, in vivo electrophysiology of STN and wide dynamic range (WDR) DHSC neurons in response to peripheral stimulation, deep brain stimulation of the STN and the selective DREADD approach. Vglut2-ires-cre mice were used to specifically target and inhibit STN glutamatergic neurons. RESULTS: STN neurons are able to detect nociceptive stimuli, encode their intensity and generate windup-like plasticity, like WDR neurons in the DHSC. These phenomena are impaired in dopamine-depleted animals, as the intensity response is altered in both spinal and subthalamic neurons. Furthermore, As with L-Dopa, STN-DBS in rats ameliorated 6-OHDA-induced allodynia, and this effect is mediated by descending brainstem projections leading to normalization of nociceptive integration in DHSC neurons. Furthermore, this therapeutic effect was reproduced by selective inhibition of STN glutamatergic neurons in Vglut2-ires-cre mice. CONCLUSION: Our study highlights the centrality of the STN in nociceptive circuits, its interaction with the DHSC and its key involvement in pain sensation in Parkinson's disease. Furthermore, our results provide for the first-time evidence that subthalamic DBS produces analgesia by normalizing the responses of spinal WDR neurons via descending brainstem pathways. These effects are due to direct inhibition, rather than activation of glutamatergic neurons in the STN or passage fibers, as shown in the DREADDs experiment.

2.
Cells ; 13(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38920628

ABSTRACT

Chronic pain is a pathological state defined as daily pain sensation over three consecutive months. It affects up to 30% of the general population. Although significant research efforts have been made in the past 30 years, only a few and relatively low effective molecules have emerged to treat chronic pain, with a considerable translational failure rate. Most preclinical models have focused on sensory neurotransmission, with particular emphasis on the dorsal horn of the spinal cord as the first relay of nociceptive information. Beyond impaired nociceptive transmission, chronic pain is also accompanied by numerous comorbidities, such as anxiety-depressive disorders, anhedonia and motor and cognitive deficits gathered under the term "pain matrix". The emergence of cutting-edge techniques assessing specific neuronal circuits allow in-depth studies of the connections between "pain matrix" circuits and behavioural outputs. Pain behaviours are assessed not only by reflex-induced responses but also by various or more complex behaviours in order to obtain the most complete picture of an animal's pain state. This review summarises the latest findings on pain modulation by brain component of the pain matrix and proposes new opportunities to unravel the mechanisms of chronic pain.


Subject(s)
Chronic Pain , Animals , Humans , Chronic Pain/physiopathology , Chronic Pain/therapy , Disease Models, Animal , Pain/physiopathology , Nerve Net/physiopathology
3.
Sci Adv ; 8(30): eabo0689, 2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35895817

ABSTRACT

Descending control from the brain to the spinal cord shapes our pain experience, ranging from powerful analgesia to extreme sensitivity. Increasing evidence from both preclinical and clinical studies points to an imbalance toward descending facilitation as a substrate of pathological pain, but the underlying mechanisms remain unknown. We used an optogenetic approach to manipulate serotonin (5-HT) neurons of the nucleus raphe magnus that project to the dorsal horn of the spinal cord. We found that 5-HT neurons exert an analgesic action in naïve mice that becomes proalgesic in an experimental model of neuropathic pain. We show that spinal KCC2 hypofunction turns this descending inhibitory control into paradoxical facilitation; KCC2 enhancers restored 5-HT-mediated descending inhibition and analgesia. Last, combining selective serotonin reuptake inhibitors (SSRIs) with a KCC2 enhancer yields effective analgesia against nerve injury-induced pain hypersensitivity. This uncovers a previously unidentified therapeutic path for SSRIs against neuropathic pain.

4.
Proc Natl Acad Sci U S A ; 119(30): e2114094119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858441

ABSTRACT

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


Subject(s)
Attention Deficit Disorder with Hyperactivity , Hyperalgesia , Pain , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Disease Models, Animal , Gyrus Cinguli/physiopathology , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Impulsive Behavior , Mice , Optogenetics , Oxidopamine/pharmacology , Pain/chemically induced , Pain/physiopathology , Sympatholytics/pharmacology
5.
Mov Disord ; 36(8): 1737-1743, 2021 08.
Article in English | MEDLINE | ID: mdl-34080714

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an opportunistic pathogen that infects the upper respiratory tract in humans and causes serious illness, including fatal pneumonia and neurological disorders. Several studies have reported that SARS-CoV-2 may worsen the symptoms of Parkinson's disease (PD), with the potential to increase mortality rates in patients with advanced disease. The potential risk of SARS-CoV-2 to induce PD has also been suggested because the virus can enter the brain, where it can trigger cellular processes involved in neurodegeneration. In this review, we will discuss the potential of SARS-CoV-2 to exacerbate and cause certain neurological disorders, including PD. We will then elucidate its impact on the brain while examining its pathways and mechanisms of action. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
COVID-19 , Nervous System Diseases , Parkinson Disease , Brain , Humans , Parkinson Disease/complications , SARS-CoV-2
6.
Prog Neurobiol ; 201: 102030, 2021 06.
Article in English | MEDLINE | ID: mdl-33711402

ABSTRACT

Animal models of pain consist of modeling a pain-like state and measuring the consequent behavior. The first animal models of neuropathic pain (NP) were developed in rodents with a total lesion of the sciatic nerve. Later, other models targeting central or peripheral branches of nerves were developed to identify novel mechanisms that contribute to persistent pain conditions in NP. Objective assessment of pain in these different animal models represents a significant challenge for pre-clinical research. Multiple behavioral approaches are used to investigate and to validate pain phenotypes including withdrawal reflex to evoked stimuli, vocalizations, spontaneous pain, but also emotional and affective behaviors. Furthermore, animal models were very useful in investigating the mechanisms of NP. This review will focus on a detailed description of rodent models of NP and provide an overview of the assessment of the sensory and emotional components of pain. A detailed inventory will be made to examine spinal mechanisms involved in NP-induced hyperexcitability and underlying the current pharmacological approaches used in clinics with the possibility to present new avenues for future treatment. The success of pre-clinical studies in this area of research depends on the choice of the relevant model and the appropriate test based on the objectives of the study.


Subject(s)
Neuralgia , Animals , Disease Models, Animal , Ion Channels , Rats, Sprague-Dawley , Sciatic Nerve
7.
J Neurosci Methods ; 348: 108997, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33188801

ABSTRACT

Chronic pain is a maladaptive neurological disease that remains a major health problem. A deepening of our knowledge on mechanisms that cause pain is a prerequisite to developing novel treatments. A large variety of animal models of pain has been developed that recapitulate the diverse symptoms of different pain pathologies. These models reproduce different pain phenotypes and remain necessary to examine the multidimensional aspects of pain and understand the cellular and molecular basis underlying pain conditions. In this review, we propose an overview of animal models, from simple organisms to rodents and non-human primates and the specific traits of pain pathologies they model. We present the main behavioral tests for assessing pain and investing the underpinning mechanisms of chronic pathological pain. The validity of animal models is analysed based on their ability to mimic human clinical diseases and to predict treatment outcomes. Refine characterization of pathological phenotypes also requires to consider pain globally using specific procedures dedicated to study emotional comorbidities of pain. We discuss the limitations of pain models when research findings fail to be translated from animal models to human clinics. But we also point to some recent successes in analgesic drug development that highlight strategies for improving the predictive validity of animal models of pain. Finally, we emphasize the importance of using assortments of preclinical pain models to identify pain subtype mechanisms, and to foster the development of better analgesics.


Subject(s)
Analgesics , Chronic Pain , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Primates
8.
Front Cell Neurosci ; 14: 214, 2020.
Article in English | MEDLINE | ID: mdl-32765223

ABSTRACT

In the central nervous system, the inhibitory GABAB receptor is the archetype of heterodimeric G protein-coupled receptors (GPCRs). Receptor interaction with partner proteins has emerged as a novel mechanism to alter GPCR signaling in pathophysiological conditions. We propose here that GABAB activity is inhibited through the specific binding of fibulin-2, an extracellular matrix protein, to the B1a subunit in a rat model of neuropathic pain. We demonstrate that fibulin-2 hampers GABAB activation, presumably through decreasing agonist-induced conformational changes. Fibulin-2 regulates the GABAB-mediated presynaptic inhibition of neurotransmitter release and weakens the GABAB-mediated inhibitory effect in neuronal cell culture. In the dorsal spinal cord of neuropathic rats, fibulin-2 is overexpressed and colocalized with B1a. Fibulin-2 may thus interact with presynaptic GABAB receptors, including those on nociceptive afferents. By applying anti-fibulin-2 siRNA in vivo, we enhanced the antinociceptive effect of intrathecal baclofen in neuropathic rats, thus demonstrating that fibulin-2 limits the action of GABAB agonists in vivo. Taken together, our data provide an example of an endogenous regulation of GABAB receptor by extracellular matrix proteins and demonstrate its functional impact on pathophysiological processes of pain sensitization.

9.
Int J Mol Sci ; 20(24)2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31817540

ABSTRACT

Windup, a progressive increase in spinal response to repetitive stimulations of nociceptive peripheral fibers, is a useful model to study central sensitization to pain. Windup is expressed by neurons in both the dorsal and ventral horn of the spinal cord. In juvenile rats, it has been demonstrated both in vivo and in vitro that windup depends on calcium-dependent intrinsic properties and their modulation by synaptic components. However, the involvement of these two components in the adults remains controversial. In the present study, by means of electromyographic and extracellular recordings, we show that windup in adults, in vivo, depends on a synaptic balance between excitatory N-methyl-D-aspartate (NMDA) receptors and inhibitory glycinergic receptors. We also demonstrate the involvement of L-type calcium channels in both the dorsal and ventral horn of the spinal cord. These results indicate that windup in adults is similar to juvenile rats and that windup properties are the same regardless of the spinal network, i.e., sensory or motor.


Subject(s)
Calcium Channels, L-Type/metabolism , Nociception , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Reflex , Synapses/metabolism , Animals , Posterior Horn Cells/cytology , Rats , Rats, Wistar
10.
Pain ; 160(2): 345-357, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30281531

ABSTRACT

Pain is associated with negative emotions such as anxiety, but the underlying neurocircuitry and modulators of the association of pain and anxiety remain unclear. The neuropeptide cholecystokinin (CCK) has both pronociceptive and anxiogenic properties, so we explored the role of CCK in anxiety and nociception in the central amygdala (CeA), a key area in control of emotions and descending pain pathways. Local infusion of CCK into the CeA of control rats increased anxiety, as measured in the light-dark box test, but had no effect on mechanical sensitivity. By contrast, intra-CeA CCK infusion 4 days after Complete Freund's Adjuvant (CFA) injection into the hindpaw resulted in analgesia, but also in loss of its anxiogenic capacity. Inflammatory conditions induced changes in the CeA CCK signaling system with an increase of CCK immunoreactivity and a decrease in CCK1, but not CCK2, receptor mRNA. In CFA rats, patch-clamp experiments revealed that CCK infusion increased CeA neuron excitability. It also partially blocked the discharge of wide dynamic range neurons in the dorsal spinal cord. These effects of CCK on CeA and spinal neurons in CFA rats were mimicked by the specific CCK2 receptor agonist, gastrin. This analgesic effect was likely mediated by identified CeA neurons projecting to the periaqueductal gray matter that express CCK receptors. Together, our data demonstrate that intra-CeA CCK infusion activated a descending CCK2 receptor-dependent pathway that inhibited spinal neuron discharge. Thus, persistent pain induces a functional switch to a newly identified analgesic capacity of CCK in the amygdala, indicating central emotion-related circuit controls pain transmission in spinal cord.


Subject(s)
Amygdala/metabolism , Cholecystokinin/metabolism , Pain/pathology , Receptor, Cholecystokinin B/metabolism , Signal Transduction/physiology , Amygdala/pathology , Animals , Dark Adaptation/drug effects , Disease Models, Animal , Exploratory Behavior/drug effects , Freund's Adjuvant/toxicity , Gastrins/therapeutic use , Glutamate Decarboxylase/metabolism , Inflammation/chemically induced , Inflammation/complications , Male , Neurons/drug effects , Neurons/physiology , Nociception/drug effects , Pain/etiology , Pain Threshold/drug effects , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/genetics , Signal Transduction/drug effects , Sincalide/therapeutic use , Tetragastrin/analogs & derivatives , Tetragastrin/therapeutic use
11.
Sci Rep ; 8(1): 15349, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337626

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental disorder characterized by impaired attention, impulsivity and hyperactivity. The "neonatal 6-hydroxydopamine" (6-OHDA) lesion is a commonly used model of ADHD in rat. However, a comprehensive assessment of ADHD-like symptoms is still missing, and data in mouse remain largely unavailable. Our aim was to analyse symptoms of ADHD in the mouse neonatal 6-OHDA model. 6-OHDA mice exhibited the major ADHD-like symptoms, i.e. hyperactivity (open field), attention deficit and impulsivity (five-choice serial reaction time task). Further, the model revealed discrete co-existing symptoms, i.e. anxiety-like (elevated plus maze test) and antisocial (social interaction) behaviours and decreased cognitive functioning (novel object recognition). The efficacy of methylphenidate, a classical psychostimulant used in the treatment of ADHD, was also evaluated. A histological analysis further supports the model validity by indicating dopamine depletion, changes in cortical thickness and abnormalities in anterior cingulate cortex neurons. A principal component analysis of the behaviour profile confirms that the 6-OHDA mouse model displayed good face and predictive validity. We conclude that neonatal dopamine depletion results in behavioural and morphological changes similar to those seen in patients and therefore could be used as a model for studying ADHD pathophysiological mechanisms and identifying therapeutic targets.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Behavior, Animal/drug effects , Disease Models, Animal , Maze Learning/drug effects , Oxidopamine/toxicity , Social Behavior , Animals , Animals, Newborn , Attention Deficit Disorder with Hyperactivity/chemically induced , Attention Deficit Disorder with Hyperactivity/physiopathology , Male , Mice , Rats
12.
Mov Disord ; 33(6): 1010-1015, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29774960

ABSTRACT

BACKGROUND: Pain is a major non motor symptom that contributes to impaired quality of life in PD. However, its mechanism is unknown. OBJECTIVES AND METHODS: We sought to identify the pain phenotypes and parallel changes in spinal integration of peripheral stimuli in a rat model of PD induced by lesions of SN dopamine neurons, using behavioral plantar and von Frey tests as well as electrophysiology of the dorsal horn. RESULTS: We show that dopamine depletion by 6-OHDA induced hypersensitivity to mechanical and thermal stimuli. These abnormal behaviors were paralleled by increased neuronal responses and hyperexcitability of wide dynamic range neurons of lamina V of the dorsal horn of the spinal cord in response to electrical stimulation of the sciatic nerve in the 6-OHDA model as compared to sham rats. CONCLUSIONS: These results provide evidence for alteration of nociceptive integration in the spinal dorsal horn neurons in 6-OHDA rats that can reflect changes in pain behavior. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Hyperalgesia/etiology , Hyperkinesis/chemically induced , Parkinson Disease, Secondary/complications , Parkinson Disease, Secondary/pathology , Spinal Cord/pathology , Action Potentials/physiology , Animals , Disease Models, Animal , Dopamine/metabolism , Functional Laterality , Hyperalgesia/pathology , Male , Neurons/physiology , Oxidopamine/toxicity , Pain Measurement , Pain Threshold/drug effects , Pain Threshold/physiology , Parkinson Disease, Secondary/chemically induced , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sympatholytics/toxicity
13.
Exp Neurol ; 307: 1-11, 2018 09.
Article in English | MEDLINE | ID: mdl-29772245

ABSTRACT

Inflammatory pain is a complex and multifactorial disorder. Apurinic/apyrimidinic endonuclease 1 (APE1), also called Redox Factor-1 (Ref-1), is constitutively expressed in the central nervous system and regulates various cellular functions including oxidative stress. In the present study, we investigated APE1 modulation and associated pain behavior changes in the complete Freund's adjuvant (CFA) model of inflammatory pain in rats. In addition we tested the anti-inflammatory effects of E3330, a selective inhibitor of APE1-redox activity, in CFA pain condition. We demonstrate that APE1 expression and subcellular distribution are significantly altered in rats at 4 days post CFA injection. We observed around 30% reduction in the overall APE1 mRNA and protein levels. Interestingly, our data point to an increased nuclear accumulation in the inflamed group as compared to the sham group. E3330 inhibitor injection in CFA rats normalized APE1 mRNA expression and changed its distribution toward cytosolic accumulation. Furthermore, intrathecal injection of E3330 decreased inflammation (i.e. reduced IL-6 expression) and alleviated pain, as assessed by measuring the paw withdrawal threshold with the von Frey test. In conclusion, our data indicate that changes in APE1 expression and sub-cellular distribution are implicated in inflammatory pain mechanisms mediated by APE1 redox functions. Further studies are required to elucidate the exact function of APE1 in inflammatory pain processes.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Oxidative Stress/physiology , Pain/metabolism , Animals , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Freund's Adjuvant/toxicity , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Pain/chemically induced , Pain/drug therapy , Propionates/pharmacology , Propionates/therapeutic use , Rats , Rats, Sprague-Dawley
14.
Mol Pain ; 13: 1744806917737934, 2017.
Article in English | MEDLINE | ID: mdl-29020860

ABSTRACT

Abstract: The dorsal horn of the spinal cord is a crucial site for pain transmission and modulation. Dorsal horn neurons of the spinal cord express group I metabotropic glutamate receptors (group I mGluRs) that exert a complex role in nociceptive transmission. In particular, group I mGluRs promote the activation of L-type calcium channels, voltage-gated channels involved in short- and long-term sensitization to pain. In this study, we analyzed the role of group I mGluRs in spinal nociceptive transmission and the possible cooperation between these receptors and L-type calcium channels in the pathophysiology of pain transmission in the dorsal horn of the spinal cord. We demonstrate that the activation of group I mGluRs induces allodynia and L-type calcium channel-dependent increase in nociceptive field potentials following sciatic nerve stimulation. Surprisingly, in a model of persistent inflammation induced by complete Freund's adjuvant, the activation of group I mGluRs induced an analgesia and a decrease in nociceptive field potentials. Among the group I mGluRs, mGluR1 promotes the activation of L-type calcium channels and increased nociceptive transmission while mGluR5 induces the opposite through the inhibitory network. These results suggest a functional switch exists in pathological conditions that can change the action of group I mGluR agonists into possible analgesic molecules, thereby suggesting new therapeutic perspectives to treat persistent pain in inflammatory settings.


Subject(s)
Hyperalgesia/physiopathology , Inflammation/metabolism , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/metabolism , Animals , Male , Posterior Horn Cells/metabolism , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/analysis , Spinal Cord/physiology , Synapses/metabolism
15.
J Physiol ; 594(22): 6607-6626, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27231046

ABSTRACT

KEY POINTS: L-type calcium channels in the CNS exist as two subunit forming channels, Cav1.2 and Cav1.3, which are involved in short- and long-term plasticity. We demonstrate that Cav1.3 but not Cav1.2 is essential for wind-up. These results identify Cav1.3 as a key conductance responsible for short-term sensitization in physiological pain transmission. We confirm the role of Cav1.2 in a model of long-term plasticity associated with neuropathic pain. Up-regulation of Cav1.2 and down-regultation of Cav1.3 in neuropathic pain underlies the switch from physiology to pathology. Finally, the results of the present study reveal that therapeutic targeting molecular pathways involved in wind-up may be not relevant in the treatment of neuropathy. ABSTRACT: Short-term central sensitization to pain temporarily increases the responsiveness of nociceptive pathways after peripheral injury. In dorsal horn neurons (DHNs), short-term sensitization can be monitored through the study of wind-up. Wind-up, a progressive increase in DHNs response following repetitive peripheral stimulations, depends on the post-synaptic L-type calcium channels. In the dorsal horn of the spinal cord, two L-type calcium channels are present, Cav1.2 and Cav1.3, each displaying specific kinetics and spatial distribution. In the present study, we used a mathematical model of DHNs in which we integrated the specific patterns of expression of each Cav subunits. This mathematical approach reveals that Cav1.3 is necessary for the onset of wind-up, whereas Cav1.2 is not and that synaptically triggered wind-up requires NMDA receptor activation. We then switched to a biological preparation in which we knocked down Cav subunits and confirmed the prominent role of Cav1.3 in both naive and spinal nerve ligation model of neuropathy (SNL). Interestingly, although a clear mechanical allodynia dependent on Cav1.2 expression was observed after SNL, the amplitude of wind-up was decreased. These results were confirmed with our model when adapting Cav1.3 conductance to the changes observed after SNL. Finally, our mathematical approach predicts that, although wind-up amplitude is decreased in SNL, plateau potentials are not altered, suggesting that plateau and wind-up are not fully equivalent. Wind-up and long-term hyperexcitability of DHNs are differentially controlled by Cav1.2 and Cav1.3, therefore confirming that short- and long-term sensitization are two different phenomena triggered by distinct mechanisms.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels/metabolism , Neuralgia/metabolism , Action Potentials/physiology , Animals , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Neuralgia/physiopathology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/physiopathology , Spinal Nerves/metabolism , Spinal Nerves/physiopathology , Synapses/metabolism
16.
Front Aging Neurosci ; 6: 87, 2014.
Article in English | MEDLINE | ID: mdl-24860498

ABSTRACT

Parkinson's disease (PD) is a neurological disorder characterized by the manifestation of motor symptoms, such as akinesia, muscle rigidity and tremor at rest. These symptoms are classically attributed to the degeneration of dopamine neurons in the pars compacta of substantia nigra (SNc), which results in a marked dopamine depletion in the striatum. It is well established that dopamine neurons in the SNc innervate not only the striatum, which is the main target, but also other basal ganglia nuclei including the two segments of globus pallidus and the subthalamic nucleus (STN). The role of dopamine and its depletion in the striatum is well known, however, the role of dopamine depletion in the pallidal complex and the STN in the genesis of their abnormal neuronal activity and in parkinsonian motor deficits is still not clearly determined. Based on recent experimental data from animal models of Parkinson's disease in rodents and non-human primates and also from parkinsonian patients, this review summarizes current knowledge on the role of dopamine in the modulation of basal ganglia neuronal activity and also the role of dopamine depletion in these nuclei in the pathophysiology of Parkinson's disease.

17.
J Comp Neurol ; 522(2): 393-413, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23818225

ABSTRACT

The γ isoform of protein kinase C (PKCγ), which is concentrated in interneurons in the inner part of lamina II (IIi ) of the dorsal horn, has been implicated in the expression of tactile allodynia. Lamina IIi PKCγ interneurons were shown to be activated by tactile inputs and to participate in local circuits through which these inputs can reach lamina I, nociceptive output neurons. That such local circuits are gated by glycinergic inhibition and that A- and C-fibers low threshold mechanoreceptors (LTMRs) terminate in lamina IIi raise the general issue of synaptic inputs to lamina IIi PKCγ interneurons. Combining light and electron microscopic immunochemistry in the rat spinal trigeminal nucleus, we show that PKCγ-immunoreactivity is mostly restricted to interneurons in lamina IIi of the medullary dorsal horn, where they constitute 1/3 of total neurons. The majority of synapses on PKCγ-immunoreactive interneurons are asymmetric (likely excitatory). PKCγ-immunoreactive interneurons appear to receive exclusively myelinated primary afferents in type II synaptic glomeruli. Neither large dense core vesicle terminals nor type I synaptic glomeruli, assumed to be the endings of unmyelinated nociceptive terminals, were found on these interneurons. Moreover, there is no vesicular glutamate transporter 3-immunoreactive bouton, specific to C-LTMRs, on PKCγ-immunoreactive interneurons. PKCγ-immunoreactive interneurons contain GABAA ergic and glycinergic receptors. At the subcellular level, PKCγ-immunoreactivity is mostly concentrated on plasma membranes, close to, but not within, postsynaptic densities. That only myelinated primary afferents were found to contact PKCγ-immunoreactive interneurons suggests that myelinated, but not unmyelinated, LTMRs play a critical role in the expression of mechanical allodynia.


Subject(s)
Hyperalgesia/metabolism , Interneurons/enzymology , Mechanoreceptors/enzymology , Protein Kinase C/metabolism , Synapses/ultrastructure , Animals , Blotting, Western , Immunohistochemistry , Interneurons/ultrastructure , Male , Mechanoreceptors/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Transmission , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/ultrastructure , Nociceptors/enzymology , Nociceptors/ultrastructure , Posterior Horn Cells/enzymology , Posterior Horn Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/enzymology
18.
J Neurosci ; 33(37): 14840-9, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027284

ABSTRACT

Burst firing has been reported as a pathological activity of subthalamic nucleus (STN) neurons in Parkinson's disease. However, the origin of bursts and their causal link with motor deficits remain unknown. Here we tested the hypothesis that dopamine D5 receptors (D5Rs), characterized by a high constitutive activity, may contribute to the emergence of burst firing in STN. We tested whether inhibiting D5R constitutive activity depresses burst firing and alleviates motor impairments in the 6-OHDA rat model of Parkinson's disease. Intrasubthalamic microinjections of either an inverse agonist of D5Rs, flupenthixol, or a D2R antagonist, raclopride, were applied. Behavioral experiments, in vivo and in vitro electrophysiological recordings, and ex vivo functional neuroanatomy studies were performed. Using [(5)S]GTPγ binding autoradiography, we show that application of flupenthixol inhibits D5R constitutive activity within the STN. Furthermore, flupenthixol reduced evoked burst in brain slices and converted pathological burst firing into physiological tonic, single-spike firing in 6-OHDA rats in vivo. This later action was mimicked by calciseptine, a Cav1 channel blocker. Moreover, the same treatment dramatically attenuated motor impairment in this model and normalized metabolic hyperactivity in both STN and substantia nigra pars reticulata, the main output structure of basal ganglia in rats. In contrast, raclopride as well as saline did not reverse burst firing and motor deficits, confirming the selective action of flupenthixol on D5Rs. These results are the first to demonstrate that subthalamic D5Rs are involved in the pathophysiology of Parkinson's disease and that administering an inverse agonist of these receptors may lessen motor symptoms.


Subject(s)
Dopamine Antagonists/therapeutic use , Flupenthixol/therapeutic use , Locomotion/physiology , Parkinson Disease/drug therapy , Receptors, Dopamine D5/metabolism , Subthalamic Nucleus/metabolism , Action Potentials/drug effects , Animals , Animals, Newborn , Disease Models, Animal , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Female , Flupenthixol/pharmacology , In Vitro Techniques , Locomotion/drug effects , Male , Neurons/drug effects , Oxidopamine/toxicity , Parkinson Disease/etiology , Raclopride/pharmacology , Rats , Rats, Wistar , Statistics, Nonparametric , Subthalamic Nucleus/drug effects , Subthalamic Nucleus/pathology
19.
EMBO J ; 31(15): 3239-51, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22692127

ABSTRACT

In the central nervous system, the inhibitory GABAB receptor is the archetype of heterodimeric G protein-coupled receptors (GPCRs). However, the regulation of GABAB dimerization, and more generally of GPCR oligomerization, remains largely unknown. We propose a novel mechanism for inhibition of GPCR activity through de-dimerization in pathological conditions. We show here that 14-3-3ζ, a GABAB1-binding protein, dissociates the GABAB heterodimer, resulting in the impairment of GABAB signalling in spinal neurons. In the dorsal spinal cord of neuropathic rats, 14-3-3ζ is overexpressed and weakens GABAB inhibition. Using anti-14-3-3ζ siRNA or competing peptides disrupts 14-3-3ζ/GABAB1 interaction and restores functional GABAB heterodimers in the dorsal horn. Importantly, both strategies greatly enhance the anti-nociceptive effect of intrathecal Baclofen in neuropathic rats. Taken together, our data provide the first example of endogenous regulation of a GPCR oligomeric state and demonstrate its functional impact on the pathophysiological process of neuropathic pain sensitization.


Subject(s)
14-3-3 Proteins/physiology , Chronic Pain/pathology , Receptors, GABA-B/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Animals , Cells, Cultured , Chronic Pain/genetics , Chronic Pain/metabolism , Disease Models, Animal , Neuralgia/genetics , Neuralgia/metabolism , Neuralgia/pathology , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Posterior Horn Cells/pathology , Protein Binding/genetics , Protein Binding/physiology , Protein Interaction Domains and Motifs/drug effects , Protein Multimerization/drug effects , Protein Multimerization/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Transgenic , Receptors, GABA-B/chemistry , Receptors, GABA-B/genetics
20.
EMBO J ; 30(18): 3830-41, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21804529

ABSTRACT

Chronic pain states are characterized by long-term sensitization of spinal cord neurons that relay nociceptive information to the brain. Among the mechanisms involved, up-regulation of Cav1.2-comprising L-type calcium channel (Cav1.2-LTC) in spinal dorsal horn have a crucial role in chronic neuropathic pain. Here, we address a mechanism of translational regulation of this calcium channel. Translational regulation by microRNAs is a key factor in the expression and function of eukaryotic genomes. Because perfect matching to target sequence is not required for inhibition, theoretically, microRNAs could regulate simultaneously multiple mRNAs. We show here that a single microRNA, miR-103, simultaneously regulates the expression of the three subunits forming Cav1.2-LTC in a novel integrative regulation. This regulation is bidirectional since knocking-down or over-expressing miR-103, respectively, up- or down-regulate the level of Cav1.2-LTC translation. Functionally, we show that miR-103 knockdown in naive rats results in hypersensitivity to pain. Moreover, we demonstrate that miR-103 is down-regulated in neuropathic animals and that miR-103 intrathecal applications successfully relieve pain, identifying miR-103 as a novel possible therapeutic target in neuropathic chronic pain.


Subject(s)
Calcium Channels, L-Type/biosynthesis , Gene Expression Regulation , MicroRNAs/metabolism , Pain , Protein Biosynthesis , Animals , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...