1
|
Pando MM, Debner EK, Jacobs BA, Jamshidi RJ, Jennings EM, Clarke WP, Berg KA. Activation of G protein gated inwardly rectifying potassium (GIRK) channels in keratinocytes mediates peripheral kappa opioid receptor-mediated antinociception. Neuropharmacology 2025; 268:110326. [PMID: 39880327 DOI: 10.1016/j.neuropharm.2025.110326] [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: 09/20/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 01/31/2025]
Abstract
Kappa opioid receptors (KOR) expressed by peripheral pain-sensing neurons (nociceptors) are a promising target for development of effective and safer analgesics for inflammatory pain that are devoid of central nervous system adverse effects. Here we sought to delineate the signaling pathways that underlie peripheral KOR-mediated antinociception in adult male and female Sprague-Dawley rats. In an inflammatory model of pain, local intraplantar (i.pl.) injection of pertussis toxin prevented antinociception induced by the KOR agonist, U50488, indicating that members of the Gi/o family mediate the antinociceptive response. Furthermore, i.pl. injection of the G protein-coupled inward-rectifying potassium (GIRK) channel blocker, TPNQ, as well as GIRK2 subunit-targeted siRNA abolished U50488-mediated antinociceptive behavioral responses in both male and female rats. Consistent with these data, i.pl. injection of ML297, a direct activator of GIRK1 subunit-containing channels, elicited peripheral antinociceptive behavior. It is well known that intraepidermal nerve fibers (IENF) that innervate the hindpaw propagate nociceptive signals to the spinal cord. However, recent studies suggest that keratinocytes, the major cell type in the epidermis, also play an active role in pain and sensory processing. Results from RT-qPCR, RNAscope and immunohistochemistry experiments confirmed that both KOR and GIRK are expressed in keratinocytes in the epidermal layer of the rat hindpaw. Knockdown of either KOR or GIRK2 subunits selectively in keratinocytes by i.pl. injection of shRNA plasmids, prevented the antinociceptive response to U50488. Taken together, these data suggest that KOR-mediated activation of GIRK channels in keratinocytes is required for peripherally-mediated antinociception.
Collapse
Affiliation(s)
- Miryam M Pando
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Emily K Debner
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Blaine A Jacobs
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Raehannah J Jamshidi
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Elaine M Jennings
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - William P Clarke
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Kelly A Berg
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
2
|
Jayakody T, Budagoda DK, Mendis K, Dilshan WD, Bethmage D, Dissasekara R, Dawe GS. Biased agonism in peptide-GPCRs: A structural perspective. Pharmacol Ther 2025; 269:108806. [PMID: 39889970 DOI: 10.1016/j.pharmthera.2025.108806] [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: 09/17/2024] [Revised: 12/13/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
G protein-coupled receptors (GPCRs) are dynamic membrane receptors that transduce extracellular signals to the cell interior by forming a ligand-receptor-effector (ternary) complex that functions via allosterism. Peptides constitute an important class of ligands that interact with their cognate GPCRs (peptide-GPCRs) to form the ternary complex. "Biased agonism", a therapeutically relevant phenomenon exhibited by GPCRs owing to their allosteric nature, has also been observed in peptide-GPCRs, leading to the development of selective therapeutics with fewer side effects. In this review, we have focused on the structural basis of signalling bias at peptide-GPCRs of classes A and B, and reviewed the therapeutic relevance of bias at peptide-GPCRs, with the hope of contributing to the discovery of novel biased peptide drugs.
Collapse
Affiliation(s)
- Tharindunee Jayakody
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | | | - Krishan Mendis
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | | | - Duvindu Bethmage
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | - Rashmi Dissasekara
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka; The Graduate School, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
3
|
Liu M, Sheng Y, He Y, Wu S, Jin C, Shen L. Progresses in Questing for the Truth of Opioid-Related Constipation in Cancer Patients. J Cell Mol Med 2025; 29:e70553. [PMID: 40281681 PMCID: PMC12031673 DOI: 10.1111/jcmm.70553] [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] [Received: 11/24/2024] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Opioids are extensively utilised to manage pain in cancer patients, but may cause constipation which significantly impacts their prognosis and quality of life. Opioid-induced constipation (OIC) lacks effective drugs and management strategies. Opioids act on the enteric nervous system, intestinal barrier, intestinal immunity and intestinal microbiota, implying that OIC is a multifactorial process. This paper aims to examine the effects of opioids on the intestine, specifically the enteric nervous system, intestinal barrier and interstitial cells of Cajal (ICCs), and elucidate the primary mechanisms underlying OIC development and deterioration. This review suggests that enteric neurons, intestinal immunity and intestinal flora could serve as potential therapeutic targets for OIC.
Collapse
Affiliation(s)
- Mengxue Liu
- Wuxi Hospital Affiliated to Nanjing University of Chinese MedicineWuxiJiangsuChina
| | - Yedong Sheng
- Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yingrong He
- Wuxi Hospital Affiliated to Nanjing University of Chinese MedicineWuxiJiangsuChina
| | - Shixiang Wu
- Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Chunhui Jin
- Wuxi Hospital Affiliated to Nanjing University of Chinese MedicineWuxiJiangsuChina
| | - Lijuan Shen
- Wuxi Hospital Affiliated to Nanjing University of Chinese MedicineWuxiJiangsuChina
| |
Collapse
|
4
|
Soleimanii A, Fallah F, Ghorbanzadeh B, Oroojan AA, Amirgholami N, Alboghobeish S. Simultaneous use of venlafaxine and calcium channel blockers on tolerance to morphine: The role of mitochondrial damage and oxidative stress in the brain. Pharmacol Biochem Behav 2024; 245:173864. [PMID: 39216833 DOI: 10.1016/j.pbb.2024.173864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND One of the reasons for tolerance to morphine is increased oxidative stress and dysfunction of cell mitochondria in the hippocampus. Venlafaxine and calcium channel blockers can protect mitochondrial function. The investigation of the role of mitochondrial damage and oxidative stress in the simultaneous use of venlafaxine and calcium channel blockers on the acute analgesic effects of morphine and the induction of tolerance to its effects in mice was assessed. METHOD In this experimental study, to induce tolerance to morphine, NMRI mice were treated with 50 mg/kg morphine for three consecutive days and 5 mg/kg morphine on the fourth day. Venlafaxine (20 mg/kg) alone or in combination with calcium channel blockers, nimodipine (10 mg/kg), and diltiazem (40 mg/kg) was administered 30 min before morphine, and the hot plate test was used. Then, hippocampal mitochondria were isolated by differential centrifugation method, and the levels of mitochondrial dehydrogenase activity, mitochondrial membrane potential, mitochondrial ROS production rate, as well as the content of glutathione and malondialdehyde in hippocampal mitochondria, were measured. RESULTS The administration of venlafaxine-nimodipine and venlafaxine-diltiazem increased morphine's acute analgesic effects (P < 0.05) and reduced the induction and expression of tolerance to the analgesic effects of morphine (P < 0.05). Morphine significantly decreased MTT and GSH and increased MDA, mitochondrial membrane damage, and ROS compared to the control group (P < 0.01). Injection of venlafaxine-nimodipine and also venlafaxine-diltiazem 30 min before morphine can improve these alterations (P < 0.05). DISCUSSION AND CONCLUSION Our data showed that the simultaneous use of venlafaxine with calcium channel blockers could increase the acute analgesic effects of morphine and reduce the induction and expression of tolerance to it. Also, the preventive and protective roles of simultaneous administration of venlafaxine and calcium channel blockers on morphine-induced mitochondrial oxidative stress and damage during the tolerance test were achieved.
Collapse
Affiliation(s)
- Asma Soleimanii
- School of medicine, Student Research Committee, Dezful University of Medical Sciences, Dezful, Iran
| | - Faezeh Fallah
- School of medicine, Student Research Committee, Dezful University of Medical Sciences, Dezful, Iran
| | - Behnam Ghorbanzadeh
- Department of Pharmacology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Ali Akbar Oroojan
- Department of Physiology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Neda Amirgholami
- Department of Pharmacology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Soheila Alboghobeish
- Department of Pharmacology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran.
| |
Collapse
|
5
|
Ferreira MDA, Lückemeyer DD, Martins F, Schran RG, da Silva AM, Gambeta E, Zamponi GW, Ferreira J. Pronociceptive role of spinal Ca v2.3 (R-type) calcium channels in a mouse model of postoperative pain. Br J Pharmacol 2024; 181:3594-3609. [PMID: 38812100 DOI: 10.1111/bph.16407] [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: 08/07/2023] [Revised: 03/05/2024] [Accepted: 03/28/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND More than 80% of patients may experience acute pain after a surgical procedure, and this is often refractory to pharmacological intervention. The identification of new targets to treat postoperative pain is necessary. There is an association of polymorphisms in the Cav2.3 gene with postoperative pain and opioid consumption. Our study aimed to identify Cav2.3 as a potential target to treat postoperative pain and to reduce opioid-related side effects. EXPERIMENTAL APPROACH A plantar incision model was established in adult male and female C57BL/6 mice. Cav2.3 expression was detected by qPCR and suppressed by siRNA treatment. The antinociceptive efficacy and safety of a Cav2.3 blocker-alone or together with morphine-was also assessed after surgery. KEY RESULTS Paw incision in female and male mice caused acute nociception and increased Cav2.3 mRNA expression in the spinal cord but not in the incised tissue. Intrathecal treatment with siRNA against Cav2.3, but not with a scrambled siRNA, prevented the development of surgery-induced nociception in both male and female mice, with female mice experiencing long-lasting effects. High doses of i.t. SNX-482, a Cav2.3 channel blocker, or morphine injected alone, reversed postoperative nociception but also induced side effects. A combination of lower doses of morphine and SNX-482 mediated a long-lasting reversal of postsurgical pain in female and male mice. CONCLUSION Our results demonstrate that Cav2.3 has a pronociceptive role in the induction of postoperative pain, indicating that it is a potential target for the development of therapeutic approaches for the treatment of postoperative pain.
Collapse
Affiliation(s)
- Marcella de Amorim Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Debora Denardin Lückemeyer
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fernanda Martins
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Roberta Giusti Schran
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Ana Merian da Silva
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Eder Gambeta
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
6
|
Borjas NC, Anstötz M, Maccaferri G. Multiple layers of diversity govern the cell type specificity of GABAergic input received by mouse subicular pyramidal neurons. J Physiol 2024; 602:4195-4213. [PMID: 39141819 PMCID: PMC11665487 DOI: 10.1113/jp286679] [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: 04/02/2024] [Accepted: 07/23/2024] [Indexed: 08/16/2024] Open
Abstract
The subiculum is a key region of the brain involved in the initiation of pathological activity in temporal lobe epilepsy, and local GABAergic inhibition is essential to prevent subicular-originated epileptiform discharges. Subicular pyramidal cells may be easily distinguished into two classes based on their different firing patterns. Here, we have compared the strength of the GABAa receptor-mediated inhibitory postsynaptic currents received by regular- vs. burst-firing subicular neurons and their dynamic modulation by the activation of μ opioid receptors. We have taken advantage of the sequential re-patching of the same cell to initially classify pyramidal neurons according to their firing patters, and then to measure GABAergic events triggered by the optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons. Activation of parvalbumin-expressing cells generated larger responses in postsynaptic burst-firing neurons whereas the opposite was observed for currents evoked by the stimulation of somatostatin-expressing interneurons. In all cases, events depended critically on ω-agatoxin IVA- but not on ω-conotoxin GVIA-sensitive calcium channels. Optogenetic GABAergic input originating from both parvalbumin- and somatostatin-expressing cells was reduced in amplitude following the exposure to a μ opioid receptor agonist. The kinetics of this pharmacological sensitivity was different in regular- vs. burst-firing neurons, but only when responses were evoked by the activation of parvalbumin-expressing neurons, whereas no differences were observed when somatostatin-expressing cells were stimulated. In conclusion, our results show that a high degree of complexity regulates the organizing principles of subicular GABAergic inhibition, with the interaction of pre- and postsynaptic diversity at multiple levels. KEY POINTS: Optogenetic stimulation of parvalbumin- and somatostatin-expressing interneurons (PVs and SOMs) triggers inhibitory postsynaptic currents (IPSCs) in both regular- and burst-firing (RFs and BFs) subicular pyramidal cells. The amplitude of optogenetically evoked IPSCs from PVs (PV-opto IPSCs) is larger in BFs whereas IPSCs generated by the light activation of SOMs (SOM-opto IPSCs) are larger in RFs. Both PV- and SOM-opto IPSCs critically depend on ω-agatoxin IVA-sensitive P/Q type voltage-gated calcium channels, whereas no major effects are observed following exposure to ω-conotoxin GVIA, suggesting no significant involvement of N-type channels. The amplitude of both PV- and SOM-opto IPSCs is reduced by the probable pharmacological activation of presynaptic μ opioid receptors, with a faster kinetics of the effect observed in PV-opto IPSCs from RFs vs. BFs, but not in SOM-opto IPSCs. These results help us understand the complex interactions between different layers of diversity regulating GABAergic input onto subicular microcircuits.
Collapse
Affiliation(s)
- Nancy Castro Borjas
- Department of Neuroscience, Feinberg School of MedicineNorthwestern UniversityChicagoUSA
| | - Max Anstötz
- Institute of Anatomy II, Medical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
| | - Gianmaria Maccaferri
- Department of Neuroscience, Feinberg School of MedicineNorthwestern UniversityChicagoUSA
| |
Collapse
|
7
|
Perez-Miller S, Gomez K, Khanna R. Peptide and Peptidomimetic Inhibitors Targeting the Interaction of Collapsin Response Mediator Protein 2 with the N-Type Calcium Channel for Pain Relief. ACS Pharmacol Transl Sci 2024; 7:1916-1936. [PMID: 39022365 PMCID: PMC11249630 DOI: 10.1021/acsptsci.4c00181] [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: 03/28/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 07/20/2024]
Abstract
Ion channels serve pleiotropic functions. Often found in complexes, their activities and functions are sculpted by auxiliary proteins. We discovered that collapsin response mediator protein 2 (CRMP2) is a binding partner and regulator of the N-type voltage-gated calcium channel (CaV2.2), a genetically validated contributor to chronic pain. Herein, we trace the discovery of a new peptidomimetic modulator of this interaction, starting from the identification and development of CBD3, a CRMP2-derived CaV binding domain peptide. CBD3 uncouples CRMP2-CaV2.2 binding to decrease CaV2.2 surface localization and calcium currents. These changes occur at presynaptic sites of nociceptive neurons and indeed, CBD3 ameliorates chronic pain in preclinical models. In pursuit of a CBD3 peptidomimetic, we exploited a unique approach to identify a dipeptide with low conformational flexibility and high solvent accessibility that anchors binding to CaV2.2. From a pharmacophore screen, we obtained CBD3063, a small-molecule that recapitulated CBD3's activity, reversing nociceptive behaviors in rodents of both sexes without sensory, affective, or cognitive effects. By disrupting the CRMP2-CaV2.2 interaction, CBD3063 exerts these effects indirectly through modulating CaV2.2 trafficking, supporting CRMP2 as an auxiliary subunit of CaV2.2. The parent peptide CBD3 was also found by us and others to have neuroprotective properties at postsynaptic sites, through N-methyl-d-aspartate receptor and plasmalemmal Na+/Ca2+ exchanger 3, potentially acting as an auxiliary subunit for these pathways as well. Our new compound is poised to address several open questions regarding CRMP2's role in regulating the CaV2.2 pathways to treat pain with the potential added benefit of neuroprotection.
Collapse
Affiliation(s)
- Samantha Perez-Miller
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
| | - Kimberly Gomez
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
| | - Rajesh Khanna
- Department
of Pharmacology & Therapeutics, College of Medicine, University of Florida, 1200 Newell Drive, ARB R5-234, Gainesville, Florida 32610-0267, United States
- Pain
and Addiction Therapeutics (PATH) Collaboratory, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
8
|
Iadarola MJ, Sapio MR, Loydpierson AJ, Mervis CB, Fehrenbacher JC, Vasko MR, Maric D, Eisenberg DP, Nash TA, Kippenhan JS, Garvey MH, Mannes AJ, Gregory MD, Berman KF. Syntaxin1A overexpression and pain insensitivity in individuals with 7q11.23 duplication syndrome. JCI Insight 2024; 9:e176147. [PMID: 38261410 DOI: 10.1172/jci.insight.176147] [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: 10/18/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Genetic modifications leading to pain insensitivity phenotypes, while rare, provide invaluable insights into the molecular biology of pain and reveal targets for analgesic drugs. Pain insensitivity typically results from Mendelian loss-of-function mutations in genes expressed in nociceptive (pain-sensing) dorsal root ganglion (DRG) neurons that connect the body to the spinal cord. We document a pain insensitivity mechanism arising from gene overexpression in individuals with the rare 7q11.23 duplication syndrome (Dup7), who have 3 copies of the approximately 1.5-megabase Williams syndrome (WS) critical region. Based on parental accounts and pain ratings, people with Dup7, mainly children in this study, are pain insensitive following serious injury to skin, bones, teeth, or viscera. In contrast, diploid siblings (2 copies of the WS critical region) and individuals with WS (1 copy) show standard reactions to painful events. A converging series of human assessments and cross-species cell biological and transcriptomic studies identified 1 likely candidate in the WS critical region, STX1A, as underlying the pain insensitivity phenotype. STX1A codes for the synaptic vesicle fusion protein syntaxin1A. Excess syntaxin1A was demonstrated to compromise neuropeptide exocytosis from nociceptive DRG neurons. Taken together, these data indicate a mechanism for producing "genetic analgesia" in Dup7 and offer previously untargeted routes to pain control.
Collapse
Affiliation(s)
- Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Amelia J Loydpierson
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Carolyn B Mervis
- Neurodevelopmental Sciences Laboratory, Department of Psychological and Brain Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael R Vasko
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke (NINDS), and
| | - Daniel P Eisenberg
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Tiffany A Nash
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - J Shane Kippenhan
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Madeline H Garvey
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael D Gregory
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| | - Karen F Berman
- Clinical and Translational Neuroscience Branch, National Institute of Mental Health (NIMH), NIH, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Goudsward HJ, Ruiz-Velasco V, Stella SL, Willing LB, Holmes GM. Coexpressed δ-, μ-, and κ-Opioid Receptors Modulate Voltage-Gated Ca 2+ Channels in Gastric-Projecting Vagal Afferent Neurons. Mol Pharmacol 2024; 105:250-259. [PMID: 38182431 PMCID: PMC10877734 DOI: 10.1124/molpharm.123.000774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/07/2024] Open
Abstract
Opioid analgesics are frequently associated with gastrointestinal side effects, including constipation, nausea, dysphagia, and reduced gastric motility. Though it has been shown that stimulation of opioid receptors expressed in enteric motor neurons contributes to opioid-induced constipation, it remains unclear whether activation of opioid receptors in gastric-projecting nodose ganglia neurons contributes to the reduction in gastric motility and emptying associated with opioid use. In the present study, whole-cell patch-clamp recordings were performed to determine the mechanism underlying opioid receptor-mediated modulation of Ca2+ currents in acutely isolated gastric vagal afferent neurons. Our results demonstrate that CaV2.2 channels provide the majority (71% ± 16%) of Ca2+ currents in gastric vagal afferent neurons. Furthermore, we found that application of oxycodone, U-50488, or deltorphin II on gastric nodose ganglia neurons inhibited Ca2+ currents through a voltage-dependent mechanism by coupling to the Gα i/o family of heterotrimeric G-proteins. Because previous studies have demonstrated that the nodose ganglia expresses low levels of δ-opioid receptors, we also determined the deltorphin II concentration-response relationship and assessed deltorphin-mediated Ca2+ current inhibition following exposure to the δ-opioid receptor antagonist ICI 174,864 (0.3 µM). The peak mean Ca2+ current inhibition following deltorphin II application was 47% ± 24% (EC50 = 302.6 nM), and exposure to ICI 174,864 blocked deltorphin II-mediated Ca2+ current inhibition (4% ± 4% versus 37% ± 20%). Together, our results suggest that analgesics targeting any opioid receptor subtype can modulate gastric vagal circuits. SIGNIFICANCE STATEMENT: This study demonstrated that in gastric nodose ganglia neurons, agonists targeting all three classical opioid receptor subtypes (μ, δ, and κ) inhibit voltage-gated Ca2+ channels in a voltage-dependent mechanism by coupling to Gαi/o. These findings suggest that analgesics targeting any opioid receptor subtype would modulate gastric vagal circuits responsible for regulating gastric reflexes.
Collapse
Affiliation(s)
- Hannah J Goudsward
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., L.B.W., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., L.B.W., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Salvatore L Stella
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., L.B.W., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Lisa B Willing
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., L.B.W., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Gregory M Holmes
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., L.B.W., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V.), Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
10
|
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
|
11
|
de Amorim Ferreira M, Ferreira J. Role of Cav2.3 (R-type) Calcium Channel in Pain and Analgesia: A Scoping Review. Curr Neuropharmacol 2024; 22:1909-1922. [PMID: 37581322 PMCID: PMC11284728 DOI: 10.2174/1570159x21666230811102700] [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: 11/21/2022] [Revised: 12/22/2022] [Accepted: 02/15/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Voltage-gated calcium channels (VGCCs) play an important role in pain development and maintenance. As Cav2.2 and Cav3.2 channels have been identified as potential drug targets for analgesics, the participation of Cav2.3 (that gives rise to R-type calcium currents) in pain and analgesia remains incompletely understood. OBJECTIVE Identify the participation of Cav2.3 in pain and analgesia. METHODS To map research in this area as well as to identify any existing gaps in knowledge on the potential role of Cav2.3 in pain signalling, we conducted this scoping review. We searched PubMed and SCOPUS databases, and 40 articles were included in this study. Besides, we organized the studies into 5 types of categories within the broader context of the role of Cav2.3 in pain and analgesia. RESULTS Some studies revealed the expression of Cav2.3 in pain pathways, especially in nociceptive neurons at the sensory ganglia. Other studies demonstrated that Cav2.3-mediated currents could be inhibited by analgesic/antinociceptive drugs either indirectly or directly. Some articles indicated that Cav2.3 modulates nociceptive transmission, especially at the pre-synaptic level at spinal sites. There are studies using different rodent pain models and approaches to reduce Cav2.3 activity or expression and mostly demonstrated a pro-nociceptive role of Cav2.3, despite some contradictory findings and deficiencies in the description of study design quality. There are three studies that reported the association of single-nucleotide polymorphisms in the Cav2.3 gene (CACNA1E) with postoperative pain and opioid consumption as well as with the prevalence of migraine in patients. CONCLUSION Cav2.3 is a target for some analgesic drugs and has a pro-nociceptive role in pain.
Collapse
Affiliation(s)
| | - Juliano Ferreira
- Graduate Program of Pharmacology, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| |
Collapse
|
12
|
Celik MÖ, Seitz V, Yergöz F, Dembla S, Blum NK, Schulz S, Stein C. Modulation of G-protein activation, calcium currents and opioid receptor phosphorylation by the pH-dependent antinociceptive agonist NFEPP. Front Mol Neurosci 2023; 16:1171855. [PMID: 37251645 PMCID: PMC10213447 DOI: 10.3389/fnmol.2023.1171855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide is a newly-designed pain killer selectively activating G-protein-coupled mu-opioid receptors (MOR) in acidic injured tissues, and therefore devoid of central side effects which are typically elicited at normal pH values in healthy tissues. However, the neuronal mechanisms underlying NFEPP's antinociceptive effects were not examined in detail so far. Voltage-dependent Ca2+ channels (VDCCs) in nociceptive neurons play a major role in the generation and inhibition of pain. In this study, we focused on the effects of NFEPP on calcium currents in rat dorsal root ganglion (DRG) neurons. The inhibitory role of the G-protein subunits Gi/o and Gβγ on VDCCs was investigated using the blockers pertussis toxin and gallein, respectively. GTPγS binding, calcium signals and MOR phosphorylation were also investigated. All experiments were performed at acidic and normal pH values using NFEPP in comparison to the conventional opioid agonist fentanyl. At low pH, NFEPP produced more efficient G-protein activation in transfected HEK293 cells and significantly reduced VDCCs in depolarized DRG neurons. The latter effect was mediated by Gβγ subunits, and NFEPP-mediated MOR phosphorylation was pH-dependent. Fentanyl's responses were not affected by pH changes. Our data indicate that NFEPP-induced MOR signaling is more effective at low pH and that the inhibition of calcium channels in DRG neurons underlies NFEPP's antinociceptive actions.
Collapse
Affiliation(s)
- Melih Özgür Celik
- Department of Experimental Anesthesiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Viola Seitz
- Department of Experimental Anesthesiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fatih Yergöz
- Department of Experimental Anesthesiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sandeep Dembla
- Department of Experimental Anesthesiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Stefan Schulz
- Department of Pharmacology, Universitätsklinikum Jena, Jena, Germany
| | - Christoph Stein
- Department of Experimental Anesthesiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
13
|
Danaf J, da Silveira Scarpellini C, Montandon G. βγ G-proteins, but not regulators of G-protein signaling 4, modulate opioid-induced respiratory rate depression. Front Physiol 2023; 14:1043581. [PMID: 37089428 PMCID: PMC10117644 DOI: 10.3389/fphys.2023.1043581] [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: 09/13/2022] [Accepted: 03/24/2023] [Indexed: 04/09/2023] Open
Abstract
Opioid medications are the mainstay of pain management but present substantial side-effects such as respiratory depression which can be lethal with overdose. Most opioid drugs, such as fentanyl, act on opioid receptors such as the G-protein-coupled µ-opioid receptors (MOR). G-protein-coupled receptors activate pertussis toxin-sensitive G-proteins to inhibit neuronal activity. Binding of opioid ligands to MOR and subsequent activation G proteins βγ is modulated by regulator of G-protein signaling (RGS). The roles of G-proteins βγ and RGS in MOR-mediated inhibition of the respiratory network are not known. Using rodent models to pharmacologically modulate G-protein signaling, we aim to determine the roles of βγ G-proteins and RGS4. We showed that inhibition of βγ G-proteins using gallein perfused in the brainstem circuits regulating respiratory depression by opioid drugs results in complete reversal of respiratory depression. Blocking of RGS4 using CCG55014 did not change the respiratory depression induced by MOR activation despite co-expression of RGS4 and MORs in the brainstem. Our results suggest that neuronal inhibition by opioid drugs is mediated by G-proteins, but not by RGS4, which supports the concept that βγ G-proteins could be molecular targets to develop opioid overdose antidotes without the risks of re-narcotization often found with highly potent opioid drugs. On the other hand, RGS4 mediates opioid analgesia, but not respiratory depression, and RGS4 may be molecular targets to develop pain therapies without respiratory liability.
Collapse
Affiliation(s)
- Jamil Danaf
- St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
| | | | - Gaspard Montandon
- St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Graven-Nielsen CS, Knoph CS, Okdahl T, Høyer KL, Krogh K, Hellström PM, Drewes AM. Opioids in the Treatment of Chronic Idiopathic Diarrhea in Humans—A Systematic Review and Treatment Guideline. J Clin Med 2023; 12:jcm12072488. [PMID: 37048572 PMCID: PMC10094889 DOI: 10.3390/jcm12072488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
In patients with chronic idiopathic diarrhea resistant to standard treatment, opioids are often used as rescue therapy. This systematic review investigated opioid effects on gut function in chronic diarrhea. PubMed and Embase were searched regarding effects of opioid agonists on the gastrointestinal tract in humans with chronic or experimentally induced diarrhea. A total of 1472 relevant articles were identified and, after thorough evaluation, 11 clinical trials were included. Generally, studies reported a reduction in stool frequency and an increase in transit time during treatment with the opioid receptor agonists loperamide, asimadoline, casokefamide, and codeine compared with placebo. Loperamide and diphenoxylate significantly improved stool consistency compared with placebo, whereas asimadoline showed no such effects. Compared with placebo, loperamide treatment caused less abdominal pain and urgency. Asimadoline showed no significant subjective improvements, but fedotozine was superior to placebo in reducing abdominal pain and bloating in selected patients. Only two relevant studies were published within the last 20 years, and standardized endpoint measures are lacking. Most trials included few participants, and further evidence is needed from larger, prospective studies. Likewise, consensus is needed to standardize endpoints for stool frequency, transit time, and consistency to conduct future meta-analyses on opioids in management of chronic idiopathic diarrhea.
Collapse
|
15
|
Kelly E, Conibear A, Henderson G. Biased Agonism: Lessons from Studies of Opioid Receptor Agonists. Annu Rev Pharmacol Toxicol 2023; 63:491-515. [PMID: 36170657 DOI: 10.1146/annurev-pharmtox-052120-091058] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In ligand bias different agonist drugs are thought to produce distinct signaling outputs when activating the same receptor. If these signaling outputs mediate therapeutic versus adverse drug effects, then agonists that selectively activate the therapeutic signaling pathway would be extremely beneficial. It has long been thought that μ-opioid receptor agonists that selectively activate G protein- over β-arrestin-dependent signaling pathways would produce effective analgesia without the adverse effects such as respiratory depression. However, more recent data indicate that most of the therapeutic and adverse effects of agonist-induced activation of the μ-opioid receptor are actually mediated by the G protein-dependent signaling pathway, and that a number of drugs described as G protein biased in fact may not be biased, but instead may be low-intrinsic-efficacy agonists. In this review we discuss the current state of the field of bias at the μ-opioid receptor and other opioid receptor subtypes.
Collapse
Affiliation(s)
- Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom;
| | - Alexandra Conibear
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom;
| | - Graeme Henderson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom;
| |
Collapse
|
16
|
Martin L, Ibrahim M, Gomez K, Yu J, Cai S, Chew LA, Bellampalli SS, Moutal A, Largent-Milnes T, Porreca F, Khanna R, Olivera BM, Patwardhan A. Conotoxin contulakin-G engages a neurotensin receptor 2/R-type calcium channel (Cav2.3) pathway to mediate spinal antinociception. Pain 2022; 163:1751-1762. [PMID: 35050960 PMCID: PMC9198109 DOI: 10.1097/j.pain.0000000000002561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/07/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Intrathecal application of contulakin-G (CGX), a conotoxin peptide and a neurotensin analogue, has been demonstrated to be safe and potentially analgesic in humans. However, the mechanism of action for CGX analgesia is unknown. We hypothesized that spinal application of CGX produces antinociception through activation of the presynaptic neurotensin receptor (NTSR)2. In this study, we assessed the mechanisms of CGX antinociception in rodent models of inflammatory and neuropathic pain. Intrathecal administration of CGX, dose dependently, inhibited thermal and mechanical hypersensitivities in rodents of both sexes. Pharmacological and clustered regularly interspaced short palindromic repeats/Cas9 editing of NTSR2 reversed CGX-induced antinociception without affecting morphine analgesia. Electrophysiological and gene editing approaches demonstrated that CGX inhibition was dependent on the R-type voltage-gated calcium channel (Cav2.3) in sensory neurons. Anatomical studies demonstrated coexpression of NTSR2 and Cav2.3 in dorsal root ganglion neurons. Finally, synaptic fractionation and slice electrophysiology recordings confirmed a predominantly presynaptic effect. Together, these data reveal a nonopioid pathway engaged by a human-tested drug to produce antinociception.
Collapse
Affiliation(s)
- Laurent Martin
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Mohab Ibrahim
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ 85742, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Jie Yu
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Lindsey A. Chew
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Shreya Sai Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Tally Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ 85742, USA
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ 85742, USA
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, USA
| | | | - Amol Patwardhan
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
17
|
Dehe L, Mousa SA, Shaqura M, Shakibaei M, Schäfer M, Treskatsch S. Naltrexone-Induced Cardiac Function Improvement is Associated With an Attenuated Inflammatory Response and Lipid Perioxidation in Volume Overloaded Rats. Front Pharmacol 2022; 13:873169. [PMID: 35847039 PMCID: PMC9280420 DOI: 10.3389/fphar.2022.873169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
In previous studies, upregulation of myocardial opioid receptors as well as the precursors of their endogenous ligands were detected in the failing heart due to chronic volume overload. Moreover, opioid receptor blockade by naltrexone improved left ventricular function. In parallel, inflammatory processes through cytokines have been confirmed to play an important role in the pathogenesis of different forms of heart failure. Thus, the present study examined the systemic and myocardial inflammatory response to chronic volume overload and its modulation by chronic naltrexone therapy. Chronic volume overload was induced in male Wistar rats by applying an infrarenal aortocaval fistula (ACF) for 28 days during which the selective opioid receptor antagonist naltrexone (n = 6) or vehicle (n = 6) were administered via a subcutaneously implanted Alzet minipump. The ultrastructural, morphometric and hemodynamic characterization of ACF animals were performed using an intraventricular conductance catheter in vivo and electron microscopy in vitro. Co-localization of mu-, delta- and kappa-opioid receptor subtypes (MOR, DOR, and KOR respectively) with the voltage gated L-type Ca2+ channel (Cav1.2), the ryanodine receptor (RyR), and mitochondria in cardiomyocytes as well as IL-6, IL-12, TNF-alpha, and Malondialdehyde (MDA) were determined using double immunofluorescence confocal microscopy, RT-PCR and ELISA, respectively. In rat left ventricular myocardium, three opioid receptor subtypes MOR, DOR, and KOR colocalized with Cav1.2, RyR and mitochondria suggesting a modulatory role of the excitation-contraction coupling. In rats with ACF-induced volume overload, signs of heart failure and myocardial ultrastructural damage, chronic naltrexone therapy improved cardiac function and reversed the systemic and myocardial inflammatory cytokine expression as well as lipid peroxidation. In conclusion, antagonism of the cardiodepressive effects of the myocardial opioid system does not only improve left ventricular function but also blunts the inflammatory response and lipid peroxidation.
Collapse
Affiliation(s)
- Lukas Dehe
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Shaaban A. Mousa
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
- *Correspondence: Shaaban A. Mousa,
| | - Mohammed Shaqura
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilians-Universität München, München, Germany
| | - Michael Schäfer
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Sascha Treskatsch
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
18
|
Hartung JE, Moy JK, Loeza-Alcocer E, Nagarajan V, Jostock R, Christoph T, Schroeder W, Gold MS. Voltage-gated calcium currents in human dorsal root ganglion neurons. Pain 2022; 163:e774-e785. [PMID: 34510139 PMCID: PMC8882208 DOI: 10.1097/j.pain.0000000000002465] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/10/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Voltage-gated calcium channels in sensory neurons underlie processes ranging from neurotransmitter release to gene expression and remain a therapeutic target for the treatment of pain. Yet virtually all we know about voltage-gated calcium channels has been obtained through the study of rodent sensory neurons and heterologously expressed channels. To address this, high voltage-activated (HVA) Ca2+ currents in dissociated human and rat dorsal root ganglion neurons were characterized with whole-cell patch clamp techniques. The HVA currents from both species shared basic biophysical and pharmacological properties. However, HVA currents in human neurons differed from those in the rat in at least 3 potentially important ways: (1) Ca2+ current density was significantly smaller, (2) the proportion of nifedipine-sensitive currents was far greater, and (3) a subpopulation of human neurons displayed relatively large constitutive current inhibition. These results highlight the need to for the study of native proteins in their native environment before initiating costly clinical trials.
Collapse
Affiliation(s)
- Jane E Hartung
- University of Pittsburgh School of Medicine, Department of Neurobiology, PA, USA
| | - Jamie K Moy
- University of Pittsburgh School of Medicine, Department of Neurobiology, PA, USA
| | | | - Vidhya Nagarajan
- University of Pittsburgh School of Medicine, Department of Neurobiology, PA, USA
| | | | | | | | - Michael S Gold
- University of Pittsburgh School of Medicine, Department of Neurobiology, PA, USA
| |
Collapse
|
19
|
Abstract
Opiates, such as morphine, and synthetic opioids, such as fentanyl, constitute a class of drugs acting on opioid receptors which have been used therapeutically and recreationally for centuries. Opioid drugs have strong analgesic properties and are used to treat moderate to severe pain, but also present side effects including opioid dependence, tolerance, addiction, and respiratory depression, which can lead to lethal overdose if not treated. This chapter explores the pathophysiology, the neural circuits, and the cellular mechanisms underlying opioid-induced respiratory depression and provides a translational perspective of the most recent research. The pathophysiology discussed includes the effects of opioid drugs on the respiratory system in patients, as well as the animal models used to identify underlying mechanisms. Using a combination of gene editing and pharmacology, the neural circuits and molecular pathways mediating neuronal inhibition by opioids are examined. By using pharmacology and neuroscience approaches, new therapies to prevent or reverse respiratory depression by opioid drugs have been identified and are currently being developed. Considering the health and economic burden associated with the current opioid epidemic, innovative research is needed to better understand the side effects of opioid drugs and to discover new therapeutic solutions to reduce the incidence of lethal overdoses.
Collapse
|
20
|
Standifer KM, Inturrisi CE, Foley KM, Pan YX. Understanding Opioid Actions, Pain and Analgesia: A Tribute to Dr. Gavril Pasternak. Cell Mol Neurobiol 2021; 41:827-834. [PMID: 33978862 PMCID: PMC11448670 DOI: 10.1007/s10571-021-01097-0] [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: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
This special issue is a tribute to our mentor, colleague and friend, Gavril W. Pasternak, MD, PhD. Homage to the breadth and depth of his work (~ 450 publications) over a 40 career in pharmacology and medicine cannot be captured fully in one special issue, but the 22 papers collected herein represent seven of the topics near and dear to Gav's heart, and the colleagues, friends and mentees who held him near to theirs. The seven themes include: (1) sites and mechanisms of opioid actions in vivo; (2) development of novel analgesic agents; (3) opioid tolerance, withdrawal and addiction: mechanisms and treatment; (4) opioid receptor splice variants; (5) novel research tools and approaches; (6) receptor signaling and crosstalk in vitro; and (7) mentorship. This introduction to the issue summarizes contributions and includes formal and personal remembrances of Gav that illustrate his personality, warmth, and dedication to making a difference in patient care and people's lives.
Collapse
Affiliation(s)
- Kelly M Standifer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | | | - Kathleen M Foley
- Memorial Sloan Kettering Cancer Center Member Emeritus, New York, NY, USA
| | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NY, USA
| |
Collapse
|