1
|
Hennessy MR, Creed SM, Gutridge AM, Rusali LE, Luo D, Sepehri B, Rhoda ES, Villegas JA, van Rijn RM, Riley AP. Discovery of Potent Kappa Opioid Receptor Agonists Derived from Akuammicine. J Med Chem 2024. [PMID: 39565354 DOI: 10.1021/acs.jmedchem.4c00736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Akuammicine (1), an alkaloid isolated from Picralima nitida, is an agonist of the kappa opioid receptor (κOR). To establish structure-activity relationships (SARs) for this structurally unique κOR ligand, a collection of semisynthetic derivatives was synthesized. Evaluating these derivatives for their ability to activate the κOR and mu opioid receptor (μOR) revealed key SAR trends and identified derivatives with enhanced κOR potency. Most notably, substitutions to the C10 position of the aryl ring led to a > 200-fold improvement in κOR potency and nearly complete selectivity for the κOR. A selection of the most potent ligands was shown to possess differing abilities recruitment of β-Arrestin-2 to the κOR, indicating they have distinct signaling properties from each other and existing κOR ligands. The discovery of these κOR agonists underscores the potential of using natural products to identify new classes of potent and selective ligands and provides new tools to probe the κOR.
Collapse
Affiliation(s)
- Madeline R Hennessy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Simone M Creed
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lisa E Rusali
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Dan Luo
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Bakhtyar Sepehri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Elizabeth S Rhoda
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - José A Villegas
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, Indiana 47907, United States
| | - Andrew P Riley
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
2
|
Staedtler ES, Sapio MR, King DM, Maric D, Ghetti A, Mannes AJ, Iadarola MJ. The μ-opioid receptor differentiates two distinct human nociceptive populations relevant to clinical pain. Cell Rep Med 2024; 5:101788. [PMID: 39413733 PMCID: PMC11513826 DOI: 10.1016/j.xcrm.2024.101788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/26/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
The shortfall in new analgesic agents is a major impediment to reducing reliance on opioid medications for control of severe pain. In both animals and man, attenuating nociceptive transmission from primary afferent neurons with a μ-opioid receptor agonist yields highly effective analgesia. Consequently, deeper molecular characterization of human nociceptive afferents expressing OPRM1, the μ-opioid receptor gene, is a key component for advancing analgesic drug discovery and understanding clinical pain control. A co-expression matrix for the μ-opioid receptor and a variety of nociceptive channels as well as δ- and κ-opioid receptors is established by multiplex in situ hybridization. Our results indicate an OPRM1-positive population with strong molecular resemblance to rodent peptidergic C-nociceptors associated with tissue damage pain and an OPRM1-negative population sharing molecular characteristics of murine non-peptidergic C-nociceptors. The empirical identification of two distinct human nociceptive populations that differ profoundly in their presumed responsiveness to opioids provides an actionable translational framework for human pain control.
Collapse
Affiliation(s)
- Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD 20892, USA
| | | | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Zhao J, Baiula M, Cuna E, Francescato M, Matalińska J, Lipiński PF, Bedini A, Gentilucci L. Identification of c[D-Trp-Phe-β-Ala-β-Ala], the First κ-Opioid Receptor-Specific Negative Allosteric Modulator. ACS Pharmacol Transl Sci 2024; 7:3192-3204. [PMID: 39416958 PMCID: PMC11475277 DOI: 10.1021/acsptsci.4c00372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Recently, the fungus secondary metabolite cyclotetrapetide c[Trp-Phe-D-Pro-Phe] (CJ-15,208) and its derivatives deserved some attention for their unusual structure and distinctive in vitro and in vivo activity. These tryptophan-containing noncationic opioid peptides can be truly regarded as versatile picklocks capable of activating all opioid receptors. Intriguingly, minimal modification of the potent κ-opioid receptor (KOR) agonist c[D-Trp-Phe-Gly-β-Ala] (3) yielded c[D-Trp-Phe-β-Ala-β-Ala] (11), the first KOR-specific negative allosteric modulator (NAM) reported to-date. KOR exerts control over numerous functions in the central nervous system, including pain, depression, stress, mood, and reward. Hence, this KOR-selective NAM looks promising for modulating the KOR in addiction and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Junwei Zhao
- Dept.
of Chemistry “G. Ciamician”, University of Bologna, Campus Navile - Ue4, via Gobetti 83, Bologna 40129, Italy
| | - Monica Baiula
- Department
of Pharmacy and Biotechnology, University
of Bologna, Bologna 40126, Italy
| | - Elisabetta Cuna
- Department
of Pharmacy and Biotechnology, University
of Bologna, Bologna 40126, Italy
| | - Marco Francescato
- Dept.
of Chemistry “G. Ciamician”, University of Bologna, Campus Navile - Ue4, via Gobetti 83, Bologna 40129, Italy
| | - Joanna Matalińska
- Department
of Neuropeptides, Mossakowski Medical Research
Institute, Polish Academy of Sciences, 5 Pawinskiego Street, Warsaw 02-106, Poland
| | - Piotr F.J. Lipiński
- Department
of Neuropeptides, Mossakowski Medical Research
Institute, Polish Academy of Sciences, 5 Pawinskiego Street, Warsaw 02-106, Poland
| | - Andrea Bedini
- Department
of Pharmacy and Biotechnology, University
of Bologna, Bologna 40126, Italy
| | - Luca Gentilucci
- Dept.
of Chemistry “G. Ciamician”, University of Bologna, Campus Navile - Ue4, via Gobetti 83, Bologna 40129, Italy
- Health
Sciences & Technologies (HST) CIRI, University of Bologna, Ozzano
Emilia 40064, Italy
| |
Collapse
|
4
|
Trojniak AE, Dang VQ, Czekner KM, Russo RJ, Mather LM, Stahl EL, Cameron MD, Bohn LM, Aubé J. Synthesis and evaluation of 3,4,5-trisubstituted triazoles as G protein-biased kappa opioid receptor agonists. Eur J Med Chem 2024; 276:116627. [PMID: 38971050 PMCID: PMC11316643 DOI: 10.1016/j.ejmech.2024.116627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Kappa opioid receptor (KOR) agonists represent promising therapeutics for pain relief due to their analgesic properties along with lower abuse potential than opioids that act at the mu opioid receptor. However, typical KOR agonists produce sedation and dysphoria. Previous studies have shown that G protein signaling-biased KOR agonists may present a means to untangle the desired analgesic properties from undesired side effects. In this paper, we report a new series of G protein signaling-biased KOR agonists entailing -S- → -CH2- replacement in a previously reported KOR agonist, triazole 1.1. With an optimized carbon linker in hand, further development of the scaffold was undertaken to investigate the appendages of the triazole core. The structure-activity relationship study of this series is described, including several analogues that display enhanced potency while maintaining G protein-signaling bias compared to triazole 1.1.
Collapse
Affiliation(s)
- Ashley E Trojniak
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7363, USA
| | - Vuong Q Dang
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Kerri M Czekner
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA; The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, FL, 33458, USA
| | - Robin J Russo
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA; The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, FL, 33458, USA
| | - Lilyan M Mather
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7363, USA
| | - Edward L Stahl
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Michael D Cameron
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Laura M Bohn
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA; The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, FL, 33458, USA
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7363, USA.
| |
Collapse
|
5
|
Varela-Rodríguez L, Calzada F, Velázquez-Domínguez JA, Hernández-Ramírez VI, Varela-Rodríguez H, Bautista E, Herrera-Martínez M, Pichardo-Hernández DL, Castellanos-Mijangos RD, Chávez-Munguía B, Talamás-Rohana P. Toxicological Evaluation of Kaempferol and Linearolactone as Treatments for Amoebic Liver Abscess Development in Mesocricetus auratus. Int J Mol Sci 2024; 25:10633. [PMID: 39408962 PMCID: PMC11477209 DOI: 10.3390/ijms251910633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Several studies with kaempferol (KP) and linearolactone (LL) have demonstrated their antiparasitic activity. However, the toxicity of these treatments is unknown. Therefore, this study aimed to evaluate the possible toxicological effects of intraperitoneal (i.p.) administration of KP or LL on the amoebic liver abscess model (ALA) in Mesocricetus auratus. An ALA was induced in male hamsters with 1.5 × 105Entamoeba histolytica (E. histolytica) trophozoites inoculated in the left hepatic lobe. The lesion evolved for 4 days, and then KP (5 mg/kg body weight/day) or LL (10 mg/kg body weight/day) was administered for 4 consecutive days. Then, magnetic resonance imaging (MRI), paraclinical analyses, and necropsy for histopathological evaluation were performed. There was similar ALA inhibition by KP (19.42%), LL (28.16%), and metronidazole, the antiamoebic control (20.87%) (p ≤ 0.05, analysis of variance [ANOVA]). There were hepatic and renal biochemical alterations in all treatment groups, mainly for KP (aspartate aminotransferase: 347.5 ± 37.5 U/L; blood urea nitrogen: 19.4 ± 1.9 g/dL; p ≤ 0.05, ANOVA). Lesions found in the organs were directly linked to the pathology. In conclusion, KP and LL decreased ALA development and exerted fewer toxicological effects compared with metronidazole. Therefore, both compounds exhibit therapeutic potential as an alternative treatment of amoebiasis caused by E. histolytica. However, additional clinical studies in different contexts are required to reaffirm this assertion.
Collapse
Affiliation(s)
- Luis Varela-Rodríguez
- Facultad de Ciencias Químicas (FCQ), Universidad Autónoma de Chihuahua (UACH), Chihuahua 31125, CP, Mexico; (L.V.-R.); (H.V.-R.)
| | - Fernando Calzada
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades UMAE-CMNSXXI-IMSS, Ciudad de México 06725, CP, Mexico;
| | - José Antonio Velázquez-Domínguez
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, Ciudad de México 07360, CP, Mexico; (J.A.V.-D.); (V.I.H.-R.); (D.L.P.-H.); (B.C.-M.)
| | - Verónica Ivonne Hernández-Ramírez
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, Ciudad de México 07360, CP, Mexico; (J.A.V.-D.); (V.I.H.-R.); (D.L.P.-H.); (B.C.-M.)
| | - Hugo Varela-Rodríguez
- Facultad de Ciencias Químicas (FCQ), Universidad Autónoma de Chihuahua (UACH), Chihuahua 31125, CP, Mexico; (L.V.-R.); (H.V.-R.)
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua (UACH), Chihuahua 31109, CP, Mexico
| | - Elihú Bautista
- Unidad de Ciencias Ambientales, IPICYT, San Luis Potosí 78216, CP, Mexico;
| | - Mayra Herrera-Martínez
- Instituto de Farmacobiología, Universidad de la Cañada (UNCA), Teotitlán de Flores Magón, Oax 68540, CP, Mexico;
| | - Diana Laura Pichardo-Hernández
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, Ciudad de México 07360, CP, Mexico; (J.A.V.-D.); (V.I.H.-R.); (D.L.P.-H.); (B.C.-M.)
| | | | - Bibiana Chávez-Munguía
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, Ciudad de México 07360, CP, Mexico; (J.A.V.-D.); (V.I.H.-R.); (D.L.P.-H.); (B.C.-M.)
| | - Patricia Talamás-Rohana
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV-IPN, Ciudad de México 07360, CP, Mexico; (J.A.V.-D.); (V.I.H.-R.); (D.L.P.-H.); (B.C.-M.)
| |
Collapse
|
6
|
Wong S, Le GH, Vasudeva S, Teopiz KM, Phan L, Meshkat S, Kwan ATH, Rhee TG, Ho R, Choi H, Cao B, Rosenblat JD, McIntyre RS. Preclinical and clinical efficacy of kappa opioid receptor antagonists for depression: A systematic review. J Affect Disord 2024; 362:816-827. [PMID: 39019223 DOI: 10.1016/j.jad.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/05/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Approximately 30 % of persons with Major Depressive Disorder (MDD) inadequately respond to conventional antidepressants. Kappa opioid receptor (KOR) antagonists, aticaprant and navacaprant, are in development as treatments for MDD. Herein, we aim to comprehensively evaluate the safety, efficacy and pharmacology of aticaprant and navacaprant for MDD. METHODS We performed a systematic review of primary research investigating aticaprant and navacaprant on PubMed, OVID, and Scopus databases from inception to April 2024. Studies that reported on the pharmacological profile and/or safety and efficacy of aticaprant and navacaprant were included. RESULTS Navacaprant monotherapy and aticaprant adjunctive therapy are in development for MDD. Navacaprant exhibits 300-fold selectivity for the KOR compared to the mu-opioid receptor, while aticaprant exhibits 30-fold selectivity. At clinically-relevant doses, navacaprant and aticaprant occupy 87-95 % and 73-94 % of KORs, respectively. Clinical trials of the foregoing agents (navacaprant as monotherapy and actiprant as adjunctive therapy) reported significant improvement in depressive symptoms and may clinically benefit measures of anhedonia. Both agents appear well-tolerated, with most adverse events mild and no known safety concerns. LIMITATIONS Aticaprant and navacaprant treatment for MDD are in early stages of clinical trials and results from Phase 3 pivotal trials are not yet available. CONCLUSIONS Kappa opioid receptor antagonists may serve as mechanistically-novel treatments for MDD and persons who inadequately respond to index conventional antidepressants. Anhedonia is debilitating and insufficiently treated with conventional antidepressants. Future research vistas should establish the efficacy and safety of KORAs in phase 3 studies in both acute and maintenance paradigms.
Collapse
Affiliation(s)
- Sabrina Wong
- Mood Disorder and Psychopharmacology Unit, University Health Network, Toronto, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada.
| | - Gia Han Le
- Mood Disorder and Psychopharmacology Unit, University Health Network, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada.
| | - Shreya Vasudeva
- Mood Disorder and Psychopharmacology Unit, University Health Network, Toronto, Canada.
| | - Kayla M Teopiz
- Brain and Cognition Discovery Foundation, Toronto, Canada.
| | - Lee Phan
- Mood Disorder and Psychopharmacology Unit, University Health Network, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada.
| | | | - Angela T H Kwan
- Brain and Cognition Discovery Foundation, Toronto, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada.
| | - Taeho Greg Rhee
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore; Division of Life Science (LIFS), Hong Kong University of Science and Technology, Hong Kong.
| | - Hayun Choi
- Brain and Cognition Discovery Foundation, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Psychiatry, Veteran Health Service Medical Center, Seoul, Republic of Korea.
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing 400715, PR China.
| | - Joshua D Rosenblat
- Mood Disorder and Psychopharmacology Unit, University Health Network, Toronto, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada.
| | - Roger S McIntyre
- Mood Disorder and Psychopharmacology Unit, University Health Network, Toronto, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada.
| |
Collapse
|
7
|
van de Wetering R, Bibi R, Biggerstaff A, Hong S, Pengelly B, Prisinzano TE, La Flamme AC, Kivell BM. Nalfurafine promotes myelination in vitro and facilitates recovery from cuprizone + rapamycin-induced demyelination in mice. Glia 2024; 72:1801-1820. [PMID: 38899723 DOI: 10.1002/glia.24583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
The kappa opioid receptor has been identified as a promising therapeutic target for promoting remyelination. In the current study, we evaluated the ability of nalfurafine to promote oligodendrocyte progenitor cell (OPC) differentiation and myelination in vitro, and its efficacy in an extended, cuprizone-induced demyelination model. Primary mouse (C57BL/6J) OPC-containing cultures were treated with nalfurafine (0.6-200 nM), clemastine (0.01-100 μM), T3 (30 ng/mL), or vehicle for 5 days. Using immunocytochemistry and confocal microscopy, we found that nalfurafine treatment increased OPC differentiation, oligodendrocyte (OL) morphological complexity, and myelination of nanofibers in vitro. Adult male mice (C57BL/6J) were given a diet containing 0.2% cuprizone and administered rapamycin (10 mg/kg) once daily for 12 weeks followed by 6 weeks of treatment with nalfurafine (0.01 or 0.1 mg/kg), clemastine (10 mg/kg), or vehicle. We quantified the number of OLs using immunofluorescence, gross myelination using black gold staining, and myelin thickness using electron microscopy. Cuprizone + rapamycin treatment produced extensive demyelination and was accompanied by a loss of mature OLs, which was partially reversed by therapeutic administration of nalfurafine. We also assessed these mice for functional behavioral changes in open-field, horizontal bar, and mouse motor skill sequence tests (complex wheel running). Cuprizone + rapamycin treatment resulted in hyperlocomotion, poorer horizontal bar scores, and less distance traveled on the running wheels. Partial recovery was observed on both the horizontal bar and complex running wheel tests over time, which was facilitated by nalfurafine treatment. Taken together, these data highlight the potential of nalfurafine as a remyelination-promoting therapeutic.
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Rabia Bibi
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Andy Biggerstaff
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Sheein Hong
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Bria Pengelly
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Anne C La Flamme
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
8
|
Yeung J, DeYoung T, Spring S, de Guzman AE, Elder MW, Beauchamp A, Wong CS, Palmert MR, Lerch JP, Nieman BJ. Sex chromosomes and hormones independently influence healthy brain development but act similarly after cranial radiation. Proc Natl Acad Sci U S A 2024; 121:e2404042121. [PMID: 39207735 PMCID: PMC11388377 DOI: 10.1073/pnas.2404042121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The course of normal development and response to pathology are strongly influenced by biological sex. For instance, female childhood cancer survivors who have undergone cranial radiation therapy (CRT) tend to display more pronounced cognitive deficits than their male counterparts. Sex effects can be the result of sex chromosome complement (XX vs. XY) and/or gonadal hormone influence. The contributions of each can be separated using the four-core genotype mouse model (FCG), where sex chromosome complement and gonadal sex are decoupled. While studies of FCG mice have evaluated brain differences in adulthood, it is still unclear how sex chromosome and sex hormone effects emerge through development in both healthy and pathological contexts. Our study utilizes longitudinal MRI with the FCG model to investigate sex effects in healthy development and after CRT in wildtype and immune-modified Ccl2-knockout mice. Our findings in normally developing mice reveal a relatively prominent chromosome effect prepubertally, compared to sex hormone effects which largely emerge later. Spatially, sex chromosome and hormone influences were independent of one another. After CRT in Ccl2-knockout mice, both male chromosomes and male hormones similarly improved brain outcomes but did so more separately than in combination. Our findings highlight the crucial role of sex chromosomes in early development and identify roles for sex chromosomes and hormones after CRT-induced inflammation, highlighting the influences of biological sex in both normal brain development and pathology.
Collapse
Affiliation(s)
- Jonas Yeung
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - Taylor DeYoung
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
| | - A Elizabeth de Guzman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Rovereto TN 38068, Italy
| | - Madeline W Elder
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
| | - Antoine Beauchamp
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - C Shun Wong
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, Toronto ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto ON M5T 1P5, Canada
| | - Mark R Palmert
- Division of Endocrinology, The Hospital for Sick Children, University of Toronto, Toronto ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto ON M5S 1A8, Canada
- Department of Physiology, University of Toronto, Toronto ON M5S 1A8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Wellcome Centre for Integrative Neuroimaging, Medical Sciences Division, University of Oxford, Oxford, OXF OX3 9DU, United Kingdom
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, OXF OX3 9DU, United Kingdom
| | - Brian J Nieman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| |
Collapse
|
9
|
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
|
10
|
Puls K, Olivé-Marti AL, Hongnak S, Lamp D, Spetea M, Wolber G. Discovery of Novel, Selective, and Nonbasic Agonists for the Kappa-Opioid Receptor Determined by Salvinorin A-Based Virtual Screening. J Med Chem 2024; 67:13788-13801. [PMID: 39088801 PMCID: PMC11345774 DOI: 10.1021/acs.jmedchem.4c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024]
Abstract
Modulating the kappa-opioid receptor (KOR) is a promising strategy for treating various human diseases. KOR agonists show potential for treating pain, pruritus, and epilepsy, while KOR antagonists show potential for treating depression, anxiety, and addiction. The diterpenoid Salvinorin A (SalA), a secondary metabolite of Salvia divinorum, is a potent and selective KOR agonist. Unlike typical opioids, SalA lacks a basic nitrogen, which encouraged us to search for nonbasic KOR ligands. Through structure-based virtual screening using 3D pharmacophore models based on the binding mode of SalA, we identified novel, nonbasic, potent, and selective KOR agonists. In vitro studies confirmed two virtual hits, SalA-VS-07 and SalA-VS-08, as highly selective for the KOR and showing G protein-biased KOR agonist activity. Both KOR ligands share a novel spiro-moiety and a nonbasic scaffold. Our findings provide novel starting points for developing therapeutics aimed at treating pain and other conditions in which KOR is a central player.
Collapse
Affiliation(s)
- Kristina Puls
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - 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
| | - Siriwat Hongnak
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - David Lamp
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - 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
| | - Gerhard Wolber
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| |
Collapse
|
11
|
Kansagra KA, Momin T, Patel HB, Shah C, Parmar G, Ghoghari A, Patel HV, Parmar DV. Safety, tolerability, pharmacokinetics, and pharmacodynamics of a novel kappa opioid receptor agonist ZYKR1: a randomized double-blind placebo-control phase 1 study in healthy adult human participants. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4737-4745. [PMID: 38141084 DOI: 10.1007/s00210-023-02912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
To perform first-in-human single-dose escalation trial of ZYKR1, which is a potent, selective, and peripherally-restricted kappa opioid receptor agonist, is the purpose of this study. This randomized, double-blind, placebo-controlled single ascending dose study conducted at Zydus Research Centre, Ahmedabad, India included healthy male participants aged 18-55 years and weighing > 50 kg. The primary objective was to evaluate the safety and tolerability of ZYKR1. The secondary objective was to evaluate the pharmacokinetics and pharmacodynamics (PD) of ZYKR1. Participants received ZYKR1 (0.5 - 6 mcg/kg) or placebo infused intravenously in 15 ± 1 min. Of total five dose groups (0.5 - 6 mcg/kg), each group included eight participants with six and two randomized to ZYKR1 and placebo, respectively. Three participants experienced six treatment-emergent adverse events (TEAEs); two were gastrointestinal disorders (nausea and vomiting at 2 mcg/kg); and four were related to the nervous system (headache (at 2 mcg/kg) and facial tingling, facial numbness and paresthesia (at 6 mcg/kg)); all TEAEs were mild and resolved without sequelae. The Cmax of ZYKR1 was achieved after 15 - 20 min of start of infusion. The mean exposures (Cmax and AUC0 - t) increased in a dose-proportional manner. The mean t1/2 ranged from 2.20 to 2.98 h across the dose range. Increase in the mean prolactin level was significantly higher in treatment groups compared with that in the placebo group. Intravenous ZYKR1 at doses up to 6 mcg/kg showed acceptable safety and tolerability and demonstrated a short half-life with principal route of excretion as renal. ZYKR1 displayed a potent PD effect reflected by increased prolactin levels, supporting further study in patients. Trial registration Clinical Trial Registry of India: CTRI/2018/07/014927. Date of registration: 18/07/2018.
Collapse
Affiliation(s)
- Kevinkumar A Kansagra
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India.
| | | | - Hardik B Patel
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Chintan Shah
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Gordhan Parmar
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Ashok Ghoghari
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Harilal V Patel
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | | |
Collapse
|
12
|
Liu H, Huang R, Zhuo Z, Zhang X, Wu L, Guo Z, Wen F, An L, Yuan H, Zhang Y, Xu Y. Activation of kappa opioid receptor suppresses post-traumatic osteoarthritis via sequestering STAT3 on the plasma membrane. Cell Commun Signal 2024; 22:335. [PMID: 38890746 PMCID: PMC11186255 DOI: 10.1186/s12964-024-01709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Kappa opioid receptor (KOR) signaling is involved in joint development and inflammation in Osteoarthritis (OA), while the biochemical mechanism remains unclarified. This study aims to investigate downstream molecular events of KOR activation, to provide novel perspectives in OA pathology. METHODS U50,488H, a selective KOR agonist, was intra-articularly injected in mice upon destabilization of the medial meniscus (DMM) as OA models, with PBS injection as control. The behavioral and histological evaluation was assessed by hot plate test and red solid green staining, respectively. Alterations in mRNA and protein expression were assessed by RNA-seq, RT-qPCR, immunohistochemistry and western blotting (WB) in chondrocytes treated with TNF-α or TNF-α + U50,488H. Proteins interacted with KOR were explored using proximity labeling followed by mass spectrometry and then testified by co-immunoprecipitation (Co-IP) assay and immunofluorescence (IF). RESULTS OA-induced pain was reduced and cartilage degeneration was alleviated upon KOR activation in DMM mice. In chondrocytes, activation of KOR reversed the upregulation of MMPs, IL-6, IL-1β and phosphorylated(p-) STAT3, stimulated by TNF-α, while the expression of NF-κB, MAPKs and AKT signaling weren't reversed. RNA-seq and IF results presented that KOR activation evidently reduced STAT3 nuclear translocation in chondrocytes upon TNF-α stimuli. The reduction may be resulted from the binding of KOR and STAT3 in the plasma membrane, revealed by proximity labeling and Co-IP results. CONCLUSIONS KOR activation protects cartilage from OA, and this protective effect is mainly exerted via sequestering STAT3 on the plasma membrane, resulting in inactivation of STAT3-dependent immune responses which otherwise contributes to OA.
Collapse
Affiliation(s)
- Haixia Liu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Renhuan Huang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziang Zhuo
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinru Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ling Wu
- iView Therapeutics, Inc., Cranbury, NJ, USA
| | - Zhen Guo
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fuping Wen
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liwei An
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hang Yuan
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yiming Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
13
|
St. Onge C, Pagare PP, Zheng Y, Arriaga M, Stevens DL, Mendez RE, Poklis JL, Halquist MS, Selley DE, Dewey WL, Banks ML, Zhang Y. Systematic Structure-Activity Relationship Study of Nalfurafine Analogues toward Development of Potentially Nonaddictive Pain Management Treatments. J Med Chem 2024; 67:9552-9574. [PMID: 38814086 PMCID: PMC11181328 DOI: 10.1021/acs.jmedchem.4c00646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Despite the availability of numerous pain medications, the current array of Food and Drug Administration-approved options falls short in adequately addressing pain states for numerous patients and consequently worsens the opioid crisis. Thus, it is imperative for basic research to develop novel and nonaddictive pain medications. Toward addressing this clinical goal, nalfurafine (NLF) was chosen as a lead and its structure-activity relationship (SAR) systematically studied through design, syntheses, and in vivo characterization of 24 analogues. Two analogues, 21 and 23, showed longer durations of action than NLF in a warm-water tail immersion assay, produced in vivo effects primarily mediated by KOR and DOR, penetrated the blood-brain barrier, and did not function as reinforcers. Additionally, 21 produced fewer sedative effects than NLF. Taken together, these results aid the understanding of NLF SAR and provide insights for future endeavors in developing novel nonaddictive therapeutics to treat pain.
Collapse
Affiliation(s)
- Celsey
M. St. Onge
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
| | - Piyusha P. Pagare
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
| | - Yi Zheng
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
| | - Michelle Arriaga
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - David L. Stevens
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Rolando E. Mendez
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Justin L. Poklis
- Department
of Pharmaceutics, Virginia Commonwealth
University, 410 North
12th Street, Richmond, Virginia 23298, United States
| | - Matthew S. Halquist
- Department
of Pharmaceutics, Virginia Commonwealth
University, 410 North
12th Street, Richmond, Virginia 23298, United States
| | - Dana E. Selley
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - William L. Dewey
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Matthew L. Banks
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
- Institute
for Drug and Alcohol Studies, 203 East Cary Street, Richmond, Virginia 23298, United States
| |
Collapse
|
14
|
Wen Q, Sun D, Yang L, Li Y. Impact of butorphanol versus sufentanil on postoperative cognition and inflammation in elderly: a pilot study. Front Aging Neurosci 2024; 16:1395725. [PMID: 38912522 PMCID: PMC11190082 DOI: 10.3389/fnagi.2024.1395725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024] Open
Abstract
Background This randomized controlled trial aimed to compare the effects of butorphanol and sufentanil on early post-operative cognitive dysfunction (POCD) and systemic inflammation in older surgical patients. Methods Patients (aged 65 years or above) undergoing surgeries with general anesthesia were randomized to either the butorphanol group (40 μg/kg during anesthesia induction) or the sufentanil group (0.4 μg/kg). Cognitive function changes during the perioperative period were assessed using the Mini-Mental State Examination (MMSE) and the Montreal Cognitive Assessment (MoCA) scale up to 3 days after surgery. POCD was defined as a Z-score or composite Z-score greater than 1.96 for both MMSE and MoCA scores. Circulating inflammatory factors, including tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 10 (IL-10), were measured using enzyme-linked immunosorbent assay. Results The study included 114 patients (median age: 71 years, 57.7% male). Compared to sufentanil, butorphanol significantly reduced the incidence of POCD on the first (11.5% versus 32.7%, p = 0.017) and third day (3.8% versus 15.4%, p = 0.046) after surgery. Additionally, patients receiving butorphanol had significantly lower circulating levels of TNF-α and IL-1β at the time of discharge from the post-anesthesia care unit and on the first and third day after surgery (p < 0.05 for all comparisons). Furthermore, circulating IL-10 levels were significantly higher in patients receiving butorphanol (p < 0.05 for all comparisons). Conclusion Administration of butorphanol during anesthesia induction, as opposed to sufentanil, was associated with a significant reduction in the early incidence of POCD in older surgical patients, possibly attributed to its impact on systemic inflammation.Clinical trial registration: The present study was registered in the China Clinical Trial Center (ChiCTR2300070805, 24/04/2023).
Collapse
Affiliation(s)
- Qiannan Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Defeng Sun
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Yang
- Department of Neuroelectrophysiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuexian Li
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
15
|
Birgül Iyison N, Abboud C, Abboud D, Abdulrahman AO, Bondar AN, Dam J, Georgoussi Z, Giraldo J, Horvat A, Karoussiotis C, Paz-Castro A, Scarpa M, Schihada H, Scholz N, Güvenc Tuna B, Vardjan N. ERNEST COST action overview on the (patho)physiology of GPCRs and orphan GPCRs in the nervous system. Br J Pharmacol 2024. [PMID: 38825750 DOI: 10.1111/bph.16389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 06/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that play a critical role in nervous system function by transmitting signals between cells and their environment. They are involved in many, if not all, nervous system processes, and their dysfunction has been linked to various neurological disorders representing important drug targets. This overview emphasises the GPCRs of the nervous system, which are the research focus of the members of ERNEST COST action (CA18133) working group 'Biological roles of signal transduction'. First, the (patho)physiological role of the nervous system GPCRs in the modulation of synapse function is discussed. We then debate the (patho)physiology and pharmacology of opioid, acetylcholine, chemokine, melatonin and adhesion GPCRs in the nervous system. Finally, we address the orphan GPCRs, their implication in the nervous system function and disease, and the challenges that need to be addressed to deorphanize them.
Collapse
Affiliation(s)
- Necla Birgül Iyison
- Department of Molecular Biology and Genetics, University of Bogazici, Istanbul, Turkey
| | - Clauda Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | | | - Ana-Nicoleta Bondar
- Faculty of Physics, University of Bucharest, Magurele, Romania
- Forschungszentrum Jülich, Institute for Computational Biomedicine (IAS-5/INM-9), Jülich, Germany
| | - Julie Dam
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Christos Karoussiotis
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Alba Paz-Castro
- Molecular Pharmacology of GPCRs research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago, Spain
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Bilge Güvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
16
|
Bao MY, Feng CY, Li XQ, He Y, Han B, Yang YN, Zhang Y, Li X. Targeting of KOR by famotidine promotes OPC maturation differentiation and CNS remyelination via STAT3 signaling pathway. Int J Biol Macromol 2024; 269:131964. [PMID: 38692525 DOI: 10.1016/j.ijbiomac.2024.131964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
This study aims to identify FDA-approved drugs that can target the kappa-opioid receptor (KOR) for the treatment of demyelinating diseases. Demyelinating diseases are characterized by myelin sheath destruction or formation that results in severe neurological dysfunction. Remission of this disease is largely dependent on the differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLGs) in demyelinating lesions. KOR is an important regulatory protein and drug target for the treatment of demyelinating diseases. However, no drug targeting KOR has been developed due to the long clinical trials for drug discovery. Here, a structure-based virtual screening was applied to identify drugs targeting KOR among 1843 drugs of FDA-approved drug libraries, and famotidine was screen out by its high affinity cooperation with KOR as well as the clinical safety. We discovered that famotidine directly promoted OPC maturation and remyelination using the complementary in vitro and in vivo models. Administration of famotidine was not only effectively enhanced CNS myelinogenesis, but also promoted remyelination. Mechanically speaking, famotidine promoted myelinogenesis or remyelination through KOR/STAT3 signaling pathway. In general, our study provided evidence of new clinical applicability of famotidine for the treatment of demyelinating diseases for which there is currently no effective therapy.
Collapse
Affiliation(s)
- Ming-Yue Bao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Chen-Yu Feng
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiu-Qing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yan He
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Bing Han
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Ya-Na Yang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
17
|
Martucci KT. Neuroimaging of opioid effects in humans across conditions of acute administration, chronic pain therapy, and opioid use disorder. Trends Neurosci 2024; 47:418-431. [PMID: 38762362 PMCID: PMC11168870 DOI: 10.1016/j.tins.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/01/2024] [Accepted: 04/21/2024] [Indexed: 05/20/2024]
Abstract
Evidence of central nervous system (CNS) exogenous opioid effects in humans has been primarily gained through neuroimaging of three participant populations: individuals after acute opioid administration, those with opioid use disorder (OUD), and those with chronic pain receiving opioid therapy. In both the brain and spinal cord, opioids alter processes of pain, cognition, and reward. Opioid-related CNS effects may persist and accumulate with longer opioid use duration. Meanwhile, opioid-induced benefits versus risks to brain health remain unclear. This review article highlights recent accumulating evidence for how exogenous opioids impact the CNS in humans. While investigation of CNS opioid effects has remained largely disparate across contexts of opioid acute administration, OUD, and chronic pain opioid therapy, integration across these contexts may enable advancement toward effective interventions.
Collapse
Affiliation(s)
- Katherine T Martucci
- Human Affect and Pain Neuroscience Lab, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Center for Translational Pain Medicine, Duke University School of Medicine, Durham, NC, USA; Duke Institute for Brain Sciences, Duke University, Durham, NC, USA.
| |
Collapse
|
18
|
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
|
19
|
Li Z, Ye R, He Q, Lu J, Sun Y, Sun X, Tang S, Hu S, Chai J, Kong L, Liu X, Chen J, Fang Y, Lan Y, Xie Q, Liu J, Shao L, Fu W, Wang Y, Li W. Discovery of an Ortho-Substituted N-Cyclopropylmethyl-7α-phenyl-6,14- endoethano-tetrahydronorthebaine Derivative as a Selective and Potent Kappa Opioid Receptor Agonist with Subsided Sedative Effect. J Med Chem 2024. [PMID: 38647397 DOI: 10.1021/acs.jmedchem.3c02439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Research into kappa opioid receptor (KOR) agonists with attenuated central-nervous-system side effects is a critical focus for developing productive and safe analgesics. Herein, a series of ortho-substituted N-cyclopropylmethyl-7α-phenyl-6,14-endoethano-tetrahydronorthebaines were designed, synthesized, and subjected to bioassays. Compound 7a exhibited high subtype selectivity and potent agonistic activity toward KOR (KOR, Ki = 3.9 nM, MOR/KOR = 270, DOR/KOR = 1075; [35S]GTPγS binding, EC50 = 3.4 nM). Additionally, this compound exhibited robust and persistent antinociceptive effects in rodent models with different animal strains (hot plate test, ED50 = 0.20-0.30 mg/kg, i.p.; abdominal constriction test, ED50 = 0.20-0.60 mg/kg, i.p.), with its KOR-mediated mechanism for antinociception firmly established. Notably, compound 7a, unlike conventional KOR agonists, displayed minimal sedation and aversion at the antinociceptive ED50 dose. This feature addresses a crucial limitation in existing KOR agonists, positioning compound 7a as a promising novel therapeutic agent.
Collapse
Affiliation(s)
- Zixiang Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Rufeng Ye
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Qian He
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jiashuo Lu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- Department of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yanting Sun
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Xiujian Sun
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Siyuan Tang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Shuyang Hu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Jingrui Chai
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Xiaoning Liu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| | - Jing Chen
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yun Fang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yingjie Lan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
20
|
Ujcikova H, Lee YS, Roubalova L, Svoboda P. The impact of multifunctional enkephalin analogs and morphine on the protein changes in crude membrane fractions isolated from the rat brain cortex and hippocampus. Peptides 2024; 174:171165. [PMID: 38307418 DOI: 10.1016/j.peptides.2024.171165] [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: 12/11/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Endogenous opioid peptides serve as potent analgesics through the opioid receptor (OR) activation. However, they often suffer from poor metabolic stability, low lipophilicity, and low blood-brain barrier permeability. Researchers have developed many strategies to overcome the drawbacks of current pain medications and unwanted biological effects produced by the interaction with opioid receptors. Here, we tested multifunctional enkephalin analogs LYS739 (MOR/DOR agonist and KOR partial antagonist) and LYS744 (MOR/DOR agonist and KOR full antagonist) under in vivo conditions in comparison with MOR agonist, morphine. We applied 2D electrophoretic resolution to investigate differences in proteome profiles of crude membrane (CM) fractions isolated from the rat brain cortex and hippocampus exposed to the drugs (10 mg/kg, seven days). Our results have shown that treatment with analog LYS739 induced the most protein changes in cortical and hippocampal samples. The identified proteins were mainly associated with energy metabolism, cell shape and movement, apoptosis, protein folding, regulation of redox homeostasis, and signal transduction. Among these, the isoform of mitochondrial ATP synthase subunit beta (ATP5F1B) was the only protein upregulation in the hippocampus but not in the brain cortex. Contrarily, the administration of analog LYS744 caused a small number of protein alterations in both brain parts. Our results indicate that the KOR full antagonism, together with MOR/DOR agonism of multifunctional opioid ligands, can be beneficial in treating chronic pain states by reducing changes in protein expression levels but retaining analgesic efficacy.
Collapse
Affiliation(s)
- Hana Ujcikova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic.
| | - Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Lenka Roubalova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Petr Svoboda
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| |
Collapse
|
21
|
Schrader TO, Lorrain KI, Bagnol D, Edu GC, Broadhead A, Baccei C, Poon MM, Stebbins KJ, Xiong Y, Lorenzana AO, Chan JR, Green AJ, Lorrain DS, Chen A. Identification and In Vivo Evaluation of Myelination Agent PIPE-3297, a Selective Kappa Opioid Receptor Agonist Devoid of β-Arrestin-2 Recruitment Efficacy. ACS Chem Neurosci 2024; 15:685-698. [PMID: 38265210 DOI: 10.1021/acschemneuro.3c00807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024] Open
Abstract
Structure-activity relationship studies led to the discovery of PIPE-3297, a fully efficacious and selective kappa opioid receptor (KOR) agonist. PIPE-3297, a potent activator of G-protein signaling (GTPγS EC50 = 1.1 nM, 91% Emax), did not elicit a β-arrestin-2 recruitment functional response (Emax < 10%). Receptor occupancy experiments performed with the novel KOR radiotracer [3H]-PIPE-3113 revealed that subcutaneous (s.c.) administration of PIPE-3297 at 30 mg/kg in mice achieved 90% occupancy of the KOR in the CNS 1 h post dose. A single subcutaneous dose of PIPE-3297 in healthy mice produced a statistically significant increase of mature oligodendrocytes (P < 0.0001) in the KOR-enriched striatum, an effect that was not observed in animals predosed with the selective KOR antagonist norbinaltorphimine. An equivalent dose given to mice in an open-field activity-monitoring system revealed a small KOR-independent decrease in total locomotor activity versus vehicle measured between 60 and 75 min post dose. Daily doses of PIPE-3297 at both 3 and 30 mg/kg s.c. reduced the disease score in the mouse experimental autoimmune encephalomyelitis (EAE) model. Visually evoked potential (VEP) N1 latencies were also significantly improved versus vehicle in both dose groups, and latencies matched those of untreated animals. Taken together, these findings highlight the potential therapeutic value of functionally selective G-protein KOR agonists in demyelinating disease, which may avoid the sedating side effects typically associated with classical nonbiased KOR agonists.
Collapse
Affiliation(s)
- Thomas O Schrader
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Kym I Lorrain
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Didier Bagnol
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Geraldine C Edu
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Alexander Broadhead
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Christopher Baccei
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Michael M Poon
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Karin J Stebbins
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Yifeng Xiong
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Ariana O Lorenzana
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Jonah R Chan
- Department of Neurology, University of California, San Francisco, San Francisco, California 94143, United States
| | - Ari J Green
- Department of Neurology, University of California, San Francisco, San Francisco, California 94143, United States
| | - Daniel S Lorrain
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| | - Austin Chen
- Contineum Therapeutics, Suite 200, 10578 Science Center Drive, San Diego, California 92121, United States
| |
Collapse
|
22
|
Shi Y, Tao H, Li X, Zhang L, Li C, Sun W, Chu M, Chen K, Zhu P, Wang Q, Gu C, Wang L, Yang X, Hao Y. κ-Opioid receptor activation attenuates osteoarthritis synovitis by regulating macrophage polarization through the NF-κB pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:82-95. [PMID: 38013468 PMCID: PMC10875361 DOI: 10.3724/abbs.2023223] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 11/29/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent and chronic joint disease that affects the aging population, causing pain and disability. Macrophages in synovium are important mediators of synovial inflammatory activity and pathological joint pain. Previous studies have demonstrated the significant involvement of κ-opioid receptor (KOR) in the regulation of pain and inflammation. Our study reveals a significant reduction in synovial KOR expression among patients and mice with OA. Here, we find that KOR activation effectively inhibits the expressions of the LPS-induced-inflammatory cytokines TNF-α and IL-6 by inhibiting macrophage M1 phenotype. Mechanistically, KOR activation effectively suppresses the proinflammatory factor secretion of macrophages by inhibiting the translocation of NF-κB into the nucleus. Our animal experiments reveal that activation of KOR effectively alleviates knee pain and prevents synovitis progression in OA mice. Consistently, KOR administration suppresses the expressions of M1 macrophage markers and the NF-κB pathway in the synovium of the knee. Collectively, our study suggests that targeting KOR may be a viable strategy for treating OA by inhibiting synovitis and improving joint pain in affected patients.
Collapse
Affiliation(s)
- Yi Shi
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Huaqiang Tao
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversitySuzhou215000China
| | - Xueyan Li
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Liyuan Zhang
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Chunhui Li
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Wen Sun
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Miao Chu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversitySuzhou215000China
| | - Kai Chen
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversitySuzhou215000China
| | - Pengfei Zhu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversitySuzhou215000China
| | - Qiang Wang
- Department of OrthopedicsChangshu Hospital Affiliated to Soochow UniversityFirst People’s Hospital of Changshu CityChangshu215500China
| | - Chengyong Gu
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Liangliang Wang
- Department of Orthopedicsthe Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical UniversityChangzhou213000China
| | - Xing Yang
- Orthopedics and Sports Medicine CenterSuzhou Municipal HospitalNanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine CenterSuzhou Municipal HospitalNanjing Medical University Affiliated Suzhou HospitalSuzhou215000China
| |
Collapse
|
23
|
Wang A, Murphy J, Shteynman L, Daksla N, Gupta A, Bergese S. Novel Opioids in the Setting of Acute Postoperative Pain: A Narrative Review. Pharmaceuticals (Basel) 2023; 17:29. [PMID: 38256863 PMCID: PMC10819619 DOI: 10.3390/ph17010029] [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: 10/25/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Although traditional opioids such as morphine and oxycodone are commonly used in the management of acute postoperative pain, novel opioids may play a role as alternatives that provide potent pain relief while minimizing adverse effects. In this review, we discuss the mechanisms of action, findings from preclinical studies and clinical trials, and potential advantages of several novel opioids. The more established include oliceridine (biased ligand activity to activate analgesia and downregulate opioid-related adverse events), tapentadol (mu-opioid agonist and norepinephrine reuptake inhibitor), and cebranopadol (mu-opioid agonist with nociceptin opioid peptide activity)-all of which have demonstrated success in the clinical setting when compared to traditional opioids. On the other hand, dinalbuphine sebacate (DNS; semi-synthetic mu partial antagonist and kappa agonist), dual enkephalinase inhibitors (STR-324, PL37, and PL265), and endomorphin-1 analog (CYT-1010) have shown good efficacy in preclinical studies with future plans for clinical trials. Rather than relying solely on mu-opioid receptor agonism to relieve pain and risk opioid-related adverse events (ORAEs), novel opioids make use of alternative mechanisms of action to treat pain while maintaining a safer side-effect profile, such as lower incidence of nausea, vomiting, sedation, and respiratory depression as well as reduced abuse potential.
Collapse
Affiliation(s)
- Ashley Wang
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.W.); (N.D.); (A.G.)
| | - Jasper Murphy
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (J.M.); (L.S.)
| | - Lana Shteynman
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (J.M.); (L.S.)
| | - Neil Daksla
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.W.); (N.D.); (A.G.)
| | - Abhishek Gupta
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.W.); (N.D.); (A.G.)
| | - Sergio Bergese
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (A.W.); (N.D.); (A.G.)
- Department of Neurosurgery, Stony Brook University Hospital, Stony Brook, NY 11794, USA
| |
Collapse
|
24
|
Muratspahić E, Deibler K, Han J, Tomašević N, Jadhav KB, Olivé-Marti AL, Hochrainer N, Hellinger R, Koehbach J, Fay JF, Rahman MH, Hegazy L, Craven TW, Varga BR, Bhardwaj G, Appourchaux K, Majumdar S, Muttenthaler M, Hosseinzadeh P, Craik DJ, Spetea M, Che T, Baker D, Gruber CW. Design and structural validation of peptide-drug conjugate ligands of the kappa-opioid receptor. Nat Commun 2023; 14:8064. [PMID: 38052802 PMCID: PMC10698194 DOI: 10.1038/s41467-023-43718-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
Despite the increasing number of GPCR structures and recent advances in peptide design, the development of efficient technologies allowing rational design of high-affinity peptide ligands for single GPCRs remains an unmet challenge. Here, we develop a computational approach for designing conjugates of lariat-shaped macrocyclized peptides and a small molecule opioid ligand. We demonstrate its feasibility by discovering chemical scaffolds for the kappa-opioid receptor (KOR) with desired pharmacological activities. The designed De Novo Cyclic Peptide (DNCP)-β-naloxamine (NalA) exhibit in vitro potent mixed KOR agonism/mu-opioid receptor (MOR) antagonism, nanomolar binding affinity, selectivity, and efficacy bias at KOR. Proof-of-concept in vivo efficacy studies demonstrate that DNCP-β-NalA(1) induces a potent KOR-mediated antinociception in male mice. The high-resolution cryo-EM structure (2.6 Å) of the DNCP-β-NalA-KOR-Gi1 complex and molecular dynamics simulations are harnessed to validate the computational design model. This reveals a network of residues in ECL2/3 and TM6/7 controlling the intrinsic efficacy of KOR. In general, our computational de novo platform overcomes extensive lead optimization encountered in ultra-large library docking and virtual small molecule screening campaigns and offers innovation for GPCR ligand discovery. This may drive the development of next-generation therapeutics for medical applications such as pain conditions.
Collapse
Affiliation(s)
- Edin Muratspahić
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Kristine Deibler
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Novo Nordisk Research Center Seattle, Novo Nordisk A/S, 530 Fairview Ave N #5000, Seattle, WA, 97403, USA
| | - Jianming Han
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nataša Tomašević
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Kirtikumar B Jadhav
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090, Vienna, Austria
| | - 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
| | - 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
| | - Roland Hellinger
- 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, Brisbane, QLD, 4072, Australia
- School of Biomedical Sciences, Faculty for Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jonathan F Fay
- Department of Biochemistry and Molecular Biology, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Mohammad Homaidur Rahman
- Department of Pharmaceutical and Administrative Sciences, Saint Louis College of Pharmacy, University of Health Sciences & Pharmacy in St. Louis, St. Louis, MO, 63110, USA
| | - Lamees Hegazy
- Department of Pharmaceutical and Administrative Sciences, Saint Louis College of Pharmacy, University of Health Sciences & Pharmacy in St. Louis, St. Louis, MO, 63110, USA
| | - Timothy W Craven
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Balazs R Varga
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gaurav Bhardwaj
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Kevin Appourchaux
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Markus Muttenthaler
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Parisa Hosseinzadeh
- Department of Bioengineering, Knight Campus, University of Oregon, Eugene, OR, 97403, USA
| | - David J Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, 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
| | - Tao Che
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington, WA, 98195, USA.
| | - Christian W Gruber
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
25
|
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
|
26
|
Guttman-Yassky E, Facheris P, Da Rosa JC, Rothenberg-Lausell C, Del Duca E, David E, Estrada Y, Liu Y, Bose S, Chowdhury M, Munera C, Goncalves J, Nograles K, Kim BS, Lebwohl M. Oral difelikefalin reduces moderate to severe pruritus and expression of pruritic and inflammatory biomarkers in subjects with atopic dermatitis. J Allergy Clin Immunol 2023; 152:916-926. [PMID: 37453614 DOI: 10.1016/j.jaci.2023.06.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/07/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Pruritus is the most common and burdensome symptom of atopic dermatitis (AD). Pruritus-targeted treatments in AD are lacking, particularly for patients with milder skin disease. OBJECTIVE We sought to evaluate the impact of the selective κ-opioid receptor agonist difelikefalin (DFK) on pruritus intensity and pruritus- and immune-related biomarkers in subjects with moderate to severe AD-related pruritus. METHODS A phase 2 clinical trial investigated the efficacy and safety of oral DFK 0.25, 0.5, and 1.0 mg in subjects with moderate to severe AD-related pruritus. A biomarker substudy evaluated the effects of DFK on the expression of pruritus, TH2-associated genes, and skin barrier-related genes. RESULTS In the clinical trial (N = 401), all DFK doses reduced itch versus placebo; however, the results were not statistically significant at week 12. In a subgroup of subjects in the trial with mild to moderate skin inflammation and moderate to severe itch (itch-dominant AD phenotype), DFK reduced itch at week 12 versus placebo. In the biomarker substudy, DFK downregulated the expression of key pruritus-related genes (eg, IL-31 and TRPV1) and the AD phenotype (eg, CCL17). Gene set variation analysis confirmed that DFK, but not placebo, downregulated pruritus-related genes and TH2 pathways. DFK improved skin barrier integrity markers and upregulated the expression of claudins and lipid metabolism-associated genes (eg, SEC14L6, ELOVL3, CYP1A2, and AKR1D1). CONCLUSIONS DFK treatment reduced itch in subjects with moderate to severe AD-related pruritus, particularly those with an "itch-dominant" AD phenotype, and had an impact on the expression of pruritus, TH2-associated genes, and skin barrier-related genes. DFK is a promising therapy for AD-related pruritus; further clinical studies are warranted.
Collapse
Affiliation(s)
| | | | | | | | | | - Eden David
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Ying Liu
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Swaroop Bose
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | | | - Brian S Kim
- Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Mark Lebwohl
- Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
27
|
Beck TC, Wilson EM, Wilkes E, Lee LW, Norris R, Valdebran M. Kappa opioid agonists in the treatment of itch: just scratching the surface? ITCH (PHILADELPHIA, PA.) 2023; 8:e0072. [PMID: 38099236 PMCID: PMC10720604 DOI: 10.1097/itx.0000000000000072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Chronic pruritus is a debilitating condition affecting 23-44 million Americans. Recently, kappa opioid agonists (KOAs) have emerged as a novel class of potent antipruritic agents. In 2021, the Food and Drug Administration approved difelikefalin (Korsuva) for the treatment of moderate-to-severe pruritus associated with chronic kidney disease in adults undergoing hemodialysis. Difelikefalin is a potent, peripherally restricted KOA that is intravenously available. Although promising, difelikefalin is currently available as an intravenous composition only, limiting the scope of use. Oral formulations of difelikefalin did not meet the primary endpoint criteria in recent phase 2 clinical trials; however, additional clinical studies are ongoing. The future for KOAs in the treatment of pruritus is encouraging. Orally active pathway-biased KOAs, such as triazole 1.1, may serve as viable alternatives with broader applications. Extended-release compositions, such as the TP-2021 ProNeura subdermal implant, may circumvent the pharmacokinetic issues associated with peptide-based KOAs. Lastly, dual-acting kappa opioid receptor agonist/mu opioid receptor antagonists are orally bioavailable and may be useful in the treatment of various forms of chronic itch. In this review, we summarize the results of KOAs in clinical and preclinical trials and discuss future directions of drug development.
Collapse
Affiliation(s)
- Tyler C. Beck
- Department of Dermatology and Dermatological Surgery, Medical University of South Carolina, Charleston, SC
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| | - Elena M. Wilson
- Department of Dermatology and Dermatological Surgery, Medical University of South Carolina, Charleston, SC
| | - Erik Wilkes
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
| | - Lara Wine Lee
- Department of Dermatology and Dermatological Surgery, Medical University of South Carolina, Charleston, SC
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Russell Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| | - Manuel Valdebran
- Department of Dermatology and Dermatological Surgery, Medical University of South Carolina, Charleston, SC
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
28
|
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: 0] [Impact Index Per Article: 0] [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
|
29
|
Dean E, Dominique A, Palillero A, Tran A, Paradis N, Wu C. Probing the Activation Mechanisms of Agonist DPI-287 to Delta-Opioid Receptor and Novel Agonists Using Ensemble-Based Virtual Screening with Molecular Dynamics Simulations. ACS OMEGA 2023; 8:32404-32423. [PMID: 37720760 PMCID: PMC10500586 DOI: 10.1021/acsomega.3c01918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023]
Abstract
Pain drugs targeting mu-opioid receptors face major addiction problems that have caused an epidemic. The delta-opioid receptor (DOR) has shown to not cause addictive effects when bound to an agonist. While the active conformation of the DOR in complex with agonist DPI-287 has been recently solved, there are still no FDA-approved agonists targeting it, providing the opportunity for structure-based virtual screening. In this study, the conformational plasticity of the DOR was probed using molecular dynamics (MD) simulations, identifying two representative conformations from clustering analysis. The two MD conformations as well as the crystal conformation of DOR were used to screen novel compounds from the ZINC database (17 million compounds), in which 69 drugs were picked as potential compounds based on their docking scores. Notably, 37 out of the 69 compounds were obtained from the simulated conformations. The binding stability of the 69 compounds was further investigated using MD simulations. Based on the MM-GBSA binding energy and the predicted drug properties, eight compounds were chosen as the most favorable, six of which were from the simulated conformations. Using a dynamic network model, the communication between the crystal agonist and the top eight molecules with the receptor was analyzed to confirm if these novel compounds share a similar activation mechanism to the crystal ligand. Encouragingly, docking of these eight compounds to the other two opioid receptors (kappa and mu) suggests their good selectivity toward DOR.
Collapse
Affiliation(s)
- Emily Dean
- Department of Molecular &
Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - AnneMarie Dominique
- Department of Molecular &
Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Americus Palillero
- Department of Molecular &
Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Annie Tran
- Department of Molecular &
Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Nicholas Paradis
- Department of Molecular &
Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Chun Wu
- Department of Molecular &
Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
30
|
Furo H, Podichetty A, Whitted M, Zhou YY, Torres F, Brimhall BB. Association Between Opioid Use Disorder and Seizure Incidents Among Alcohol Use Disorder Patients. Subst Abuse 2023; 17:11782218231181563. [PMID: 37426877 PMCID: PMC10326460 DOI: 10.1177/11782218231181563] [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: 02/20/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023]
Abstract
Many previous studies have discussed an association between alcohol use disorder (AUD) and seizure incidents. There are also case reports of seizures during opioid withdrawals. Therefore, it is possible that AUD patients may have a higher risk of seizures if they also have opioid use disorder (OUD). However, it remains unproven whether AUD patients with a dual diagnosis of OUD have higher seizure incidents, to our knowledge. This study explored seizure incidents among the patients with a dual diagnosis of AUD and OUD as well as seizures among AUD only or OUD only patients. This study utilized de-identified data from 30 777 928 hospital inpatient encounters at 948 healthcare systems over 4 years (9/1/2018-8/31/2022) from the Vizient® Clinical Database for this study. Applying the International Classification of Diseases 10th Revision (ICD-10) diagnostic codes, AUD (1 953 575), OUD (768 982), and seizure (1 209 471) encounters were retrieved from the database to examine the effects of OUD on seizure incidence among AUD patients. This study also stratified patient encounters for demographic factors such as gender, age, and race, as well as the Vizient-categorized primary payer. Greatest gender differences were identified among AUD followed by OUD, and seizure patient groups. The mean age for seizure incidents was 57.6 years, while that of AUD was 54.7 years, and OUD 48.9 years. The greatest proportion of patients in all 3 groups were White, followed by Black, with Medicare being the most common primary payer in all 3 categories. Seizure incidents were statistically more common (P < .001, chi-square) in patients with a dual diagnosis of AUD and OUD (8.07%) compared to those with AUD only (7.55%). The patients with the dual diagnosis had a higher odd ratio than those with AUD only or OUD only. These findings across more than 900 health systems provide a greater understanding of seizure risks. Consequently, this information may help in triaging AUD and OUD patients in certain higher-risk demographic groups.
Collapse
Affiliation(s)
- Hiroko Furo
- Department of Psychiatry and Behavioral
Sciences, The University of Texas Health at San Antonio, San Antonio, TX, USA
- Department of Pathology and Laboratory
Medicine, The University of Texas Health at San Antonio, San Antonio, TX, USA
- Department of Biomedical Informatics,
State University of New York (SUNY) at Buffalo, Buffalo, NY, USA
- Department of Family Medicine, State
University of New York (SUNY) at Buffalo, Buffalo, NY, USA
| | - Ankita Podichetty
- McWilliams School of Biomedical
Informatics, The University of Texas Health Science Center at Houston, Houston, TX,
USA
| | - Marisa Whitted
- Department of Pathology and Laboratory
Medicine, The University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Yi Yuan Zhou
- Department of Pathology and Laboratory
Medicine, The University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Francis Torres
- Department of Pathology and Laboratory
Medicine, University Health System, San Antonio, TX, USA
| | - Bradley B. Brimhall
- Department of Pathology and Laboratory
Medicine, The University of Texas Health at San Antonio, San Antonio, TX, USA
| |
Collapse
|
31
|
van de Wetering R, Ewald A, Welsh S, Kornberger L, Williamson SE, McElroy BD, Butelman ER, Prisinzano TE, Kivell BM. The Kappa Opioid Receptor Agonist 16-Bromo Salvinorin A Has Anti-Cocaine Effects without Significant Effects on Locomotion, Food Reward, Learning and Memory, or Anxiety and Depressive-like Behaviors. Molecules 2023; 28:4848. [PMID: 37375403 DOI: 10.3390/molecules28124848] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Kappa opioid receptor (KOR) agonists have preclinical antipsychostimulant effects; however, adverse side effects have limited their therapeutic development. In this preclinical study, conducted in Sprague Dawley rats, B6-SJL mice, and non-human primates (NHPs), we evaluated the G-protein-biased analogue of salvinorin A (SalA), 16-bromo salvinorin A (16-BrSalA), for its anticocaine effects, side effects, and activation of cellular signaling pathways. 16-BrSalA dose-dependently decreased the cocaine-primed reinstatement of drug-seeking behavior in a KOR-dependent manner. It also decreased cocaine-induced hyperactivity, but had no effect on responding for cocaine on a progressive ratio schedule. Compared to SalA, 16-BrSalA had an improved side effect profile, with no significant effects in the elevated plus maze, light-dark test, forced swim test, sucrose self-administration, or novel object recognition; however, it did exhibit conditioned aversive effects. 16-BrSalA increased dopamine transporter (DAT) activity in HEK-293 cells coexpressing DAT and KOR, as well as in rat nucleus accumbens and dorsal striatal tissue. 16-BrSalA also increased the early phase activation of extracellular-signal-regulated kinases 1 and 2, as well as p38 in a KOR-dependent manner. In NHPs, 16-BrSalA caused dose-dependent increases in the neuroendocrine biomarker prolactin, similar to other KOR agonists, at doses without robust sedative effects. These findings highlight that G-protein-biased structural analogues of SalA can have improved pharmacokinetic profiles and fewer side effects while maintaining their anticocaine effects.
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Amy Ewald
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Susan Welsh
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Lindsay Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA
| | - Samuel E Williamson
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
32
|
Margolis EB, Moulton MG, Lambeth PS, O'Meara MJ. The life and times of endogenous opioid peptides: Updated understanding of synthesis, spatiotemporal dynamics, and the clinical impact in alcohol use disorder. Neuropharmacology 2023; 225:109376. [PMID: 36516892 PMCID: PMC10548835 DOI: 10.1016/j.neuropharm.2022.109376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The opioid G-protein coupled receptors (GPCRs) strongly modulate many of the central nervous system structures that contribute to neurological and psychiatric disorders including pain, major depressive disorder, and substance use disorders. To better treat these and related diseases, it is essential to understand the signaling of their endogenous ligands. In this review, we focus on what is known and unknown about the regulation of the over two dozen endogenous peptides with high affinity for one or more of the opioid receptors. We briefly describe which peptides are produced, with a particular focus on the recently proposed possible synthesis pathways for the endomorphins. Next, we describe examples of endogenous opioid peptide expression organization in several neural circuits and how they appear to be released from specific neural compartments that vary across brain regions. We discuss current knowledge regarding the strength of neural activity required to drive endogenous opioid peptide release, clues about how far peptides diffuse from release sites, and their extracellular lifetime after release. Finally, as a translational example, we discuss the mechanisms of action of naltrexone (NTX), which is used clinically to treat alcohol use disorder. NTX is a synthetic morphine analog that non-specifically antagonizes the action of most endogenous opioid peptides developed in the 1960s and FDA approved in the 1980s. We review recent studies clarifying the precise endogenous activity that NTX prevents. Together, the works described here highlight the challenges and opportunities the complex opioid system presents as a therapeutic target.
Collapse
Affiliation(s)
- Elyssa B Margolis
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
| | - Madelyn G Moulton
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Philip S Lambeth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
33
|
Stefanucci A, Minosi P, Pieretti S, Tanguturi P, Molnar G, Scioli G, Marinaccio L, Della Valle A, Streicher JM, Mollica A. Design of Analgesic Trivalent Peptides with Low Withdrawal Symptoms: Probing the Antinociceptive Profile of Novel Linear and Cyclic Peptides as Opioid Pan Ligands. ACS Chem Neurosci 2023; 14:506-515. [PMID: 36651179 DOI: 10.1021/acschemneuro.3c00005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The discovery of efficacious and safe analgesics with reduced side effects is the foremost challenge in the pain field. In this work, we report the in vitro and in vivo evaluation of linear and cyclic analogues of biphalin with the aim to complete the series of structural modifications previously applied in the development of opioid peptides incorporating a xylene bridge. Replacement of Tyr1,1' by Dmt (2,5-dimethyltyrosine) in the linear biphalin analogue AM94 and cyclic analogue MACE4 resulted in two new compounds (namely, MJ2 and MJ5) endowed with improved KOR/MOR/DOR binding affinity. Both compounds showed a strong antinociceptive profile in in vivo models of nociception, allodynia, and hyperalgesia via the tail flick, hot plate, and formalin tests after intracerebroventricular and subcutaneous administration. One of these ligands, MJ2, was also tested in tolerance and dependence studies, exhibiting very little withdrawal symptoms.
Collapse
Affiliation(s)
- Azzurra Stefanucci
- Dipartimento di Farmacia, Universitá; di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - Paola Minosi
- Centro Nazionale Ricerca e Valutazione Preclinica e Clinica dei Farmaci, Istituto Superiore di Sanita, Viale Regina Elena 299, 00161 Rome, Italy
| | - Stefano Pieretti
- Centro Nazionale Ricerca e Valutazione Preclinica e Clinica dei Farmaci, Istituto Superiore di Sanita, Viale Regina Elena 299, 00161 Rome, Italy
| | | | - Gabriella Molnar
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85012, United States
| | - Giuseppe Scioli
- Dipartimento di Farmacia, Universitá; di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - Lorenza Marinaccio
- Dipartimento di Farmacia, Universitá; di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - Alice Della Valle
- Dipartimento di Farmacia, Universitá; di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| | - John M Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85012, United States.,Comprehensive Pain and Addiction Center, University of Arizona, Tucson, Arizona 85012, United States
| | - Adriano Mollica
- Dipartimento di Farmacia, Universitá; di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
34
|
Santino F, Gentilucci L. Design of κ-Opioid Receptor Agonists for the Development of Potential Treatments of Pain with Reduced Side Effects. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010346. [PMID: 36615540 PMCID: PMC9822356 DOI: 10.3390/molecules28010346] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
The κ-opioid receptor (KOR) has recently emerged as an alternative therapeutic target for the development of pain medications, without deleterious side effects associated with the μ-opioid receptor (MOR). However, modulation of KOR is currently under investigation for the treatment of depression, mood disorders, psychiatric comorbidity, and specific drug addictions. However, KOR agonists also trigger adverse effects including sedation, dysphoria, and hallucinations. In this respect, there is currently much debate on alternative paradigms. Recent effort has been devoted in search of biased ligands capable of selectively activating favorable signaling over signaling associated with unwanted side effects. On the other hand, the use of partial agonists is expected to allow the analgesia to be produced at dosages lower than those required to produce the adverse effects. More empirically, the unwanted central effects can be also avoided by using peripherally restricted agonists. In this review, we discuss the more recent trends in the design of KOR-selective, biased or partial, and finally, peripherally acting agonists. Special emphasis is given on the discussion of the most recent approaches for controlling functional selectivity of KOR-specific ligands.
Collapse
|
35
|
Dumitrascuta M, Martin C, Ballet S, Spetea M. Bifunctional Peptidomimetic G Protein-Biased Mu-Opioid Receptor Agonist and Neuropeptide FF Receptor Antagonist KGFF09 Shows Efficacy in Visceral Pain without Rewarding Effects after Subcutaneous Administration in Mice. Molecules 2022; 27:8785. [PMID: 36557917 PMCID: PMC9780937 DOI: 10.3390/molecules27248785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
There is still an unmet clinical need to develop new pharmaceuticals for effective and safe pain management. Current pharmacotherapy offers unsatisfactory solutions due to serious side effects related to the chronic use of opioid drugs. Prescription opioids produce analgesia through activation of the mu-opioid receptor (MOR) and are major contributors to the current opioid crisis. Multifunctional ligands possessing activity at more than one receptor represent a prominent therapeutic approach for the treatment of pain with fewer adverse effects. We recently reported on the design of a bifunctional MOR agonist/neuropeptide FF receptor (NPFFR) antagonist peptididomimetic, KGFF09 (H-Dmt-DArg-Aba-βAla-Bpa-Phe-NH2), and its antinociceptive effects after subcutaneous (s.c.) administration in acute and persistent pain in mice with reduced propensity for unwanted side effects. In this study, we further investigated the antinociceptive properties of KGFF09 in a mouse model of visceral pain after s.c. administration and the potential for opioid-related liabilities of rewarding and sedation/locomotor dysfunction following chronic treatment. KGFF09 produced a significant dose-dependent inhibition of the writhing behavior in the acetic acid-induced writhing assay with increased potency when compared to morphine. We also demonstrated the absence of harmful effects caused by typical MOR agonists, i.e., rewarding effects (conditioned-place preference test) and sedation/locomotor impairment (open-field test), at a dose shown to be highly effective in inhibiting pain behavior. Consequently, KGFF09 displayed a favorable benefit/side effect ratio regarding these opioid-related side effects compared to conventional opioid analgesics, such as morphine, underlining the development of dual MOR agonists/NPFFR antagonists as improved treatments for various pain conditions.
Collapse
Affiliation(s)
- Maria Dumitrascuta
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Charlotte Martin
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Steven Ballet
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - 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
|
36
|
Lee YS. Peptidomimetics and Their Applications for Opioid Peptide Drug Discovery. Biomolecules 2022; 12:biom12091241. [PMID: 36139079 PMCID: PMC9496382 DOI: 10.3390/biom12091241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Despite various advantages, opioid peptides have been limited in their therapeutic uses due to the main drawbacks in metabolic stability, blood-brain barrier permeability, and bioavailability. Therefore, extensive studies have focused on overcoming the problems and optimizing the therapeutic potential. Currently, numerous peptide-based drugs are being marketed thanks to new synthetic strategies for optimizing metabolism and alternative routes of administration. This tutorial review briefly introduces the history and role of natural opioid peptides and highlights the key findings on their structure-activity relationships for the opioid receptors. It discusses details on opioid peptidomimetics applied to develop therapeutic candidates for the treatment of pain from the pharmacological and structural points of view. The main focus is the current status of various mimetic tools and the successful applications summarized in tables and figures.
Collapse
Affiliation(s)
- Yeon Sun Lee
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|