1
|
Duron DI, Tanguturi P, Campbell CS, Chou K, Bejarano P, Gabriel KA, Bowden JL, Mishra S, Brackett C, Barlow D, Houseknecht KL, Blagg BSJ, Streicher JM. Inhibiting spinal cord-specific hsp90 isoforms reveals a novel strategy to improve the therapeutic index of opioid treatment. Sci Rep 2024; 14:14715. [PMID: 38926482 PMCID: PMC11208559 DOI: 10.1038/s41598-024-65637-6] [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: 04/05/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
Opioids are the gold standard for the treatment of chronic pain but are limited by adverse side effects. In our earlier work, we showed that Heat shock protein 90 (Hsp90) has a crucial role in regulating opioid signaling in spinal cord; Hsp90 inhibition in spinal cord enhances opioid anti-nociception. Building on these findings, we injected the non-selective Hsp90 inhibitor KU-32 by the intrathecal route into male and female CD-1 mice, showing that morphine anti-nociceptive potency was boosted by 1.9-3.5-fold in acute and chronic pain models. At the same time, tolerance was reduced from 21-fold to 2.9 fold and established tolerance was rescued, while the potency of constipation and reward was unchanged. These results demonstrate that spinal Hsp90 inhibition can improve the therapeutic index of morphine. However, we also found that systemic non-selective Hsp90 inhibition blocked opioid pain relief. To avoid this effect, we used selective small molecule inhibitors and CRISPR gene editing to identify 3 Hsp90 isoforms active in spinal cord (Hsp90α, Hsp90β, and Grp94) while only Hsp90α was active in brain. We thus hypothesized that a systemically delivered selective inhibitor to Hsp90β or Grp94 could selectively inhibit spinal cord Hsp90 activity, resulting in enhanced opioid therapy. We tested this hypothesis using intravenous delivery of KUNB106 (Hsp90β) and KUNG65 (Grp94), showing that both drugs enhanced morphine anti-nociceptive potency while rescuing tolerance. Together, these results suggest that selective inhibition of spinal cord Hsp90 isoforms is a novel, translationally feasible strategy to improve the therapeutic index of opioids.
Collapse
Affiliation(s)
- David I Duron
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Parthasaradhireddy Tanguturi
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Christopher S Campbell
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Kerry Chou
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Paul Bejarano
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Katherin A Gabriel
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Jessica L Bowden
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA
| | - Sanket Mishra
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN, USA
| | - Christopher Brackett
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN, USA
| | - Deborah Barlow
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - Karen L Houseknecht
- Department of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, ME, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, College of Science, University of Notre Dame, Notre Dame, IN, USA
| | - John M Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Box 245050, LSN563, 1501 N. Campbell Ave., Tucson, AZ, 85724, USA.
- Comprehensive Center for Pain and Addiction, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
2
|
Durant KM, Whitesell A, Dasse KD. A review of fetal cell lines used during drug development: Focus on COVID-19 vaccines, transplant medications, and biologics. Am J Health Syst Pharm 2024; 81:e336-e344. [PMID: 38347743 DOI: 10.1093/ajhp/zxae031] [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] [Indexed: 06/25/2024] Open
Abstract
PURPOSE The recent coronavirus disease 2019 (COVID-19) pandemic and vaccine mandates have increased the number of patient questions related to how fetal cell lines are used during drug development and final manufacturing. This article describes our literature search and review of COVID-19 vaccines, transplant medications, and biologics whose development included use of fetal cell lines. SUMMARY A detailed literature search was conducted to identify the common fetal cell lines used in COVID-19 vaccine development; the two most prevalent fetal cell lines identified were HEK-293 and PER.C6. Subsequent literatures searches were conducted to identify transplant medications and biologics whose development included use of the HEK-293 or PER.C6 cell lines. For the COVID-19 vaccines, only the viral vector vaccine by Janssen was found to contain proteins produced by PER.C6 in the final preparation administered to patients, and Novavax is the only vaccine for which fetal cell lines were not directly involved in any portion of drug development. For transplant medications, many medications were studied in fetal cell lines in postmarketing studies after Food and Drug Administration approval; however, none of these medications contained fetal cells or would expose a patient to a fetal cell line. Many new biologics and cellular therapies for genetic diseases and malignancies have been directly developed from HEK-293 fetal cells or contain proteins produced directly from fetal cell lines. CONCLUSION There were very few drugs reviewed that were found to contain HEK-293 or PER.C6 fetal cells or proteins derived directly from fetal cell lines; however, use of fetal cell lines in biologics and gene therapies will continue to increase. Healthcare providers should be mindful of patients' beliefs while also correcting common misconceptions about how these fetal cell lines are used throughout drug development and manufacturing.
Collapse
Affiliation(s)
- Karin M Durant
- Department of Pharmacy Services, Michigan Medicine, University of Michigan Health, Ann Arbor, MI
- University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Ashlyn Whitesell
- Department of Pharmacy Services, Michigan Medicine, University of Michigan Health, Ann Arbor, MI
- University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Kathy D Dasse
- Department of Pharmacy Services, Michigan Medicine, University of Michigan Health, Ann Arbor, MI
- University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Sun S, Li P, Wang J, Zhao D, Yang T, Zhou P, Su R, Zheng Z, Li S. Novel Scaffold Agonists of the α 2A Adrenergic Receptor Identified via Ensemble-Based Strategy. Molecules 2024; 29:1097. [PMID: 38474611 DOI: 10.3390/molecules29051097] [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: 02/07/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
The α2A adrenergic receptor (α2A-AR) serves as a critical molecular target for sedatives and analgesics. However, α2A-AR ligands with an imidazole ring also interact with an imidazoline receptor as well as other proteins and lead to undesirable effects, motivating us to develop more novel scaffold α2A-AR ligands. For this purpose, we employed an ensemble-based ligand discovery strategy, integrating long-term molecular dynamics (MD) simulations and virtual screening, to identify new potential α2A-AR agonists with novel scaffold. Our results showed that compounds SY-15 and SY-17 exhibited significant biological effects in the preliminary evaluation of protein kinase A (PKA) redistribution assays. They also reduced levels of intracellular cyclic adenosine monophosphate (cAMP) in a dose-dependent manner. Upon treatment of the cells with 100 μM concentrations of SY-15 and SY-17, there was a respective decrease in the intracellular cAMP levels by 63.43% and 53.83%. Subsequent computational analysis was conducted to elucidate the binding interactions of SY-15 and SY-17 with the α2A-AR. The binding free energies of SY-15 and SY-17 calculated by MD simulations were -45.93 and -71.97 kcal/mol. MD simulations also revealed that both compounds act as bitopic agonists, occupying the orthosteric site and a novel exosite of the receptor simultaneously. Our findings of integrative computational and experimental approaches could offer the potential to enhance ligand affinity and selectivity through dual-site occupancy and provide a novel direction for the rational design of sedatives and analgesics.
Collapse
Affiliation(s)
- Shiyang Sun
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Pengyun Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiaqi Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dongsheng Zhao
- Academy of Military Medical Sciences, Beijing 100850, China
| | - Tingting Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Peilan Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zhibing Zheng
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Song Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
4
|
Zhu H, Liu X, Wang X, Li Y, Ma F, Tan B, Zhou P, Fu F, Su R. Gβγ subunit inhibitor decreases DOM-induced head twitch response via the PLCβ/IP3/Ca 2+/ERK and cAMP signaling pathways. Eur J Pharmacol 2023; 957:176038. [PMID: 37657742 DOI: 10.1016/j.ejphar.2023.176038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023]
Abstract
AIMS (-)-2,5-dimethoxy-4-methylamphetamine (DOM) induces the head-twitch response (HTR) primarily by activating the serotonin 5-hydroxytryptamine 2A receptor (5-HT2A receptor) in mice. However, the mechanisms underlying 5-HT2A receptor activation and the HTR remain elusive. Gβγ subunits are a potential treatment target in numerous diseases. The present study investigated the mechanism whereby Gβγ subunits influence DOM-induced HTR. MAIN METHODS The effects of the Gβγ inhibitor 3',4',5',6'-tetrahydroxyspiro[2-benzofuran-3,9'-xanthene]-1-one (gallein) and antagonistic peptide βARKct (β-adrenergic receptor kinase C-terminal fragment) on DOM-induced HTR were studied via an HTR test. The activation of the phospholipase C β (PLCβ)/inositol triphosphate (IP3)/calcium (Ca2+) signaling pathway and extracellular signal-regulated kinase (ERK) following Gβγ subunit inhibition was detected by western blotting, Homogeneous Time-Resolved Fluorescence (HTRF) inositol phosphate (IP1) assay and Fluorometric Imaging Plate Reader (FLIPR) calcium 6 assay. The Gβγ subunit-mediated regulation of cyclic adenosine monophosphate (cAMP) was assessed via a GloSensor™ cAMP assay. KEY FINDINGS The Gβγ subunit inhibitors gallein and βARKct reduced DOM-induced HTR in C57BL/6J mice. Like the 5-HT2A receptor-selective antagonist (R)-[2,3-di(methoxy)phenyl]-[1-[2-(4-fluorophenyl)ethyl]piperidin-4-yl]methanol (M100907), gallein inhibited PLCβ phosphorylation (pPLCβ), IP1 production, Ca2+ transients, ERK1/2 phosphorylation (pERK1/2) and cAMP accumulation induced by DOM in human embryonic kidney (HEK) 293T cells stably or transiently transfected with the human 5-HT2A receptor. Moreover, PLCβ protein inhibitor 1-[6-[[(8R,9S,13S,14S,17S)-3-methoxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-17-yl]amino]hexyl]pyrrole-2,5-dione (U73122) (10 nmol/mouse), intracellular Ca2+ blocker 6-[6-[6-[5-acetamido-4,6-dihydroxy-2-(sulfooxymethyl)oxan-3-yl]oxy-2-carboxy-4-hydroxy-5-sulfooxyoxan-3-yl]oxy-2-(hydroxymethyl)-5-(sulfoamino)-4-sulfooxyoxan-3-yl]oxy-3,4-dihydroxy-5-sulfooxyoxane-2-carboxylic acid (heparin) (5 nmol/mouse), L-type Ca2+ channel blocker 3-O-(2-methoxyethyl) 5-O-propan-2-yl 2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3,5-dicarboxylate (nimodipine) (4 mg/kg), mitogen extracellular regulating kinase 1/2 (MEK1/2) inhibitor (Z)-3-amino-3-(4-aminophenyl)sulfanyl-2-[2-(trifluoromethyl)phenyl]prop-2-enenitrile (SL327) (30 mg/kg), and Gαs protein selective antagonist 4,4',4″,4‴-(Carbonylbis-(imino-5,1,3-benzenetriylbis(carbonylimino)))tetrakisbenzene-1,3-disulfonic acid (NF449) (10 nmol/mouse) reduced DOM-induced HTR in C57BL/6J mice. SIGNIFICANCE The Gβγ subunits potentially mediate the HTR after 5-HT2A receptor activation via the PLCβ/IP3/Ca2+/ERK1/2 and cAMP signaling pathways. Inhibitors targeting the Gβγ subunits potentially inhibit the hallucinogenic effects of 5-HT2A receptor agonists.
Collapse
Affiliation(s)
- Huili Zhu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China; School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Xiaoqian Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Xiaoxuan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Yulei Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Fang Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Bo Tan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Peilan Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China.
| | - Fenghua Fu
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China.
| |
Collapse
|
5
|
Tian X, Zhang J, Wang S, Gao H, Sun Y, Liu X, Fu W, Tan B, Su R. Tyrosine 7.43 is important for mu-opioid receptor downstream signaling pathways activated by fentanyl. Front Pharmacol 2022; 13:919325. [PMID: 36120357 PMCID: PMC9478952 DOI: 10.3389/fphar.2022.919325] [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: 04/13/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
G protein–coupled receptors can signal through both G proteins and ß-arrestin2. For the µ-opioid receptor (MOR), early experimental evidence from a single study suggested that G protein signaling mediates analgesia and sedation, whereas ß-arrestin signaling mediates respiratory depression and constipation. Then, receptor mutations were used to clarify which residues interact with ligands to selectively regulate signals in a ligand-specific manner. However, there is no systematic study on how to determine these residues and clarify the molecular mechanism of their influence on signal pathways. We have therefore used molecular docking to predict the amino acid sites that affect the binding of ligands and MOR. Then, the corresponding sites were mutated to determine the effect of the structural determinant of MOR on Gi/o protein and ß-arrestin pathways. The pharmacological and animal behavioral experiments in combination with molecular dynamics simulations were used to elucidate the molecular mechanism of key residues governing the signaling. Without affecting ligand binding to MOR, MORY7.43A attenuated the activation of both Gi/o protein and ß-arrestin signaling pathways stimulated by fentanyl, whereas it did not change these two pathways stimulated by morphine. Likewise, the activation peak time of extracellular regulated protein kinases was significantly prolonged at MORY7.43A compared with that at MORwildtype stimulated by fentanyl, but there was no difference stimulated by morphine. In addition, MORY7.43A significantly enhanced analgesia by fentanyl but not by morphine in the mice behavioral experiment. Furthermore, the molecular dynamics simulations showed that H6 moves toward the cellular membrane. H6 of the fentanyl–Y7.43A system moved outward more than that in the morphine–Y7.43A system. Y7.43 mutation disrupted hydrophobic interactions between W6.48 and Y7.43 in the fentanyl–Y7.43A system but not in the morphine–Y7.43A system. Our results have disclosed novel mechanisms of Y7.43 mutation affecting MOR signaling pathways. Y7.43 mutation reduced the activation of the Gi/o protein pathway and blocked the ß-arrestin2 recruitment, increased the H6 outward movement of MOR, and disrupted hydrophobic interactions. This may be responsible for the enhanced fentanyl analgesia. These findings are conducive to designing new drugs from the perspective of ligand and receptor binding, and Y7.43 is also expected to be a key site to structure optimization of synthesized compounds.
Collapse
Affiliation(s)
- Xiangyun Tian
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Junjie Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Shaowen Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Huan Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- School of Pharmacy, Yantai University, Yantai, China
| | - Yi Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaoqian Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Bo Tan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Bo Tan, , ; Ruibin Su, ,
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Bo Tan, , ; Ruibin Su, ,
| |
Collapse
|
6
|
Maiti S, Picard D. Cytosolic Hsp90 Isoform-Specific Functions and Clinical Significance. Biomolecules 2022; 12:1166. [PMID: 36139005 PMCID: PMC9496497 DOI: 10.3390/biom12091166] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
The heat shock protein 90 (Hsp90) is a molecular chaperone and a key regulator of proteostasis under both physiological and stress conditions. In mammals, there are two cytosolic Hsp90 isoforms: Hsp90α and Hsp90β. These two isoforms are 85% identical and encoded by two different genes. Hsp90β is constitutively expressed and essential for early mouse development, while Hsp90α is stress-inducible and not necessary for survivability. These two isoforms are known to have largely overlapping functions and to interact with a large fraction of the proteome. To what extent there are isoform-specific functions at the protein level has only relatively recently begun to emerge. There are studies indicating that one isoform is more involved in the functionality of a specific tissue or cell type. Moreover, in many diseases, functionally altered cells appear to be more dependent on one particular isoform. This leaves space for designing therapeutic strategies in an isoform-specific way, which may overcome the unfavorable outcome of pan-Hsp90 inhibition encountered in previous clinical trials. For this to succeed, isoform-specific functions must be understood in more detail. In this review, we summarize the available information on isoform-specific functions of mammalian Hsp90 and connect it to possible clinical applications.
Collapse
Affiliation(s)
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH-1211 Geneve, Switzerland
| |
Collapse
|
7
|
Pan- and isoform-specific inhibition of Hsp90: Design strategy and recent advances. Eur J Med Chem 2022; 238:114516. [DOI: 10.1016/j.ejmech.2022.114516] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022]
|
8
|
Skoie IM, Bårdsen K, Nilsen MM, Eidem LE, Grimstad T, Dalen I, Omdal R. Fatigue and expression of heat shock genes in plaque type psoriasis. Clin Exp Dermatol 2021; 47:1068-1077. [PMID: 34921435 DOI: 10.1111/ced.15068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Chronic fatigue is common in psoriasis, and heat shock proteins (HSP) have been postulated to influence fatigue. OBJECTIVE To evaluate gene expression patterns of selected HSPs in psoriasis patients with high versus low fatigue. METHODS Fatigue was assessed using the fatigue Visual Analoge Scale and disease activity by the Psoriasis Area and Severity Index. Peripheral blood transcriptional profiles (RNA-seq) of HSP genes from 10 patients with high fatigue were compared with 10 patients with low fatigue. HSPB11, HSPBAP1, HSPA14, HSPA9P1, HSP90B1 and HSP90AB1 contributed most to separation of the two groups in a principal component analysis. Four of these genes (HSPB11, HSPA14, HSP90B1, HSP90AB1) were further investigated by reverse transcription quantitative real-time polymerase reaction (RT-qPCR) in 20 patients with high and 20 with low fatigue scores. RESULTS Both RNA-seq and RT-qPCR analyses revealed a tendency to higher expression levels of HSPB11 and lower expression of HSP90B1 in the high fatigue group versus the low fatigue group. Psoriasis disease activity had no influence on the expression levels of the studied HSP genes. CONCLUSION Overall, the results suggest that some HSPs are involved in generation of fatigue in psoriasis supporting the hypothesis that downregulatory innate immune responses influence fatigue.
Collapse
Affiliation(s)
| | - Kjetil Bårdsen
- Research Department, Stavanger University Hospital, Stavanger
| | - Mari M Nilsen
- Department Chemistry, Bioscience and Environmental Engineering, University of Stavanger
| | - Live E Eidem
- Research Department, Stavanger University Hospital, Stavanger
| | - Tore Grimstad
- Department of Gastroenterology, Stavanger University Hospital, Stavanger.,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen
| | - Ingvild Dalen
- Section of Biostatistics, Stavanger University Stavanger
| | - Roald Omdal
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen.,Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
9
|
Zhang Y, Zhou P, Lu F, Su R, Gong Z. A20-Binding Inhibitor of Nuclear Factor- κB Targets β-Arrestin2 to Attenuate Opioid Tolerance. Mol Pharmacol 2021; 100:170-180. [PMID: 34031190 DOI: 10.1124/molpharm.120.000211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/26/2021] [Indexed: 11/22/2022] Open
Abstract
Opioids play an important role in pain relief, but repeated exposure results in tolerance and dependence. To make opioids more effective and useful, research in the field has focused on reducing the tolerance and dependence for chronic pain relief. Here, we showed the effect of A20-binding inhibitor of nuclear factor-κB (ABIN-1) in modulating morphine function. We used hot-plate tests and conditioned place preference (CPP) tests to show that overexpression of ABIN-1 in the mouse brain attenuated morphine dependence. These effects of ABIN-1 are most likely mediated through the formation of ABIN-1-β-arrestin2 complexes, which accelerate β-arrestin2 degradation by ubiquitination. With the degradation of β-arrestin2, ABIN-1 overexpression also decreased μ opioid receptor (MOR) phosphorylation and internalization after opioid treatment, affecting the β-arrestin2-dependent signaling pathway to regulate morphine tolerance. Importantly, the effect of ABIN-1 on morphine tolerance was abolished in β-arrestin2-knockout mice. Taken together, these results suggest that the interaction between ABIN-1 and β-arrestin2 inhibits MOR internalization to attenuate morphine tolerance, revealing a novel mechanism for MOR regulation. Hence, ABIN-1 may be a therapeutic target to regulate MOR internalization, thus providing a foundation for a novel treatment strategy for alleviating morphine tolerance and dependence. SIGNIFICANCE STATEMENT: A20-binding inhibitor of nuclear factor-κB (ABIN-1) overexpression in the mouse brain attenuated morphine tolerance and dependence. The likely mechanism for this finding is that ABIN-1-β-arrestin2 complex formation facilitated β-arrestin2 degradation by ubiquitination. ABIN-1 targeted β-arrestin2 to regulate morphine tolerance. Therefore, the enhancement of ABIN-1 is an important strategy to prevent morphine tolerance and dependence.
Collapse
Affiliation(s)
- Yixin Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Peilan Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Fengfeng Lu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zehui Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
10
|
Injection of minocycline into the periaqueductal gray attenuates morphine withdrawal signs. Neurosci Lett 2020; 736:135283. [PMID: 32739271 DOI: 10.1016/j.neulet.2020.135283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/07/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022]
Abstract
This study investigated the effects of minocycline microinjections, into the midbrain periaqueductal gray (PAG), on morphine withdrawal and the expression of pannexin-1 (panx1), phosphorylated mammalian target of rapamycin (p-mTOR), protein kinase A (PKA), and cAMP response element-binding protein (CREB). Rats were injected with morphine, intraperitoneally, at increasing doses, twice per day, to establish animal models of morphine exposure. Minocycline was administered into the PAG before the first intraperitoneal (i.p.) injection of morphine each day, on days 1-4. On the last day of the experiment, all rats were injected with naloxone, and morphine withdrawal was observed, and then changes in the expression levels of ionized calcium-binding adaptor molecule 1 (Iba1) and its downstream factors, panx1, p-mTOR, PKA, and CREB were evaluated by western blot and immunohistochemistry analyses. Morphine withdrawal increased microglial activation, whereas minocycline could inhibit microglial activation and withdrawal and the downregulation of panx1, p-mTOR, PKA, and CREB expression, reducing the effects of morphine withdrawal.
Collapse
|