1
|
Jubileum E, Binzen U, Treede RD, Greffrath W. Temporal and spatial summation of laser heat stimuli in cultured nociceptive neurons of the rat. Pflugers Arch 2022; 474:1003-1019. [PMID: 35867188 PMCID: PMC9393153 DOI: 10.1007/s00424-022-02728-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/04/2022]
Abstract
We studied the efficacy of a near-infrared laser (1475 nm) to activate rat dorsal root ganglion (DRG) neurons with short punctate radiant heat pulses (55 µm diameter) and investigated temporal and spatial summation properties for the transduction process for noxious heat at a subcellular level. Strength-duration curves (10–80 ms range) indicated a minimum power of 30.2mW for the induction of laser-induced calcium transients and a chronaxia of 13.9 ms. However, threshold energy increased with increasing stimulus duration suggesting substantial radial cooling of the laser spot. Increasing stimulus duration demonstrated suprathreshold intensity coding of calcium transients with less than linear gains (Stevens exponents 0.29/35mW, 0.38/60mW, 0.46/70mW). The competitive TRPV1 antagonist capsazepine blocked responses to short near-threshold stimuli and significantly reduced responses to longer duration suprathreshold heat. Heating 1/3 of the soma of a neuron was sufficient to induce calcium transients significantly above baseline (p < 0.05), but maximum amplitude was only achieved by centering the laser over the entire neuron. Heat-induced calcium increase was highest in heated cell parts but rapidly reached unstimulated areas reminiscent of spreading depolarization and opening of voltage-gated calcium channels. Full intracellular equilibrium took about 3 s, consistent with a diffusion process. In summary, we investigated transduction mechanisms for noxious laser heat pulses in native sensory neurons at milliseconds temporal and subcellular spatial resolution and characterized strength duration properties, intensity coding, and spatial summation within single neurons. Thermal excitation of parts of a nociceptor spread via both membrane depolarization and intracellular calcium diffusion.
Collapse
Affiliation(s)
- Elisabeth Jubileum
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.,Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, Rheinhessen Clinics, Hartmühlenweg 2-4, 55122, Mainz, Germany.,Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Uta Binzen
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.,Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Wolfgang Greffrath
- Department of Neurophysiology, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
2
|
Körner J, Lampert A. Functional subgroups of rat and human sensory neurons: a systematic review of electrophysiological properties. Pflugers Arch 2022; 474:367-385. [PMID: 35031856 PMCID: PMC8924089 DOI: 10.1007/s00424-021-02656-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/23/2021] [Accepted: 12/14/2021] [Indexed: 11/15/2022]
Abstract
Sensory neurons are responsible for the generation and transmission of nociceptive signals from the periphery to the central nervous system. They encompass a broadly heterogeneous population of highly specialized neurons. The understanding of the molecular choreography of individual subpopulations is essential to understand physiological and pathological pain states. Recently, it became evident that species differences limit transferability of research findings between human and rodents in pain research. Thus, it is necessary to systematically compare and categorize the electrophysiological data gained from human and rodent dorsal root ganglia neurons (DRGs). In this systematic review, we condense the available electrophysiological data defining subidentities in human and rat DRGs. A systematic search on PUBMED yielded 30 studies on rat and 3 studies on human sensory neurons. Defined outcome parameters included current clamp, voltage clamp, cell morphology, pharmacological readouts, and immune reactivity parameters. We compare evidence gathered for outcome markers to define subgroups, offer electrophysiological parameters for the definition of neuronal subtypes, and give a framework for the transferability of electrophysiological findings between species. A semiquantitative analysis revealed that for rat DRGs, there is an overarching consensus between studies that C-fiber linked sensory neurons display a lower action potential threshold, higher input resistance, a larger action potential overshoot, and a longer afterhyperpolarization duration compared to other sensory neurons. They are also more likely to display an infliction point in the falling phase of the action potential. This systematic review points out the need of more electrophysiological studies on human sensory neurons.
Collapse
Affiliation(s)
- Jannis Körner
- Institute of Physiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.,Clinic of Anesthesiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
3
|
Garami A, Shimansky YP, Rumbus Z, Vizin RCL, Farkas N, Hegyi J, Szakacs Z, Solymar M, Csenkey A, Chiche DA, Kapil R, Kyle DJ, Van Horn WD, Hegyi P, Romanovsky AA. Hyperthermia induced by transient receptor potential vanilloid-1 (TRPV1) antagonists in human clinical trials: Insights from mathematical modeling and meta-analysis. Pharmacol Ther 2020; 208:107474. [PMID: 31926897 DOI: 10.1016/j.pharmthera.2020.107474] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel alter body temperature (Tb) in laboratory animals and humans: most cause hyperthermia; some produce hypothermia; and yet others have no effect. TRPV1 can be activated by capsaicin (CAP), protons (low pH), and heat. First-generation (polymodal) TRPV1 antagonists potently block all three TRPV1 activation modes. Second-generation (mode-selective) TRPV1 antagonists potently block channel activation by CAP, but exert different effects (e.g., potentiation, no effect, or low-potency inhibition) in the proton mode, heat mode, or both. Based on our earlier studies in rats, only one mode of TRPV1 activation - by protons - is involved in thermoregulatory responses to TRPV1 antagonists. In rats, compounds that potently block, potentiate, or have no effect on proton activation cause hyperthermia, hypothermia, or no effect on Tb, respectively. A Tb response occurs when a TRPV1 antagonist blocks (in case of hyperthermia) or potentiates (hypothermia) the tonic TRPV1 activation by protons somewhere in the trunk, perhaps in muscles, and - via the acido-antithermogenic and acido-antivasoconstrictor reflexes - modulates thermogenesis and skin vasoconstriction. In this work, we used a mathematical model to analyze Tb data from human clinical trials of TRPV1 antagonists. The analysis suggests that, in humans, the hyperthermic effect depends on the antagonist's potency to block TRPV1 activation not only by protons, but also by heat, while the CAP activation mode is uninvolved. Whereas in rats TRPV1 drives thermoeffectors by mediating pH signals from the trunk, but not Tb signals, our analysis suggests that TRPV1 mediates both pH and thermal signals driving thermoregulation in humans. Hence, in humans (but not in rats), TRPV1 is likely to serve as a thermosensor of the thermoregulation system. We also conducted a meta-analysis of Tb data from human trials and found that polymodal TRPV1 antagonists (ABT-102, AZD1386, and V116517) increase Tb, whereas the mode-selective blocker NEO6860 does not. Several strategies of harnessing the thermoregulatory effects of TRPV1 antagonists in humans are discussed.
Collapse
Affiliation(s)
- Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
| | - Yury P Shimansky
- Department of Neurobiology, Barrow Neurological Institute, Dignity Health, Phoenix, AZ, USA
| | - Zoltan Rumbus
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Robson C L Vizin
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA
| | - Nelli Farkas
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Judit Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Zsolt Szakacs
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Margit Solymar
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Alexandra Csenkey
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Peter Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary; Department of Translational Medicine, First Department of Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andrej A Romanovsky
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA; School of Molecular Sciences, Arizona State University, Tempe, AZ, USA; Zharko Pharma Inc., Olympia, WA, USA.
| |
Collapse
|
4
|
The capsaicin receptor TRPV1 is the first line defense protecting from acute non damaging heat: a translational approach. J Transl Med 2020; 18:28. [PMID: 31952468 PMCID: PMC6966804 DOI: 10.1186/s12967-019-02200-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Pain is the vital sense preventing tissue damage by harmful noxious stimuli. The capsaicin receptor TRPV1 is activated by noxious temperatures, however, acute heat pain is only marginally affected in mice after TRPV1 knockout but completely eliminated in mice lacking TRPV1 positive fibers. Exploring contribution of candidate signal transduction mechanisms to heat pain in humans needs translational models. Methods We used focused, non-damaging, short near-infrared laser heat stimuli (wavelength 1470/1475 nm) to study the involvement of TRPV1-expressing nerve fibers in the encoding of heat pain intensity. Human psychophysics (both sexes) were compared to calcium transients in native rat DRG neurons and heterologously expressing HEK293 cells. Results Heating of dermal and epidermal nerve fibers in humans with laser stimuli of ≥ 2.5 mJ (≥ 25 ms, 100 mW) induced pain that increased linearly as a function of stimulus intensity in double logarithmic space across two orders of magnitude and was completely abolished by desensitization using topical capsaicin. In DRG neurons and TRPV1-expressing HEK cells, heat sensitivity was restricted to capsaicin sensitive cells. Strength duration curves (2–10 ms range) and thresholds (DRGs 0.56 mJ, HEK cells 0.52 mJ) were nearly identical. Tachyphylaxis upon repetitive stimulation occurred in HEK cells (54%), DRGs (59%), and humans (25%). Conclusion TRPV1-expressing nociceptors encode transient non-damaging heat pain in humans, thermal gating of TRPV1 is similar in HEK cells and DRG neurons, and TRPV1 tachyphylaxis is an important modulator of heat pain sensitivity. These findings suggest that TRPV1 expressed in dermal and epidermal populations of nociceptors serves as first line defense against heat injury.
Collapse
|
5
|
Abstract
Metastatic bone pain is a complex, poorly understood process. Understanding the unique mechanisms causing cancer-induced bone pain may lead to potential therapeutic targets. This article discusses the effects of osteoclast overstimulation within the tumor microenvironment; the role of inflammatory factors at the tumor-nociceptor interface; the development of structural instability, causing mechanical nerve damage; and, ultimately, the neuroplastic changes in the setting of sustained pain. Several adjuvant therapies are available to attenuate metastatic bone pain. This article discusses the role of pharmacologic therapies, surgery, kyphoplasty, vertebroplasty, and radiofrequency ablation.
Collapse
Affiliation(s)
- Nicholas Figura
- Department of Radiation Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Joshua Smith
- Department of Supportive Care Medicine, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Hsiang-Hsuan Michael Yu
- Department of Radiation Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
6
|
Abstract
Pain research is based broadly on physiological disciplines and its development follows the methodological progress of the era, from classical psychophysiology to electrophysiological investigations at peripheral and central nociceptive systems, single cells and ion channels to modern imaging of nociceptive processing. Physiological pain research in Germany has long been part of an interdisciplinary research network extending beyond all political boundaries, and this situation has continued since molecular techniques started to dominate all biomedical research. Current scientific questions, such as intracellular nociceptive signal mechanisms, interactions with other physiological systems including the immune system, or the genetic basis of epidemic and chronic pain diseases can only be solved interdisciplinary and with international collaboration.
Collapse
Affiliation(s)
- K Messlinger
- Institut für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Deutschland.
| | - H O Handwerker
- Institut für Physiologie und Pathophysiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstr. 17, 91054, Erlangen, Deutschland
| |
Collapse
|
7
|
Liu B, Qin F. Use Dependence of Heat Sensitivity of Vanilloid Receptor TRPV2. Biophys J 2016; 110:1523-1537. [PMID: 27074678 DOI: 10.1016/j.bpj.2016.03.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 02/24/2016] [Accepted: 03/07/2016] [Indexed: 11/30/2022] Open
Abstract
Thermal TRP channels mediate temperature transduction and pain sensation. The vanilloid receptor TRPV2 is involved in detection of noxious heat in a subpopulation of high-threshold nociceptors. It also plays a critical role in development of thermal hyperalgesia, but the underlying mechanism remains uncertain. Here we analyze the heat sensitivity of the TRPV2 channel. Heat activation of the channel exhibits strong use dependence. Prior heat activation can profoundly alter its subsequent temperature responsiveness, causing decreases in both temperature activation threshold and slope sensitivity of temperature dependence while accelerating activation time courses. Notably, heat and agonist activations differ in cross use-dependence. Prior heat stimulation can dramatically sensitize agonist responses, but not conversely. Quantitative analyses indicate that the use dependence in heat sensitivity is pertinent to the process of temperature sensing by the channel. The use dependence of TRPV2 reveals that the channel can have a dynamic temperature sensitivity. The temperature sensing structures within the channel have multiple conformations and the temperature activation pathway is separate from the agonist activation pathway. Physiologically, the use dependence of TRPV2 confers nociceptors with a hypersensitivity to heat and thus provides a mechanism for peripheral thermal hyperalgesia.
Collapse
Affiliation(s)
- Beiying Liu
- Department of Physiology/Biophysics, State University of New York-Buffalo, Buffalo, New York
| | - Feng Qin
- Department of Physiology/Biophysics, State University of New York-Buffalo, Buffalo, New York.
| |
Collapse
|
8
|
Wang X, Hu R, Liang J, Li Z, Sun W, Pan X. 5-HT7 Receptors Are Not Involved in Neuropeptide Release in Primary Cultured Rat Trigeminal Ganglion Neurons. J Mol Neurosci 2016; 59:251-9. [PMID: 26892478 DOI: 10.1007/s12031-016-0727-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 01/26/2016] [Indexed: 12/20/2022]
Abstract
Migraine is a common but complex neurological disorder. Its precise mechanisms are not fully understood. Increasing indirect evidence indicates that 5-HT7 receptors may be involved; however, their role remains unknown. Our previous in vivo study showed that selective blockade of 5-HT7 receptors caused decreased serum levels of calcitonin gene-related peptide (CGRP) in the external jugular vein following electrical stimulation of the trigeminal ganglion (TG) in an animal model of migraine. In the present study, we used an in vitro model of cultured TG cells to further investigate whether 5-HT7 receptors are directly responsible for the release of CGRP and substance P from TG neurons. We stimulated rat primary cultured TG neurons with capsaicin or potassium chloride (KCl) to mimic neurogenic inflammation, resulting in release of CGRP and substance P. 5-HT7 receptors were abundantly expressed in TG neurons. Greater than 93 % of 5-HT7 receptor-positive neurons co-expressed CGRP and 56 % co-expressed substance P. Both the capsaicin- and KCl-induced release of CGRP and substance P were unaffected by pretreatment of cultured TG cells with the selective 5-HT7 receptor agonist AS19 and antagonist SB269970. This study demonstrates for the first time that 5-HT7 receptors are abundantly co-expressed with CGRP and substance P in rat primary TG neurons and suggests that they are not responsible for the release of CGRP and substance P from cultured TG neurons evoked by capsaicin or KCl.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Department of Neurology, Guangzhou First People's Hospital, Affiliated to Guangzhou Medical University, No. 1 Panfu Road, Guangzhou, 510180, China
| | - Rong Hu
- Department of Neurology, Guangzhou First People's Hospital, Affiliated to Guangzhou Medical University, No. 1 Panfu Road, Guangzhou, 510180, China
| | - Jianbo Liang
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ze Li
- Department of Neurology, Guangzhou First People's Hospital, Affiliated to Guangzhou Medical University, No. 1 Panfu Road, Guangzhou, 510180, China
| | - Weiwen Sun
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neurosciences and the Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang Dong Road, Guangzhou, 510260, China
| | - Xiaoping Pan
- Department of Neurology, Guangzhou First People's Hospital, Affiliated to Guangzhou Medical University, No. 1 Panfu Road, Guangzhou, 510180, China.
| |
Collapse
|
9
|
Niederberger E, Kuner R, Geißlinger G. [Pharmacological aspects of pain research in Germany]. Schmerz 2015; 29:531-8. [PMID: 26294077 DOI: 10.1007/s00482-015-0042-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In spite of several approved analgesics, the therapy of pain still constitutes a challenge due to the fact that the drugs do not exert sufficient efficacy or are associated with severe side effects. Therefore, the development of new and improved painkillers is still of great importance. A number of highly qualified scientists in Germany are investigating signal transduction pathways in pain, effectivity of new drugs and the so far incompletely investigated mechanisms of well-known analgesics in preclinical and clinical studies. The highlights of pharmacological pain research in Germany are summarized in this article.
Collapse
Affiliation(s)
- E Niederberger
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Deutschland
| | - R Kuner
- Pharmakologisches Institut, Universität Heidelberg, Im Neuenheimer Feld 584, 69120, Heidelberg, Deutschland
| | - G Geißlinger
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Deutschland.
| |
Collapse
|
10
|
Kim MJ, Lee SY, Yang KY, Nam SH, Kim HJ, Kim YJ, Bae YC, Ahn DK. Differential regulation of peripheral IL-1β-induced mechanical allodynia and thermal hyperalgesia in rats. Pain 2014; 155:723-732. [PMID: 24406203 DOI: 10.1016/j.pain.2013.12.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 12/18/2013] [Accepted: 12/30/2013] [Indexed: 11/29/2022]
Abstract
This study examined the differential mechanisms of mechanical allodynia and thermal hyperalgesia after injection of interleukin (IL) 1β into the orofacial area of male Sprague-Dawley rats. The subcutaneous administration of IL-1β produced both mechanical allodynia and thermal hyperalgesia. Although a pretreatment with iodoresiniferatoxin (IRTX), a transient receptor potential vanilloid 1 (TRPV1) antagonist, did not affect IL-1β-induced mechanical allodynia, it significantly abolished IL-1β-induced thermal hyperalgesia. On the other hand, a pretreatment with D-AP5, an N-methyl-d-aspartate (NMDA) receptor antagonist, and NBQX, an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist, blocked IL-1β-induced mechanical allodynia. Pretreatment with H89, a protein kinase A (PKA) inhibitor, blocked IL-1β-induced mechanical allodynia but not thermal hyperalgesia. In contrast, pretreatment with chelerythrine, a protein kinase C (PKC) inhibitor, inhibited IL-1β-induced thermal hyperalgesia. Subcutaneous injections of 2% lidocaine, a local anesthetic agent, blocked IL-1β-induced thermal hyperalgesia but not IL-1β-induced mechanical allodynia. In the resiniferatoxin (RTX)-pretreated rats, a subcutaneous injection of IL-1β did not produce thermal hyperalgesia due to the depletion of TRPV1 in the primary afferent fibers. Double immunofluorescence revealed the colocalization of PKA with neurofilament 200 (NF200) and of PKC with the calcitonin gene-related peptide (CGRP) in the trigeminal ganglion. Furthermore, NMDA receptor 1 (NR1) and TRPV1 predominantly colocalize with PKA and PKC, respectively, in the trigeminal ganglion. These results suggest that IL-1β-induced mechanical allodynia is mediated by sensitized peripheral NMDA/AMPA receptors through PKA-mediated signaling in the large-diameter primary afferent nerve fibers, whereas IL-1β-induced thermal hyperalgesia is mediated by sensitized peripheral TRPV1 receptors through PKC-mediated signaling in the small-diameter primary afferent nerve fibers.
Collapse
Affiliation(s)
- Min J Kim
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea Department of Pediatric Dentistry, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea Department of Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Shin YH, Namkoong E, Choi S, Bae JS, Jin M, Hwang SM, Arote R, Choi SY, Park K. Capsaicin regulates the NF-κB pathway in salivary gland inflammation. J Dent Res 2013; 92:547-52. [PMID: 23603336 DOI: 10.1177/0022034513487376] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Salivary gland epithelial cells (SGEC) release several cytokines that play important roles in the inflammatory process. In this study, we examined whether capsaicin can modulate cytokine release in SGEC. After cells were stimulated with polyinosinic-polycytidylic acid [poly(I:C)] or lipopolysaccharide (LPS), mRNA transcript and protein levels were detected by reverse-transcriptase-polymerase chain-reaction (RT-PCR), real-time PCR, and enzyme-linked immunosorbent assay (ELISA). These findings demonstrated that the increases in TNFα and IL-6 mRNA transcripts were highest at 3 hrs and 1 hr after incubation with poly(I:C) and LPS, respectively. Pre-treatment of the cells with 10 μµ capsaicin, however, significantly inhibited mRNA transcripts and its protein levels. The simultaneous application of 10 μµ capsazepine with capsaicin did not block the inhibitory effect of capsaicin. Furthermore, the inhibitory effect of capsaicin was also shown in primary cultured cells from TRPV1(-/-) mice. We found that both poly(I:C) and LPS induced IκB-α degradation and phosphorylation, which resulted in NF-κB activation, and capsaicin inhibited this NF-κB pathway. These results demonstrate that SGEC release pro-inflammatory cytokines mediated by TLR, and capsaicin inhibits this process through the NF-κB pathway. This study suggests that capsaicin could potentially alleviate inflammation in salivary glands.
Collapse
Affiliation(s)
- Y-H Shin
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhu YJ, Lu TJ. A multi-scale view of skin thermal pain: from nociception to pain sensation. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2010; 368:521-559. [PMID: 20047938 DOI: 10.1098/rsta.2009.0234] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
All biological bodies live in a thermal environment, including the human body, where skin is the interface with a protecting function. When the temperature is out of the normal physiological range, skin fails to protect, and the pain sensation is evoked. Furthermore, in medicine, with advances in laser, microwave and similar technologies, various thermal therapeutic methods have been widely used to cure disease/injury involving skin tissue. However, the corresponding problem of pain relief has limited further application and development of these thermal treatments. Skin thermal pain is induced through both direct (i.e. an increase/decrease in temperature) and indirect (e.g. thermomechanical and thermochemical) ways, and is governed by complicated thermomechanical-chemical-neurophysiological responses. However, a complete understanding of the underlying mechanisms is still far from clear. In this article, starting from an engineering perspective, we aim to recast the biological behaviour of skin in engineering system parlance. Then, by coupling the concepts of engineering with established methods in neuroscience, we attempt to establish multi-scale modelling of skin thermal pain through ion channel to pain sensation. The model takes into account skin morphological plausibility, the thermomechanical response of skin tissue and the biophysical and neurological mechanisms of pain sensation.
Collapse
Affiliation(s)
- Y J Zhu
- Stomatological Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | | |
Collapse
|
13
|
Greffrath W, Schwarz ST, Büsselberg D, Treede RD. Heat-induced action potential discharges in nociceptive primary sensory neurons of rats. J Neurophysiol 2009; 102:424-36. [PMID: 19439674 DOI: 10.1152/jn.90916.2008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although several transducer molecules for noxious stimuli have been identified, little is known about the transformation of the resulting generator currents into action potentials (APs). Therefore we investigated the transformation process for stepped noxious heat stimuli (42-47 degrees C, 3-s duration) into membrane potential changes and subsequent AP discharges using the somata of acutely dissociated small dorsal root ganglion (DRG) neurons (diameter<or=32.5 microm) of adult rats as a model for their own peripheral terminals. Three types of heat-induced membrane potential changes were differentiated: type 1, heat-induced AP discharges (approximately 37% of the neurons); type 2, heat-induced membrane depolarization (40%); and type 3, responses not exceeding those of switching the superfusion (23%). Warming neurons from room temperature to 35 degrees C increased their background conductance, nearly doubled the AP threshold current, and led to smaller and narrower APs. Adaptation of heat-induced AP discharges was seen in about half of the type 1 neurons. The remaining half displayed accelerating discharges to both heat stimuli and depolarizing current injection. Repeated heat stimulation induced marked suppression of AP discharges. Under rapid calcium buffering using BAPTA, repolarization of heat-induced APs stopped at a plateau potential slowly decreasing from +16.5+/-2.9 to -2.2+/-5.5 mV, resulting in no further AP discharges. This study demonstrates that heat-induced AP discharges can be elicited in the soma of a subgroup of DRG neurons. These discharges display suppression on repetitive stimulation, but either adaptation or sensitization during prolonged stimuli. AP threshold and AP shape during these discharges suggest temperature dependence of background conductance and repolarizing currents.
Collapse
Affiliation(s)
- Wolfgang Greffrath
- Division of Neurophysiology, Center of Biomedicine and Medical Technology Mannheim, CBTM Ludolf-Krehl-Strasse 13-17, Medical Faculty Mannheim, Ruprecht-Karls-University Heidelberg, D-68167 Mannheim, Germany.
| | | | | | | |
Collapse
|
14
|
St. Pierre M, Reeh PW, Zimmermann K. Differential effects of TRPV channel block on polymodal activation of rat cutaneous nociceptors in vitro. Exp Brain Res 2009; 196:31-44. [DOI: 10.1007/s00221-009-1808-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 04/07/2009] [Indexed: 02/02/2023]
|
15
|
Relationship between oral burn and temperature in chili spiced pork patties evaluated by time–intensity. Food Qual Prefer 2009. [DOI: 10.1016/j.foodqual.2008.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
16
|
Challenges in cancer pain management–bone pain. Eur J Cancer 2008; 44:1083-90. [DOI: 10.1016/j.ejca.2008.03.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 03/06/2008] [Indexed: 11/21/2022]
|
17
|
Tender GC, Li YY, Cui JG. Vanilloid receptor 1-positive neurons mediate thermal hyperalgesia and tactile allodynia. Spine J 2008; 8:351-8. [PMID: 18029293 DOI: 10.1016/j.spinee.2007.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 04/11/2007] [Accepted: 08/27/2007] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT The vanilloid receptor 1 (VR1) is expressed by the type II A-delta and C-fiber neurons, functioning as a molecular integrator for nociception. VR1 can be selectively ablated by resiniferatoxin (RTX), an ultra-potent excitotoxic agonist, when injected into sensory ganglia. PURPOSE To evaluate the role of the VR1-positive neurons in neuropathic pain. STUDY DESIGN Photochemical injury to rat sciatic nerve (Gazelius model). METHODS Two groups of rats underwent the photochemical injury and RTX treatment. RTX was injected in the dorsal root ganglia (DRGs) of the L3, L4, L5, and L6 nerve roots, either after or before the nerve injury. The animals were tested for thermal hyperalgesia (noxious heat stimuli) and mechanical allodynia (von Frey filaments). Immunohistochemical analysis of the DRGs was performed after euthanasia. RESULTS In the tactile allodynic rats, RTX injection in the DRGs improved the average withdrawal threshold from 1.62 g to 5.68 g. Immunohistochemical labeling showed that almost all VR1-positive neurons were eliminated. When RTX was administrated into the ipsilateral DRGs before the nerve injury, this treatment prevented the development of tactile allodynia in 12 out of 14 rats. Immunohistochemical staining revealed that the VR1-positive neurons were eliminated in the rats that did not develop tactile allodynia, whereas they were still present in the allodynic rats. CONCLUSIONS VR1-positive neurons are essential for the development of mechanical allodynia. In rats already exhibiting neuropathic pain, the VR1-positive neurons mediate the most sensitive part of mechanical allodynia. RTX injection in sensory ganglia may represent a novel treatment for neuropathic pain.
Collapse
Affiliation(s)
- Gabriel C Tender
- Department of Neurosurgery, Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
18
|
Hagenacker T, Ledwig D, Büsselberg D. Feedback mechanisms in the regulation of intracellular calcium ([Ca2+]i) in the peripheral nociceptive system: role of TRPV-1 and pain related receptors. Cell Calcium 2007; 43:215-27. [PMID: 17673288 DOI: 10.1016/j.ceca.2007.05.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 05/17/2007] [Accepted: 05/30/2007] [Indexed: 11/21/2022]
Abstract
Multimodal stimuli like heat, cold, bacterial or mechanical events are able to elicit pain, which is necessary to guarantee survival. However, the control of pain is of major clinical importance. The perception and transduction of pain is differentially modulated in the peripheral and central nervous system (CNS): while peripheral structures modulate these signals, the perception of pain occurs in the CNS. In recent years major advances have been made in the understanding of the processes which are involved in pain sensation. For the peripheral pain reception, the importance of specific pain receptors of the transition receptor pore (TRP)-family (e.g. the TRPV-1 receptor) has been analyzed. These receptors/channels are localized at the cell membrane of nociceptive neurones as well as in membranes of intracellular calcium stores like the endoplasmic reticulum. While the associated channel conducts different ions, a major proportion is calcium. Therefore, this review focuses on (1) the modulations of intracellular calcium ([Ca2+]i) initiated by the activation of pain receptors and (2) the consequences of [Ca2+]i changes for the processing of pain signals at the peripheral side. The possible interference of TRPV-1 induced [Ca2+]i modulations to the function of other membrane receptors and channels, like voltage gated calcium, sodium or potassium channels, or co-expressed CB1-receptors will be discussed. The latter interactions are of specific interest since the analgetic properties of endo- and exo-cannabinoids are mediated by CB1 receptors and their activation significantly modulates the calcium induced release of pain related transmitters. Furthermore, multiple cross links between different pain modulating intracellular pathways and their dependence on [Ca2+]i modulations will be illuminated. Overall, this review will summarize new insights resulting in the understanding of the prominent influence of [Ca2+]i for processes which are involved in pain sensation.
Collapse
Affiliation(s)
- T Hagenacker
- Universitätsklinikum Essen, Institut für Physiologie, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | |
Collapse
|
19
|
Rau KK, Jiang N, Johnson RD, Cooper BY. Heat sensitization in skin and muscle nociceptors expressing distinct combinations of TRPV1 and TRPV2 protein. J Neurophysiol 2007; 97:2651-62. [PMID: 17287441 DOI: 10.1152/jn.00840.2006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recordings were made from small and medium diameter dorsal root ganglia (DRG) neurons that expressed transient receptor potential (TRP) proteins. Physiologically characterized skin nociceptors expressed either TRPV1 (type 2) or TRPV2 (type 4) in isolation. Other nociceptors co-expressed both TRP proteins and innervated deep tissue sites (gastrocnemius muscle, distal colon; type 5, type 8) and skin (type 8). Subpopulations of myelinated (type 8) and unmyelinated (type 5) nociceptors co-expressed both TRPs. Cells that expressed TRPV1 were excellent transducers of intense heat. Proportional inward currents were obtained from a threshold of approximately 46.5 to approximately 56 degrees C. In contrast, cells expressing TRPV2 alone (52 degrees C threshold) did not reliably transduce the intensity of thermal events. Studies were undertaken to assess the capacity of skin and deep nociceptors to exhibit sensitization to repeated intense thermal stimuli [heat-heat sensitization (HHS)]. Only nociceptors that expressed TRPV2, alone or in combination with TRPV1, exhibited HHS. HHS was shown to be Ca(2+) dependent in either case. Intracellular Ca(2+) dependent pathways to HHS varied with the pattern of TRP protein expression. Cells co-expressing both TRPs modulated heat reactivity through serine/threonine phosphorylation or PLA(2)-dependent pathways. Cells expressing only TRPV2 may have relied on tyrosine kinases for HHS. We conclude that heat sensitization in deep and superficial capsaicin and capsaicin-insensitive C and Adelta nociceptors varies with the distribution of TRPV1 and TRPV2 proteins. The expression pattern of these proteins are specific to subclasses of physiologically identified C and A fiber nociceptors with highly restricted tissue targets.
Collapse
MESH Headings
- Animals
- Capsaicin/analogs & derivatives
- Capsaicin/pharmacology
- Colon/innervation
- Colon/metabolism
- Colon/physiology
- Histocytochemistry
- Hot Temperature
- Male
- Muscle, Skeletal/innervation
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiology
- Muscle, Smooth/innervation
- Muscle, Smooth/metabolism
- Muscle, Smooth/physiology
- Muscles/innervation
- Muscles/metabolism
- Muscles/physiology
- Nerve Fibers, Myelinated/physiology
- Nerve Fibers, Unmyelinated/physiology
- Neurofilament Proteins/biosynthesis
- Neurons, Afferent/drug effects
- Nociceptors/metabolism
- Nociceptors/physiology
- Patch-Clamp Techniques
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/physiology
- Skin/innervation
- Skin/metabolism
- TRPV Cation Channels/biosynthesis
Collapse
Affiliation(s)
- K K Rau
- Dept. of Oral Surgery and Diagnostic Sciences, Div. of Neuroscience, Box 100416, JHMHC, Univ. of Florida College of Dentistry, Gainesville, FL 32610, USA
| | | | | | | |
Collapse
|
20
|
Firner M, Greffrath W, Treede RD. Phosphorylation of extracellular signal-related protein kinase is required for rapid facilitation of heat-induced currents in rat dorsal root ganglion neurons. Neuroscience 2006; 143:253-63. [PMID: 16973292 DOI: 10.1016/j.neuroscience.2006.07.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Revised: 06/20/2006] [Accepted: 07/23/2006] [Indexed: 12/29/2022]
Abstract
A subgroup of dorsal root ganglion (DRG) neurons responds to noxious heat with an influx of cations carried by specific ion channels such as the transient receptor potential channel of the vanilloid receptor type, subtype 1 (TRPV1). Application of capsaicin induces a reversible facilitation of these currents. This facilitation could be an interaction of two agonists at their common receptor or be caused by an influx of calcium ions into the cell. Calcium influx into the cell can activate protein kinases such as the extracellular signal-related protein kinase (ERK) pathway. This study explored the kinetics, calcium-dependency and intracellular signals following application of capsaicin and leading to facilitation of heat-induced currents (Iheat) in rat DRG neurons. Application of 0.5 microM capsaicin caused a 2.65-fold increase of Iheat within 2 s, which was significantly correlated to a small capsaicin-induced current. Intracellular application of 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA), a fast calcium chelator, did not change capsaicin-induced currents or Iheat itself, but inhibited facilitation of Iheat by capsaicin. ERK is activated by calcium influx and membrane depolarization via the mitogen-activated protein kinase/extracellular signal-related protein kinase kinase (MEK). Application of the MEK inhibitor U0126 also inhibited facilitation of Iheat by capsaicin. We conclude that the MEK/ERK cascade is an intracellular signaling pathway playing a vital role in the regulation of nociceptive neurons' sensitivity. The very fast kinetics (less than two seconds) are only explainable with a membrane-attached or at least membrane-near localization of these kinases.
Collapse
Affiliation(s)
- M Firner
- Institute of Physiology and Pathophysiology, Johannes Gutenberg University, Saarstrasse 21, D-55099 Mainz, Germany
| | | | | |
Collapse
|
21
|
Abstract
The capsaicin receptor TRPV1, once discovered as a receptor for pungent spices, is a polymodal sensor molecule for painful chemical and thermal stimuli. However, TRPV1 plays an important role not only for the integration of acute painful stimuli but also in the genesis of inflammatory processes. The persistent functional sensitization of TRPV1 as well as an up-regulation of its expression may contribute to the development and maintenance of chronic pain states. Thus, TRPV1 is an excellent target for a rational pharmacological treatment of pain. Several additional physiological and pathophysiological functions of TRPV1 are assumed beyond nociception and pain. Activation of TRPV1 seems to contribute to the etiology and pathogenesis of inflammatory diseases concerning, e.g., the gastrointestinal tract, the bladder, and the respiratory system. Therefore, the therapeutic potential of a pharmacological manipulation of TRPV1 may not be restricted to a symptomatic therapy of pain.
Collapse
Affiliation(s)
- W Greffrath
- Institut für Physiologie und Pathophysiologie, Johannes-Gutenberg-Universität Mainz.
| |
Collapse
|
22
|
Tomaszewski A, Büsselberg D. Cisplatin modulates voltage gated channel currents of dorsal root ganglion neurons of rats. Neurotoxicology 2006; 28:49-58. [PMID: 16945417 DOI: 10.1016/j.neuro.2006.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 07/13/2006] [Accepted: 07/13/2006] [Indexed: 10/24/2022]
Abstract
The anticancer drug cis-diammindichloroplatin (CDDP, cisplatin) causes severe side effects like peripheral sensitive neuropathy. The toxicity of CDDP has been linked to changes in intracellular calcium homeostasis ([Ca2+]i). Voltage activated calcium channel currents (ICa(V)) are important for the regulation of [Ca2+]i; therefore, this study was designed to examine the effect of CDDP on ICa(V) in comparison to voltage activated potassium (IK(V)) and sodium (INa(V)) channel currents using the whole cell patch clamp method on dorsal root ganglion neurons of rats. In small neurons (<or=Ø20 microm) CDDP reduced peak and sustained ICa(V) concentration dependently (1-100 microM). The IC50 was 23.9+/-4.5 microM (+/-S.D.) for the peak current with a Hill-coefficient of 0.6+/-0.1 and 38.8+/-6.1 microM for the sustained current (Hill-coefficient: 0.7+/-0.1). IK(V) were reduced by 20.9+/-4.8% (10 microM) and INa(V) were only reduced by 9.2%+/-7.2% (10 microM). ICa(V) of large neurons (>or=Ø25 microm) were less sensitive to CDDP. The peak ICa(V) was reduced by 14.1+/-2.3% and IK(V) by 12.8+/-3.4% (100 microM). The sensitivity of INa(V) in large neurons to CDDP was not different compared to small neurons. We conclude that the reduction of ICa(V) in small cells may be responsible for the neurotoxic side effects CDDP causes in sensory neurons.
Collapse
Affiliation(s)
- Anke Tomaszewski
- Universität Duisburg-Essen, Universitätsklinik, Institut für Physiologie, Hufelandstrasse 55, 45122 Essen, Germany
| | | |
Collapse
|
23
|
Simonetti M, Fabbro A, D'Arco M, Zweyer M, Nistri A, Giniatullin R, Fabbretti E. Comparison of P2X and TRPV1 receptors in ganglia or primary culture of trigeminal neurons and their modulation by NGF or serotonin. Mol Pain 2006; 2:11. [PMID: 16566843 PMCID: PMC1448213 DOI: 10.1186/1744-8069-2-11] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 03/28/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cultured sensory neurons are a common experimental model to elucidate the molecular mechanisms of pain transduction typically involving activation of ATP-sensitive P2X or capsaicin-sensitive TRPV1 receptors. This applies also to trigeminal ganglion neurons that convey pain inputs from head tissues. Little is, however, known about the plasticity of these receptors on trigeminal neurons in culture, grown without adding the neurotrophin NGF which per se is a powerful algogen. The characteristics of such receptors after short-term culture were compared with those of ganglia. Furthermore, their modulation by chronically-applied serotonin or NGF was investigated. RESULTS Rat or mouse neurons in culture mainly belonged to small and medium diameter neurons as observed in sections of trigeminal ganglia. Real time RT-PCR, Western blot analysis and immunocytochemistry showed upregulation of P2X(3) and TRPV1 receptors after 1-4 days in culture (together with their more frequent co-localization), while P2X(2) ones were unchanged. TRPV1 immunoreactivity was, however, lower in mouse ganglia and cultures. Intracellular Ca(2+) imaging and whole-cell patch clamping showed functional P2X and TRPV1 receptors. Neurons exhibited a range of responses to the P2X agonist alpha, beta-methylene-adenosine-5'-triphosphate indicating the presence of homomeric P2X(3) receptors (selectively antagonized by A-317491) and heteromeric P2X(2/3) receptors. The latter were observed in 16 % mouse neurons only. Despite upregulation of receptors in culture, neurons retained the potential for further enhancement of P2X(3) receptors by 24 h NGF treatment. At this time point TRPV1 receptors had lost the facilitation observed after acute NGF application. Conversely, chronically-applied serotonin selectively upregulated TRPV1 receptors rather than P2X(3) receptors. CONCLUSION Comparing ganglia and cultures offered the advantage of understanding early adaptive changes of nociception-transducing receptors of trigeminal neurons. Culturing did not prevent differential receptor upregulation by algogenic substances like NGF or serotonin, indicating that chronic application led to distinct plastic changes in the molecular mechanisms mediating pain on trigeminal nociceptors.
Collapse
Affiliation(s)
- Manuela Simonetti
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| | - Alessandra Fabbro
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| | - Marianna D'Arco
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| | - Marina Zweyer
- Department of Normal Human Morphology, University of Trieste, Via Manzoni 16, 34138 Trieste, Italy
| | - Andrea Nistri
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| | - Rashid Giniatullin
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| | - Elsa Fabbretti
- Neurobiology Sector, International School for Advanced Studies (SISSA), Via Beirut 4, 34014 Trieste, Italy
| |
Collapse
|
24
|
Chapter 4 TRPV1: A Polymodal Sensor in the Nociceptor Terminal. CURRENT TOPICS IN MEMBRANES 2006. [DOI: 10.1016/s1063-5823(06)57003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
25
|
Hagenacker T, Splettstoesser F, Greffrath W, Treede RD, Büsselberg D. Capsaicin differentially modulates voltage-activated calcium channel currents in dorsal root ganglion neurones of rats. Brain Res 2005; 1062:74-85. [PMID: 16269136 DOI: 10.1016/j.brainres.2005.09.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Revised: 09/16/2005] [Accepted: 09/25/2005] [Indexed: 11/30/2022]
Abstract
It is discussed whether capsaicin, an agonist of the pain mediating TRPV1 receptor, decreases or increases voltage-activated calcium channel (VACC) currents (I(Ca(V))). I(Ca(V)) were isolated in cultured dorsal root ganglion (DRG) neurones of rats using the whole cell patch clamp method and Ba2+ as charge carrier. In large diameter neurones (>35 micorm), a concentration of 50 microM was needed to reduce I(Ca(V)) (activated by depolarizations to 0 mV) by 80%, while in small diameter neurones (< or =30 microm), the IC50 was 0.36 microM. This effect was concentration dependent with a threshold below 0.025 microM and maximal blockade (>80%) at 5 microM. The current-voltage relation was shifted to the hyperpolarized direction with an increase of the current between -40 and -10 mV and a decrease between 0 and +50 mV. Isolation of L-, N-, and T-type calcium channels resulted in differential effects when 0.1 microM capsaicin was applied. While T-type channel currents were equally reduced over the voltage range, L-type channel currents were additionally shifted to the hyperpolarized direction by 10 to 20 mV. N-type channel currents expressed either a shift (3 cells) or a reduction of the current (4 cells) or both (3 cells). Thus, capsaicin increases I(Ca(V)) at negative and decreases I(Ca(V)) at positive voltages by differentially affecting L-, N-, and T-type calcium channels. These effects of capsaicin on different VACCs in small DRG neurones, which most likely express the TRPV1 receptor, may represent another mechanism of action of the pungent substance capsaicin in addition to opening of TRPV1.
Collapse
Affiliation(s)
- Tim Hagenacker
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | | | | | |
Collapse
|
26
|
Lu G, Henderson D, Liu L, Reinhart PH, Simon SA. TRPV1b, a functional human vanilloid receptor splice variant. Mol Pharmacol 2005; 67:1119-27. [PMID: 15644492 DOI: 10.1124/mol.104.009852] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Transient receptor potential (TRP) genes encode a family of related ion-channel subunits. This family consists of cation-selective, calcium-permeable channels that include a group of vanilloid receptor channels (TRPV) implicated in pain and inflammation. These channels are activated by diverse stimuli, including capsaicin, lipids, membrane deformation, heat, and protons. Six members of the TRPV family have been identified that differ predominantly in their activation properties. However, in neurons, TRPV channels do not account for the observed diversity of responses to activators. By probing human and rat brain cDNA libraries to identify TRPV subunits, we identified a novel human TRPV1 RNA splice variant, TRPV1b, which forms functional ion channels that are activated by temperature (threshold, approximately 47 degrees C), but not by capsaicin or protons. Channels with similar activation properties were found in trigeminal ganglion neurons, suggesting that TRPV1b receptors are expressed in these cells and contribute to thermal nociception.
Collapse
Affiliation(s)
- Gang Lu
- Box 3209, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
27
|
Pethö G, Izydorczyk I, Reeh PW. Effects of TRPV1 receptor antagonists on stimulated iCGRP release from isolated skin of rats and TRPV1 mutant mice. Pain 2004; 109:284-290. [PMID: 15157689 DOI: 10.1016/j.pain.2004.01.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2003] [Accepted: 01/12/2004] [Indexed: 11/26/2022]
Abstract
Capsaicin antagonists including ruthenium red, capsazepine and iodo-resiniferatoxin (I-RTX) have recently been shown to inhibit the activation by noxious heat of the capsaicin receptor (TRPV1) expressed in non-neuronal host cells, and natively, in cultured dorsal root ganglion cells. Noxious heat has been shown to release immunoreactive calcitonin gene-related peptide (iCGRP) from the isolated rat skin. In this model, ruthenium red, I-RTX as well as capsazepine 10 microM caused no alteration in iCGRP release at 32 degrees C by themselves whereas capsazepine 100 microM doubled it reversibly. In wild-type mice 100 microM capsazepine also stimulated iCGRP release while it was without effect in TRPV1 knockout littermates. In the rat skin, both ruthenium red and capsazepine (10/100 microM) reduced and abolished, respectively, capsaicin-induced iCGRP release while I-RTX (1/10 microM) was ineffective. Only ruthenium red 100 microM showed an unspecific effect inhibiting iCGRP release induced by KCl. Ruthenium red and capsazepine (10/100 microM) caused no significant alteration of iCGRP release induced by heat stimulation at 47 degrees C. Employing 45 degrees C stimulation intensity, capsazepine and I-RTX (in the higher concentrations) showed a significant facilitatory effect on the heat response suggesting a partial agonistic action of the compounds. It is concluded that noxious heat-induced iCGRP release in the isolated rat skin occurs through a mechanism that is not inhibited by TRPV1 antagonism reflecting a different pharmacological profile of noxious heat transduction in terminals of sensory neurons compared to that in cultured cell bodies and TRPV1-transfected host cells.
Collapse
Affiliation(s)
- Gábor Pethö
- Institute of Physiology and Experimental Pathophysiology, University Erlangen/Nürnberg, Universitaetsstr. 17, D-91054 Erlangen, Germany
| | | | | |
Collapse
|
28
|
Greffrath W, Binzen U, Schwarz ST, Saaler-Reinhardt S, Treede RD. Co-expression of heat sensitive vanilloid receptor subtypes in rat dorsal root ganglion neurons. Neuroreport 2004; 14:2251-5. [PMID: 14625457 DOI: 10.1097/00001756-200312020-00023] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Expression of the heat sensitive cation channels TRPV1 and TRPV2 was investigated by immunofluorescence in rat dorsal root ganglion (DRG) neurons. TRPV1-positive neurons were more frequent and had smaller diameters than TRPV2-positive neurons (35.7% vs 7.3%; 22.3 microm vs 27.6 microm), but size distributions overlapped and significant co-expression was seen in 20.7% of TRPV2-positive neurons (1.7% of all). Expression patterns did not differ between tissue sections typically used in immunocytochemistry and dissociated DRG neurons typically used in electrophysiology. Rectangular temperature pulses revealed two patterns of heat-evoked inward currents in small DRG neurons: low-threshold rapidly activating and high-threshold slowly activating. Slowly activating heat-evoked currents have not been described previously, but correspond to the type I heat responses of primary nociceptive afferents, which have been suggested to be mediated by TRPV2.
Collapse
Affiliation(s)
- Wolfgang Greffrath
- Institute of Physiology and Pathophysiology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | |
Collapse
|
29
|
Khasabova IA, Harding-Rose C, Simone DA, Seybold VS. Differential effects of CB1 and opioid agonists on two populations of adult rat dorsal root ganglion neurons. J Neurosci 2004; 24:1744-53. [PMID: 14973253 PMCID: PMC6730464 DOI: 10.1523/jneurosci.4298-03.2004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inhibition of primary afferent neurons contributes to the antihyperalgesic effects of opioid and CB1 receptor agonists. Two bioassays were used to compare the effects of the CB1 receptor agonist CP 55,940 and morphine on dissociated adult rat DRG neurons. Both agonists inhibited the increase in free intracellular Ca2+ concentration evoked by depolarization; however, effects of CP 55,940 occurred primarily in large neurons (cell area, >800 microm2), whereas morphine inhibited the response in smaller neurons. Cotreatment with selective blockers of L-, N-, and P/Q-type voltage-dependent Ca2+ channels indicated that CB1 receptors on DRG neurons couple solely with N-type channels but opioid receptors couple with multiple subtypes. Experiments with selective agonists and antagonists of opioid receptors indicated that mu and delta, but not kappa, receptors contributed to the inhibitory effect of morphine on voltage-dependent Ca2+ influx. Because Ca2+ channels underlie release of transmitters from neurons, the effects of opioid agonists and CP 55,940 on depolarization-evoked release of calcitonin gene-related peptide (CGRP) were compared. Morphine inhibited release through delta receptors but CP 55,940 had no effect. Colocalization of CGRP with delta-opioid but not mu-opioid or CB1 receptor immunoreactivity in superficial laminae of the dorsal horn of the spinal cord was consistent with the data for agonist inhibition of peptide release. Therefore, CB1 and opioid agonists couple with different voltage-dependent Ca2+ channels in different populations of DRG neurons. Furthermore, differences occur in the distribution of receptors between the cell body and terminals of DRG neurons. The complementary action of CB1 and opioid receptor agonists on populations of DRG neurons provides a rationale for their combined use in modulation of somatosensory input to the spinal cord.
Collapse
Affiliation(s)
- I A Khasabova
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
30
|
Caudle RM, Karai L, Mena N, Cooper BY, Mannes AJ, Perez FM, Iadarola MJ, Olah Z. Resiniferatoxin-induced loss of plasma membrane in vanilloid receptor expressing cells. Neurotoxicology 2004; 24:895-908. [PMID: 14637384 DOI: 10.1016/s0161-813x(03)00146-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Resiniferatoxin (RTX), a potent analog of capsaicin, was evaluated electrophysiologically in dorsal root ganglion (DRG) cells and cell lines ectopically expressing the vanilloid receptor type 1 (VR1) to determine if cell phenotype influenced RTXs neurotoxic properties. Furthermore, capsaicin and heat activation of VR1 were evaluated in these cells to determine if cellular damage was unique to RTX activation of the receptors. RTX application to DRG cells identified as type 1, 2 or 5, cell types known to express VR1, induced large inward currents. RTX did not induce currents in DRG cells that do not express the receptor (type 4 cells). In cell lines ectopically expressing VR1, RTX-induced similar currents. RTX produced no effect in non-transfected cells. After exposure to RTX both DRG cells and transfected cells failed to respond to subsequent applications of the agonist. In addition, whole cell capacitance was reduced up to 70%. The decrease in capacitance was associated with the loss of plasma membrane, as determined by confocal microscopy. Cell phenotype, other than VR1 expression, did not influence the response to RTX. Interestingly, capsaicin and heat activation of vanilloid receptors also decreased cell capacitance, but the loss of membrane was not as great as with RTX and responses to these stimuli were not lost after the initial exposure. The loss of cell membrane required elevated intracellular levels of Ca2+. From these data it was concluded that the loss of cell membrane was dependent on the presence of both VR1 and intracellular Ca2+ accumulation, but not on cell phenotype.
Collapse
Affiliation(s)
- Robert M Caudle
- Department of Oral and Maxillofacial Surgery, University of Florida College of Dentistry and the University of Florida McKnight Brain Institute, 1600 Archer Road, P.O. Box 100416, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Truong H, McGinnis L, Dindo L, Honda CN, Giesler GJ. Identification of dorsal root ganglion neurons that innervate the common bile duct of rats. Exp Brain Res 2003; 155:477-84. [PMID: 14689139 DOI: 10.1007/s00221-003-1753-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2003] [Accepted: 10/03/2003] [Indexed: 10/26/2022]
Abstract
Pain originating in the bile duct is common and many patients who have suffered from it report that it is one of the most intense forms of pain that they have experienced. Many uncertainties remain about the mechanisms underlying pain originating in the bile duct. For example, the dorsal root ganglion (DRG) neurons that give rise to the sensory innervation of the common bile duct (CBD) have not been identified and examined in any species. The goal of the present study was to determine the number, distribution, and size of DRG neurons that innervate the CBD in rats. Injections of WGA-HRP or CTB-HRP were restricted to the lumen of the bile duct. Injections of WGA-HRP labeled a mean number of about 500 DRG neurons bilaterally throughout all thoracic and upper lumbar levels. Injections of CTB-HRP labeled smaller numbers of DRG neurons. Application of colchicine onto the surface of the CBD reduced the number of cells labeled following injections of WGA-HRP into the lumen of the CBD by roughly 86%, suggesting that tracer had not spread in large amounts out of the CBD and labeled afferent fibers in other tissues. Approximately 85% of the neurons labeled with WGA-HRP had cell bodies that were classified as small; the remainder were medium in size. Injections of CTB-HRP labeled cell bodies of varying sizes, including a few large diameter cell bodies. These results indicate that a large number of primarily small DRG cells, located bilaterally at many segmental levels, provide a rich innervation of the common bile duct.
Collapse
Affiliation(s)
- H Truong
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
32
|
An intact intermediate filament network is required for collateral sprouting of small diameter nerve fibers. J Neurosci 2003. [PMID: 14561858 DOI: 10.1523/jneurosci.23-28-09312.2003] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Expression of the intermediate filament (IF) protein peripherin is initiated during development at the time of axonal extension and increases during regeneration of nerve fibers. To test whether the IF network is essential for neuron process outgrowth in the mature organism in vivo, we disrupted endogenous peripherin IF in small-sized dorsal root ganglion (DRG) neurons in transgenic mice via expression of a mutant peripherin transgene under control of peripherin gene regulatory sequences. Anatomical and functional analyses showed that these neurons send peripheral and central axonal projections to correct targets, express correct neuropeptides, and mediate acute pain responses normally. However, disruption of IF significantly impaired the ability of uninjured small-sized DRG neurons to sprout collateral axons into adjacent denervated skin, indicating a critical role for intact IF in plasticity, specifically in compensatory nociceptive nerve sprouting.
Collapse
|
33
|
Galoyan SM, Petruska JC, Mendell LM. Mechanisms of sensitization of the response of single dorsal root ganglion cells from adult rat to noxious heat. Eur J Neurosci 2003; 18:535-41. [PMID: 12911749 DOI: 10.1046/j.1460-9568.2003.02775.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We investigated the regulation by nerve growth factor of the response of sensory neurons to noxious heat (>43 degrees C). In dissociated dorsal root ganglion neurons (<30 micro m) from adult rat we demonstrated, using perforated patch clamp recording, that the inward current elicited in response to noxious heating is enhanced by nerve growth factor and reduced by capsazepine. The tachyphylaxis observed in response to the second of two heat pulses was reversed in most cells when nerve growth factor was introduced into the medium during the 5 min between the two heat stimuli, similar to findings using capsaicin [X. Shu & L.M. Mendell (1999) Neurosci. Lett.274, 159-162]. The threshold temperature did not change systematically after nerve growth factor. Using antibodies to TRPV1 and trkA in a subset of cells from which we recorded, we found a virtually perfect correlation between expression of TRPV1 and sensitivity to noxious heat. In addition, trkA expression was perfectly correlated with the ability of nerve growth factor to reverse tachyphylaxis. Thus, this physiological test is a reliable measure of trkA expression in cells sensitive to noxious heat. In agreement with studies in heterologous cells expressing trkA and TRPV1, pharmacologically blocking phospholipase C abolished the effect of nerve growth factor on heat-evoked currents in cells verified to express trkA. We conclude that the response of dorsal root ganglion neurons to noxious heat is conditioned by nerve growth factor in the same way as their response to capsaicin and that these responses require the presence of trkA and TRPV1.
Collapse
Affiliation(s)
- Samuel M Galoyan
- Department of Neurobiology and Behavior, State University of New York at Stony Brook, Life Sciences Building, Room 550, Stony Brook, NY 11794-5230, USA
| | | | | |
Collapse
|
34
|
Abstract
The past year has seen a doubling in the number of heat-sensitive ion channels to six, and four of these channels are from the TRPV family. These channels characteristically have Q(10) values of >10 above the thermal threshold, very different from the Q(10) values of 1.5-2.0 seen in most ion channels. Cells expressing TRPV1 show similar temperature sensitivity to small capsaicin-sensitive nociceptor neurons, consistent with these neurons expressing homomers of TRPV1. A-delta fibres exhibit properties that may be explained by TRPV2 containing channels which is present in large diameter sensory neurons that do not express TRPV1. TRPV3 has a lower temperature threshold and may contribute to warm-sensitive channels together with TRPV1. Warm sensation may also be transduced by TRPV4 expressing sensory neurons and hypothalamic neurons. We can now look forward to further work defining the properties of the recombinant channels in more detail and a re-analysis of endogenous i(heat) currents in thermosensitive neurons and other cells. Data from the study of mice in which TRPV2, TRPV3 or TRPV4 have been deleted are also eagerly awaited.
Collapse
Affiliation(s)
- Christopher D Benham
- Neurology, GlaxoSmithKline Research and Development Ltd., New Frontiers Science Park (North), Third Avenue, Harlow, Essex CM19 5AW, UK.
| | | | | |
Collapse
|
35
|
Kim CS, Kawada T, Kim BS, Han IS, Choe SY, Kurata T, Yu R. Capsaicin exhibits anti-inflammatory property by inhibiting IkB-a degradation in LPS-stimulated peritoneal macrophages. Cell Signal 2003; 15:299-306. [PMID: 12531428 DOI: 10.1016/s0898-6568(02)00086-4] [Citation(s) in RCA: 192] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Capsaicin, a major ingredient of hot pepper, was considered to exhibit an anti-inflammatory property. In order to clarify the signalling mechanism underlying the anti-inflammatory action of capsaicin, we investigated the effect of capsaicin on the production of inflammatory molecules in lipopolysaccharide (LPS)-stimulated murine peritoneal macrophages. The level of PGE2 was measured by EIA. The expression levels of COX-2, iNOS, IkB-a, and vanilloid receptor-1 (VR-1) were determined at the protein and mRNA levels. Significant inhibition of the production of LPS-induced PGE2 by capsaicin was observed in a dose-dependent manner. Capsaicin did not affect the COX-2 expression at either the protein or mRNA level, but inhibited the enzyme activity of COX-2 and the expression of the iNOS protein. Capsaicin completely blocked LPS-induced disappearance of IkB-a and therefore inactivated NF-kB. The inhibitory action of capsaicin on PGE2 production was not abolished by capsazepine, a specific antagonist to VR-1. A high expression level of the VR-1 like protein (VRL-1) was observed in peritoneal macrophages, while the expression of VR-1 was not detected. These findings suggest that the anti-inflammatory action of capsaicin may occur through a novel mechanism, not by a VR-1 receptor-mediated one. Both capsaicin and capsazepine may be a promising drug candidates for ameliorating inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Chu-Sook Kim
- Department of Food Science and Nutrition, University of Ulsan, Mugeo-dong, Nam-ku, Ulsan 680-749, South Korea
| | | | | | | | | | | | | |
Collapse
|
36
|
Quantitative single-cell differences in mu-opioid receptor mRNA distinguish myelinated and unmyelinated nociceptors. J Neurosci 2003. [PMID: 12514199 DOI: 10.1523/jneurosci.23-01-00034.2003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A remarkable feature of opioids is that they inhibit pain that persists from previous injuries without eliminating either the initial pain of a new injury or the protective reflexes triggered by it. Here we ask whether selective expression of the mu-opioid receptor (MOR) gene in primary nociceptors (pain-sensing neurons) might contribute to this aspect of opioid specificity. We quantified single-cell levels of MOR mRNA and measured opioid inhibition of Ca channels on identified nociceptors and low-threshold mechanosensors (non-nociceptors) isolated from rats. Negligibly few non-nociceptors express MOR mRNA, thereby rendering nonpain sensations insensitive to opioids. Nearly half of nociceptors of all size classes also fail to express MOR mRNA or to respond to opioids. Among the opioid-responsive nociceptors, a gene dose-response relationship exists such that maximal opioid inhibition occurs when the MOR mRNA concentration of a cell is >15 pm. Almost all large, myelinated nociceptors express MOR mRNA below this level, whereas small, unmyelinated nociceptors are likely to express above it. Because myelinated nociceptors mediate anti-nociceptive reflexes, the data suggest that fine control of the MOR mRNA level contributes to a complex neural trait: the ability of opioids to suppress persistent pain without preventing response to a new injury.
Collapse
|
37
|
Khasabova IA, Simone DA, Seybold VS. Cannabinoids attenuate depolarization-dependent Ca2+ influx in intermediate-size primary afferent neurons of adult rats. Neuroscience 2003; 115:613-25. [PMID: 12421626 DOI: 10.1016/s0306-4522(02)00449-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
CB1 receptors have been localized to primary afferent neurons, but little is known about the direct effect of cannabinoids on these neurons. The depolarization-evoked increase in the concentration of free intracellular calcium ([Ca(2+)](i)), measured by microfluorimetry, was used as a bioassay for the effect of cannabinoids on isolated, adult rat primary afferent neurons 20-28 h after dissociation of dorsal root ganglia. Cannabinoid agonists CP 55,940 (100 nM) and WIN 55,212-2 (1 microM) had no effect on the mean K(+)-evoked increase in [Ca(2+)](i) in neurons with a somal area<800 microm(2), but the ligands attenuated the evoked increase in [Ca(2+)](i) by 35% in neurons defined as intermediate in size (800-1500 microm(2)). The effects of CP 55,940 and WIN 55,212-2 were mediated by the CB1 receptor on the basis of relative effective concentrations, blockade by the CB1 receptor antagonist SR141716A and lack of effect of WIN 55,212-3. Intermediate-size neurons rarely responded to capsaicin (100 nM). Although cannabinoid agonists generally did not inhibit depolarization-evoked increases in [Ca(2+)](i) in small neurons, immunocytochemical studies indicated that CB1 receptor-immunoreactivity occurred in this population. CB1 receptor-immunoreactive neurons ranged in size from 227 to 2995 microm(2) (mean somal area of 1044 microm(2)). In double labeling studies, CB1 receptor-immunoreactivity co-localized with labeling for calcitonin gene-related peptide and RT97, a marker for myelination, in some primary afferent neurons. The decrease in evoked Ca(2+) influx indicates that cannabinoids decrease conductance through voltage-dependent calcium channels in a subpopulation of primary afferent neurons. Modulation of calcium channels is one mechanism by which cannabinoids may decrease transmitter release from primary afferent neurons. An effect on voltage-dependent calcium channels, however, represents only one possible effect of cannabinoids on primary afferent neurons. Identifying the mechanisms by which cannabinoids modulate nociceptive neurons will increase our understanding of how cannabinoids produce anti-nociception in normal animals and animals with tissue injury.
Collapse
Affiliation(s)
- I A Khasabova
- Department of Neuroscience, University of Minnesota, 6-145 Jackson Hall, 321 Church Street S.E., Minneapolis 55455, USA
| | | | | |
Collapse
|
38
|
Prostaglandin and protein kinase A-dependent modulation of vanilloid receptor function by metabotropic glutamate receptor 5: potential mechanism for thermal hyperalgesia. J Neurosci 2002. [PMID: 12196566 DOI: 10.1523/jneurosci.22-17-07444.2002] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In addition to its role as a CNS neurotransmitter, glutamate has been shown recently to be an important component of the peripheral inflammation response. We demonstrated previously that the group I metabotropic glutamate receptors (mGluRs) mGlu1 and mGlu5 are expressed in the peripheral terminals of sensory neurons and that activation of group I mGluRs in the skin increases thermal sensitivity. In the present study, we provide evidence suggesting that group I mGluRs increase thermal sensitivity by enhancing vanilloid (capsaicin) receptor function. We show that mGlu5 potentiates capsaicin responses in mouse sensory neurons by the phospholipase C pathway but not by activation of protein kinase C. Rather, the effects are mediated by the metabolism of diacylglycerol and the production of prostaglandins via the cyclooxygenase pathway, leading to activation of the cAMP-dependent protein kinase subsequent to prostanoid receptor activation. Behavioral thermal sensitization in mice induced by intraplantar injection of mGlu1/5 agonists was also blocked by inhibitors of protein kinase A and cyclooxygenase, suggesting that a similar signaling pathway operates in vivo. These results demonstrate a novel signaling pathway in sensory neurons and provide a plausible mechanism for the enhancement of thermal sensitivity that occurs with inflammation and after activation of mGluRs on peripheral sensory neuron terminals.
Collapse
|
39
|
Greffrath W, Nemenov MI, Schwarz S, Baumgärtner U, Vogel H, Arendt-Nielsen L, Treede RD. Inward currents in primary nociceptive neurons of the rat and pain sensations in humans elicited by infrared diode laser pulses. Pain 2002; 99:145-55. [PMID: 12237192 DOI: 10.1016/s0304-3959(02)00071-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Radiant heat is often used to study nociception in vivo. We now used infrared radiation generated by a diode laser stimulator (wavelength 980 nm) to investigate transduction mechanisms for noxious heat stimuli in acutely dissociated dorsal root ganglion (DRG) neurons of rats in vitro. The laser stimulator offered the unique opportunity to test whether the same stimuli also elicit pain sensations in humans. A specific heat-induced current (I(heat)) was elicited in six of 13 small DRG neurons (diameter < or =30 microm) tested in the whole-cell configuration of the patch-clamp mode. Current responses in the seven heat-insensitive neurons were within the range explainable by the temperature dependence of the recording setup. I(heat) was characterized by: (1) non-linearity of its amplitude during a suprathreshold heat ramp as well as with stimuli of increasing intensity with an estimated threshold of 42 +/- 1 degrees C; (2) fast rise time and even faster decay time (t(1/2) = 96.5 +/- 5.9 and 27.7 +/- 1.5 ms, respectively); and (3) rate dependence of its induction. All three heat-sensitive neurons tested were also sensitive to capsaicin. The mean threshold for the induction of I(heat) was 2.8 +/- 0.3 J mm(-2). The threshold for the induction of action potentials by depolarizing current pulses was significantly reduced after laser stimulation, suggesting a sensitization at the transformation stage. No such change was seen in heat-insensitive neurons that underwent the same heat stimuli. The same diode laser elicited pain sensations and laser-evoked potentials in human subjects. No significant differences were seen between the pain thresholds in hairy and in glabrous skin, probably due to the deep penetration of this laser radiation. The mean pain threshold for stimuli > or =200 ms in humans was 2.5 +/- 0.2 J mm(-2) (n = 11), and did not differ from the thresholds for the induction of I(heat) in vitro. Our results indicate that I(heat) in primary sensory neurons can be activated by infrared laser pulses that are painful in humans.
Collapse
Affiliation(s)
- Wolfgang Greffrath
- Institute of Physiology and Pathophysiology, Johannes Gutenberg University, Saarstrasse 21, D-55099 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Shin J, Cho H, Hwang SW, Jung J, Shin CY, Lee SY, Kim SH, Lee MG, Choi YH, Kim J, Haber NA, Reichling DB, Khasar S, Levine JD, Oh U. Bradykinin-12-lipoxygenase-VR1 signaling pathway for inflammatory hyperalgesia. Proc Natl Acad Sci U S A 2002; 99:10150-5. [PMID: 12097645 PMCID: PMC126639 DOI: 10.1073/pnas.152002699] [Citation(s) in RCA: 283] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The capsaicin-sensitive vanilloid receptor (VR1) was recently shown to play an important role in inflammatory pain (hyperalgesia), but the underlying mechanism is unknown. We hypothesized that pain-producing inflammatory mediators activate capsaicin receptors by inducing the production of fatty acid agonists of VR1. This study demonstrates that bradykinin, acting at B2 bradykinin receptors, excites sensory nerve endings by activating capsaicin receptors via production of 12-lipoxygenase metabolites of arachidonic acid. This finding identifies a mechanism that might be targeted in the development of new therapeutic strategies for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Jieun Shin
- Sensory Research Center, National Creative Research Initiatives, Seoul National University, San 56-1, Shinlim, Kwanak-Gu, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Pain is the most disruptive influence on the quality of life of cancer patients. Although significant advances are being made in cancer treatment and diagnosis, the basic neurobiology of cancer pain is poorly understood. New insights into these mechanisms are now arising from animal models, and have the potential to fundamentally change the way that cancer pain is controlled.
Collapse
Affiliation(s)
- Patrick W Mantyh
- Department of Preventive Sciences, University of Minnesota, Minneapolis 55455, USA.
| | | | | | | |
Collapse
|
42
|
Greffrath W, Kirschstein T, Nawrath H, Treede RD. Acetylsalicylic acid reduces heat responses in rat nociceptive primary sensory neurons--evidence for a new mechanism of action. Neurosci Lett 2002; 320:61-4. [PMID: 11849764 DOI: 10.1016/s0304-3940(02)00033-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Acetylsalicylic acid (ASA) is thought to exert its peripheral analgesic effects via inhibition of cyclooxygenase. We now studied the effects of ASA on heat responses in primary nociceptive neurons by whole-cell patch-clamp and calcium microfluorimetry experiments. Heat-evoked inward currents in acutely dissociated rat dorsal root ganglion neurons were significantly reduced by ASA in a dose-dependent and reversible manner (IC(50) 375 nM, Hill slope -2.2, maximum effect 55%). Heat-evoked calcium transients (measured with FURA-2) were reversibly reduced by 53+/-14% (P<0.05) by co-application of 1 microM ASA. The low IC(50) value, the rapid occurrence, and the reversibility of the observed effects make it unlikely that inhibition of prostaglandin synthesis is involved in the inhibition of nociceptive heat responses by ASA, and suggest a more direct effect on heat transduction mechanisms.
Collapse
Affiliation(s)
- Wolfgang Greffrath
- Institute of Physiology and Pathophysiology, Johannes Gutenberg University, Saarstrasse 21, D-55099 Mainz, Germany
| | | | | | | |
Collapse
|
43
|
Abstract
Noxious heat may act as an endogenous activator of the ionotropic capsaicin receptor (VR1) and of its recently found homologue VRL1, expressed in rat dorsal root ganglion cells and present along their nerve fibres. We have previously reported that capsaicin induces receptor-mediated and Ca++-dependent calcitonin gene-related peptide (CGRP) release from axons of the isolated rat sciatic nerve. Here we extended the investigation to noxious heat stimulation and the transduction mechanisms involved. Heat stimulation augmented the CGRP release from desheathed sciatic nerves in a log-linear manner with a Q10 of approximately 15 and a threshold between 40 and 42 degrees C. The increases were 1.75-fold at 42 degrees C, 3.8-fold at 45 degrees C and 29.1-fold at 52 degrees C; in Ca++-free solution these heat responses were abolished or reduced by 71 and 92%, respectively. Capsazepine (10 microm) and Ruthenium Red (1 microm) used as capsaicin receptor/channel antagonists did not significantly inhibit the heat-induced release. Pretreatment of the nerves with capsaicin (100 microm for 30 min) caused complete desensitization to 1 microm capsaicin, but a significant heat response remained, indicating that heat sensitivity is not restricted to capsaicin-sensitive fibres. The sciatic nerve axons responded to heat, potassium and capsaicin stimulation with a Ca++-dependent CGRP release. Blockade of the capsaicin receptor/channels had little effect on the heat-induced neuropeptide release. We conclude therefore that other heat-activated ion channels than VR1 and VRL1 in capsaicin-sensitive and -insensitive nerve fibres may cause excitation, axonal Ca++ influx and subsequent CGRP release.
Collapse
Affiliation(s)
- S K Sauer
- Department of Physiology and Experimental Pathophysiology, University of Erlangen, Universitätsstr. 17, D-91054 Erlangen, Germany.
| | | | | |
Collapse
|
44
|
Abstract
The detection of painful stimuli occurs primarily at the peripheral terminals of specialized sensory neurons called nociceptors. These small-diameter neurons transduce signals of a chemical, mechanical, or thermal nature into action potentials and transmit this information to the central nervous system, ultimately eliciting a perception of pain or discomfort. Little is known about the proteins that detect noxious stimuli, especially those of a physical nature. Here we review recent advances in the molecular characterization of the capsaicin (vanilloid) receptor, an excitatory ion channel expressed by nociceptors, which contributes to the detection and integration of pain-producing chemical and thermal stimuli. The analysis of vanilloid receptor gene knockout mice confirms the involvement of this channel in pain sensation, as well as in hypersensitivity to noxious stimuli following tissue injury. At the same time, these studies demonstrate the existence of redundant mechanisms for the sensation of heat-evoked pain.
Collapse
Affiliation(s)
- M J Caterina
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
45
|
Abstract
The sensation of pain alerts us to real or impending injury and triggers appropriate protective responses. Unfortunately, pain often outlives its usefulness as a warning system and instead becomes chronic and debilitating. This transition to a chronic phase involves changes within the spinal cord and brain, but there is also remarkable modulation where pain messages are initiated - at the level of the primary sensory neuron. Efforts to determine how these neurons detect pain-producing stimuli of a thermal, mechanical or chemical nature have revealed new signalling mechanisms and brought us closer to understanding the molecular events that facilitate transitions from acute to persistent pain.
Collapse
Affiliation(s)
- D Julius
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94143, USA.
| | | |
Collapse
|
46
|
Greffrath W, Kirschstein T, Nawrath H, Treede R. Changes in cytosolic calcium in response to noxious heat and their relationship to vanilloid receptors in rat dorsal root ganglion neurons. Neuroscience 2001; 104:539-50. [PMID: 11377853 DOI: 10.1016/s0306-4522(01)00088-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Heat transduction mechanisms in primary nociceptive afferents have been suggested to involve a vanilloid receptor channel with high calcium permeability. To characterize the changes in free cytosolic calcium evoked by noxious heat stimuli (< or =51 degrees C, 10s), we performed microfluorometric measurements in acutely dissociated small dorsal root ganglion neurons (< or =32.5 microm) of adult rats using the dye FURA-2. Only neurons that responded with a reversible increase in intracellular calcium to high potassium were evaluated. Heat-induced calcium transients (exceeding mean + 3S.D. of the temperature dependence of the dye) were found in 66 of 105 neurons. These transients increased non-linearly with temperature. In contrast, heat-insensitive neurons showed a small linear increase of intracellular calcium throughout the range of 12-49 degrees C, similar to cardiac muscle cells. The vanilloid receptor agonist capsaicin induced calcium transients in 72 of 99 neurons. Capsaicin sensitivity and heat sensitivity were significantly associated (P<0.001, chi(2)-test), but 16 of 34 heat-insensitive cells responded to capsaicin and four of 49 heat-sensitive cells were capsaicin insensitive. The competitive vanilloid receptor antagonist capsazepine (10 microM) reversibly reduced the heat-induced calcium transients by 47+/-13%. In contrast, high potassium-induced calcium transients were not affected by pre-incubation with capsazepine. In calcium-free extracellular solution, the heat-induced rise in intracellular calcium was reduced by 76+/-5%. Heat-induced calcium transients were also reversibly reduced by 75+/-6% in sodium-free solution and by 62+/-7% with the L-type calcium channel blocker nifedipine (5 microM). These results indicate that noxious heat rapidly increases intracellular calcium in nociceptive primary sensory neurons. Heat-sensitive vanilloid receptors are involved in the induction of calcium transients, and calcium is also released from intracellular stores, but the main fraction of calcium passes through voltage-operated calcium channels.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Calcium Channels/drug effects
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Capsaicin/pharmacology
- Cells, Cultured/cytology
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Cytosol/drug effects
- Cytosol/metabolism
- Female
- Fluorescent Dyes/pharmacokinetics
- Fura-2/pharmacokinetics
- Ganglia, Spinal/cytology
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Hot Temperature/adverse effects
- Male
- Myocardium/metabolism
- Neurons, Afferent/cytology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Nociceptors/cytology
- Nociceptors/drug effects
- Nociceptors/metabolism
- Pain/metabolism
- Pain/physiopathology
- Potassium/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Drug/drug effects
- Receptors, Drug/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Thermosensing/drug effects
- Thermosensing/physiology
Collapse
Affiliation(s)
- W Greffrath
- Institute of Physiology and Pathophysiology, Johannes Gutenberg University, D-55099, Mainz, Germany
| | | | | | | |
Collapse
|
47
|
Abstract
The recently cloned vanilloid receptor (VR1) is postulated to account for heat and capsaicin sensitivity in unmyelinated afferents. We sought to determine whether heat and capsaicin sensitivity also coexist in myelinated nociceptive afferents. Action potential (AP) activity was recorded from single A-fiber nociceptors that innervated the hairy skin in monkey. Before intradermal injection of capsaicin (10 microg/10 microl) into the receptive field, nociceptors were classified as heat-sensitive (threshold, </=53 degrees C, 1 sec) or heat-insensitive afferents and as mechanically sensitive (von Frey threshold, <6 bar) or mechanically insensitive afferents. All heat-sensitive afferents (n = 16) were insensitive to mechanical stimuli but responded to the intradermal injection of capsaicin (69 +/- 7 APs in 10 min). Responsiveness to mechanical stimuli, thermal stimuli, and capsaicin varied in their receptive fields; the majority of receptive field sites (24 of 36) were responsive to only one or two stimulus modalities, whereas only eight sites responded to all three modalities. For most heat-insensitive afferents, the activity induced by the capsaicin injection did not exceed the activity induced by needle insertion alone. However, the largest response to capsaicin (314 +/- 98 APs in 10 min) was observed for five afferents that were insensitive to heat as well as mechanical stimuli and therefore may be classified as cutaneous chemoreceptors. These results suggest that A-fiber nociceptors play a role in the pain and hyperalgesia associated with capsaicin injection. Our finding that a subgroup of capsaicin-sensitive A-fiber nociceptors are insensitive to heat predicts the existence of heat-insensitive capsaicin receptors.
Collapse
|
48
|
Sprague J, Harrison C, Rowbotham DJ, Smart D, Lambert DG. Temperature-dependent activation of recombinant rat vanilloid VR1 receptors expressed in HEK293 cells by capsaicin and anandamide. Eur J Pharmacol 2001; 423:121-5. [PMID: 11448475 DOI: 10.1016/s0014-2999(01)01123-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Capsaicin activates vanilloid (VR1) receptors found on sensory neurons. These ligand-gated ion channels are also sensitive to low pH, elevated temperature and the endocannabinoid, anandamide. In this study, we have measured capsaicin- and anandamide-induced elevations in intracellular calcium concentrations ([Ca(2+)](i)) in fura-2 loaded HEK293 cells stably expressing the rat VR1 receptor at 22, 37 and 50 degrees C. Both capsaicin and anandamide produced a concentration-dependent elevation in [Ca(2+)](i) at all temperatures. pEC(50) values were 7.74 and 5.69 at 22 degrees C and 6.90 and 5.15 at 37 degrees C for capsaicin and anandamide, respectively. At 50 degrees C, the pEC(50) value for capsaicin was 6.36 but the response to anandamide did not saturate. Responses to both agonists were sensitive to ruthenium red and capsazepine at all temperatures. This temperature-dependent reduction in potency may result from desensitization.
Collapse
Affiliation(s)
- J Sprague
- University Department of Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester LE1 5WW, UK
| | | | | | | | | |
Collapse
|
49
|
Savidge JR, Ranasinghe SP, Rang HP. Comparison of intracellular calcium signals evoked by heat and capsaicin in cultured rat dorsal root ganglion neurons and in a cell line expressing the rat vanilloid receptor, VR1. Neuroscience 2001; 102:177-84. [PMID: 11226681 DOI: 10.1016/s0306-4522(00)00447-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The cloning of the receptor for capsaicin, vanilloid receptor 1, has shown it to be non-selective cation channel with a high calcium permeability which can be opened by noxious heat as well as capsaicin. Here we compare the calcium signals produced by native and recombinant capsaicin receptors when activated by either heat or capsaicin by imaging intracellular calcium levels ([Ca2+](i)) in rat dorsal root ganglion neurons and Chinese hamster ovary cells transfected with the rat vanilloid receptor, vanilloid receptor 1. Vanilloid receptor 1 transfected cells and a subset of dorsal root ganglion neurons responded to both capsaicin and to heating to 50 degrees C with rapid, substantial and reversible rises in [Ca2+](i). All except one of the dorsal root ganglion neurons responsive to capsaicin also showed sensitivity to heat, and most, but not all, heat-sensitive neurons also responded to capsaicin. Both capsaicin and heat responses were dependent on the presence of extracellular Ca2+. Non-transfected Chinese hamster ovary cells and non-responsive dorsal root ganglion neurons showed only small rises in [Ca2+](i) in response to heat which did not depend on the presence of external Ca2+. Responsive dorsal root ganglion neurons and vanilloid receptor 1 transfected cells showed a clear temperature threshold, above which [Ca2+](i) increased rapidly. This was estimated to be 42.6+/-0.3 degrees C for vanilloid receptor 1 transfected cells and 42.0+/-0.6 degrees C for dorsal root ganglion neurons. The competitive capsaicin antagonist capsazepine (10microM) abolished [Ca2+](i) increases stimulated by capsaicin in both dorsal root ganglion neurons and vanilloid receptor 1 transfected cells. However, responses to heat of a similar magnitude in the same cells were inhibited by only 37% by capsazepine (10microM). In vanilloid receptor 1 transfected cells, Ruthenium Red (10microM) blocked responses to both capsaicin and heat. These results demonstrate that imaging of [Ca2+](i) can identify dorsal root ganglion neurons which are responsive to both heat and capsaicin. They show that heat and capsaicin responses mediated by native and recombinant capsaicin receptors are similar with respect to the characteristics and pharmacology examined, suggesting that expression of recombinant vanilloid receptor 1 in cell lines accurately reproduces the properties of the native receptor.
Collapse
Affiliation(s)
- J R Savidge
- Novartis Institute for Medical Sciences, 5 Gower Place, WC1E 6BN, London, UK
| | | | | |
Collapse
|
50
|
Oh GS, Pae HO, Seo WG, Kim NY, Pyun KH, Kim IK, Shin M, Chung HT. Capsazepine, a vanilloid receptor antagonist, inhibits the expression of inducible nitric oxide synthase gene in lipopolysaccharide-stimulated RAW264.7 macrophages through the inactivation of nuclear transcription factor-kappa B. Int Immunopharmacol 2001; 1:777-84. [PMID: 11357890 DOI: 10.1016/s1567-5769(01)00012-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
High amounts of nitric oxide (NO) production following the induction of inducible NO synthase (iNOS) gene expression has been implicated in the pathogenesis of inflammatory diseases. Capsaicin, a vanilloid receptor agonist, is known to have an inhibitory effect on NO production in macrophages. In the present study, we have found that capsazepine (CAPZ), a vanilloid receptor antagonist, also inhibited NO and iNOS protein syntheses induced by lipopolysaccharide in RAW264.7 macrophages via the suppression of iNOS mRNA. The mechanistic studies showed that CAPZ inhibited the expression of iNOS mRNA through the inactivation of nuclear transcription factor-kappa B (NF-kappa B). Thus, capsazepine may be a useful candidate for the development of a drug to treat inflammatory diseases related to iNOS gene overexpression.
Collapse
Affiliation(s)
- G S Oh
- Medicinal Resources Research Center (MRRC), Wonkwang University, Chonbuk, South Korea
| | | | | | | | | | | | | | | |
Collapse
|