1
|
Luo L, Cheng Y, Wang H, Li L, Niu H, Yang Y, Zhou Q, He J, Xu J. Lidocaine-A Promising Candidate for the Treatment of Cancer-Induced Bone Pain: A Narrative Review. Adv Ther 2025; 42:2587-2605. [PMID: 40232625 DOI: 10.1007/s12325-025-03192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025]
Abstract
Pain is one of the most common symptoms in patients with cancer, with cancer-induced bone pain (CIBP) significantly affecting their quality of life. Opioids are commonly used as first-line treatments for cancer pain, but their use requires caution due to non-mechanistic analgesia and significant side effects. As a result, there is a need for new non-opioid drugs that target cancer pain through specific mechanisms. Recent studies on the anticancer effects of lidocaine have highlighted its potential benefits in both treating cancer and alleviating cancer-induced pain. This article discusses the mechanism of action and clinical applications of lidocaine in cancer pain management, and suggests new treatment approaches for patients with CIBP.
Collapse
Affiliation(s)
- Lihan Luo
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuqi Cheng
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Hanxi Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Li Li
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Hanyun Niu
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuzhu Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Qianqian Zhou
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiannan He
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| | - Jianhong Xu
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
2
|
Yang Y, Yang W, Zhang R, Wang Y. Peripheral Mechanism of Cancer-Induced Bone Pain. Neurosci Bull 2024; 40:815-830. [PMID: 37798428 PMCID: PMC11178734 DOI: 10.1007/s12264-023-01126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/28/2023] [Indexed: 10/07/2023] Open
Abstract
Cancer-induced bone pain (CIBP) is a type of ongoing or breakthrough pain caused by a primary bone tumor or bone metastasis. CIBP constitutes a specific pain state with distinct characteristics; however, it shares similarities with inflammatory and neuropathic pain. At present, although various therapies have been developed for this condition, complete relief from CIBP in patients with cancer is yet to be achieved. Hence, it is urgent to study the mechanism underlying CIBP to develop efficient analgesic drugs. Herein, we focused on the peripheral mechanism associated with the initiation of CIBP, which involves tissue injury in the bone and changes in the tumor microenvironment (TME) and dorsal root ganglion. The nerve-cancer and cancer-immunocyte cross-talk in the TME creates circumstances that promote tumor growth and metastasis, ultimately leading to CIBP. The peripheral mechanism of CIBP and current treatments as well as potential therapeutic targets are discussed in this review.
Collapse
Affiliation(s)
- Yachen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Wei Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Ruofan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Zhongshan-Fudan Joint Innovation Center, Zhongshan, 528437, China.
| |
Collapse
|
3
|
IQM-PC332, a Novel DREAM Ligand with Antinociceptive Effect on Peripheral Nerve Injury-Induced Pain. Int J Mol Sci 2022; 23:ijms23042142. [PMID: 35216258 PMCID: PMC8876042 DOI: 10.3390/ijms23042142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 02/04/2023] Open
Abstract
Neuropathic pain is a form of chronic pain arising from damage of the neural cells that sense, transmit or process sensory information. Given its growing prevalence and common refractoriness to conventional analgesics, the development of new drugs with pain relief effects constitutes a prominent clinical need. In this respect, drugs that reduce activity of sensory neurons by modulating ion channels hold the promise to become effective analgesics. Here, we evaluated the mechanical antinociceptive effect of IQM-PC332, a novel ligand of the multifunctional protein downstream regulatory element antagonist modulator (DREAM) in rats subjected to chronic constriction injury of the sciatic nerve as a model of neuropathic pain. IQM-PC332 administered by intraplantar (0.01–10 µg) or intraperitoneal (0.02–1 µg/kg) injection reduced mechanical sensitivity by ≈100% of the maximum possible effect, with ED50 of 0.27 ± 0.05 µg and 0.09 ± 0.01 µg/kg, respectively. Perforated-patch whole-cell recordings in isolated dorsal root ganglion (DRG) neurons showed that IQM-PC332 (1 and 10 µM) reduced ionic currents through voltage-gated K+ channels responsible for A-type potassium currents, low, T-type, and high voltage-activated Ca2+ channels, and transient receptor potential vanilloid-1 (TRPV1) channels. Furthermore, IQM-PC332 (1 µM) reduced electrically evoked action potentials in DRG neurons from neuropathic animals. It is suggested that by modulating multiple DREAM–ion channel signaling complexes, IQM-PC332 may serve a lead compound of novel multimodal analgesics.
Collapse
|
4
|
Trevisan G, Oliveira SM. Animal Venom Peptides Cause Antinociceptive Effects by Voltage-gated Calcium Channels Activity Blockage. Curr Neuropharmacol 2022; 20:1579-1599. [PMID: 34259147 PMCID: PMC9881091 DOI: 10.2174/1570159x19666210713121217] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022] Open
Abstract
Pain is a complex phenomenon that is usually unpleasant and aversive. It can range widely in intensity, quality, and duration and has diverse pathophysiologic mechanisms and meanings. Voltage-gated sodium and calcium channels are essential to transmitting painful stimuli from the periphery until the dorsal horn of the spinal cord. Thus, blocking voltage-gated calcium channels (VGCCs) can effectively control pain refractory to treatments currently used in the clinic, such as cancer and neuropathic pain. VGCCs blockers isolated of cobra Naja naja kaouthia (α-cobratoxin), spider Agelenopsis aperta (ω-Agatoxin IVA), spider Phoneutria nigriventer (PhTx3.3, PhTx3.4, PhTx3.5, PhTx3.6), spider Hysterocrates gigas (SNX-482), cone snails Conus geographus (GVIA), Conus magus (MVIIA or ziconotide), Conus catus (CVID, CVIE and CVIF), Conus striatus (SO- 3), Conus fulmen (FVIA), Conus moncuri (MoVIA and MoVIB), Conus regularis (RsXXIVA), Conus eburneus (Eu1.6), Conus victoriae (Vc1.1.), Conus regius (RgIA), and spider Ornithoctonus huwena (huwentoxin-I and huwentoxin-XVI) venoms caused antinociceptive effects in different acute and chronic pain models. Currently, ziconotide is the only clinical used N-type VGCCs blocker peptide for chronic intractable pain. However, ziconotide causes different adverse effects, and the intrathecal route of administration also impairs its use in a more significant number of patients. In this sense, peptides isolated from animal venoms or their synthetic forms that act by modulating or blocking VGCCs channels seem to be a relevant prototype for developing new analgesics efficacious and well tolerated by patients.
Collapse
Affiliation(s)
- Gabriela Trevisan
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil
| | - Sara Marchesan Oliveira
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil
| |
Collapse
|
5
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
6
|
Biet M, Dansereau M, Sarret P, Dumaine R. The neuronal potassium current I A is a potential target for pain during chronic inflammation. Physiol Rep 2021; 9:e14975. [PMID: 34405579 PMCID: PMC8371350 DOI: 10.14814/phy2.14975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/10/2021] [Accepted: 06/25/2021] [Indexed: 11/24/2022] Open
Abstract
Voltage-gated ion channels play a key role in the action potential (AP) initiation and its propagation in sensory neurons. Modulation of their activity during chronic inflammation creates a persistent pain state. In this study, we sought to determine how peripheral inflammation caused by complete Freund's adjuvant (CFA) alters the fast sodium (INa ), L-type calcium (ICaL ), and potassium (IK ) currents in primary afferent fibers to increase nociception. In our model, intraplantar administration of CFA induced mechanical allodynia and thermal hyperalgesia at day 14 post-injection. Using whole-cell patch-clamp recording in dissociated small (C), medium (Aδ), and large-sized (Aβ) rat dorsal root ganglion (DRG) neurons, we found that CFA prolonged the AP duration and increased the amplitude of the tetrodotoxin-resistant (TTX-r) INa in Aβ fibers. In addition, CFA accelerated the recovery of INa from inactivation in C and Aδ nociceptive fibers but enhanced the late sodium current (INaL ) only in Aδ and Aβ neurons. Inflammation similarly reduced the amplitude of ICaL in each neuronal cell type. Fourteen days after injection, CFA reduced both components of IK (IKdr and IA ) in Aδ fibers. We also found that IA was significantly larger in C and Aδ neurons in normal conditions and during chronic inflammation. Our data, therefore, suggest that targeting the transient potassium current IA represents an efficient way to shift the balance toward antinociception during inflammation, since its activation will selectively decrease the AP duration in nociceptive fibers. Altogether, our data indicate that complex interactions between IK , INa , and ICaL reduce pain threshold by concomitantly enhancing the activity of nociceptive neurons and reducing the inhibitory action of Aβ fibers during chronic inflammation.
Collapse
MESH Headings
- Action Potentials
- Animals
- Calcium Channels, L-Type/metabolism
- Cells, Cultured
- Ganglia, Spinal/cytology
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/physiology
- Male
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Neurons, Afferent/physiology
- Nociception
- Nociceptive Pain/metabolism
- Nociceptive Pain/physiopathology
- Potassium Channels, Voltage-Gated/metabolism
- Rats
- Rats, Sprague-Dawley
- Sodium Channel Blockers/pharmacology
- Sodium Channels/metabolism
- Tetrodotoxin/pharmacology
Collapse
Affiliation(s)
- Michael Biet
- Département de Pharmacologie et PhysiologieInstitut de pharmacologie de SherbrookeCentre de Recherche du Centre Hospitalier Universitaire de SherbrookeFaculté de médecine et des Sciences de la SantéUniversité de SherbrookeSherbrookeQuébecCanada
| | - Marc‐André Dansereau
- Département de Pharmacologie et PhysiologieInstitut de pharmacologie de SherbrookeCentre de Recherche du Centre Hospitalier Universitaire de SherbrookeFaculté de médecine et des Sciences de la SantéUniversité de SherbrookeSherbrookeQuébecCanada
| | - Philippe Sarret
- Département de Pharmacologie et PhysiologieInstitut de pharmacologie de SherbrookeCentre de Recherche du Centre Hospitalier Universitaire de SherbrookeFaculté de médecine et des Sciences de la SantéUniversité de SherbrookeSherbrookeQuébecCanada
| | - Robert Dumaine
- Département de Pharmacologie et PhysiologieInstitut de pharmacologie de SherbrookeCentre de Recherche du Centre Hospitalier Universitaire de SherbrookeFaculté de médecine et des Sciences de la SantéUniversité de SherbrookeSherbrookeQuébecCanada
| |
Collapse
|
7
|
Cheng J, Deng Y, Zhou J. Role of the Ubiquitin System in Chronic Pain. Front Mol Neurosci 2021; 14:674914. [PMID: 34122010 PMCID: PMC8194701 DOI: 10.3389/fnmol.2021.674914] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023] Open
Abstract
As a significant public health issue, chronic pain, mainly neuropathic pain (NP) and inflammatory pain, has a severe impact. The underlying mechanisms of chronic pain are enigmatic at present. The roles of ubiquitin have been demonstrated in various physiological and pathological conditions and underscore its potential as therapeutic targets. The dysfunction of the component of the ubiquitin system that occurs during chronic pain is rapidly being discovered. These results provide insight into potential molecular mechanisms of chronic pain. Chronic pain is regulated by ubiquitination, SUMOylation, ubiquitin ligase, and deubiquitinating enzyme (DUB), etc. Insight into the mechanism of the ubiquitin system regulating chronic pain might contribute to relevant therapeutic targets and the development of novel analgesics.
Collapse
Affiliation(s)
| | | | - Jun Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
|
9
|
Analgesic effect and related amino acids regulation of ginsenoside Rg3 in mouse pain models. Life Sci 2019; 239:117083. [DOI: 10.1016/j.lfs.2019.117083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/07/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
|
10
|
Antinociceptive activity of ethanolic extract of Azadirachta indica A. Juss (Neem, Meliaceae) fruit through opioid, glutamatergic and acid-sensitive ion pathways in adult zebrafish (Danio rerio). Biomed Pharmacother 2018; 108:408-416. [DOI: 10.1016/j.biopha.2018.08.160] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 11/20/2022] Open
|
11
|
Wu J, Peng S, Xiao L, Cheng X, Kuang H, Zhu M, Zhang D, Jiang C, Liu T. Cell-Type Specific Distribution of T-Type Calcium Currents in Lamina II Neurons of the Rat Spinal Cord. Front Cell Neurosci 2018; 12:370. [PMID: 30386213 PMCID: PMC6199353 DOI: 10.3389/fncel.2018.00370] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/28/2018] [Indexed: 12/19/2022] Open
Abstract
Spinal lamina II (substantia gelatinosa, SG) neurons integrate nociceptive information from the primary afferents and are classified according to electrophysiological (tonic firing, delayed firing, single spike, initial burst, phasic firing, gap firing and reluctant firing) or morphological (islet, central, vertical, radial and unclassified) criteria. T-type calcium (Cav3) channels play an essential role in the central mechanism of pathological pain, but the electrophysiological properties and the cell-type specific distribution of T-type channels in SG neurons have not been fully elucidated. To investigate the electrophysiological and morphological features of T-type channel-expressing or -lacking neurons, voltage- and current-clamp recordings were performed on either transverse or parasagittal spinal cord slices. Recording made in transverse spinal cord slices showed that an inward current (I T) was observed in 44.5% of the SG neurons that was fully blocked by Ni2+ and TTA-A2. The amplitude of I T depended on the magnitude and the duration of hyperpolarization pre-pulse. The voltage for eliciting and maximizing I T were -70 mV and -35 mV, respectively. In addition, we found that most of the I T-expressing neurons are tonic firing neurons and exhibit more negative action potential (AP) threshold and smaller difference of AP threshold and resting membrane potential (RMP) than those neurons lacking I T. Consistently, a specific T-type calcium channel blocker TTA-P2 increased the AP threshold and enlarged the difference between AP threshold and membrane potential (Ihold = 0). Meanwhile, the morphological analysis indicated that most of the I T-expressing neurons are islet neurons. In conclusion, we identify a cell-type specific distribution and the function of T-type channels in SG neurons. These findings might provide new insights into the mechanisms underlying the contribution of T-type channels in sensory transmission.
Collapse
Affiliation(s)
- Jing Wu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sicong Peng
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Linghui Xiao
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoe Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haixia Kuang
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mengye Zhu
- Department of Pain Clinic, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Daying Zhang
- Department of Pain Clinic, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Changyu Jiang
- Jisheng Han Academician Workstation for Pain Medicine, Nanshan Hospital, Shenzhen, China
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Jisheng Han Academician Workstation for Pain Medicine, Nanshan Hospital, Shenzhen, China.,Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
12
|
Joksimovic SL, Joksimovic SM, Tesic V, García-Caballero A, Feseha S, Zamponi GW, Jevtovic-Todorovic V, Todorovic SM. Selective inhibition of Ca V3.2 channels reverses hyperexcitability of peripheral nociceptors and alleviates postsurgical pain. Sci Signal 2018; 11:eaao4425. [PMID: 30154101 PMCID: PMC6193449 DOI: 10.1126/scisignal.aao4425] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Pain-sensing sensory neurons of the dorsal root ganglion (DRG) can become sensitized or hyperexcitable in response to surgically induced peripheral tissue injury. We investigated the potential role and molecular mechanisms of nociceptive ion channel dysregulation in acute pain conditions such as those resulting from skin and soft tissue incision. We used selective pharmacology, electrophysiology, and mouse genetics to link increased current densities arising from the CaV3.2 isoform of T-type calcium channels (T-channels) to nociceptive sensitization using a clinically relevant rodent model of skin and deep tissue incision. Furthermore, knockdown of the CaV3.2-targeting deubiquitinating enzyme USP5 or disruption of USP5 binding to CaV3.2 channels in peripheral nociceptors resulted in a robust antihyperalgesic effect in vivo and substantial T-current reduction in vitro. Our study provides mechanistic insight into the role of plasticity in CaV3.2 channel activity after surgical incision and identifies potential targets for perioperative pain that may greatly decrease the need for narcotics and potential for drug abuse.
Collapse
Affiliation(s)
- Sonja L Joksimovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
- Pharmacology Graduate Program, School of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Srdjan M Joksimovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vesna Tesic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Agustin García-Caballero
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1 Canada
| | - Simon Feseha
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1 Canada
| | - Vesna Jevtovic-Todorovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA.
- Neuroscience Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
13
|
Hidaka S, Kanai Y, Takehana S, Syoji Y, Kubota Y, Uotsu N, Yui K, Shimazu Y, Takeda M. Systemic administration of α-lipoic acid suppresses excitability of nociceptive wide-dynamic range neurons in rat spinal trigeminal nucleus caudalis. Neurosci Res 2018; 144:14-20. [PMID: 29885345 DOI: 10.1016/j.neures.2018.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/25/2018] [Accepted: 06/04/2018] [Indexed: 11/16/2022]
Abstract
Although a modulatory role has been reported for α-lipoic acid (LA) on T-type Ca2+ channels in the nervous system, the acute effects of LA in vivo, particularly on nociceptive transmission in the trigeminal system, remain to be determined. The aim of the present study was to investigate whether acute intravenous LA administration to rats attenuates the excitability of wide dynamic range (WDR) spinal trigeminal nucleus caudalis (SpVc) neurons in response to nociceptive and non-nociceptive mechanical stimulation in vivo. Extracellular single unit recordings were made from seventeen SpVc neurons in response to orofacial mechanical stimulation of pentobarbital-anesthetized rats. Responses to both non-noxious and noxious mechanical stimuli were analyzed in the present study. The mean firing frequency of SpVc WDR neurons in response to both non-noxious and noxious mechanical stimuli was significantly and dose-dependently inhibited by LA (1-100 mM, i.v.) and maximum inhibition of the discharge frequency of both non-noxious and noxious mechanical stimuli was seen within 5 min. These inhibitory effects lasted for approximately 10 min. These results suggest that acute intravenous LA administration suppresses trigeminal sensory transmission, including nociception, via possibly blocking T-type Ca2+ channels. LA may be used as a therapeutic agent for the treatment of trigeminal nociceptive pain.
Collapse
Affiliation(s)
- S Hidaka
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Y Kanai
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - S Takehana
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Y Syoji
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Y Kubota
- FANCL Health Science Research Center, Research Institute, FANCL Corporation, 12-13, Kamishinano, Totsuka-ku, Yokohama, Kanagawa, 244-0806, Japan
| | - N Uotsu
- FANCL Health Science Research Center, Research Institute, FANCL Corporation, 12-13, Kamishinano, Totsuka-ku, Yokohama, Kanagawa, 244-0806, Japan
| | - K Yui
- FANCL Health Science Research Center, Research Institute, FANCL Corporation, 12-13, Kamishinano, Totsuka-ku, Yokohama, Kanagawa, 244-0806, Japan
| | - Y Shimazu
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - M Takeda
- Laboratory of Food and Physiological Sciences, Department of Life and Food Sciences, School of Life and Environmental Sciences, Azabu University, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan.
| |
Collapse
|
14
|
Kerckhove N, Pereira B, Soriot-Thomas S, Alchaar H, Deleens R, Hieng VS, Serra E, Lanteri-Minet M, Arcagni P, Picard P, Lefebvre-Kuntz D, Maindet C, Mick G, Balp L, Lucas C, Creach C, Letellier M, Martinez V, Navez M, Delbrouck D, Kuhn E, Piquet E, Bozzolo E, Brosse C, Lietar B, Marcaillou F, Hamdani A, Leroux-Bromberg N, Perier Y, Vergne-Salle P, Gov C, Delage N, Gillet D, Romettino S, Richard D, Mallet C, Bernard L, Lambert C, Dubray C, Duale C, Eschalier A. Efficacy and safety of a T-type calcium channel blocker in patients with neuropathic pain: A proof-of-concept, randomized, double-blind and controlled trial. Eur J Pain 2018; 22:1321-1330. [PMID: 29577519 DOI: 10.1002/ejp.1221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND T-type calcium channels have been shown to play an important role in the initiation and maintenance of neuropathic pain and represent a promising therapeutic target for new analgesic treatments. Ethosuximide (ETX), an anticonvulsant and a T-type channel blocker has shown analgesic effect in several chronic pain models but has not yet been evaluated in patients with neuropathic pain. METHODS This proof-of-concept, multicentre, double-blind, controlled and randomized trial compared the efficacy and safety of ETX (given as add-on therapy) to an inactive control (IC) in 114 patients with non-diabetic peripheral neuropathic pain. After a 7-day run-in period, eligible patients aged over 18 years were randomly assigned (1:1) to ETX or IC for 6 weeks. The primary outcome was the difference between groups in the pain intensity (% of change from the baseline to end of treatment) assessed in the intention-to-treat population. This study is registered with EudraCT (2013-004801-26) and ClinicalTrials.gov (NCT02100046). RESULTS The study was stopped during the interim analysis due to the high number of adverse events in the active treatment group. ETX failed to reduce total pain and showed a poor tolerance in comparison to IC. In the per-protocol analysis, ETX significantly reduced pain intensity by 15.6% (95% CI -25.8; -5.4) from baseline compared to IC (-7.8%, 95% CI -14.3; -1.3; p = 0.033), but this result must be interpreted with caution because of a small subgroup of patients. CONCLUSION Ethosuximide did not reduce the severity of neuropathic pain and induces, at the doses used, many adverse events. SIGNIFICANCE This article shows that ETX is not effective to treat neuropathic pain. Nevertheless, per-protocol analysis suggests a possible analgesic effect of ETX. Thus, our work adds significant knowledge to preclinical and clinical data on the benefits of T-type calcium channel inhibition for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- N Kerckhove
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France.,Analgesia Institute, Université Clermont Auvergne, Clermont-Ferrand, France
| | - B Pereira
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | | | - H Alchaar
- Université Nice Côte-d'Azur, CHU Nice - Hôpital de Cimiez, Fédération Hospitalo-Universitaire INOVPAIN, CETD, Nice, France
| | | | | | - E Serra
- CHU Amiens Picardie, CETD, CRC, Amiens, France
| | - M Lanteri-Minet
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France.,Université Nice Côte-d'Azur, CHU Nice - Hôpital de Cimiez, Fédération Hospitalo-Universitaire INOVPAIN, CETD, Nice, France
| | - P Arcagni
- CHU Saint-Etienne, CETD, Saint-Etienne, France
| | - P Picard
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | | | - C Maindet
- CHU Grenoble Alpes, CETD, Grenoble, France
| | - G Mick
- CH Voiron, UETD, Voiron, France
| | - L Balp
- CH Lons-le-Saunier, CETD, Lons-le-Saunier, France
| | - C Lucas
- Université Lille Nord de France, CHRU Lille, CETD, Lille, France
| | - C Creach
- CHU Saint-Etienne, CETD, Saint-Etienne, France
| | | | - V Martinez
- AP-HP - Hôpital Raymond Poincaré, CETD, Paris, France
| | - M Navez
- CHU Saint-Etienne, CETD, Saint-Etienne, France
| | | | - E Kuhn
- CHU Nantes, CETD, Nantes, France
| | - E Piquet
- Université Nice Côte-d'Azur, CHU Nice - Hôpital de Cimiez, Fédération Hospitalo-Universitaire INOVPAIN, CETD, Nice, France
| | - E Bozzolo
- Université Nice Côte-d'Azur, CHU Nice - Hôpital de Cimiez, Fédération Hospitalo-Universitaire INOVPAIN, CETD, Nice, France
| | - C Brosse
- CHU Saint-Etienne, CETD, Saint-Etienne, France
| | - B Lietar
- CHU Saint-Etienne, CETD, Saint-Etienne, France
| | - F Marcaillou
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | - A Hamdani
- Cancer Centre Oscar-Lambret, Lille, France
| | | | - Y Perier
- CH Avranches, CETD, Avranches, France
| | | | - C Gov
- HCL - Hôpital Neurologique, CETD, Lyon, France
| | - N Delage
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | | | - S Romettino
- Université Nice Côte-d'Azur, CHU Nice - Hôpital de Cimiez, Fédération Hospitalo-Universitaire INOVPAIN, CETD, Nice, France
| | - D Richard
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | - C Mallet
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | - L Bernard
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | - C Lambert
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France
| | - C Dubray
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France.,Analgesia Institute, Université Clermont Auvergne, Clermont-Ferrand, France
| | - C Duale
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France.,Analgesia Institute, Université Clermont Auvergne, Clermont-Ferrand, France
| | - A Eschalier
- Service de Pharmacologie Médicale, Direction de la Recherche Clinique et de l'Innovation, CETD, CIC, CNRS, SIGMA Clermont, ICCF, Service de Pharmacie, Université Clermont Auvergne, CHU Clermont-Ferrand, INSERM - NEURO-DOL, Clermont-Ferrand, France.,Analgesia Institute, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
15
|
McArthur JR, Motin L, Gleeson EC, Spiller S, Lewis RJ, Duggan PJ, Tuck KL, Adams DJ. Inhibition of human N- and T-type calcium channels by an ortho-phenoxyanilide derivative, MONIRO-1. Br J Pharmacol 2017; 175:2284-2295. [PMID: 28608537 DOI: 10.1111/bph.13910] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/24/2017] [Accepted: 06/05/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Voltage-gated calcium channels are involved in nociception in the CNS and in the periphery. N-type (Cav 2.2) and T-type (Cav 3.1, Cav 3.2 and Cav 3.3) voltage-gated calcium channels are particularly important in studying and treating pain and epilepsy. EXPERIMENTAL APPROACH In this study, whole-cell patch clamp electrophysiology was used to assess the potency and mechanism of action of a novel ortho-phenoxylanilide derivative, MONIRO-1, against a panel of voltage-gated calcium channels including Cav 1.2, Cav 1.3, Cav 2.1, Cav 2.2, Cav 2.3, Cav 3.1, Cav 3.2 and Cav 3.3. KEY RESULTS MONIRO-1 was 5- to 20-fold more potent at inhibiting human T-type calcium channels, hCav 3.1, hCav 3.2 and hCav 3.3 (IC50 : 3.3 ± 0.3, 1.7 ± 0.1 and 7.2 ± 0.3 μM, respectively) than N-type calcium channel, hCav 2.2 (IC50 : 34.0 ± 3.6 μM). It interacted with L-type calcium channels Cav 1.2 and Cav 1.3 with significantly lower potency (IC50 > 100 μM) and did not inhibit hCav 2.1 or hCav 2.3 channels at concentrations as high as 100 μM. State- and use-dependent inhibition of hCav 2.2 channels was observed, whereas stronger inhibition occurred at high stimulation frequencies for hCav 3.1 channels suggesting a different mode of action between these two channels. CONCLUSIONS AND IMPLICATIONS Selectivity, potency, reversibility and multi-modal effects distinguish MONIRO-1 from other low MW inhibitors acting on Cav channels involved in pain and/or epilepsy pathways. High-frequency firing increased the affinity for MONIRO-1 for both hCav 2.2 and hCav 3.1 channels. Such Cav channel modulators have potential clinical use in the treatment of epilepsies, neuropathic pain and other nociceptive pathophysiologies. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Health Innovations Research Institute, RMIT University, Melbourne, VIC, Australia
| | - Leonid Motin
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Health Innovations Research Institute, RMIT University, Melbourne, VIC, Australia
| | - Ellen C Gleeson
- CSIRO Manufacturing, Bag 10, Clayton South, VIC, Australia.,School of Chemistry, Monash University, Clayton, VIC, Australia
| | - Sandro Spiller
- School of Chemistry, Monash University, Clayton, VIC, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Peter J Duggan
- CSIRO Manufacturing, Bag 10, Clayton South, VIC, Australia.,School of Chemical and Physical Sciences, Flinders University, Adelaide, SA, Australia
| | - Kellie L Tuck
- School of Chemistry, Monash University, Clayton, VIC, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia.,Health Innovations Research Institute, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Lin SF, Wang B, Zhang FM, Fei YH, Gu JH, Li J, Bi LB, Liu XJ. T-type calcium channels, but not Cav3.2, in the peripheral sensory afferents are involved in acute itch in mice. Biochem Biophys Res Commun 2017; 487:801-806. [DOI: 10.1016/j.bbrc.2017.04.127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 04/23/2017] [Indexed: 12/31/2022]
|
17
|
Yabuki Y, Matsuo K, Izumi H, Haga H, Yoshida T, Wakamori M, Kakei A, Sakimura K, Fukuda T, Fukunaga K. Pharmacological properties of SAK3, a novel T-type voltage-gated Ca 2+ channel enhancer. Neuropharmacology 2017; 117:1-13. [PMID: 28093211 DOI: 10.1016/j.neuropharm.2017.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/27/2016] [Accepted: 01/12/2017] [Indexed: 11/30/2022]
Abstract
T-type voltage-gated Ca2+ channels (T-VGCCs) function in the pathophysiology of epilepsy, pain and sleep. However, their role in cognitive function remains unclear. We previously reported that the cognitive enhancer ST101, which stimulates T-VGCCs in rat cortical slices, was a potential Alzheimer's disease therapeutic. Here, we introduce a more potent T-VGCC enhancer, SAK3 (ethyl 8'-methyl-2',4-dioxo-2-(piperidin-1-yl)-2'H-spiro[cyclopentane-1,3'-imidazo [1,2-a]pyridin]-2-ene-3-carboxylate), and characterize its pharmacological properties in brain. Based on whole cell patch-clamp analysis, SAK3 (0.01-10 nM) significantly enhanced Cav3.1 currents in neuro2A cells ectopically expressing Cav3.1. SAK3 (0.1-10 nM nM) also enhanced Cav3.3 but not Cav3.2 currents in the transfected cells. Notably, Cav3.1 and Cav3.3 T-VGCCs were localized in cholinergic neurve systems in hippocampus and in the medial septum. Indeed, acute oral administration of SAK3 (0.5 mg/kg, p.o.), but not ST101 (0.5 mg/kg, p.o.) significantly enhanced acetylcholine (ACh) release in the hippocampal CA1 region of naïve mice. Moreover, acute SAK3 (0.5 mg/kg, p.o.) administration significantly enhanced hippocampal ACh levels in olfactory-bulbectomized (OBX) mice, rescuing impaired memory-related behaviors. Treatment of OBX mice with the T-VGCC-specific blocker NNC 55-0396 (12.5 mg/kg, i.p.) antagonized both enhanced ACh release and memory improvements elicited by SAK3 administration. We also observed that SAK3-induced ACh releases were significantly blocked in the hippocampus from Cav3.1 knockout (KO) mice. These findings suggest overall that T-VGCCs play a key role in cognition by enhancing hippocampal ACh release and that the cognitive enhancer SAK3 could be a candidate therapeutic in Alzheimer's disease.
Collapse
Affiliation(s)
- Yasushi Yabuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kazuya Matsuo
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hidaka Haga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takashi Yoshida
- Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Minoru Wakamori
- Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Akikazu Kakei
- Department of Chemistry and Material Engineering, Faculty of Engineering, Shinshu University, Nagano, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takaichi Fukuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
18
|
De Prá SDT, Ferro PR, Milioli AM, Rigo FK, Chipindo OJ, Camponogara C, Casoti R, Manfron MP, de Oliveira SM, Ferreira J, Trevisan G. Antinociceptive activity and mechanism of action of hydroalcoholic extract and dichloromethane fraction of Amphilophium crucigerum seeds in mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 195:283-297. [PMID: 27864110 DOI: 10.1016/j.jep.2016.11.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 10/16/2016] [Accepted: 11/15/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The medicinal plant generally known as monkey's comb (Amphilophium crucigerum) has been popularly described for the treatment of neuropathic and inflammatory pain, specially seeds preparations. AIM OF THE STUDY The goal of the present study was to evaluate the antinociceptive effect of the crude extract (Crd) and dichloromethane fraction (Dcm) of A. crucigerum seeds, and investigate the involvement of transient receptor potential vanilloid 1 (TRPV1) receptor in this effect. MATERIALS AND METHODS Male Swiss mice were used in this study. The effects of Crd and Dcm was tested on capsaicin-induced Ca2+ influx or the specific binding of [3H]-resiniferatoxin. Moreover, after treatment with Crd or Dcm, animals were exposed to acute pain (hot water tail-flick and capsaicin intraplantar test) or chronic pain models (injection of complete Freund's adjuvant or partial ligation of the sciatic nerve). Acute adverse effects were also noted: locomotor activity, corporal temperature, hepatic or renal damage, gastrointestinal transit alteration, and ulcerogenic activity. RESULTS The oral administration of Crd or Dcm resulted in an antinociceptive effect in the hot water tail-flick (48°C) and capsaicin intraplantar tests. Furthermore, these preparations exhibited antinociceptive and anti-inflammatory effects in a chronic inflammatory pain model, and antinociceptive effects in a neuropathic pain model. Moreover, Crd and Dcm reduced capsaicin-induced Ca2+ influx and diminished the [3H]-resiniferatoxin specific binding to spinal cord membranes. Acute adverse events were not found with Crd or Dcm administration. CONCLUSION In conclusion, our results support the analgesic effect of A. crucigerum and suggest the presence of compounds that may act as TRPV1 antagonists.
Collapse
Affiliation(s)
- Samira Dal Toé De Prá
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88006-000 Criciúma (SC), Brazil.
| | - Paula Ronsani Ferro
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88006-000 Criciúma (SC), Brazil.
| | - Alessandra Marcon Milioli
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88006-000 Criciúma (SC), Brazil.
| | - Flávia Karine Rigo
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88006-000 Criciúma (SC), Brazil.
| | - Orlando Justo Chipindo
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88006-000 Criciúma (SC), Brazil.
| | - Camila Camponogara
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria (RS), Brazil.
| | - Rosana Casoti
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade de São Paulo (USP - Ribeirão Preto), 14040-903 Ribeirão Preto (SP), Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria (RS), Brazil.
| | - Melânia Palermo Manfron
- Programa de Pós-Graduação em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria (RS), Brazil.
| | - Sara Marchesan de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria (RS), Brazil.
| | - Juliano Ferreira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina (UFSC), 88049-900 Florianópolis (RS), Brazil.
| | - Gabriela Trevisan
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), 88006-000 Criciúma (SC), Brazil; Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa Maria (RS), Brazil.
| |
Collapse
|
19
|
Kerckhove N, Mallet C, Pereira B, Chenaf C, Duale C, Dubray C, Eschalier A. Assessment of the effectiveness and safety of Ethosuximide in the Treatment of non-Diabetic Peripheral Neuropathic Pain: EDONOT-protocol of a randomised, parallel, controlled, double-blinded and multicentre clinical trial. BMJ Open 2016; 6:e013530. [PMID: 27986742 PMCID: PMC5168699 DOI: 10.1136/bmjopen-2016-013530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Currently available analgesics are ineffective in 30-50% of patients suffering from neuropathic pain and often induce deleterious side effects. T-type calcium channel blockers (mibefradil, ethosuximide, NNC 55-0396) are of great interest for the development of new symptomatic treatments of neuropathic pain, due to their various effects on pain perception. Interestingly, ethosuximide, which has already been approved for treating epilepsy, is available on the European market for clinical use. Despite numerous preclinical data demonstrating an antinociceptive effect of ethosuximide in various animal models of neuropathic pain, no clinical studies have been published to date on the analgesic efficacy of ethosuximide in patients with neuropathic pain. METHODS AND ANALYSIS The Ethosuximide in the Treatment of non-Diabetic Peripheral Neuropathic Pain (EDONOT) trial is a randomised, parallel, controlled, double-blinded, multicentre clinical study. It is the first clinical trial to evaluate the efficacy and safety of ethosuximide in the treatment of non-diabetic peripheral neuropathic pain. Adult patients exhibiting peripheral neuropathic pain (Numeric Rating Scale (NRS) ≥4 and Douleur Neuropathique 4 (DN4)≥4) for at least 3 months and under stable analgesic treatment for at least 1 month will be included. Patients (n=220) will be randomly assigned to receive either ethosuximide or control treatment for 6 weeks following a 1 week run-in period. The primary end point is the intensity of neuropathic pain, assessed by NRS (0-10) before and after 6 weeks of treatment. The secondary end points are safety (adverse events are collected during the study: daily by the patient on the logbook and during planned phone calls by investigators), the intensity and features of neuropathic pain (assessed by Brief Pain Inventory (BPI) and Neuropathic Pain Symptom Inventory (NPSI) questionnaires) and health-related quality of life (assessed by Medical Outcome Study Short Form 12 (MOS SF-12) and Leeds questionnaires). ETHICS AND COMMUNICATION The study was approved by an independent ethics committee (CPP Sud-Est VI, France, IRB00008526) and registered by the French competent authority (Agence nationale de sécurité du médicament (ANSM)). TRIAL REGISTRATION NUMBER NCT02100046, Recruiting.
Collapse
Affiliation(s)
- Nicolas Kerckhove
- Service de Pharmacologie/Toxicologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
- CHU Clermont-Ferrand, Direction de la Recherche Clinique et des Innovations, villa annexe IFSI, Clermont-Ferrand, France
- Institut Analgesia, Clermont Université, Clermont-Ferrand, France
| | - Christophe Mallet
- Clermont Université, Clermont-Ferrand, France
- INSERM, U1107 “Neuro-Dol”, Clermont-Ferrand, France
| | - Bruno Pereira
- CHU Clermont-Ferrand, Direction de la Recherche Clinique et des Innovations, villa annexe IFSI, Clermont-Ferrand, France
| | - Chouki Chenaf
- Service de Pharmacologie/Toxicologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
- Clermont Université, Clermont-Ferrand, France
- INSERM, U1107 “Neuro-Dol”, Clermont-Ferrand, France
| | - Christian Duale
- CHU Clermont-Ferrand, Centre de Pharmacologie Clinique, Clermont-Ferrand, France
- INSERM, CIC1405, Clermont-Ferrand, France
| | - Claude Dubray
- Clermont Université, Clermont-Ferrand, France
- INSERM, U1107 “Neuro-Dol”, Clermont-Ferrand, France
- CHU Clermont-Ferrand, Centre de Pharmacologie Clinique, Clermont-Ferrand, France
- INSERM, CIC1405, Clermont-Ferrand, France
| | - Alain Eschalier
- Service de Pharmacologie/Toxicologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
- Institut Analgesia, Clermont Université, Clermont-Ferrand, France
- Clermont Université, Clermont-Ferrand, France
- INSERM, U1107 “Neuro-Dol”, Clermont-Ferrand, France
| |
Collapse
|
20
|
Cain SM, Ahn S, Garcia E, Zhang Y, Waheed Z, Tyson JR, Yang Y, Van Sung T, Phillips AG, Snutch TP. Heantos-4, a natural plant extract used in the treatment of drug addiction, modulates T-type calcium channels and thalamocortical burst-firing. Mol Brain 2016; 9:94. [PMID: 27919294 PMCID: PMC5139062 DOI: 10.1186/s13041-016-0274-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/21/2016] [Indexed: 11/10/2022] Open
Abstract
Heantos-4 is a refined combination of plant extracts currently approved to treat opiate addiction in Vietnam. In addition to its beneficial effects on withdrawal and prevention of relapse, reports of sedation during clinical treatment suggest that arousal networks in the brain may be recruited during Heantos administration. T-type calcium channels are implicated in the generation of sleep rhythms and in this study we examined whether a Heantos-4 extraction modulates T-type calcium channel currents generated by the Cav3.1, Cav3.2 and Ca3.3 subtypes. Utilizing whole-cell voltage clamp on exogenously expressed T-type calcium channels we find that Heantos inhibits Cav3.1 and Cav3.3 currents, while selectively potentiating Cav3.2 currents. We further examined the effects of Heantos-4 extract on low-threshold burst-firing in thalamic neurons which contribute to sleep oscillations. Using whole-cell current clamp in acute thalamic brain slices Heantos-4 suppressed rebound burst-firing in ventrobasal thalamocortical neurons, which express primarily Cav3.1 channels. Conversely, Heantos-4 had no significant effect on the burst-firing properties of thalamic reticular neurons, which express a mixed population of Cav3.2 and Cav3.3 channels. Examining Heantos-4 effects following oral administration in a model of absence epilepsy revealed the potential to exacerbate seizure activity. Together, the findings indicate that Heantos-4 has selective effects both on specific T-type calcium channel isoforms and distinct populations of thalamic neurons providing a putative mechanism underlying its effects on sedation and on the thalamocortical network.
Collapse
Affiliation(s)
- Stuart M Cain
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Soyon Ahn
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
| | - Esperanza Garcia
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Yiming Zhang
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Zeina Waheed
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - John R Tyson
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Yi Yang
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Tran Van Sung
- Institute of Chemistry, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Anthony G Phillips
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 219-2185 East Mall, Vancouver, BC, V6T 1Z4, Canada. .,Department of Psychiatry, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
21
|
Siegrist R, Pozzi D, Jacob G, Torrisi C, Colas K, Braibant B, Mawet J, Pfeifer T, de Kanter R, Roch C, Kessler M, Corminboeuf O, Bezençon O. Structure–Activity Relationship, Drug Metabolism and Pharmacokinetics Properties Optimization, and in Vivo Studies of New Brain Penetrant Triple T-Type Calcium Channel Blockers. J Med Chem 2016; 59:10661-10675. [DOI: 10.1021/acs.jmedchem.6b01356] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Romain Siegrist
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Davide Pozzi
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Gaël Jacob
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Caterina Torrisi
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Kilian Colas
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Bertrand Braibant
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Jacques Mawet
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Thomas Pfeifer
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Ruben de Kanter
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Catherine Roch
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Melanie Kessler
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Olivier Corminboeuf
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| | - Olivier Bezençon
- Drug Discovery Chemistry,
Biology and Pharmacology, Actelion Pharmaceuticals Ltd., Gewerbestrasse
16, CH-4123 Allschwil, Switzerland
| |
Collapse
|
22
|
Shiue SJ, Wang CH, Wang TY, Chen YC, Cheng JK. Chronic intrathecal infusion of T-type calcium channel blockers attenuates Ca V3.2 upregulation in nerve-ligated rats. ACTA ACUST UNITED AC 2016; 54:81-87. [PMID: 27765616 DOI: 10.1016/j.aat.2016.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVE T-type channel (TCC) CaV3.2 plays a pivotal role in pain transmission. In this study, we examined the effects of intrathecal TCC blockers on CaV3.2 expression in a L5/6 spinal nerve ligation (SNL) pain model. The neurotoxicity of TCC blockers were also evaluated. METHODS Male Sprague-Dawley rats (200-250 g) were used for right L5/6 SNL to induce neuropathic pain. Intrathecal infusion of saline or TCC blockers [mibefradil (0.7 μg/h) or ethosuximide (60 μg/h)] was started after surgery for 7 days. Fluorescent immunohistochemistry and Western blotting were used to determine the expression pattern and protein level of CaV3.2. Hematoxylin-eosin and toluidine blue staining were used to evaluate the neurotoxicity of tested agents. RESULTS Seven days after SNL, CaV3.2 protein levels were upregulated in ipsi-lateral L5/6 spinal cord and dorsal root ganglia (DRG) in immunofluorescence and Western blotting studies. Compared with the saline-treated group, rats receiving mibefradil or ethosuximide showed significant lower CaV3.2 expression in the spinal cord and DRG. No obvious histopathologic change in hematoxylin-eosin and toluidine blue staining were observed in all tested groups. CONCLUSION In this study, we demonstrate that SNL-induced CaV3.2 upregulation in the spinal cord and DRG was attenuated by intrathecal infusion of mibefradil or ethosuximide. No obvious neurotoxicity effects were observed in all the tested groups. Our data suggest that continuous intrathecal infusion of TCC blockers may be considered as a promising alternative for the treatment of nerve injury-induced pain.
Collapse
Affiliation(s)
- Sheng-Jie Shiue
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chi-Hsu Wang
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Tao-Yeuan Wang
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Department of Pathology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Chun Chen
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Jen-Kun Cheng
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.
| |
Collapse
|
23
|
T-type calcium channel blocker Z944 restores cortical synchrony and thalamocortical connectivity in a rat model of neuropathic pain. Pain 2016; 157:255-263. [PMID: 26683108 DOI: 10.1097/j.pain.0000000000000362] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Oscillations are fundamental to communication between neuronal ensembles. We previously reported that pain in awake rats enhances synchrony in primary somatosensory cortex (S1) and attenuates coherence between S1 and ventral posterolateral (VPL) thalamus. Here, we asked whether similar changes occur in anesthetized rats and whether pain modulates phase-amplitude coupling between VPL and S1. We also hypothesized that the suppression of burst firing in VPL using Z944, a novel T-type calcium channel blocker, restores S1 synchrony and thalamocortical connectivity. Local field potentials were recorded from S1 and VPL in anesthetized rats 7 days after sciatic chronic constriction injury (CCI). In rats with CCI, low-frequency (4-12 Hz) synchrony in S1 was enhanced, whereas VPL-S1 coherence and theta-gamma phase-amplitude coupling were attenuated. Moreover, Granger causality showed decreased informational flow from VPL to S1. Systemic or intrathalamic delivery of Z944 to rats with CCI normalized these changes. Systemic Z944 also reversed thermal hyperalgesia and conditioned place preference. These data suggest that pain-induced cortical synchrony and thalamocortical disconnectivity are directly related to burst firing in VPL.
Collapse
|
24
|
Wormuth C, Lundt A, Henseler C, Müller R, Broich K, Papazoglou A, Weiergräber M. Review: Ca v2.3 R-type Voltage-Gated Ca 2+ Channels - Functional Implications in Convulsive and Non-convulsive Seizure Activity. Open Neurol J 2016; 10:99-126. [PMID: 27843503 PMCID: PMC5080872 DOI: 10.2174/1874205x01610010099] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/16/2016] [Accepted: 06/24/2016] [Indexed: 11/22/2022] Open
Abstract
Background: Researchers have gained substantial insight into mechanisms of synaptic transmission, hyperexcitability, excitotoxicity and neurodegeneration within the last decades. Voltage-gated Ca2+ channels are of central relevance in these processes. In particular, they are key elements in the etiopathogenesis of numerous seizure types and epilepsies. Earlier studies predominantly targeted on Cav2.1 P/Q-type and Cav3.2 T-type Ca2+ channels relevant for absence epileptogenesis. Recent findings bring other channels entities more into focus such as the Cav2.3 R-type Ca2+ channel which exhibits an intriguing role in ictogenesis and seizure propagation. Cav2.3 R-type voltage gated Ca2+ channels (VGCC) emerged to be important factors in the pathogenesis of absence epilepsy, human juvenile myoclonic epilepsy (JME), and cellular epileptiform activity, e.g. in CA1 neurons. They also serve as potential target for various antiepileptic drugs, such as lamotrigine and topiramate. Objective: This review provides a summary of structure, function and pharmacology of VGCCs and their fundamental role in cellular Ca2+ homeostasis. We elaborate the unique modulatory properties of Cav2.3 R-type Ca2+ channels and point to recent findings in the proictogenic and proneuroapoptotic role of Cav2.3 R-type VGCCs in generalized convulsive tonic–clonic and complex-partial hippocampal seizures and its role in non-convulsive absence like seizure activity. Conclusion: Development of novel Cav2.3 specific modulators can be effective in the pharmacological treatment of epilepsies and other neurological disorders.
Collapse
Affiliation(s)
- Carola Wormuth
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Andreas Lundt
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Christina Henseler
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Ralf Müller
- Department of Psychiatry and Psychotherapy, University of Cologne, Faculty of Medicine, Cologne, Germany
| | - Karl Broich
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Anna Papazoglou
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Marco Weiergräber
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| |
Collapse
|
25
|
Tibbs GR, Posson DJ, Goldstein PA. Voltage-Gated Ion Channels in the PNS: Novel Therapies for Neuropathic Pain? Trends Pharmacol Sci 2016; 37:522-542. [DOI: 10.1016/j.tips.2016.05.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/24/2016] [Accepted: 05/03/2016] [Indexed: 12/19/2022]
|
26
|
Inflammatory mediator bradykinin increases population of sensory neurons expressing functional T-type Ca(2+) channels. Biochem Biophys Res Commun 2016; 473:396-402. [PMID: 26944020 PMCID: PMC4840015 DOI: 10.1016/j.bbrc.2016.02.118] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/28/2016] [Indexed: 02/06/2023]
Abstract
T-type Ca2+ channels are important regulators of peripheral sensory neuron excitability. Accordingly, T-type Ca2+ currents are often increased in various pathological pain conditions, such as inflammation or nerve injury. Here we investigated effects of inflammation on functional expression of T-type Ca2+ channels in small-diameter cultured dorsal root ganglion (DRG) neurons. We found that overnight treatment of DRG cultures with a cocktail of inflammatory mediators bradykinin (BK), adenosine triphosphate (ATP), norepinephrine (NE) and prostaglandin E2 (PGE2) strongly increased the population size of the small-diameter neurons displaying low-voltage activated (LVA, T-type) Ca2+ currents while having no effect on the peak LVA current amplitude. When applied individually, BK and ATP also increased the population size of LVA-positive neurons while NE and PGE2 had no effect. The PLC inhibitor U-73122 and B2 receptor antagonist, Hoe-140, both abolished the increase of the population of LVA-positive DRG neurons. Inflammatory treatment did not affect CaV3.2 mRNA or protein levels in DRG cultures. Furthermore, an ubiquitination inhibitor, MG132, did not increase the population of LVA-positive neurons. Our data suggest that inflammatory mediators BK and ATP increase the abundance of LVA-positive DRG neurons in total neuronal population by stimulating the recruitment of a ‘reserve pool’ of CaV3.2 channels, particularly in neurons that do not display measurable LVA currents under control conditions. Inflammatory mediators bradykinin and ATP increase the population of DRG neurons displaying T-type Ca2+ currents. This effect is mediated by the phospholipase C signaling cascade. Increase in the proportion is not correlated with the increased expression or decreased degradation of the CaV3.2 channel.
Collapse
|
27
|
Elies J, Scragg JL, Boyle JP, Gamper N, Peers C. Regulation of the T-type Ca(2+) channel Cav3.2 by hydrogen sulfide: emerging controversies concerning the role of H2 S in nociception. J Physiol 2016; 594:4119-29. [PMID: 26804000 DOI: 10.1113/jp270963] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/26/2015] [Indexed: 12/22/2022] Open
Abstract
Ion channels represent a large and growing family of target proteins regulated by gasotransmitters such as nitric oxide, carbon monoxide and, as described more recently, hydrogen sulfide. Indeed, many of the biological actions of these gases can be accounted for by their ability to modulate ion channel activity. Here, we report recent evidence that H2 S is a modulator of low voltage-activated T-type Ca(2+) channels, and discriminates between the different subtypes of T-type Ca(2+) channel in that it selectively modulates Cav3.2, whilst Cav3.1 and Cav3.3 are unaffected. At high concentrations, H2 S augments Cav3.2 currents, an observation which has led to the suggestion that H2 S exerts its pro-nociceptive effects via this channel, since Cav3.2 plays a central role in sensory nerve excitability. However, at more physiological concentrations, H2 S is seen to inhibit Cav3.2. This inhibitory action requires the presence of the redox-sensitive, extracellular region of the channel which is responsible for tonic metal ion binding and which particularly distinguishes this channel isoform from Cav3.1 and 3.3. Further studies indicate that H2 S may act in a novel manner to alter channel activity by potentiating the zinc sensitivity/affinity of this binding site. This review discusses the different reports of H2 S modulation of T-type Ca(2+) channels, and how such varying effects may impact on nociception given the role of this channel in sensory activity. This subject remains controversial, and future studies are required before the impact of T-type Ca(2+) channel modulation by H2 S might be exploited as a novel approach to pain management.
Collapse
Affiliation(s)
- Jacobo Elies
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jason L Scragg
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - John P Boyle
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nikita Gamper
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK.,Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Chris Peers
- Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
28
|
Ondacova K, Karmazinova M, Lazniewska J, Weiss N, Lacinova L. Modulation of Cav3.2 T-type calcium channel permeability by asparagine-linked glycosylation. Channels (Austin) 2016; 10:175-84. [PMID: 26745591 DOI: 10.1080/19336950.2016.1138189] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Low-voltage-gated T-type calcium channels are expressed throughout the nervous system where they play an essential role in shaping neuronal excitability. Defects in T-type channel expression have been linked to various neuronal disorders including neuropathic pain and epilepsy. Currently, little is known about the cellular mechanisms controlling the expression and function of T-type channels. Asparagine-linked glycosylation has recently emerged as an essential signaling pathway by which the cellular environment can control expression of T-type channels. However, the role of N-glycans in the conducting function of T-type channels remains elusive. In the present study, we used human Cav3.2 glycosylation-deficient channels to assess the role of N-glycosylation on the gating of the channel. Patch-clamp recordings of gating currents revealed that N-glycans attached to hCav3.2 channels have a minimal effect on the functioning of the channel voltage-sensor. In contrast, N-glycosylation on specific asparagine residues may have an essential role in the conducting function of the channel by enhancing the channel permeability and / or the pore opening of the channel. Our data suggest that modulation of N-linked glycosylation of hCav3.2 channels may play an important physiological role, and could also support the alteration of T-type currents observed in disease states.
Collapse
Affiliation(s)
- Katarina Ondacova
- a Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences , Bratislava , Slovakia
| | - Maria Karmazinova
- a Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences , Bratislava , Slovakia
| | - Joanna Lazniewska
- b Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. , Prague , Czech Republic
| | - Norbert Weiss
- b Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. , Prague , Czech Republic
| | - Lubica Lacinova
- a Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences , Bratislava , Slovakia
| |
Collapse
|
29
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 790] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
30
|
Effect of the T-type channel blocker KYS-05090S in mouse models of acute and neuropathic pain. Pflugers Arch 2015; 468:193-9. [PMID: 26354962 DOI: 10.1007/s00424-015-1733-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 08/29/2015] [Accepted: 09/02/2015] [Indexed: 12/11/2022]
Abstract
T-type channels are important contributors to the initiation and the maintenance of chronic pain states. Blocking T-type channels is therefore a possible therapeutic strategy for relieving pain. Here, we report the Cav3.2 T-type channel blocking action of a previously reported small organic molecule, KYS-05090S. This compound was able to reduce transiently expressed Cav3.2 currents with low micromolar affinity and mediated a hyperpolarizing shift in half-inactivation potential. KYS-05090S was then tested in models of acute and neuropathic pain. KYS-05090S (10 μg/10 μl delivered intrathecally) significantly reduced acute pain induced by formalin in both the tonic and inflammatory phases. Its antinociceptive effect was not observed when delivered to Cav3.2 null-mice revealing a Cav3.2-dependent mechanism. KYS-05090S also reduced neuropathic pain in a model of partial sciatic nerve injury. Those results indicate that KYS-05090S mediates a potent analgesic effect in inflammatory and neuropathic pain through T-type channel modulation, suggesting that its scaffold could be explored as a new class of analgesic compounds.
Collapse
|
31
|
Monteil A, Chausson P, Boutourlinsky K, Mezghrani A, Huc-Brandt S, Blesneac I, Bidaud I, Lemmers C, Leresche N, Lambert RC, Lory P. Inhibition of Cav3.2 T-type Calcium Channels by Its Intracellular I-II Loop. J Biol Chem 2015; 290:16168-76. [PMID: 25931121 DOI: 10.1074/jbc.m114.634261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Indexed: 11/06/2022] Open
Abstract
Voltage-dependent calcium channels (Cav) of the T-type family (Cav3.1, Cav3.2, and Cav3.3) are activated by low threshold membrane depolarization and contribute greatly to neuronal network excitability. Enhanced T-type channel activity, especially Cav3.2, contributes to disease states, including absence epilepsy. Interestingly, the intracellular loop connecting domains I and II (I-II loop) of Cav3.2 channels is implicated in the control of both surface expression and channel gating, indicating that this I-II loop plays an important regulatory role in T-type current. Here we describe that co-expression of this I-II loop or its proximal region (Δ1-Cav3.2; Ser(423)-Pro(542)) together with recombinant full-length Cav3.2 channel inhibited T-type current without affecting channel expression and membrane incorporation. Similar T-type current inhibition was obtained in NG 108-15 neuroblastoma cells that constitutively express Cav3.2 channels. Of interest, Δ1-Cav3.2 inhibited both Cav3.2 and Cav3.1 but not Cav3.3 currents. Efficacy of Δ1-Cav3.2 to inhibit native T-type channels was assessed in thalamic neurons using viral transduction. We describe that T-type current was significantly inhibited in the ventrobasal neurons that express Cav3.1, whereas in nucleus reticularis thalami neurons that express Cav3.2 and Cav3.3 channels, only the fast inactivating T-type current (Cav3.2 component) was significantly inhibited. Altogether, these data describe a new strategy to differentially inhibit Cav3 isoforms of the T-type calcium channels.
Collapse
Affiliation(s)
- Arnaud Monteil
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, Plateforme de Vectorologie, Biocampus Montpellier CNRS UMS 3426, INSERM US009, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Patrick Chausson
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Katia Boutourlinsky
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Alexandre Mezghrani
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Sylvaine Huc-Brandt
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Iulia Blesneac
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Isabelle Bidaud
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Céline Lemmers
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, Plateforme de Vectorologie, Biocampus Montpellier CNRS UMS 3426, INSERM US009, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Nathalie Leresche
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Régis C Lambert
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Philippe Lory
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France,
| |
Collapse
|
32
|
Regulation of neuronal cav3.1 channels by cyclin-dependent kinase 5 (Cdk5). PLoS One 2015; 10:e0119134. [PMID: 25760945 PMCID: PMC4356599 DOI: 10.1371/journal.pone.0119134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 01/19/2015] [Indexed: 11/25/2022] Open
Abstract
Low voltage-activated (LVA) T-type Ca2+ channels activate in response to subthreshold membrane depolarizations and therefore represent an important source of Ca2+ influx near the resting membrane potential. In neurons, these proteins significantly contribute to control relevant physiological processes including neuronal excitability, pacemaking and post-inhibitory rebound burst firing. Three subtypes of T-type channels (Cav3.1 to Cav3.3) have been identified, and using functional expression of recombinant channels diverse studies have validated the notion that T-type Ca2+ channels can be modulated by various endogenous ligands as well as by second messenger pathways. In this context, the present study reveals a previously unrecognized role for cyclin-dependent kinase 5 (Cdk5) in the regulation of native T-type channels in N1E-115 neuroblastoma cells, as well as recombinant Cav3.1channels heterologously expressed in HEK-293 cells. Cdk5 and its co-activators play critical roles in the regulation of neuronal differentiation, cortical lamination, neuronal cell migration and axon outgrowth. Our results show that overexpression of Cdk5 causes a significant increase in whole cell patch clamp currents through T-type channels in N1E-115 cells, while siRNA knockdown of Cdk5 greatly reduced these currents. Consistent with this, overexpression of Cdk5 in HEK-293 cells stably expressing Cav3.1channels upregulates macroscopic currents. Furthermore, using site-directed mutagenesis we identified a major phosphorylation site at serine 2234 within the C-terminal region of the Cav3.1subunit. These results highlight a novel role for Cdk5 in the regulation of T-type Ca2+ channels.
Collapse
|
33
|
François A, Schüetter N, Laffray S, Sanguesa J, Pizzoccaro A, Dubel S, Mantilleri A, Nargeot J, Noël J, Wood JN, Moqrich A, Pongs O, Bourinet E. The Low-Threshold Calcium Channel Cav3.2 Determines Low-Threshold Mechanoreceptor Function. Cell Rep 2015; 10:370-382. [PMID: 25600872 DOI: 10.1016/j.celrep.2014.12.042] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 11/14/2014] [Accepted: 12/18/2014] [Indexed: 12/12/2022] Open
Abstract
The T-type calcium channel Cav3.2 emerges as a key regulator of sensory functions, but its expression pattern within primary afferent neurons and its contribution to modality-specific signaling remain obscure. Here, we elucidate this issue using a unique knockin/flox mouse strain wherein Cav3.2 is replaced by a functional Cav3.2-surface-ecliptic GFP fusion. We demonstrate that Cav3.2 is a selective marker of two major low-threshold mechanoreceptors (LTMRs), Aδ- and C-LTMRs, innervating the most abundant skin hair follicles. The presence of Cav3.2 along LTMR-fiber trajectories is consistent with critical roles at multiple sites, setting their strong excitability. Strikingly, the C-LTMR-specific knockout uncovers that Cav3.2 regulates light-touch perception and noxious mechanical cold and chemical sensations and is essential to build up that debilitates allodynic symptoms of neuropathic pain, a mechanism thought to be entirely A-LTMR specific. Collectively, our findings support a fundamental role for Cav3.2 in touch/pain pathophysiology, validating their critic pharmacological relevance to relieve mechanical and cold allodynia.
Collapse
Affiliation(s)
- Amaury François
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Niklas Schüetter
- Department of Physiology, University of Saarland, School of Medicine, Kirrberger Straße 1, 66424 Homburg, Germany
| | - Sophie Laffray
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Juan Sanguesa
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Anne Pizzoccaro
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Stefan Dubel
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Annabelle Mantilleri
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, 13288 Marseille, France
| | - Joel Nargeot
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Jacques Noël
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; Université de Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS, 660 route des lucioles, 06560 Valbonne, France
| | - John N Wood
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, 13288 Marseille, France
| | - Olaf Pongs
- Department of Physiology, University of Saarland, School of Medicine, Kirrberger Straße 1, 66424 Homburg, Germany
| | - Emmanuel Bourinet
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France.
| |
Collapse
|
34
|
Terada Y, Kawabata A. H2S and Pain: A Novel Aspect for Processing of Somatic, Visceral and Neuropathic Pain Signals. Handb Exp Pharmacol 2015; 230:217-230. [PMID: 26162837 DOI: 10.1007/978-3-319-18144-8_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Hydrogen sulfide (H2S) formed by multiple enzymes including cystathionine-γ-lyase (CSE) targets Cav3.2 T-type Ca2+ channels (T-channels) and transient receptor potential ankyrin-1 (TRPA1). Intraplantar and intracolonic administration of H2S donors promotes somatic and visceral pain, respectively, via activation of Cav3.2 and TRPA1 in rats and/or mice. Injection of H2S donors into the plantar tissues, pancreatic duct, colonic lumen, or bladder causes T-channel-dependent excitation of nociceptors, determined as phosphorylation of ERK or expression of Fos in the spinal dorsal horn. Electrophysiological studies demonstrate that exogenous and/or endogenous H2S facilitates membrane currents through T-channels in NG108-15 cells and isolated mouse dorsal root ganglion (DRG) neurons that abundantly express Cav3.2 and also in Cav3.2-transfected HEK293 cells. In mice with cerulein-induced pancreatitis and cyclophosphamide-induced cystitis, visceral pain and/or referred hyperalgesia are inhibited by CSE inhibitors and by pharmacological blockade or genetic silencing of Cav3.2, and CSE protein is upregulated in the pancreas and bladder. In rats with neuropathy induced by L5 spinal nerve cutting or by repeated administration of paclitaxel, an anticancer drug, the neuropathic hyperalgesia is reversed by inhibitors of CSE or T-channels and by silencing of Cav3.2. Upregulation of Cav3.2 protein in DRG is detectable in the former, but not in the latter, neuropathic pain models. Thus, H2S appears to function as a nociceptive messenger by facilitating functions of Cav3.2 and TRPA1, and the enhanced function of the CSE/H2S/Cav3.2 pathway is considered to be involved in the pancreatitis- and cystitis-related pain and in neuropathic pain.
Collapse
Affiliation(s)
- Yuka Terada
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, Higashi-Osaka, 577-8502, Japan
| | | |
Collapse
|
35
|
Arranz-Tagarro JA, de los Ríos C, García AG, Padín JF. Recent patents on calcium channel blockers: emphasis on CNS diseases. Expert Opin Ther Pat 2014; 24:959-77. [DOI: 10.1517/13543776.2014.940892] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
36
|
Harraz OF, Abd El-Rahman RR, Bigdely-Shamloo K, Wilson SM, Brett SE, Romero M, Gonzales AL, Earley S, Vigmond EJ, Nygren A, Menon BK, Mufti RE, Watson T, Starreveld Y, Furstenhaupt T, Muellerleile PR, Kurjiaka DT, Kyle BD, Braun AP, Welsh DG. Ca(V)3.2 channels and the induction of negative feedback in cerebral arteries. Circ Res 2014; 115:650-61. [PMID: 25085940 DOI: 10.1161/circresaha.114.304056] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE T-type (CaV3.1/CaV3.2) Ca(2+) channels are expressed in rat cerebral arterial smooth muscle. Although present, their functional significance remains uncertain with findings pointing to a variety of roles. OBJECTIVE This study tested whether CaV3.2 channels mediate a negative feedback response by triggering Ca(2+) sparks, discrete events that initiate arterial hyperpolarization by activating large-conductance Ca(2+)-activated K(+) channels. METHODS AND RESULTS Micromolar Ni(2+), an agent that selectively blocks CaV3.2 but not CaV1.2/CaV3.1, was first shown to depolarize/constrict pressurized rat cerebral arteries; no effect was observed in CaV3.2(-/-) arteries. Structural analysis using 3-dimensional tomography, immunolabeling, and a proximity ligation assay next revealed the existence of microdomains in cerebral arterial smooth muscle which comprised sarcoplasmic reticulum and caveolae. Within these discrete structures, CaV3.2 and ryanodine receptor resided in close apposition to one another. Computational modeling revealed that Ca(2+) influx through CaV3.2 could repetitively activate ryanodine receptor, inducing discrete Ca(2+)-induced Ca(2+) release events in a voltage-dependent manner. In keeping with theoretical observations, rapid Ca(2+) imaging and perforated patch clamp electrophysiology demonstrated that Ni(2+) suppressed Ca(2+) sparks and consequently spontaneous transient outward K(+) currents, large-conductance Ca(2+)-activated K(+) channel mediated events. Additional functional work on pressurized arteries noted that paxilline, a large-conductance Ca(2+)-activated K(+) channel inhibitor, elicited arterial constriction equivalent, and not additive, to Ni(2+). Key experiments on human cerebral arteries indicate that CaV3.2 is present and drives a comparable response to moderate constriction. CONCLUSIONS These findings indicate for the first time that CaV3.2 channels localize to discrete microdomains and drive ryanodine receptor-mediated Ca(2+) sparks, enabling large-conductance Ca(2+)-activated K(+) channel activation, hyperpolarization, and attenuation of cerebral arterial constriction.
Collapse
Affiliation(s)
- Osama F Harraz
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Rasha R Abd El-Rahman
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Kamran Bigdely-Shamloo
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Sean M Wilson
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Suzanne E Brett
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Monica Romero
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Albert L Gonzales
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Scott Earley
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Edward J Vigmond
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Anders Nygren
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Bijoy K Menon
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Rania E Mufti
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Tim Watson
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Yves Starreveld
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Tobias Furstenhaupt
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Philip R Muellerleile
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - David T Kurjiaka
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Barry D Kyle
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Andrew P Braun
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.)
| | - Donald G Welsh
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes (O.F.H., R.R.A.E.-R., K.B.-S., S.E.B., R.E.M., B.D.K., A.P.B., D.G.W.), Department of Electrical and Computer Engineering (K.B.-S., E.J.V., A.N.), Department of Clinical Neurosciences (B.K.M., T.W., Y.S.), and Microscopy Imaging Facility (T.F.), University of Calgary, Calgary, Alberta, Canada; Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Division of Pharmacology, Loma Linda University, CA (S.M.W., M.R.); Department of Biomedical Sciences, Colorado State University, Fort Collins (A.L.G.); Department of Pharmacology, University of Nevada, Reno (S.E.); LIRYC Institute and Lab IMB, University of Bordeaux, Bordeaux, France (E.J.V.); and Department of Biomedical Sciences, Grand Valley State University, Allendale, MI (P.R.M., D.T.K.). dwelsh@ucalgary
| |
Collapse
|
37
|
|