1
|
Kalogriopoulos NA, Tei R, Yan Y, Klein PM, Ravalin M, Cai B, Soltesz I, Li Y, Ting A. Synthetic GPCRs for programmable sensing and control of cell behaviour. Nature 2025; 637:230-239. [PMID: 39633047 DOI: 10.1038/s41586-024-08282-3] [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: 02/28/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
Synthetic receptors that mediate antigen-dependent cell responses are transforming therapeutics, drug discovery and basic research1,2. However, established technologies such as chimeric antigen receptors3 can only detect immobilized antigens, have limited output scope and lack built-in drug control3-7. Here we engineer synthetic G-protein-coupled receptors (GPCRs) that are capable of driving a wide range of native or non-native cellular processes in response to a user-defined antigen. We achieve modular antigen gating by engineering and fusing a conditional auto-inhibitory domain onto GPCR scaffolds. Antigen binding to a fused nanobody relieves auto-inhibition and enables receptor activation by drug, thus generating programmable antigen-gated G-protein-coupled engineered receptors (PAGERs). We create PAGERs that are responsive to more than a dozen biologically and therapeutically important soluble and cell-surface antigens in a single step from corresponding nanobody binders. Different PAGER scaffolds allow antigen binding to drive transgene expression, real-time fluorescence or endogenous G-protein activation, enabling control of diverse cellular functions. We demonstrate multiple applications of PAGER, including induction of T cell migration along a soluble antigen gradient, control of macrophage differentiation, secretion of therapeutic antibodies and inhibition of neuronal activity in mouse brain slices. Owing to its modular design and generalizability, we expect PAGERs to have broad utility in discovery and translational science.
Collapse
Affiliation(s)
| | - Reika Tei
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Yuqi Yan
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Matthew Ravalin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Bo Cai
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Yulong Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, Beijing, China
| | - Alice Ting
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Department of Biology, Stanford University, Stanford, CA, USA.
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
- Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Singh K, Yadav A, Khan S, Shukla A, Alam M, Verma AK, Tiwari N, Khan F, Yadav PN, Dev K. Baicalein isolated from Oroxylum indicum acts as a potent µ- and κ-opioid receptor antagonist agent via the reversal of agonist-mediated cAMP inhibition. Nat Prod Res 2024:1-9. [PMID: 39205489 DOI: 10.1080/14786419.2024.2396452] [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: 01/09/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The opioid receptors play a pivotal role in the treatment of several neuropsychiatric and neurological disorders. Oroxylum indicum (L.) Kurtz is a very important medicinal plant with several therapeutic applications. It is a main constituent of the Ayurvedic formulation 'Dashmool' used for multifaceted disorders by the Indians. However, the constituents of this plant in neurological conditions have not been well studied. Here, we performed activity-guided isolation of compounds for opioid receptor modulator activity. In the study, we found that the isolated compound baicalein (3) has shown the most potent and competitive antagonistic activity at 20 mg/kg dose in vivo experiments. The acute dose of 3 (20 mg/kg) and pan opioid receptor antagonist naloxone (20 mg/kg) block the morphine-induced antinociception and the paw withdrawal latency decreases up to 8.3 s and 9.6 s, respectively. The in silico studies also support our in vitro data that compound 3 binds with MOR and KOR.
Collapse
Affiliation(s)
- Kishan Singh
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
| | - Anubhav Yadav
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
- Neuroscience & Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sana Khan
- Department of Metabolic and Structural Biology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Arti Shukla
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Maksood Alam
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | | | - Neerja Tiwari
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Feroz Khan
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
- Department of Metabolic and Structural Biology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Prem Narayan Yadav
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
- Neuroscience & Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kapil Dev
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
| |
Collapse
|
3
|
Goode TD, Alipio JB, Besnard A, Pathak D, Kritzer-Cheren MD, Chung A, Duan X, Sahay A. A dorsal hippocampus-prodynorphinergic dorsolateral septum-to-lateral hypothalamus circuit mediates contextual gating of feeding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606427. [PMID: 39149322 PMCID: PMC11326193 DOI: 10.1101/2024.08.02.606427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Adaptive regulation of feeding depends on linkage of internal states and food outcomes with contextual cues. Human brain imaging has identified dysregulation of a hippocampal-lateral hypothalamic area (LHA) network in binge eating, but mechanistic instantiation of underlying cell-types and circuitry is lacking. Here, we identify an evolutionary conserved and discrete Prodynorphin (Pdyn)-expressing subpopulation of Somatostatin (Sst)-expressing inhibitory neurons in the dorsolateral septum (DLS) that receives primarily dorsal, but not ventral, hippocampal inputs. DLS(Pdyn) neurons inhibit LHA GABAergic neurons and confer context- and internal state-dependent calibration of feeding. Viral deletion of Pdyn in the DLS mimicked effects seen with optogenetic silencing of DLS Pdyn INs, suggesting a potential role for DYNORPHIN-KAPPA OPIOID RECEPTOR signaling in contextual regulation of food-seeking. Together, our findings illustrate how the dorsal hippocampus has evolved to recruit an ancient LHA feeding circuit module through Pdyn DLS inhibitory neurons to link contextual information with regulation of food consumption.
Collapse
Affiliation(s)
- Travis D Goode
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Jason Bondoc Alipio
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Devesh Pathak
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Michael D Kritzer-Cheren
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Ain Chung
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, CA
- Department of Physiology, University of California, San Francisco, CA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
4
|
Li Y, Wu M, Fu Y, Xue J, Yuan F, Qu T, Rissanou AN, Wang Y, Li X, Hu H. Therapeutic stapled peptides: Efficacy and molecular targets. Pharmacol Res 2024; 203:107137. [PMID: 38522761 DOI: 10.1016/j.phrs.2024.107137] [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: 12/06/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
Peptide stapling, by employing a stable, preformed alpha-helical conformation, results in the production of peptides with improved membrane permeability and enhanced proteolytic stability, compared to the original peptides, and provides an effective solution to accelerate the rapid development of peptide drugs. Various reviews present peptide stapling chemistries, anchoring residues and one- or two-component cyclization, however, therapeutic stapled peptides have not been systematically summarized, especially focusing on various disease-related targets. This review highlights the latest advances in therapeutic peptide drug development facilitated by the application of stapling technology, including different stapling techniques, synthetic accessibility, applicability to biological targets, potential for solving biological problems, as well as the current status of development. Stapled peptides as therapeutic drug candidates have been classified and analysed mainly by receptor- and ligand-based stapled peptide design against various diseases, including cancer, infectious diseases, inflammation, and diabetes. This review is expected to provide a comprehensive reference for the rational design of stapled peptides for different diseases and targets to facilitate the development of therapeutic peptides with enhanced pharmacokinetic and biological properties.
Collapse
Affiliation(s)
- Yulei Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| | - Minghao Wu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yinxue Fu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jingwen Xue
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fei Yuan
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Tianci Qu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Anastassia N Rissanou
- Theoretical & Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, Athens 11635, Greece
| | - Yilin Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 131 Dong'an Road, Shanghai 200032, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| | - Honggang Hu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China.
| |
Collapse
|
5
|
Kalogriopoulos NA, Tei R, Yan Y, Ravalin M, Li Y, Ting A. Synthetic G protein-coupled receptors for programmable sensing and control of cell behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589622. [PMID: 38659921 PMCID: PMC11042292 DOI: 10.1101/2024.04.15.589622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Synthetic receptors that mediate antigen-dependent cell responses are transforming therapeutics, drug discovery, and basic research. However, established technologies such as chimeric antigen receptors (CARs) can only detect immobilized antigens, have limited output scope, and lack built-in drug control. Here, we engineer synthetic G protein-coupled receptors (GPCRs) capable of driving a wide range of native or nonnative cellular processes in response to user-defined antigen. We achieve modular antigen gating by engineering and fusing a conditional auto-inhibitory domain onto GPCR scaffolds. Antigen binding to a fused nanobody relieves auto-inhibition and enables receptor activation by drug, thus generating Programmable Antigen-gated G protein-coupled Engineered Receptors (PAGERs). We create PAGERs responsive to more than a dozen biologically and therapeutically important soluble and cell surface antigens, in a single step, from corresponding nanobody binders. Different PAGER scaffolds permit antigen binding to drive transgene expression, real-time fluorescence, or endogenous G protein activation, enabling control of cytosolic Ca 2+ , lipid signaling, cAMP, and neuronal activity. Due to its modular design and generalizability, we expect PAGER to have broad utility in discovery and translational science.
Collapse
|
6
|
Didik S, Golosova D, Xu B, Staruschenko A. Opioids and the Kidney: A Compendium. KIDNEY360 2023; 4:1816-1823. [PMID: 37927032 PMCID: PMC10758516 DOI: 10.34067/kid.0000000000000291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Opioids are a class of medications used in pain management. Unfortunately, long-term use, overprescription, and illicit opioid use have led to one of the greatest threats to mankind: the opioid crisis. Accompanying the classical analgesic properties of opioids, opioids produce a myriad of effects including euphoria, immunosuppression, respiratory depression, and organ damage. It is essential to ascertain the physiological role of the opioid/opioid receptor axis to gain an in-depth understanding of the effects of opioid use. This knowledge will aid in the development of novel therapeutic interventions to combat the increasing mortality rate because of opioid misuse. This review describes the current knowledge of opioids, including the opioid epidemic and opioid/opioid receptor physiology. Furthermore, this review intricately relates opioid use to kidney damage, navigates kidney structure and physiology, and proposes potential ways to prevent opioid-induced kidney damage.
Collapse
Affiliation(s)
- Steven Didik
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- James A. Haley Veteran's Hospital, Tampa, Florida
| | - Daria Golosova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Biyang Xu
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- James A. Haley Veteran's Hospital, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
| |
Collapse
|
7
|
Rehrauer KJ, Cunningham CW. IUPHAR Review - Bivalent and bifunctional opioid receptor ligands as novel analgesics. Pharmacol Res 2023; 197:106966. [PMID: 37865129 DOI: 10.1016/j.phrs.2023.106966] [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: 08/25/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Though efficacious in managing chronic, severe pain, opioid analgesics are accompanied by significant adverse effects including constipation, tolerance, dependence, and respiratory depression. The life-threatening risks associated with µ opioid receptor agonist-based analgesics challenges their use in clinic. A rational approach to combatting these adverse effects is to develop agents that incorporate activity at a second pharmacologic target in addition to µ opioid receptor activation. The promise of such bivalent or bifunctional ligands is the development of an analgesic with an improved side effect profile. In this review, we highlight ongoing efforts in the development of bivalent and bifunctional analgesics that combine µ agonism with efficacy at κ and δ opioid receptors, the nociceptin opioid peptide (NOP) receptor, σ receptors, and cannabinoid receptors. Several examples of bifunctional analgesics in preclinical and clinical development are highlighted, as are strategies being employed toward the rational design of novel agents.
Collapse
Affiliation(s)
- Kyle J Rehrauer
- Department of Pharmaceutical and Administrative Sciences, Concordia University Wisconsin School of Pharmacy, 12800 N. Lake Shore Drive, Mequon, WI 53092, USA
| | - Christopher W Cunningham
- Department of Pharmaceutical and Administrative Sciences, Concordia University Wisconsin School of Pharmacy, 12800 N. Lake Shore Drive, Mequon, WI 53092, USA; CUW Center for Structure-Based Drug Discovery and Development, Concordia University Wisconsin School of Pharmacy, 12800 N. Lake Shore Drive, Mequon, WI 53092, USA.
| |
Collapse
|
8
|
Collier AD, Abdulai AR, Leibowitz SF. Utility of the Zebrafish Model for Studying Neuronal and Behavioral Disturbances Induced by Embryonic Exposure to Alcohol, Nicotine, and Cannabis. Cells 2023; 12:2505. [PMID: 37887349 PMCID: PMC10605371 DOI: 10.3390/cells12202505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
It is estimated that 5% of pregnant women consume drugs of abuse during pregnancy. Clinical research suggests that intake of drugs during pregnancy, such as alcohol, nicotine and cannabis, disturbs the development of neuronal systems in the offspring, in association with behavioral disturbances early in life and an increased risk of developing drug use disorders. After briefly summarizing evidence in rodents, this review focuses on the zebrafish model and its inherent advantages for studying the effects of embryonic exposure to drugs of abuse on behavioral and neuronal development, with an emphasis on neuropeptides known to promote drug-related behaviors. In addition to stimulating the expression and density of peptide neurons, as in rodents, zebrafish studies demonstrate that embryonic drug exposure has marked effects on the migration, morphology, projections, anatomical location, and peptide co-expression of these neurons. We also describe studies using advanced methodologies that can be applied in vivo in zebrafish: first, to demonstrate a causal relationship between the drug-induced neuronal and behavioral disturbances and second, to discover underlying molecular mechanisms that mediate these effects. The zebrafish model has great potential for providing important information regarding the development of novel and efficacious therapies for ameliorating the effects of early drug exposure.
Collapse
Affiliation(s)
| | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
9
|
Zhu Y, Wang K, Ma T, Ji Y, Lou Y, Fu X, Lu Y, Liu Y, Dang W, Zhang Q, Yin F, Wang K, Yu B, Zhang H, Lai J, Wang Y. Nucleus accumbens D1/D2 circuits control opioid withdrawal symptoms in mice. J Clin Invest 2023; 133:e163266. [PMID: 37561576 PMCID: PMC10503809 DOI: 10.1172/jci163266] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
The nucleus accumbens (NAc) is the most promising target for drug use disorder treatment. Deep brain stimulation (DBS) of NAc is effective for drug use disorder treatment. However, the mechanisms by which DBS produces its therapeutic effects remain enigmatic. Here, we define a behavioral cutoff criterion to distinguish depressive-like behaviors and non-depressive-like behaviors in mice after morphine withdrawal. We identified a basolateral amygdala (BLA) to NAc D1 medium spiny neuron (MSN) pathway that controls depressive-like behaviors after morphine withdrawal. Furthermore, the paraventricular nucleus of thalamus (PVT) to NAc D2 MSN pathway controls naloxone-induced acute withdrawal symptoms. Optogenetically induced long-term potentiation with κ-opioid receptor (KOR) antagonism enhanced BLA to NAc D1 MSN signaling and also altered the excitation/inhibition balance of NAc D2 MSN signaling. We also verified that a new 50 Hz DBS protocol reversed morphine withdrawal-evoked abnormal plasticity in NAc. Importantly, this refined DBS treatment effectively alleviated naloxone-induced withdrawal symptoms and depressive-like behaviors and prevented stress-induced reinstatement. Taken together, the results demonstrated that input- and cell type-specific synaptic plasticity underlies morphine withdrawal, which may lead to novel targets for the treatment of opioid use disorder.
Collapse
Affiliation(s)
- Yongsheng Zhu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Kejia Wang
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Tengfei Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuanyuan Ji
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Yin Lou
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Xiaoyu Fu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Ye Lu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Yige Liu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Wei Dang
- The Sixth Ward, Xi’an Mental Health Center, Xi’an, China
| | - Qian Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Fangyuan Yin
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Kena Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Bing Yu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hongbo Zhang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Jianghua Lai
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
| | - Yunpeng Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, National Biosafety Evidence Foundation, Xi’an Jiaotong University, Xi’an, China
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
10
|
Muratspahić E, White AM, Ciotu CI, Hochrainer N, Tomašević N, Koehbach J, Lewis RJ, Spetea M, Fischer MJM, Craik DJ, Gruber CW. Development of a Selective Peptide κ-Opioid Receptor Antagonist by Late-Stage Functionalization with Cysteine Staples. J Med Chem 2023; 66:11843-11854. [PMID: 37632447 PMCID: PMC10510397 DOI: 10.1021/acs.jmedchem.3c00426] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Indexed: 08/28/2023]
Abstract
The κ-opioid receptor (KOR) is an attractive target for the development of novel drugs. KOR agonists are potentially safer pain medications, whereas KOR antagonists are promising drug candidates for the treatment of neuropsychiatric disorders. Hitherto, the vast majority of selective drug leads that have been developed for KOR are small molecules. In this study, novel peptide probes were designed by using an endogenous dynorphin A1-13 sequence as a template for peptide stapling via late-stage cysteine functionalization. Leveraging this strategy, we developed a stable and potent KOR antagonist, CSD-CH2(1,8)-NH2, with approximately 1000-fold improved selectivity for KOR over μ- and δ-opioid receptors. Its potent competitive KOR antagonism was verified in KOR-expressing cells, peripheral dorsal root ganglion neurons, and using the tail-flick and rotarod tests in mice. This work highlights the value of cysteine stapling to develop selective peptide probes to modulate central KOR function, as innovative peptide drug candidates for the treatment of KOR-related illnesses.
Collapse
Affiliation(s)
- Edin Muratspahić
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Andrew M. White
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Cosmin I. Ciotu
- Center
for Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nadine Hochrainer
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Nataša Tomašević
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes Koehbach
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Richard J. Lewis
- Institute
for Molecular Bioscience, The University
of Queensland, 4072 Brisbane, Queensland, Australia
| | - Mariana Spetea
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Michael J. M. Fischer
- Center
for Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, 1090 Vienna, Austria
| | - David J. Craik
- Institute
for Molecular Bioscience, Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Christian W. Gruber
- Center
for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
11
|
Pinheiro PDSM, Franco LS, Fraga CAM. The Magic Methyl and Its Tricks in Drug Discovery and Development. Pharmaceuticals (Basel) 2023; 16:1157. [PMID: 37631072 PMCID: PMC10457765 DOI: 10.3390/ph16081157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/06/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
One of the key scientific aspects of small-molecule drug discovery and development is the analysis of the relationship between its chemical structure and biological activity. Understanding the effects that lead to significant changes in biological activity is of paramount importance for the rational design and optimization of bioactive molecules. The "methylation effect", or the "magic methyl" effect, is a factor that stands out due to the number of examples that demonstrate profound changes in either pharmacodynamic or pharmacokinetic properties. In many cases, this has been carried out rationally, but in others it has been the product of serendipitous observations. This paper summarizes recent examples that provide an overview of the current state of the art and contribute to a better understanding of the methylation effect in bioactive small-molecule drug candidates.
Collapse
Affiliation(s)
- Pedro de Sena Murteira Pinheiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (P.d.S.M.P.); (L.S.F.)
| | - Lucas Silva Franco
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (P.d.S.M.P.); (L.S.F.)
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), CCS, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (P.d.S.M.P.); (L.S.F.)
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), CCS, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
12
|
Yasmin N, Collier AD, Karatayev O, Abdulai AR, Yu B, Fam M, Khalizova N, Leibowitz SF. Subpopulations of hypocretin/orexin neurons differ in measures of their cell proliferation, dynorphin co-expression, projections, and response to embryonic ethanol exposure. Sci Rep 2023; 13:8448. [PMID: 37231149 PMCID: PMC10213024 DOI: 10.1038/s41598-023-35432-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
Numerous studies in animals demonstrate that embryonic exposure to ethanol (EtOH) at low-moderate doses stimulates neurogenesis and increases the number of hypothalamic neurons expressing the peptide, hypocretin/orexin (Hcrt). A recent study in zebrafish showed that this effect on the Hcrt neurons in the anterior hypothalamus (AH) is area specific, evident in the anterior (aAH) but not posterior (pAH) part of this region. To understand specific factors that may determine the differential sensitivity to EtOH of these Hcrt subpopulations, we performed additional measures in zebrafish of their cell proliferation, co-expression of the opioid dynorphin (Dyn), and neuronal projections. In association with the increase in Hcrt neurons in the aAH but not pAH, EtOH significantly increased only in the aAH the proliferation of Hcrt neurons and their number lacking Dyn co-expression. The projections of these subpopulations differed markedly in their directionality, with those from the pAH primarily descending to the locus coeruleus and those from the aAH ascending to the subpallium, and they were both stimulated by EtOH, which induced specifically the most anterior subpallium-projecting Hcrt neurons to become ectopically expressed beyond the aAH. These differences between the Hcrt subpopulations suggest they are functionally distinct in their regulation of behavior.
Collapse
Affiliation(s)
- Nushrat Yasmin
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Adam D Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Olga Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Abdul R Abdulai
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Boyi Yu
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Milisia Fam
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Nailya Khalizova
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
13
|
Recent Advances in the Synthesis and Applications of m-Aryloxy Phenols. Molecules 2023; 28:molecules28062657. [PMID: 36985628 PMCID: PMC10056990 DOI: 10.3390/molecules28062657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Since phenol derivatives have high potential as building blocks for the synthesis of bioactive natural products and conducting polymers, many synthesis methods have been invented. In recent years, innovative synthetic methods have been developed for the preparation of m-aryloxy phenols, which has allowed for the preparation of complex m-aryloxy phenols with functional groups, such as esters, nitriles, and halogens, that impart specific properties of these compounds. This review provides an overview of recent advances in synthetic strategies for m-aryloxy phenols and their potential biological activities. This paper highlights the importance of m-aryloxy phenols in various industries, including plastics, adhesives, and coatings, and it discusses their applications as antioxidants, ultraviolet absorbers, and flame retardants.
Collapse
|
14
|
Zhao J, Carbone J, Farruggia G, Janecka A, Gentilucci L, Calonghi N. Synthesis and Antiproliferative Activity against Cancer Cells of Indole-Aryl-Amide Derivatives. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010265. [PMID: 36615458 PMCID: PMC9822155 DOI: 10.3390/molecules28010265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
Indoles constitute a large family of heterocyclic compounds widely occurring in nature which are present in a number of bioactive natural and synthetic compounds, including anticancer agents or atypical opioid agonists. As a result, exponential increases in the development of novel methods for the synthesis of indole-containing compounds have been reported in the literature. A series of indole-aryl amide derivatives 1-7 containing tryptamine or an indolylacetic acid nucleus were designed, synthesized, and evaluated as opioid ligands. These new indole derivatives showed negligible to very low affinity for μ- and δ-opioid receptor (OR). On the other hand, compounds 2, 5 and 7 showed Ki values in the low μM range for κ-OR. Since indoles are well known for their anticancer potential, their effect against a panel of tumor cell lines was tested. The target compounds were evaluated for their in vitro cytotoxicity in HT29, HeLa, IGROV-1, MCF7, PC-3, and Jurkat J6 cells. Some of the synthesized compounds showed good activity against the selected tumor cell lines, with the exception of IGROV1. In particular, compound 5 showed a noteworthy selectivity towards HT29 cells, a malignant colonic cell line, without affecting healthy human intestinal cells. Further studies revealed that 5 caused the cell cycle arrest in the G1 phase and promoted apoptosis in HT29 cells.
Collapse
Affiliation(s)
- Junwei Zhao
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Jacopo Carbone
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giovanna Farruggia
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- Correspondence: (L.G.); (N.C.); Tel.: +39-05-1209-9570 (L.G.); +39-05-1209-1231 (N.C.)
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
- Correspondence: (L.G.); (N.C.); Tel.: +39-05-1209-9570 (L.G.); +39-05-1209-1231 (N.C.)
| |
Collapse
|
15
|
Cippitelli A, Zribi G, Toll L. PPL-103: A mixed opioid partial agonist with desirable anti-cocaine properties. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110599. [PMID: 35798174 DOI: 10.1016/j.pnpbp.2022.110599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022]
Abstract
Cocaine use disorder (CUD) is a persistent public health problem for which no effective medications are available. PPL-103 is an opioid receptor ligand with partial agonist activity at mu, kappa and delta opioid receptors, with a greater efficacy for kappa and low efficacy at mu receptors. Because chronic cocaine use induces changes in the kappa opioid receptor/dynorphin system, we hypothesized that a kappa partial agonist, such as PPL-103, would attenuate the aversive properties of the upregulated kappa system, resulting in effective treatment approach for CUD. We tested the effects of PPL-103 on cocaine self-administration models that recapitulate core aspects of CUD in humans. We found that PPL-103 reduced both long and short access cocaine self-administration, motivation to respond for cocaine, and binge-like cocaine taking, in rats. Operant responding for food, fentanyl and locomotor behavior were not altered at doses that decreased cocaine infusions. Repeated PPL-103 treatment did not lead to tolerance development. PPL-103 also reduced both priming- and cue-induced reinstatement of cocaine seeking, being more effective in the former. Surprisingly, PPL-103 reduced self-administration parameters and reinstatement in rats previously treated with the long-acting kappa receptor antagonist JDTic more potently than in non-JDTic treated animals, whereas naltrexone injected to rats subsequent to JDTic administration increased self-administration, suggesting that the partial mu agonist activity, rather than kappa agonism is important for reduction in cocaine taking and seeking. However, partial kappa activation seems to increase safety by limiting dysphoria, tolerance and addiction development. PPL-103 displays a desirable profile as a possible CUD pharmacotherapy.
Collapse
Affiliation(s)
- Andrea Cippitelli
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States.
| | - Gilles Zribi
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Lawrence Toll
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
16
|
Ji MJ, Gao ZQ, Yang J, Cai JH, Li KX, Wang J, Zhang H, Zhou CH, Cao JL, Liu C. Dynorphin promotes stress-induced depressive behaviors by inhibiting ventral pallidal neurons in rats. Acta Physiol (Oxf) 2022; 236:e13882. [PMID: 36039689 DOI: 10.1111/apha.13882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 01/29/2023]
Abstract
AIM Endogenous dynorphin signaling via kappa opioid receptors (KORs) plays a key role in producing the depressive and aversive consequences of stress. We investigated the behavioral effects of the dynorphin/KOR system in the ventral pallidum (VP) and studied the underlying mechanisms. METHODS To investigate the effects of dynorphin on the VP, we conducted behavioral experiments after microinjection of drugs or shRNA and brain-slice electrophysiological recordings. Histological tracing and molecular biological experiments were used to identify the distribution of KORs and the possible sources of dynorphin projections to the VP. RESULTS An elevated dynorphin concentration and increased KOR activity were observed in the VP after acute stress. Infusion of dynorphin-A into the VP produced depressive-like phenotypes including anhedonia and despair and anxiety behaviors, but did not alter locomotor behavior. Mechanistically, dynorphin had an inhibitory effect on VP neurons-reducing their firing rate and inhibiting excitatory transmission-through direct activation of KORs and modulation of downstream G-protein-gated inwardly rectifying potassium (GIRK) channels and high-voltage gated calcium channels (VGCCs). Tracing revealed direct innervation of VP neurons by dynorphin-positive projections; potential sources of these dynorphinergic projections include the nucleus accumbens, amygdala, and hypothalamus. Blockade of dynorphin/KOR signaling in the VP by drugs or viral knock-down of KORs significantly reduced despair behavior in rats. CONCLUSIONS Endogenous dynorphinergic modulation of the VP plays a critical role in mediating depressive reactions to stress.
Collapse
Affiliation(s)
- Miao-Jin Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Zhi-Qiang Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Jiao Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Ji-Heng Cai
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Ke-Xue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Jie Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Hua Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Chao Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
17
|
Mousavifar L, Lewicky JD, Taponard A, Bagul R, Rivat M, Abdullayev S, Martel AL, Fraleigh NL, Nakamura A, Veyrier FJ, Le HT, Roy R. Synthesis & Evaluation of Novel Mannosylated Neoglycolipids for Liposomal Delivery System Applications. Pharmaceutics 2022; 14:2300. [PMID: 36365120 PMCID: PMC9692915 DOI: 10.3390/pharmaceutics14112300] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 07/03/2024] Open
Abstract
Glycosylated NPs, including liposomes, are known to target various receptors involved in cellular carbohydrate transport, of which the mannoside binding receptors are attracting particular attention for their expression on various immune cells, cancers, and cells involved in maintaining central nervous system (CNS) integrity. As part of our interest in NP drug delivery, mannosylated glycoliposomal delivery systems formed from the self-assembly of amphiphilic neoglycolipids were developed, with a C12-alkyl mannopyranoside (ML-C12) being identified as a lead compoundcapable of entrapping, protecting, and improving the delivery of structurally diverse payloads. However, ML-C12 was not without limitations in both the synthesis of the glycolipids, and the physicochemical properties of the resulting glycoliposomes. Herein, the chemical syntheses of a novel series of mannosylated neoglycolipids are reported with the goal of further improving on the previous ML-C12 glyconanoparticles. The current work aimed to use a self-contingent strategy which overcomes previous synthetic limitations to produce neoglycolipids that have one exposed mannose residue, an aromatic scaffold, and two lipid tails with varied alkyl chains. The azido-ending carbohydrates and the carboxylic acid-ending lipid tails were ligated using a new one-pot modified Staudinger chemistry that differed advantageously to previous syntheses. The formation of stable neoglycoliposomes of controllable and ideal sizes (≈100-400 nm) was confirmed via dynamic light scattering (DLS) experiments and transmission electron microscopy (TEM). Beyond chemical advantages, the present study further aimed to establish potential improvements in the biological activity of the neoglycoliposomes. Concanavalin A (Con A) agglutination studies demonstrated efficient and stable cross-linking abilities dependent on the length of the linkers and lipid tails. The efficacy of the glycoliposomes in improving cytosolic uptake was investigated using Nile Red as probe in immune and cancer cell lines. Preliminary ex vivo safety assessments showed that the mannosylated glycoliposomes are hemocompatible, and non-immunogenic. Finally, using a model peptide therapeutic, the relative entrapment capacity and plasma stability of the optimal glycoliposome delivery system was evaluated and compared to the previous neoglycoliposomes. Overall, the new lead glycoliposome showed improved biological activity over ML-C12, in addition to having several chemical benefits including the lack of stereocenters, a longer linker allowing better sugar availability, and ease of synthesis using novel one-pot modified Staudinger chemistry.
Collapse
Affiliation(s)
- Leila Mousavifar
- Glycosciences and Nanomaterial Laboratory, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, QC H3C 3P8, Canada
| | - Jordan D. Lewicky
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, ON P3E 2H2, Canada
| | - Alexis Taponard
- Glycosciences and Nanomaterial Laboratory, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, QC H3C 3P8, Canada
| | - Rahul Bagul
- Glycosciences and Nanomaterial Laboratory, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, QC H3C 3P8, Canada
| | - Madleen Rivat
- Glycosciences and Nanomaterial Laboratory, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, QC H3C 3P8, Canada
| | - Shuay Abdullayev
- Glycosciences and Nanomaterial Laboratory, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, QC H3C 3P8, Canada
| | - Alexandrine L. Martel
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, ON P3E 2H2, Canada
| | - Nya L. Fraleigh
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, ON P3E 2H2, Canada
| | - Arnaldo Nakamura
- Armand-Frappier Santé Biotechnologie Research Centre, Institut National de la Recherche Scientifique, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada
| | - Frédéric J. Veyrier
- Armand-Frappier Santé Biotechnologie Research Centre, Institut National de la Recherche Scientifique, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada
| | - Hoang-Thanh Le
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, ON P3E 2H2, Canada
- Medicinal Sciences Division, NOSM University, 935 Ramsey Lake Road, Sudbury, ON P3E 2C6, Canada
| | - René Roy
- Glycosciences and Nanomaterial Laboratory, Université du Québec à Montréal, P.O. Box 8888, Succ. Centre-Ville, Montréal, QC H3C 3P8, Canada
| |
Collapse
|
18
|
Khan MIH, Sawyer BJ, Akins NS, Le HV. A systematic review on the kappa opioid receptor and its ligands: New directions for the treatment of pain, anxiety, depression, and drug abuse. Eur J Med Chem 2022; 243:114785. [PMID: 36179400 DOI: 10.1016/j.ejmech.2022.114785] [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: 07/08/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
Kappa opioid receptor (KOR) is a member of the opioid receptor system, the G protein-coupled receptors that are expressed throughout the peripheral and central nervous systems and play crucial roles in the modulation of antinociception and a variety of behavioral states like anxiety, depression, and drug abuse. KOR agonists are known to produce potent analgesic effects and have been used clinically for the treatment of pain, while KOR antagonists have shown efficacy in the treatment of anxiety and depression. This review summarizes the history, design strategy, discovery, and development of KOR ligands. KOR agonists are classified as non-biased, G protein-biased, and β-arrestin recruitment-biased, according to their degrees of bias. The mechanisms and associated effects of the G protein signaling pathway and β-arrestin recruitment signaling pathway are also discussed. Meanwhile, KOR antagonists are classified as long-acting and short-acting, based on their half-lives. In addition, we have special sections for mixed KOR agonists and selective peripheral KOR agonists. The mechanisms of action and pharmacokinetic, pharmacodynamic, and behavioral studies for each of these categories are also discussed in this review.
Collapse
Affiliation(s)
- Md Imdadul H Khan
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Benjamin J Sawyer
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Nicholas S Akins
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Hoang V Le
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
19
|
Wedemeyer MJ, Jennings EM, Smith HR, Chavera TS, Jamshidi RJ, Berg KA, Clarke WP. 14-3-3γ mediates the long-term inhibition of peripheral kappa opioid receptor antinociceptive signaling by norbinaltorphimine. Neuropharmacology 2022; 220:109251. [PMID: 36126728 DOI: 10.1016/j.neuropharm.2022.109251] [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: 06/02/2022] [Revised: 08/16/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
Long-term inhibition of kappa opioid receptor (KOR) signaling in peripheral pain-sensing neurons is a potential obstacle for development of peripherally-restricted KOR agonists that produce analgesia. Such a long-term inhibitory mechanism is invoked from activation of c-Jun N-terminal kinase (JNK) that follows a single injection of the KOR antagonist norbinaltorphimine (norBNI). This effect requires protein synthesis of an unknown mediator in peripheral pain-sensing neurons. Using 2D difference gel electrophoresis with tandem mass spectrometry, we have identified that the scaffolding protein 14-3-3γ is upregulated in peripheral sensory neurons following activation of JNK with norBNI. Knockdown of 14-3-3γ by siRNA eliminates the long-term reduction in KOR-mediated cAMP signaling by norBNI in peripheral sensory neurons in culture. Similarly, knockdown of 14-3-3γ in the rat hind paw abolished the norBNI-mediated long-term reduction in peripheral KOR-mediated antinociception. Further, overexpression of 14-3-3γ in KOR expressing CHO cells prevented KOR-mediated inhibition of cAMP signaling. These long-term effects are selective for KOR as heterologous regulation of other receptor systems was not observed. These data suggest that 14-3-3γ is both necessary and sufficient for the long-term inhibition of KOR by norBNI in peripheral sensory neurons.
Collapse
Affiliation(s)
- Michael J Wedemeyer
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Elaine M Jennings
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hudson R Smith
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Teresa S Chavera
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Raehannah J Jamshidi
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kelly A Berg
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - William P Clarke
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
20
|
Deji C, Yan P, Ji Y, Yan X, Feng Y, Liu J, Liu Y, Wei S, Zhu Y, Lai J. The Basolateral Amygdala to Ventral Hippocampus Circuit Controls Anxiety-Like Behaviors Induced by Morphine Withdrawal. Front Cell Neurosci 2022; 16:894886. [PMID: 35726232 PMCID: PMC9205755 DOI: 10.3389/fncel.2022.894886] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/26/2022] [Indexed: 12/02/2022] Open
Abstract
Anxiety is one of the most common comorbid conditions reported in people with opioid dependence. The basolateral amygdala (BLA) and ventral hippocampus (vHip) are critical brain regions for fear and anxiety. The kappa opioid receptor (KOR) is present in the mesolimbic regions involved in emotions and addiction. However, the precise circuits and molecular basis underlying anxiety associated with chronic opioid use are poorly understood. Using a mouse model, we demonstrated that anxiety-like behaviors appeared in the first 2 weeks after morphine withdrawal. Furthermore, the BLA and vHip were activated in mice experiencing anxiety after morphine withdrawal (Mor-A). KORs in the BLA to vHip projections were significantly increased in the Mor-A group. Optogenetic/chemogenetic inhibition of BLA inputs ameliorated anxiety-like behaviors and facilitated conditioned place preference (CPP) extinction in Mor-A mice. Knockdown of the BLA to vHip circuit KOR alleviated the anxiety-like behaviors but did not affect CPP extinction or reinstatement. Furthermore, combined treatment of inhibition of the BLA to vHip circuit and KOR antagonists mitigated anxiety-like behaviors and prevented stress-induced CPP reinstatement after morphine withdrawal. These results revealed a previously unknown circuit associated with the emotional component of opioid withdrawal and indicated that restoration of synaptic deficits with KOR antagonists might be effective in the treatment of anxiety associated with morphine withdrawal.
Collapse
|
21
|
Brice-Tutt AC, Eans SO, Yakovlev D, Aldrich JV, McLaughlin JP. An analog of [d-Trp]CJ-15,208 exhibits kappa opioid receptor antagonism following oral administration and prevents stress-induced reinstatement of extinguished morphine conditioned place preference. Pharmacol Biochem Behav 2022; 217:173405. [PMID: 35584724 DOI: 10.1016/j.pbb.2022.173405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/26/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Opioid use disorder (OUD) relapse rates are discouragingly high, underscoring the need for new treatment options. The macrocyclic tetrapeptide natural product CJ-15,208 and its stereoisomer [d-Trp]CJ-15,208 demonstrate kappa opioid receptor (KOR) antagonist activity upon oral administration which prevents stress-induced reinstatement of cocaine-seeking behavior. In order to further explore the structure-activity relationships and expand the potential therapeutic applications of KOR antagonism for the treatment of OUD, we screened a series of 24 analogs of [d-Trp]CJ-15,208 with the goal of enhancing KOR antagonist activity. From this screening, analog 22 arose as a compound of interest, demonstrating dose-dependent KOR antagonism after central and oral administration lasting at least 2.5 h. In further oral testing, analog 22 lacked respiratory, locomotor, or reinforcing effects, consistent with the absence of opioid agonism. Pretreatment with analog 22 (30 mg/kg, p.o.) prevented stress-induced reinstatement of extinguished morphine conditioned place preference and reduced some signs of naloxone-precipitated withdrawal in mice physically dependent on morphine. Collectively, these data support the therapeutic potential of KOR antagonists to support abstinence in OUD and ameliorate opioid withdrawal.
Collapse
Affiliation(s)
- Ariana C Brice-Tutt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Dmitry Yakovlev
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America.
| |
Collapse
|
22
|
Puls K, Olivé-Marti AL, Pach S, Pinter B, Erli F, Wolber G, Spetea M. In Vitro, In Vivo and In Silico Characterization of a Novel Kappa-Opioid Receptor Antagonist. Pharmaceuticals (Basel) 2022; 15:680. [PMID: 35745598 PMCID: PMC9229160 DOI: 10.3390/ph15060680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Kappa-opioid receptor (KOR) antagonists are promising innovative therapeutics for the treatment of the central nervous system (CNS) disorders. The new scaffold opioid ligand, Compound A, was originally found as a mu-opioid receptor (MOR) antagonist but its binding/selectivity and activation profile at the KOR and delta-opioid receptor (DOR) remain elusive. In this study, we present an in vitro, in vivo and in silico characterization of Compound A by revealing this ligand as a KOR antagonist in vitro and in vivo. In the radioligand competitive binding assay, Compound A bound at the human KOR, albeit with moderate affinity, but with increased affinity than to the human MOR and without specific binding at the human DOR, thus displaying a preferential KOR selectivity profile. Following subcutaneous administration in mice, Compound A effectively reverse the antinociceptive effects of the prototypical KOR agonist, U50,488. In silico investigations were carried out to assess the structural determinants responsible for opioid receptor subtype selectivity of Compound A. Molecular docking, molecular dynamics simulations and dynamic pharmacophore (dynophore) generation revealed differences in the stabilization of the chlorophenyl moiety of Compound A within the opioid receptor binding pockets, rationalizing the experimentally determined binding affinity values. This new chemotype bears the potential for favorable ADMET properties and holds promise for chemical optimization toward the development of potential therapeutics.
Collapse
Affiliation(s)
- Kristina Puls
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany; (K.P.); (S.P.)
| | - Aina-Leonor Olivé-Marti
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| | - Szymon Pach
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany; (K.P.); (S.P.)
| | - Birgit Pinter
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| | - Filippo Erli
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| | - Gerhard Wolber
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany; (K.P.); (S.P.)
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (A.-L.O.-M.); (B.P.); (F.E.)
| |
Collapse
|
23
|
Leconte C, Mongeau R, Noble F. Traumatic Stress-Induced Vulnerability to Addiction: Critical Role of the Dynorphin/Kappa Opioid Receptor System. Front Pharmacol 2022; 13:856672. [PMID: 35571111 PMCID: PMC9091501 DOI: 10.3389/fphar.2022.856672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Substance use disorders (SUD) may emerge from an individual’s attempt to limit negative affective states and symptoms linked to stress. Indeed, SUD is highly comorbid with chronic stress, traumatic stress, or post-traumatic stress disorder (PTSD), and treatments approved for each pathology individually often failed to have a therapeutic efficiency in such comorbid patients. The kappa-opioid receptor (KOR) and its endogenous ligand dynorphin (DYN), seem to play a key role in the occurrence of this comorbidity. The DYN/KOR function is increased either in traumatic stress or during drug use, dependence acquisition and DYN is released during stress. The behavioural effects of stress related to the DYN/KOR system include anxiety, dissociative and depressive symptoms, as well as increased conditioned fear response. Furthermore, the DYN/KOR system is implicated in negative reinforcement after the euphoric effects of a drug of abuse ends. During chronic drug consumption DYN/KOR functions increase and facilitate tolerance and dependence. The drug-seeking behaviour induced by KOR activation can be retrieved either during the development of an addictive behaviour, or during relapse after withdrawal. DYN is known to be one of the most powerful negative modulators of dopamine signalling, notably in brain structures implicated in both reward and fear circuitries. KOR are also acting as inhibitory heteroreceptors on serotonin neurons. Moreover, the DYN/KOR system cross-regulate with corticotropin-releasing factor in the brain. The sexual dimorphism of the DYN/KOR system could be the cause of the gender differences observed in patients with SUD or/and traumatic stress-related pathologies. This review underlies experimental and clinical results emphasizing the DYN/KOR system as common mechanisms shared by SUD or/and traumatic stress-related pathologies, and suggests KOR antagonist as a new pharmacological strategy to treat this comorbidity.
Collapse
|
24
|
Sturaro C, Malfacini D, Argentieri M, Djeujo FM, Marzola E, Albanese V, Ruzza C, Guerrini R, Calo’ G, Molinari P. Pharmacology of Kappa Opioid Receptors: Novel Assays and Ligands. Front Pharmacol 2022; 13:873082. [PMID: 35529436 PMCID: PMC9068900 DOI: 10.3389/fphar.2022.873082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
The present study investigated the in vitro pharmacology of the human kappa opioid receptor using multiple assays, including calcium mobilization in cells expressing chimeric G proteins, the dynamic mass redistribution (DMR) label-free assay, and a bioluminescence resonance energy transfer (BRET) assay that allows measurement of receptor interaction with G protein and β-arrestin 2. In all assays, dynorphin A, U-69,593, and [D-Pro10]dyn(1-11)-NH2 behaved as full agonists with the following rank order of potency [D-Pro10]dyn(1-11)-NH2 > dynorphin A ≥ U-69,593. [Dmt1,Tic2]dyn(1-11)-NH2 behaved as a moderate potency pure antagonist in the kappa-β-arrestin 2 interaction assay and as low efficacy partial agonist in the other assays. Norbinaltorphimine acted as a highly potent and pure antagonist in all assays except kappa-G protein interaction, where it displayed efficacy as an inverse agonist. The pharmacological actions of novel kappa ligands, namely the dynorphin A tetrameric derivative PWT2-Dyn A and the palmitoylated derivative Dyn A-palmitic, were also investigated. PWT2-Dyn A and Dyn A-palmitic mimicked dynorphin A effects in all assays showing similar maximal effects but 3–10 fold lower potency. In conclusion, in the present study, multiple in vitro assays for the kappa receptor have been set up and pharmacologically validated. In addition, PWT2-Dyn A and Dyn A-palmitic were characterized as potent full agonists; these compounds are worthy of further investigation in vivo for those conditions in which the activation of the kappa opioid receptor elicits beneficial effects e.g. pain and pruritus.
Collapse
Affiliation(s)
- Chiara Sturaro
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Davide Malfacini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- *Correspondence: Davide Malfacini,
| | - Michela Argentieri
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Francine M. Djeujo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Erika Marzola
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Valentina Albanese
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, Ferrara, Italy
- Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
- Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, Ferrara, Italy
| | - Girolamo Calo’
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Paola Molinari
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| |
Collapse
|
25
|
Ma H, Brust T, Frankowski KJ, Lovell KM, Cameron MD, Bohn LM, Aubé J. Advances in Sulfonamide Kappa Opioid Receptor Antagonists: Structural Refinement and Evaluation of CNS Clearance. ACS Chem Neurosci 2022; 13:1315-1332. [PMID: 35410469 DOI: 10.1021/acschemneuro.2c00140] [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] [Indexed: 11/29/2022] Open
Abstract
Focused modification of a sulfonamide-based kappa opioid receptor (KOR) antagonist series previously reported by this laboratory was investigated. A total of 32 analogues were prepared to explore linker replacement, constraint manipulation, and aryl group or amine substitution. All analogues were assayed for KOR antagonist activity, and the initial lead compound was assessed for in vivo CNS penetration. The most improved analogue possessed a 4-fold increase of potency (IC50 = 18.9 ± 4.4 nM) compared with the lead compound (IC50 = 83.5 ± 20 nM) from an earlier work. The initial lead compound was found to attain suitable brain levels and to possess a shorter clearance time than canonical KOR antagonists such as JDTic.
Collapse
Affiliation(s)
- Huiyong Ma
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, 125 Mason Farm Road, CB 7363, Chapel Hill, North Carolina 27599, United States
- Department of Medicinal Chemistry, University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Tarsis Brust
- Departments of Molecular Therapeutics and Neuroscience, The Scripps Research Institute, 130 Scripps Way, #2A2, Jupiter, Florida 33458, United States
| | - Kevin J Frankowski
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, 125 Mason Farm Road, CB 7363, Chapel Hill, North Carolina 27599, United States
- Department of Medicinal Chemistry, University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| | - Kimberly M Lovell
- Departments of Molecular Therapeutics and Neuroscience, The Scripps Research Institute, 130 Scripps Way, #2A2, Jupiter, Florida 33458, United States
| | - Michael D Cameron
- Department of Molecular Therapeutics, The Scripps Research Institute, 130 Scripps Way, #2A1, Jupiter, Florida 33458, United States
| | - Laura M Bohn
- Departments of Molecular Therapeutics and Neuroscience, The Scripps Research Institute, 130 Scripps Way, #2A2, Jupiter, Florida 33458, United States
| | - Jeffrey Aubé
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, 125 Mason Farm Road, CB 7363, Chapel Hill, North Carolina 27599, United States
- Department of Medicinal Chemistry, University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, United States
| |
Collapse
|
26
|
Yao C, Jiang X, Ye X, Xie T, Bai R. Antidepressant Drug Discovery and Development: Mechanism and Drug Design Based on Small Molecules. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Chuansheng Yao
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Xiaoying Jiang
- College of Material, Chemistry and Chemical Engineering Key Laboratory of Organosilicon Chemistry and Material Technology Ministry of Education, Hangzhou Normal University Hangzhou 311121 P.R. China
| | - Xiang‐Yang Ye
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Tian Xie
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| | - Renren Bai
- School of Pharmacy Hangzhou Normal University Hangzhou 311121 PR China
- Key Laboratory of Elemene Class Anti‐Cancer Chinese Medicine of Zhejiang Province Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province Collaborative Innovation Center of Chinese Medicines from Zhejiang Province Hangzhou Normal University Hangzhou 311121 PR China
| |
Collapse
|
27
|
Abraham AD, Casello SM, Land BB, Chavkin C. Optogenetic stimulation of dynorphinergic neurons within the dorsal raphe activate kappa opioid receptors in the ventral tegmental area and ablation of dorsal raphe prodynorphin or kappa receptors in dopamine neurons blocks stress potentiation of cocaine reward. ADDICTION NEUROSCIENCE 2022; 1. [PMID: 36176476 PMCID: PMC9518814 DOI: 10.1016/j.addicn.2022.100005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Behavioral stress exposure increases the risk of drug-taking in individuals with substance use disorders by mechanisms involving the dynorphins, which are the endogenous neuropeptides for the kappa opioid receptor (KOR). KOR agonists have been shown to encode dysphoria, aversion, and changes in reward valuation, and kappa opioid antagonists are in clinical development for treating substance use disorders. In this study, we confirmed that KORs were expressed in dopaminergic neurons in the ventral tegmental area (VTA) of male C57BL6/J mice. Genetic ablation of KORs from dopamine neurons blocked the potentiating effects of repeated forced swim stress on cocaine conditioned place preference (CPP). KOR activation inhibited dopamine neuron GCaMP6m calcium activity in VTA during swim stress and caused a rebound enhancement during the period after stress exposure. Transient optogenetic inhibition of VTA dopamine neurons with AAV5-DIO-SwiChR was acutely aversive in a real time place preference assay and blunted cocaine CPP when inhibition was administered concurrently with cocaine conditioning. However, when inhibition preceded cocaine conditioning by 30 min, cocaine CPP was enhanced. Retrograde tracing with CAV2-DIO-ZsGreen identified a population of prodynorphinCre neurons in the dorsal raphe nucleus (DRN) projecting to the VTA. Optogenetic stimulation of dynorphinergic neurons within the DRN by Channelrhodopsin2 activated KOR in VTA and ablation of prodynorphin blocked stress potentiation of cocaine CPP. Together, these studies demonstrate the presence of a dynorphin/KOR midbrain circuit that projects from the DRN to VTA and is involved in altering the dynamic response of dopamine neuron activity to enhance drug reward learning.
Collapse
|
28
|
Missig G, Fritsch EL, Mehta N, Damon ME, Jarrell EM, Bartlett AA, Carroll FI, Carlezon WA. Blockade of kappa-opioid receptors amplifies microglia-mediated inflammatory responses. Pharmacol Biochem Behav 2022; 212:173301. [PMID: 34826432 PMCID: PMC8748402 DOI: 10.1016/j.pbb.2021.173301] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 01/03/2023]
Abstract
Brain kappa-opioid receptors (KORs) are implicated in the pathophysiology of depressive and anxiety disorders, stimulating interest in the therapeutic potential of KOR antagonists. Research on KOR function has tended to focus on KOR-expressing neurons and pathways such as the mesocorticolimbic dopamine system. However, KORs are also expressed on non-neuronal cells including microglia, the resident immune cells in the brain. The effects of KOR antagonists on microglia are not understood despite the potential contributions of these cells to overall responsiveness to this class of drugs. Previous work in vitro suggests that KOR activation suppresses proinflammatory signaling mediated by immune cells including microglia. Here, we examined how KOR antagonism affects microglia function in vivo, together with its effects on physiological and behavioral responses to an immune challenge. Pretreatment with the prototypical KOR antagonist JDTic potentiates levels of proinflammatory cytokines (IL-1β, IL-6) in blood following administration of lipopolysaccharide (LPS), an immune-activating agent, without triggering effects on its own. Using magnetic-activated cell sorting (MACs), we found that KOR antagonism potentiates LPS-induced cytokine expression within microglia. This effect is accompanied by potentiation of LPS-induced hyperthermia, although reductions in body weight and locomotion were not affected. Histological analyses confirm that LPS produces visible changes in microglia morphology consistent with activation, but this effect is not altered by KOR antagonism. Considering that inflammation is increasingly implicated in depressive and anxiety disorders, these findings raise the possibility that KOR antagonist actions on microglia may detract from actions on neurons that contribute to their therapeutic potential.
Collapse
Affiliation(s)
- Galen Missig
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| | - Emma L. Fritsch
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| | - Niyati Mehta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| | - Miles E. Damon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| | - Erica M. Jarrell
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| | - Andrew A. Bartlett
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| | - F. Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, P. O. Box 12194, Research Triangle Park NC, 27709, USA
| | - William A. Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA, 02478, USA
| |
Collapse
|
29
|
Fundamentals of the Dynorphins/Kappa Opioid Receptor System: From Distribution to Signaling and Function. Handb Exp Pharmacol 2022; 271:3-21. [PMID: 33754230 PMCID: PMC9013522 DOI: 10.1007/164_2021_433] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This chapter provides a general introduction to the dynorphins (DYNs)/kappa opioid receptor (KOR) system, including DYN peptides, neuroanatomy of the DYNs/KOR system, cellular signaling, and in vivo behavioral effects of KOR activation and inhibition. It is intended to serve as a primer for the book and to provide a basic background for the chapters in the book.
Collapse
|
30
|
Aldrich JV, McLaughlin JP. Peptide Kappa Opioid Receptor Ligands and Their Potential for Drug Development. Handb Exp Pharmacol 2022; 271:197-220. [PMID: 34463847 DOI: 10.1007/164_2021_519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ligands for kappa opioid receptors (KOR) have potential uses as non-addictive analgesics and for the treatment of pruritus, mood disorders, and substance abuse. These areas continue to have major unmet medical needs. Significant advances have been made in recent years in the preclinical development of novel opioid peptides, notably ones with structural features that inherently impart stability to proteases. Following a brief discussion of the potential therapeutic applications of KOR agonists and antagonists, this review focuses on two series of novel opioid peptides, all-D-amino acid tetrapeptides as peripherally selective KOR agonists for the treatment of pain and pruritus without centrally mediated side effects, and macrocyclic tetrapeptides based on CJ-15,208 that can exhibit different opioid profiles with potential applications such as analgesics and treatments for substance abuse.
Collapse
Affiliation(s)
- Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
31
|
Ko MC, Husbands SM. Pleiotropic Effects of Kappa Opioid Receptor-Related Ligands in Non-human Primates. Handb Exp Pharmacol 2022; 271:435-452. [PMID: 33274403 PMCID: PMC8175454 DOI: 10.1007/164_2020_419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The kappa opioid receptor (KOR)-related ligands have been demonstrated in preclinical studies for several therapeutic potentials. This chapter highlights (1) how non-human primates (NHP) studies facilitate the research and development of ligands targeting the KOR, (2) effects of the endogenous opioid peptide, dynorphin A-(1-17), and its analogs in NHP, and (3) pleiotropic effects and therapeutic applications of KOR-related ligands. In particular, synthetic ligands targeting the KOR have been extensively studied in NHP in three therapeutic areas, i.e., the treatment for itch, pain, and substance use disorders. As the KORs are widely expressed in the peripheral and central nervous systems, pleiotropic effects of KOR-related ligands, such as discriminative stimulus effects, neuroendocrine effects (e.g., prolactin release and stimulation of hypothalamic-pituitary-adrenal axis), and diuresis, in NHP are discussed. Centrally acting KOR agonists are known to produce adverse effects including dysphoria, hallucination, and sedation. Nonetheless, with strategic advances in medicinal chemistry, three classes of KOR-related agonists, i.e., peripherally restricted KOR agonists, mixed KOR/mu opioid receptor partial agonists, and G protein-biased KOR agonists, warrant additional NHP studies to improve our understanding of their functional efficacy, selectivity, and tolerability. Pharmacological studies in NHP which carry high translational significance will facilitate future development of KOR-based medications.
Collapse
Affiliation(s)
- Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
32
|
Baynard C, Prisinzano TE, Butelman ER. Rapid-Onset Anti-Stress Effects of a Kappa-Opioid Receptor Antagonist, LY2795050, Against Immobility in an Open Space Swim Paradigm in Male and Female Mice. Front Pharmacol 2021; 12:775317. [PMID: 34880762 PMCID: PMC8645979 DOI: 10.3389/fphar.2021.775317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The kappa-opioid receptor (KOR) / dynorphin system is implicated with behavioral and neurobiological effects of stress exposure (including heavy exposure to drugs of abuse) in translational animal models. Thus some KOR-antagonists can decrease the aversive, depressant-like and anxiety-like effects caused by stress exposure. The first generation of selective KOR-antagonists have slow onsets (hours) and extremely long durations of action (days-weeks), in vivo. A new generation of KOR antagonists with rapid onset and shorter duration of action can potentially decrease the effects of stress exposure in translational models, and may be of interest for medication development. This study examined the rapid onset anti-stress effects of one of the shorter acting novel KOR-antagonists (LY2795050, (3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide)) in a single-session open space swim (OSS) stress paradigm (15 min duration), in adult male and female C57BL/6 J mice. LY2795050 (0.32 mg/kg, i.p.) had rapid onset (within 15 min) and short duration (<3 h) of KOR-antagonist effects, based on its blockade of the locomotor depressant effects of the KOR-agonist U50,488 (10 mg/kg). LY2795050 (0.32 mg/kg), when administered only 1 min prior to the OSS stress paradigm, decreased immobility in males, but not females. With a slightly longer pretreatment time (15 min), this dose of LY2795050 decreased immobility in both males and females. A 10-fold smaller dose of LY2795050 (0.032 mg/kg) was inactive in the OSS, showing dose-dependence of this anti-stress effect. Overall, these studies show that a novel KOR-antagonist can produce very rapid onset anti-immobility effects in this model of acute stress exposure.
Collapse
Affiliation(s)
- Caroline Baynard
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| |
Collapse
|
33
|
Zhou Q, Zhang Z, Long S, Li W, Wang B, Liang N. Opioids in cancer: The κ‑opioid receptor (Review). Mol Med Rep 2021; 25:44. [PMID: 34878160 PMCID: PMC8674701 DOI: 10.3892/mmr.2021.12560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
The κ‑opioid receptor (KOR) is one of the primary receptors of opioids and serves a vital role in the regulation of pain, anesthesia, addiction and other pathological and physiological processes. KOR is associated with several types of cancer and may influence cancer progression. It has been proposed that KOR may represent a new tumor molecular marker and provide a novel basis for molecular targeted therapies for cancer. However, the association between KOR and cancer remains to be explored comprehensively. The present review introduces KOR and its association with different types of cancer. Improved understanding of KOR may facilitate development of novel antitumor therapies.
Collapse
Affiliation(s)
- Qier Zhou
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhiwei Zhang
- Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Songkai Long
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wanjun Li
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Baiyun Wang
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Na Liang
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
34
|
Spetea M, Schmidhammer H. Recent Chemical and Pharmacological Developments on 14-Oxygenated- N-methylmorphinan-6-ones. Molecules 2021; 26:5677. [PMID: 34577147 PMCID: PMC8464912 DOI: 10.3390/molecules26185677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Adequate pain management, particularly chronic pain, remains a major challenge associated with modern-day medicine. Current pharmacotherapy offers unsatisfactory long-term solutions due to serious side effects related to the chronic administration of analgesic drugs. Morphine and structurally related derivatives (e.g., oxycodone, oxymorphone, buprenorphine) are highly effective opioid analgesics, mediating their effects via the activation of opioid receptors, with the mu-opioid receptor subtype as the primary molecular target. However, they also cause addiction and overdose deaths, which has led to a global opioid crisis in the last decades. Therefore, research efforts are needed to overcome the limitations of present pain therapies with the aim to improve treatment efficacy and to reduce complications. This review presents recent chemical and pharmacological advances on 14-oxygenated-N-methylmorphinan-6-ones, in the search of safer pain therapeutics. We focus on drug design strategies and structure-activity relationships on specific modifications in positions 5, 6, 14 and 17 on the morphinan skeleton, with the goal of aiding the discovery of opioid analgesics with more favorable pharmacological properties, potent analgesia and fewer undesirable effects. Targeted molecular modifications on the morphinan scaffold can afford novel opioids as bi- or multifunctional ligands targeting multiple opioid receptors, as attractive alternatives to mu-opioid receptor selective analgesics.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria;
| | | |
Collapse
|
35
|
Hillhouse TM, Olson KM, Hallahan JE, Rysztak LG, Sears BF, Meurice C, Ostovar M, Koppenhaver PO, West JL, Jutkiewicz EM, Husbands SM, Traynor JR. The Buprenorphine Analogue BU10119 Attenuates Drug-Primed and Stress-Induced Cocaine Reinstatement in Mice. J Pharmacol Exp Ther 2021; 378:287-299. [PMID: 34183434 PMCID: PMC11047085 DOI: 10.1124/jpet.121.000524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/22/2021] [Indexed: 01/01/2023] Open
Abstract
There are no Food and Drug Administration-approved medications for cocaine use disorder, including relapse. The μ-opioid receptor (MOPr) partial agonist buprenorphine alone or in combination with naltrexone has been shown to reduce cocaine-positive urine tests and cocaine seeking in rodents. However, there are concerns over the abuse liability of buprenorphine. Buprenorphine's partial agonist and antagonist activity at the nociception receptor (NOPr) and κ-opioid receptor (KOPr), respectively, may contribute to its ability to inhibit cocaine seeking. Thus, we hypothesized that a buprenorphine derivative that exhibits antagonist activity at MOPr and KOPr with enhanced agonist activity at the NOPr could provide a more effective treatment. Here we compare the pharmacology of buprenorphine and two analogs, BU10119 and BU12004, in assays for antinociception and for cocaine- and stress-primed reinstatement in the conditioned place preference paradigm. In vitro and in vivo assays showed that BU10119 acts as an antagonist at MOPr, KOPr, and δ-opioid receptor (DOPr) and a partial agonist at NOPr, whereas BU12004 showed MOPr partial agonist activity and DOPr, KOPr, and NOPr antagonism. BU10119 and buprenorphine but not BU12004 lessened cocaine-primed reinstatement. In contrast, BU10119, BU12004, and buprenorphine blocked stress-primed reinstatement. The selective NOPr agonist SCH221510 but not naloxone decreased cocaine-primed reinstatement. Together, these findings are consistent with the concept that NOPr agonism contributes to the ability of BU10119 and buprenorphine to attenuate reinstatement of cocaine-conditioned place preference in mice. The findings support the development of buprenorphine analogs lacking MOPr agonism with increased NOPr agonism for relapse prevention to cocaine addiction. SIGNIFICANCE STATEMENT: There are no Food and Drug Administration-approved medications for cocaine use disorder. Buprenorphine has shown promise as a treatment for cocaine relapse prevention; however, there are concerns over the abuse liability of buprenorphine. Here we show a buprenorphine analogue, BU10119, which lacks μ-opioid receptor agonism and inhibits cocaine-primed and stress-primed reinstatement in a conditioned place-preference paradigm. The results suggest the development of BU10119 for the management of relapse to cocaine seeking.
Collapse
MESH Headings
- Animals
- Buprenorphine/pharmacology
- Buprenorphine/analogs & derivatives
- Mice
- Male
- Cocaine/pharmacology
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
- Cocaine-Related Disorders/drug therapy
- Mice, Inbred C57BL
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Drug-Seeking Behavior/drug effects
- Humans
- Receptors, Opioid/metabolism
- Receptors, Opioid/agonists
- Narcotic Antagonists/pharmacology
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
Collapse
Affiliation(s)
- Todd M Hillhouse
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Keith M Olson
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - James E Hallahan
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Lauren G Rysztak
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Bryan F Sears
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Claire Meurice
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Mehrnoosh Ostovar
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Peyton O Koppenhaver
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Joshua L West
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Emily M Jutkiewicz
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Stephen M Husbands
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - John R Traynor
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| |
Collapse
|
36
|
Improving the Utility of a Dynorphin Peptide Analogue Using Mannosylated Glycoliposomes. Int J Mol Sci 2021; 22:ijms22157996. [PMID: 34360762 PMCID: PMC8348236 DOI: 10.3390/ijms22157996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/27/2022] Open
Abstract
Peptide therapeutics offer numerous advantages in the treatment of diseases and disorders of the central nervous system (CNS). However, they are not without limitations, especially in terms of their pharmacokinetics where their metabolic lability and low blood–brain barrier penetration hinder their application. Targeted nanoparticle delivery systems are being tapped for their ability to improve the delivery of therapeutics into the brain non-invasively. We have developed a family of mannosylated glycoliposome delivery systems for targeted drug delivery applications. Herein, we demonstrate via in vivo distribution studies the potential of these glycoliposomes to improve the utility of CNS active therapeutics using dynantin, a potent and selective dynorphin peptide analogue antagonist of the kappa opioid receptor (KOR). Glycoliposomal entrapment protected dynantin against known rapid metabolic degradation and ultimately improved brain levels of the peptide by approximately 3–3.5-fold. Moreover, we linked this improved brain delivery with improved KOR antagonist activity by way of an approximately 30–40% positive modulation of striatal dopamine levels 20 min after intranasal administration. Overall, the results clearly highlight the potential of our glycoliposomes as a targeted delivery system for therapeutic agents of the CNS.
Collapse
|
37
|
Analogs of the κ opioid receptor antagonist arodyn cyclized by ring-closing metathesis retain κ opioid receptor affinity, selectivity and κ opioid receptor antagonism. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02758-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
38
|
Bloodgood DW, Hardaway JA, Stanhope CM, Pati D, Pina MM, Neira S, Desai S, Boyt KM, Palmiter RD, Kash TL. Kappa opioid receptor and dynorphin signaling in the central amygdala regulates alcohol intake. Mol Psychiatry 2021; 26:2187-2199. [PMID: 32099099 PMCID: PMC8124770 DOI: 10.1038/s41380-020-0690-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/14/2020] [Accepted: 02/13/2020] [Indexed: 01/09/2023]
Abstract
Excessive alcohol drinking has been shown to modify brain circuitry to predispose individuals for future alcohol abuse. Previous studies have implicated the central nucleus of the amygdala (CeA) as an important site for mediating the somatic symptoms of withdrawal and for regulating alcohol intake. In addition, recent work has established a role for both the Kappa Opioid Receptor (KOR) and its endogenous ligand dynorphin in mediating these processes. However, it is unclear whether these effects are due to dynorphin or KOR arising from within the CeA itself or other input brain regions. To directly examine the role of preprodynorphin (PDYN) and KOR expression in CeA neurons, we performed region-specific conditional knockout of these genes and assessed the effects on the Drinking in the Dark (DID) and Intermittent Access (IA) paradigms. Conditional gene knockout resulted in sex-specific responses wherein PDYN knockout decreased alcohol drinking in both male and female mice, whereas KOR knockout decreased drinking in males only. We also found that neither PDYN nor KOR knockout protected against anxiety caused by alcohol drinking. Lastly, a history of alcohol drinking did not alter synaptic transmission in PDYN neurons in the CeA of either sex, but excitability of PDYN neurons was increased in male mice only. Taken together, our findings indicate that PDYN and KOR signaling in the CeA plays an important role in regulating excessive alcohol consumption and highlight the need for future studies to examine how this is mediated through downstream effector regions.
Collapse
Affiliation(s)
- Daniel W Bloodgood
- Bowles Center for Alcohol Studies, Curriculum in Neuroscience, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - J Andrew Hardaway
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Christina M Stanhope
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Dipanwita Pati
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Melanie M Pina
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Sofia Neira
- Bowles Center for Alcohol Studies, Curriculum in Neuroscience, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Shivani Desai
- Department of Biology, University of North Carolina College of Arts and Sciences, Chapel Hill, NC, USA
| | - Kristen M Boyt
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Curriculum in Neuroscience, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
- Bowles Center for Alcohol Studies, Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
39
|
Saleh AH, Abdelwaly A, Darwish KM, Eissa AAHM, Chittiboyina A, Helal MA. Deciphering the molecular basis of the kappa opioid receptor selectivity: A Molecular Dynamics study. J Mol Graph Model 2021; 106:107940. [PMID: 34015577 DOI: 10.1016/j.jmgm.2021.107940] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022]
Abstract
Selective antagonists for the kappa opioid receptor (KOP) have the potential to treat opiate and alcohol addiction, as well as depression and other CNS disorders. Over the years, the development of KOP-selective antagonists yielded very few successful compounds. Recently, N-substituted trans-3,4-dimethyl-4-(3-hydroxyphenyl)piperidines have emerged as a novel class of pure opioid receptor antagonists, including the marketed Mu opioid receptor (MOP) peripheral antagonist Alvimopan and the potent KOP antagonist JDTic. However, the selectivity determinants of this class of compounds towards the opioid receptor subtypes are still vague and understudied. In this work, we have performed Molecular Dynamics (MD) simulation to gain insights into the differential binding of this class of compounds into KOP, as exemplified by Alvimopan and JDTic. Our study indicated that the selectivity largely depends on ligands interaction with the selectivity pocket formed by Val108, Thr111, and Val118, supported by two additional polar and hydrophobic contacts with Asp138 and Trp287, respectively. Our results also demonstrate, for the first time, that non-morphinan ligands can still adopt the "message-address model" for KOP efficacy and selectivity by binding to Glu297.
Collapse
Affiliation(s)
- Amr H Saleh
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt
| | - Ahmad Abdelwaly
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt
| | - Khaled M Darwish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Amal A H M Eissa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amar Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS, 38677, United States
| | - Mohamed A Helal
- University of Science and Technology, Biomedical Sciences Program, Zewail City of Science and Technology, October Gardens, 6th of October, Giza, 12578, Egypt; Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
40
|
Lazaridou A, Paschali M, Edwards RR, Gilligan C. Is Buprenorphine Effective for Chronic Pain? A Systematic Review and Meta-analysis. PAIN MEDICINE 2021; 21:3691-3699. [PMID: 32330264 DOI: 10.1093/pm/pnaa089] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The objective was to perform a systematic review and meta-analysis of the literature on the effects of buprenorphine on chronic pain outcomes (i.e., patient-reported pain intensity) in patients with and without opioid use disorder (OUD). DESIGN Ovid/Medline, PubMed, Embase, and the Cochrane Library were searched for studies that explored the effectiveness (in reducing pain) of buprenorphine treatment for chronic pain patients with and without a history of OUD. Randomized controlled trials and observational studies were included in the review. METHODS Two separate searches were conducted to identify buprenorphine trials that included chronic pain patients either with or without OUD. Five studies used validated pain report measures and included a chronic pain population with OUD. Nine studies used validated report measures and included chronic pain patients without OUD. Meta-analysis was performed using the R, version 3.2.2, Metafor package, version 1.9-7. RESULTS The meta-analysis revealed that buprenorphine has a beneficial effect on pain intensity overall, with a small mean effect size in patients with comorbid chronic pain and OUD and a moderate- to large-sized effect in chronic pain patients without OUD. CONCLUSIONS Our results indicate that buprenorphine is modestly beneficial in reducing pain intensity in patients without OUD. Although informative, these findings should be carefully interpreted due to the small amount of data available and the variation in study designs.
Collapse
Affiliation(s)
- Asimina Lazaridou
- Department of Anesthesiology, Harvard Medical School, Brigham & Women's Hospital, Chestnut Hill, Massachusetts, USA
| | - Myrella Paschali
- Department of Anesthesiology, Harvard Medical School, Brigham & Women's Hospital, Chestnut Hill, Massachusetts, USA
| | - Robert R Edwards
- Department of Anesthesiology, Harvard Medical School, Brigham & Women's Hospital, Chestnut Hill, Massachusetts, USA
| | - Christopher Gilligan
- Department of Anesthesiology, Harvard Medical School, Brigham & Women's Hospital, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
41
|
Zaidi SA, Katritch V. Structural Characterization of KOR Inactive and Active States for 3D Pharmacology and Drug Discovery. Handb Exp Pharmacol 2021; 271:41-64. [PMID: 33945028 DOI: 10.1007/164_2021_461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The structure of the human kappa opioid receptor (KOR) in complex with the long-acting antagonist JDTic was solved crystallographically in 2012 and, along with structures of other opioid receptors, revolutionized our understanding of opioid system function and pharmacology. More recently, active state KOR structure was also determined, giving important insights into activation mechanisms of the receptor. In this review, we will discuss how the understanding of atomistic structures of KOR established a key platform for deciphering details of subtype and functional selectivity of KOR-targeting ligands and for discovery of new chemical probes with potentially beneficial pharmacological profiles.
Collapse
Affiliation(s)
- Saheem A Zaidi
- Department of Quantitative and Computational Biology, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Vsevolod Katritch
- Department of Quantitative and Computational Biology, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA. .,Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
42
|
Ryu J, Stone P, Lee S, Payne B, Gorse K, Lafrenaye A. Buprenorphine alters microglia and astrocytes acutely following diffuse traumatic brain injury. Sci Rep 2021; 11:8620. [PMID: 33883663 PMCID: PMC8060410 DOI: 10.1038/s41598-021-88030-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
Traumatic brain injury (TBI) is a common phenomenon, accounting for significant cost and adverse health effects. While there is information about focal pathologies following TBI, knowledge of more diffuse processes is lacking, particularly regarding how analgesics affect this pathology. As buprenorphine is the most commonly used analgesic in experimental TBI models, this study investigated the acute effects of the opioid analgesic buprenorphine (Bup-SR-Lab) on diffuse neuronal/glial pathology, neuroinflammation, cell damage, and systemic physiology. We utilized a model of central fluid percussion injury (CFPI) in adult male rats treated with a single subcutaneous bolus of Bup-SR-Lab or saline 15 min post-injury. Microscopic assessments were performed at 1 day post-injury. Cell impermeable dextran was infused intraventricularly prior to sacrifice to assess neuronal membrane disruption. Axonal injury was assessed by investigating labeling of the anterogradely transported amyloid precursor protein. Neuroinflammation was assessed by analyzing Iba-1 + microglial and GFAP + astrocyte histological/morphological features as well as cytokine levels in both regions of interest (ROIs). Myelin pathology was assessed by evaluating the expression of myelin basic protein (MBP) and the propensity of MBP + myelin debris. Acute physiologic data showed no difference between groups except for reduction in weight loss following cFPI in Bup treated animals compared to saline. There were no discernable differences in axonal injury or membrane disruption between treatment groups. Cytokine levels were consistent between Bup and saline treated animals, however, microglia and astrocytes revealed region specific histological changes at 1d following Bup treatment. Myelin integrity and overall MBP expression showed no differences between Bup and saline treated animals, but there were significant regional differences in MBP expression between the cortex and thalamus. These data suggest effects of Bup treatment on weight following CFPI and potential regional specificity of Bup-associated microglial and astrocyte alterations, but very little change in other acute pathology at 1-day post-injury. Overall, this preliminary study indicates that use of Bup-SR-Lab in preclinical work does have effects on acute glial pathology, however, longer term studies will be needed to assess potential effects of Bup treatment on more chronic pathological progressions.
Collapse
Affiliation(s)
- Jane Ryu
- grid.224260.00000 0004 0458 8737Virginia Commonwealth University, 1101 E. Marshall St., Box 980709, Richmond, VA 23298 USA
| | - Phillip Stone
- grid.224260.00000 0004 0458 8737Virginia Commonwealth University, 1101 E. Marshall St., Box 980709, Richmond, VA 23298 USA
| | | | - Brighton Payne
- grid.266671.20000 0000 9565 4349University of Mary Washington, Fredericksburg, VA USA
| | - Karen Gorse
- grid.224260.00000 0004 0458 8737Virginia Commonwealth University, 1101 E. Marshall St., Box 980709, Richmond, VA 23298 USA
| | - Audrey Lafrenaye
- grid.224260.00000 0004 0458 8737Virginia Commonwealth University, 1101 E. Marshall St., Box 980709, Richmond, VA 23298 USA
| |
Collapse
|
43
|
Marchette RCN, Gregory-Flores A, Tunstall BJ, Carlson ER, Jackson SN, Sulima A, Rice KC, Koob GF, Vendruscolo LF. κ-Opioid receptor antagonism reverses heroin withdrawal-induced hyperalgesia in male and female rats. Neurobiol Stress 2021; 14:100325. [PMID: 33997152 PMCID: PMC8095052 DOI: 10.1016/j.ynstr.2021.100325] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/18/2021] [Accepted: 04/06/2021] [Indexed: 10/29/2022] Open
Abstract
Although opioids are potent analgesics, a consequence of chronic opioid use is hyperalgesia during withdrawal, which may contribute to opioid misuse. Dynorphin, the endogenous ligand of κ-opioid receptors (KORs), is upregulated in opioid-dependent rats and in animal models of chronic pain. However, the role of KORs in opioid withdrawal-induced hyperalgesia remains to be determined. We hypothesized that KOR antagonism would reverse opioid withdrawal-induced hyperalgesia in opioid-dependent rats. Male and female Wistar rats received daily injections of heroin (2-6 mg/kg, SC) and were tested for mechanical sensitivity in the electronic von Frey test 4-6 h into withdrawal. Female rats required significantly more heroin than male rats to reach comparable levels of both heroin-induced analgesia and hyperalgesia (6 mg/kg vs. 2 mg/kg). Once hyperalgesia was established, we tested the effects of the KOR antagonists nor-binaltorphimine (norBNI; 30 mg/kg, SC) and 5'-guanidinonaltrindole (5'GNTI; 30 mg/kg, SC). When the animals continued to receive their daily heroin treatment (or saline treatment in the repeated saline group) five times per week throughout the experiment, both KOR antagonists reversed heroin withdrawal-induced hyperalgesia. The anti-hyperalgesia effect of norBNI was more prolonged in males than in females (14 days vs. 7 days), whereas 5'GNTI had more prolonged effects in females than in males (14 days vs. 4 days). The behavioral effects of 5'GNTI coincided with higher 5'GNTI levels in the brain than in plasma when measured at 24 h, whereas 5'GNTI did not reverse hyperalgesia at 30 min posttreatment when 5'GNTI levels were higher in plasma than in the brain. Finally, we tested the effects of 5'GNTI on naloxone-induced and spontaneous signs of opioid withdrawal and found no effect in either male or female rats. These findings indicate a functional role for KORs in heroin withdrawal-induced hyperalgesia that is observed in rats of both sexes.
Collapse
Affiliation(s)
- Renata C N Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Adriana Gregory-Flores
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Brendan J Tunstall
- Department of Pharmacology, Addiction Science, and Toxicology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Erika R Carlson
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Shelley N Jackson
- Structural Biology Core, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Agnieszka Sulima
- Drug Design and Synthesis Section, National Institute on Drug Abuse, Intramural Research Program, Bethesda, MD, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse, Intramural Research Program, Bethesda, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
44
|
Gisemba SA, Ferracane MJ, Murray TF, Aldrich JV. Conformational Constraint between Aromatic Residue Side Chains in the "Message" Sequence of the Peptide Arodyn Using Ring Closing Metathesis Results in a Potent and Selective Kappa Opioid Receptor Antagonist. J Med Chem 2021; 64:3153-3164. [PMID: 33688737 DOI: 10.1021/acs.jmedchem.0c01984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Kappa opioid receptor (KOR) antagonists have recently shown potential for treating drug addiction and mood disorders. The linear acetylated dynorphin A analog arodyn (Ac[Phe1,2,3,Arg4,d-Ala8]dynorphin A-(1-11)NH2), synthesized in our laboratory, demonstrated potent and selective KOR antagonism. Cyclization of arodyn could potentially stabilize the bioactive conformation and enhance its metabolic stability. The cyclization strategy employed involved ring closing metathesis between adjacent meta- or para-substituted Tyr(allyl) residues in the "message" sequence that were predicted in a docking study to yield analogs that would bind to the KOR with binding poses similar to arodyn. Consistent with the modeling, the resulting analogs retained KOR affinity similar to arodyn; the peptides involving cyclization between para O-allyl groups also retained high KOR selectivity, with one analog exhibiting KOR antagonist potency (KB = 15 nM) similar to arodyn. These promising cyclized analogs with constrained aromatic residues represent novel leads for further exploration of KOR pharmacology.
Collapse
Affiliation(s)
- Solomon A Gisemba
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Michael J Ferracane
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida 32610, United States.,Department of Chemistry, University of Redlands, Redlands, California 92373, United States
| | - Thomas F Murray
- Departments of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, Nebraska 68102, United States
| | - Jane V Aldrich
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
45
|
Can the migration process influence the clinical expression of heroin use disorder in migrants to Italy? CNS Spectr 2021; 26:62-70. [PMID: 31969204 DOI: 10.1017/s1092852919001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND For some time now, there has been a strong consensus that the migration process can influence the onset, course, development, outcome, and clinical aspects of psychiatric pathologies. METHODS In this study, we have analyzed the influence of the migration process on the clinical expression of heroin use disorder (HUD). In a naturalistic case-control study, we compared, both at univariate and multivariate level, 30 migrant HUD (M-HUD) patients with 30 age/gender-matched Italian HUD (IT-HUD) patients. We also analyzed demographic data, drug addiction history, psychopathological symptoms, addictive behavior, and emotional reactivity to life events. RESULTS Compared with IT-HUD pairs, at HUD Agonist Opioid Treatment, M-HUD patients were characterized by inadequate income and the presence of legal problems. They were more frequently at stage 3 of heroin addiction, with a concomitantly less frequent use of stimulants. Their age at the onset of heroin use was greater than that of subjects in the IT-HUD group. HUD post-traumatic stress disorder spectrum was present and was more severe in all M-HUD patients, but grief reactions and maladaptive behavior were the most discriminant traits. No differences were found in terms of addictive behaviors related to heroin craving or with respect to the severity/typology of psychopathology specific to HUD. CONCLUSIONS The migratory process does not seem to be correlated with addictive behaviors or with psychopathology specific to HUD. It partly affects HUD history, and specifically correlates with emotional reactivity to loss and traumatic life events, so suggesting that in M-HUD individuals, the link between the migratory syndrome and HUD is very close.
Collapse
|
46
|
Spetea M, Schmidhammer H. Kappa Opioid Receptor Ligands and Pharmacology: Diphenethylamines, a Class of Structurally Distinct, Selective Kappa Opioid Ligands. Handb Exp Pharmacol 2021; 271:163-195. [PMID: 33454858 DOI: 10.1007/164_2020_431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The kappa opioid receptor (KOR), a G protein-coupled receptor, and its endogenous ligands, the dynorphins, are prominent members of the opioid neuromodulatory system. The endogenous kappa opioid system is expressed in the central and peripheral nervous systems, and has a key role in modulating pain in central and peripheral neuronal circuits and a wide array of physiological functions and neuropsychiatric behaviors (e.g., stress, reward, emotion, motivation, cognition, epileptic seizures, itch, and diuresis). We review the latest advances in pharmacology of the KOR, chemical developments on KOR ligands with advances and challenges, and therapeutic and potential applications of KOR ligands. Diverse discovery strategies of KOR ligands targeting natural, naturally derived, and synthetic compounds with different scaffolds, as small molecules or peptides, with short or long-acting pharmacokinetics, and central or peripheral site of action, are discussed. These research efforts led to ligands with distinct pharmacological properties, as agonists, partial agonists, biased agonists, and antagonists. Differential modulation of KOR signaling represents a promising strategy for developing pharmacotherapies for several human diseases, either by activating (treatment of pain, pruritus, and epilepsy) or blocking (treatment of depression, anxiety, and addiction) the receptor. We focus on the recent chemical and pharmacological advances on diphenethylamines, a new class of structurally distinct, selective KOR ligands. Design strategies and investigations to define structure-activity relationships together with in vivo pharmacology of diphenethylamines as agonists, biased agonists, and antagonists and their potential use as therapeutics are discussed.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
47
|
Zhang L, McCarthy DM, Eskow Jaunarajs KL, Biederman J, Spencer TJ, Bhide PG. Frontal Cortical Monoamine Release, Attention, and Working Memory in a Perinatal Nicotine Exposure Mouse Model Following Kappa Opioid Receptor Antagonism. Cereb Cortex 2021; 31:483-496. [PMID: 32869057 DOI: 10.1093/cercor/bhaa238] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 02/27/2024] Open
Abstract
Perinatal nicotine exposure (PNE) produces frontal cortical hypo-dopaminergic state and attention and working memory deficits consistent with neurodevelopmental disorders such as attention deficit hyperactivity disorder (ADHD). Methylphenidate alleviates ADHD symptoms by increasing extracellular dopamine and noradrenaline. Kappa opioid receptor (KOR) antagonism may be another mechanism to achieve the same results because KOR activation inhibits frontal cortical dopamine release. We administered the selective KOR antagonist norbinaltorphimine (norBNI) (20 mg/kg; intraperitoneal) or methylphenidate (0.75 mg/kg; intraperitoneal) to PNE mouse model and examined frontal cortical monoamine release, attention, and working memory. Both compounds increased dopamine and noradrenaline release but neither influenced serotonin release. Both compounds improved object-based attention and working memory in the PNE group, with norBNI's effects evident at 2.5 h and 5.5 h but absent at 24 h. Methylphenidate's effects were evident at 0.5 h but not at 2.5 h. norBNI's effects temporally coincided with frontal cortical c-Jun N-terminal kinase phosphorylation. norBNI did not alter tissue dopamine content in the nucleus accumbens, offering preliminary support for lack of reinforcement.
Collapse
Affiliation(s)
- Lin Zhang
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Deirdre M McCarthy
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | | | - Joseph Biederman
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Thomas J Spencer
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pradeep G Bhide
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
48
|
Margolis EB, Wallace TL, Van Orden LJ, Martin WJ. Differential effects of novel kappa opioid receptor antagonists on dopamine neurons using acute brain slice electrophysiology. PLoS One 2020; 15:e0232864. [PMID: 33373369 PMCID: PMC7771853 DOI: 10.1371/journal.pone.0232864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 12/01/2020] [Indexed: 12/03/2022] Open
Abstract
Activation of the kappa opioid receptor (KOR) contributes to the aversive properties of stress, and modulates key neuronal circuits underlying many neurobehavioral disorders. KOR agonists directly inhibit ventral tegmental area (VTA) dopaminergic neurons, contributing to aversive responses (Margolis et al. 2003, 2006); therefore, selective KOR antagonists represent a novel therapeutic approach to restore circuit function. We used whole cell electrophysiology in acute rat midbrain slices to evaluate pharmacological properties of four novel KOR antagonists: BTRX-335140, BTRX-395750, PF-04455242, and JNJ-67953964. Each compound concentration-dependently reduced the outward current induced by the KOR selective agonist U-69,593. BTRX-335140 and BTRX-395750 fully blocked U-69,593 currents (IC50 = 1.2 ± 0.9 and 1.2 ± 1.3 nM, respectively). JNJ-67953964 showed an IC50 of 3.0 ± 4.6 nM. PF-04455242 exhibited partial antagonist activity asymptoting at 55% blockade (IC50 = 6.7 ± 15.1 nM). In 3/8 of neurons, 1 μM PF-04455242 generated an outward current independent of KOR activation. BTRX-335140 (10 nM) did not affect responses to saturating concentrations of the mu opioid receptor (MOR) agonist DAMGO or the delta opioid receptor (DOR) agonist DPDPE, while JNJ-67953964 (10 nM) partially blocked DAMGO and DPDPE responses. Importantly, BTRX-335140 (10 nM) rapidly washed out with complete recovery of U-69,593 responses within 10 min. Collectively, we show electrophysiological evidence of key differences amongst KOR antagonists that could impact their therapeutic potential and have not been observed using recombinant systems. The results of this study demonstrate the value of characterizing compounds in native neuronal tissue and within circuits implicated in the neurobehavioral disorders of interest.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzamides/pharmacology
- Biphenyl Compounds/pharmacology
- Dopaminergic Neurons/drug effects
- Dopaminergic Neurons/metabolism
- Electrophysiology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Male
- Membrane Potentials/drug effects
- Mesencephalon/metabolism
- Narcotic Antagonists/pharmacology
- Oxadiazoles/pharmacology
- Patch-Clamp Techniques/methods
- Piperidines/pharmacology
- Pyrrolidines/pharmacology
- Quinolines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/metabolism
- Sulfonamides/pharmacology
- Ventral Tegmental Area/drug effects
Collapse
Affiliation(s)
- Elyssa B. Margolis
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States of America
- * E-mail:
| | - Tanya L. Wallace
- BlackThorn Therapeutics, San Francisco, CA, United States of America
| | | | - William J. Martin
- BlackThorn Therapeutics, San Francisco, CA, United States of America
| |
Collapse
|
49
|
Schmidhammer H, Erli F, Guerrieri E, Spetea M. Development of Diphenethylamines as Selective Kappa Opioid Receptor Ligands and Their Pharmacological Activities. Molecules 2020; 25:E5092. [PMID: 33147885 PMCID: PMC7663249 DOI: 10.3390/molecules25215092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 02/01/2023] Open
Abstract
Among the opioid receptors, the kappa opioid receptor (KOR) has been gaining substantial attention as a promising molecular target for the treatment of numerous human disorders, including pain, pruritus, affective disorders (i.e., depression and anxiety), drug addiction, and neurological diseases (i.e., epilepsy). Particularly, the knowledge that activation of the KOR, opposite to the mu opioid receptor (MOR), does not produce euphoria or leads to respiratory depression or overdose, has stimulated the interest in discovering ligands targeting the KOR as novel pharmacotherapeutics. However, the KOR mediates the negative side effects of dysphoria/aversion, sedation, and psychotomimesis, with the therapeutic promise of biased agonism (i.e., selective activation of beneficial over deleterious signaling pathways) for designing safer KOR therapeutics without the liabilities of conventional KOR agonists. In this review, the development of new KOR ligands from the class of diphenethylamines is presented. Specifically, we describe the design strategies, synthesis, and pharmacological activities of differently substituted diphenethylamines, where structure-activity relationships have been extensively studied. Ligands with distinct profiles as potent and selective agonists, G protein-biased agonists, and selective antagonists, and their potential use as therapeutic agents (i.e., pain treatment) and research tools are described.
Collapse
MESH Headings
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/therapeutic use
- Humans
- Ligands
- Pain/drug therapy
- Pain/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/chemistry
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/metabolism
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (F.E.); (E.G.)
| | | | | | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (F.E.); (E.G.)
| |
Collapse
|
50
|
Reichard KL, Newton KA, Rivera ZMG, Sotero de Menezes PM, Schattauer SS, Land BB, Chavkin C. Regulation of Kappa Opioid Receptor Inactivation Depends on Sex and Cellular Site of Antagonist Action. Mol Pharmacol 2020; 98:548-558. [PMID: 32913138 DOI: 10.1124/molpharm.120.000124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
The prototypical member of the receptor-inactivating kappa opioid receptor (KOR) antagonists, norbinaltorphimine (norBNI), produces prolonged receptor inactivation by a cJun kinase mechanism. These antagonists have potential therapeutic utility in the treatment of stress disorders; however, additional preclinical characterization is necessary to understand important aspects of their action. In this study, we report that norBNI does not work as effectively in female mice as in males because of estrogen regulation of G protein receptor kinase (GRK); pretreatment of ovary-intact female mice with the selective GRK2/3 inhibitor, Compound 101, made females equally sensitive to norBNI as males. Prior observations suggested that in vivo treatment with norBNI does not produce long-lasting inhibition of KOR regulation of dopamine release in the nucleus accumbens. We assessed the persistence of norBNI receptor inactivation in subcellular compartments. Fast-scan cyclic voltammetry recordings confirmed that presynaptic inhibition of dopamine release by the KOR agonist U69,593 was not blocked by in vivo pretreatment with norBNI under conditions that prevented KOR-mediated aversion and analgesia. We employed a novel in vivo proxy sensor of KOR activation, adenovirus associated double floxed inverted-HyPerRed, and demonstrated that KOR activation stimulates cJun kinase-dependent reactive oxygen species (ROS) production in somatic regions of ventral tegmental area dopamine neurons, but did not activate ROS production in dopamine terminals. The compartment selective action helps explain how dopamine somatic, but not terminally expressed, KORs are inactivated by norBNI. These results further elucidate molecular signaling mechanisms mediating receptor-inactivating KOR antagonist action and advance medication development for this novel class of stress-resilience medications. SIGNIFICANCE STATEMENT: Kappa opioid receptor (KOR) antagonists are being developed as novel proresilience therapeutics for the treatment of mood and substance use disorders. This study showed that the long-acting KOR antagonists are affected by both the sex of the animal and the subcellular compartment in which the receptor is expressed.
Collapse
Affiliation(s)
- Kathryn L Reichard
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Keionna A Newton
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Zeena M G Rivera
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Paulo M Sotero de Menezes
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Selena S Schattauer
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Benjamin B Land
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Charles Chavkin
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| |
Collapse
|