1
|
Calderon-Rivera A, Gomez K, Rodríguez-Palma EJ, Khanna R. SUMOylation and DeSUMOylation: Tug of War of Pain Signaling. Mol Neurobiol 2025; 62:3305-3321. [PMID: 39276308 DOI: 10.1007/s12035-024-04478-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024]
Abstract
SUMOylation is a post-translational modification that attaches a small ubiquitin-like modifier (SUMO) group to a target protein via SUMO ligases, while deSUMOylation refers to the removal of this SUMO group by sentrin-specific proteases (SENPs). Although the functions of these processes have been well described in the nucleus, the role of SUMOylation and deSUMOylation in regulating ion channels is emerging as a novel area of study. Despite this, their contributions to pain signaling remain less clear. Therefore, this review consolidates the current evidence on the link(s) between SUMOylation, deSUMOylation, and pain, with a specific focus on ion channels expressed in the sensory system. Additionally, we explore the role of SUMOylation in the expression and function of kinases, vesicle proteins, and transcription factors, which result in the modulation of certain ion channels contributing to pain. Altogether, this review aims to highlight the relationship between SUMOylation and deSUMOylation in the modulation of ion channels, ultimately exploring the potential therapeutic role of these processes in chronic pain.
Collapse
Affiliation(s)
- Aida Calderon-Rivera
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA
| | - Kimberly Gomez
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA
| | - Erick J Rodríguez-Palma
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA
| | - Rajesh Khanna
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, Gainesville, FL, 32610, USA.
- Pain and Addiction Therapeutics (PATH) Collaboratory, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
2
|
Baggio DF, Gambeta E, Souza IA, Huang S, Zamponi GW, Chichorro JG. Ca V3.2 T-type calcium channels contribute to CGRP- induced allodynia in a rodent model of experimental migraine. J Headache Pain 2024; 25:219. [PMID: 39695919 DOI: 10.1186/s10194-024-01921-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Migraine is a painful neurological syndrome characterized by attacks of throbbing headache, of moderate to severe intensity, which is associated with photo- and phono- sensitivity as well as nausea and vomiting. It affects about 15% of the world's population being 2-3 times more prevalent in females. The calcitonin gene-related peptide (CGRP) is a key mediator in the pathophysiology of migraine, and a significant advance in the field has been the development of anti-CGRP therapies. The trigeminal ganglion (TG) is thought to be an important site of action for these drugs. Moreover, experimental migraine can be induced by CGRP injection in the TG. The signaling pathway induced by CGRP in the TG is not fully understood, but studies suggest that voltage-gated calcium channels contribute to CGRP effects relevant to migraine. OBJECTIVE We hypothesised that CGRP injection in the TG enhances CaV3.2 T-type calcium channel currents to contribute to the development of periorbital mechanical allodynia. RESULTS A Co-Immunoprecipitation assay in tsA-201 cells revealed that CaV3.2 channels form a complex with RAMP-1, a component of the CGRP receptor. Constitutive CGRPR activity was able to inhibit CaV3.2 channels and induce a depolarizing shift in both activation and inactivation curves. Incubation of TG neurons with CGRP increased T-type current density by ~ 3.6 fold, an effect that was not observed in TG neurons from CaV3.2 knockout mice. Incubation of TG neurons with Z944, a pan T-type channel blocker, resulted in an approximately 80% inhibition of T-type currents. In vivo, this treatment abolished the development of periorbital mechanical allodynia induced by CGRP in male and female mice. Likewise, CaV3.2 knockout mice did not develop periorbital mechanical allodynia after intraganglionic CGRP injection. Finally, we demonstrated that the CGRP effect depends on the activation of its canonical GPCR, followed by protein kinase A activation. CONCLUSION The present study suggests that CGRP modulates CaV3.2 in the TG, an effect possibly mediated by the canonical CGRP receptor and PKA activation. The increase in T-type currents in the TG may represent a contributing factor for the initiation and maintenance of the headache pain during migraine.
Collapse
Affiliation(s)
- Darciane F Baggio
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, PR, Brazil
| | - Eder Gambeta
- Department of Clinical Neuroscience, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ivana A Souza
- Department of Clinical Neuroscience, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sun Huang
- Department of Clinical Neuroscience, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald W Zamponi
- Department of Clinical Neuroscience, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Juliana G Chichorro
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, PR, Brazil.
| |
Collapse
|
3
|
Li X, Hu C, Luo S, Dai F, Li C, Zhou W, Wang J, Chen H, Wang Z, Long T, Jiang L, Tang C. Cav3.2 deletion attenuates nonalcoholic fatty liver disease in mice. Gene 2024; 929:148812. [PMID: 39116959 DOI: 10.1016/j.gene.2024.148812] [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: 03/16/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and represents the main cause of liver cirrhosis and hepatocellular carcinoma. Cav3.2 is a T-type calcium channel that is widely present in tissues throughout the body and plays a vital role in energy and metabolic balance. However, the effects of Cav3.2 on the NFALD remain unclear. Here, we investigated the role of Cav3.2 channel in the development and progression of NAFLD. After 16 weeks on a high-fat diets (HFD), Cav3.2 knockout (Cav3.2 KO) improved hepatic steatosis, liver injury and metabolic syndrome in an NAFLD mouse model. We provided evidence that Cav3.2 KO inhibited HFD-induced hepatic oxidative stress, inflammation and hepatocyte apoptosis. In addition, Cav3.2 KO also attenuated hepatic lipid accumulation, oxidative stress, inflammation and hepatocyte apoptosis in palmitic acid/oleic acid (PAOA)-treated primary hepatocytes. These results suggest that therapeutic approaches targeting Cav3.2 provide effective approaches for treating NAFLD.
Collapse
Affiliation(s)
- Xue Li
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Departmentof Anesthesiology, Anhui Provincial Cancer Hospital. Hefei, Anhui 230031, China
| | - Chengyun Hu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Departmentof Anesthesiology, Anhui Provincial Cancer Hospital. Hefei, Anhui 230031, China
| | - Shanshan Luo
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Departmentof Anesthesiology, Anhui Provincial Cancer Hospital. Hefei, Anhui 230031, China
| | - Feibiao Dai
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Departmentof Anesthesiology, Anhui Provincial Cancer Hospital. Hefei, Anhui 230031, China
| | - Chuanyao Li
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Wanjun Zhou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jiawu Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Departmentof Anesthesiology, Anhui Provincial Cancer Hospital. Hefei, Anhui 230031, China
| | - Hao Chen
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Zhen Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Tengfei Long
- Department of Radiotherapy, Hefei Ion Medical Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230088, China.
| | - Lai Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Chaoliang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China; Departmentof Anesthesiology, Anhui Provincial Cancer Hospital. Hefei, Anhui 230031, China.
| |
Collapse
|
4
|
Antunes FTT, Gandini MA, Gadotti VM, Quintão NLM, Santin JR, Souza IA, David LS, Snutch TP, Hildebrand M, Zamponi GW. Contribution of T-type calcium channel isoforms to cold and mechanical sensitivity in naïve and oxaliplatin-treated mice of both sexes. Br J Pharmacol 2024; 181:5062-5078. [PMID: 39295452 DOI: 10.1111/bph.17337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND AND PURPOSE The chemotherapy agent oxaliplatin can give rise to oxaliplatin-induced peripheral neuropathy (OIPN). Here, we investigated whether T-type calcium channels (Cav3) contribute to OIPN. EXPERIMENTAL APPROACH We chronically treated mice with oxaliplatin and assessed pain responses and changes in expression of Cav3.2 calcium channels. We also tested the effects of T-type channel blockers on cold sensitivity in wild-type and Cav3.2 null mice. KEY RESULTS Oxaliplatin treatment led to mechanical and cold hypersensitivity in male and female mice. Mechanical hypersensitivity persisted in Cav3.2 null mice of both sexes. Intraperitoneal or intrathecal delivery of pan T-type channel inhibitors attenuated mechanical hypersensitivity in wild-type but not Cav3.2 null mice. Remarkably cold hypersensitivity occurred in female but not male Cav3.2 null mice even without oxaliplatin treatment. Unexpectedly, intrathecal, intraplantar or intraperitoneal delivery of T-type channel inhibitors Z944 or TTA-P2 transiently induced cold hypersensitivity in both male and female wild-type mice. Acute knockdown of specific Cav3 isoforms revealed that the depletion of Cav3.1 in males and depletion of either Cav3.1 or Cav3.2 in females triggered cold hypersensitivity. Finally, reducing Cav3.2 expression by disrupting the interactions between Cav3.2 and the deubiquitinase USP5 with the small organic molecule II-2 reversed oxaliplatin-induced mechanical and cold hypersensitivity and importantly did not trigger cold allodynia. CONCLUSION AND IMPLICATIONS Altogether, our data indicate that T-type channels differentially contribute to the regulation of cold and mechanical hypersensitivity, and raise the possibility that T-type channel blockers could promote cold allodynia.
Collapse
Affiliation(s)
- Flavia T T Antunes
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Maria A Gandini
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
- School of Health Sciences, Postgraduate Program in Pharmaceutical Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
| | - Nara Lins Meira Quintão
- School of Health Sciences, Postgraduate Program in Pharmaceutical Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
| | - José Roberto Santin
- School of Health Sciences, Postgraduate Program in Pharmaceutical Sciences, Universidade do Vale do Itajaí (UNIVALI), Itajaí, Brazil
| | - Ivana A Souza
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | | | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | | | - Gerald W Zamponi
- Department of Clinical Neurosciences, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
5
|
Maddox JW, Ordemann GJ, de la Rosa Vázquez JAM, Huang A, Gault C, Wisner SR, Randall K, Futagi D, Salem NA, Mayfield D, Zemelman BV, DeVries S, Hoon M, Lee A. A non-conducting role of the Ca v1.4 Ca 2+ channel drives homeostatic plasticity at the cone photoreceptor synapse. eLife 2024; 13:RP94908. [PMID: 39531384 PMCID: PMC11556788 DOI: 10.7554/elife.94908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
In congenital stationary night blindness, type 2 (CSNB2)-a disorder involving the Cav1.4 (L-type) Ca2+ channel-visual impairment is mild considering that Cav1.4 mediates synaptic release from rod and cone photoreceptors. Here, we addressed this conundrum using a Cav1.4 knockout (KO) mouse and a knock-in (G369i KI) mouse expressing a non-conducting Cav1.4. Surprisingly, Cav3 (T-type) Ca2+ currents were detected in cones of G369i KI mice and Cav1.4 KO mice but not in cones of wild-type mouse, ground squirrels, and macaque retina. Whereas Cav1.4 KO mice are blind, G369i KI mice exhibit normal photopic (i.e. cone-mediated) visual behavior. Cone synapses, which fail to form in Cav1.4 KO mice, are present, albeit enlarged, and with some errors in postsynaptic wiring in G369i KI mice. While Cav1.4 KO mice lack evidence of cone synaptic responses, electrophysiological recordings in G369i KI mice revealed nominal transmission from cones to horizontal cells and bipolar cells. In CSNB2, we propose that Cav3 channels maintain cone synaptic output provided that the nonconducting role of Cav1.4 in cone synaptogenesis remains intact. Our findings reveal an unexpected form of homeostatic plasticity that relies on a non-canonical role of an ion channel.
Collapse
Affiliation(s)
- J Wesley Maddox
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Gregory J Ordemann
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | | | - Angie Huang
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Christof Gault
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Serena R Wisner
- Department of Ophthalmology and Visual Sciences, University of Wisconsin- MadisonMadisonUnited States
- Neuroscience Training Program, University of Wisconsin-MadisonMadisonUnited States
| | - Kate Randall
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Daiki Futagi
- Department of Ophthalmology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Nihal A Salem
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Dayne Mayfield
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Boris V Zemelman
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| | - Steven DeVries
- Department of Ophthalmology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin- MadisonMadisonUnited States
- McPherson Eye Research InstituteMadisonUnited States
| | - Amy Lee
- Department of Neuroscience, University of Texas-AustinAustinUnited States
| |
Collapse
|
6
|
Sokolaj E, Assareh N, Anderson K, Aubrey KR, Vaughan CW. Cannabis constituents for chronic neuropathic pain; reconciling the clinical and animal evidence. J Neurochem 2024; 168:3685-3698. [PMID: 37747128 DOI: 10.1111/jnc.15964] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/26/2023]
Abstract
Chronic neuropathic pain is a debilitating pain syndrome caused by damage to the nervous system that is poorly served by current medications. Given these problems, clinical studies have pursued extracts of the plant Cannabis sativa as alternative treatments for this condition. The vast majority of these studies have examined cannabinoids which contain the psychoactive constituent delta-9-tetrahydrocannabinol (THC). While there have been some positive findings, meta-analyses of this clinical work indicates that this effectiveness is limited and hampered by side-effects. This review focuses on how recent preclinical studies have predicted the clinical limitations of THC-containing cannabis extracts, and importantly, point to how they might be improved. This work highlights the importance of targeting channels and receptors other than cannabinoid CB1 receptors which mediate many of the side-effects of cannabis.
Collapse
Affiliation(s)
- Eddy Sokolaj
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Neda Assareh
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Kristen Anderson
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Karin R Aubrey
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Christopher W Vaughan
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Salib AMN, Crane MJ, Jamieson AM, Lipscombe D. Peripheral Ca V2.2 Channels in the Skin Regulate Prolonged Heat Hypersensitivity during Neuroinflammation. eNeuro 2024; 11:ENEURO.0311-24.2024. [PMID: 39433408 PMCID: PMC11599794 DOI: 10.1523/eneuro.0311-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Neuroinflammation can lead to chronic maladaptive pain affecting millions of people worldwide. Neurotransmitters, cytokines, and ion channels are implicated in neuroimmune cell signaling, but their roles in specific behavioral responses are not fully elucidated. Voltage-gated CaV2.2 channel activity in skin controls rapid and transient heat hypersensitivity induced by intradermal (i.d.) capsaicin via IL-1ɑ cytokine signaling. CaV2.2 channels are not, however, involved in mechanical hypersensitivity that developed in the i.d. capsaicin animal model. Here, we show that CaV2.2 channels are also critical for heat hypersensitivity induced by i.d. complete Freund adjuvant (CFA). i.d. CFA, a model of chronic neuroinflammation, involves ongoing cytokine signaling for days leading to pronounced edema and hypersensitivity to sensory stimuli. Peripheral CaV2.2 channel activity in the skin was required for the full development and week-long time course of heat hypersensitivity induced by i.d. CFA, but paw edema and mechanical hypersensitivity were independent of CaV2.2 channel activity. CFA induced increases in several cytokines in hindpaw fluid including IL-6 which was also dependent on CaV2.2 channel activity. Using IL-6-specific neutralizing antibodies in vivo, we show that IL-6 contributes to heat hypersensitivity and that neutralizing both IL-1ɑ and IL-6 was even more effective at reducing the magnitude and duration of CFA-induced heat hypersensitivity. Our findings demonstrate a functional link between CaV2.2 channel activity and the release of IL-6 in the skin and show that CaV2.2 channels have a privileged role in the induction and maintenance of heat hypersensitivity during chronic forms of neuroinflammation in the skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Departments of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Meredith J Crane
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Amanda M Jamieson
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Diane Lipscombe
- Departments of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
8
|
Dai F, Hu C, Li X, Zhang Z, Wang H, Zhou W, Wang J, Geng Q, Dong Y, Tang C. Cav3.2 channel regulates cerebral ischemia/reperfusion injury: a promising target for intervention. Neural Regen Res 2024; 19:2480-2487. [PMID: 38526284 PMCID: PMC11090426 DOI: 10.4103/1673-5374.390966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/05/2023] [Accepted: 10/25/2023] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00028/figure1/v/2024-03-08T184507Z/r/image-tiff Calcium influx into neurons triggers neuronal death during cerebral ischemia/reperfusion injury. Various calcium channels are involved in cerebral ischemia/reperfusion injury. Cav3.2 channel is a main subtype of T-type calcium channels. T-type calcium channel blockers, such as pimozide and mibefradil, have been shown to prevent cerebral ischemia/reperfusion injury-induced brain injury. However, the role of Cav3.2 channels in cerebral ischemia/reperfusion injury remains unclear. Here, in vitro and in vivo models of cerebral ischemia/reperfusion injury were established using middle cerebral artery occlusion in mice and high glucose hypoxia/reoxygenation exposure in primary hippocampal neurons. The results showed that Cav3.2 expression was significantly upregulated in injured hippocampal tissue and primary hippocampal neurons. We further established a Cav3.2 gene-knockout mouse model of cerebral ischemia/reperfusion injury. Cav3.2 knockout markedly reduced infarct volume and brain water content, and alleviated neurological dysfunction after cerebral ischemia/reperfusion injury. Additionally, Cav3.2 knockout attenuated cerebral ischemia/reperfusion injury-induced oxidative stress, inflammatory response, and neuronal apoptosis. In the hippocampus of Cav3.2-knockout mice, calcineurin overexpression offset the beneficial effect of Cav3.2 knockout after cerebral ischemia/reperfusion injury. These findings suggest that the neuroprotective function of Cav3.2 knockout is mediated by calcineurin/nuclear factor of activated T cells 3 signaling. Findings from this study suggest that Cav3.2 could be a promising target for treatment of cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Feibiao Dai
- Graduate School, Wannan Medical College, Wuhu, Anhui Province, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Chengyun Hu
- Graduate School, Wannan Medical College, Wuhu, Anhui Province, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Xue Li
- Graduate School, Wannan Medical College, Wuhu, Anhui Province, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Zhetao Zhang
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Hongtao Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Wanjun Zhou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Jiawu Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Qingtian Geng
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| | - Yongfei Dong
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Chaoliang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Core Facility Center for Medical Sciences, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, Anhui Province, China
| |
Collapse
|
9
|
Patel KV, Gadotti VM, Garcia-Caballero A, Antunes FTT, Ali MY, Zamponi GW, Derksen DJ. Development of Tetrahydroquinoline-Based Inhibitors for Chronic Pain. ACS Chem Neurosci 2024. [PMID: 39377454 DOI: 10.1021/acschemneuro.4c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024] Open
Abstract
Chronic pain affects a substantial portion of the population, posing a significant health challenge. Current treatments often come with limitations and side effects, necessitating novel therapeutic approaches. Our study focuses on disrupting the Cav3.2-USP5 interaction as a strategy for chronic pain management. Through structure-activity relationship studies of a tetrahydroquinoline (THQ) scaffold, we identified a family of lead molecules that demonstrated potent inhibition of the Cav3.2-USP5 interaction. In vitro pharmacokinetic assessments and in vivo studies support the efficacy and drug-like properties of the lead compounds in mouse models of acute and chronic pain. Dependence on the Cav3.2 channels was validated in Cav3.2 null mice, consistent with the proposed mode of action of these small molecules. These findings provide a novel chronic pain treatment strategy, highlighting the potential of these small molecules for further development.
Collapse
Affiliation(s)
- Ketul V Patel
- Department of Chemistry, University of Calgary, Calgary T2N 1N4, Alberta, Canada
- Zymedyne Therapeutics, Calgary T2N 4G4, Alberta, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Agustin Garcia-Caballero
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
- Zymedyne Therapeutics, Calgary T2N 4G4, Alberta, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Darren J Derksen
- Department of Chemistry, University of Calgary, Calgary T2N 1N4, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| |
Collapse
|
10
|
Wang J, Wang Z, Zhang K, Cui Y, Zhou J, Liu J, Li H, Zhao M, Jiang J. The role of the ubiquitin system in the onset and reversal of neuropathic pain. Biomed Pharmacother 2024; 179:117127. [PMID: 39191026 DOI: 10.1016/j.biopha.2024.117127] [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: 05/21/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 08/29/2024] Open
Abstract
Neuropathic pain (NP) remains one of the world's most difficult problems, and people suffering from NP have their quality of life affected to a great extent and constantly suffer from pain. Sensitization of injurious receptors, ectopic firing of afferent nerves after nerve injury, and coupling between sympathetic and sensory neurons are involved in the onset or development of NP, but the pathogenesis of NP is still not well understood. We found that the ubiquitin system is involved in the pathogenesis of NP and has a crucial role in it. The ubiquitin system can be involved in the onset or reversal of NP by affecting ion channels, cellular signal transduction, glial cells, and the regulation of non-coding RNAs. This provides new ideas for the treatment of NP. The ubiquitin system may be a new effective target for the treatment of NP. A continued, in-depth understanding of the mechanisms of the ubiquitin system involved in NP could further refine the study of analgesic targets and improve pharmacological studies.
Collapse
Affiliation(s)
- Jialin Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhijing Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kexin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanping Cui
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingruo Zhou
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiazhou Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huanyi Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingxia Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingjing Jiang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Dube CJ, Zhang Y, Saha S, Lai M, Gibert MK, Escalante M, Hudson K, Wong D, Marcinkiewicz P, Yener U, Sun Y, Xu E, Sorot A, Mulcahy E, Kefas B, Hanif F, Guessous F, Vernon A, Patel MK, Schiff D, Zong H, Purow B, Holland E, Sonkusare S, Sontheimer H, Abounader R. Microenvironment T-Type calcium channels regulate neuronal and glial processes to promote glioblastoma growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609229. [PMID: 39229003 PMCID: PMC11370607 DOI: 10.1101/2024.08.22.609229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Glioblastoma (GBM) is the most common primary malignant brain tumor. The aim of this study was to elucidate the role of microenvironment and intrinsic T-type calcium channels (Cav3) in regulating tumor growth and progression. Methods We grafted syngeneic GBM cells into Cav3.2 knockout mice to assess the role of microenvironment T-Type calcium channels on GBM tumor growth. We performed single-cell RNA-seq (scRNA-seq) of tumors from WT and Cav3.2 KO mice to elucidate the regulation of tumors by the microenvironment. We used neurons from WT and Cav3.2 KO mice in co-culture with GBM stem cells (GSC) to assess the effects of Cav3.2 on neuron/GSC synaptic connections and tumor cell growth. Results Cav3.2 KO in the microenvironment led to significant reduction of GBM growth and prolongation of animal survival. scRNA-seq showed that microenvironment Cav3.2 regulates neuronal and glial biological processes. Microenvironment Cav3.2 downregulated numerous genes associated with regulating the OPC cell state in GBM tumors such as SOX10 and Olig2. Neuronal Cav3.2 promoted neuron/GSC synaptic connections and GSC growth. Treatment of GSCs with the Cav3 blocker mibefradil downregulated genes associated with neuronal processes. The Cav3 blocker drug mibefradil synergized with temozolomide (TMZ) and radiation to reduce in vivo tumor growth and prolong animal survival. Conclusions Together these data reveal a role for microenvironment Cav3 in promoting GBM tumor progression through regulating neuronal and glial processes particularly associated with the OPC-cell state. Targeting both intrinsic and microenvironment Cav3 with the inhibitor mibefradil significantly enhanced the anti-GBM effects of TMZ and radiation.
Collapse
Affiliation(s)
- Collin J. Dube
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ying Zhang
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Shekhar Saha
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Michelle Lai
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Myron K. Gibert
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Miguel Escalante
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Kadie Hudson
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Doris Wong
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Pawel Marcinkiewicz
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ulas Yener
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yunan Sun
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Esther Xu
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Aditya Sorot
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Elizabeth Mulcahy
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Benjamin Kefas
- Pharmacy, University of Virginia, Charlottesville, VA 22908, USA
| | - Farina Hanif
- Department of Biochemistry, Dow International Medical College, Dow University of Health Sciences, Karachi, 75270, Pakistan
| | - Fadilla Guessous
- Laboratory of Onco-Pathology, Biology and Cancer Environment, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Ashley Vernon
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Manoj K. Patel
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| | - David Schiff
- University of Virginia Department of Neurology, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| | - Hui Zong
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| | - Benjamin Purow
- Laboratory of Onco-Pathology, Biology and Cancer Environment, Faculty of Medicine, Mohammed VI University of Sciences and Health, Casablanca, Morocco
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| | | | - Swapnil Sonkusare
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| | - Roger Abounader
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Department of Neurology, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| |
Collapse
|
12
|
Salib AMN, Crane MJ, Jamieson AM, Lipscombe D. Peripheral Ca V2.2 channels in skin regulate prolonged heat hypersensitivity during neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603149. [PMID: 39071304 PMCID: PMC11275762 DOI: 10.1101/2024.07.13.603149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Neuroinflammation can lead to chronic maladaptive pain affecting millions of people worldwide. Neurotransmitters, cytokines, and ion channels are implicated in neuro-immune cell signaling but their roles in specific behavioral responses are not fully elucidated. Voltage-gated CaV2.2 channel activity in skin controls rapid and transient heat hypersensitivity induced by intradermal capsaicin via IL-1α cytokine signaling. CaV2.2 channels are not, however, involved in mechanical hypersensitivity that developed in the same animal model. Here, we show that CaV2.2 channels are also critical for heat hypersensitivity induced by the intradermal (id) Complete Freund's Adjuvant (CFA) model of chronic neuroinflammation that involves ongoing cytokine signaling for days. Ongoing CFA-induced cytokine signaling cascades in skin lead to pronounced edema, and hypersensitivity to sensory stimuli. Peripheral CaV2.2 channel activity in skin is required for the full development and week-long time course of heat hypersensitivity induced by id CFA. CaV2.2 channels, by contrast, are not involved in paw edema and mechanical hypersensitivity. CFA induced increases in cytokines in hind paws including IL-6 which was dependent on CaV2.2 channel activity. Using IL-6 specific neutralizing antibodies, we show that IL-6 contributes to heat hypersensitivity and, neutralizing both IL-1α and IL-6 was even more effective at reducing the magnitude and duration of CFA-induced heat hypersensitivity. Our findings demonstrate a functional link between CaV2.2 channel activity and the release of IL-6 in skin and show that CaV2.2 channels have a privileged role in the induction and maintenance of heat hypersensitivity during chronic forms of neuroinflammation in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience & the Carney Institute for Brain Science Brown University, Providence, RI 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience & the Carney Institute for Brain Science Brown University, Providence, RI 02912, USA
| |
Collapse
|
13
|
Pozzi E, Terribile G, Cherchi L, Di Girolamo S, Sancini G, Alberti P. Ion Channel and Transporter Involvement in Chemotherapy-Induced Peripheral Neurotoxicity. Int J Mol Sci 2024; 25:6552. [PMID: 38928257 PMCID: PMC11203899 DOI: 10.3390/ijms25126552] [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: 04/21/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The peripheral nervous system can encounter alterations due to exposure to some of the most commonly used anticancer drugs (platinum drugs, taxanes, vinca alkaloids, proteasome inhibitors, thalidomide), the so-called chemotherapy-induced peripheral neurotoxicity (CIPN). CIPN can be long-lasting or even permanent, and it is detrimental for the quality of life of cancer survivors, being associated with persistent disturbances such as sensory loss and neuropathic pain at limb extremities due to a mostly sensory axonal polyneuropathy/neuronopathy. In the state of the art, there is no efficacious preventive/curative treatment for this condition. Among the reasons for this unmet clinical and scientific need, there is an uncomplete knowledge of the pathogenetic mechanisms. Ion channels and transporters are pivotal elements in both the central and peripheral nervous system, and there is a growing body of literature suggesting that they might play a role in CIPN development. In this review, we first describe the biophysical properties of these targets and then report existing data for the involvement of ion channels and transporters in CIPN, thus paving the way for new approaches/druggable targets to cure and/or prevent CIPN.
Collapse
Affiliation(s)
- Eleonora Pozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.P.); (L.C.); (S.D.G.)
| | - Giulia Terribile
- Human Physiology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.T.); (G.S.)
| | - Laura Cherchi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.P.); (L.C.); (S.D.G.)
| | - Sara Di Girolamo
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.P.); (L.C.); (S.D.G.)
| | - Giulio Sancini
- Human Physiology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.T.); (G.S.)
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (E.P.); (L.C.); (S.D.G.)
- Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
14
|
Huang J, Fan X, Jin X, Lyu C, Guo Q, Liu T, Chen J, Davakan A, Lory P, Yan N. Structural basis for human Ca v3.2 inhibition by selective antagonists. Cell Res 2024; 34:440-450. [PMID: 38605177 PMCID: PMC11143251 DOI: 10.1038/s41422-024-00959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
The Cav3.2 subtype of T-type calcium channels has been targeted for developing analgesics and anti-epileptics for its role in pain and epilepsy. Here we present the cryo-EM structures of Cav3.2 alone and in complex with four T-type calcium channel selective antagonists with overall resolutions ranging from 2.8 Å to 3.2 Å. The four compounds display two binding poses. ACT-709478 and TTA-A2 both place their cyclopropylphenyl-containing ends in the central cavity to directly obstruct ion flow, meanwhile extending their polar tails into the IV-I fenestration. TTA-P2 and ML218 project their 3,5-dichlorobenzamide groups into the II-III fenestration and place their hydrophobic tails in the cavity to impede ion permeation. The fenestration-penetrating mode immediately affords an explanation for the state-dependent activities of these antagonists. Structure-guided mutational analysis identifies several key residues that determine the T-type preference of these drugs. The structures also suggest the role of an endogenous lipid in stabilizing drug binding in the central cavity.
Collapse
Affiliation(s)
- Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Xiao Fan
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Laboratory of Neurophysiology and Behavior, The Rockefeller University, New York, NY, USA.
| | - Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Chen Lyu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qinmeng Guo
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tao Liu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiaofeng Chen
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Amaël Davakan
- IGF, Université de Montpellier, CNRS, INSERM, LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Philippe Lory
- IGF, Université de Montpellier, CNRS, INSERM, LabEx 'Ion Channel Science and Therapeutics', Montpellier, France
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
- Institute of Bio-Architecture and Bio-Interactions, Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong, China.
| |
Collapse
|
15
|
Xia GQ, Xu M, Sun C, Zhang ZL, Li XQ. Elevated microRNA-214-3p level ameliorates neuroinflammation after spinal cord ischemia-reperfusion injury by inhibiting Nmb/Cav3.2 pathway. Int Immunopharmacol 2024; 133:112031. [PMID: 38631219 DOI: 10.1016/j.intimp.2024.112031] [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: 01/09/2024] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Neuromedin B (Nmb) plays a pivotal role in the transmission of neuroinflammation, particularly during spinal cord ischemia-reperfusion injury (SCII). However, the detailed molecular mechanisms underlying this process remain elusive. METHODS The SCII model was established by clamping the abdominal aorta of male Sprague-Dawley (SD) rats for 60 min. The protein expression levels of Nmb, Cav3.2, and IL-1β were detected by Western blotting, while miR-214-3p expression was quantified by qRT-PCR. The targeted regulation between miR-214-3p and Nmb was investigated using a dual-luciferase reporter gene assay. The cellular localization of Nmb and Cav3.2 with cell-specific markers was visualized by immunofluorescence staining. The specific roles of miR-214-3p on the Nmb/Cav3.2 interactions in SCII-injured rats were explored by intrathecal injection of Cav3.2-siRNA, PD168368 (a specific NmbR inhibitor) and synthetic miR-214-3p agomir and antagomir in separate experiments. Additionally, hind-limb motor function was evaluated using the modified Tarlov scores. RESULTS Compared to the Sham group, the protein expression levels of Nmb, Cav3.2, and the proinflammatory factor Interleukin(IL)-1β were significantly elevated at 24 h post-SCII. Intrathecal injection of PD168368 and Cav3.2-siRNA significantly suppressed the expression of Cav3.2 and IL-1β compared to the SCII group. The miRDB database and dual-luciferase reporter gene assay identified Nmb as a direct target of miR-214-3p. As expected, in vivo overexpression of miR-214-3p by agomir-214-3p pretreatment significantly inhibited the increases in Nmb, Cav3.2 and IL-1β expression and improved lower limb motor function in SCII-injured rats, while antagomiR-214-3p pretreatment reversed these effects. CONCLUSIONS Nmb protein levels positively correlated with Cav3.2 expression in SCII rats. Upregulating miR-214-3p ameliorated hind-limb motor function and protected against neuroinflammation via inhibiting the aberrant Nmb/Cav3.2 interactions and downstream IL-1β release. These findings provide novel therapeutic targets for clinical prevention and treatment of SCII.
Collapse
Affiliation(s)
- Guo-Qiang Xia
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Miao Xu
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Cong Sun
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Zai-Li Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Xiao-Qian Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| |
Collapse
|
16
|
Salib AMN, Crane MJ, Lee SH, Wainger BJ, Jamieson AM, Lipscombe D. Interleukin-1α links peripheral Ca V2.2 channel activation to rapid adaptive increases in heat sensitivity in skin. Sci Rep 2024; 14:9051. [PMID: 38643253 PMCID: PMC11032389 DOI: 10.1038/s41598-024-59424-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Neurons have the unique capacity to adapt output in response to changes in their environment. Within seconds, sensory nerve endings can become hypersensitive to stimuli in response to potentially damaging events. The underlying behavioral response is well studied, but several of the key signaling molecules that mediate sensory hypersensitivity remain unknown. We previously discovered that peripheral voltage-gated CaV2.2 channels in nerve endings in skin are essential for the rapid, transient increase in sensitivity to heat, but not to mechanical stimuli, that accompanies intradermal capsaicin. Here we report that the cytokine interleukin-1α (IL-1α), an alarmin, is necessary and sufficient to trigger rapid heat and mechanical hypersensitivity in skin. Of 20 cytokines screened, only IL-1α was consistently detected in hind paw interstitial fluid in response to intradermal capsaicin and, similar to behavioral sensitivity to heat, IL-1α levels were also dependent on peripheral CaV2.2 channel activity. Neutralizing IL-1α in skin significantly reduced capsaicin-induced changes in hind paw sensitivity to radiant heat and mechanical stimulation. Intradermal IL-1α enhances behavioral responses to stimuli and, in culture, IL-1α enhances the responsiveness of Trpv1-expressing sensory neurons. Together, our data suggest that IL-1α is the key cytokine that underlies rapid and reversible neuroinflammatory responses in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Sang Hun Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Brian J Wainger
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
17
|
Salib AMN, Crane MJ, Lee SH, Wainger BJ, Jamieson AM, Lipscombe D. Interleukin-1α links peripheral Ca V2.2 channel activation to rapid adaptive increases in heat sensitivity in skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.17.572072. [PMID: 38585803 PMCID: PMC10996502 DOI: 10.1101/2023.12.17.572072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Neurons have the unique capacity to adapt output in response to changes in their environment. Within seconds, sensory nerve endings can become hypersensitive to stimuli in response to potentially damaging events. The underlying behavioral response is well studied, but several of the key signaling molecules that mediate sensory hypersensitivity remain unknown. We previously discovered that peripheral voltage-gated CaV2.2 channels in nerve endings in skin are essential for the rapid, transient increase in sensitivity to heat, but not to mechanical stimuli, that accompanies intradermal capsaicin. Here we report that the cytokine interleukin-1α (IL-1α), an alarmin, is necessary and sufficient to trigger rapid heat and mechanical hypersensitivity in skin. Of 20 cytokines screened, only IL-1α was consistently detected in hind paw interstitial fluid in response to intradermal capsaicin and, similar to behavioral sensitivity to heat, IL-1α levels were also dependent on peripheral CaV2.2 channel activity. Neutralizing IL-1α in skin significantly reduced capsaicin-induced changes in hind paw sensitivity to radiant heat and mechanical stimulation. Intradermal IL-1α enhances behavioral responses to stimuli and, in culture, IL-1α enhances the responsiveness of Trpv1-expressing sensory neurons. Together, our data suggest that IL-1α is the key cytokine that underlies rapid and reversible neuroinflammatory responses in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Sang Hun Lee
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Brian J Wainger
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| |
Collapse
|
18
|
Bispat AS, Cardoso FC, Hasan MM, Dongol Y, Wilcox R, Lewis RJ, Duggan PJ, Tuck KL. Inhibition of N-type calcium channels by phenoxyaniline and sulfonamide analogues. RSC Med Chem 2024; 15:916-936. [PMID: 38516585 PMCID: PMC10953480 DOI: 10.1039/d3md00714f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/26/2024] [Indexed: 03/23/2024] Open
Abstract
Building on previous investigations, structural modifications to the neuronal calcium ion channel blocker MONIRO-1 and related compounds were conducted that included replacement of the amide linker with an aniline and isosteric sulfonamide moiety, and the previously used strategy of substitution of the guanidinium group with less hydrophilic amine functionalities. A comprehensive SAR study revealed a number of phenoxyaniline and sulfonamide compounds that were more potent or had similar potency for the CaV2.2 and CaV3.2 channel compared to MONIRO-1 when evaluated in a FLIPR-based intracellular calcium response assay. Cytotoxicity investigations indicated that the sulfonamide analogues were well tolerated by Cos-7 cells at dosages required to inhibit both calcium ion channels. The sulfonamide derivatives were the most promising CaV2.2 inhibitors developed by us to date due, possessing high stability in plasma, low toxicity (estimated therapeutic index > 10), favourable CNS MPO scores (4.0-4.4) and high potency and selectivity, thereby, making this class of compounds suitable candidates for future in vivo studies.
Collapse
Affiliation(s)
- Anjie S Bispat
- School of Chemistry, Monash University Victoria 3800 Australia
- CSIRO Manufacturing, Research Way Clayton Victoria 3168 Australia
| | - Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland St Lucia QLD 4072 Australia
| | - Md Mahadhi Hasan
- Institute for Molecular Bioscience, The University of Queensland St Lucia QLD 4072 Australia
| | - Yashad Dongol
- Institute for Molecular Bioscience, The University of Queensland St Lucia QLD 4072 Australia
| | - Ricki Wilcox
- School of Chemistry, Monash University Victoria 3800 Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland St Lucia QLD 4072 Australia
| | - Peter J Duggan
- CSIRO Manufacturing, Research Way Clayton Victoria 3168 Australia
- College of Science and Engineering, Flinders University Adelaide South Australia 5042 Australia
| | - Kellie L Tuck
- School of Chemistry, Monash University Victoria 3800 Australia
| |
Collapse
|
19
|
Weiss N, Zamponi GW. The T-type calcium channelosome. Pflugers Arch 2024; 476:163-177. [PMID: 38036777 DOI: 10.1007/s00424-023-02891-z] [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: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
T-type calcium channels perform crucial physiological roles across a wide spectrum of tissues, spanning both neuronal and non-neuronal system. For instance, they serve as pivotal regulators of neuronal excitability, contribute to cardiac pacemaking, and mediate the secretion of hormones. These functions significantly hinge upon the intricate interplay of T-type channels with interacting proteins that modulate their expression and function at the plasma membrane. In this review, we offer a panoramic exploration of the current knowledge surrounding these T-type channel interactors, and spotlight certain aspects of their potential for drug-based therapeutic intervention.
Collapse
Affiliation(s)
- Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
20
|
Raut NG, Maile LA, Oswalt LM, Mitxelena I, Adlakha A, Sprague KL, Rupert AR, Bokros L, Hofmann MC, Patritti-Cram J, Rizvi TA, Queme LF, Choi K, Ratner N, Jankowski MP. Schwann cells modulate nociception in neurofibromatosis 1. JCI Insight 2024; 9:e171275. [PMID: 38258905 PMCID: PMC10906222 DOI: 10.1172/jci.insight.171275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 11/28/2023] [Indexed: 01/24/2024] Open
Abstract
Pain of unknown etiology is frequent in individuals with the tumor predisposition syndrome neurofibromatosis 1 (NF1), even when tumors are absent. Nerve Schwann cells (SCs) were recently shown to play roles in nociceptive processing, and we find that chemogenetic activation of SCs is sufficient to induce afferent and behavioral mechanical hypersensitivity in wild-type mice. In mouse models, animals showed afferent and behavioral hypersensitivity when SCs, but not neurons, lacked Nf1. Importantly, hypersensitivity corresponded with SC-specific upregulation of mRNA encoding glial cell line-derived neurotrophic factor (GDNF), independently of the presence of tumors. Neuropathic pain-like behaviors in the NF1 mice were inhibited by either chemogenetic silencing of SC calcium or by systemic delivery of GDNF-targeting antibodies. Together, these findings suggest that alterations in SCs directly modulate mechanical pain and suggest cell-specific treatment strategies to ameliorate pain in individuals with NF1.
Collapse
Affiliation(s)
- Namrata G.R. Raut
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Laura A. Maile
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Leila M. Oswalt
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Irati Mitxelena
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Aaditya Adlakha
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kourtney L. Sprague
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ashley R. Rupert
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lane Bokros
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Megan C. Hofmann
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jennifer Patritti-Cram
- Graduate Program in Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Cancer Biology and Experimental Hematology and
| | - Tilat A. Rizvi
- Division of Cancer Biology and Experimental Hematology and
| | - Luis F. Queme
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Pediatric Pain Research Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kwangmin Choi
- Division of Cancer Biology and Experimental Hematology and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nancy Ratner
- Division of Cancer Biology and Experimental Hematology and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michael P. Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Pediatric Pain Research Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
21
|
Willemen HLDM, Santos Ribeiro PS, Broeks M, Meijer N, Versteeg S, Tiggeler A, de Boer TP, Małecki JM, Falnes PØ, Jans J, Eijkelkamp N. Inflammation-induced mitochondrial and metabolic disturbances in sensory neurons control the switch from acute to chronic pain. Cell Rep Med 2023; 4:101265. [PMID: 37944527 PMCID: PMC10694662 DOI: 10.1016/j.xcrm.2023.101265] [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: 11/16/2022] [Revised: 07/24/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
Pain often persists in patients with an inflammatory disease, even when inflammation has subsided. The molecular mechanisms leading to this failure in pain resolution and the transition to chronic pain are poorly understood. Mitochondrial dysfunction in sensory neurons links to chronic pain, but its role in resolution of inflammatory pain is unclear. Transient inflammation causes neuronal plasticity, called hyperalgesic priming, which impairs resolution of pain induced by a subsequent inflammatory stimulus. We identify that hyperalgesic priming in mice increases the expression of a mitochondrial protein (ATPSc-KMT) and causes mitochondrial and metabolic disturbances in sensory neurons. Inhibition of mitochondrial respiration, knockdown of ATPSCKMT expression, or supplementation of the affected metabolite is sufficient to restore resolution of inflammatory pain and prevents chronic pain development. Thus, inflammation-induced mitochondrial-dependent disturbances in sensory neurons predispose to a failure in resolution of inflammatory pain and development of chronic pain.
Collapse
Affiliation(s)
- Hanneke L D M Willemen
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Patrícia Silva Santos Ribeiro
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Melissa Broeks
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Nils Meijer
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Sabine Versteeg
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Annefien Tiggeler
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 Utrecht, the Netherlands
| | - Jędrzej M Małecki
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Pål Ø Falnes
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway; CRES-O - Centre for Embryology and Healthy Development, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Judith Jans
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands
| | - Niels Eijkelkamp
- Center for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht University, 3508 Utrecht, the Netherlands.
| |
Collapse
|
22
|
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
|
23
|
Ślęczkowska M, Misra K, Santoro S, Gerrits MM, Hoeijmakers JGJ. Ion Channel Genes in Painful Neuropathies. Biomedicines 2023; 11:2680. [PMID: 37893054 PMCID: PMC10604193 DOI: 10.3390/biomedicines11102680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Neuropathic pain (NP) is a typical symptom of peripheral nerve disorders, including painful neuropathy. The biological mechanisms that control ion channels are important for many cell activities and are also therapeutic targets. Disruption of the cellular mechanisms that govern ion channel activity can contribute to pain pathophysiology. The voltage-gated sodium channel (VGSC) is the most researched ion channel in terms of NP; however, VGSC impairment is detected in only <20% of painful neuropathy patients. Here, we discuss the potential role of the other peripheral ion channels involved in sensory signaling (transient receptor potential cation channels), neuronal excitation regulation (potassium channels), involuntary action potential generation (hyperpolarization-activated cyclic nucleotide-gated channels), thermal pain (anoctamins), pH modulation (acid sensing ion channels), and neurotransmitter release (calcium channels) related to pain and their prospective role as therapeutic targets for painful neuropathy.
Collapse
Affiliation(s)
- Milena Ślęczkowska
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands;
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Kaalindi Misra
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, INSPE, 20132 Milan, Italy; (K.M.); (S.S.)
| | - Silvia Santoro
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, INSPE, 20132 Milan, Italy; (K.M.); (S.S.)
| | - Monique M. Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands;
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
24
|
Liu H, Lauzadis J, Gunaratna K, Sipple E, Kaczocha M, Puopolo M. Inhibition of T-Type Calcium Channels With TTA-P2 Reduces Chronic Neuropathic Pain Following Spinal Cord Injury in Rats. THE JOURNAL OF PAIN 2023; 24:1681-1695. [PMID: 37169156 DOI: 10.1016/j.jpain.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Spinal cord injury (SCI)-induced neuropathic pain (SCI-NP) develops in up to 60 to 70% of people affected by traumatic SCI, leading to a major decline in quality of life and increased risk for depression, anxiety, and addiction. Gabapentin and pregabalin, together with antidepressant drugs, are commonly prescribed to treat SCI-NP, but their efficacy is unsatisfactory. The limited efficacy of current pharmacological treatments for SCI-NP likely reflects our limited knowledge of the underlying mechanism(s) responsible for driving the maintenance of SCI-NP. The leading hypothesis in the field supports a major role for spontaneously active injured nociceptors in driving the maintenance of SCI-NP. Recent data from our laboratory provided additional support for this hypothesis and identified the T-type calcium channels as key players in driving the spontaneous activity of SCI-nociceptors, thus providing a rational pharmacological target to treat SCI-NP. To test whether T-type calcium channels contribute to the maintenance of SCI-NP, male and female SCI and sham rats were treated with TTA-P2 (a blocker of T-type calcium channels) to determine its effects on mechanical hypersensitivity (as measured with the von Frey filaments) and spontaneous ongoing pain (as measured with the conditioned place preference paradigm), and compared them to the effects of gabapentin, a blocker of high voltage-activated calcium channels. We found that both TTA-P2 and gabapentin reduced mechanical hypersensitivity in male and females SCI rats, but surprisingly only TTA-P2 reduced spontaneous ongoing pain in male SCI rats. PERSPECTIVES: SCI-induced neuropathic pain, and in particular the spontaneous ongoing pain component, is notoriously very difficult to treat. Our data provide evidence that inhibition of T-type calcium channels reduces spontaneous ongoing pain in SCI rats, supporting a clinically relevant role for T-type channels in the maintenance of SCI-induced neuropathic pain.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Kavindu Gunaratna
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Erin Sipple
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York.
| |
Collapse
|
25
|
Ivasiuk A, Matvieienko M, Kononenko NI, Duzhyy DE, Korogod SM, Voitenko N, Belan P. Diabetes-Induced Amplification of Nociceptive DRG Neuron Output by Upregulation of Somatic T-Type Ca 2+ Channels. Biomolecules 2023; 13:1320. [PMID: 37759720 PMCID: PMC10526307 DOI: 10.3390/biom13091320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
The development of pain symptoms in peripheral diabetic neuropathy (PDN) is associated with the upregulation of T-type Ca2+ channels (T-channels) in the soma of nociceptive DRG neurons. Moreover, a block of these channels in DRG neurons effectively reversed mechanical and thermal hyperalgesia in animal diabetic models, indicating that T-channel functioning in these neurons is causally linked to PDN. However, no particular mechanisms relating the upregulation of T-channels in the soma of nociceptive DRG neurons to the pathological pain processing in PDN have been suggested. Here we have electrophysiologically identified voltage-gated currents expressed in nociceptive DRG neurons and developed a computation model of the neurons, including peripheral and central axons. Simulations showed substantially stronger sensitivity of neuronal excitability to diabetes-induced T-channel upregulation at the normal body temperature compared to the ambient one. We also found that upregulation of somatic T-channels, observed in these neurons under diabetic conditions, amplifies a single action potential invading the soma from the periphery into a burst of multiple action potentials further propagated to the end of the central axon. We have concluded that the somatic T-channel-dependent amplification of the peripheral nociceptive input to the spinal cord demonstrated in this work may underlie abnormal nociception at different stages of diabetes development.
Collapse
Affiliation(s)
- Arsentii Ivasiuk
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Maksym Matvieienko
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Nikolai I. Kononenko
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Dmytro E. Duzhyy
- Department of Sensory Signaling, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine;
| | - Sergiy M. Korogod
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
| | - Nana Voitenko
- Department of Biomedicine and Neuroscience, Kyiv Academic University of NAS of Ukraine, 03142 Kyiv, Ukraine
- Research Center, Dobrobut Academy Medical School, 03022 Kyiv, Ukraine
| | - Pavel Belan
- Department of Molecular Biophysics, Bogomoletz Institute of Physiology of NAS of Ukraine, 01024 Kyiv, Ukraine; (A.I.); (M.M.); (N.I.K.); (S.M.K.)
- Department of Biomedicine and Neuroscience, Kyiv Academic University of NAS of Ukraine, 03142 Kyiv, Ukraine
| |
Collapse
|
26
|
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
|
27
|
He BH, Diatchenko L, Ingelmo P. Genetic risk factors for chronic postsurgical pain in children: A narrative review. Eur J Anaesthesiol 2023; 40:472-483. [PMID: 37199409 DOI: 10.1097/eja.0000000000001847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Genetic risk factors for chronic postsurgical pain in adults have been established, but little is known whether the same associations exist in children. It is even less clear how much influence single nucleotide polymorphisms can exert on the phenotypic expression of chronic postsurgical pain in children in general. To this effect, a search was made for original articles which met the following criteria: evaluation of postsurgical pain in children with known genetic mutations or, conversely, evaluation of atypical pain trajectories of postsurgical children assessing for possible genetic mutations that may explain the phenotype. All titles and abstracts retrieved were reviewed for suitability for inclusion. The references of the selected articles were also checked for additional relevant papers. To assess the transparency and quality of the genetic studies both STrengthening the REporting of Genetic Association studies scores and Q-Genie scores were applied. Overall, there is a paucity of information regarding the link between genetic mutations and eventual chronic postsurgical pain development although there is some information on acute postoperative pain. Evidence has shown that the contribution of genetic risk factors to chronic postsurgical pain development appears to be minor, with its clinical relevance yet to be described. More advanced techniques in systems biology (proteomics, transcriptomics) suggest promising avenues for investigating the disease.
Collapse
Affiliation(s)
- Billy Haitian He
- From the Department of Anesthesia, McGill University, Montréal, Québec, Canada (BHH), Department of Anesthesia and Faculty of Dentistry, McGill University, Montréal, Québec, Canada (LD), Alan Edwards Centre for Research on Pain, McGill University, Montréal, Québec, Canada (LD, PI), Edwards Family Interdisciplinary Center for Complex Pain. Montreal Children's Hospital. McGill University Health Center. Montréal, Québec, Canada (PI), Research Institute, McGill University Health Center. Montréal, Québec, Canada (PI)
| | | | | |
Collapse
|
28
|
Xu YM, Wijeratne EMK, Calderon-Rivera A, Loya-López S, Perez-Miller S, Khanna R, Gunatilaka AAL. Argentatin C Analogues with Potential Antinociceptive Activity and Other Triterpenoid Constituents from the Aerial Parts of Parthenium incanum. ACS OMEGA 2023; 8:20085-20095. [PMID: 37305315 PMCID: PMC10249386 DOI: 10.1021/acsomega.3c02302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
Four new triterpenes, 25-dehydroxy-25-methoxyargentatin C (1), 20S-hydroxyargentatin C (2), 20S-hydroxyisoargentatin C (3), and 24-epi-argentatin C (4), together with 10 known triterpenes (5-14) were isolated from the aerial parts of Parthenium incanum. The structures of 1-4 were elucidated by detailed analysis of their spectroscopic data, and the known compounds 5-14 were identified by comparison of their spectroscopic data with those reported. Since argentatin C (11) was found to exhibit antinociceptive activity by decreasing the excitability of rat and macaque dorsal root ganglia (DRG) neurons, 11 and its new analogues 1-4 were evaluated for their ability to decrease the excitability of rat DRG neurons. Of the argentatin C analogues tested, 25-dehydroxy-25-methoxyargentatin C (1) and 24-epi-argentatin C (4) decreased neuronal excitability in a manner comparable to 11. Preliminary structure-activity relationships for the action potential-reducing effects of argentatin C (11) and its analogues 1-4, and their predicted binding sites in pain-relevant voltage-gated sodium and calcium channels (VGSCs and VGCCs) in DRG neurons are presented.
Collapse
Affiliation(s)
- Ya-ming Xu
- Southwest
Center for Natural Products Research, School of Natural Resources
and the Environment, College of Agriculture and Life Sciences, University of Arizona, 1064 E. Lowell St., Tucson, Arizona 85719, United States
| | - E. M. Kithsiri Wijeratne
- Southwest
Center for Natural Products Research, School of Natural Resources
and the Environment, College of Agriculture and Life Sciences, University of Arizona, 1064 E. Lowell St., Tucson, Arizona 85719, United States
| | - Aida Calderon-Rivera
- NYU
Pain Research Center and Department of Molecular Pathobiology, College
of Dentistry, New York University, 433 First Avenue, New York, New York 10010, United States
| | - Santiago Loya-López
- NYU
Pain Research Center and Department of Molecular Pathobiology, College
of Dentistry, New York University, 433 First Avenue, New York, New York 10010, United States
| | - Samantha Perez-Miller
- NYU
Pain Research Center and Department of Molecular Pathobiology, College
of Dentistry, New York University, 433 First Avenue, New York, New York 10010, United States
| | - Rajesh Khanna
- NYU
Pain Research Center and Department of Molecular Pathobiology, College
of Dentistry, New York University, 433 First Avenue, New York, New York 10010, United States
- Department
of Neuroscience and Physiology and Neuroscience Institute, School
of Medicine, New York University, New York, New York 10010, United States
| | - A. A. Leslie Gunatilaka
- Southwest
Center for Natural Products Research, School of Natural Resources
and the Environment, College of Agriculture and Life Sciences, University of Arizona, 1064 E. Lowell St., Tucson, Arizona 85719, United States
| |
Collapse
|
29
|
Munikishore R, Liu R, Zhang S, Zhao QS, Nian Y, Zuo Z. Structurally modified Cyclovirobuxine-D Buxus alkaloids as effective analgesic agents through Ca v3.2 T-Type calcium channel inhibition. Bioorg Chem 2023; 135:106493. [PMID: 36996509 DOI: 10.1016/j.bioorg.2023.106493] [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: 01/14/2023] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Cyclovirobuxine-D (CVB-D) is a Buxus alkaloid and a major active constituent in the Chinese medicinal herb Buxus microphylls. Traditionally, the natural alkaloid cyclovirobuxine-D has a long history of use as a traditional Chinese medicine for cardiovascular diseases as well as to treat a wide variety of medical conditions. As we found that CVB-D inhibited T-type calcium channels, we designed and synthesized a variety of fragments and analogues and evaluated them for the first time as new Cav3.2 inhibitors. Compounds 2-7 exhibited potency against Cav 3.2 channels, and two of them were more active than their parent molecules. As a result of the in vivo experiments, both compounds 3 and 4 showed significantly reduced writhes in the acetic acid-induced writhing test. Studies of molecular modeling have identified possible mechanism(s) of Cav3.2 binding. Moreover, the relationship between structure and activity was studied in a preliminary manner. Our results indicated that compounds 3 and 4 could play an important role in the discovery and development of novel analgesics.
Collapse
Affiliation(s)
- Rachakunta Munikishore
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan Province, People's Republic of China; Sadashiva Life Sciences, Research and Development Division, Navodaya Industrial Park, IDA Cherlapally, Hyderabad 500051, India
| | - Rui Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan Province, People's Republic of China
| | - Shuqun Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan Province, People's Republic of China
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan Province, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China.
| | - Yin Nian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan Province, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China.
| | - Zhili Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan Province, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China.
| |
Collapse
|
30
|
Zhou M, Yang M, Wen H, Xu S, Han C, Wu Y. O1-conotoxin Tx6.7 cloned from the genomic DNA of Conus textile that inhibits calcium currents. J Venom Anim Toxins Incl Trop Dis 2023; 29:e20220085. [PMID: 37283723 PMCID: PMC10241523 DOI: 10.1590/1678-9199-jvatitd-2022-0085] [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: 12/08/2022] [Accepted: 04/13/2023] [Indexed: 06/08/2023] Open
Abstract
Background Conotoxins exhibit great potential as neuropharmacology tools and therapeutic candidates due to their high affinity and specificity for ion channels, neurotransmitter receptors or transporters. The traditional methods to discover new conotoxins are peptide purification from the crude venom or gene amplification from the venom duct. Methods In this study, a novel O1 superfamily conotoxin Tx6.7 was directly cloned from the genomic DNA of Conus textile using primers corresponding to the conserved intronic sequence and 3' UTR elements. The mature peptide of Tx6.7 (DCHERWDWCPASLLGVIYCCEGLICFIAFCI) was synthesized by solid-phase chemical synthesis and confirmed by mass spectrometry. Results Patch clamp experiments on rat DRG neurons showed that Tx6.7 inhibited peak calcium currents by 59.29 ± 2.34% and peak potassium currents by 22.33 ± 7.81%. In addition, patch clamp on the ion channel subtypes showed that 10 μM Tx6.7 inhibited 56.61 ± 3.20% of the hCaV1.2 currents, 24.67 ± 0.91% of the hCaV2.2 currents and 7.30 ± 3.38% of the hNaV1.8 currents. Tx6.7 had no significant toxicity to ND7/23 cells and increased the pain threshold from 0.5 to 4 hours in the mouse hot plate assay. Conclusion Our results suggested that direct cloning of conotoxin sequences from the genomic DNA of cone snails would be an alternative approach to obtaining novel conotoxins. Tx6.7 could be used as a probe tool for ion channel research or a therapeutic candidate for novel drug development.
Collapse
Affiliation(s)
- Maojun Zhou
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics,
State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, NHC Key
Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University,
Changsha, Hunan, China
| | - Huiling Wen
- School of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi,
China
| | - Shun Xu
- Guangdong Provincial Key Laboratory of Medical Molecular
Diagnostics, Guangdong Medical University, Dongguan, China
| | - Cuifang Han
- Guangdong Provincial Key Laboratory of Medical Molecular
Diagnostics, Guangdong Medical University, Dongguan, China
| | - Yun Wu
- Guangdong Provincial Key Laboratory of Medical Molecular
Diagnostics, Guangdong Medical University, Dongguan, China
| |
Collapse
|
31
|
Chiliquinga AJ, Acosta B, Ogonaga-Borja I, Villarruel-Melquiades F, de la Garza J, Gariglio P, Ocádiz-Delgado R, Ramírez A, Sánchez-Pérez Y, García-Cuellar CM, Bañuelos C, Camacho J. Ion Channels as Potential Tools for the Diagnosis, Prognosis, and Treatment of HPV-Associated Cancers. Cells 2023; 12:1376. [PMID: 37408210 PMCID: PMC10217072 DOI: 10.3390/cells12101376] [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: 02/15/2023] [Revised: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 07/07/2023] Open
Abstract
The human papilloma virus (HPV) group comprises approximately 200 genetic types that have a special affinity for epithelial tissues and can vary from producing benign symptoms to developing into complicated pathologies, such as cancer. The HPV replicative cycle affects various cellular and molecular processes, including DNA insertions and methylation and relevant pathways related to pRb and p53, as well as ion channel expression or function. Ion channels are responsible for the flow of ions across cell membranes and play very important roles in human physiology, including the regulation of ion homeostasis, electrical excitability, and cell signaling. However, when ion channel function or expression is altered, the channels can trigger a wide range of channelopathies, including cancer. In consequence, the up- or down-regulation of ion channels in cancer makes them attractive molecular markers for the diagnosis, prognosis, and treatment of the disease. Interestingly, the activity or expression of several ion channels is dysregulated in HPV-associated cancers. Here, we review the status of ion channels and their regulation in HPV-associated cancers and discuss the potential molecular mechanisms involved. Understanding the dynamics of ion channels in these cancers should help to improve early diagnosis, prognosis, and treatment in the benefit of HPV-associated cancer patients.
Collapse
Affiliation(s)
| | - Brenda Acosta
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Ingrid Ogonaga-Borja
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Fernanda Villarruel-Melquiades
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Jaime de la Garza
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Rodolfo Ocádiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Ana Ramírez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Calzada Universidad 14418, Tijuana 22390, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Claudia M. García-Cuellar
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), Tlalpan, Ciudad de Mexico CP 14080, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de Mexico CP 07360, Mexico
| |
Collapse
|
32
|
Derre A, Soler N, Billoux V, Benizri S, Vialet B, Rivat C, Barthélémy P, Carroll P, Pattyn A, Venteo S. FXYD2 antisense oligonucleotide provides an efficient approach for long-lasting relief of chronic peripheral pain. JCI Insight 2023; 8:161246. [PMID: 37154155 DOI: 10.1172/jci.insight.161246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 03/22/2023] [Indexed: 05/10/2023] Open
Abstract
Chronic pain, whether of inflammatory or neuropathic origin, affects about 18% of the population of developed countries, and most current treatments are only moderately effective and/or cause serious side effects. Therefore, the development of novel therapeutic approaches still represents a major challenge. The Na,K-ATPase modulator FXYD2 is critically required for the maintenance of neuropathic pain in rodents. Here, we set up a therapeutic protocol based on the use of chemically modified antisense oligonucleotides (ASOs) to inhibit FXYD2 expression and treat chronic pain. We identified an ASO targeting a 20-nucleotide stretch in the FXYD2 mRNA that is evolutionarily conserved between rats and humans and is a potent inhibitor of FXYD2 expression. We used this sequence to synthesize lipid-modified forms of ASO (FXYD2-LASO) to facilitate their entry into dorsal root ganglia neurons. We established that intrathecal or intravenous injections of FXYD2-LASO in rat models of neuropathic or inflammatory pain led to a virtually complete alleviation of their pain symptoms, without causing obvious side effects. Remarkably, by using 2'-O-2-methoxyethyl chemical stabilization of the ASO (FXYD2-LASO-Gapmer), we could significantly prolong the therapeutic action of a single treatment up to 10 days. This study establishes FXYD2-LASO-Gapmer administration as a promising and efficient therapeutic strategy for long-lasting relief of chronic pain conditions in human patients.
Collapse
Affiliation(s)
- Alexandre Derre
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Noelian Soler
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Valentine Billoux
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Sebastien Benizri
- ARNA Laboratory, University of Bordeaux, INSERM U1212, UMR CNRS 5320, Bordeaux, France
| | - Brune Vialet
- ARNA Laboratory, University of Bordeaux, INSERM U1212, UMR CNRS 5320, Bordeaux, France
| | - Cyril Rivat
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Philippe Barthélémy
- ARNA Laboratory, University of Bordeaux, INSERM U1212, UMR CNRS 5320, Bordeaux, France
| | - Patrick Carroll
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Alexandre Pattyn
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Stephanie Venteo
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| |
Collapse
|
33
|
Abd Elmaaboud MA, Awad MM, El-Shaer RAA, Kabel AM. The immunomodulatory effects of ethosuximide and sodium butyrate on experimentally induced fibromyalgia: The interaction between IL-4, synaptophysin, and TGF-β1/NF-κB signaling. Int Immunopharmacol 2023; 118:110061. [PMID: 36989891 DOI: 10.1016/j.intimp.2023.110061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/01/2023] [Accepted: 03/18/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND AND AIMS Fibromyalgia is a widespread chronic pain syndrome associated with several comorbid conditions that affect the quality of patients' life. Its pathogenesis is complex, and the treatment strategies are limited by partial efficacy and potential adverse effects. So, our aim was to investigate the possible ameliorative effects of ethosuximide and sodium butyrate on fibromyalgia and compare their effects to pregabalin. MATERIALS AND METHODS In a mouse model of reserpine induced fibromyalgia, the effect of ethosuximide, sodium butyrate, and pregabalin was investigated. Evaluation of mechanical allodynia, cold hypersensitivity, anxiety, cognitive impairment, and depression was performed. Also, the brain and spinal cord tissue serotonin, dopamine and glutamate in addition to the serum levels of interleukin (IL)-4 and transforming growth factor beta 1 (TGF-β1) were assayed. Moreover, the expression of nuclear factor kappa B (NF-κB) synaptophysin was immunoassayed in the hippocampal tissues. KEY FINDINGS Ethosuximide and sodium butyrate restored the behavioral tests to the normal values except for the antidepressant effect which was evident only with ethosuximide. Both drugs elevated the levels of the anti-inflammatory cytokines IL-4 and TGF-β1, reduced the hippocampal NF-κB, and increased synaptophysin expression with superiority of sodium butyrate. Ethosuximide reduced only spinal cord and brain glutamate while improved brain dopamine while sodium butyrate elevated spinal cord dopamine and serotonin with no effect on glutamate. Also, sodium butyrate elevated brain serotonin and reduced glutamate with no effect on brain dopamine. SIGNIFICANCE Each of sodium butyrate and ethosuximide would serve as a promising therapeutic modality for management of fibromyalgia and its comorbid conditions.
Collapse
Affiliation(s)
| | - Marwa M Awad
- Department of physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rehab A A El-Shaer
- Department of physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ahmed M Kabel
- Department of pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt.
| |
Collapse
|
34
|
Ali MY, Gadotti VM, Huang S, Garcia-Caballero A, Antunes FTT, Jung HA, Choi JS, Zamponi GW. Icariside II, a Prenyl-Flavonol, Alleviates Inflammatory and Neuropathic Pain by Inhibiting T-Type Calcium Channels and USP5-Cav3.2 Interactions. ACS Chem Neurosci 2023; 14:1859-1869. [PMID: 37116219 DOI: 10.1021/acschemneuro.3c00083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Cav3.2 channels play an important role in the afferent nociceptive pathway, which is responsible for both physiological and pathological pain transmission. Cav3.2 channels are upregulated during neuropathic pain or peripheral inflammation in part due to an increased association with the deubiquitinase USP5. In this study, we investigated nine naturally occurring flavonoid derivatives which we tested for their abilities to inhibit transiently expressed Cav3.2 channels and their interactions with USP5. Icariside II (ICA-II), one of the flavonols studied, inhibited the biochemical interactions between USP5 and Cav3.2 and concomitantly and effectively blocked Cav3.2 channels. Molecular docking analysis predicts that ICA-II binds to the cUBP domain and the Cav3.2 interaction region. In addition, ICA-II was predicted to interact with residues in close proximity to the Cav3.2 channel's fenestrations, thus accounting for the observed blocking activity. In mice with inflammatory and neuropathic pain, ICA-II inhibited both phases of the formalin-induced nocifensive responses and abolished thermal hyperalgesia induced by injection of complete Freund's adjuvant (CFA) into the hind paw. Furthermore, ICA-II produced significant and long-lasting thermal anti-hyperalgesia in female mice, whereas Cav3.2 null mice were resistant to the action of ICA-II. Altogether, our data show that ICA-II has analgesic activity via an action on Cav3.2 channels.
Collapse
Affiliation(s)
- Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Sun Huang
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
| | - Agustin Garcia-Caballero
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
| |
Collapse
|
35
|
Calderon-Rivera A, Gomez K, Loya-López S, Wijeratne EK, Stratton H, Tang C, Duran P, Masterson K, Alsbiei O, Gunatilaka AL, Khanna R. Betulinic acid analogs inhibit N- and T-type voltage-gated calcium channels to attenuate nerve-injury associated neuropathic and formalin models of pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 13:100116. [PMID: 36687466 PMCID: PMC9853350 DOI: 10.1016/j.ynpai.2023.100116] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Over the past three decades, there has been a significant growth in the use of natural products, with approximately 80% of individuals using them for some aspect of primary healthcare. Our laboratories have identified and studied natural compounds with analgesic effects from dry land plants or their associated fungus during the past ten years. Here, we isolated and characterized thirteen betulin analogs and fifteen betulinic acid analogs for their capacity to prevent calcium influx brought on by depolarization in sensory neurons. The in vitro inhibition of voltage-gated calcium channels by the top drugs was then assessed using whole cell patch clamp electrophysiology. In vivo experiments, conducted at two sites, evaluated the best compound in acute and tonic, neuropathic, inflammatory, post-operative and visceral models of pain. We found that the betulinic acid analog 8 inhibited calcium influx in rat dorsal root ganglion neurons by inhibiting N- (CaV2.2) and T- (CaV3) type voltage-gated calcium channels. Moreover, intrathecal delivery of analog 8 had analgesic activity in both spared nerve injury model of neuropathic pain and acute and tonic pain induced by formalin. The results presented herein highlight the potential antinociceptive properties of betulinic acid analog 8 and set the stage for the development of novel non-opioid pain therapeutics based on the triterpenoid scaffold of betulinic acid.
Collapse
Affiliation(s)
- Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York University, New York, NY, United States
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York University, New York, NY, United States
| | - Santiago Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York University, New York, NY, United States
| | - E.M. Kithsiri Wijeratne
- Natural Products Center, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, United States
| | - Harrison Stratton
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Cheng Tang
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York University, New York, NY, United States
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York University, New York, NY, United States
| | - Kyleigh Masterson
- NYU Pain Research Center, New York University, New York, NY, United States
| | - Omar Alsbiei
- NYU Pain Research Center, New York University, New York, NY, United States
| | - A.A. Leslie Gunatilaka
- Natural Products Center, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, AZ, United States
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York University, New York, NY, United States
| |
Collapse
|
36
|
Su D, Gong Y, Li S, Yang J, Nian Y. Cyclovirobuxine D, a cardiovascular drug from traditional Chinese medicine, alleviates inflammatory and neuropathic pain mainly via inhibition of voltage-gated Ca v3.2 channels. Front Pharmacol 2022; 13:1081697. [PMID: 36618940 PMCID: PMC9811679 DOI: 10.3389/fphar.2022.1081697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cyclovirobuxine D (CVB-D), the main active constituent of traditional Chinese medicine Buxus microphylla, was developed as a safe and effective cardiovascular drug in China. B. microphylla has also been used to relieve various pain symptoms for centuries. In this study, we examined and uncovered strong and persistent analgesic effects of cyclovirobuxine D against several mouse models of pain, including carrageenan- and CFA-induced inflammatory pain and paclitaxel-mediated neuropathic hypersensitivity. Cyclovirobuxine D shows comparable analgesic effects by intraplantar or intraperitoneal administration. Cyclovirobuxine D potently inhibits voltage-gated Cav2.2 and Cav3.2 channels but has negligible effects on a diverse group of nociceptive ion channels distributed in primary afferent neurons, including Nav1.7, Nav1.8, TRPV1, TPRA1, TRPM8, ASIC3, P2X2 and P2X4. Moreover, inhibition of Cav3.2, rather than Cav2.2, plays a dominant role in attenuating the excitability of isolated dorsal root ganglion neurons and pain relieving effects of cyclovirobuxine D. Our work reveals that a currently in-use cardiovascular drug has strong analgesic effects mainly via blockade of Cav3.2 and provides a compelling rationale and foundation for conducting clinical studies to repurpose cyclovirobuxine D in pain management.
Collapse
Affiliation(s)
- Deyuan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms/Key Laboratory of Bioactive Peptides of Yunnan Province, Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ye Gong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Songyu Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Yin Nian
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
37
|
Calderon-Rivera A, Loya-Lopez S, Gomez K, Khanna R. Plant and fungi derived analgesic natural products targeting voltage-gated sodium and calcium channels. Channels (Austin) 2022; 16:198-215. [PMID: 36017978 PMCID: PMC9423853 DOI: 10.1080/19336950.2022.2103234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Voltage-gated sodium and calcium channels (VGSCs and VGCCs) play an important role in the modulation of physiologically relevant processes in excitable cells that range from action potential generation to neurotransmission. Once their expression and/or function is altered in disease, specific pharmacological approaches become necessary to mitigate the negative consequences of such dysregulation. Several classes of small molecules have been developed with demonstrated effectiveness on VGSCs and VGCCs; however, off-target effects have also been described, limiting their use and spurring efforts to find more specific and safer molecules to target these channels. There are a great number of plants and herbal preparations that have been empirically used for the treatment of diseases in which VGSCs and VGCCs are involved. Some of these natural products have progressed to clinical trials, while others are under investigation for their action mechanisms on signaling pathways, including channels. In this review, we synthesize information from ~30 compounds derived from natural sources like plants and fungi and delineate their effects on VGSCs and VGCCs in human disease, particularly pain. [Figure: see text].
Collapse
Affiliation(s)
- Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA
| | - Santiago Loya-Lopez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA,NYU Pain Research Center, New York University, New York, NY, USA,CONTACT Rajesh Khanna
| |
Collapse
|
38
|
Shin SM, Lauzadis J, Itson-Zoske B, Cai Y, Fan F, Natarajan GK, Kwok WM, Puopolo M, Hogan QH, Yu H. Targeting intrinsically disordered regions facilitates discovery of calcium channels 3.2 inhibitory peptides for adeno-associated virus-mediated peripheral analgesia. Pain 2022; 163:2466-2484. [PMID: 35420557 PMCID: PMC9562599 DOI: 10.1097/j.pain.0000000000002650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Ample data support a prominent role of peripheral T-type calcium channels 3.2 (Ca V 3.2) in generating pain states. Development of primary sensory neuron-specific inhibitors of Ca V 3.2 channels is an opportunity for achieving effective analgesic therapeutics, but success has been elusive. Small peptides, especially those derived from natural proteins as inhibitory peptide aptamers (iPAs), can produce highly effective and selective blockade of specific nociceptive molecular pathways to reduce pain with minimal off-target effects. In this study, we report the engineering of the potent and selective iPAs of Ca V 3.2 from the intrinsically disordered regions (IDRs) of Ca V 3.2 intracellular segments. Using established prediction algorithms, we localized the IDRs in Ca V 3.2 protein and identified several Ca V 3.2iPA candidates that significantly reduced Ca V 3.2 current in HEK293 cells stably expressing human wide-type Ca V 3.2. Two prototype Ca V 3.2iPAs (iPA1 and iPA2) derived from the IDRs of Ca V 3.2 intracellular loops 2 and 3, respectively, were expressed selectively in the primary sensory neurons of dorsal root ganglia in vivo using recombinant adeno-associated virus (AAV), which produced sustained inhibition of calcium current conducted by Ca V 3.2/T-type channels and significantly attenuated both evoked and spontaneous pain behavior in rats with neuropathic pain after tibial nerve injury. Recordings from dissociated sensory neurons showed that AAV-mediated Ca V 3.2iPA expression suppressed neuronal excitability, suggesting that Ca V 3.2iPA treatment attenuated pain by reversal of injury-induced neuronal hypersensitivity. Collectively, our results indicate that Ca V 3.2iPAs are promising analgesic leads that, combined with AAV-mediated delivery in anatomically targeted sensory ganglia, have the potential to be a selective peripheral Ca V 3.2-targeting strategy for clinical treatment of pain.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Fan Fan
- Department of Pharmacology and Toxicology, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Gayathri K. Natarajan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
39
|
Gomez K, Khanna R. Ca V 3.2 calcium channels: new players in facial pain. Pain 2022; 163:e1166-e1168. [PMID: 35442930 PMCID: PMC9568618 DOI: 10.1097/j.pain.0000000000002652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry,
New York University, New York, NY, 10010, USA
- NYU Pain Research Center, 433 First Avenue, 8
Floor, New York University, New York, NY, 10010, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry,
New York University, New York, NY, 10010, USA
- NYU Pain Research Center, 433 First Avenue, 8
Floor, New York University, New York, NY, 10010, USA
| |
Collapse
|
40
|
Pathophysiology of Post-Traumatic Trigeminal Neuropathic Pain. Biomolecules 2022; 12:biom12121753. [PMID: 36551181 PMCID: PMC9775491 DOI: 10.3390/biom12121753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/29/2022] Open
Abstract
Trigeminal nerve injury is one of the causes of chronic orofacial pain. Patients suffering from this condition have a significantly reduced quality of life. The currently available management modalities are associated with limited success. This article reviews some of the common causes and clinical features associated with post-traumatic trigeminal neuropathic pain (PTNP). A cascade of events in the peripheral and central nervous system function is involved in the pathophysiology of pain following nerve injuries. Central and peripheral processes occur in tandem and may often be co-dependent. Due to the complexity of central mechanisms, only peripheral events contributing to the pathophysiology have been reviewed in this article. Future investigations will hopefully help gain insight into trigeminal-specific events in the pathophysiology of the development and maintenance of neuropathic pain secondary to nerve injury and enable the development of new therapeutic modalities.
Collapse
|
41
|
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: 1.3] [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
|
42
|
Duran P, Loya-López S, Ran D, Tang C, Calderon-Rivera A, Gomez K, Stratton HJ, Huang S, Xu YM, Wijeratne EMK, Perez-Miller S, Shan Z, Cai S, Gabrielsen AT, Dorame A, Masterson KA, Alsbiei O, Madura CL, Luo G, Moutal A, Streicher J, Zamponi GW, Gunatilaka AAL, Khanna R. The natural product argentatin C attenuates postoperative pain via inhibition of voltage-gated sodium and T-type voltage-gated calcium channels. Br J Pharmacol 2022; 180:1267-1285. [PMID: 36245395 DOI: 10.1111/bph.15974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE Postoperative pain occurs in as many as 70% of surgeries performed worldwide. Postoperative pain management still relies on opioids despite their negative consequences, resulting in a public health crisis. Therefore, it is important to develop alternative therapies to treat chronic pain. Natural products derived from medicinal plants are potential sources of novel biologically active compounds for development of safe analgesics. In this study, we screened a library of natural products to identify small molecules that target the activity of voltage-gated sodium and calcium channels that have important roles in nociceptive sensory processing. EXPERIMENTAL APPROACH Fractions derived from the Native American medicinal plant, Parthenium incanum, were assessed using depolarization-evoked calcium influx in rat dorsal root ganglion (DRG) neurons. Further separation of these fractions yielded a cycloartane-type triterpene identified as argentatin C, which was additionally evaluated using whole-cell voltage and current-clamp electrophysiology, and behavioural analysis in a mouse model of postsurgical pain. KEY RESULTS Argentatin C blocked the activity of both voltage-gated sodium and low-voltage-activated (LVA) calcium channels in calcium imaging assays. Docking analysis predicted that argentatin C may bind to NaV 1.7-1.9 and CaV 3.1-3.3 channels. Furthermore, argentatin C decreased Na+ and T-type Ca2+ currents as well as excitability in rat and macaque DRG neurons, and reversed mechanical allodynia in a mouse model of postsurgical pain. CONCLUSION AND IMPLICATIONS These results suggest that the dual effect of argentatin C on voltage-gated sodium and calcium channels supports its potential as a novel treatment for painful conditions.
Collapse
Affiliation(s)
- Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Santiago Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Dongzhi Ran
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Cheng Tang
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA.,NYU Pain Research Center, New York, New York, USA.,Department of Biochemistry and Molecular Biology, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Harrison J Stratton
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Sun Huang
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ya-Ming Xu
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - E M Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Samantha Perez-Miller
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Zhiming Shan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Anna T Gabrielsen
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Angie Dorame
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Kyleigh A Masterson
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Omar Alsbiei
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Cynthia L Madura
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Guoqin Luo
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - John Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA.,NYU Pain Research Center, New York, New York, USA
| |
Collapse
|
43
|
Liu XG. Normalization of Neuroinflammation: A New Strategy for Treatment of Persistent Pain and Memory/Emotional Deficits in Chronic Pain. J Inflamm Res 2022; 15:5201-5233. [PMID: 36110505 PMCID: PMC9469940 DOI: 10.2147/jir.s379093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic pain, which affects around 1/3 of the world population and is often comorbid with memory deficit and mood depression, is a leading source of suffering and disability. Studies in past decades have shown that hyperexcitability of primary sensory neurons resulting from abnormal expression of ion channels and central sensitization mediated pathological synaptic plasticity, such as long-term potentiation in spinal dorsal horn, underlie the persistent pain. The memory/emotional deficits are associated with impaired synaptic connectivity in hippocampus. Dysregulation of numerous endogenous proteins including receptors and intracellular signaling molecules is involved in the pathological processes. However, increasing knowledge contributes little to clinical treatment. Emerging evidence has demonstrated that the neuroinflammation, characterized by overproduction of pro-inflammatory cytokines and glial activation, is reliably detected in humans and animals with chronic pain, and is sufficient to induce persistent pain and memory/emotional deficits. The abnormal expression of ion channels and pathological synaptic plasticity in spinal dorsal horn and in hippocampus are resulting from neuroinflammation. The neuroinflammation is initiated and maintained by the interactions of circulating monocytes, glial cells and neurons. Obviously, unlike infectious diseases and cancer, which are caused by pathogens or malignant cells, chronic pain is resulting from alterations of cells and molecules which have numerous physiological functions. Therefore, normalization (counterbalance) but not simple inhibition of the neuroinflammation is the right strategy for treating neuronal disorders. Currently, no such agent is available in clinic. While experimental studies have demonstrated that intracellular Mg2+ deficiency is a common feature of chronic pain in animal models and supplement Mg2+ are capable of normalizing the neuroinflammation, activation of upregulated proteins that promote recovery, such as translocator protein (18k Da) or liver X receptors, has a similar effect. In this article, relevant experimental and clinical evidence is reviewed and discussed.
Collapse
Affiliation(s)
- Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
44
|
T-Type Calcium Channels: A Mixed Blessing. Int J Mol Sci 2022; 23:ijms23179894. [PMID: 36077291 PMCID: PMC9456242 DOI: 10.3390/ijms23179894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
The role of T-type calcium channels is well established in excitable cells, where they preside over action potential generation, automaticity, and firing. They also contribute to intracellular calcium signaling, cell cycle progression, and cell fate; and, in this sense, they emerge as key regulators also in non-excitable cells. In particular, their expression may be considered a prognostic factor in cancer. Almost all cancer cells express T-type calcium channels to the point that it has been considered a pharmacological target; but, as the drugs used to reduce their expression are not completely selective, several complications develop, especially within the heart. T-type calcium channels are also involved in a specific side effect of several anticancer agents, that act on microtubule transport, increase the expression of the channel, and, thus, the excitability of sensory neurons, and make the patient more sensitive to pain. This review puts into context the relevance of T-type calcium channels in cancer and in chemotherapy side effects, considering also the cardiotoxicity induced by new classes of antineoplastic molecules.
Collapse
|
45
|
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]
|
46
|
Gomez K, Tang C, Tan B, Perez-Miller S, Ran D, Loya S, Calderon-Rivera A, Stratton HJ, Duran P, Masterson KA, Gabrielsen AT, Alsbiei O, Dorame A, Serafini M, Moutal A, Wang J, Khanna R. Stereospecific Effects of Benzimidazolonepiperidine Compounds on T-Type Ca 2+ Channels and Pain. ACS Chem Neurosci 2022; 13:2035-2047. [PMID: 35671441 DOI: 10.1021/acschemneuro.2c00256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
T-type calcium channels activate in response to subthreshold membrane depolarizations and represent an important source of Ca2+ influx near the resting membrane potential. These channels regulate neuronal excitability and have been linked to pain. For this reason, T-type calcium channels are suitable molecular targets for the development of new non-opioid analgesics. Our previous work identified an analogue of benzimidazolonepiperidine, 5bk, that preferentially inhibited CaV3.2 channels and reversed mechanical allodynia. In this study, we synthesized and screened a small library of 47 compounds derived from 5bk. We found several compounds that inhibited the Ca2+ influx in DRG neurons of all sizes. After separating the enantiomers of each active compound, we found two compounds, 3-25-R and 3-14-3-S, that potently inhibited the Ca2+ influx. Whole-cell patch clamp recordings from small- to medium-sized DRG neurons revealed that both compounds decreased total Ca2+. Application of 3-14-3-S (but not 3-25-R) blocked transiently expressed CaV3.1-3.3 channels with a similar IC50 value. 3-14-3-S decreased T-type, but not N-type, Ca2+ currents in DRG neurons. Furthermore, intrathecal delivery of 3-14-3-S relieved tonic, neuropathic, and inflammatory pain in preclinical models. 3-14-3-S did not exhibit any activity against G protein-coupled opioid receptors. Preliminary docking studies also suggest that 3-14-3-S can bind to the central pore domain of T-type channels. Together, our chemical characterization and functional and behavioral data identify a novel T-type calcium channel blocker with in vivo efficacy in experimental models of tonic, neuropathic, and inflammatory pain.
Collapse
Affiliation(s)
- Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States
| | - Cheng Tang
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States.,The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Bin Tan
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, William Levine Hall, Room 320, 160 Frelinghuysen Road, Piscataway, New Jersey 0885, United States
| | - Samantha Perez-Miller
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States
| | - Dongzhi Ran
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Santiago Loya
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States
| | - Harrison J Stratton
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States
| | - Kyleigh A Masterson
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Anna T Gabrielsen
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Omar Alsbiei
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Angie Dorame
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Maria Serafini
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Aubin Moutal
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd. Schwitalla Hall, Room 432, Saint Louis, Missouri 63104, United States
| | - Jun Wang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, 433 First Avenue, 8th Floor, New York, New York 10010, United States
| |
Collapse
|
47
|
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: 30] [Impact Index Per Article: 10.0] [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
|
48
|
CaV3.2 calcium channels contribute to trigeminal neuralgia. Pain 2022; 163:2315-2325. [PMID: 35467587 DOI: 10.1097/j.pain.0000000000002651] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/23/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Trigeminal neuralgia (TN) is a rare but debilitating disorder characterized by excruciating facial pain, with a higher incidence in women. Recent studies demonstrated that TN patients present mutations in the gene encoding the CaV3.2 T-type calcium channel, an important player in peripheral pain pathways. Here we characterize the role of CaV3.2 channels in TN at two levels. First, we examined the biophysical properties of CACNA1H variants found in TN patients. Second, we investigated the role of CaV3.2 in an animal model of trigeminal neuropathic pain. Whole cell patch clamp recordings from four different mutants expressed in tsA-201 cells (E286K in the pore loop of domain I, H526Y, G563R and P566T in the domain I-II linker) identified a loss-of-function in activation in the E286K mutation and gain-of-function in the G563R and P566T mutations. Moreover, a loss-of-function in inactivation was observed with the E286K and H526Y mutations. Cell surface biotinylation revealed no difference in channel trafficking among the variants. The G563R mutant also caused a gain-of-function in the firing properties of transfected trigeminal ganglion neurons. In female and male mice, constriction of the infraorbital nerve (CION) induced facial thermal heat hyperalgesia. Block of T-type channels with Z944 resulted in antihyperalgesia. The effect of Z944 was absent in CaV3.2-/- mice, indicating that CaV3.2 is the molecular target of the antihyperalgesic Z944 effect. Finally, ELISA analysis revealed increased CaV3.2 channel expression in the spinal trigeminal subnucleus caudalis. Altogether, the present study demonstrates an important role of CaV3.2 channels in trigeminal pain.
Collapse
|
49
|
Papazoglou A, Arshaad MI, Henseler C, Daubner J, Broich K, Hescheler J, Ehninger D, Haenisch B, Weiergräber M. Ca v3 T-Type Voltage-Gated Ca 2+ Channels and the Amyloidogenic Environment: Pathophysiology and Implications on Pharmacotherapy and Pharmacovigilance. Int J Mol Sci 2022; 23:3457. [PMID: 35408817 PMCID: PMC8998330 DOI: 10.3390/ijms23073457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/07/2022] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) were reported to play a crucial role in neurotransmitter release, dendritic resonance phenomena and integration, and the regulation of gene expression. In the septohippocampal system, high- and low-voltage-activated (HVA, LVA) Ca2+ channels were shown to be involved in theta genesis, learning, and memory processes. In particular, HVA Cav2.3 R-type and LVA Cav3 T-type Ca2+ channels are expressed in the medial septum-diagonal band of Broca (MS-DBB), hippocampal interneurons, and pyramidal cells, and ablation of both channels was proven to severely modulate theta activity. Importantly, Cav3 Ca2+ channels contribute to rebound burst firing in septal interneurons. Consequently, functional impairment of T-type Ca2+ channels, e.g., in null mutant mouse models, caused tonic disinhibition of the septohippocampal pathway and subsequent enhancement of hippocampal theta activity. In addition, impairment of GABA A/B receptor transcription, trafficking, and membrane translocation was observed within the septohippocampal system. Given the recent findings that amyloid precursor protein (APP) forms complexes with GABA B receptors (GBRs), it is hypothesized that T-type Ca2+ current reduction, decrease in GABA receptors, and APP destabilization generate complex functional interdependence that can constitute a sophisticated proamyloidogenic environment, which could be of potential relevance in the etiopathogenesis of Alzheimer's disease (AD). The age-related downregulation of T-type Ca2+ channels in humans goes together with increased Aβ levels that could further inhibit T-type channels and aggravate the proamyloidogenic environment. The mechanistic model presented here sheds new light on recent reports about the potential risks of T-type Ca2+ channel blockers (CCBs) in dementia, as observed upon antiepileptic drug application in the elderly.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Muhammad Imran Arshaad
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (K.B.); (B.H.)
| | - Jürgen Hescheler
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany;
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Dan Ehninger
- Translational Biogerontology, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany;
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Britta Haenisch
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (K.B.); (B.H.)
- German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
- Center for Translational Medicine, Medical Faculty, University of Bonn, 53113 Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (A.P.); (M.I.A.); (C.H.); (J.D.)
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany; (K.B.); (B.H.)
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany;
- Center of Physiology and Pathophysiology, Faculty of Medicine, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| |
Collapse
|
50
|
Elwing JE, Atassi H, Rogers BD, Sayuk GS. Emerging therapies in the management of Irritable Bowel Syndrome (IBS). Expert Opin Emerg Drugs 2022; 27:55-73. [PMID: 35266839 DOI: 10.1080/14728214.2022.2052043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a common, symptom-based disorder of chronic abdominal pain and altered bowel habits. The pathogenesis of IBS is multifactorial, leading to the potential for the development of multiple, diverse treatment strategies. This mechanistic heterogeneity also leads to the realization that available therapies are only effective in a subset of IBS suffers. Current US Food and Drug Administration (FDA) approved therapies for IBS with diarrhea (IBS-D) and IBS with constipation (IBS-C) are reviewed. Limited symptom responses and side effect experiences lead to considerable patient dissatisfaction with currently available IBS treatments. Only a small percentage of IBS patients are on prescription therapies underscoring the potential market and need for additional therapeutic options. AREAS COVERED : Expanding on currently available therapies, the serotonergic and endogenous opioid receptor systems continue to be a focus of future IBS treatment development. Additional novel emerging therapies include the endogenous cannabinoid system, bile acid secretion and sequestration, and exploit our enhanced understanding of visceral sensory signaling and intestinal secretomotor function. EXPERT OPINION While challenges remain for the future development of IBS therapies, the diverse etiologies underlying the disorder present an opportunity for novel therapies. Hence, great potential is anticipated for future IBS treatment options.
Collapse
Affiliation(s)
- Jill E Elwing
- St. Louis Veterans Affairs Medical Center, St. Louis, MO, USA
| | - Hadi Atassi
- Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
| | - Benjamin D Rogers
- Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA.,Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory S Sayuk
- St. Louis Veterans Affairs Medical Center, St. Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|