1
|
Sekiguchi F, Tsubota M, Kawabata A. Sulfide and polysulfide as pronociceptive mediators: Focus on Ca v3.2 function enhancement and TRPA1 activation. J Pharmacol Sci 2024; 155:113-120. [PMID: 38797535 DOI: 10.1016/j.jphs.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/29/2024] Open
Abstract
Reactive sulfur species including sulfides, polysulfides and cysteine hydropersulfide play extensive roles in health and disease, which involve modification of protein functions through the interaction with metals bound to the proteins, cleavage of cysteine disulfide (S-S) bonds and S-persulfidation of cysteine residues. Sulfides over a wide micromolar concentration range enhance the activity of Cav3.2 T-type Ca2+ channels by eliminating Zn2+ bound to the channels, thereby promoting somatic and visceral pain. Cav3.2 is under inhibition by Zn2+ in physiological conditions, so that sulfides function to reboot Cav3.2 from Zn2+ inhibition and increase the excitability of nociceptors. On the other hand, polysulfides generated from sulfides activate TRPA1 channels via cysteine S-persulfidation, thereby facilitating somatic, but not visceral, pain. Thus, Cav3.2 function enhancement by sulfides and TRPA1 activation by polysulfides, synergistically accelerate somatic pain signals. The increased activity of the sulfide/Cav3.2 system, in particular, appears to have a great impact on pathological pain, and may thus serve as a therapeutic target for treatment of neuropathic and inflammatory pain including visceral pain.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
2
|
Hosseindoost S, Askari Rad M, Inanloo SH, Rahimi M, Dehghan S, Orandi A, Dehpour AR, Majedi H. The analgesic effects of botulinum neurotoxin by modulating pain-related receptors; A literature review. Mol Pain 2024; 20:17448069241275099. [PMID: 39093638 PMCID: PMC11339750 DOI: 10.1177/17448069241275099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/12/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
Botulinum neurotoxins (BoNTs), produced by Clostridium botulinum, have been used for the treatment of various central and peripheral neurological conditions. Recent studies have suggested that BoNTs may also have a beneficial effect on pain conditions. It has been hypothesized that one of the mechanisms underlying BoNTs' analgesic effects is the inhibition of pain-related receptors' transmission to the neuronal cell membrane. BoNT application disrupts the integration of synaptic vesicles with the cellular membrane, which is responsible for transporting various receptors, including pain receptors such as TRP channels, calcium channels, sodium channels, purinergic receptors, neurokinin-1 receptors, and glutamate receptors. BoNT also modulates the opioidergic system and the GABAergic system, both of which are involved in the pain process. Understanding the cellular and molecular mechanisms underlying these effects can provide valuable insights for the development of novel therapeutic approaches for pain management. This review aims to summarize the experimental evidence of the analgesic functions of BoNTs and discuss the cellular and molecular mechanisms by which they can act on pain conditions by inhibiting the transmission of pain-related receptors.
Collapse
Affiliation(s)
- Saereh Hosseindoost
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Maziyar Askari Rad
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Inanloo
- Department of Urology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Rahimi
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Orandi
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Majedi
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Anesthesia, Critical Care, and Pain Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Liu Q, Lu Z, Ren H, Fu L, Wang Y, Bu H, Ma M, Ma L, Huang C, Wang J, Zang W, Cao J, Fan X. Cav3.2 T-Type calcium channels downregulation attenuates bone cancer pain induced by inhibiting IGF-1/HIF-1α signaling pathway in the rat spinal cord. J Bone Oncol 2023; 42:100495. [PMID: 37583441 PMCID: PMC10423893 DOI: 10.1016/j.jbo.2023.100495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Background Bone cancer pain (BCP) is one of the most ubiquitous and refractory symptoms of cancer patients that needs to be urgently addressed. Substantial studies have revealed the pivotal role of Cav3.2 T-type calcium channels in chronic pain, however, its involvement in BCP and the specific molecular mechanism have not been fully elucidated. Methods The expression levels of Cav3.2, insulin-like growth factor 1(IGF-1), IGF-1 receptor (IGF-1R) and hypoxia-inducible factor-1α (HIF-1α) were detected by Western blot in tissues and cells. X-ray and Micro CT used to detect bone destruction in rats. Immunofluorescence was used to detect protein expression and spatial location in the spinal dorsal horn. Electrophoretic mobility shift assay used to verify the interaction between HIF-1α and Cav3.2. Results The results showed that the expression of Cav3.2 channel was upregulated and blockade of this channel alleviated mechanical allodynia and thermal hyperalgesia in BCP rats. Additionally, inhibition of IGF-1/IGF-1R signaling not only reversed the BCP-induced upregulation of Cav3.2 and HIF-1α, but also decreased nociceptive hypersensitivity in BCP rats. Inhibition of IGF-1 increased Cav3.2 expression levels, which were abolished by pretreatment with HIF-1α siRNA in PC12 cells. Furthermore, nuclear HIF-1α bound to the promoter of Cav3.2 to regulate the Cav3.2 transcription level, and knockdown of HIF-1α suppresses the IGF-1-induced upregulation of Cav3.2 and pain behaviors in rats with BCP. Conclusion These findings suggest that spinal Cav3.2 T-type calcium channels play a central role during the development of bone cancer pain in rats via regulation of the IGF-1/IGF-1R/HIF-1α pathway.
Collapse
Affiliation(s)
- Qingying Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhongyuan Lu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huan Ren
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lijun Fu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yueliang Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huilian Bu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Minyu Ma
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Letian Ma
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chen Huang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Weidong Zang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
4
|
Rangel-Galván M, Rangel-Galván V, Rangel-Huerta A. T-type calcium channel modulation by hydrogen sulfide in neuropathic pain conditions. Front Pharmacol 2023; 14:1212800. [PMID: 37529702 PMCID: PMC10387653 DOI: 10.3389/fphar.2023.1212800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/05/2023] [Indexed: 08/03/2023] Open
Abstract
Neuropathic pain can appear as a direct or indirect nerve damage lesion or disease that affects the somatosensory nervous system. If the neurons are damaged or indirectly stimulated, immune cells contribute significantly to inflammatory and neuropathic pain. After nerve injury, peripheral macrophages/spinal microglia accumulate around damaged neurons, producing endogenous hydrogen sulfide (H2S) through the cystathionine-γ-lyase (CSE) enzyme. H2S has a pronociceptive modulation on the Cav3.2 subtype, the predominant Cav3 isoform involved in pain processes. The present review provides relevant information about H2S modulation on the Cav3.2 T-type channels in neuropathic pain conditions. We have discussed that the dual effect of H2S on T-type channels is concentration-dependent, that is, an inhibitory effect is seen at low concentrations of 10 µM and an augmentation effect on T-current at 100 µM. The modulation mechanism of the Cav3.2 channel by H2S involves the direct participation of the redox/Zn2+ affinity site located in the His191 in the extracellular loop of domain I of the channel, involving a group of extracellular cysteines, comprising C114, C123, C128, and C1333, that can modify the local redox environment. The indirect interaction pathways involve the regulation of the Cav3.2 channel through cytokines, kinases, and post-translational regulators of channel expression. The findings conclude that the CSE/H2S/Cav3.2 pathway could be a promising therapeutic target for neuropathic pain disorders.
Collapse
Affiliation(s)
- Maricruz Rangel-Galván
- Biothecnology Department, Metropolitan Polytechnic University of Puebla, Puebla, Puebla, Mexico
| | - Violeta Rangel-Galván
- Nursing and Physiotherapy Department, University of Professional Development, Tijuana, Baja California, Mexico
| | - Alejandro Rangel-Huerta
- Faculty of Computer Science, Meritorious Autonomous University of Puebla, Puebla, Puebla, Mexico
| |
Collapse
|
5
|
Sekiguchi F, Koike N, Shimada Y, Sugimoto K, Masuda H, Nakamura T, Yamaguchi H, Tanabe G, Marumoto S, Kasanami Y, Tsubota M, Ohkubo T, Yoshida S, Kawabata A. A hydrolysate of poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132) suppresses Ca v3.2-dependent pain by sequestering exogenous and endogenous sulfide. Redox Biol 2023; 59:102579. [PMID: 36563535 PMCID: PMC9800310 DOI: 10.1016/j.redox.2022.102579] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132), an organogermanium, is hydrolyzed to 3-(trihydroxygermyl)propanoic acid (THGP) in aqueous solutions, and reduces inflammation, pain and cancer, whereas the underlying mechanisms remain unknown. Sulfides including H2S, a gasotransmitter, generated from l-cysteine by some enzymes including cystathionine-γ-lyase (CSE), are pro-nociceptive, since they enhance Cav3.2 T-type Ca2+ channel activity expressed in the primary afferents, most probably by canceling the channel inhibition by Zn2+ linked via coordinate bonding to His191 of Cav3.2. Given that germanium is reactive to sulfur, we tested whether THGP would directly trap sulfide, and inhibit sulfide-induced enhancement of Cav3.2 activity and sulfide-dependent pain in mice. Using mass spectrometry and 1H NMR techniques, we demonstrated that THGP directly reacted with sulfides including Na2S and NaSH, and formed a sulfur-containing reaction product, which decreased in the presence of ZnCl2. In Cav3.2-transfected HEK293 cells, THGP inhibited the sulfide-induced enhancement of T-type Ca2+ channel-dependent membrane currents. In mice, THGP, administered systemically or locally, inhibited the mechanical allodynia caused by intraplantar Na2S. In the mice with cyclophosphamide-induced cystitis and cerulein-induced pancreatitis, which exhibited upregulation of CSE in the bladder and pancreas, respectively, systemic administration of THGP as well as a selective T-type Ca2+ channel inhibitor suppressed the cystitis-related and pancreatitis-related visceral pain. These data suggest that THGP traps sulfide and inhibits sulfide-induced enhancement of Cav3.2 activity, leading to suppression of Cav3.2-dependent pain caused by sulfide applied exogenously and generated endogenously.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Nene Koike
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Yasuhiro Shimada
- Asai Germanium Research Institute Co., Ltd., Suzuranoka, Hakodate, Hokkaido, 042-0958, Japan
| | - Kaho Sugimoto
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Hiroshi Masuda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Takashi Nakamura
- Asai Germanium Research Institute Co., Ltd., Suzuranoka, Hakodate, Hokkaido, 042-0958, Japan
| | - Hiroaki Yamaguchi
- Yamagata University Graduate School of Medicine, Iida-nishi 2-2-2, Yamagata, 990-9585, Japan; Department of Pharmacy, Yamagata University Hospital, Iida-nishi 2-2-2, Yamagata, 990-9585, Japan
| | - Genzoh Tanabe
- Laboratory of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Shinsuke Marumoto
- Joint Research Center, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yoshihito Kasanami
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan
| | - Tsuyako Ohkubo
- Division of Basic Medical Sciences and Fundamental Nursing, Faculty of Nursing, Fukuoka Nursing College, Fukuoka, 814-0193, Japan
| | - Shigeru Yoshida
- Department of Life Science, Faculty of Science and Engineering, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Kowakae 3-4-1, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
6
|
Vitamin C Modes of Action in Calcium-Involved Signaling in the Brain. Antioxidants (Basel) 2023; 12:antiox12020231. [PMID: 36829790 PMCID: PMC9952025 DOI: 10.3390/antiox12020231] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Vitamin C (ascorbic acid) is well known for its potent antioxidant properties, as it can neutralize ROS and free radicals, thereby protecting cellular elements from oxidative stress. It predominantly exists as an ascorbate anion and after oxidation to dehydroascorbic acid and further breakdown, is removed from the cells. In nervous tissue, a progressive decrease in vitamin C level or its prolonged deficiency have been associated with an increased risk of disturbances in neurotransmission, leading to dysregulation in brain function. Therefore, understanding the regulatory function of vitamin C in antioxidant defence and identification of its molecular targets deserves more attention. One of the key signalling ions is calcium and a transient rise in its concentration is crucial for all neuronal processes. Extracellular Ca2+ influx (through specific ion channels) or Ca2+ release from intracellular stores (endoplasmic reticulum, mitochondria) are precisely controlled. Ca2+ regulates the functioning of the CNS, including growth, development, myelin formation, synthesis of catecholamines, modulation of neurotransmission and antioxidant protection. A growing body of evidence indicates a unique role for vitamin C in these processes. In this short review, we focus on vitamin C in the regulation of calcium-involved pathways under physiological and stress conditions in the brain.
Collapse
|
7
|
Huerta de la Cruz S, Medina-Terol GJ, Sánchez-López A, Centurión D. TRPA1, but not TRPV1, is involved in the increase of the non-adrenergic non-cholinergic outflow induced by hydrogen sulfide in pithed rats. Peptides 2022; 157:170861. [PMID: 35973467 DOI: 10.1016/j.peptides.2022.170861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/20/2022] [Accepted: 08/11/2022] [Indexed: 01/21/2023]
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter that modulates the peripheral transmission regulating the vascular tone. In vitro studies have suggested that H2S induces vasodilation by stimulating capsaicin-sensitive sensory neurons. This study was designed to determine the effects of H2S on the non-adrenergic/non-cholinergic (NANC) outflow in the pithed rat, and the underlying mechanisms. For that purpose, 72 male Wistar rats were anesthetized, pithed and the carotid, femoral and jugular veins were cannulated and then divided into two main sets. The first set of animals (n = 48) was used to determine the effect of NaHS (H2S donor) on the vasodepressor responses induced by: 1) NANC outflow electrical stimulation (n = 24); and 2) i.v. bolus of α-CGRP (n = 24) and subdivided into 4 groups (n = 6 each): 1) control group (without infusion); continuous infusion of: 2) PBS (vehicle; 0.02 ml/kg·min); 3) NaHS 10 μg/kg·min; and 4) NaHS 18 μg/kg·min. The second set of animals (n = 24) received an i.v. bolus of either (1) HC 030031 (TRPA1 channel antagonist; 18 μg/kg; n = 12) or (2) capsazepine (TRPV1 channel antagonist; 100 μg/kg; n = 12) in presence and absence of 18 µg/kg·min NaHS i.v. continuous infusion to determine the underlying mechanism of the NaHS effect on the NANC outflow. Our results show that NaHS infusion increased the vasodepressor responses induced by electrical stimulation, but not by α-CGRP, effect that was abolished by HC030031 and remained unaffected after capsazepine. These data suggest that activation of TRPA1 channels, but no TRPV1, is responsible for the NaHS-induced NANC neurotransmission stimulation.
Collapse
Affiliation(s)
- Saúl Huerta de la Cruz
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Del. Tlalpan, C.P. 14330 México D.F., Mexico.
| | - Grecia J Medina-Terol
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Del. Tlalpan, C.P. 14330 México D.F., Mexico.
| | - Araceli Sánchez-López
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Del. Tlalpan, C.P. 14330 México D.F., Mexico.
| | - David Centurión
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Del. Tlalpan, C.P. 14330 México D.F., Mexico.
| |
Collapse
|
8
|
The T-type calcium channel Ca V 3.2 regulates bladder afferent responses to mechanical stimuli. Pain 2022; 164:1012-1026. [PMID: 36279179 PMCID: PMC10108591 DOI: 10.1097/j.pain.0000000000002795] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 09/09/2022] [Indexed: 11/06/2022]
Abstract
ABSTRACT The bladder wall is innervated by a complex network of afferent nerves that detect bladder stretch during filling. Sensory signals, generated in response to distension, are relayed to the spinal cord and brain to evoke physiological and painful sensations and regulate urine storage and voiding. Hyperexcitability of these sensory pathways is a key component in the development of chronic bladder hypersensitivity disorders including interstitial cystitis/bladder pain syndrome and overactive bladder syndrome. Despite this, the full array of ion channels that regulate bladder afferent responses to mechanical stimuli have yet to be determined. Here, we investigated the role of low-voltage-activated T-type calcium (Ca V 3) channels in regulating bladder afferent responses to distension. Using single-cell reverse-transcription polymerase chain reaction and immunofluorescence, we revealed ubiquitous expression of Ca V 3.2, but not Ca V 3.1 or Ca V 3.3, in individual bladder-innervating dorsal root ganglia neurons. Pharmacological inhibition of Ca V 3.2 with TTA-A2 and ABT-639, selective blockers of T-type calcium channels, dose-dependently attenuated ex-vivo bladder afferent responses to distension in the absence of changes to muscle compliance. Further evaluation revealed that Ca V 3.2 blockers significantly inhibited both low- and high-threshold afferents, decreasing peak responses to distension, and delayed activation thresholds, thereby attenuating bladder afferent responses to both physiological and noxious distension. Nocifensive visceromotor responses to noxious bladder distension in vivo were also significantly reduced by inhibition of Ca V 3 with TTA-A2. Together, these data provide evidence of a major role for Ca V 3.2 in regulating bladder afferent responses to bladder distension and nociceptive signalling to the spinal cord.
Collapse
|
9
|
Discovery of pimozide derivatives as novel T-type calcium channel inhibitors with little binding affinity to dopamine D2 receptors for treatment of somatic and visceral pain. Eur J Med Chem 2022; 243:114716. [DOI: 10.1016/j.ejmech.2022.114716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022]
|
10
|
Harding EK, Zamponi GW. Central and peripheral contributions of T-type calcium channels in pain. Mol Brain 2022; 15:39. [PMID: 35501819 PMCID: PMC9063214 DOI: 10.1186/s13041-022-00923-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
AbstractChronic pain is a severely debilitating condition that reflects a long-term sensitization of signal transduction in the afferent pain pathway. Among the key players in this pathway are T-type calcium channels, in particular the Cav3.2 isoform. Because of their biophysical characteristics, these channels are ideally suited towards regulating neuronal excitability. Recent evidence suggests that T-type channels contribute to excitability of neurons all along the ascending and descending pain pathways, within primary afferent neurons, spinal dorsal horn neurons, and within pain-processing neurons in the midbrain and cortex. Here we review the contribution of T-type channels to neuronal excitability and function in each of these neuronal populations and how they are dysregulated in chronic pain conditions. Finally, we discuss their molecular pharmacology and the potential role of these channels as therapeutic targets for chronic pain.
Collapse
|
11
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
12
|
Joksimovic SL, Jevtovic-Todorovic V, Todorovic SM. The Mechanisms of Plasticity of Nociceptive Ion Channels in Painful Diabetic Neuropathy. FRONTIERS IN PAIN RESEARCH 2022; 3:869735. [PMID: 35419564 PMCID: PMC8995507 DOI: 10.3389/fpain.2022.869735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Treating pain in patients suffering from small fiber neuropathies still represents a therapeutic challenge for health care providers and drug developers worldwide. Unfortunately, none of the currently available treatments can completely reverse symptoms of either gain or loss of peripheral nerve sensation. Therefore, there is a clear need for novel mechanism-based therapies for peripheral diabetic neuropathy (PDN) that would improve treatment of this serious condition. In this review, we summarize the current knowledge on the mechanisms and causes of peripheral sensory neurons damage in diabetes. In particular, we focused on the subsets of voltage-gated sodium channels, TRP family of ion channels and a CaV3.2 isoform of T-type voltage-gated calcium channels. However, even though their potential is well-validated in multiple rodent models of painful PDN, clinical trials with specific pharmacological blockers of these channels have failed to exhibit therapeutic efficacy. We argue that understanding the development of diabetes and causal relationship between hyperglycemia, glycosylation, and other post-translational modifications may lead to the development of novel therapeutics that would efficiently alleviate painful PDN by targeting disease-specific mechanisms rather than individual nociceptive ion channels.
Collapse
Affiliation(s)
- Sonja L Joksimovic
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO, United States
| | | | - Slobodan M Todorovic
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO, United States
- Neuroscience Graduate Program, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
13
|
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: 2] [Impact Index Per Article: 1.0] [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
|
14
|
Targeting T-type/CaV3.2 channels for chronic pain. Transl Res 2021; 234:20-30. [PMID: 33422652 PMCID: PMC8217081 DOI: 10.1016/j.trsl.2021.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 01/09/2023]
Abstract
T-type calcium channels regulate neuronal excitability and are important contributors of pain processing. CaV3.2 channels are the major isoform expressed in nonpeptidergic and peptidergic nociceptive neurons and are emerging as promising targets for pain treatment. Numerous studies have shown that CaV3.2 expression and/or activity are significantly increased in spinal dorsal horn and in dorsal root ganglia neurons in different inflammatory and neuropathic pain models. Pharmacological campaigns to inhibit the functional expression of CaV3.2 for treatment of pain have focused on the development of direct channel blockers, but none have produced lead candidates. Targeting the proteins that regulate the trafficking or transcription, and the ones that modify the channels via post-translational modifications are alternative means to regulate expression and function of CaV3.2 channels and hence to develop new drugs to control pain. Here we synthesize data supporting a role for CaV3.2 in numerous pain modalities and then discuss emerging opportunities for the indirect targeting of CaV3.2 channels.
Collapse
|
15
|
Macrophage as a Peripheral Pain Regulator. Cells 2021; 10:cells10081881. [PMID: 34440650 PMCID: PMC8392675 DOI: 10.3390/cells10081881] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/30/2022] Open
Abstract
A neuroimmune crosstalk is involved in somatic and visceral pathological pain including inflammatory and neuropathic components. Apart from microglia essential for spinal and supraspinal pain processing, the interaction of bone marrow-derived infiltrating macrophages and/or tissue-resident macrophages with the primary afferent neurons regulates pain signals in the peripheral tissue. Recent studies have uncovered previously unknown characteristics of tissue-resident macrophages, such as their origins and association with regulation of pain signals. Peripheral nerve macrophages and intestinal resident macrophages, in addition to adult monocyte-derived infiltrating macrophages, secrete a variety of mediators, such as tumor necrosis factor-α, interleukin (IL)-1β, IL-6, high mobility group box 1 and bone morphogenic protein 2 (BMP2), that regulate the excitability of the primary afferents. Neuron-derived mediators including neuropeptides, ATP and macrophage-colony stimulating factor regulate the activity or polarization of diverse macrophages. Thus, macrophages have multitasks in homeostatic conditions and participate in somatic and visceral pathological pain by interacting with neurons.
Collapse
|
16
|
A modulator of the low-voltage-activated T-type calcium channel that reverses HIV glycoprotein 120-, paclitaxel-, and spinal nerve ligation-induced peripheral neuropathies. Pain 2021; 161:2551-2570. [PMID: 32541387 DOI: 10.1097/j.pain.0000000000001955] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The voltage-gated calcium channels CaV3.1-3.3 constitute the T-type subfamily, whose dysfunctions are associated with epilepsy, psychiatric disorders, and chronic pain. The unique properties of low-voltage-activation, faster inactivation, and slower deactivation of these channels support their role in modulation of cellular excitability and low-threshold firing. Thus, selective T-type calcium channel antagonists are highly sought after. Here, we explored Ugi-azide multicomponent reaction products to identify compounds targeting T-type calcium channel. Of the 46 compounds tested, an analog of benzimidazolonepiperidine-5bk (1-{1-[(R)-{1-[(1S)-1-phenylethyl]-1H-1,2,3,4-tetrazol-5-yl}(thiophen-3-yl)methyl]piperidin-4-yl}-2,3-dihydro-1H-1,3-benzodiazol-2-one) modulated depolarization-induced calcium influx in rat sensory neurons. Modulation of T-type calcium channels by 5bk was further confirmed in whole-cell patch clamp assays in dorsal root ganglion (DRG) neurons, where pharmacological isolation of T-type currents led to a time- and concentration-dependent regulation with a low micromolar IC50. Lack of an acute effect of 5bk argues against a direct action on T-type channels. Genetic knockdown revealed CaV3.2 to be the isoform preferentially modulated by 5bk. High voltage-gated calcium, as well as tetrodotoxin-sensitive and -resistant sodium, channels were unaffected by 5bk. 5bk inhibited spontaneous excitatory postsynaptic currents and depolarization-evoked release of calcitonin gene-related peptide from lumbar spinal cord slices. Notably, 5bk did not bind human mu, delta, or kappa opioid receptors. 5bk reversed mechanical allodynia in rat models of HIV-associated neuropathy, chemotherapy-induced peripheral neuropathy, and spinal nerve ligation-induced neuropathy, without effects on locomotion or anxiety. Thus, 5bk represents a novel T-type modulator that could be used to develop nonaddictive pain therapeutics.
Collapse
|
17
|
Tomita S, Sekiguchi F, Kasanami Y, Naoe K, Tsubota M, Wake H, Nishibori M, Kawabata A. Ca v3.2 overexpression in L4 dorsal root ganglion neurons after L5 spinal nerve cutting involves Egr-1, USP5 and HMGB1 in rats: An emerging signaling pathway for neuropathic pain. Eur J Pharmacol 2020; 888:173587. [PMID: 32971090 DOI: 10.1016/j.ejphar.2020.173587] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
Overexpression of Cav3.2 T-type Ca2+ channels in L4 dorsal root ganglion (DRG) participates in neuropathic pain after L5 spinal nerve cutting (L5SNC) in rats. The L5SNC-induced neuropathic pain also involves high mobility group box 1 (HMGB1), a damage-associated molecular pattern protein, and its target, the receptor for advanced glycation end-products (RAGE). We thus studied the molecular mechanisms for the L5SNC-induced Cav3.2 overexpression as well as neuropathic pain in rats by focusing on; 1) possible involvement of early growth response 1 (Egr-1), known to regulate transcriptional expression of Cav3.2, and ubiquitin-specific protease 5 (USP5) that protects Cav3.2 from proteasomal degradation, and 2) possible role of HMGB1/RAGE as an upstream signal. Protein levels of Cav3.2 as well as Egr-1 in L4 DRG significantly increased in the early (day 6) and persistent (day 14) phases of neuropathy after L5SNC, while USP5 protein in L4 DRG did not increase on day 6, but day 14. An anti-HMGB1-neutralizing antibody or a low molecular weight heparin, a RAGE antagonist, prevented the development of neuropathic pain and upregulation of Egr-1 and Cav3.2 in L4 DRG after L5SNC. L5SNC increased macrophages accumulating in the sciatic nerves, and the cytoplasm/nuclear ratio of immunoreactive HMGB1 in those macrophages. Our findings suggest that L5SNC-induced Cav3.2 overexpression in L4 DRG and neuropathic pain involves Egr-1 upregulation downstream of the macrophage-derived HMGB1/RAGE pathway, and that the delayed upregulation of USP5 might contribute to the persistent Cav3.2 overexpression and neuropathy.
Collapse
Affiliation(s)
- Shiori Tomita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yoshihito Kasanami
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Katsuki Naoe
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly Known As Kinki University), 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
18
|
Role of H 2S in pain: Growing evidences of mystification. Eur J Pharmacol 2020; 883:173322. [PMID: 32619675 DOI: 10.1016/j.ejphar.2020.173322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/31/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
There have been studies suggesting the pain attenuating as well as pain inducing actions of hydrogen sulfide (H2S). Exogenous administrated H2S may be antinociceptive or pronociceptive, while the endogenous H2S is pronociceptive. Experimental studies have shown that pharmacological inhibitors of H2S biosynthetic enzymes may attenuate nociceptive as well as neuropathic pain. It suggests that nerve injury or inflammatory agents may induce the expression of H2S biosynthetic enzymes to increase the endogenous production of H2S, which acts as a pain neurotransmitter to produce pain. The endogenous H2S may act through different mechanisms including opening of T-type calcium channels, activation of voltage-gated sodium channels, suppression of potassium channels, activation of TRPA1, TRPV1 and TRPC6 channels, upregulation of spinal NMDA receptors and sensitization of purinergic receptors. Exogenous administration of H2S/precursors/donors attenuates or facilitates pain. It may be hypothesized that local administration of H2S may cause pain; while it's systemic administration may attenuate pain. The doses of H2S may also influence the pain response and H2S in low doses may contribute in reducing pain, while H2S in high doses may contribute in relieving pain. Accordingly, enzymatic inhibitors of H2S synthesis or systemic administration of slow H2S releasing agents/low dose H2S donors may be useful in attenuating nociceptive and neuropathic pain. The present review describes the dual role of H2S in pain attenuation and pain induction along with possible mechanisms.
Collapse
|
19
|
Zhou Y, Cai S, Gomez K, Wijeratne EMK, Ji Y, Bellampalli SS, Luo S, Moutal A, Gunatilaka AAL, Khanna R. 1-O-Acetylgeopyxin A, a derivative of a fungal metabolite, blocks tetrodotoxin-sensitive voltage-gated sodium, calcium channels and neuronal excitability which correlates with inhibition of neuropathic pain. Mol Brain 2020; 13:73. [PMID: 32393368 PMCID: PMC7216607 DOI: 10.1186/s13041-020-00616-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/04/2020] [Indexed: 01/03/2023] Open
Abstract
Chronic pain can be the result of an underlying disease or condition, medical treatment, inflammation, or injury. The number of persons experiencing this type of pain is substantial, affecting upwards of 50 million adults in the United States. Pharmacotherapy of most of the severe chronic pain patients includes drugs such as gabapentinoids, re-uptake blockers and opioids. Unfortunately, gabapentinoids are not effective in up to two-thirds of this population and although opioids can be initially effective, their long-term use is associated with multiple side effects. Therefore, there is a great need to develop novel non-opioid alternative therapies to relieve chronic pain. For this purpose, we screened a small library of natural products and their derivatives in the search for pharmacological inhibitors of voltage-gated calcium and sodium channels, which are outstanding molecular targets due to their important roles in nociceptive pathways. We discovered that the acetylated derivative of the ent-kaurane diterpenoid, geopyxin A, 1-O-acetylgeopyxin A, blocks voltage-gated calcium and tetrodotoxin-sensitive voltage-gated sodium channels but not tetrodotoxin-resistant sodium channels in dorsal root ganglion (DRG) neurons. Consistent with inhibition of voltage-gated sodium and calcium channels, 1-O-acetylgeopyxin A reduced reduce action potential firing frequency and increased firing threshold (rheobase) in DRG neurons. Finally, we identified the potential of 1-O-acetylgeopyxin A to reverse mechanical allodynia in a preclinical rat model of HIV-induced sensory neuropathy. Dual targeting of both sodium and calcium channels may permit block of nociceptor excitability and of release of pro-nociceptive transmitters. Future studies will harness the core structure of geopyxins for the generation of antinociceptive drugs.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Clinical Laboratory, the First Hospital of Jilin University, Changchun, 130021, China
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - E M Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources & the Environment, College of Agriculture & Life Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Yingshi Ji
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Shreya S Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources & the Environment, College of Agriculture & Life Sciences, The University of Arizona, Tucson, AZ, 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ, 85724, USA.
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, Tucson, AZ, 85724, USA.
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, AZ, 85724, USA.
| |
Collapse
|
20
|
Martínez-Hernández E, Zeglin A, Almazan E, Perissinotti P, He Y, Koob M, Martin JL, Piedras-Rentería ES. KLHL1 Controls Ca V3.2 Expression in DRG Neurons and Mechanical Sensitivity to Pain. Front Mol Neurosci 2020; 12:315. [PMID: 31969803 PMCID: PMC6960199 DOI: 10.3389/fnmol.2019.00315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/05/2019] [Indexed: 11/29/2022] Open
Abstract
Dorsal root ganglion (DRG) neurons process pain signaling through specialized nociceptors located in their peripheral endings. It has long been established low voltage-activated (LVA) CaV3.2 calcium channels control neuronal excitability during sensory perception in these neurons. Silencing CaV3.2 activity with antisense RNA or genetic ablation results in anti-nociceptive, anti-hyperalgesic and anti-allodynic effects. CaV3.2 channels are regulated by many proteins (Weiss and Zamponi, 2017), including KLHL1, a neuronal actin-binding protein that stabilizes channel activity by recycling it back to the plasma membrane through the recycling endosome. We explored whether manipulation of KLHL1 levels and thereby function as a CaV3.2 modifier can modulate DRG excitability and mechanical pain transmission or sensitivity to pain. We first assessed the mechanical sensitivity threshold and DRG properties in the KLHL1 KO mouse model. KO DRG neurons exhibited smaller T-type current density compared to WT without significant changes in voltage dependence, as expected in the absence of its modulator. Western blot analysis confirmed CaV3.2 but not CaV3.1, CaV3.3, CaV2.1, or CaV2.2 protein levels were significantly decreased; and reduced neuron excitability and decreased pain sensitivity were also found in the KLHL1 KO model. Analogously, transient down-regulation of KLHL1 levels in WT mice with viral delivery of anti-KLHL1 shRNA also resulted in decreased pain sensitivity. These two experimental approaches confirm KLHL1 as a physiological modulator of excitability and pain sensitivity, providing a novel target to control peripheral pain.
Collapse
Affiliation(s)
- Elizabeth Martínez-Hernández
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Neuroscience Division of the Cardiovascular Institute, Loyola University Chicago, Maywood, IL, United States
| | - Alissa Zeglin
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Erik Almazan
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
| | - Paula Perissinotti
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Neuroscience Division of the Cardiovascular Institute, Loyola University Chicago, Maywood, IL, United States
| | - Yungui He
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Michael Koob
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Jody L. Martin
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Neuroscience Division of the Cardiovascular Institute, Loyola University Chicago, Maywood, IL, United States
| | - Erika S. Piedras-Rentería
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, United States
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Neuroscience Division of the Cardiovascular Institute, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
21
|
Cdk5-Dependent Phosphorylation of Ca V3.2 T-Type Channels: Possible Role in Nerve Ligation-Induced Neuropathic Allodynia and the Compound Action Potential in Primary Afferent C Fibers. J Neurosci 2019; 40:283-296. [PMID: 31744861 DOI: 10.1523/jneurosci.0181-19.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 11/05/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Voltage-gated T-type Ca2+ (CaV3) channels regulate diverse physiological events, including neuronal excitability, and have been linked to several pathological conditions such as absence epilepsy, cardiovascular diseases, and neuropathic pain. It is also acknowledged that calcium/calmodulin-dependent protein kinase II and protein kinases A and C regulate the activity of T-type channels. Interestingly, peripheral nerve injury induces tactile allodynia and upregulates CaV3.2 channels and cyclin-dependent kinase 5 (Cdk5) in dorsal root ganglia (DRG) and spinal dorsal horn. Here, we report that recombinant CaV3.2 channels expressed in HEK293 cells are regulatory targets of Cdk5. Site-directed mutagenesis showed that the relevant sites for this regulation are residues S561 and S1987. We also found that Cdk5 may regulate CaV3.2 channel functional expression in rats with mechanical allodynia induced by spinal nerve ligation (SNL). Consequently, the Cdk5 inhibitor olomoucine affected the compound action potential recorded in the spinal nerves, as well as the paw withdrawal threshold. Likewise, Cdk5 expression was upregulated after SNL in the DRG. These findings unveil a novel mechanism for how phosphorylation may regulate CaV3.2 channels and suggest that increased channel activity by Cdk5-mediated phosphorylation after SNL contributes nerve injury-induced tactile allodynia.SIGNIFICANCE STATEMENT Neuropathic pain is a current public health challenge. It can develop as a result of injury or nerve illness. It is acknowledged that the expression of various ion channels can be altered in neuropathic pain, including T-type Ca2+ channels that are expressed in sensory neurons, where they play a role in the regulation of cellular excitability. The present work shows that the exacerbated expression of Cdk5 in a preclinical model of neuropathic pain increases the functional expression of CaV3.2 channels. This finding is relevant for the understanding of the molecular pathophysiology of the disease. Additionally, this work may have a substantial translational impact, since it describes a novel molecular pathway that could represent an interesting therapeutic alternative for neuropathic pain.
Collapse
|
22
|
Nguyen HD, Okada T, Sekiguchi F, Tsubota M, Nishikawa H, Kawabata A, Toyooka N. Prenylflavanones as Novel T-Type Calcium Channel Blockers Useful for Pain Therapy. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19873441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Prenylated flavonoids have attracted much attention due to their promising and diverse bioactivities on multitarget tissues. To the best of our knowledge, our recent studies demonstrated first that (2 S)-6-prenylnaringenin (6-PNG), a hop component, blocks Cav3.2 T-type calcium channels (T-channels) and alleviates neuropathic and visceral pain with little side effects; it also indicated first that other natural prenylflavanones (PFVNs), such as sophoraflavanone G and (2 S)-8-PNG, or synthetic 6-PFVNs including (2 R/S)-6-PNG and its derivatives are capable of blocking T-channels and useful for pain therapy. Through the structure-activity relationship studies on the synthetic 6-PFVNs, we identified 6-(3-ethylpent-2-enyl)-5,7-dihydroxy-2-(2-hydroxyphenyl)chroman-4-one (8j or KTt-45) as the most potent blocker of Cav3.2 T-channels. It is interesting to recognize a prenylated flavonoid, belonging to other sub-classes, as a novel T-channel blocker. Therefore, this article will review some of our recent studies to introduce a new branch to researchers studying on prenylated flavonoids.
Collapse
Affiliation(s)
- Huy Du Nguyen
- Graduate School of Innovative Life Science, University of Toyama, Japan
| | - Takuya Okada
- Graduate School of Innovative Life Science, University of Toyama, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Hiroyuki Nishikawa
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, University of Toyama, Japan
- Graduate School of Science and Engineering, University of Toyama, Japan
| |
Collapse
|
23
|
Matsui K, Tsubota M, Fukushi S, Koike N, Masuda H, Kasanami Y, Miyazaki T, Sekiguchi F, Ohkubo T, Yoshida S, Mukai Y, Oita A, Takada M, Kawabata A. Genetic deletion of Ca v3.2 T-type calcium channels abolishes H 2S-dependent somatic and visceral pain signaling in C57BL/6 mice. J Pharmacol Sci 2019; 140:310-312. [PMID: 31492577 DOI: 10.1016/j.jphs.2019.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 10/26/2022] Open
Abstract
We tested whether genetic deletion of Cav3.2 T-type Ca2+ channels abolishes hydrogen sulfide (H2S)-mediated pain signals in mice. In Cav3.2-expressing HEK293 cells, Na2S, an H2S donor, at 100 μM clearly increased Ba2+ currents, as assessed by whole-cell patch-clamp recordings. In wild-type C57BL/6 mice, intraplantar and intracolonic administration of Na2S evoked mechanical allodynia and visceral nociceptive behavior, respectively, which were abolished by TTA-A2, a T-type Ca2+ channel blocker. In Cav3.2-knockout mice of a C57BL/6 background, Na2S caused neither somatic allodynia nor colonic nociception. Our study thus provides definitive evidence for an essential role of Cav3.2 in H2S-dependent somatic and colonic pain.
Collapse
Affiliation(s)
- Kazuki Matsui
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan; Department of Pharmacy, National Cerebral and Cardiovascular Center, Suita, 565-8565, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Saaya Fukushi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Nene Koike
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Hiroshi Masuda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Yoshihito Kasanami
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Takaya Miyazaki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Tsuyako Ohkubo
- Division of Basic Medical Sciences and Fundamental Nursing, Faculty of Nursing, Fukuoka Nursing College, Fukuoka, 814-0193, Japan
| | - Shigeru Yoshida
- Department of Life Science, Faculty of Science and Engineering, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Yutaro Mukai
- Department of Pharmacy, National Cerebral and Cardiovascular Center, Suita, 565-8565, Japan
| | - Akira Oita
- Department of Pharmacy, National Cerebral and Cardiovascular Center, Suita, 565-8565, Japan
| | - Mitsutaka Takada
- Division of Clinical Drug Informatics, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
24
|
Liu Q, Chen W, Fan X, Wang J, Fu S, Cui S, Liao F, Cai J, Wang X, Huang Y, Su L, Zhong L, Yi M, Liu F, Wan Y. Upregulation of interleukin-6 on Cav3.2 T-type calcium channels in dorsal root ganglion neurons contributes to neuropathic pain in rats with spinal nerve ligation. Exp Neurol 2019; 317:226-243. [DOI: 10.1016/j.expneurol.2019.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/22/2019] [Accepted: 03/09/2019] [Indexed: 10/27/2022]
|
25
|
Tsubota M, Uebo K, Miki K, Sekiguchi F, Ishigami A, Kawabata A. Dietary ascorbic acid restriction in GNL/SMP30-knockout mice unveils the role of ascorbic acid in regulation of somatic and visceral pain sensitivity. Biochem Biophys Res Commun 2019; 511:705-710. [PMID: 30827506 DOI: 10.1016/j.bbrc.2019.02.102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 10/27/2022]
Abstract
Cav3.2 T-type Ca2+ channels are expressed in the primary afferents and play a pronociceptive role. The activity of Cav3.2 is enhanced by H2S, a gasotransmitter, and suppressed by ascorbic acid (vitamin C) through metal-catalyzed oxidation of the Zn2+-binding His191 in Cav3.2. Since rodents, but not humans, are capable of synthesizing ascorbic acid, the present study examined the role of ascorbic acid in nociceptive processing, using the mice lacking GNL/SMP30, an enzyme essential for ascorbic acid biosynthesis. Intraplantar and intracolonic administration of NaHS, an H2S donor, caused somatic allodynia and referred hyperalgesia, respectively, and repeated treatment with paclitaxel produced neuropathic allodynia in wild-type mice, all of which were suppressed by ascorbic acid or T-type Ca2+ channel blockers. Dietary ascorbic acid restriction caused dramatic decreases in plasma and tissue ascorbic acid levels in GNL/SMP30-knockout, but not wild-type, mice. The ascorbic acid restriction enhanced the somatic and visceral hypersensitivity following intraplantar and intracolonic NaHS, respectively, and paclitaxel-induced neuropathy in GNL/SMP30-knockout mice, while it had no such effect in wild-type mice. Together, our data unveil the critical role of ascorbic acid in regulating somatic and visceral pain sensitivity and support accumulating clinical evidence for the usefulness of ascorbic acid in pain management.
Collapse
Affiliation(s)
- Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8502, Japan
| | - Kenta Uebo
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8502, Japan
| | - Koki Miki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8502, Japan
| | - Akihiko Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
26
|
Shimada T, Takahashi K, Tominaga M, Ohta T. Identification of molecular targets for toxic action by persulfate, an industrial sulfur compound. Neurotoxicology 2019; 72:29-37. [PMID: 30738091 DOI: 10.1016/j.neuro.2019.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/18/2019] [Accepted: 02/04/2019] [Indexed: 02/02/2023]
Abstract
Persulfate salts are broadly used as industrial chemicals and exposure to them causes occupational asthma, occupational rhinitis and contact dermatitis. However, the mechanisms underlying these toxic actions are not fully elucidated. Transient receptor potential (TRP) vanilloid 1 (V1), ankyrin 1 (A1) and melastatin 8 (M8) are non-selective cation channels preferentially expressing sensory neurons. These channels are known to be involved in respiratory and skin diseases. In the present study, we investigated the effects of sodium persulfate on these TRP channels. In wild-type mouse sensory neurons, persulfate evoked [Ca2+]i increases that were inhibited by removal of extracellular Ca2+ or blockers of TRPA1 but not by those of TRPV1 and TRPM8. Persulfate failed to evoke [Ca2+]i responses in neurons from TRPA1(-/-) mice, but did evoke them in neurons from TRPV1(-/-) mice. In HEK 293 cells expressing mouse TRPA1 (mTRPA1-HEK), persulfate induced [Ca2+]i increases. Moreover, in HEK 293 cells expressing mouse TRPV1 (mTRPV1-HEK), a high concentration of persulfate also evoked [Ca2+]i increases. Similar [Ca2+]i responses were observed in HEK 293 cells expressing human TRPA1 and human TRPV1. Current responses were also elicited by persulfate in mTRPA1- and mTRPV1-HEK. Analysis using mutated channels revealed that persulfate acted on electrophilic agonist-sensitive cysteine residues of TRPA1, and it indirectly activated TRPV1 due to the external acidification, because of the disappearance of [Ca2+]i responses in acid-insensitive mTRPV1 mutant. These results demonstrate that persulfate activates nociceptive TRPA1 and TRPV1 channels. It is suggested that activation of these nociceptive channels may be involved in respiratory and skin injuries caused by exposure to this industrial sulfur compound. Thus, selective TRPA1 and TRPV1 channel blockers may be effective to remedy persulfate-induced toxic actions.
Collapse
Affiliation(s)
- Takahisa Shimada
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Kenji Takahashi
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Japan
| | - Toshio Ohta
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, Japan.
| |
Collapse
|
27
|
Cheng XE, Ma LX, Feng XJ, Zhu MY, Zhang DY, Xu LL, Liu T. Antigen retrieval pre-treatment causes a different expression pattern of Cav3.2 in rat and mouse spinal dorsal horn. Eur J Histochem 2019; 63. [PMID: 30678436 PMCID: PMC6346256 DOI: 10.4081/ejh.2019.2988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/20/2019] [Indexed: 11/27/2022] Open
Abstract
Cav3 channels consist of three isoforms, Cav3.1 (α1G), Cav3.2 (α1H), and Cav3.3 (α1I), which produce low-threshold spikes that trigger burst firings in nociceptive neurons of the spinal dorsal horn (SDH) and dorsal root ganglion (DRG). Although Cav3.2 plays a crucial role in pathological pain, it is distribution in SDH still remains controversial. One study showed that Cav3.2 is ubiquitously expressed in neurons, but another study implied that Cav3.2 is expressed restricted to astrocytes. To unravel these discrepancies, we used methods of immunohistochemistry either with or without antigen retrieval (AR) pre-treatment to detect Cav3 in SDH and DRG from both rats and mice. Moreover, Cav3.2 mRNA was detected in mice SDH using in situ hybridization. We found that the expression pattern of Cav3.2 but not Cav3.1 and Cav3.3 in SDH were largely different with or without AR pre-treatment, which showed a neuron- like and an astrocyte-like appearance, respectively. Double staining further demonstrated that Cav3.2 was mainly costained with the neuronal marker NeuN in the presence of AR but was with glial fibrillary acidic protein (GFAP, marker for astrocytes) in the absence of AR pre-treatment. Importantly, Cav3.2 mRNA was mainly colocalized with Cav3.2 but not GFAP. Together, our findings indicate that AR pretreatment or not impacts the expression pattern of Cav3.2, which may make a significant contribution to the future study of Cav3.2 in SDH.
Collapse
Affiliation(s)
- Xiao E Cheng
- The First Affiliated Hospital of Nanchang University, Center for Experimental Medicine.
| | | | | | | | | | | | | |
Collapse
|
28
|
Roa-Coria JE, Pineda-Farias JB, Barragán-Iglesias P, Quiñonez-Bastidas GN, Zúñiga-Romero Á, Huerta-Cruz JC, Reyes-García JG, Flores-Murrieta FJ, Granados-Soto V, Rocha-González HI. Possible involvement of peripheral TRP channels in the hydrogen sulfide-induced hyperalgesia in diabetic rats. BMC Neurosci 2019; 20:1. [PMID: 30602386 PMCID: PMC6317195 DOI: 10.1186/s12868-018-0483-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Peripheral diabetic neuropathy can be painful and its symptoms include hyperalgesia, allodynia and spontaneous pain. Hydrogen sulfide (H2S) is involved in diabetes-induced hyperalgesia and allodynia. However, the molecular target through which H2S induces hyperalgesia in diabetic animals is unclear. The aim of this study was to determine the possible involvement of transient receptor potential (TRP) channels in H2S-induced hyperalgesia in diabetic rats. RESULTS Streptozotocin (STZ) injection produced hyperglycemia in rats. Intraplantar injection of NaHS (an exogenous donor of H2S, 3-100 µg/paw) induced hyperalgesia, in a time-dependent manner, in formalin-treated diabetic rats. NaHS-induced hyperalgesia was partially prevented by local intraplantar injection of capsazepine (0.3-3 µg/paw), HC-030031 (100-316 µg/paw) and SKF-96365 (10-30 µg/paw) blockers, at 21 days post-STZ injection. At the doses used, these blockers did not modify formalin-induced nociception. Moreover, capsazepine (0.3-30 µg/paw), HC-030031 (100-1000 µg/paw) and SKF-96365 (10-100 µg/paw) reduced formalin-induced nociception in diabetic rats. Contralateral injection of the highest doses used did not modify formalin-induced flinching behavior. Hyperglycemia, at 21 days, also increased protein expression of cystathionine-β-synthase enzyme (CBS) and TRPC6, but not TRPA1 nor TRPV1, channels in dorsal root ganglia (DRG). Repeated injection of NaHS enhanced CBS and TRPC6 expression, but hydroxylamine (HA) prevented the STZ-induced increase of CBS protein. In addition, daily administration of SKF-96365 diminished TRPC6 protein expression, whereas NaHS partially prevented the decrease of SKF-96365-induced TRPC6 expression. Concordantly, daily intraplantar injection of NaHS enhanced, and HA prevented STZ-induced intraepidermal fiber loss, respectively. CBS was expressed in small- and medium-sized cells of DRG and co-localized with TRPV1, TRPA1 and TRPC6 in IB4-positive neurons. CONCLUSIONS Our data suggest that H2S leads to hyperalgesia in diabetic rats through activation of TRPV1, TRPA1 and TRPC channels and, subsequent intraepidermal fibers loss. CBS enzyme inhibitors or TRP-channel blockers could be useful for treatment of painful diabetic neuropathy.
Collapse
Affiliation(s)
- José Eduardo Roa-Coria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
| | - Jorge Baruch Pineda-Farias
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Unidad Coapa, Calzada de los Tenorios 235, Col. Granjas Coapa, 14330 Ciudad de México, Mexico
| | - Paulino Barragán-Iglesias
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Unidad Coapa, Calzada de los Tenorios 235, Col. Granjas Coapa, 14330 Ciudad de México, Mexico
| | - Geovanna Nallely Quiñonez-Bastidas
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
| | - Ángel Zúñiga-Romero
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
| | - Juan Carlos Huerta-Cruz
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Secretaría de Salud, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, Mexico
| | - Juan Gerardo Reyes-García
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
| | - Francisco Javier Flores-Murrieta
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
- Unidad de Investigación en Farmacología, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Secretaría de Salud, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, Mexico
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Unidad Coapa, Calzada de los Tenorios 235, Col. Granjas Coapa, 14330 Ciudad de México, Mexico
| | - Héctor Isaac Rocha-González
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
| |
Collapse
|
29
|
Feng XJ, Ma LX, Jiao C, Kuang HX, Zeng F, Zhou XY, Cheng XE, Zhu MY, Zhang DY, Jiang CY, Liu T. Nerve injury elevates functional Cav3.2 channels in superficial spinal dorsal horn. Mol Pain 2019; 15:1744806919836569. [PMID: 30803310 PMCID: PMC6458665 DOI: 10.1177/1744806919836569] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/02/2019] [Accepted: 02/12/2019] [Indexed: 01/23/2023] Open
Abstract
Cav3 channels play an important role in modulating chronic pain. However, less is known about the functional changes of Cav3 channels in superficial spinal dorsal horn in neuropathic pain states. Here, we examined the effect of partial sciatic nerve ligation (PSNL) on either expression or electrophysiological properties of Cav3 channels in superficial spinal dorsal horn. Our in vivo studies showed that the blockers of Cav3 channels robustly alleviated PSNL-induced mechanical allodynia and thermal hyperalgesia, which lasted at least 14 days following PSNL. Meanwhile, PSNL triggered an increase in both mRNA and protein levels of Cav3.2 but not Cav3.1 or Cav3.3 in rats. However, in Cav3.2 knockout mice, PSNL predominantly attenuated mechanical allodynia but not thermal hyperalgesia. In addition, the results of whole-cell patch-clamp recordings showed that both the overall proportion of Cav3 current-expressing neurons and the Cav3 current density in individual neurons were elevated in spinal lamina II neurons from PSNL rats, which could not be recapitulated in Cav3.2 knockout mice. Altogether, our findings reveal that the elevated functional Cav3.2 channels in superficial spinal dorsal horn may contribute to the mechanical allodynia in PSNL-induced neuropathic pain model.
Collapse
Affiliation(s)
- Xiao-Jin Feng
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Long-Xian Ma
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cui Jiao
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hai-Xia Kuang
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Zeng
- Department of Pain Clinic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xue-Ying Zhou
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-E Cheng
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meng-Ye Zhu
- Department of Pain Clinic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Da-Ying Zhang
- Department of Pain Clinic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chang-Yu Jiang
- Jisheng Han Academician Workstation for Pain Medicine, Nanshan Hospital, Shenzhen, China
| | - Tao Liu
- Center for Experimental Medicine, the First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Pediatrics, the First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Tsubota M, Kawabata A. [Regulation of Ca v3.2-mediated pain signals by hydrogen sulfide]. Nihon Yakurigaku Zasshi 2019; 154:128-132. [PMID: 31527362 DOI: 10.1254/fpj.154.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Hydrogen sulfide (H2S), an endogenous gasotransmitter, is generated from L-cysteine by 3 distinct enzymes including cystathionine-γ-lyase (CSE), and targets multiple molecules, thereby playing various roles in health and disease. H2S triggers or accelerates somatic pain and visceral nociceptive signals in the pancreas, colon and bladder by enhancing the activity of Cav3.2 T-type calcium channels. H2S also activates TRPA1, which participates in H2S-induced somatic pain signaling. However, Cav3.2 predominantly mediates colonic nociception by H2S, because genetic deletion of TRPA1 does not reduce H2S-induced colonic pain. The functional upregulation of the CSE/H2S/Cav3.2 system is involved in neuropathic pain and visceral pain accompanying pancreatitis and cystitis. Cav3.2 also appears to participate in irritable bowel syndrome (IBS), although the role of endogenous H2S generation by CSE in IBS is still open to question. In this review, we describe how H2S regulates pain signals, particularly by interacting with Cav3.2.
Collapse
Affiliation(s)
- Maho Tsubota
- Division of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University
| | - Atsufumi Kawabata
- Division of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University
| |
Collapse
|
31
|
Tomita S, Sekiguchi F, Deguchi T, Miyazaki T, Ikeda Y, Tsubota M, Yoshida S, Nguyen HD, Okada T, Toyooka N, Kawabata A. Critical role of Ca v3.2 T-type calcium channels in the peripheral neuropathy induced by bortezomib, a proteasome-inhibiting chemotherapeutic agent, in mice. Toxicology 2018; 413:33-39. [PMID: 30552955 DOI: 10.1016/j.tox.2018.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/30/2023]
Abstract
Bortezomib, a first-line agent for treatment of multiple myeloma, exhibits anticancer activity through proteasome inhibition. However, bortezomib-induced peripheral neuropathy (BIPN) is one of the most serious side effects. Since decreased proteasomal degradation of Cav3.2 T-type calcium channels in the primary afferents is involved in persistent pain, we investigated whether BIPN involves increased protein levels of Cav3.2 in mice. Six repeated i.p. administrations of bortezomib for 12 days developed persistent mechanical allodynia. Systemic administration of novel T-type calcium channel blockers, (2R/S)-6-prenylnaringenin and KTt-45, and of TTA-A2, the well-known blocker, reversed the BIPN. Ascorbic acid, known to block Cav3.2, but not Cav3.1 or 3.3, and silencing of Cav3.2 gene also suppressed BIPN. Protein levels of Cav3.2 in the dorsal root ganglion (DRG) at L4-L6 levels increased throughout days 1-21 after the onset of bortezomib treatment. Protein levels of USP5, a deubiquitinating enzyme that specifically inhibits proteasomal degradation of Cav3.2, increased in DRG on days 3-21, but not day 1, in bortezomib-treated mice. In DRG-derived ND7/23 cells, bortezomib increased protein levels of Cav3.2 and T-channel-dependent currents, as assessed by a patch-clamp method, but did not upregulate expression of Cav3.2 mRNA or USP5 protein. MG-132, another proteasome inhibitor, also increased Cav3.2 protein levels in the cultured cells. Given the previous evidence for USP5 induction following nociceptor excitation, our data suggest that BIPN involves the increased protein levels of Cav3.2 in nociceptors through inhibition of proteasomal degradation of Cav3.2 by bortezomib itself and then by USP5 that is upregulated probably in an activity-dependent manner.
Collapse
Affiliation(s)
- Shiori Tomita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Tomoyo Deguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Takaya Miyazaki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Yuya Ikeda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan
| | - Shigeru Yoshida
- Department of Life Science, Faculty of Science and Engineering, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Huy Du Nguyen
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-855, Japan
| | - Takuya Okada
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-855, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-855, Japan; Graduate School of Science and Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formerly known as Kinki University), Higashi-Osaka, 577-8802, Japan.
| |
Collapse
|
32
|
Kang XJ, Chi YN, Chen W, Liu FY, Cui S, Liao FF, Cai J, Wan Y. Increased expression of Ca V3.2 T-type calcium channels in damaged DRG neurons contributes to neuropathic pain in rats with spared nerve injury. Mol Pain 2018; 14:1744806918765808. [PMID: 29592785 PMCID: PMC5888807 DOI: 10.1177/1744806918765808] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Ion channels are very important in the peripheral sensitization in neuropathic pain. Our present study aims to investigate the possible contribution of CaV3.2 T-type calcium channels in damaged dorsal root ganglion neurons in neuropathic pain. We established a neuropathic pain model of rats with spared nerve injury. In these model rats, it was easy to distinguish damaged dorsal root ganglion neurons (of tibial nerve and common peroneal nerve) from intact dorsal root ganglion neurons (of sural nerves). Our results showed that CaV3.2 protein expression increased in medium-sized neurons from the damaged dorsal root ganglions but not in the intact ones. With whole cell patch clamp recording technique, it was found that after-depolarizing amplitudes of the damaged medium-sized dorsal root ganglion neurons increased significantly at membrane potentials of −85 mV and −95 mV. These results indicate a functional up-regulation of CaV3.2 T-type calcium channels in the damaged medium-sized neurons after spared nerve injury. Behaviorally, blockade of CaV3.2 with antisense oligodeoxynucleotides could significantly reverse mechanical allodynia. These results suggest that CaV3.2 T-type calcium channels in damaged medium-sized dorsal root ganglion neurons might contribute to neuropathic pain after peripheral nerve injury.
Collapse
Affiliation(s)
- Xue-Jing Kang
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Ye-Nan Chi
- 1 Neuroscience Research Institute, Peking University, Beijing, China.,2 Department of Anesthesiology, Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Wen Chen
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Feng-Yu Liu
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Shuang Cui
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Fei-Fei Liao
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - Jie Cai
- 1 Neuroscience Research Institute, Peking University, Beijing, China
| | - You Wan
- 1 Neuroscience Research Institute, Peking University, Beijing, China.,3 Key Lab for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| |
Collapse
|
33
|
Wang D, Ragnarsson L, Lewis RJ. T-type Calcium Channels in Health and Disease. Curr Med Chem 2018; 27:3098-3122. [PMID: 30277145 DOI: 10.2174/0929867325666181001112821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Low Voltage-Activated (LVA) T-type calcium channels are characterized by transient current and Low Threshold Spikes (LTS) that trigger neuronal firing and oscillatory behavior. Combined with their preferential localization in dendrites and their specific "window current", T-type calcium channels are considered to be key players in signal amplification and synaptic integration. Assisted by the emerging pharmacological tools, the structural determinants of channel gating and kinetics, as well as novel physiological and pathological functions of T-type calcium channels, are being uncovered. In this review, we provide an overview of structural determinants in T-type calcium channels, their involvement in disorders and diseases, the development of novel channel modulators, as well as Structure-Activity Relationship (SAR) studies that lead to rational drug design.
Collapse
Affiliation(s)
- Dan Wang
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, the University of Queensland, Brisbane Qld 4072, Australia
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, the University of Queensland, Brisbane Qld 4072, Australia
| | - Richard J Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, the University of Queensland, Brisbane Qld 4072, Australia
| |
Collapse
|
34
|
Stemkowski PL, Garcia-Caballero A, Gadotti VM, M'Dahoma S, Chen L, Souza IA, Zamponi GW. Identification of interleukin-1 beta as a key mediator in the upregulation of Cav3.2-USP5 interactions in the pain pathway. Mol Pain 2018; 13:1744806917724698. [PMID: 28741432 PMCID: PMC5560507 DOI: 10.1177/1744806917724698] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We recently reported that nerve injury or peripheral inflammation triggers an upregulation of the deubiquitinase, USP5 in mouse dorsal root ganglion and spinal dorsal horn. This leads to dysregulated ubiquitination of Cav3.2 T-type calcium channels, thus increasing Cav3.2 channel plasma membrane expression and nociceptive signaling in the primary afferent pain pathway. This phenomenon could be recapitulated by noninvasive, optogenetic activation of transient receptor potential vanilloid-1–expressing nociceptors, indicating that neuronal activity is a key player in this process. Given the relevance of the pro-inflammatory cytokine interleukin-1 beta in many forms of pathological pain, we hypothesized that interleukin-1 beta may be a critical cofactor required to drive upregulation of interactions between USP5 and Cav3.2 channels. Here, we report that gene expression, as well as protein levels for interleukin-1 beta and the endogenous interleukin-1 receptor-I antagonist, IL-1Ra are unaltered following conditioning stimulation of optogenetically targeted cutaneous nociceptors, indicating that neuronal activity is not a driver of interleukin-1 beta signaling. In contrast, co-immunoprecipitation experiments revealed that intrathecal administration of interleukin-1 beta in wild-type mice led to an increase in the interaction between USP5 and Cav3.2 in the spinal dorsal horn. Moreover, disruption of the interaction between USP5 and Cav3.2 with TAT peptides suppressed acute nocifensive responses produced by interleukin-1 beta, which was similar to that achieved by elimination of T-type channel activity with the channel blockers, mibefradil, or TTA-A2. Finally, this upregulation could be maintained in dorsal root ganglion neuron cultures exposed overnight to interleukin-1 beta, while the copresence of interleukin-1 receptor antagonist or the dampening of neuronal cell activity with tetrodotoxin attenuated this response. Altogether, our findings identify interleukin-1 beta as an upstream trigger for the upregulation of interactions between USP5 and Cav3.2 channels in the pain pathway, presumably by triggering increased firing activity in afferent fibers.
Collapse
Affiliation(s)
- Patrick L Stemkowski
- 1Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Agustin Garcia-Caballero
- 1Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Vinicius M Gadotti
- 1Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Said M'Dahoma
- 1Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Lina Chen
- 1Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Ivana A Souza
- 1Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | | |
Collapse
|
35
|
Sekiguchi F, Fujita T, Deguchi T, Yamaoka S, Tomochika K, Tsubota M, Ono S, Horaguchi Y, Ichii M, Ichikawa M, Ueno Y, Koike N, Tanino T, Nguyen HD, Okada T, Nishikawa H, Yoshida S, Ohkubo T, Toyooka N, Murata K, Matsuda H, Kawabata A. Blockade of T-type calcium channels by 6-prenylnaringenin, a hop component, alleviates neuropathic and visceral pain in mice. Neuropharmacology 2018; 138:232-244. [PMID: 29913186 DOI: 10.1016/j.neuropharm.2018.06.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/30/2018] [Accepted: 06/14/2018] [Indexed: 10/14/2022]
Abstract
Since Cav3.2 T-type Ca2+ channels (T-channels) expressed in the primary afferents and CNS contribute to intractable pain, we explored T-channel-blocking components in distinct herbal extracts using a whole-cell patch-clamp technique in HEK293 cells stably expressing Cav3.2 or Cav3.1, and purified and identified sophoraflavanone G (SG) as an active compound from SOPHORAE RADIX (SR). Interestingly, hop-derived SG analogues, (2S)-6-prenylnaringenin (6-PNG) and (2S)-8-PNG, but not naringenin, also blocked T-channels; IC50 (μM) of SG, (2S)-6-PNG and (2S)-8-PNG was 0.68-0.75 for Cav3.2 and 0.99-1.41 for Cav3.1. (2S)-6-PNG and (2S)-8-PNG, but not SG, exhibited reversible inhibition. The racemic (2R/S)-6-PNG as well as (2S)-6-PNG potently blocked Cav3.2, but exhibited minor effect on high-voltage-activated Ca2+ channels and voltage-gated Na+ channels in differentiated NG108-15 cells. In mice, the mechanical allodynia following intraplantar (i.pl.) administration of an H2S donor was abolished by oral or i.p. SR extract and by i.pl. SG, (2S)-6-PNG or (2S)-8-PNG, but not naringenin. Intraperitoneal (2R/S)-6-PNG strongly suppressed visceral pain and spinal ERK phosphorylation following intracolonic administration of an H2S donor in mice. (2R/S)-6-PNG, administered i.pl. or i.p., suppressed the neuropathic allodynia induced by partial sciatic nerve ligation or oxaliplatin, an anti-cancer agent, in mice. (2R/S)-6-PNG had little or no effect on open-field behavior, motor performance or cardiovascular function in mice, and on the contractility of isolated rat aorta. (2R/S)-6-PNG, but not SG, was detectable in the brain after their i.p. administration in mice. Our data suggest that 6-PNG, a hop component, blocks T-channels, and alleviates neuropathic and visceral pain with little side effects.
Collapse
Affiliation(s)
- Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Tomoyo Fujita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Takahiro Deguchi
- Division of Natural Drug Resources, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Sakura Yamaoka
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Ken Tomochika
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Sumire Ono
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Yamato Horaguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Maki Ichii
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Mio Ichikawa
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Yumiko Ueno
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Nene Koike
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Tadatoshi Tanino
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| | - Huy Du Nguyen
- Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-8555, Japan
| | - Takuya Okada
- Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-8555, Japan
| | - Hiroyuki Nishikawa
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Shigeru Yoshida
- Department of Life Science, Faculty of Science and Engineering, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Tsuyako Ohkubo
- Division of Basic Medical Sciences and Fundamental Nursing, Faculty of Nursing, Fukuoka Nursing College, Fukuoka, 814-0193, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-8555, Japan; Graduate School of Science and Engineering, University of Toyama, Toyama, 930-8555, Japan
| | - Kazuya Murata
- Division of Natural Drug Resources, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Hideaki Matsuda
- Division of Natural Drug Resources, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
36
|
Upregulation of Ca v3.2 T-type calcium channels in adjacent intact L4 dorsal root ganglion neurons in neuropathic pain rats with L5 spinal nerve ligation. Neurosci Res 2018; 142:30-37. [PMID: 29684385 DOI: 10.1016/j.neures.2018.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023]
Abstract
Besides the injured peripheral dorsal root ganglion (DRG) neurons, the adjacent intact DRG neurons also have important roles in neuropathic pain. Ion channels including Cav3.2 T-type calcium channel in the DRG neurons are important in the development of neuropathic pain. In the present study, we aimed to examine the expression of Cav3.2 T-type calcium channels in the intact DRG neurons in neuropathic pain. A neuropathic pain model of rat with lumbar 5 (L5) spinal nerve ligation (SNL) was established, in which the L4 DRG was separated from the axotomized L5 DRG, and the molecular, morphological and electrophysiological changes of Cav3.2 T-type calcium channels in L4 DRG neurons were investigated. Western blotting showed that total and membrane protein levels of Cav3.2 in L4 DRG neurons increased, and voltage-dependent patch clamp recordings revealed an increased T-type current density with a curve shift to the left in steady-state activation in the acutely isolated L4 DRG neurons in neuropathic pain rats. Immunofluorescent staining further showed that the membrane expression of Cav3.2 increased in CGRP-, IB4-positive small neurons and NF200-positive large ones. In conclusion, the membrane expression and the function of Cav3.2 T-type calcium channels are increased in the intact L4 DRG neurons in neuropathic pain rats with peripheral nerve injury like SNL.
Collapse
|
37
|
Chen W, Chi YN, Kang XJ, Liu QY, Zhang HL, Li ZH, Zhao ZF, Yang Y, Su L, Cai J, Liao FF, Yi M, Wan Y, Liu FY. Accumulation of Ca v3.2 T-type Calcium Channels in the Uninjured Sural Nerve Contributes to Neuropathic Pain in Rats with Spared Nerve Injury. Front Mol Neurosci 2018; 11:24. [PMID: 29472842 PMCID: PMC5809483 DOI: 10.3389/fnmol.2018.00024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/17/2018] [Indexed: 11/13/2022] Open
Abstract
Injuries to peripheral nerve fibers induce neuropathic pain. But the involvement of adjacent uninjured fibers to pain is not fully understood. The present study aims to investigate the possible contribution of Cav3.2 T-type calcium channels in uninjured afferent nerve fibers to neuropathic pain in rats with spared nerve injury (SNI). Aβ-, Aδ- and C-fibers of the uninjured sural nerve were sensitized revealed by in vivo single-unit recording, which were accompanied by accumulation of Cav3.2 T-type calcium channel proteins shown by Western blotting. Application of mibefradil, a T-type calcium channel blocker, to sural nerve receptive fields increased mechanical thresholds of Aβ-, Aδ- and C-fibers, confirming the functional involvement of accumulated channels in the sural nerve in SNI rats. Finally, perineural application of mibefradil or TTA-P2 to the uninjured sural nerve alleviated mechanical allodynia in SNI rats. These results suggest that axonal accumulation of Cav3.2 T-type calcium channels plays an important role in the uninjured sural nerve sensitization and contributes to neuropathic pain.
Collapse
Affiliation(s)
- Wen Chen
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Ye-Nan Chi
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Anesthesiology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xue-Jing Kang
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Qing-Ying Liu
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Hao-Lin Zhang
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Zhi-Hua Li
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zi-Fang Zhao
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Yin Yang
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Li Su
- Neuroscience Research Institute, Peking University, Beijing, China.,Center of Medical and Health Analysis, Peking University, Beijing, China
| | - Jie Cai
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Fei-Fei Liao
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Ming Yi
- Neuroscience Research Institute, Peking University, Beijing, China
| | - You Wan
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
| | - Feng-Yu Liu
- Neuroscience Research Institute, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
38
|
Tsubota M, Okawa Y, Irie Y, Maeda M, Ozaki T, Sekiguchi F, Ishikura H, Kawabata A. Involvement of the cystathionine-γ-lyase/Ca v3.2 pathway in substance P-induced bladder pain in the mouse, a model for nonulcerative bladder pain syndrome. Neuropharmacology 2018; 133:254-263. [PMID: 29407215 DOI: 10.1016/j.neuropharm.2018.01.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 11/29/2017] [Accepted: 01/25/2018] [Indexed: 11/28/2022]
Abstract
Hydrogen sulfide (H2S) formed by cystathionine-γ-lyase (CSE) enhances the activity of Cav3.2 T-type Ca2+ channels, contributing to the bladder pain accompanying hemorrhagic cystitis caused by systemic administration of cyclophosphamide (CPA) in mice. Given clinical and fundamental evidence for the involvement of the substance P/NK1 receptor systems in bladder pain syndrome (BPS)/interstitial cystitis (IC), we created an intravesical substance P-induced bladder pain model in mice and analyzed the possible involvement of the CSE/Cav3.2 pathway. Bladder pain/cystitis was induced by i.p. CPA or intravesical substance P in female mice. Bladder pain was evaluated by counting nociceptive behavior and by detecting referred hyperalgesia in the lower abdomen and hindpaw. The isolated bladder tissue was weighed to estimate bladder swelling and subjected to histological observation and Western blotting. Intravesical substance P caused profound referred hyperalgesia accompanied by little bladder swelling or edema 6-24 h after the administration, in contrast to i.p. CPA-induced nociceptive behavior/referred hyperalgesia with remarkable bladder swelling/edema and urothelial damage. The bladder pain and/or cystitis symptoms caused by substance P or CPA were prevented by the NK1 receptor antagonist. CSE in the bladder was upregulated by substance P or CPA, and the NK1 antagonist prevented the CPA-induced CSE upregulation. A CSE inhibitor, a T-type Ca2+ channel blocker and gene silencing of Cav3.2 abolished the intravesical substance P-induced referred hyperalgesia. The intravesical substance P-induced pain in mice is useful as a model for nonulcerative BPS, and involves the activation of the NK1 receptor/CSE/H2S/Cav3.2 cascade.
Collapse
Affiliation(s)
- Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Yasumasa Okawa
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Yuhei Irie
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan; Division of Emergency and Critical Care Medicine, Fukuoka University, Hospital, Fukuoka 814-0180, Japan
| | - Mariko Maeda
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Tomoka Ozaki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | - Hiroyasu Ishikura
- Division of Emergency and Critical Care Medicine, Fukuoka University, Hospital, Fukuoka 814-0180, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan.
| |
Collapse
|
39
|
TRPV1 Nociceptor Activity Initiates USP5/T-type Channel-Mediated Plasticity. Cell Rep 2017; 17:2901-2912. [PMID: 27974205 DOI: 10.1016/j.celrep.2016.11.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/30/2016] [Accepted: 11/14/2016] [Indexed: 12/23/2022] Open
Abstract
Peripheral nerve injury and tissue inflammation result in upregulation of the deubiquitinase USP5, thus causing a dysregulation of T-type calcium channel activity and increased pain sensitivity. Here, we have explored the role of afferent fiber activity in this process. Conditioning stimulation of optogenetically targeted cutaneous TRPV1 expressing nociceptors, but not that of non-nociceptive fibers, resulted in enhanced expression of USP5 in mouse dorsal root ganglia and spinal dorsal horn, along with decreased withdrawal thresholds for thermal and mechanical stimuli that abated after 24 hr. This sensitization was drastically reduced by an interfering peptide that prevented USP5-Cav3.2 association. Sensitization was relieved by pharmacological block of TRPV1 afferents, but not of myelinated neurons. In spinal cord slice recordings, we could optogenetically trigger an activity-dependent potentiation of presynaptic neurotransmission in the spinal dorsal horn that relied on Cav3.2 channel activity. This neuronal-activity-induced USP5 upregulation may underlie a protective, transient sensitization of the pain pathway.
Collapse
|
40
|
Downregulations of TRPM8 expression and membrane trafficking in dorsal root ganglion mediate the attenuation of cold hyperalgesia in CCI rats induced by GFRα3 knockdown. Brain Res Bull 2017; 135:8-24. [PMID: 28867384 DOI: 10.1016/j.brainresbull.2017.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/07/2017] [Accepted: 08/04/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cold hyperalgesia is an intractable sensory abnormality commonly seen in peripheral neuropathies. Although glial cell line-derived neurotrophic factor family receptor alpha3 (GFRα3) is required for the formation of pathological cold pain has been revealed, potential transduction mechanism is poorly elucidated. We have previously demonstrated the contribution of enhanced activity of transient receptor potential melastatin 8 (TRPM8) to cold hyperalgesia in neuropathic pain using a rat model of chronic constriction injury (CCI) to the sciatic nerve. Recently, the enhancement of TRPM8 activity is attributed to the increased TRPM8 plasma membrane trafficking. In addition, TRPM8 can be sensitized by the activation of GFRα3, leading to increased cold responses in vivo. The aim of this study was to investigate whether GFRα3 could influence cold hyperalgesia of CCI rats via modulating TRPM8 expression and plasma membrane trafficking in dorsal root ganglion (DRG). METHODS Mechanical allodynia, cold and heat hyperalgesia were measured on 1day before CCI and the 1st, 4th, 7th, 10th and 14th day after CCI. TRPM8 total expression and membrane trafficking as well as GFRα3 expression in DRG were detected by immunofluorescence and western blot. Furthermore, GFRα3 small interfering RNA (siRNA) was intrathecally administrated to reduce GFRα3 expression in DRG, and the effects of GFRα3 knockdown on CCI-induced behavioral sensitization as well as TRPM8 total expression and membrane trafficking in both mRNA and protein levels were investigated, and the change in coexpression of TRPM8 with GFRα3 was also evaluated. Then, the effect of GFRα3 activation with artemin on pain behavior of CCI rats pretreated with the selective TRPM8 antagonist RQ-00203078 was observed. RESULTS Here we found that TRPM8 total expression and plasma membrane trafficking as well as GFRα3 expression in DRG were initially increased on the 4th day after CCI, and maintained at the peak level from the 10th to the 14th day, which entirely conformed with the induction and maintenance of behavioral-reflex facilitation following CCI. The coexpression of TRPM8 with GFRα3, which was mainly located in peptidergic C-fibers DRG neurons, was also increased after CCI. Downregulation of GFRα3 protein in DRG attenuated CCI-induced cold hyperalgesia without affecting mechanical allodynia and heat hyperalgesia, and reduced the upregulations of TRPM8 total expression and plasma membrane trafficking as well as coexpression of TRPM8 with GFRα3 induced by CCI. Additionally, the inhibition of TRPM8 abolished the influence of GFRα3 activation on cold hyperalgesia after CCI. CONCLUSION Our results demonstrate that GFRα3 knockdown specially inhibits cold hyperalgesia following CCI via decreasing the expression level and plasma membrane trafficking of TRPM8 in DRG. GFRα3 and its downstream mediator, TRPM8, represent a new analgesia axis which can be further exploited in sensitized cold reflex under the condition of chronic pain.
Collapse
|
41
|
Hayano Y, Takasu K, Koyama Y, Yamada M, Ogawa K, Minami K, Asaki T, Kitada K, Kuwabara S, Yamashita T. Dorsal horn interneuron-derived Netrin-4 contributes to spinal sensitization in chronic pain via Unc5B. J Exp Med 2016; 213:2949-2966. [PMID: 27856613 PMCID: PMC5154943 DOI: 10.1084/jem.20160877] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/01/2016] [Accepted: 10/24/2016] [Indexed: 11/28/2022] Open
Abstract
Hayano et al. show that Netrin-4, which is originally identified as an axon guidance molecule, is capable of enhancing sensitivity to sensory input and can contribute to neuropathic pain. The findings provide evidence for a previously unknown pain-inducing signal from spinal cord interneurons. Because of the incomplete understanding of the molecular mechanisms that underlie chronic pain, the currently available treatments for this type of pain remain inefficient. In this study, we show that Netrin-4, a member of the axon guidance molecule family, was expressed in dorsal horn inner lamina II excitatory interneurons in the rat spinal cord. A similar expression pattern for Netrin-4 was also observed in human spinal cord. Behavioral analysis revealed that tactile and heat hyperalgesia after peripheral nerve injury or inflammation were abolished in Netrin-4–mutant rats. Transient suppression of Netrin-4 or its receptor Unc5B after injury could also prevent allodynia. Conversely, intrathecal administration of Netrin-4 protein to naive rats enhanced excitatory synaptic transmission in the dorsal horn and induced allodynia, suggesting that Netrin-4 is involved in spinal sensitization. Furthermore, the Unc5B receptor and subsequent activation of the tyrosine phosphatase SHP2 mediated Netrin-4–induced pain signaling in the spinal cord. These results identify Netrin-4 as a novel protein regulating spinal sensitization leading to chronic pain. Our findings provide evidence for the function of Netrin in the adult nervous system, as well as a previously unknown function in inducing pain signals from dorsal horn interneurons.
Collapse
Affiliation(s)
- Yasufumi Hayano
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Keiko Takasu
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Yoshihisa Koyama
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Moe Yamada
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Koichi Ogawa
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Kazuhisa Minami
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Toshiyuki Asaki
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Kazuhiro Kitada
- Department of Biological Sciences, Graduate School of Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
42
|
Rose P, Moore PK, Zhu YZ. H 2S biosynthesis and catabolism: new insights from molecular studies. Cell Mol Life Sci 2016; 74:1391-1412. [PMID: 27844098 PMCID: PMC5357297 DOI: 10.1007/s00018-016-2406-8] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/07/2016] [Accepted: 11/01/2016] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide (H2S) has profound biological effects within living organisms and is now increasingly being considered alongside other gaseous signalling molecules, such as nitric oxide (NO) and carbon monoxide (CO). Conventional use of pharmacological and molecular approaches has spawned a rapidly growing research field that has identified H2S as playing a functional role in cell-signalling and post-translational modifications. Recently, a number of laboratories have reported the use of siRNA methodologies and genetic mouse models to mimic the loss of function of genes involved in the biosynthesis and degradation of H2S within tissues. Studies utilising these systems are revealing new insights into the biology of H2S within the cardiovascular system, inflammatory disease, and in cell signalling. In light of this work, the current review will describe recent advances in H2S research made possible by the use of molecular approaches and genetic mouse models with perturbed capacities to generate or detoxify physiological levels of H2S gas within tissues.
Collapse
Affiliation(s)
- Peter Rose
- School of Life Science, University of Lincoln, Brayford Pool, Lincoln, Lincolnshire, LN6 7TS, UK. .,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China.
| | - Philip K Moore
- Department of Pharmacology, National University of Singapore, Lee Kong Chian Wing, UHL #05-02R, 21 Lower Kent Ridge Road, Singapore, 119077, Singapore
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
43
|
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: 0.9] [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
|
44
|
Therapeutic potential of RQ-00311651, a novel T-type Ca2+ channel blocker, in distinct rodent models for neuropathic and visceral pain. Pain 2016; 157:1655-1665. [DOI: 10.1097/j.pain.0000000000000565] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Gui Y, Li A, Qiu B, Chen F, Chen L, Liu D, Chen S, Zhou W, Zhou H. Endogenous CBS–H2S Pathway Contributes to the Development of CCI-Induced Neuropathic Pain. Neurochem Res 2016; 41:1381-9. [DOI: 10.1007/s11064-016-1842-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/11/2016] [Accepted: 01/20/2016] [Indexed: 01/26/2023]
|
46
|
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: 1.8] [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
|
47
|
Zhao S, Liu FF, Wu YM, Jiang YQ, Guo YX, Wang XL. Upregulation of spinal NMDA receptors mediates hydrogen sulfide-induced hyperalgesia. J Neurol Sci 2016; 363:176-81. [PMID: 27000247 DOI: 10.1016/j.jns.2016.02.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 02/04/2016] [Accepted: 02/23/2016] [Indexed: 01/23/2023]
Abstract
Hydrogen sulfide (H2S) is an endogenous neurotransmitter that importantly regulates various physiological and pathological events including pain signal transduction. In this study, we investigated the role of spinal NMDA receptors in the nociception induced by intraplantar injection of NaHS, an H2S donor. Intraplantar injection of NaHS into hindpaw significantly decreased the paw withdrawal threshold (PWT) in contralateral hindpaw. However, intraplantar formalin injection did not produce PWT in contralateral hindpaw. Intrathecal injection of methemoglobin, a H2S scavenger, abolished hyperalgesia induced by NaHS. In addition, NaHS-induced hyperalgesia was partly, but significantly, attenuated by intrathecal injection of hydroxylamine, a cystathionine-β-synthase (CBS) inhibitor. RT-PCR and western blotting analysis revealed that NR2B mRNA and protein levels were increased in the spinal dorsal horn, but not in dorsal root ganglion (DRG) in rats subjected to NaHS intraplantar injection. Collectively, these data suggest that peripheral injection of H2S donor causes hyperalgesia through increase in NR2B expression and production of H2S in the spinal cord.
Collapse
Affiliation(s)
- Shuang Zhao
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, China
| | - Fei-Fei Liu
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, China
| | - Yu-Ming Wu
- Department of Physiology, HeBei Medical University, Shijiazhuang, HeBei 050017, China
| | - Yu-Qing Jiang
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, China
| | - Yue-Xian Guo
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, China
| | - Xiu-Li Wang
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, China.
| |
Collapse
|
48
|
Aoki Y, Tsubota M, Nishimoto Y, Maeda Y, Sekiguchi F, Kawabata A. Selective sensitization of C-fiber nociceptors by hydrogen sulfide. J Pharmacol Sci 2016; 130:38-41. [PMID: 26826903 DOI: 10.1016/j.jphs.2015.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/28/2015] [Indexed: 10/22/2022] Open
Abstract
We examined the effects of intraplantar (i.pl.) administration of NaHS, an H2S donor, known to cause T-type Ca(2+) channel (T-channel)-dependent mechanical hyperalgesia, on responsiveness to electric stimulation with 5, 250 and 2000 Hz sine waves (SW) that selectively excites C, Aδ and Aβ fibers, respectively. NaHS, given i.pl., caused behavioral hypersensitivity to SW stimulation at 5 Hz, but not 250 or 2000 Hz, in rats. NaHS also enhanced phosphorylation of spinal ERK following 5 Hz SW stimulation. Three distinct T-channel blockers abolished the NaHS-induced behavioral hypersensitivity to 5 Hz SW stimulation. Thus, H2S selectively sensitizes C-fiber nociceptors via T-channels.
Collapse
Affiliation(s)
- Yuka Aoki
- Laboratory of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Japan
| | - Yuta Nishimoto
- Laboratory of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Japan
| | - Yumi Maeda
- Laboratory of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology & Pathophysiology, Faculty of Pharmacy, Kindai University, Higashi-Osaka 577-8502, Japan.
| |
Collapse
|
49
|
van den Born JC, Hammes HP, Greffrath W, van Goor H, Hillebrands JL. Gasotransmitters in Vascular Complications of Diabetes. Diabetes 2016; 65:331-45. [PMID: 26798119 DOI: 10.2337/db15-1003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In the past decades three gaseous signaling molecules-so-called gasotransmitters-have been identified: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S). These gasotransmitters are endogenously produced by different enzymes in various cell types and play an important role in physiology and disease. Despite their specific functions, all gasotransmitters share the capacity to reduce oxidative stress, induce angiogenesis, and promote vasorelaxation. In patients with diabetes, a lower bioavailability of the different gasotransmitters is observed when compared with healthy individuals. As yet, it is unknown whether this reduction precedes or results from diabetes. The increased risk for vascular disease in patients with diabetes, in combination with the extensive clinical, financial, and societal burden, calls for action to either prevent or improve the treatment of vascular complications. In this Perspective, we present a concise overview of the current data on the bioavailability of gasotransmitters in diabetes and their potential role in the development and progression of diabetes-associated microvascular (retinopathy, neuropathy, and nephropathy) and macrovascular (cerebrovascular, coronary artery, and peripheral arterial diseases) complications. Gasotransmitters appear to have both inhibitory and stimulatory effects in the course of vascular disease development. This Perspective concludes with a discussion on gasotransmitter-based interventions as a therapeutic option.
Collapse
Affiliation(s)
- Joost C van den Born
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Greffrath
- Department of Neurophysiology, Centre for Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | |
Collapse
|
50
|
|