Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Med Sci (Paris) ; 40(1): 64-71, 2024 Jan.
Article in French | MEDLINE | ID: mdl-38299905

ABSTRACT

Head pain and notably orofacial pain differs from spinal pain on pathophysiological, clinical, therapeutic and prognostic levels. Its high prevalence, important impact on quality of life and significant socio-economical burden justify specific study of such type of pain. Among them, neuropathic orofacial pain resulting from disease or trauma of the trigeminal nervous system is among the most difficult types of pain to diagnose and to treat. Deciphering of underlying peripheral and central mechanisms has allowed numerous conceptual, clinical and therapeutic advances, notably the role of neural and non neural cell types, such as glia, immunocytes, vascular endothelial cells or the role of trigeminal sensory complex neural circuitry reconfiguration in the development of post-traumatic trigeminal neuropathic pain. Cellular interactions within the trigeminal ganglion, allowing a better understanding of several painful dental, ocular or cephalalgic comorbidities, are also described.


Title: Spécificités de la douleur neuropathique oro-faciale. Abstract: Les douleurs de la région céphalique ­ et notamment les douleurs oro-faciales ­ diffèrent des douleurs spinales sur les plans physiopathologique, clinique, thérapeutique et pronostique. Leur prévalence élevée, leur fort retentissement sur la qualité de vie individuelle et leur impact économique et sociétal important justifient une étude spécifique. Parmi ces douleurs, les douleurs neuropathiques, résultant d'une maladie ou d'un traumatisme du système nerveux trigéminal, sont parmi les plus difficiles à diagnostiquer et à soigner. L'étude des mécanismes neurobiologiques, périphériques et centraux les sous-tendant a permis de nombreuses avancées conceptuelles, cliniques et thérapeutiques, avec, par exemple, la mise en évidence du rôle des cellules nerveuses et non nerveuses, telles que la glie, les immunocytes, les cellules endothéliales vasculaires ou le rôle de la reconfiguration de la circuiterie nerveuse au niveau du complexe sensitif trigéminal, dans la genèse des douleurs neuropathiques post-lésionnelles. Les interactions cellulaires au sein du ganglion trigéminal, susceptibles d'éclairer la compréhension de certaines comorbidités douloureuses dentaires, oculaires ou céphalalgiques, sont également décrites.


Subject(s)
Neuralgia , Trigeminal Neuralgia , Humans , Endothelial Cells/metabolism , Quality of Life , Neuralgia/etiology , Neuralgia/therapy , Facial Pain/etiology , Facial Pain/therapy , Facial Pain/diagnosis , Trigeminal Neuralgia/diagnosis , Headache
2.
Nutrients ; 14(8)2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35458170

ABSTRACT

Conflicting evidence exists concerning the effects of nutrient intake in osteoarthritis (OA). A systematic literature review and meta-analysis were performed using PubMed, EMBASE, and Cochrane Library up to November 2021 to assess the effects of nutrients on pain, stiffness, function, quality of life, and inflammation markers. We obtained 52 references including 50 on knee OA. Twelve studies compared 724 curcumin patients and 714 controls. Using the standardized mean difference, improvement was significant with regard to pain and function in the curcumin group compared to placebo, but not with active treatment (i.e., nonsteroidal anti-inflammatory drugs, chondroitin, or paracetamol). Three studies assessed the effects of ginger on OA symptoms in 166 patients compared to 164 placebo controls. Pain was the only clinical parameter that significantly decreased. Vitamin D supplementation caused a significant decrease in pain and function. Omega-3 and vitamin E caused no changes in OA parameters. Herbal formulations effects were significant only for stiffness compared to placebo, but not with active treatment. A significant decrease in inflammatory markers was found, especially with ginger. Thus, curcumin and ginger supplementation can have a favorable impact on knee OA symptoms. Other studies are needed to better assess the effects of omega-3 and vitamin D.


Subject(s)
Curcumin , Osteoarthritis, Knee , Zingiber officinale , Curcumin/therapeutic use , Dietary Supplements , Humans , Pain/drug therapy , Quality of Life , Vitamin D/therapeutic use
4.
Neuron ; 109(1): 73-90.e7, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33181066

ABSTRACT

The spinal dorsal horn is a major site for the induction and maintenance of mechanical allodynia, but the circuitry that underlies this clinically important form of pain remains unclear. The studies presented here provide strong evidence that the neural circuits conveying mechanical allodynia in the dorsal horn differ by the nature of the injury. Calretinin (CR) neurons in lamina II inner convey mechanical allodynia induced by inflammatory injuries, while protein kinase C gamma (PKCγ) neurons at the lamina II/III border convey mechanical allodynia induced by neuropathic injuries. Cholecystokinin (CCK) neurons located deeper within the dorsal horn (laminae III-IV) are important for both types of injuries. Interestingly, the Maf+ subset of CCK neurons is composed of transient vesicular glutamate transporter 3 (tVGLUT3) neurons, which convey primarily dynamic allodynia. Identification of an etiology-based circuitry for mechanical allodynia in the dorsal horn has important implications for the mechanistic and clinical understanding of this condition.


Subject(s)
Hyperalgesia/metabolism , Nerve Net/metabolism , Pain Measurement/methods , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Injuries/metabolism , Amino Acid Transport Systems, Acidic/metabolism , Animals , Female , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/chemistry , Nerve Net/pathology , Spinal Cord Dorsal Horn/chemistry , Spinal Cord Dorsal Horn/pathology , Spinal Cord Injuries/pathology
5.
J Neural Transm (Vienna) ; 127(4): 505-525, 2020 04.
Article in English | MEDLINE | ID: mdl-32239353

ABSTRACT

The dorsal horns of the spinal cord and the trigeminal nuclei in the brainstem contain neuron populations that are critical to process sensory information. Neurons in these areas are highly heterogeneous in their morphology, molecular phenotype and intrinsic properties, making it difficult to identify functionally distinct cell populations, and to determine how these are engaged in pathophysiological conditions. There is a growing consensus concerning the classification of neuron populations, based on transcriptomic and transductomic analyses of the dorsal horn. These approaches have led to the discovery of several molecularly defined cell types that have been implicated in cutaneous mechanical allodynia, a highly prevalent and difficult-to-treat symptom of chronic pain, in which touch becomes painful. The main objective of this review is to provide a contemporary view of dorsal horn neuronal populations, and describe recent advances in our understanding of on how they participate in cutaneous mechanical allodynia.


Subject(s)
Chronic Pain , Hyperalgesia , Inflammation , Neuralgia , Posterior Horn Cells , Animals , Chronic Pain/immunology , Chronic Pain/metabolism , Chronic Pain/physiopathology , Hyperalgesia/immunology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Neuralgia/immunology , Neuralgia/metabolism , Neuralgia/physiopathology , Posterior Horn Cells/classification , Posterior Horn Cells/cytology , Posterior Horn Cells/metabolism
6.
Science ; 354(6312): 578-584, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27811268

ABSTRACT

The mammalian nervous system encodes many different forms of pain, from those that arise as a result of short-term low-grade interactions with noxious thermal, chemical, or mechanical sources to more serious forms of pain induced by trauma and disease. In this Review, we highlight recent advances in our understanding of the neural circuits that encode these types of pain. Promising therapeutic strategies based on recent advances are also highlighted.


Subject(s)
Neural Pathways/physiology , Nociceptors/physiology , Pain/physiopathology , Animals , Brain/physiopathology , Humans , Hyperalgesia/physiopathology , Neural Pathways/anatomy & histology , Pain/drug therapy , Pain Management/methods , Presynaptic Terminals/physiology , Spinal Cord/anatomy & histology , Spinal Cord/physiopathology
7.
Anesthesiology ; 124(5): 1136-52, 2016 May.
Article in English | MEDLINE | ID: mdl-26745451

ABSTRACT

BACKGROUND: Tissue injury enhances pain sensitivity both at the site of tissue damage and in surrounding uninjured skin (secondary hyperalgesia). Secondary hyperalgesia encompasses several pain symptoms including pain to innocuous punctate stimuli or static mechanical allodynia. How injury-induced barrage from C-fiber nociceptors produces secondary static mechanical allodynia has not been elucidated. METHODS: Combining behavioral, immunohistochemical, and Western blot analysis, the authors investigated the cell and molecular mechanisms underlying the secondary static mechanical allodynia in the rat medullary dorsal horn (MDH) using the capsaicin model (n = 4 to 5 per group). RESULTS: Intradermal injection of capsaicin (25 µg) into the vibrissa pad produces a spontaneous pain and a secondary static mechanical allodynia. This allodynia is associated with the activation of a neuronal network encompassing lamina I-outer lamina III, including interneurons expressing the γ isoform of protein kinase C (PKCγ) within inner lamina II (IIi) of MDH. PKCγ is concomitantly phosphorylated (+351.4 ± 79.2%, mean ± SD; P = 0.0003). Mechanical allodynia and innocuous punctate stimulus-evoked laminae I to III neuronal activation can be replicated after intracisternally applied γ-aminobutyric acid receptor type A (GABAA) antagonist (bicuculline: 0.05 µg) or reactive oxygen species (ROS) donor (tert-butyl hydroperoxide: 50 to 250 ng). Conversely, intracisternal PKCγ antagonist, GABAA receptor agonist, or ROS scavenger prevent capsaicin-induced static mechanical allodynia and neuronal activation. CONCLUSIONS: Sensitization of lamina IIi PKCγ interneurons is required for the manifestation of secondary static mechanical allodynia but not for spontaneous pain. Such sensitization is driven by ROS and GABAAergic disinhibition. ROS released during intense C-fiber nociceptor activation might produce a GABAAergic disinhibition of PKCγ interneurons. Innocuous punctate inputs carried by Aδ low-threshold mechanoreceptors onto PKCγ interneurons can then gain access to the pain transmission circuitry of superficial MDH, producing pain.


Subject(s)
Hyperalgesia/physiopathology , Hyperalgesia/psychology , Interneurons , Nerve Fibers, Unmyelinated , Pain/physiopathology , Pain/psychology , Protein Kinase C/metabolism , Animals , Bicuculline/pharmacology , Capsaicin , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/chemically induced , Male , Mechanoreceptors/drug effects , Pain/chemically induced , Phosphorylation , Posterior Horn Cells , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, GABA-B/drug effects
9.
Neuron ; 87(4): 797-812, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26291162

ABSTRACT

Persistent mechanical hypersensitivity that occurs in the setting of injury or disease remains a major clinical problem largely because the underlying neural circuitry is still not known. Here we report the functional identification of key components of the elusive dorsal horn circuit for mechanical allodynia. We show that the transient expression of VGLUT3 by a discrete population of neurons in the deep dorsal horn is required for mechanical pain and that activation of the cells in the adult conveys mechanical hypersensitivity. The cells, which receive direct low threshold input, point to a novel location for circuit initiation. Subsequent analysis of c-Fos reveals the circuit extends dorsally to nociceptive lamina I projection neurons, and includes lamina II calretinin neurons, which we show also convey mechanical allodynia. Lastly, using inflammatory and neuropathic pain models, we show that multiple microcircuits in the dorsal horn encode this form of pain.


Subject(s)
Amino Acid Transport Systems, Acidic/biosynthesis , Hyperalgesia/metabolism , Nerve Net/metabolism , Pain/metabolism , Spinal Cord Dorsal Horn/metabolism , Touch , Animals , Hyperalgesia/pathology , Mice , Mice, Knockout , Nerve Net/pathology , Organ Culture Techniques , Pain/pathology , Spinal Cord Dorsal Horn/pathology
10.
Pain ; 156(9): 1714-1728, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25961142

ABSTRACT

Mechanical allodynia, a cardinal symptom of persistent pain, is associated with the unmasking of usually blocked local circuits within the superficial spinal or medullary dorsal horn (MDH) through which low-threshold mechanical inputs can gain access to the lamina I nociceptive output neurons. Specific interneurons located within inner lamina II (IIi) and expressing the gamma isoform of protein kinase C (PKCγ⁺) have been shown to be key elements for such circuits. However, their morphologic and electrophysiologic features are still unknown. Using whole-cell patch-clamp recordings and immunohistochemical techniques in slices of adult rat MDH, we characterized such lamina IIi PKCγ⁺ interneurons and compared them with neighboring PKCγ⁻ interneurons. Our results reveal that PKCγ⁺ interneurons display very specific activity and response properties. Compared with PKCγ⁻ interneurons, they exhibit a smaller membrane input resistance and rheobase, leading to a lower threshold for action potentials. Consistently, more than half of PKCγ⁺ interneurons respond with tonic firing to step current. They also receive a weaker excitatory synaptic drive. Most PKCγ⁺ interneurons express Ih currents. The neurites of PKCγ⁺ interneurons arborize extensively within lamina IIi, can spread dorsally into lamina IIo, but never reach lamina I. In addition, at least 2 morphologically and functionally different subpopulations of PKCγ⁺ interneurons can be identified: central and radial PKCγ⁺ interneurons. The former exhibit a lower membrane input resistance, rheobase and, thus, action potential threshold, and less PKCγ⁺ immunoreactivity than the latter. These 2 subpopulations might thus differently contribute to the gating of dorsally directed circuits within the MDH underlying mechanical allodynia.


Subject(s)
Interneurons/physiology , Medulla Oblongata/cytology , Membrane Potentials/physiology , Protein Kinase C/metabolism , Spinal Cord Dorsal Horn/cytology , Analysis of Variance , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials , Imaging, Three-Dimensional , In Vitro Techniques , Interneurons/classification , Male , Neurites , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
11.
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
12.
Pain ; 152(6): 1340-1348, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21392888

ABSTRACT

Glycine inhibitory dysfunction provides a useful experimental model for studying the mechanism of dynamic mechanical allodynia, a widespread and intractable symptom of neuropathic pain. In this model, allodynia expression relies on N-methyl-d-aspartate receptors (NMDARs), and it has been shown that astrocytes can regulate their activation through the release of the NMDAR coagonist d-serine. Recent studies also suggest that astrocytes potentially contribute to neuropathic pain. However, the involvement of astrocytes in dynamic mechanical allodynia remains unknown. Here, we show that after blockade of glycine inhibition, orofacial tactile stimuli activated medullary dorsal horn (MDH) astrocytes, but not microglia. Accordingly, the glia inhibitor fluorocitrate, but not the microglia inhibitor minocycline, prevented allodynia. Fluorocitrate also impeded activation of astrocytes and blocked activation of the superficial MDH neural circuit underlying allodynia, as revealed by study of Fos expression. MDH astrocytes are thus required for allodynia. They may also produce d-serine because astrocytic processes were selectively immunolabeled for serine racemase, the d-serine synthesizing enzyme. Accordingly, selective degradation of d-serine with d-amino acid oxidase applied in vivo prevented allodynia and activation of the underlying neural circuit. Conversely, allodynia blockade by fluorocitrate was reversed by exogenous d-serine. These results suggest the following scenario: removal of glycine inhibition makes tactile stimuli able to activate astrocytes; activated astrocytes may provide d-serine to enable NMDAR activation and thus allodynia. Such a contribution of astrocytes to pathological pain fuels the emerging concept that astrocytes are critical players in pain signaling. Glycine disinhibition makes tactile stimuli able to activate astrocytes, which may provide d-serine to enable NMDA receptor activation and thus allodynia.


Subject(s)
Citrates/therapeutic use , Glycine/metabolism , Hyperalgesia/drug therapy , Serine/metabolism , Analysis of Variance , Animals , Astrocytes/chemistry , Astrocytes/drug effects , CD11b Antigen/metabolism , Citrates/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Male , Oncogene Proteins v-fos/metabolism , Pain Measurement , Rats , Rats, Sprague-Dawley , Spinal Nerve Roots/cytology , Strychnine/therapeutic use , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...