open access publication

Article, 2024

Structural insights into vesicular monoamine storage and drug interactions

Nature, ISSN 0028-0836, 1476-4687, Volume 629, 8010, Pages 235-243, 10.1038/s41586-024-07290-7

Contributors

Ye, Jin 0000-0003-1004-5344 [1] Chen, Huaping [1] Wang, Kaituo 0000-0002-5922-7109 [2] Wang, Yi [1] Ammerman, Aaron [1] Awasthi, Samjhana [1] Xu, Jinbin 0000-0002-2120-8287 [1] Liu, Bin 0000-0002-6581-780X (Corresponding author) [3] Li, Weikai (Corresponding author) [1]

Affiliations

  1. [1] Washington University in St. Louis
  2. [NORA names: United States; America, North; OECD];
  3. [2] University of Copenhagen
  4. [NORA names: KU University of Copenhagen; University; Denmark; Europe, EU; Nordic; OECD];
  5. [3] University of Minnesota
  6. [NORA names: United States; America, North; OECD]

Abstract

Biogenic monoamines—vital transmitters orchestrating neurological, endocrinal and immunological functions1–5—are stored in secretory vesicles by vesicular monoamine transporters (VMATs) for controlled quantal release6,7. Harnessing proton antiport, VMATs enrich monoamines around 10,000-fold and sequester neurotoxicants to protect neurons8–10. VMATs are targeted by an arsenal of therapeutic drugs and imaging agents to treat and monitor neurodegenerative disorders, hypertension and drug addiction1,8,11–16. However, the structural mechanisms underlying these actions remain unclear. Here we report eight cryo-electron microscopy structures of human VMAT1 in unbound form and in complex with four monoamines (dopamine, noradrenaline, serotonin and histamine), the Parkinsonism-inducing MPP+, the psychostimulant amphetamine and the antihypertensive drug reserpine. Reserpine binding captures a cytoplasmic-open conformation, whereas the other structures show a lumenal-open conformation stabilized by extensive gating interactions. The favoured transition to this lumenal-open state contributes to monoamine accumulation, while protonation facilitates the cytoplasmic-open transition and concurrently prevents monoamine binding to avoid unintended depletion. Monoamines and neurotoxicants share a binding pocket that possesses polar sites for specificity and a wrist-and-fist shape for versatility. Variations in this pocket explain substrate preferences across the SLC18 family. Overall, these structural insights and supporting functional studies elucidate the mechanism of vesicular monoamine transport and provide the basis to develop therapeutics for neurodegenerative diseases and substance abuse.

Keywords

MPP+, VMAT1, abuse, accumulation, action, agents, amphetamine, antihypertensive drug reserpine, antiporter, binding, binding pocket, complex, conformation, cryo-electron, cryo-electron microscopy structure, depletion, disease, disorders, drug, drug interactions, drug reserpine, family, form, functional studies, functions1, gate, gating interactions, hypertension, images, imaging agents, insights, interaction, mechanism, monoamine, monoamine accumulation, monoamine storage, monoamine transporters, neurodegenerative diseases, neurodegenerative disorders, neurotoxicants, pocket, polar sites, preferences, proton, proton antiporter, psychostimulant amphetamine, psychostimulants, reserpine, reserpine binding, secretory vesicles, shape, sites, specificity, state, storage, structural insights, structural mechanics, structure, study, substance abuse, substances, substrate, substrate preference, therapeutic drugs, therapeutics, transition, transmitter, transport, unbound form, variation, versatility, vesicles, vesicular monoamine transporter

Funders

  • Lundbeck Foundation
  • American Heart Association
  • W. M. Keck Foundation
  • National Institute of Allergy and Infectious Diseases
  • National Institute of General Medical Sciences
  • National Institute of Neurological Disorders and Stroke
  • National Heart Lung and Blood Institute

Data Provider: Digital Science