1
|
Sun X, Yin L, Qiao Z, Younus M, Chen G, Wu X, Li J, Kang X, Xu H, Zhou L, Li Y, Gao M, Du X, Hang Y, Lin Z, Sun L, Wang Q, Jiao R, Wang L, Hu M, Wang Y, Huang R, Li Y, Wu Q, Shang S, Guo S, Lei Q, Shu H, Zheng L, Wang S, Zhu F, Zuo P, Liu B, Wang C, Zhang Q, Zhou Z. Action Potential Firing Patterns Regulate Dopamine Release via Voltage-Sensitive Dopamine D2 Autoreceptors in Mouse Striatum In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2412229. [PMID: 39731325 DOI: 10.1002/advs.202412229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/09/2024] [Indexed: 12/29/2024]
Abstract
Dopamine (DA) in the striatum is vital for motor and cognitive behaviors. Midbrain dopaminergic neurons generate both tonic and phasic action potential (AP) firing patterns in behavior mice. Besides AP numbers, whether and how different AP firing patterns per se modulate DA release remain largely unknown. Here by using in vivo and ex vivo models, it is shown that the AP frequency per se modulates DA release through the D2 receptor (D2R), which contributes up to 50% of total DA release. D2R has a voltage-sensing site at D131 and can be deactivated in a frequency-dependent manner by membrane depolarization. This voltage-dependent D2R inhibition of DA release is mediated via the facilitation of voltage-gated Ca2+ channels (VGCCs). Collectively, this work establishes a novel mechanism that APs per se modulate DA overflow by disinhibiting the voltage-sensitive autoreceptor D2R and thus the facilitation of VGCCs, providing a pivotal pathway and insight into mammalian DA-dependent functions in vivo.
Collapse
Affiliation(s)
- Xiaoxuan Sun
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Lili Yin
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhongjun Qiao
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Muhammad Younus
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Guoqing Chen
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xi Wu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Jie Li
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xinjiang Kang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Huadong Xu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Li Zhou
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yinglin Li
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Min Gao
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Xingyu Du
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yuqi Hang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhaohan Lin
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Liyuan Sun
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Qinglong Wang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Ruiying Jiao
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Lun Wang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Meiqin Hu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yuan Wang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Rong Huang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yiman Li
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Qihui Wu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Shujiang Shang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Shu Guo
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Qian Lei
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Haifeng Shu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Lianghong Zheng
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Shirong Wang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Feipeng Zhu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Panli Zuo
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Bing Liu
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Changhe Wang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Neuroscience Research Center, Institute of Mitochondrial Biology and Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, and the Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Quanfeng Zhang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center and Institute of Molecular Medicine, College of Future Technology, Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| |
Collapse
|
2
|
Rinne A, Bünemann M. Charge Movements and Conformational Changes: Biophysical Properties and Physiology of Voltage-Dependent GPCRs. Biomolecules 2024; 14:1652. [PMID: 39766359 PMCID: PMC11674552 DOI: 10.3390/biom14121652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
G protein-coupled receptors (GPCRs) regulate multiple cellular functions and represent important drug targets. More than 20 years ago, it was noted that GPCR activation (agonist binding) and signaling (G protein activation) are dependent on the membrane potential (VM). While it is now proven that many GPCRs display an intrinsic voltage dependence, the molecular mechanisms of how GPCRs sense depolarization of the plasma membrane are less well defined. This review summarizes the current knowledge of voltage-dependent signaling in GPCRs. We describe how voltage dependence was discovered in muscarinic receptors, present an overview of GPCRs that are regulated by voltage, and show how biophysical properties of GPCRs led to the discovery of voltage-sensing mechanisms in those receptors. Furthermore, we summarize physiological functions that have been shown to be regulated by voltage-dependent GPCR signaling of endogenous receptors in excitable tissues, such as the nervous system or the heart. Finally, we discuss challenges that remain in analyzing voltage-dependent signaling of GPCRs in vivo and present an outlook on experimental applications of the interesting concept of GPCR signaling.
Collapse
Affiliation(s)
- Andreas Rinne
- Department of Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy Bucharest, 050474 Bucharest, Romania
| | - Moritz Bünemann
- Institute of Pharmacology and Clinical Pharmacy, Biochemical Pharmaceutical Center (BPC) Marburg, University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
3
|
Boutonnet M, Bünemann M, Perroy J. The voltage sensitivity of G-protein coupled receptors: Unraveling molecular mechanisms and physiological implications. Pharmacol Ther 2024; 264:108741. [PMID: 39489434 DOI: 10.1016/j.pharmthera.2024.108741] [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: 07/16/2024] [Revised: 09/11/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
In the landscape of proteins controlled by membrane voltage (Vm), like voltage-gated ionotropic channels, the emergence of the voltage sensitivity within the vast family of G-protein coupled receptors (GPCRs) marked a significant milestone at the onset of the 21st century. Since its discovery, extensive research has been devoted to understanding the intricate relationship between Vm and GPCRs. Approximately 30 GPCRs out of a family comprising more than 800 receptors have been implicated in Vm-dependent positive and negative regulation. GPCRs stand out as the quintessential regulators of synaptic transmission in neurons, where they encounter substantial variations in Vm. However, the molecular mechanism underlying the Vm sensor of GPCRs remains enigmatic, hindered by the scarcity of mutant GPCRs insensitive to Vm yet functionally intact, impeding a comprehensive understanding of this unique property in physiology. Nevertheless, two decades of dedicated research have furnished numerous insights into the molecular aspects of GPCR Vm-sensing, accompanied by recently proposed physiological roles as well as pharmacological potential, which we encapsulate in this review. The Vm sensitivity of GPCRs emerges as a pivotal attribute, shedding light on previously unforeseen roles in synaptic transmission and extending beyond, underscoring its significance in cellular signaling and physiological processes.
Collapse
Affiliation(s)
- Marin Boutonnet
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Julie Perroy
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
4
|
Maldifassi MC, Guerra-Fernández MJ, Ponce D, Alfonso-Bueno S, Maripillán J, Vielma AH, Báez-Matus X, Marengo FD, Acuña-Castillo C, Sáez JC, Martínez AD, Cárdenas AM. Autocrine activation of P2X7 receptors mediates catecholamine secretion in chromaffin cells. Br J Pharmacol 2024; 181:2905-2922. [PMID: 38679932 DOI: 10.1111/bph.16371] [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: 10/20/2022] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE ATP is highly accumulated in secretory vesicles and secreted upon exocytosis from neurons and endocrine cells. In adrenal chromaffin granules, intraluminal ATP reaches concentrations over 100 mM. However, how these large amounts of ATP contribute to exocytosis has not been investigated. EXPERIMENTAL APPROACH Exocytotic events in bovine and mouse adrenal chromaffin cells were measured with single cell amperometry. Cytosolic Ca2+ measurements were carried out in Fluo-4 loaded cells. Submembrane Ca2+ was examined in PC12 cells transfected with a membrane-tethered Ca2+ indicator Lck-GCaMP3. ATP release was measured using the luciferin/luciferase assay. Knockdown of P2X7 receptors was induced with short interfering RNA (siRNA). Direct Ca2+ influx through this receptor was measured using a P2X7 receptor-GCamp6 construct. KEY RESULTS ATP induced exocytosis in chromaffin cells, whereas the ectonucleotidase apyrase reduced the release events induced by the nicotinic agonist dimethylphenylpiperazinium (DMPP), high KCl, or ionomycin. The purinergic agonist BzATP also promoted a secretory response that was dependent on extracellular Ca2+. A740003, a P2X7 receptor antagonist, abolished secretory responses of these secretagogues. Exocytosis was also diminished in chromaffin cells when P2X7 receptors were silenced using siRNAs and in cells of P2X7 receptor knockout mice. In PC12 cells, DMPP induced ATP release, triggering Ca2+ influx through P2X7 receptors. Furthermore, BzATP, DMPP, and KCl allowed the formation of submembrane Ca2+ microdomains inhibited by A740003. CONCLUSION AND IMPLICATIONS Autocrine activation of P2X7 receptors constitutes a crucial feedback system that amplifies the secretion of catecholamines in chromaffin cells by favouring submembrane Ca2+ microdomains.
Collapse
Affiliation(s)
- María Constanza Maldifassi
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - María José Guerra-Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniela Ponce
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Samuel Alfonso-Bueno
- Instituto de Fisiología, Biología Molecular y Neurociencias. CONICET. Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jaime Maripillán
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alex H Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ximena Báez-Matus
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Fernando D Marengo
- Instituto de Fisiología, Biología Molecular y Neurociencias. CONICET. Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Claudio Acuña-Castillo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, USACH, Santiago, Chile
| | - Juan C Sáez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
5
|
Boutonnet M, Carpena C, Bouquier N, Chastagnier Y, Font-Ingles J, Moutin E, Tricoire L, Chemin J, Perroy J. Voltage tunes mGlu 5 receptor function, impacting synaptic transmission. Br J Pharmacol 2024; 181:1793-1811. [PMID: 38369690 DOI: 10.1111/bph.16317] [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: 09/07/2023] [Revised: 12/08/2023] [Accepted: 12/29/2023] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND AND PURPOSE Voltage sensitivity is a common feature of many membrane proteins, including some G-protein coupled receptors (GPCRs). However, the functional consequences of voltage sensitivity in GPCRs are not well understood. EXPERIMENTAL APPROACH In this study, we investigated the voltage sensitivity of the post-synaptic metabotropic glutamate receptor mGlu5 and its impact on synaptic transmission. Using biosensors and electrophysiological recordings in non-excitable HEK293T cells or neurons. KEY RESULTS We found that mGlu5 receptor function is optimal at resting membrane potentials. We observed that membrane depolarization significantly reduced mGlu5 receptor activation, Gq-PLC/PKC stimulation, Ca2+ release and mGlu5 receptor-gated currents through transient receptor potential canonical, TRPC6, channels or glutamate ionotropic NMDA receptors. Notably, we report a previously unknown activity of the NMDA receptor at the resting potential of neurons, enabled by mGlu5. CONCLUSIONS AND IMPLICATIONS Our findings suggest that mGlu5 receptor activity is directly regulated by membrane voltage which may have a significant impact on synaptic processes and pathophysiological functions.
Collapse
Affiliation(s)
- Marin Boutonnet
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Camille Carpena
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Yan Chastagnier
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Joan Font-Ingles
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
- SpliceBio, Barcelona, Spain
| | - Enora Moutin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Ludovic Tricoire
- Neuroscience Paris Seine, Institut de biologie Paris Seine, Sorbonne universite, CNRS, INSERM, Paris, France
| | - Jean Chemin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
6
|
Liu W, Gao T, Li N, Shao S, Liu B. Vesicle fusion and release in neurons under dynamic mechanical equilibrium. iScience 2024; 27:109793. [PMID: 38736547 PMCID: PMC11088343 DOI: 10.1016/j.isci.2024.109793] [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] [Indexed: 05/14/2024] Open
Abstract
Vesicular fusion plays a pivotal role in cellular processes, involving stages like vesicle trafficking, fusion pore formation, content release, and membrane integration or separation. This dynamic process is regulated by a complex interplay of protein assemblies, osmotic forces, and membrane tension, which together maintain a mechanical equilibrium within the cell. Changes in cellular mechanics or external pressures prompt adjustments in this equilibrium, highlighting the system's adaptability. This review delves into the synergy between intracellular proteins, structural components, and external forces in facilitating vesicular fusion and release. It also explores how cells respond to mechanical stress, maintaining equilibrium and offering insights into vesicle fusion mechanisms and the development of neurological disorders.
Collapse
Affiliation(s)
- Wenhao Liu
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
| | - Tianyu Gao
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
| | - Na Li
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Shuai Shao
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Bo Liu
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Faculty of Medicine, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
7
|
Tauber M, Ben-Chaim Y. Voltage Sensors Embedded in G Protein-Coupled Receptors. Int J Mol Sci 2024; 25:5295. [PMID: 38791333 PMCID: PMC11120775 DOI: 10.3390/ijms25105295] [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: 04/18/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Some signaling processes mediated by G protein-coupled receptors (GPCRs) are modulated by membrane potential. In recent years, increasing evidence that GPCRs are intrinsically voltage-dependent has accumulated. A recent publication challenged the view that voltage sensors are embedded in muscarinic receptors. Herein, we briefly discuss the evidence that supports the notion that GPCRs themselves are voltage-sensitive proteins and an alternative mechanism that suggests that voltage-gated sodium channels are the voltage-sensing molecules involved in such processes.
Collapse
Affiliation(s)
| | - Yair Ben-Chaim
- Department of Natural Sciences, The Open University of Israel, Ra’anana 4353701, Israel
| |
Collapse
|
8
|
Artalejo AR, Arribas-Blázquez M, Barahona MV, Llorente-Sáez C, Olivos-Oré LA. María Teresa Miras Portugal: a pioneer in the study of purinoceptors in chromaffin cells. Purinergic Signal 2024; 20:109-113. [PMID: 36941507 PMCID: PMC10997576 DOI: 10.1007/s11302-023-09934-1] [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: 02/06/2023] [Accepted: 03/09/2023] [Indexed: 03/23/2023] Open
Abstract
María Teresa Miras Portugal devoted most of her scientific life to the study of purinergic signalling. In an important part of her work, she used a model system: the chromaffin cells of the adrenal medulla. It was in these cells that she identified diadenosine polyphosphates, from which she proceeded to the study of adrenomedullary purinome: nucleotide synthesis and degradation, adenosine transport, nucleotide uptake into chromaffin granules, exocytotic release of nucleotides and autocrine regulation of chromaffin cell function via purinoceptors. This short review will focus on the current state of knowledge of the purinoceptors of adrenal chromaffin cells, a subject to which María Teresa made seminal contributions and which she continued to study until the end of her scientific life.
Collapse
Affiliation(s)
- Antonio R Artalejo
- Departament of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040, Madrid, Spain.
| | - Marina Arribas-Blázquez
- Departament of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040, Madrid, Spain
| | - María Victoria Barahona
- Departament of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040, Madrid, Spain
| | - Celia Llorente-Sáez
- Departament of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040, Madrid, Spain
| | - Luis Alcides Olivos-Oré
- Departament of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040, Madrid, Spain
| |
Collapse
|
9
|
López-Correa JM, König C, Vellido A. GPCR molecular dynamics forecasting using recurrent neural networks. Sci Rep 2023; 13:20995. [PMID: 38017062 PMCID: PMC10684758 DOI: 10.1038/s41598-023-48346-4] [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: 04/13/2023] [Accepted: 11/25/2023] [Indexed: 11/30/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are a large superfamily of cell membrane proteins that play an important physiological role as transmitters of extracellular signals. Signal transmission through the cell membrane depends on conformational changes in the transmembrane region of the receptor, which makes the investigation of the dynamics in these regions particularly relevant. Molecular dynamics (MD) simulations provide a wealth of data about the structure, dynamics, and physiological function of biological macromolecules by modelling the interactions between their atomic constituents. In this study, a Recurrent and Convolutional Neural Network (RNN) model, namely Long Short-Term Memory (LSTM), is used to predict the dynamics of two GPCR states and three specific simulations of each one, through their activation path and focussing on specific receptor regions. Active and inactive states of the GPCRs are analysed in six scenarios involving APO, Full Agonist (BI 167107) and Partial Inverse Agonist (carazolol) of the receptor. Four Machine Learning models with increasing complexity in terms of neural network architecture are evaluated, and their results discussed. The best method achieves an overall RMSD lower than 0.139 Å and the transmembrane helices are the regions showing the minimum prediction errors and minimum relative movements of the protein.
Collapse
Affiliation(s)
| | - Caroline König
- Universitat Politècnica de Catalunya, Barcelona, Spain
- IDEAI-UPC - Research Center, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Alfredo Vellido
- Universitat Politècnica de Catalunya, Barcelona, Spain.
- IDEAI-UPC - Research Center, Universitat Politècnica de Catalunya, Barcelona, Spain.
| |
Collapse
|
10
|
Mollaei P, Barati Farimani A. Activity Map and Transition Pathways of G Protein-Coupled Receptor Revealed by Machine Learning. J Chem Inf Model 2023; 63:2296-2304. [PMID: 37036101 PMCID: PMC10131220 DOI: 10.1021/acs.jcim.3c00032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 04/11/2023]
Abstract
Approximately, one-third of all U.S. Food and Drug Administration approved drugs target G protein-coupled receptors (GPCRs). However, more knowledge of protein structure-activity correlation is required to improve the efficacy of the drugs targeting GPCRs. In this study, we developed a machine learning model to predict the activation state and activity level of the receptors with high prediction accuracy. Furthermore, we applied this model to thousands of molecular dynamics trajectories to correlate residue-level conformational changes of a GPCR to its activity level. Finally, the most probable transition pathway between activation states of a receptor can be identified using the state-activity information. In addition, with this model, we can associate the contribution of each amino acid to the activation process. Using this method, we can design drugs that mainly target principal amino acids driving the transition between activation states of GPCRs. Our advanced method is generalizable to all GPCR classes and provides mechanistic insight into the activation mechanism in the receptors.
Collapse
Affiliation(s)
- Parisa Mollaei
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
| | - Amir Barati Farimani
- Department
of Mechanical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh, Pennsylvania 15213, United States
- Machine
Learning Department, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
11
|
Van Gelder P, Audenaert E, Calders P, Leybaert L. A new look at osteoarthritis: Threshold potentials and an analogy to hypocalcemia. FRONTIERS IN AGING 2023; 4:977426. [PMID: 36970729 PMCID: PMC10031104 DOI: 10.3389/fragi.2023.977426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023]
Abstract
Cartilage is a tissue that consist of very few cells embedded in a highly negatively charged extracellular matrix (ECM). This tissue is dealing with several electrical potentials which have been shown to control the production of ECM. Cartilage is present at joints and is constantly prone to degradation. Failing to repair the damage will result in the occurrence of osteoarthritis (OA). This perspective aims to link biophysical insights with biomolecular research in order to provide an alternative view on the possible causes of OA. Firstly, we hypothesize the existence of a threshold potential, which should be reached in order to initiate repair but if not met, unrepaired damage will evolve to OA. Measurements of the magnitude of this threshold electrical potential would be a helpful diagnostic tool. Secondly, since electrical potential alterations can induce chondrocytes to synthesize ECM, a cellular sensor must be present. We here propose an analogy to the hypocalcemia ‘unshielding’ situation to comprehend electrical potential generation and explore possible sensing mechanisms translating the electrical message into cellular responses. A better understanding of the cellular voltage sensors and down-stream signalling mechanisms may lead to the development of novel treatments for cartilage regeneration.
Collapse
Affiliation(s)
- P. Van Gelder
- Department of Rehabilitation Sciences, Ghent University, Ghent, Belgium
| | - E. Audenaert
- Department of Orthopaedic Surgery and Traumatology, Ghent University, Ghent, Belgium
| | - P. Calders
- Department of Rehabilitation Sciences, Ghent University, Ghent, Belgium
| | - L. Leybaert
- Department of Basic and Applied Medical Sciences (BAMS), Physiology Group, Ghent University, Ghent, Belgium
- *Correspondence: L. Leybaert,
| |
Collapse
|
12
|
Davies A, Tomas A. Appreciating the potential for GPCR crosstalk with ion channels. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 195:101-120. [PMID: 36707150 DOI: 10.1016/bs.pmbts.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
G protein-coupled receptors (GPCRs) are expressed by most tissues in the body and are exploited pharmacologically in a variety of pathological conditions including diabetes, cardiovascular disease, neurological diseases, and cancers. Numerous cell signaling pathways can be regulated by GPCR activation, depending on the specific GPCR, ligand and cell type. Ion channels are among the many effector proteins downstream of these signaling pathways. Saliently, ion channels are also recognized as druggable targets, and there is evidence that their activity may regulate GPCR function via membrane potential and cytoplasmic ion concentration. Overall, there appears to be a large potential for crosstalk between ion channels and GPCRs. This might have implications not only for targeting GPCRs for drug development, but also opens the possibility of co-targeting them with ion channels to achieve improved therapeutic outcomes. In this review, we highlight the large variety of possible GPCR-ion channel crosstalk modes.
Collapse
Affiliation(s)
- Amy Davies
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
13
|
Li Q, Peng J, Luo Y, Zhou J, Li T, Cao L, Peng S, Zuo Z, Wang Z. Far infrared light irradiation enhances Aβ clearance via increased exocytotic microglial ATP and ameliorates cognitive deficit in Alzheimer's disease-like mice. J Neuroinflammation 2022; 19:145. [PMID: 35701825 PMCID: PMC9195249 DOI: 10.1186/s12974-022-02521-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 06/01/2022] [Indexed: 12/12/2022] Open
Abstract
Background Exposure to sunlight may decrease the risk of developing Alzheimer’s disease (AD), and visible and near infrared light have been proposed as a possible therapeutic strategy for AD. Here, we investigated the effects of the visible, near infrared and far infrared (FIR) light on the cognitive ability of AD mice, and found that FIR light also showed potential in the improvement of cognitive dysfunction in AD. However, the related mechanism remains to be elucidated. Methods Morris water maze was used to evaluate the cognitive ability of APPswe/PSEN1dE9 double-transgenic AD mice after light treatment. Western blot was carried out to detect the expression of protein involved in synaptic function and amyloid-β (Aβ) production. The protein amount of interleukin (IL)-1β, IL-6, Aβ1-40 and Aβ1-42 were determined using enzyme-linked immunosorbent assay. The mRNA level of receptors was performed using real-time quantitative polymerase chain reaction. Immunostaining was performed to characterize the Aβ burden and microglial Aβ phagocytosis in the brain of AD mice. The Aβ phagocytosis of primary cultured microglia and BV2 were assessed by flow cytometry. The energy metabolism changes were evaluated using related assay kits, including adenosine triphosphate (ATP), lactate content, mitochondrial respiratory chain complex enzymatic activity and oxidized/reduced nicotinamide adenine dinucleotide assay kits. Results Our results showed that FIR light reduced Aβ burden, a hallmark of AD neuropathology, alleviated neuroinflammation, restored the expression of the presynaptic protein synaptophysin, and ameliorated learning and memory impairment in the AD mice. FIR light enhanced mitochondrial oxidative phosphorylation pathway to increase ATP production. This increased intracellular ATP promoted the extracellular ATP release from microglia stimulated by Aβ, leading to the enhanced Aβ phagocytosis through phosphoinositide 3-kinase/mammalian target of rapamycin pathways for Aβ clearance. Conclusions Our findings have uncovered a previously unappreciated function of FIR light in inducing microglial phagocytosis to clean Aβ, which may be the mechanisms for FIR light to improve cognitive dysfunction in AD mice. These results suggest that FIR light treatment is a potential therapeutic strategy for AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02521-y.
Collapse
Affiliation(s)
- Qingyong Li
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China.,Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jun Peng
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China
| | - Yuelian Luo
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China
| | - Jiaxin Zhou
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China
| | - Tailin Li
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China
| | - Lin Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China
| | - Shuling Peng
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, 22901, USA. .,Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA.
| | - Zhi Wang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 YanJiang West Road, Guangzhou, 510289, Guangdong Province, China.
| |
Collapse
|
14
|
Prediction of GPCR activity using Machine Learning. Comput Struct Biotechnol J 2022; 20:2564-2573. [PMID: 35685352 PMCID: PMC9163700 DOI: 10.1016/j.csbj.2022.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 11/20/2022] Open
Abstract
GPCRs are the target for one-third of the FDA-approved drugs, however; the development of new drug molecules targeting GPCRs is limited by the lack of mechanistic understanding of the GPCR structure–activity-function relationship. To modulate the GPCR activity with highly specific drugs and minimal side-effects, it is necessary to quantitatively describe the important structural features in the GPCR and correlate them to the activation state of GPCR. In this study, we developed 3 ML approaches to predict the conformation state of GPCR proteins. Additionally, we predict the activity level of GPCRs based on their structure. We leverage the unique advantages of each of the 3 ML approaches, interpretability of XGBoost, minimal feature engineering for 3D convolutional neural network, and graph representation of protein structure for graph neural network. By using these ML approaches, we are able to predict the activation state of GPCRs with high accuracy (91%–95%) and also predict the activation state of GPCRs with low error (MAE of 7.15–10.58). Furthermore, the interpretation of the ML approaches allows us to determine the importance of each of the features in distinguishing between the GPCRs conformations.
Collapse
|
15
|
Rozenfeld E, Tauber M, Ben-Chaim Y, Parnas M. GPCR voltage dependence controls neuronal plasticity and behavior. Nat Commun 2021; 12:7252. [PMID: 34903750 PMCID: PMC8668892 DOI: 10.1038/s41467-021-27593-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/29/2021] [Indexed: 12/25/2022] Open
Abstract
G-protein coupled receptors (GPCRs) play a paramount role in diverse brain functions. Almost 20 years ago, GPCR activity was shown to be regulated by membrane potential in vitro, but whether the voltage dependence of GPCRs contributes to neuronal coding and behavioral output under physiological conditions in vivo has never been demonstrated. Here we show that muscarinic GPCR mediated neuronal potentiation in vivo is voltage dependent. This voltage dependent potentiation is abolished in mutant animals expressing a voltage independent receptor. Depolarization alone, without a muscarinic agonist, results in a nicotinic ionotropic receptor potentiation that is mediated by muscarinic receptor voltage dependency. Finally, muscarinic receptor voltage independence causes a strong behavioral effect of increased odor habituation. Together, this study identifies a physiological role for the voltage dependency of GPCRs by demonstrating crucial involvement of GPCR voltage dependence in neuronal plasticity and behavior. Thus, this study suggests that GPCR voltage dependency plays a role in many diverse neuronal functions including learning and memory.
Collapse
Affiliation(s)
- Eyal Rozenfeld
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Merav Tauber
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, 43107, Israel
| | - Yair Ben-Chaim
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, 43107, Israel
| | - Moshe Parnas
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
16
|
Rituper B, Guček A, Lisjak M, Gorska U, Šakanović A, Bobnar ST, Lasič E, Božić M, Abbineni PS, Jorgačevski J, Kreft M, Verkhratsky A, Platt FM, Anderluh G, Stenovec M, Božič B, Coorssen JR, Zorec R. Vesicle cholesterol controls exocytotic fusion pore. Cell Calcium 2021; 101:102503. [PMID: 34844123 DOI: 10.1016/j.ceca.2021.102503] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022]
Abstract
In some lysosomal storage diseases (LSD) cholesterol accumulates in vesicles. Whether increased vesicle cholesterol affects vesicle fusion with the plasmalemma, where the fusion pore, a channel between the vesicle lumen and the extracellular space, is formed, is unknown. Super-resolution microscopy revealed that after stimulation of exocytosis, pituitary lactotroph vesicles discharge cholesterol which transfers to the plasmalemma. Cholesterol depletion in lactotrophs and astrocytes, both exhibiting Ca2+-dependent exocytosis regulated by distinct Ca2+sources, evokes vesicle secretion. Although this treatment enhanced cytosolic levels of Ca2+ in lactotrophs but decreased it in astrocytes, this indicates that cholesterol may well directly define the fusion pore. In an attempt to explain this mechanism, a new model of cholesterol-dependent fusion pore regulation is proposed. High-resolution membrane capacitance measurements, used to monitor fusion pore conductance, a parameter related to fusion pore diameter, confirm that at resting conditions reducing cholesterol increases, while enrichment with cholesterol decreases the conductance of the fusion pore. In resting fibroblasts, lacking the Npc1 protein, a cellular model of LSD in which cholesterol accumulates in vesicles, the fusion pore conductance is smaller than in controls, showing that vesicle cholesterol controls fusion pore and is relevant for pathophysiology of LSD.
Collapse
Affiliation(s)
- Boštjan Rituper
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Alenka Guček
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Marjeta Lisjak
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Urszula Gorska
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Aleksandra Šakanović
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Saša Trkov Bobnar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Eva Lasič
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Mićo Božić
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Prabhodh S Abbineni
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, United States of America
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Alexei Verkhratsky
- Celica Biomedical, 1000, Ljubljana, Slovenia; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, United Kingdom; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, United Kingdom
| | - Gregor Anderluh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Bojan Božič
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Jens R Coorssen
- Department of Health Sciences, Faculty of Applied Health Sciences and Department of Biological Sciences, Faculty of Mathematics & Science, Brock University, St Catherine's, Ontario, Canada
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia; Celica Biomedical, 1000, Ljubljana, Slovenia.
| |
Collapse
|