2701
|
Xiao B, Patapoutian A. Scratching the surface: a role of pain-sensing TRPA1 in itch. Nat Neurosci 2011; 14:540-2. [DOI: 10.1038/nn.2813] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
2702
|
Li H, Hui F, Jianmin X. Effects of hydrocortisone sodium succinate on voltage-gated sodium current in trigeminal ganglion neurons of rat. Neurol Res 2011; 33:295-9. [PMID: 21513649 DOI: 10.1179/016164110x12714125204515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE The effects of hydrocortisone sodium succinate on voltage-gated sodium current (I(Na)) were investigated by using the patch-clamp technique in trigeminal ganglion (TG) neurons of rat. METHODS I(Na) was recorded by whole-cell patch-clamp techniques after different concentrations of hydrocortisone sodium succinate were perfused in TG neurons of rat. RESULTS The results showed that hydrocortisone sodium succinate could inhibit I(Na) in concentration-dependent manner. Hydrocortisone sodium succinate 0.1, 0.3, 1, 3 μmol/l reduced I(Na) by 19.4±4.3, 26.7±3.9, 38.1±6.1, 69.6±5.4% respectively. The IC(50) was 1.58 μmol/l. This inhibitory effect occurred quickly (within 1 minute). However, hydrocortisone sodium succinate had no significant effect on the activation and inactivation courses of I(Na). CONCLUSION It is suggested that the rapid inhibition of I(Na) in TG neurons by hydrocortisone sodium succinate is probably related to non-genomic effect. This inhibition might participate in the relaxation of pain in some emergency states.
Collapse
Affiliation(s)
- He Li
- Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | |
Collapse
|
2703
|
Wang H, Xu H, Wu LJ, Kim SS, Chen T, Koga K, Descalzi G, Gong B, Vadakkan KI, Zhang X, Kaang BK, Zhuo M. Identification of an adenylyl cyclase inhibitor for treating neuropathic and inflammatory pain. Sci Transl Med 2011; 3:65ra3. [PMID: 21228397 DOI: 10.1126/scitranslmed.3001269] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neuropathic pain, often caused by nerve injury, is commonly observed among patients with different diseases. Because its basic mechanisms are poorly understood, effective medications are limited. Previous investigations of basic pain mechanisms and drug discovery efforts have focused mainly on early sensory neurons such as dorsal root ganglion and spinal dorsal horn neurons, and few synaptic-level studies or new drugs are designed to target the injury-related cortical plasticity that accompanies neuropathic pain. Our previous work has demonstrated that calcium-stimulated adenylyl cyclase 1 (AC1) is critical for nerve injury-induced synaptic changes in the anterior cingulate cortex. Through rational drug design and chemical screening, we have identified a lead candidate AC1 inhibitor, NB001, which is relatively selective for AC1 over other adenylate cyclase isoforms. Using a variety of behavioral tests and toxicity studies, we have found that NB001, when administered intraperitoneally or orally, has an analgesic effect in animal models of neuropathic pain, without any apparent side effects. Our study thus shows that AC1 could be a productive therapeutic target for neuropathic pain and describes a new agent for the possible treatment of neuropathic pain.
Collapse
Affiliation(s)
- Hansen Wang
- Department of Physiology, Faculty of Medicine, University of Toronto Centre for the Study of Pain, Medical Sciences Building, 1 King's College Circle, Toronto, Ontario M5S1A8, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2704
|
Abstract
Chronic pain is estimated to be the third most prevalent health problem in the world. Although scientists have made great strides in our understanding of the molecular mechanisms through which chronic pain develops, this knowledge has not been translated into new therapies. In this issue of Science Translational Medicine, Wang and colleagues report on the development of a selective antagonist of type 1 adenylate cyclase (AC1), which is induced in subsets of neurons in the central nervous system during the development of neuropathic pain. Blockade of AC1 significantly alleviates the mechanical hypersensitivity that occurs in a mouse model of neuropathic pain without affecting acute pain responsiveness or cognitive and motor function. These features make AC1 a potential therapeutic target for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Reza Sharif-Naeini
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
2705
|
Takahashi Y, Hasegawa-Moriyama M, Sakurai T, Inada E. The macrophage-mediated effects of the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuate tactile allodynia in the early phase of neuropathic pain development. Anesth Analg 2011; 113:398-404. [PMID: 21490083 DOI: 10.1213/ane.0b013e31821b220c] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Neuroinflammation triggered by macrophage infiltration into sites of peripheral nerve injury may result in neuropathic pain. Peroxisome proliferator-activated receptor (PPAR)γ signaling regulates the properties of macrophages. However, the macrophage-mediated effects of PPARγ signaling on neuropathic pain triggered by peripheral inflammation have not been investigated. METHODS To evaluate the peripheral effects of PPARγ signaling on tactile allodynia, we administered the blood-brain barrier-impermeant PPARγ agonist, rosiglitazone, after partial sciatic nerve ligation (PSNL) as (1) systemic treatment during different phases of neuropathic pain development, (2) local injection to the PSNL site in the early phase, or (3) peritoneal macrophages pretreated with rosiglitazone transplanted into the PSNL site. In addition, the direct effect of rosiglitazone was evaluated in peritoneal macrophages activated with interferon-γ. RESULTS Systemic rosiglitazone treatment early in the course of progressive inflammation ameliorated tactile allodynia, macrophage infiltration, and production of proinflammatory mediators including cyclooxygenase-2, inducible nitric oxide synthase, and matrix metalloprotease 9 at the PSNL site. Local injection of rosiglitazone and transplantation of rosiglitazone-treated peritoneal macrophages at the ligation site significantly improved tactile allodynia. In peritoneal macrophages, rosiglitazone down-regulated interferon-γ-induced gene expression of cyclooxygenase-2 and inducible nitric oxide synthase and attenuated the chemotactic response to monocyte chemotactic protein-1. DISCUSSION Rosiglitazone treatment in the early phase of neuropathic pain significantly alleviated the development of tactile allodynia by regulating macrophage infiltration and production of proinflammatory molecules at the inflamed site. Our results indicate that the activation of PPARγ signaling in macrophages during the early phase may suppress neuropathic pain development.
Collapse
Affiliation(s)
- Yoshika Takahashi
- Department of Anesthesiology and Pain Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | | | | | | |
Collapse
|
2706
|
Nassini R, Gees M, Harrison S, De Siena G, Materazzi S, Moretto N, Failli P, Preti D, Marchetti N, Cavazzini A, Mancini F, Pedretti P, Nilius B, Patacchini R, Geppetti P. Oxaliplatin elicits mechanical and cold allodynia in rodents via TRPA1 receptor stimulation. Pain 2011; 152:1621-1631. [PMID: 21481532 DOI: 10.1016/j.pain.2011.02.051] [Citation(s) in RCA: 256] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 02/18/2011] [Accepted: 02/28/2011] [Indexed: 02/09/2023]
Abstract
Platinum-based anticancer drugs cause neurotoxicity. In particular, oxaliplatin produces early-developing, painful, and cold-exacerbated paresthesias. However, the mechanism underlying these bothersome and dose-limiting adverse effects is unknown. We hypothesized that the transient receptor potential ankyrin 1 (TRPA1), a cation channel activated by oxidative stress and cold temperature, contributes to mechanical and cold hypersensitivity caused by oxaliplatin and cisplatin. Oxaliplatin and cisplatin evoked glutathione-sensitive relaxation, mediated by TRPA1 stimulation and the release of calcitonin gene-related peptide from sensory nerve terminals in isolated guinea pig pulmonary arteries. No calcium response was observed in cultured mouse dorsal root ganglion neurons or in naïve Chinese hamster ovary (CHO) cells exposed to oxaliplatin or cisplatin. However, oxaliplatin, and with lower potency, cisplatin, evoked a glutathione-sensitive calcium response in CHO cells expressing mouse TRPA1. One single administration of oxaliplatin produced mechanical and cold hyperalgesia in rats, an effect selectively abated by the TRPA1 antagonist HC-030031. Oxaliplatin administration caused mechanical and cold allodynia in mice. Both responses were absent in TRPA1-deficient mice. Administration of cisplatin evoked mechanical allodynia, an effect that was reduced in TRPA1-deficient mice. TRPA1 is therefore required for oxaliplatin-evoked mechanical and cold hypersensitivity, and contributes to cisplatin-evoked mechanical allodynia. Channel activation is most likely caused by glutathione-sensitive molecules, including reactive oxygen species and their byproducts, which are generated after tissue exposure to platinum-based drugs from cells surrounding nociceptive nerve terminals.
Collapse
Affiliation(s)
- Romina Nassini
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy Department of Molecular Cell Biology, Katholieke Universiteit, Leuven, Belgium Department of Pharmacology, Chiesi Farmaceutici, Parma, Italy Department of Pharmaceutical Chemistry, University of Ferrara, Ferrara, Italy Chemistry Department, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2707
|
Zylka MJ. Pain-relieving prospects for adenosine receptors and ectonucleotidases. Trends Mol Med 2011; 17:188-96. [PMID: 21236731 PMCID: PMC3078941 DOI: 10.1016/j.molmed.2010.12.006] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Revised: 12/12/2010] [Accepted: 12/15/2010] [Indexed: 01/09/2023]
Abstract
Adenosine receptor agonists have potent antinociceptive effects in diverse preclinical models of chronic pain. By contrast, the efficacy of adenosine and adenosine receptor agonists in treating pain in humans is unclear. Two ectonucleotidases that generate adenosine in nociceptive neurons were recently identified. When injected spinally, these enzymes have long-lasting adenosine A(1) receptor-dependent antinociceptive effects in inflammatory and neuropathic pain models. Furthermore, recent findings indicate that spinal adenosine A(2A) receptor activation can enduringly inhibit neuropathic pain symptoms. Collectively, these studies suggest the possibility of treating chronic pain in humans by targeting specific adenosine receptor subtypes in anatomically defined regions with agonists or with ectonucleotidases that generate adenosine.
Collapse
Affiliation(s)
- Mark J Zylka
- Department of Cell and Molecular Physiology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
2708
|
Richardson GP, de Monvel JB, Petit C. How the Genetics of Deafness Illuminates Auditory Physiology. Annu Rev Physiol 2011; 73:311-34. [DOI: 10.1146/annurev-physiol-012110-142228] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Guy P. Richardson
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, United Kingdom;
| | - Jacques Boutet de Monvel
- Unité de Génétique et Physiologie de l'Audition, Département de Neuroscience, Institut Pasteur, F-75724 Paris cedex 15, France; ,
- Inserm UMRS 587, F-75015 Paris, France
- Université Pierre & Marie Curie, F-75005 Paris, France
| | - Christine Petit
- Unité de Génétique et Physiologie de l'Audition, Département de Neuroscience, Institut Pasteur, F-75724 Paris cedex 15, France; ,
- Inserm UMRS 587, F-75015 Paris, France
- Université Pierre & Marie Curie, F-75005 Paris, France
- Collège de France, F-75005 Paris, France
| |
Collapse
|
2709
|
McCoy DD, Knowlton WM, McKemy DD. Scraping through the ice: uncovering the role of TRPM8 in cold transduction. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1278-87. [PMID: 21411765 DOI: 10.1152/ajpregu.00631.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The proper detection of environmental temperatures is essential for the optimal growth and survival of organisms of all shapes and phyla, yet only recently have the molecular mechanisms for temperature sensing been elucidated. The discovery of temperature-sensitive ion channels of the transient receptor potential (TRP) superfamily has been pivotal in explaining how temperatures are sensed in vivo, and here we will focus on the lone member of this cohort, TRPM8, which has been unequivocally shown to be cold sensitive. TRPM8 is expressed in somatosensory neurons that innervate peripheral tissues such as the skin and oral cavity, and recent genetic evidence has shown it to be the principal transducer of cool and cold stimuli. It is remarkable that this one channel, unlike other thermosensitive TRP channels, is associated with both innocuous and noxious temperature transduction, as well as cold hypersensitivity during injury and, paradoxically, cold-mediated analgesia. With ongoing research, the field is getting closer to answering a number of fundamental questions regarding this channel, including the cellular mechanisms of TRPM8 modulation, the molecular context of TRPM8 expression, as well as the full extent of the role of TRPM8 in cold signaling in vivo. These findings will further our understanding of basic thermotransduction and sensory coding, and may have important implications for treatments for acute and chronic pain.
Collapse
Affiliation(s)
- Daniel D McCoy
- Neurobiology, University of Southern California, Los Angeles, California 90089, USA
| | | | | |
Collapse
|
2710
|
Gangadharan V, Wang R, Ulzhöfer B, Luo C, Bardoni R, Bali KK, Agarwal N, Tegeder I, Hildebrandt U, Nagy GG, Todd AJ, Ghirri A, Häussler A, Sprengel R, Seeburg PH, MacDermott AB, Lewin GR, Kuner R. Peripheral calcium-permeable AMPA receptors regulate chronic inflammatory pain in mice. J Clin Invest 2011; 121:1608-23. [PMID: 21383497 DOI: 10.1172/jci44911] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 01/12/2011] [Indexed: 01/13/2023] Open
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type (AMPA-type) glutamate receptors (AMPARs) play an important role in plasticity at central synapses. Although there is anatomical evidence for AMPAR expression in the peripheral nervous system, the functional role of such receptors in vivo is not clear. To address this issue, we generated mice specifically lacking either of the key AMPAR subunits, GluA1 or GluA2, in peripheral, pain-sensing neurons (nociceptors), while preserving expression of these subunits in the central nervous system. Nociceptor-specific deletion of GluA1 led to disruption of calcium permeability and reduced capsaicin-evoked activation of nociceptors. Deletion of GluA1, but not GluA2, led to reduced mechanical hypersensitivity and sensitization in models of chronic inflammatory pain and arthritis. Further analysis revealed that GluA1-containing AMPARs regulated the responses of nociceptors to painful stimuli in inflamed tissues and controlled the excitatory drive from the periphery into the spinal cord. Consequently, peripherally applied AMPAR antagonists alleviated inflammatory pain by specifically blocking calcium-permeable AMPARs, without affecting physiological pain or eliciting central side effects. These findings indicate an important pathophysiological role for calcium-permeable AMPARs in nociceptors and may have therapeutic implications for the treatment chronic inflammatory pain states.
Collapse
|
2711
|
Kim H, Chung G, Jo H, Kim Y, Bae Y, Jung S, Kim JS, Oh S. Characterization of Dental Nociceptive Neurons. J Dent Res 2011; 90:771-6. [DOI: 10.1177/0022034511399906] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Selective blockade of nociceptive neurons can be achieved by the delivery of permanently charged sodium channel blockers through the pores of nociceptive ion channels. To assess the feasibility of this application in the dental area, we investigated the electrophysiological and neurochemical characteristics of nociceptive dental primary afferent (DPA) neurons. DPA neurons were identified within trigeminal ganglia labeling with a retrograde fluorescent dye applied to the upper molars of adult rats. Electrophysiological studies revealed that the majority of dental primary afferent neurons showed characteristics of nociceptive neurons, such as sensitivity to capsaicin and the presence of a hump in action potential. Immunohistochemical analysis revealed a large proportion of DPA neurons to be IB4-positive and to express TRPV1 and P2X3. Single-cell RT-PCR revealed mRNA expression of various nociceptive channels, including the temperature-sensitive TRPV1, TRPA1, TRPM8 channels, the extracellular ATP receptor channels P2X2 and P2X3, as well as the nociceptor-specific sodium channel, NaV1.8. In conclusion, DPA neurons have the electrophysiological characteristics of nociceptors and express several nociceptor-specific ion channels. Analysis of these data may assist in the search for a new route of entry for the delivery of membrane-impermeant local anesthetics. Abbreviations: AP, action potential; DiI, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate; DPA, dental primary afferent; FITC, fluorescein 5(6)-isothiocyanate; IB4, isolectin-B4; RT-PCR, reverse-transcription polymerase chain-reaction; TRP, transient receptor potential.
Collapse
Affiliation(s)
- H.Y. Kim
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, 28 Yeongeon-Dong, Jongno-Gu, Seoul 110-749, Republic of Korea
| | - G. Chung
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, 28 Yeongeon-Dong, Jongno-Gu, Seoul 110-749, Republic of Korea
| | - H.J. Jo
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, 28 Yeongeon-Dong, Jongno-Gu, Seoul 110-749, Republic of Korea
| | - Y.S. Kim
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Republic of Korea
| | - Y.C. Bae
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Republic of Korea
| | - S.J. Jung
- Department of Physiology, School of Medicine, Hanyang University, Seoul 133-791, Republic of Korea
| | - J.-S. Kim
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, 28 Yeongeon-Dong, Jongno-Gu, Seoul 110-749, Republic of Korea
| | - S.B. Oh
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, 28 Yeongeon-Dong, Jongno-Gu, Seoul 110-749, Republic of Korea
| |
Collapse
|
2712
|
Jongen JL, Holstege JC. Propagation of Spinal Nociceptive Activity in the Spatial and Temporal Domains. Neuroscientist 2011; 18:8-14. [DOI: 10.1177/1073858410391813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nociceptive stimuli are transmitted through thinly myelinated or unmyelinated primary afferent fibers called nociceptors, which terminate mainly in the superficial dorsal horn of the spinal cord. While most nociceptive fibers terminate in the spinal segment of the entrance, (collateral) fibers may ascend and descend several segments upon their entry into the spinal cord, which is reflected in the receptive fields of central nociceptive neurons. In chronic pain states like inflammatory or neuropathic pain, the area of nociceptive activity may expand even further in rostrocaudal and mediolateral directions. Also, within minutes (inflammatory pain) or days (neuropathic pain), an increased sensitivity of peripheral and central nociceptive neurons will develop, which is referred to as sensitization. While anatomical, physiological, and psychophysical techniques have focused on one particular aspect of central sensitization at a time, functional imaging techniques like functional MRI, intrinsic optical imaging, and autofluorescent flavoprotein imaging (AFI) are able to capture both spatial and temporal dimensions of central sensitization simultaneously. AFI and other neuroimaging techniques may clarify fundamental aspects relating to the spread of nociceptive activity within the spinal cord and may thus provide a practical tool to test the efficacy of new analgesic drugs or procedures in animals and ultimately in humans.
Collapse
Affiliation(s)
| | - Jan C. Holstege
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
2713
|
Verlust des Sensibilitäts- und Schmerzempfindens. MED GENET-BERLIN 2011. [DOI: 10.1007/s11825-010-0260-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Zusammenfassung
Die Weiterleitung von Informationen des Körpers über Berührung, Druck, Dehnung, Scherung, Hitze oder Kälte erfolgt durch sensorische Nervenzellen. Bei überschwelliger Reizstärke resultiert ein akuter Schmerz als Warnsignal für den Organismus. Eine Degeneration von Nervenzellen, die an der Signalvermittlung beteiligt sind, ist das wesentliche Kennzeichen der hereditären sensorischen und autonomen Neuropathien (HSAN) und zeigt die zentrale Bedeutung des sensorischen Nervensystems für die Protektion des Körpers. Der Sensibilitätsverlust bei HSAN-Patienten führt zum Auftreten von Ulzerationen an Händen und Füßen sowie lokalen Knochentzündungen und erfordert im fortgeschrittenen Krankheitsverlauf Amputationen. Bei einigen Formen der Erkrankung ist der Sensibilitätsverlust bereits angeboren, was bei betroffenen Kindern zu Selbstverstümmelungen führt. Eine Degeneration autonomer sowie motorischer Nervenfasern kann den Krankheitsverlauf begleiten. Die HSAN wird aufgrund ihrer klinischen Verlaufsformen in 5 Gruppen unterteilt. Bislang sind Mutationen in 9 Genen als ursächlich beschrieben worden. Dieser Übersichtsartikel soll Einblicke in die Klinik, Genetik und Pathophysiologie dieser Erkrankungen geben.
Collapse
|
2714
|
Gallio M, Ofstad TA, Macpherson LJ, Wang JW, Zuker CS. The coding of temperature in the Drosophila brain. Cell 2011; 144:614-24. [PMID: 21335241 PMCID: PMC3336488 DOI: 10.1016/j.cell.2011.01.028] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 11/03/2010] [Accepted: 01/24/2011] [Indexed: 11/30/2022]
Abstract
Thermosensation is an indispensable sensory modality. Here, we study temperature coding in Drosophila, and show that temperature is represented by a spatial map of activity in the brain. First, we identify TRP channels that function in the fly antenna to mediate the detection of cold stimuli. Next, we identify the hot-sensing neurons and show that hot and cold antennal receptors project onto distinct, but adjacent glomeruli in the Proximal-Antennal-Protocerebrum (PAP) forming a thermotopic map in the brain. We use two-photon imaging to reveal the functional segregation of hot and cold responses in the PAP, and show that silencing the hot- or cold-sensing neurons produces animals with distinct and discrete deficits in their behavioral responses to thermal stimuli. Together, these results demonstrate that dedicated populations of cells orchestrate behavioral responses to different temperature stimuli, and reveal a labeled-line logic for the coding of temperature information in the brain.
Collapse
Affiliation(s)
- Marco Gallio
- Departments of Neurobiology and Neurosciences, University of California at San Diego, La Jolla, 92093, USA
| | | | | | | | | |
Collapse
|
2715
|
Dickinson BD, Head CA, Gitlow S, Osbahr AJ. Maldynia: pathophysiology and management of neuropathic and maladaptive pain--a report of the AMA Council on Science and Public Health. PAIN MEDICINE 2011; 11:1635-53. [PMID: 21044254 DOI: 10.1111/j.1526-4637.2010.00986.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Because of disparate taxonomic arrays for classification, the American Academy of Pain Medicine has proposed categorizing pain on a neurobiologic basis as eudynia (nociceptive pain), Greek for "good pain," or maldynia (maladaptive pain), Greek for "bad pain." The latter has been viewed as maladaptive because it may occur in the absence of ongoing noxious stimuli and does not promote healing and repair. OBJECTIVE To address recent findings on the pathogenesis of pain following neural injury and consider whether the development of maladaptive pain justifies its classification as a disease and to briefly discuss the scope of pharmacologic and non-pharmacologic approaches employed in patients with such pain. METHODS English language reports on studies using human subjects were selected from a PubMed search of the literature from 1995 to August 2010 and from the Cochrane Library. Further information was obtained from Internet sites of medical specialty and other societies devoted to pain management. RESULTS Neural damage to either the peripheral or central nervous system provokes multiple processes including peripheral and central sensitization, ectopic activity, neuronal cell death, disinhibition, altered gene expression, and abnormal sprouting and cellular connectivity. A series of neuro-immune interactions underlie many of these mechanisms. Imaging studies have shown that such damage is characterized by functional, structural, and chemical changes in the brain. Such pain is maladaptive in the sense that it occurs in the absence of ongoing noxious stimuli and does not promote healing and repair. CONCLUSION As defined, maldynia is a multidimensional process that may warrant consideration as a chronic disease not only affecting sensory and emotional processing but also producing an altered brain state based on both functional imaging and macroscopic measurements. However, the absolute clinical value of this definition is not established.
Collapse
Affiliation(s)
- Barry D Dickinson
- Council on Science and Public Health, American Medical Association, Chicago, Illinois 60654, USA.
| | | | | | | |
Collapse
|
2716
|
Lundgren O, Jodal M, Jansson M, Ryberg AT, Svensson L. Intestinal epithelial stem/progenitor cells are controlled by mucosal afferent nerves. PLoS One 2011; 6:e16295. [PMID: 21347406 PMCID: PMC3036584 DOI: 10.1371/journal.pone.0016295] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 12/22/2010] [Indexed: 01/07/2023] Open
Abstract
Background The maintenance of the intestinal epithelium is of great importance for the survival of the organism. A possible nervous control of epithelial cell renewal was studied in rats and mice. Methods Mucosal afferent nerves were stimulated by exposing the intestinal mucosa to capsaicin (1.6 mM), which stimulates intestinal external axons. Epithelial cell renewal was investigated in the jejunum by measuring intestinal thymidine kinase (TK) activity, intestinal 3H-thymidine incorporation into DNA, and the number of crypt cells labeled with BrdU. The influence of the external gut innervation was minimized by severing the periarterial nerves. Principal Findings Luminal capsaicin increased all the studied variables, an effect nervously mediated to judge from inhibitory effects on TK activity or 3H-thymidine incorporation into DNA by exposing the mucosa to lidocaine (a local anesthetic) or by giving four different neurotransmitter receptor antagonists i.v. (muscarinic, nicotinic, neurokinin1 (NK1) or calcitonin gene related peptide (CGRP) receptors). After degeneration of the intestinal external nerves capsaicin did not increase TK activity, suggesting the involvement of an axon reflex. Intra-arterial infusion of Substance P (SP) or CGRP increased intestinal TK activity, a response abolished by muscarinic receptor blockade. Immunohistochemistry suggested presence of M3 and M5 muscarinic receptors on the intestinal stem/progenitor cells. We propose that the stem/progenitor cells are controlled by cholinergic nerves, which, in turn, are influenced by mucosal afferent neuron(s) releasing acetylcholine and/or SP and/or CGRP. In mice lacking the capsaicin receptor, thymidine incorporation into DNA and number of crypt cells labeled with BrdU was lower than in wild type animals suggesting that nerves are important also in the absence of luminal capsaicin, a conclusion also supported by the observation that atropine lowered thymidine incorporation into DNA by 60% in control rat segments. Conclusion Enteric nerves are of importance in maintaining the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Ove Lundgren
- Section of Physiology, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
2717
|
Delmas P, Hao J, Rodat-Despoix L. Molecular mechanisms of mechanotransduction in mammalian sensory neurons. Nat Rev Neurosci 2011; 12:139-53. [PMID: 21304548 DOI: 10.1038/nrn2993] [Citation(s) in RCA: 306] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The somatosensory system mediates fundamental physiological functions, including the senses of touch, pain and proprioception. This variety of functions is matched by a diverse array of mechanosensory neurons that respond to force in a specific fashion. Mechanotransduction begins at the sensory nerve endings, which rapidly transform mechanical forces into electrical signals. Progress has been made in establishing the functional properties of mechanoreceptors, but it has been remarkably difficult to characterize mechanotranducer channels at the molecular level. However, in the past few years, new functional assays have provided insights into the basic properties and molecular identity of mechanotransducer channels in mammalian sensory neurons. The recent identification of novel families of proteins as mechanosensing molecules will undoubtedly accelerate our understanding of mechanotransduction mechanisms in mammalian somatosensation.
Collapse
Affiliation(s)
- Patrick Delmas
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, UMR 6231, Centre National de la Recherche Scientifique, Université de la Méditerranée, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex 15, France.
| | | | | |
Collapse
|
2718
|
Valente P, Fernández-Carvajal A, Camprubí-Robles M, Gomis A, Quirce S, Viana F, Fernández-Ballester G, González-Ros JM, Belmonte C, Planells-Cases R, Ferrer-Montiel A. Membrane-tethered peptides patterned after the TRP domain (TRPducins) selectively inhibit TRPV1 channel activity. FASEB J 2011; 25:1628-40. [PMID: 21307333 DOI: 10.1096/fj.10-174433] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel is a thermosensory receptor implicated in diverse physiological and pathological processes. The TRP domain, a highly conserved region in the C terminus adjacent to the internal channel gate, is critical for subunit tetramerization and channel gating. Here, we show that cell-penetrating, membrane-anchored peptides patterned after this protein domain are moderate and selective TRPV1 antagonists both in vitro and in vivo, blocking receptor activity in intact rat primary sensory neurons and their peripheral axons with mean decline time of 30 min. The most potent lipopeptide, TRP-p5, blocked all modes of TRPV1 gating with micromolar efficacy (IC(50)<10 μM), without significantly affecting other thermoTRP channels. In contrast, its retrosequence or the corresponding sequences of other TRPV channels did not alter TRPV1 channel activity (IC(50)>100 μM). TRP-p5 did not affect the capsaicin sensitivity of the vanilloid receptor. Our data suggest that TRP-p5 interferes with protein-protein interactions at the level of the TRP domain that are essential for the "conformational" change that leads to gate opening. Therefore, these palmitoylated peptides, which we termed TRPducins, are noncompetitive, voltage-independent, sequence-specific TRPV1 blockers. Our findings indicate that TRPducin-like peptides may embody a novel molecular strategy that can be exploited to generate a selective pharmacological arsenal for the TRP superfamily of ion channels.
Collapse
Affiliation(s)
- Pierluigi Valente
- Instituto de Biología Molecular y Celular, Universidad Miguel Herna´ndez, Elche, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2719
|
Miura M, Sasaki M, Mizukoshi K, Shibasaki M, Izumi Y, Shimosato G, Amaya F. Peripheral sensitization caused by insulin-like growth factor 1 contributes to pain hypersensitivity after tissue injury. Pain 2011; 152:888-895. [PMID: 21296499 DOI: 10.1016/j.pain.2011.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 12/03/2010] [Accepted: 01/04/2011] [Indexed: 01/25/2023]
Abstract
Sensitization of primary afferent neurons is one of the most important components of pain hypersensitivity after tissue injury. Insulin-like growth factor 1 (IGF-1), involved in wound repair in injured tissue, also plays an important role in maintaining neuronal function. In the present study, we investigated the effect of tissue IGF-1 on nociceptive sensitivity of primary afferent neurons. Local administration of IGF-1 induced thermal and mechanical pain hypersensitivity in a dose-dependent manner, and was attenuated by IGF-1 receptor (IGF1R) inhibition. Tissue but not plasma IGF-1 levels, as determined by enzyme-linked immunosorbent assay, significantly increased after plantar incision. Immunohistochemistry revealed that IGF1R was predominantly expressed in neurons as well as in satellite glial cells in the dorsal root ganglion (DRG). Double-labeling immunohistochemistry showed that IGF1R expression colocalized with peripherin and TRPV1, but not with NF200 in DRG neurons. The IGF1R inhibitor successfully alleviated mechanical allodynia, heat hyperalgesia, and spontaneous pain behavior observed after plantar incision. Expression of phosphorylated Akt in DRG neurons significantly increased after plantar incision and was suppressed by IGF1R inhibition. These results demonstrate that increased tissue IGF-1 production sensitizes primary afferent neurons via the IGF1R/Akt pathway to facilitate pain hypersensitivity after tissue damage.
Collapse
Affiliation(s)
- Mayumi Miura
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan Pain Mechanism Research Group, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
2720
|
Liu Y, Ma Q. Generation of somatic sensory neuron diversity and implications on sensory coding. Curr Opin Neurobiol 2011; 21:52-60. [PMID: 20888752 PMCID: PMC3029488 DOI: 10.1016/j.conb.2010.09.003] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 08/27/2010] [Accepted: 09/03/2010] [Indexed: 11/30/2022]
Abstract
Neurons in the dorsal root ganglia (DRG) are composed of a variety of sensory modalities, three of which are pain-sensing nociceptors, temperature-sensing thermoceptors, and itch-sensing pruriceptors. All these neurons are emerged from a common pool of embryonic DRG neurons that are marked by the expression of the neurotrophin receptor TrkA. Here we discuss how intrinsic transcription factors interface with target-derived signals to specify these functionally distinct sensory neurons. We will also discuss how this control mechanism provides a developmental perspective for the coding of somatic sensations.
Collapse
Affiliation(s)
- Yang Liu
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, 1 Jimmy Fund Way, Boston, Massachusetts USA 02115
| | - Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, 1 Jimmy Fund Way, Boston, Massachusetts USA 02115
| |
Collapse
|
2721
|
Ghilardi JR, Freeman KT, Jimenez-Andrade JM, Mantyh WG, Bloom AP, Bouhana KS, Trollinger D, Winkler J, Lee P, Andrews SW, Kuskowski MA, Mantyh PW. Sustained blockade of neurotrophin receptors TrkA, TrkB and TrkC reduces non-malignant skeletal pain but not the maintenance of sensory and sympathetic nerve fibers. Bone 2011; 48:389-98. [PMID: 20854944 PMCID: PMC3020250 DOI: 10.1016/j.bone.2010.09.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 09/01/2010] [Accepted: 09/14/2010] [Indexed: 10/19/2022]
Abstract
Current therapies for treating skeletal pain have significant limitations as available drugs (non-steroidal anti-inflammatory drugs and opiates) have significant unwanted side effects. Targeting nerve growth factor (NGF) or its cognate receptor tropomysin receptor kinase A (TrkA) has recently become an attractive target for inhibition of adult skeletal pain. Here we explore whether sustained administration of a selective small molecule Trk inhibitor that blocks TrkA, TrkB and TrkC kinase activity with nanomolar affinity reduces skeletal pain while allowing the maintenance of sensory and sympathetic neurons in the adult mouse. Twice-daily administration of a Trk inhibitor was begun 1 day post fracture and within 8 h of acute administration fracture pain-related behaviors were reduced by 50% without significant sedation, weight gain or inhibition of fracture healing. Following administration of the Trk inhibitor for 7 weeks, there was no significant decline in the density of unmyelinated or myelinated sensory nerve fibers, sympathetic nerve fibers, measures of acute thermal pain, acute mechanical pain, or general neuromuscular function. The present results suggest that sustained administration of a peripherally selective TrkA, B and C inhibitor significantly reduces skeletal pain without having any obvious detrimental effects on adult sensory and sympathetic nerve fibers or early fracture healing. As with any potential therapeutic advance, understanding whether the benefits of Trk blockade are associated with any risks or unexpected effects will be required to fully appreciate the patient populations that may benefit from this therapeutic approach.
Collapse
Affiliation(s)
| | | | | | - William G. Mantyh
- Department of Pharmacology University of Arizona, Tucson, AZ 85724, USA
| | - Aaron P. Bloom
- Department of Pharmacology University of Arizona, Tucson, AZ 85724, USA
| | | | | | | | | | | | | | - Patrick W. Mantyh
- Research Service VA Medical Center, Minneapolis, MN 55417, USA
- Department of Pharmacology University of Arizona, Tucson, AZ 85724, USA
- Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
2722
|
Hache G, Coudore F, Gardier AM, Guiard BP. Monoaminergic Antidepressants in the Relief of Pain: Potential Therapeutic Utility of Triple Reuptake Inhibitors (TRIs). Pharmaceuticals (Basel) 2011. [PMCID: PMC4053958 DOI: 10.3390/ph4020285] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Over 75% of depressed patients suffer from painful symptoms predicting a greater severity and a less favorable outcome of depression. Imaging, anatomical and functional studies have demonstrated the existence of common brain structures, neuronal pathways and neurotransmitters in depression and pain. In particular, the ascending serotonergic and noradrenergic pathways originating from the raphe nuclei and the locus coeruleus; respectively, send projections to the limbic system. Such pathways control many of the psychological functions that are disturbed in depression and in the perception of pain. On the other hand, the descending pathways, from monoaminergic nuclei to the spinal cord, are specifically implicated in the inhibition of nociception providing rationale for the use of serotonin (5-HT) and/or norepinephrine (NE) reuptake inhibitors (SSRIs, NRIs, SNRIs), in the relief of pain. Compelling evidence suggests that dopamine (DA) is also involved in the pathophysiology and treatment of depression. Indeed, recent insights have demonstrated a central role for DA in analgesia through an action at both the spinal and suprasinal levels including brain regions such as the periaqueductal grey (PAG), the thalamus, the basal ganglia and the limbic system. In this context, dopaminergic antidepressants (i.e., containing dopaminergic activity), such as bupropion, nomifensine and more recently triple reuptake inhibitors (TRIs), might represent new promising therapeutic tools in the treatment of painful symptoms with depression. Nevertheless, whether the addition of the dopaminergic component produces more robust effects than single- or dual-acting agents, has yet to be demonstrated. This article reviews the main pathways regulating pain transmission in relation with the monoaminergic systems. It then focuses on the current knowledge regarding the in vivo pharmacological properties and mechanism of action of monoaminergic antidepressants including SSRIs, NRIs, SNRIs and TRIs. Finally, a synthesis of the preclinical studies supporting the efficacy of these antidepressants in analgesia is also addressed in order to highlight the relative contribution of 5-HT, NE and DA to nociception.
Collapse
Affiliation(s)
- Guillaume Hache
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: 011-331-46-83-53-61
| | | | | | | |
Collapse
|
2723
|
Viana F. Chemosensory properties of the trigeminal system. ACS Chem Neurosci 2011; 2:38-50. [PMID: 22778855 PMCID: PMC3369707 DOI: 10.1021/cn100102c] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/14/2010] [Indexed: 11/28/2022] Open
Abstract
The capacity of cutaneous, including trigeminal endings, to detect chemicals is known as chemesthesis or cutaneous chemosensation. This sensory function involves the activation of nociceptor and thermoreceptor endings and has a protective or defensive function, as many of these substances are irritants or poisonous. However, humans have also developed a liking for the distinct sharpness or pungency of many foods, beverages, and spices following activation of the same sensory afferents. Our understanding of the cellular and molecular mechanisms of chemosensation in the trigeminal system has experienced enormous progress in the past decade, following the cloning and functional characterization of several ion channels activated by physical and chemical stimuli. This brief review attempts to summarize our current knowledge in this field, including a functional description of various sensory channels, especially TRP channels, involved in trigeminal chemosensitivy. Finally, some of these new findings are discussed in the context of the pathophysiology of trigeminal chemosensation, including pain, pruritus, migraine, cough, airway inflammation, and ophthalmic diseases.
Collapse
Affiliation(s)
- Félix Viana
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550, San Juan de Alicante, Spain.
| |
Collapse
|
2724
|
Jiménez E, Zafra F, Pérez-Sen R, Delicado EG, Miras-Portugal MT, Aragón C, López-Corcuera B. P2Y purinergic regulation of the glycine neurotransmitter transporters. J Biol Chem 2011; 286:10712-24. [PMID: 21245148 DOI: 10.1074/jbc.m110.167056] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The sodium- and chloride-coupled glycine neurotransmitter transporters (GLYTs) control the availability of glycine at glycine-mediated synapses. The mainly glial GLYT1 is the key regulator of the glycine levels in glycinergic and glutamatergic pathways, whereas the neuronal GLYT2 is involved in the recycling of synaptic glycine from the inhibitory synaptic cleft. In this study, we report that stimulation of P2Y purinergic receptors with 2-methylthioadenosine 5'-diphosphate in rat brainstem/spinal cord primary neuronal cultures and adult rat synaptosomes leads to the inhibition of GLYT2 and the stimulation of GLYT1 by a paracrine regulation. These effects are mainly mediated by the ADP-preferring subtypes P2Y(1) and P2Y(13) because the effects are partially reversed by the specific antagonists N(6)-methyl-2'-deoxyadenosine-3',5'-bisphosphate and pyridoxal-5'-phosphate-6-azo(2-chloro-5-nitrophenyl)-2,4-disulfonate and are totally blocked by suramin. P2Y(12) receptor is additionally involved in GLYT1 stimulation. Using pharmacological approaches and siRNA-mediated protein knockdown methodology, we elucidate the molecular mechanisms of GLYT regulation. Modulation takes place through a signaling cascade involving phospholipase C activation, inositol 1,4,5-trisphosphate production, intracellular Ca(2+) mobilization, protein kinase C stimulation, nitric oxide formation, cyclic guanosine monophosphate production, and protein kinase G-I (PKG-I) activation. GLYT1 and GLYT2 are differentially sensitive to NO/cGMP/PKG-I both in brain-derived preparations and in heterologous systems expressing the recombinant transporters and P2Y(1) receptor. Sensitivity to 2-methylthioadenosine 5'-diphosphate by GLYT1 and GLYT2 was abolished by small interfering RNA (siRNA)-mediated knockdown of nitric-oxide synthase. Our data may help define the role of GLYTs in nociception and pain sensitization.
Collapse
Affiliation(s)
- Esperanza Jiménez
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
2725
|
Yao J, Liu B, Qin F. Kinetic and energetic analysis of thermally activated TRPV1 channels. Biophys J 2011; 99:1743-53. [PMID: 20858418 DOI: 10.1016/j.bpj.2010.07.022] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 06/25/2010] [Accepted: 07/02/2010] [Indexed: 12/27/2022] Open
Abstract
Thermal TRP channels are important for thermal sensation and nociception, but their gating mechanisms have remained elusive. With optically generated submillisecond temperature steps from 22°C to >60°C, we have directly measured the activation and deactivation kinetics of TRPV1 channels, and from the measurements we determined the energetics of thermal gating. We show that activation by temperature follows single exponential time courses. It occurs in a few milliseconds and is significantly faster than activation by agonists. The gating has characteristics of a melting process involving large compensatory enthalpy (>100 kcal/mol) and entropy changes with little free energy change. The reaction path is asymmetrical with temperature mainly driving the opening while the closing has nominal but negative temperature dependence (i.e., sensitivity to cold). Both voltage and agonists alter the slope of the temperature-dependent gating curve as well as shifting the midpoint. However, compared to the energetic effect of temperature on gating, the effect of voltage is small. Our data on the interdependence between voltage and direct temperature responses are not fit to a model involving independent stimuli but instead support a temperature-sensing mechanism that is coupled to charge movement or agonist binding.
Collapse
Affiliation(s)
- Jing Yao
- Department of Physiology and Biophysical Sciences, State University of New York, Buffalo, New York, USA
| | | | | |
Collapse
|
2726
|
Gong N, Gao ZY, Wang YC, Li XY, Huang JL, Hashimoto K, Wang YX. A series of D-amino acid oxidase inhibitors specifically prevents and reverses formalin-induced tonic pain in rats. J Pharmacol Exp Ther 2011; 336:282-93. [PMID: 20952482 DOI: 10.1124/jpet.110.172353] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We have found that mutation of D-amino acid oxidase (DAO) diminished formalin-induced tonic pain. The present research further studied the analgesic effects of a series of DAO inhibitors in this model. 5-Chlorobenzo[d]isoxazol-3-ol (CBIO), 4H-thieno[3,2-b]pyrrole-5-carboxylic acid (compound 8), 5-methylpyrazole-3-carboxylic acid (AS057278), sodium benzoate, and 4-nitro-3-pyrazole carboxylic acid (NPCA) inhibited rat spinal cord-derived DAO activity in a concentration-dependent manner, with maximal inhibition of 100% and potency rank of CBIO > compound 8 > AS057278 > sodium benzoate > NPCA. In rats, intrathecal injections of CBIO, compound 8, AS057278, and sodium benzoate but not NPCA specifically prevented formalin-induced tonic pain but not acute nociception, with the same potency order as in the DAO activity assay. The highly potent analgesia of DAO inhibitors was evidenced by CBIO, which prevented 50% pain at 0.06 μg, approximately 5-fold the potency of morphine. CBIO given after formalin challenge also reversed the established pain state to the same degree as prevention. The antihyperalgesic potencies of these DAO inhibitors were highly correlated to their inhibitions of spinal DAO activity. Maximum inhibition of pain by these compounds was approximately 60%, comparable with that of the N-methyl-D-aspartic acid receptor antagonist (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801), suggesting that a larger portion of formalin-induced tonic pain is "DAO-sensitive," whereas the remaining 40% of tonic pain and acute nociception is "DAO-insensitive." These findings, combined with our previous DAO gene mutation and induction results, indicate spinal DAO mediates both induction and maintenance of formalin-induced tonic pain and further validate spinal DAO as a novel and efficacious target molecule for the treatment of chronic pain.
Collapse
Affiliation(s)
- Nian Gong
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, No. 6 Biomedicine Building (Suite 106), 800 Dongchuan Road, Shanghai 2002 40, China
| | | | | | | | | | | | | |
Collapse
|
2727
|
Cheng CF, Chen IL, Cheng MH, Lian WS, Lin CC, Kuo TB, Chen CC. Acid-Sensing Ion Channel 3, But Not Capsaicin Receptor TRPV1, Plays a Protective Role in Isoproterenol-Induced Myocardial Ischemia in Mice. Circ J 2011; 75:174-8. [DOI: 10.1253/circj.cj-10-0490] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ching-Feng Cheng
- Institute of Biomedical Sciences, Academia Sinica
- Department of Medical Research, Tzu Chi General Hospital and Department of Pediatrics, Tzu Chi University
| | - I-Lun Chen
- Institute of Biomedical Sciences, Academia Sinica
- Institute of Zoology, National Taiwan University
| | | | | | | | | | | |
Collapse
|
2728
|
Iadarola MJ, Mannes AJ. The vanilloid agonist resiniferatoxin for interventional-based pain control. Curr Top Med Chem 2011; 11:2171-9. [PMID: 21671877 PMCID: PMC4289604 DOI: 10.2174/156802611796904942] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 11/26/2010] [Indexed: 11/22/2022]
Abstract
The idea of selectively targeting nociceptive transmission at the level of the peripheral nervous system is attractive from multiple perspectives, particularly the potential lack of non-specific (non-targeted) CNS side effects. Out of the multiple TRP channels involved in nociception, TRPV1 is a strong candidate based on its biophysical conductance properties and its expression in inflammation-sensitive dorsal root ganglion neurons and their axons and central and peripheral nerve terminals. While TRPV1 antagonists have undergone extensive medicinal chemical and pharmacological investigation, for TRPV1 agonists nature has provided an optimized compound in RTX. RTX is not suitable for systemic administration, but it is highly adaptable to a variety of pain problems when used by local administration. This can include routes as diverse as subcutaneous, intraganglionic or intrathecal (CSF space around the spinal cord). The present review focuses on the molecular and preclinical animal experiments that form the underpinnings of our clinical trial of intrathecal RTX for pain in advanced cancer. As such this represents a new approach to pain control that emerges from a long line of research on capsaicin and other vanilloids, their physiological actions, and the molecular biology of the capsaicin receptor TRPV1.
Collapse
Affiliation(s)
- Michael J. Iadarola
- Neurobiology and Pain Therapeutics Section, Laboratory of Sensory Biology, National Institute of Dental and Cranio-facial Research, National Institutes of Health, Bethesda MD 20892, USA
| | - Andrew J. Mannes
- Neurobiology and Pain Therapeutics Section, Laboratory of Sensory Biology, National Institute of Dental and Cranio-facial Research, National Institutes of Health, Bethesda MD 20892, USA
| |
Collapse
|
2729
|
Nociceptors, Pain, and Spinal Manipulation. Pain Manag 2011. [DOI: 10.1016/b978-1-4377-0721-2.00137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
2730
|
Binshtok AM. Mechanisms of nociceptive transduction and transmission: a machinery for pain sensation and tools for selective analgesia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 97:143-77. [PMID: 21708310 DOI: 10.1016/b978-0-12-385198-7.00006-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many surgical and dental procedures depend on use of local anesthetics to reversibly eliminate pain. By the blockade of voltage-gated sodium channels, local anesthetics prevent the transmission of nociceptive information. However, since all local anesthetics act non-selectively on all types of axons they also cause a loss of innocuous sensation, motor paralysis and autonomic block. Thus, approaches that produce only a selective blockade of pain fibers are of great potential clinical importance. In this chapter we will review the recent findings describing mechanisms of pain transduction and transmission and introduce novel therapeutic approaches to produce pain-selective analgesia.
Collapse
Affiliation(s)
- Alexander M Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada and Center for Research on Pain, The Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
2731
|
Affiliation(s)
- Cheng-Han Lee
- Institute of Neuroscience, National Yang-Ming University
- Institute of Biomedical Sciences, Academia Sinica
| | - Synthia H. Sun
- Institute of Neuroscience, National Yang-Ming University
| | - Shing-Hong Lin
- Institute of Biomedical Sciences, Academia Sinica
- Graduate Institute of Life Sciences, National Defense Medical Center
| | - Chih-Cheng Chen
- Institute of Neuroscience, National Yang-Ming University
- Institute of Biomedical Sciences, Academia Sinica
| |
Collapse
|
2732
|
Abstract
Transient receptor potential (TRP) channels represent a superfamily of cation channels found in all eukaryotes. The C. elegans genome encodes seventeen TRP channels covering all of the seven TRP subfamilies. Genetic analyses in C. elegans have implicated TRP channels in a wide spectrum of behavioral and physiological processes, ranging from sensory transduction (e.g. chemosensation, touch sensation, proprioception and osmosensation) to fertilization, drug dependence, organelle biogenesis, apoptosis, gene expression, and neurotransmitter/hormone release. Many C. elegans TRP channels share similar activation and regulatory mechanisms with their vertebrate counterparts. Studies in C. elegans have also revealed some previously unrecognized functions and regulatory mechanisms of TRP channels. C. elegans represents an excellent genetic model organism for the study of function and regulation of TRP channels in vivo.
Collapse
Affiliation(s)
- Rui Xiao
- Department of Molecular and Integrative Physiology, Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|
2733
|
Brand A, Smith ESJ, Lewin GR, Park TJ. Functional neurokinin and NMDA receptor activity in an animal naturally lacking substance P: the naked mole-rat. PLoS One 2010; 5:e15162. [PMID: 21200438 PMCID: PMC3006196 DOI: 10.1371/journal.pone.0015162] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 10/26/2010] [Indexed: 12/19/2022] Open
Abstract
Naked mole-rats are extremely unusual among mammals in that their cutaneous C-fibers lack the neuropeptide Substance P (SP). In other mammals, SP plays an important role in nociception: it is released from C-fibers onto spinal neurons where it facilitates NMDA receptor activity and causes sensitization that can last for minutes, hours or days. In the present study, we tested the effects of intrathecal application of: 1) SP, 2) an SP antagonist (GR-82334), and 3) an NMDA antagonist (APV) on heat-evoked foot withdrawal. In the naked mole-rat, at a high enough concentration, application of SP caused a large, immediate, and long-lasting sensitization of foot withdrawal latency that was transiently reversed by application of either antagonist. However, neither SP nor NMDA antagonists had an effect when administered alone to naïve animals. In contrast, both antagonists induced an increase in basal withdrawal latency in mice. These results indicate that spinal neurons in naked mole-rats have functional SP and NMDA receptors, but that these receptors do not participate in heat-evoked foot withdrawal unless SP is experimentally introduced. We propose that the natural lack of SP in naked mole-rat C-fibers may have resulted during adaptation to living in a chronically high carbon dioxide, high ammonia environment that, in other mammals, would stimulate C-fibers and evoke nocifensive behavior.
Collapse
Affiliation(s)
- Antje Brand
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ewan St. J. Smith
- Department of Neuroscience, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gary R. Lewin
- Department of Neuroscience, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Thomas J. Park
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
2734
|
Liu Y, Abdel Samad O, Zhang L, Duan B, Tong Q, Lopes C, Ji RR, Lowell BB, Ma Q. VGLUT2-dependent glutamate release from nociceptors is required to sense pain and suppress itch. Neuron 2010; 68:543-56. [PMID: 21040853 DOI: 10.1016/j.neuron.2010.09.008] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2010] [Indexed: 12/12/2022]
Abstract
Itch can be suppressed by painful stimuli, but the underlying neural basis is unknown. We generated conditional null mice in which vesicular glutamate transporter type 2 (VGLUT2)-dependent synaptic glutamate release from mainly Nav1.8-expressing nociceptors was abolished. These mice showed deficits in pain behaviors, including mechanical pain, heat pain, capsaicin-evoked pain, inflammatory pain, and neuropathic pain. The pain deficits were accompanied by greatly enhanced itching, as suggested by (1) sensitization of both histamine-dependent and histamine-independent itch pathways and (2) development of spontaneous scratching and skin lesions. Strikingly, intradermal capsaicin injection promotes itch responses in these mutant mice, as opposed to pain responses in control littermates. Consequently, coinjection of capsaicin was no longer able to mask itch evoked by pruritogenic compounds. Our studies suggest that synaptic glutamate release from a group of peripheral nociceptors is required to sense pain and suppress itch. Elimination of VGLUT2 in these nociceptors creates a mouse model of chronic neurogenic itch.
Collapse
Affiliation(s)
- Yang Liu
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, 1 Jimmy Fund Way, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
2735
|
Administration of a tropomyosin receptor kinase inhibitor attenuates sarcoma-induced nerve sprouting, neuroma formation and bone cancer pain. Mol Pain 2010; 6:87. [PMID: 21138586 PMCID: PMC3004846 DOI: 10.1186/1744-8069-6-87] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 12/07/2010] [Indexed: 12/20/2022] Open
Abstract
Pain often accompanies cancer and most current therapies for treating cancer pain have significant unwanted side effects. Targeting nerve growth factor (NGF) or its cognate receptor tropomyosin receptor kinase A (TrkA) has become an attractive target for attenuating chronic pain. In the present report, we use a mouse model of bone cancer pain and examine whether oral administration of a selective small molecule Trk inhibitor (ARRY-470, which blocks TrkA, TrkB and TrkC kinase activity at low nm concentrations) has a significant effect on cancer-induced pain behaviors, tumor-induced remodeling of sensory nerve fibers, tumor growth and tumor-induced bone remodeling. Early/sustained (initiated day 6 post cancer cell injection), but not late/acute (initiated day 18 post cancer cell injection) administration of ARRY-470 markedly attenuated bone cancer pain and significantly blocked the ectopic sprouting of sensory nerve fibers and the formation of neuroma-like structures in the tumor bearing bone, but did not have a significant effect on tumor growth or bone remodeling. These data suggest that, like therapies that target the cancer itself, the earlier that the blockade of TrkA occurs, the more effective the control of cancer pain and the tumor-induced remodeling of sensory nerve fibers. Developing targeted therapies that relieve cancer pain without the side effects of current analgesics has the potential to significantly improve the quality of life and functional status of cancer patients.
Collapse
|
2736
|
VGLUT2 expression in primary afferent neurons is essential for normal acute pain and injury-induced heat hypersensitivity. Proc Natl Acad Sci U S A 2010; 107:22296-301. [PMID: 21135246 DOI: 10.1073/pnas.1013413108] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Dorsal root ganglia (DRG) neurons, including the nociceptors that detect painful thermal, mechanical, and chemical stimuli, transmit information to spinal cord neurons via glutamatergic and peptidergic neurotransmitters. However, the specific contribution of glutamate to pain generated by distinct sensory modalities or injuries is not known. Here we generated mice in which the vesicular glutamate transporter 2 (VGLUT2) is ablated selectively from DRG neurons. We report that conditional knockout (cKO) of the Slc17a6 gene encoding VGLUT2 from the great majority of nociceptors profoundly decreased VGLUT2 mRNA and protein in these neurons, and reduced firing of lamina I spinal cord neurons in response to noxious heat and mechanical stimulation. In behavioral assays, cKO mice showed decreased responsiveness to acute noxious heat, mechanical, and chemical (capsaicin) stimuli, but responded normally to cold stimulation and in the formalin test. Strikingly, although tissue injury-induced heat hyperalgesia was lost in the cKO mice, mechanical hypersensitivity developed normally. In a model of nerve injury-induced neuropathic pain, the magnitude of heat hypersensitivity was diminished in cKO mice, but both the mechanical allodynia and the microgliosis generated by nerve injury were intact. These findings suggest that VGLUT2 expression in nociceptors is essential for normal perception of acute pain and heat hyperalgesia, and that heat and mechanical hypersensitivity induced by peripheral injury rely on distinct (VGLUT2 dependent and VGLUT2 independent, respectively) primary afferent mechanisms and pathways.
Collapse
|
2737
|
Richter F, Natura G, Löser S, Schmidt K, Viisanen H, Schaible HG. Tumor necrosis factor causes persistent sensitization of joint nociceptors to mechanical stimuli in rats. ACTA ACUST UNITED AC 2010; 62:3806-14. [DOI: 10.1002/art.27715] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
2738
|
Joseph EK, Levine JD. Mu and delta opioid receptors on nociceptors attenuate mechanical hyperalgesia in rat. Neuroscience 2010; 171:344-50. [PMID: 20736053 PMCID: PMC2956864 DOI: 10.1016/j.neuroscience.2010.08.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 08/16/2010] [Accepted: 08/18/2010] [Indexed: 11/15/2022]
Abstract
Sensitization to mechanical stimuli is important in most pain syndromes. We evaluated the populations of nociceptors mediating mechanical hyperalgesia and those mediating mu-opioid receptor (MOR) and delta-opioid receptor (DOR) agonist-induced inhibition of hyperalgesia, in the rat. We found that: (1) intradermal injection of both the endogenous ligand for the Ret receptor, glia-derived growth factor (GDNF), and the ligand for the tropomyosin receptor kinase A (TrkA) receptor, nerve growth factor (NGF)-which are present on distinct populations of nociceptors-both produce mechanical hyperalgesia; (2) DOR agonist 4-[(R)-[(2S,5R)-4-allyl-2,5-dimethylpiperazin-1-yl](3-methoxyphenyl)methyl]-N,N-diethylbenzamide (SNC) but not MOR agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) inhibit GDNF-induced hyperalgesia; (3) both DAMGO and SNC inhibit NGF hyperalgesia, even in rats pretreated with isolectin B4 (IB4)-saporin, a toxin that destroys IB4-binding neurons; (4) co-administration of low doses of DAMGO and SNC produce enhanced analgesia, and; (5) repeated administration of DAMGO produces cross-tolerance to the analgesic effect of SNC. These findings demonstrate that, most nociceptors have a role in mechanical hyperalgesia, only the DOR agonist inhibits GDNF hyperalgesia, and MOR and DOR are co-localized on a functionally important population of TrkA-positive nociceptors.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/therapeutic use
- Animals
- Cholera Toxin/metabolism
- Cysteine/adverse effects
- Cysteine/analogs & derivatives
- Disease Models, Animal
- Drug Synergism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/therapeutic use
- Glial Cell Line-Derived Neurotrophic Factor/adverse effects
- Horseradish Peroxidase/metabolism
- Hyperalgesia/chemically induced
- Hyperalgesia/drug therapy
- Hyperalgesia/metabolism
- Hyperalgesia/pathology
- Male
- Nociceptors/metabolism
- Pain Threshold/drug effects
- Pain Threshold/physiology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/metabolism
- S-Nitrosothiols/adverse effects
- Vasodilator Agents/adverse effects
Collapse
Affiliation(s)
- E K Joseph
- Division of Neuroscience, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | |
Collapse
|
2739
|
Hematopoietic colony-stimulating factors: new players in tumor-nerve interactions. J Mol Med (Berl) 2010; 89:321-9. [PMID: 21079906 PMCID: PMC3055988 DOI: 10.1007/s00109-010-0697-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Revised: 10/02/2010] [Accepted: 10/28/2010] [Indexed: 12/30/2022]
Abstract
A variety of cancers are accompanied by debilitating pain, which constitutes the primary reason for poor quality of life in cancer patients. There is an urgent demand for the development of specific mechanism-based therapies against cancer pain. Recently, important advances have been made in mechanisms contributing to cancer pain. A notable finding was that the tumor-derived hematopoietic growth factors, granulocyte- and granulocyte-macrophage-colony-stimulating factors (G-CSF/GM-CSF), subserve important functions in the generation of pain hypersensitivity in tumor-affected regions. In this context, their receptors were unexpectedly found on pain-sensing nerves and were observed to be functionally linked to nociceptive sensitization and tumor-induced pain. Here, we review evidence supporting a role for G-/GM-CSF in sensitization of pain-sensing nerves, the underlying signaling pathways and the cross-talk with other pronociceptive cytokines, peptides and modulators derived from immune cells, osteoclasts and tumor cells. These findings hold implications in the therapy of pain in disease states, such as cancer and rheumatoid arthritis.
Collapse
|
2740
|
Neely GG, Hess A, Costigan M, Keene AC, Goulas S, Langeslag M, Griffin RS, Belfer I, Dai F, Smith S, Diatchenko L, Gupta V, Xia CP, Amann S, Kreitz S, Heindl-Erdmann C, Wolz S, Ly CV, Arora S, Sarangi R, Dan D, Novatchkova M, Rosenzweig M, Gibson D, Truong D, Schramek D, Zoranovic T, Cronin SJF, Angjeli B, Brune K, Dietzl G, Maixner W, Meixner A, Thomas W, Pospisilik JA, Alenius M, Kress M, Subramaniam S, Garrity PA, Bellen HJ, Woolf CJ, Penninger JM. A genome-wide Drosophila screen for heat nociception identifies α2δ3 as an evolutionarily conserved pain gene. Cell 2010; 143:628-38. [PMID: 21074052 PMCID: PMC3040441 DOI: 10.1016/j.cell.2010.09.047] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 08/09/2010] [Accepted: 09/24/2010] [Indexed: 02/02/2023]
Abstract
Worldwide, acute, and chronic pain affects 20% of the adult population and represents an enormous financial and emotional burden. Using genome-wide neuronal-specific RNAi knockdown in Drosophila, we report a global screen for an innate behavior and identify hundreds of genes implicated in heat nociception, including the α2δ family calcium channel subunit straightjacket (stj). Mice mutant for the stj ortholog CACNA2D3 (α2δ3) also exhibit impaired behavioral heat pain sensitivity. In addition, in humans, α2δ3 SNP variants associate with reduced sensitivity to acute noxious heat and chronic back pain. Functional imaging in α2δ3 mutant mice revealed impaired transmission of thermal pain-evoked signals from the thalamus to higher-order pain centers. Intriguingly, in α2δ3 mutant mice, thermal pain and tactile stimulation triggered strong cross-activation, or synesthesia, of brain regions involved in vision, olfaction, and hearing.
Collapse
Affiliation(s)
- G. Gregory Neely
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
- Neuroscience Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - Andreas Hess
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Michael Costigan
- Program in Neurobiology, Children’s Hospital Boston and Department of Neurobiology Harvard Medical School Boston, MA 02115
| | - Alex C. Keene
- Biology Department, NYU, 100 Washington Square East, New York, NY 10003
| | - Spyros Goulas
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Michiel Langeslag
- Div. Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Fritz-Pregl-Strasse 3, A-6020 Innsbruck, Austria
| | - Robert S Griffin
- Neural Plasticity Research Group, Department of Anesthesia and Critical Care, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Inna Belfer
- Molecular epidemiology of Pain Program, Dept. of Anaesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Feng Dai
- Molecular epidemiology of Pain Program, Dept. of Anaesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shad Smith
- Center for Neurosensory Disorders, University of North Carolina at Chapel Hill, NC, USA
| | - Luda Diatchenko
- Center for Neurosensory Disorders, University of North Carolina at Chapel Hill, NC, USA
| | - Vaijayanti Gupta
- Strand Life Sciences Pvt. Ltd., 237 C V Raman Avenue, Rajmahal Vilas, Bangalore, India
| | - Cui-ping Xia
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Sabina Amann
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Silke Kreitz
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Cornelia Heindl-Erdmann
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Susanne Wolz
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Cindy V. Ly
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Suchir Arora
- Strand Life Sciences Pvt. Ltd., 237 C V Raman Avenue, Rajmahal Vilas, Bangalore, India
| | - Rinku Sarangi
- Strand Life Sciences Pvt. Ltd., 237 C V Raman Avenue, Rajmahal Vilas, Bangalore, India
| | - Debasis Dan
- Strand Life Sciences Pvt. Ltd., 237 C V Raman Avenue, Rajmahal Vilas, Bangalore, India
| | - Maria Novatchkova
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Mark Rosenzweig
- National Center for Behavioral Genomics, Department of Biology, Brandeis University, Waltham, MA 02458, USA
| | - Dustin Gibson
- Center for Neurosensory Disorders, University of North Carolina at Chapel Hill, NC, USA
| | - Darwin Truong
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Daniel Schramek
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Tamara Zoranovic
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Shane J. F. Cronin
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Belinda Angjeli
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Kay Brune
- Department of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nuremberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Georg Dietzl
- Howard Hughes Medical Institute, Stanford University, 318 Campus Drive, Stanford, California 94305, USA
| | - William Maixner
- Center for Neurosensory Disorders, University of North Carolina at Chapel Hill, NC, USA
| | - Arabella Meixner
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| | - Winston Thomas
- Deltagen, Inc. 1900 South Norfolk St., Suite 105, San Mateo, CA 9440
| | | | - Mattias Alenius
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping, SE-581 85, Sweden
| | - Michaela Kress
- Div. Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Fritz-Pregl-Strasse 3, A-6020 Innsbruck, Austria
| | - Sai Subramaniam
- Strand Life Sciences Pvt. Ltd., 237 C V Raman Avenue, Rajmahal Vilas, Bangalore, India
| | - Paul A. Garrity
- National Center for Behavioral Genomics, Department of Biology, Brandeis University, Waltham, MA 02458, USA
| | - Hugo J. Bellen
- HHMI, Department of Molecular and Human Genetics, Department of Neuroscience, and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Clifford J. Woolf
- Program in Neurobiology, Children’s Hospital Boston and Department of Neurobiology Harvard Medical School Boston, MA 02115
| | - Josef M. Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3-5, A-1030 Vienna
| |
Collapse
|
2741
|
Abstract
The description of itch (formally known as pruritus) as an "unpleasant sensation that elicits the desire or reflex to scratch" (Ikoma et al., 2006) is immediately familiar. Research in the field of pruritoception has added to our understanding of this area of sensory neurobiology as it pertains to both normal and pathological conditions. In particular, much progress has been made on the mechanisms and circuits of itch, which we review here.
Collapse
Affiliation(s)
- Kush N. Patel
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Howard Hughes Medical Institute
| |
Collapse
|
2742
|
Ma Q. Labeled lines meet and talk: population coding of somatic sensations. J Clin Invest 2010; 120:3773-8. [PMID: 21041959 DOI: 10.1172/jci43426] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The somatic sensory system responds to stimuli of distinct modalities, including touch, pain, itch, and temperature sensitivity. In the past century, great progress has been made in understanding the coding of these sensory modalities. From this work, two major features have emerged. First, there are specific neuronal circuits or labeled lines transmitting specific sensory information from the skin to the brain. Second, the generation of specific sensations often involves crosstalk among distinct labeled lines. These features suggest that population coding is the mechanism underlying somatic sensation.
Collapse
Affiliation(s)
- Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
2743
|
Abstract
To paraphrase Cole Porter's famous 1926 song, "What is this thing called pain? This funny thing called pain, just who can solve its mystery?" Pain, like love, is all consuming: when you have it, not much else matters, and there is nothing you can do about it. Unlike love, however, we are actually beginning to tease apart the mystery of pain. The substantial progress made over the last decade in revealing the genes, molecules, cells, and circuits that determine the sensation of pain offers new opportunities to manage it, as revealed in this Review series by some of the foremost experts in the field.
Collapse
Affiliation(s)
- Clifford J Woolf
- Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
2744
|
Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest 2010; 120:3760-72. [PMID: 21041958 DOI: 10.1172/jci42843] [Citation(s) in RCA: 786] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Specialized peripheral sensory neurons known as nociceptors alert us to potentially damaging stimuli at the skin by detecting extremes in temperature and pressure and injury-related chemicals, and transducing these stimuli into long-ranging electrical signals that are relayed to higher brain centers. The activation of functionally distinct cutaneous nociceptor populations and the processing of information they convey provide a rich diversity of pain qualities. Current work in this field is providing researchers with a more thorough understanding of nociceptor cell biology at molecular and systems levels and insight that will allow the targeted design of novel pain therapeutics.
Collapse
Affiliation(s)
- Adrienne E Dubin
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, USA.
| | | |
Collapse
|
2745
|
Abstract
To paraphrase Cole Porter's famous 1926 song, "What is this thing called pain? This funny thing called pain, just who can solve its mystery?" Pain, like love, is all consuming: when you have it, not much else matters, and there is nothing you can do about it. Unlike love, however, we are actually beginning to tease apart the mystery of pain. The substantial progress made over the last decade in revealing the genes, molecules, cells, and circuits that determine the sensation of pain offers new opportunities to manage it, as revealed in this Review series by some of the foremost experts in the field.
Collapse
Affiliation(s)
- Clifford J Woolf
- Department of Neurology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
2746
|
Dimitrov EL, Petrus E, Usdin TB. Tuberoinfundibular peptide of 39 residues (TIP39) signaling modulates acute and tonic nociception. Exp Neurol 2010; 226:68-83. [PMID: 20696160 PMCID: PMC2955778 DOI: 10.1016/j.expneurol.2010.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 07/26/2010] [Accepted: 08/02/2010] [Indexed: 11/19/2022]
Abstract
Tuberoinfundibular peptide of 39 residues (TIP39) synthesizing neurons at the caudal border of the thalamus and in the lateral pons project to areas rich in its receptor, the parathyroid hormone 2 receptor (PTH2R). These areas include many involved in processing nociceptive information. Here we examined the potential role of TIP39 signaling in nociception using a PTH2R antagonist (HYWH) and mice with deletion of TIP39's coding sequence or PTH2R null mutation. Intracerebroventricular (icv) infusion of HYWH significantly inhibited nociceptive responses in tail-flick and hot-plate tests and attenuated the nociceptive response to hindpaw formalin injection. TIP39-KO and PTH2R-KO had increased response latency in the 55°C hot-plate test and reduced responses in the hindpaw formalin test. The tail-flick test was not affected in either KO line. Thermal hypoalgesia in KO mice was dose-dependently reversed by systemic administration of the cannabinoid receptor 1 (CB1) antagonist rimonabant, which did not affect nociception in wild-type (WT). Systemic administration of the cannabinoid agonist CP 55,940 did not affect nociception in KO mice at a dose effective in WT. WT mice administered HYWH icv, and both KOs, had significantly increased stress-induced analgesia (SIA). Rimonabant blocked the increased SIA in TIP39-KO, PTH2R-KO or after HYWH infusion. CB1 and FAAH mRNA were decreased and increased, respectively, in the basolateral amygdala of TIP39-KO mice. These data suggest that TIP39 signaling modulates nociception, very likely by inhibiting endocannabinoid circuitry at a supraspinal level. We infer a new central mechanism for endocannabinoid regulation, via TIP39 acting on the PTH2R in discrete brain regions.
Collapse
MESH Headings
- Amidohydrolases/metabolism
- Animals
- Cannabinoid Receptor Modulators/metabolism
- Formaldehyde
- In Situ Hybridization
- Injections, Intraventricular
- Injections, Spinal
- Male
- Mice
- Mice, Knockout
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Neuropeptides/administration & dosage
- Neuropeptides/pharmacology
- Nociceptors/drug effects
- Nociceptors/physiology
- Pain/pathology
- Pain/physiopathology
- Pain Measurement/drug effects
- Piperidines/pharmacology
- Pyrazoles/pharmacology
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/biosynthesis
- Receptor, Parathyroid Hormone, Type 2/biosynthesis
- Receptor, Parathyroid Hormone, Type 2/genetics
- Rimonabant
- Signal Transduction/physiology
- Stress, Psychological/psychology
- Synapses/physiology
- Vesicular Glutamate Transport Protein 2/biosynthesis
- Vesicular Glutamate Transport Protein 2/genetics
Collapse
Affiliation(s)
- Eugene L. Dimitrov
- Section on Fundamental Neuroscience, National Institute of Mental Health, 35 Convent Drive, Room 1B-213, Bethesda, MD 20892, USA
| | - Emily Petrus
- Section on Fundamental Neuroscience, National Institute of Mental Health, 35 Convent Drive, Room 1B-215, Bethesda, MD 20892, USA
| | - Ted B. Usdin
- Section on Fundamental Neuroscience, National Institute of Mental Health, 35 Convent Drive, Room 1B-215, Bethesda, MD 20892, USA
| |
Collapse
|
2747
|
Gascon E, Moqrich A. Heterogeneity in primary nociceptive neurons: From molecules to pathology. Arch Pharm Res 2010; 33:1489-507. [DOI: 10.1007/s12272-010-1003-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 08/19/2010] [Accepted: 08/20/2010] [Indexed: 01/17/2023]
|
2748
|
Multiple desensitization mechanisms of mechanotransducer channels shape firing of mechanosensory neurons. J Neurosci 2010; 30:13384-95. [PMID: 20926665 DOI: 10.1523/jneurosci.2926-10.2010] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
How desensitization of mechanotransducer currents regulates afferent signal generation in mammalian sensory neurons is essentially unknown. Here, we dissected desensitization mechanisms of mechanotransducer channels in rat sensory neurons that mediate the sense of touch and pain. We identified four types of mechanotransducer currents that distribute differentially in cutaneous nociceptors and mechanoreceptors and that differ in desensitization rates. Desensitization of mechanotransducer channels in mechanoreceptors was fast and mediated by channel inactivation and adaptation, which reduces the mechanical force sensed by the transduction channel. Both processes were promoted by negative voltage. These properties of mechanotransducer channels suited them to encode the dynamic parameters of the stimulus. In contrast, inactivation and adaptation of mechanotransducer channels in nociceptors had slow time courses and were suited to encode duration of the stimulus. Thus, desensitization properties of mechanotransducer currents relate to their functions as sensors of phasic and tonic stimuli and enable sensory neurons to achieve efficient stimulus representation.
Collapse
|
2749
|
|
2750
|
Farrar JT. Advances in clinical research methodology for pain clinical trials. Nat Med 2010; 16:1284-93. [DOI: 10.1038/nm.2249] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|