Your browser doesn't support javascript.
loading
A µ-opioid receptor modulator that works cooperatively with naloxone.
O'Brien, Evan S; Rangari, Vipin Ashok; El Daibani, Amal; Eans, Shainnel O; Hammond, Haylee R; White, Elizabeth; Wang, Haoqing; Shiimura, Yuki; Krishna Kumar, Kaavya; Jiang, Qianru; Appourchaux, Kevin; Huang, Weijiao; Zhang, Chensong; Kennedy, Brandon J; Mathiesen, Jesper M; Che, Tao; McLaughlin, Jay P; Majumdar, Susruta; Kobilka, Brian K.
Afiliación
  • O'Brien ES; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
  • Rangari VA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, USA.
  • El Daibani A; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, USA.
  • Eans SO; Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA.
  • Hammond HR; Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA.
  • White E; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
  • Wang H; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
  • Shiimura Y; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
  • Krishna Kumar K; Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan.
  • Jiang Q; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
  • Appourchaux K; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, USA.
  • Huang W; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, USA.
  • Zhang C; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
  • Kennedy BJ; Division of CryoEM and Bioimaging, SSRL, SLAC National Acceleration Laboratory, Menlo Park, CA, USA.
  • Mathiesen JM; Lotus Separations, Princeton University, Princeton, NJ, USA.
  • Che T; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
  • McLaughlin JP; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, USA.
  • Majumdar S; Department of Pharmacodynamics, University of Florida, Gainesville, FL, USA. jpmclaughlin@ufl.edu.
  • Kobilka BK; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy at St Louis and Washington University School of Medicine, St Louis, MO, USA. susrutam@wustl.edu.
Nature ; 631(8021): 686-693, 2024 Jul.
Article en En | MEDLINE | ID: mdl-38961287
ABSTRACT
The µ-opioid receptor (µOR) is a well-established target for analgesia1, yet conventional opioid receptor agonists cause serious adverse effects, notably addiction and respiratory depression. These factors have contributed to the current opioid overdose epidemic driven by fentanyl2, a highly potent synthetic opioid. µOR negative allosteric modulators (NAMs) may serve as useful tools in preventing opioid overdose deaths, but promising chemical scaffolds remain elusive. Here we screened a large DNA-encoded chemical library against inactive µOR, counter-screening with active, G-protein and agonist-bound receptor to 'steer' hits towards conformationally selective modulators. We discovered a NAM compound with high and selective enrichment to inactive µOR that enhances the affinity of the key opioid overdose reversal molecule, naloxone. The NAM works cooperatively with naloxone to potently block opioid agonist signalling. Using cryogenic electron microscopy, we demonstrate that the NAM accomplishes this effect by binding a site on the extracellular vestibule in direct contact with naloxone while stabilizing a distinct inactive conformation of the extracellular portions of the second and seventh transmembrane helices. The NAM alters orthosteric ligand kinetics in therapeutically desirable ways and works cooperatively with low doses of naloxone to effectively inhibit various morphine-induced and fentanyl-induced behavioural effects in vivo while minimizing withdrawal behaviours. Our results provide detailed structural insights into the mechanism of negative allosteric modulation of the µOR and demonstrate how this can be exploited in vivo.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Receptores Opioides mu / Evaluación Preclínica de Medicamentos / Bibliotecas de Moléculas Pequeñas / Analgésicos Opioides / Naloxona Idioma: En Revista: Nature Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Receptores Opioides mu / Evaluación Preclínica de Medicamentos / Bibliotecas de Moléculas Pequeñas / Analgésicos Opioides / Naloxona Idioma: En Revista: Nature Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos