1
|
Jin S, Bellier J, Wells A, LIopis PM, Anekal PV, Tresback JS, Caldarone BJ, Liu L, Li S, Dettmer U, Ramalingam N, Selkoe DJ. Inhibition of hippocampal mossy fiber plasticity and episodic memory by human Aβ oligomers is prevented by enhancing cAMP signaling in Alzheimer's mice. Alzheimers Dement 2025; 21:e70194. [PMID: 40302031 PMCID: PMC12040739 DOI: 10.1002/alz.70194] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION Early episodic memory impairment in Alzheimer's disease (AD) is linked to synaptic dysfunction from amyloid β-protein oligomers (oAβ), particularly affecting the dentate gyrus mossy fiber-CA3 pathway. The APPNL-G-F mouse model exhibits early deficits in mossy fiber long-term potentiation (mf-LTP). METHODS We administered the β-adrenergic receptor agonist isoproterenol (ISO) in vivo and phosphodiesterase type 4 inhibitor GSK356278 in vitro to assess their impact on mf-LTP and contextual fear memory. Fluorescence lifetime imaging (FLIM)-Förster resonance energy transfer (FRET) microscopy was used to visualize impaired and rescued cyclic adenosine monophosphate (cAMP) signaling in dentate gyrus neurons. RESULTS ISO prevented mf-LTP impairment at 3-4 mo and improved memory by 7 mo. GSK356278 inhibited mf-LTP deficits in a dose-dependent manner. ISO also reduced hyperphosphorylation of synapsin I and microgliosis. DISCUSSION These findings suggest that β-AR activation and phosphodiesterase 4 (PDE4) inhibition mitigate oAβ-induced memory deficits, supporting enhanced cAMP signaling as a therapeutic target for early AD. HIGHLIGHTS Early episodic memory deficits in AD linked to oAβ-induced synaptic dysfunction. Isoproterenol and GSK356278 improve mossy fiber-LTP and fear memory deficits. FLIM-FRET shows treatments restore cAMP signaling in dentate gyrus neurons. Isoproterenol reduces synapsin I hyperphosphorylation and microgliosis. Enhancing cAMP signaling may help mitigate early memory deficits in AD.
Collapse
Affiliation(s)
- Shan‐Xue Jin
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Jean‐Pierre Bellier
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | | | | | | | - Jason S. Tresback
- Center for Nanoscale SystemsHarvard UniversityCambridgeMassachusettsUSA
| | | | - Lei Liu
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Shaomin Li
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| | - Dennis J. Selkoe
- Ann Romney Center for Neurologic DiseasesDepartment of NeurologyHarvard Medical School and Brigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
2
|
Sloan DC, Liao Y, Ray F, Muntean BS. The G protein modifier KCTD5 tunes the decoding of neuromodulatory signals necessary for motor function in striatal neurons. PLoS Biol 2025; 23:e3003117. [PMID: 40233107 PMCID: PMC12021292 DOI: 10.1371/journal.pbio.3003117] [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: 07/29/2024] [Revised: 04/24/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
G proteins (Gα and Gβγ subtypes) drive adenylyl cyclase type 5 (AC5) synthesis of cAMP in striatal neurons, which is essential for motor coordination. KCTD5 directly interacts with Gβγ to delimit signaling events, yet downstream impact of KCTD5 in striatal circuits is not known. Here, generation of a conditional Kctd5 knockout mouse identified that loss of striatal KCTD5 leads to a dystonic phenotype, coordination deficits, and skewed transitions between behavioral syllables. 2-photon imaging of a cAMP biosensor revealed electrically evoked dopaminergic responses were significantly augmented in the absence of KCTD5 in striatal circuits. cAMP sensitization was rescued in situ by expression of a Gβγ-scavenging nanobody and motor deficits were partially rescued in vivo by pharmacological antagonism of the indirect striatal cAMP pathway. Therefore, KCTD5 acts as a brake on cAMP signaling in striatal neurons important for tuning dopaminergic signaling and motor coordination.
Collapse
Affiliation(s)
- Douglas C. Sloan
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Yini Liao
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Forest Ray
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Brian S. Muntean
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| |
Collapse
|
3
|
Bompierre S, Byelyayeva Y, Mota E, Lefevre M, Pumo A, Kehler J, Castro LRV, Vincent P. Cross-pathway integration of cAMP signals through cGMP and calcium-regulated phosphodiesterases in mouse striatal cholinergic interneurons. Br J Pharmacol 2025; 182:1236-1253. [PMID: 39604216 DOI: 10.1111/bph.17400] [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: 02/05/2024] [Revised: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND AND PURPOSE Acetylcholine plays a key role in striatal function. Firing properties of striatal cholinergic interneurons depend on intracellular cAMP through the regulation of Ih currents. Yet, the dynamics of cyclic nucleotide signalling in these neurons have remained elusive. EXPERIMENTAL APPROACH We used highly selective FRET biosensors and pharmacological compounds to analyse the functional contribution of phosphodiesterases in striatal cholinergic interneurons in mouse brain slices. KEY RESULTS PDE1A, PDE3A and PDE4 appear as the main controllers of cAMP levels in striatal cholinergic interneurons. The calcium signal elicited through NMDA or metabotropic glutamate receptors activates PDE1A, which degrades both cAMP and cGMP. Interestingly, the nitric oxide/cGMP pathway amplifies cAMP signalling via PDE3A inhibition-a mechanism hitherto unexplored in a neuronal context. CONCLUSIONS AND IMPLICATIONS The expression pattern of specific PDE enzymes in striatal cholinergic interneurons, by integrating diverse intracellular pathways, can adjust cAMP responses bidirectionally. These properties eventually allow striatal cholinergic interneurons to dynamically regulate their overall activity and modulate acetylcholine release. Remarkably, this effect is the opposite of the cGMP-induced inhibition of cAMP signals involving PDE2A in striatal medium-sized spiny neurons, which provides important insights for the understanding of signal integration in the striatum.
Collapse
Affiliation(s)
- Ségolène Bompierre
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | | | - Elia Mota
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Marion Lefevre
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Anna Pumo
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | | | - Liliana R V Castro
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
| | - Pierre Vincent
- CNRS, Biological Adaptation and Ageing, Sorbonne Université, Paris, France
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
4
|
Davies I, Adriaenssens AE, Scott WR, Carling D, Murphy KG, Minnion JS, Bloom SR, Jones B, Tan TMM. Chronic GIPR agonism results in pancreatic islet GIPR functional desensitisation. Mol Metab 2025; 92:102094. [PMID: 39788289 PMCID: PMC11786100 DOI: 10.1016/j.molmet.2025.102094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVES There is renewed interest in targeting the glucose-dependent insulinotropic polypeptide receptor (GIPR) for treatment of obesity and type 2 diabetes. G-protein coupled receptor desensitisation is suggested to reduce the long-term efficacy of glucagon-like-peptide 1 receptor (GLP-1R) agonists and may similarly affect the efficacy of GIPR agonists. We explored the extent of pancreatic GIPR functional desensitisation with sustained agonist exposure. METHODS A long-acting GIPR agonist, GIP108, was used to probe the effect of sustained agonist exposure on cAMP responses in dispersed pancreatic islets using live cell imaging, with rechallenge cAMP responses after prior agonist treatment used to quantify functional desensitisation. Receptor internalisation and β-arrestin-2 activation were investigated in vitro using imaging-based assays. Pancreatic mouse GIPR desensitisation was assessed in vivo via intraperitoneal glucose tolerance testing. RESULTS GIP108 treatment led to weight loss and improved glucose homeostasis in mice. Prolonged exposure to GIPR agonists produced homologous functional GIPR desensitisation in isolated islets. GIP108 pre-treatment in vivo also reduced the subsequent anti-hyperglycaemic response to GIP re-challenge. GIPR showed minimal agonist-induced internalisation or β-arrestin-2 activation. CONCLUSIONS Although GIP108 chronic treatment improved glucose tolerance, it also resulted in partial desensitisation of the pancreatic islet GIPR. This suggests that ligands with reduced desensitisation tendency might lead to improved in vivo efficacy. Understanding whether pancreatic GIPR desensitisation affects the long-term benefits of GIPR agonists in humans is vital to design effective metabolic pharmacotherapies.
Collapse
Affiliation(s)
- Iona Davies
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - Alice E Adriaenssens
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, United Kingdom
| | - William R Scott
- MRC Laboratory of Medical Sciences, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - David Carling
- MRC Laboratory of Medical Sciences, London, United Kingdom
| | - Kevin G Murphy
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - James S Minnion
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - Stephen R Bloom
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom.
| | - Tricia M-M Tan
- Section of Endocrinology and Investigative Medicine, Imperial College London, United Kingdom.
| |
Collapse
|
5
|
Černe U, Horvat A, Sanjković E, Kozoderc N, Kreft M, Zorec R, Scholz N, Vardjan N. Ca 2+ excitability of glia to neuromodulator octopamine in Drosophila living brain is greater than that of neurons. Acta Physiol (Oxf) 2025; 241:e14270. [PMID: 39801347 PMCID: PMC11726276 DOI: 10.1111/apha.14270] [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: 07/01/2024] [Revised: 11/13/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
AIM Octopamine in the Drosophila brain has a neuromodulatory role similar to that of noradrenaline in mammals. After release from Tdc2 neurons, octopamine/tyramine may trigger intracellular Ca2+ signaling via adrenoceptor-like receptors on neural cells, modulating neurotransmission. Octopamine/tyramine receptors are expressed in neurons and glia, but how each of these cell types responds to octopamine remains elusive. This study aimed to characterize Ca2+ responses of neurons and astrocytes to neuromodulatory octopamine signals. METHODS We expressed Ca2+ indicator jGCaMP7b in specific cell type in adult Drosophila brains and performed intracellular Ca2+ imaging in the brain optic lobes upon bath application of octopamine by confocal microscopy. RESULTS Octopamine-stimulated Ca2+ responses in neurons were different from those of glial cells. The amplitude of octopamine-mediated Ca2+ signals in neurons was 3.4-fold greater than in astrocytes. However, astrocytes were more sensitive to octopamine; the median effective concentration that triggered Ca2+ responses was nearly 6-fold lower in astrocytes than in neurons. In both cell types, Ca2+ transients are shaped by Gq and Gs protein-coupled octopamine/tyramine receptors. Our snRNA-seq database screening uncovered differential expression patterns of these receptors between brain cell types, which may explain the difference in Ca2+ signaling. CONCLUSION In the brain optic lobes, astrocytes, not neurons, appear to be the sole responders to low concentration octopamine signals, and therefore likely drive synaptic plasticity and visual processing. Given the interconnectivity of the optic lobes with other brain regions, octopaminergic signals acting through the optic lobe astrocytes may also influence higher-order brain functions including learning and memory.
Collapse
Grants
- P40 OD018537 NIH HHS
- Deutsche Forschungsgemeinschaft (FOR 2149, 265903901/P01; CRC 1423, 421152132/B06)
- Slovenian Research and Innovation Agency (P3-0310, J3-2523, J3-50104, MR+ 2019, I0-0034, I0-0022: MRIC-Carl Zeiss Reference Centre for Laser Confocal Microscopy)
- European Cooperation in Science and Technology (COST) action CA18133 (European Research Network on Signal Transduction (ERNEST))
- European Cooperation in Science and Technology (COST) action CA18133 (European Research Network on Signal Transduction (ERNEST))
- Slovenian Research and Innovation Agency (P3‐0310, J3‐2523, J3‐50104, MR+ 2019, I0‐0034, I0‐0022: MRIC‐Carl Zeiss Reference Centre for Laser Confocal Microscopy)
- Deutsche Forschungsgemeinschaft (FOR 2149, 265903901/P01; CRC 1423, 421152132/B06)
Collapse
Affiliation(s)
- Urška Černe
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| | - Anemari Horvat
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| | - Ena Sanjković
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
| | - Nika Kozoderc
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
- Department of Biology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| | - Nicole Scholz
- Division of General Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of BiochemistryLeipzig UniversityLeipzigGermany
| | - Nina Vardjan
- Laboratory of Neuroendocrinology‐Molecular Cell Physiology, Faculty of Medicine, Institute of PathophysiologyUniversity of LjubljanaLjubljanaSlovenia
- Laboratory of Cell Engineering, Celica BiomedicalLjubljanaSlovenia
| |
Collapse
|
6
|
Liao Y, Muntean BS. KCTD1 regulation of Adenylyl cyclase type 5 adjusts striatal cAMP signaling. Proc Natl Acad Sci U S A 2024; 121:e2406686121. [PMID: 39413138 PMCID: PMC11513970 DOI: 10.1073/pnas.2406686121] [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: 04/02/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024] Open
Abstract
Dopamine transfers information to striatal neurons, and disrupted neurotransmission leads to motor deficits observed in movement disorders. Striatal dopamine converges downstream to Adenylyl Cyclase Type 5 (AC5)-mediated synthesis of cAMP, indicating the essential role of signal transduction in motor physiology. However, the relationship between dopamine decoding and AC5 regulation is unknown. Here, we utilized an unbiased global protein stability screen to identify Potassium Channel Tetramerization Domain 1 (KCTD1) as a key regulator of AC5 level that is mechanistically tied to N-linked glycosylation. We then implemented a CRISPR/SaCas9 approach to eliminate KCTD1 in striatal neurons expressing a Förster resonance energy transfer (FRET)-based cAMP biosensor. 2-photon imaging of striatal neurons in intact circuits uncovered that dopaminergic signaling was substantially compromised in the absence of KCTD1. Finally, knockdown of KCTD1 in genetically defined dorsal striatal neurons significantly altered motor behavior in mice. These results reveal that KCTD1 acts as an essential modifier of dopaminergic signaling by stabilizing striatal AC5.
Collapse
Affiliation(s)
- Yini Liao
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA30912
| | - Brian S. Muntean
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA30912
| |
Collapse
|
7
|
Singhrao N, Flores-Tamez VA, Moustafa YA, Reddy GR, Burns AE, Pinkerton KE, Chen CY, Navedo MF, Nieves-Cintrón M. Nicotine Impairs Smooth Muscle cAMP Signaling and Vascular Reactivity. Microcirculation 2024; 31:e12871. [PMID: 38805589 PMCID: PMC11303104 DOI: 10.1111/micc.12871] [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: 11/14/2023] [Revised: 03/29/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
OBJECTIVE This study aimed to determine nicotine's impact on receptor-mediated cyclic adenosine monophosphate (cAMP) synthesis in vascular smooth muscle (VSM). We hypothesize that nicotine impairs β adrenergic-mediated cAMP signaling in VSM, leading to altered vascular reactivity. METHODS The effects of nicotine on cAMP signaling and vascular function were systematically tested in aortic VSM cells and acutely isolated aortas from mice expressing the cAMP sensor TEpacVV (Camper), specifically in VSM (e.g., CamperSM). RESULTS Isoproterenol (ISO)-induced β-adrenergic production of cAMP in VSM was significantly reduced in cells from second-hand smoke (SHS)-exposed mice and cultured wild-type VSM treated with nicotine. The decrease in cAMP synthesis caused by nicotine was verified in freshly isolated arteries from a mouse that had cAMP sensor expression in VSM (e.g., CamperSM mouse). Functionally, the changes in cAMP signaling in response to nicotine hindered ISO-induced vasodilation, but this was reversed by immediate PDE3 inhibition. CONCLUSIONS These results imply that nicotine alters VSM β adrenergic-mediated cAMP signaling and vasodilation, which may contribute to the dysregulation of vascular reactivity and the development of vascular complications for nicotine-containing product users.
Collapse
Affiliation(s)
- Navid Singhrao
- Department of Pharmacology, University of California, Davis, USA
| | | | | | | | - Abby E. Burns
- Department of Pharmacology, University of California, Davis, USA
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, California, USA
| | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, USA
| | | |
Collapse
|
8
|
Zhang Y, Gao J, Li N, Xu P, Qu S, Cheng J, Wang M, Li X, Song Y, Xiao F, Yang X, Liu J, Hong H, Mu R, Li X, Wang Y, Xu H, Xie Y, Gao T, Wang G, Aa J. Targeting cAMP in D1-MSNs in the nucleus accumbens, a new rapid antidepressant strategy. Acta Pharm Sin B 2024; 14:667-681. [PMID: 38322327 PMCID: PMC10840425 DOI: 10.1016/j.apsb.2023.12.005] [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: 06/13/2023] [Revised: 09/11/2023] [Accepted: 11/14/2023] [Indexed: 02/08/2024] Open
Abstract
Studies have suggested that the nucleus accumbens (NAc) is implicated in the pathophysiology of major depression; however, the regulatory strategy that targets the NAc to achieve an exclusive and outstanding anti-depression benefit has not been elucidated. Here, we identified a specific reduction of cyclic adenosine monophosphate (cAMP) in the subset of dopamine D1 receptor medium spiny neurons (D1-MSNs) in the NAc that promoted stress susceptibility, while the stimulation of cAMP production in NAc D1-MSNs efficiently rescued depression-like behaviors. Ketamine treatment enhanced cAMP both in D1-MSNs and dopamine D2 receptor medium spiny neurons (D2-MSNs) of depressed mice, however, the rapid antidepressant effect of ketamine solely depended on elevating cAMP in NAc D1-MSNs. We discovered that a higher dose of crocin markedly increased cAMP in the NAc and consistently relieved depression 24 h after oral administration, but not a lower dose. The fast onset property of crocin was verified through multicenter studies. Moreover, crocin specifically targeted at D1-MSN cAMP signaling in the NAc to relieve depression and had no effect on D2-MSN. These findings characterize a new strategy to achieve an exclusive and outstanding anti-depression benefit by elevating cAMP in D1-MSNs in the NAc, and provide a potential rapid antidepressant drug candidate, crocin.
Collapse
Affiliation(s)
- Yue Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jingwen Gao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Na Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Xu
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China
| | - Shimeng Qu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jinqian Cheng
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Mingrui Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xueru Li
- School of Foreign Languages, China Pharmaceutical University, Nanjing 211198, China
| | - Yaheng Song
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Xiao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyu Yang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jihong Liu
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Ronghao Mu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Youmei Wang
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China
| | - Hui Xu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Xie
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Tianming Gao
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
9
|
Hinds CE, Peace E, Chen S, Davies I, El Eid L, Tomas A, Tan T, Minnion J, Jones B, Bloom SR. Abolishing β-arrestin recruitment is necessary for the full metabolic benefits of G protein-biased glucagon-like peptide-1 receptor agonists. Diabetes Obes Metab 2024; 26:65-77. [PMID: 37795639 DOI: 10.1111/dom.15288] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023]
Abstract
AIM Earlier studies have shown that peptide glucagon-like peptide-1 receptor (GLP-1R) agonists with reduced β-arrestin recruitment show enhanced anti-hyperglycaemic efficacy through avoidance of GLP-1R desensitization. However, the ligand modifications needed to decrease β-arrestin recruitment usually also reduces GLP-1R affinity, therefore higher doses are needed. Here we aimed to develop new, long-acting, G protein-biased GLP-1R agonists with acute signalling potency comparable with semaglutide, to provide insights into specific experimental and therapeutic scenarios. MATERIALS AND METHODS New GLP-1R agonist peptides were assessed using a variety of in vitro and in vivo assays. RESULTS First, we show that very substantial reductions in β-arrestin recruitment efficacy are required to realize fully the benefits of GLP-1R agonism on blood glucose lowering in mice, with more moderate reductions being less effective. Secondly, our lead compound (SRB107) performs substantially better than semaglutide for effects on blood glucose and weight loss, which may be jointly attributable to its biased agonist action and protracted pharmacokinetics. Thirdly, we show that biased agonist-specific GLP-1R internalization profiles occur at clinically relevant pharmacological concentrations. Finally, we show that SRB107 cAMP signalling is differentially modulated by single and double GLP1R coding variants seen in human populations, with implications for GLP-1R agonist pharmacogenomics. CONCLUSIONS Completely abolishing β-arrestin recruitment improves the anti-hyperglycaemic effects of GLP-1R agonists in mice.
Collapse
Affiliation(s)
- Charlotte E Hinds
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Ellie Peace
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Shiqian Chen
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Iona Davies
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Liliane El Eid
- Section of Cell Biology, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology, Imperial College London, London, UK
| | - Tricia Tan
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - James Minnion
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Stephen R Bloom
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| |
Collapse
|
10
|
Zhou F, Tichy AM, Imambocus BN, Sakharwade S, Rodriguez Jimenez FJ, González Martínez M, Jahan I, Habib M, Wilhelmy N, Burre V, Lömker T, Sauter K, Helfrich-Förster C, Pielage J, Grunwald Kadow IC, Janovjak H, Soba P. Optimized design and in vivo application of optogenetically functionalized Drosophila dopamine receptors. Nat Commun 2023; 14:8434. [PMID: 38114457 PMCID: PMC10730509 DOI: 10.1038/s41467-023-43970-0] [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: 05/18/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Neuromodulatory signaling via G protein-coupled receptors (GPCRs) plays a pivotal role in regulating neural network function and animal behavior. The recent development of optogenetic tools to induce G protein-mediated signaling provides the promise of acute and cell type-specific manipulation of neuromodulatory signals. However, designing and deploying optogenetically functionalized GPCRs (optoXRs) with accurate specificity and activity to mimic endogenous signaling in vivo remains challenging. Here we optimize the design of optoXRs by considering evolutionary conserved GPCR-G protein interactions and demonstrate the feasibility of this approach using two Drosophila Dopamine receptors (optoDopRs). These optoDopRs exhibit high signaling specificity and light sensitivity in vitro. In vivo, we show receptor and cell type-specific effects of dopaminergic signaling in various behaviors, including the ability of optoDopRs to rescue the loss of the endogenous receptors. This work demonstrates that optoXRs can enable optical control of neuromodulatory receptor-specific signaling in functional and behavioral studies.
Collapse
Affiliation(s)
- Fangmin Zhou
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 3800, Clayton, Victoria, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 3800, Clayton, Victoria, Australia
| | - Bibi Nusreen Imambocus
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Shreyas Sakharwade
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Francisco J Rodriguez Jimenez
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
- ZIEL-Institute of Life and Health, Technical University of Munich, School of Life Sciences, 85354, Freising, Germany
| | - Marco González Martínez
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
| | - Ishrat Jahan
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
| | - Margarita Habib
- Neurobiology and Genetics, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Nina Wilhelmy
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Vanessa Burre
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Tatjana Lömker
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Kathrin Sauter
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | | | - Jan Pielage
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Ilona C Grunwald Kadow
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
- ZIEL-Institute of Life and Health, Technical University of Munich, School of Life Sciences, 85354, Freising, Germany
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 3800, Clayton, Victoria, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 3800, Clayton, Victoria, Australia
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, 5042, Bedford Park, South Australia, Australia
| | - Peter Soba
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany.
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
11
|
Masuho I, Kise R, Gainza P, Von Moo E, Li X, Tany R, Wakasugi-Masuho H, Correia BE, Martemyanov KA. Rules and mechanisms governing G protein coupling selectivity of GPCRs. Cell Rep 2023; 42:113173. [PMID: 37742189 PMCID: PMC10842385 DOI: 10.1016/j.celrep.2023.113173] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023] Open
Abstract
G protein-coupled receptors (GPCRs) convert extracellular stimuli into intracellular signaling by coupling to heterotrimeric G proteins of four classes: Gi/o, Gq, Gs, and G12/13. However, our understanding of the G protein selectivity of GPCRs is incomplete. Here, we quantitatively measure the enzymatic activity of GPCRs in living cells and reveal the G protein selectivity of 124 GPCRs with the exact rank order of their G protein preference. Using this information, we establish a classification of GPCRs by functional selectivity, discover the existence of a G12/13-coupled receptor, G15-coupled receptors, and a variety of subclasses for Gi/o-, Gq-, and Gs-coupled receptors, culminating in development of the predictive algorithm of G protein selectivity. We further identify the structural determinants of G protein selectivity, allowing us to synthesize non-existent GPCRs with de novo G protein selectivity and efficiently identify putative pathogenic variants.
Collapse
Affiliation(s)
- Ikuo Masuho
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA; Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA; Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA.
| | - Ryoji Kise
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Pablo Gainza
- Laboratory of Protein Design and Immunoengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ee Von Moo
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Xiaona Li
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Ryosuke Tany
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Hideko Wakasugi-Masuho
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA; Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Bruno E Correia
- Laboratory of Protein Design and Immunoengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Kirill A Martemyanov
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA.
| |
Collapse
|
12
|
Knuth ER, Foster HR, Jin E, Merrins MJ. Leucine suppresses glucagon secretion from pancreatic islets by directly modulating α-cell cAMP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.551113. [PMID: 37577685 PMCID: PMC10418066 DOI: 10.1101/2023.07.31.551113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Objective Pancreatic islets are nutrient sensors that regulate organismal blood glucose homeostasis. Glucagon release from the pancreatic α-cell is important under fasted, fed, and hypoglycemic conditions, yet metabolic regulation of α-cells remains poorly understood. Here, we identified a previously unexplored role for physiological levels of leucine, which is classically regarded as a β-cell fuel, in the intrinsic regulation of α-cell glucagon release. Methods GcgCreERT:CAMPER and GcgCreERT:GCaMP6s mice were generated to perform dynamic, high-throughput functional measurements of α-cell cAMP and Ca2+ within the intact islet. Islet perifusion assays were used for simultaneous, time-resolved measurements of glucagon and insulin release from mouse and human islets. The effects of leucine were compared with glucose and the mitochondrial fuels 2-aminobicyclo(2,2,1)heptane-2-carboxylic acid (BCH, non-metabolized leucine analog that activates glutamate dehydrogenase), α-ketoisocaproate (KIC, leucine metabolite), and methyl-succinate (complex II fuel). CYN154806 (Sstr2 antagonist), diazoxide (KATP activator, which prevents Ca2+-dependent exocytosis from α, β, and δ-cells), and dispersed α-cells were used to inhibit islet paracrine signaling and identify α-cell intrinsic effects. Results Mimicking the effect of glucose, leucine strongly suppressed amino acid-stimulated glucagon secretion. Mechanistically, leucine dose-dependently reduced α-cell cAMP at physiological concentrations, with an IC50 of 57, 440, and 1162 μM at 2, 6, and 10 mM glucose, without affecting α-cell Ca2+. Leucine also reduced α-cell cAMP in islets treated with Sstr2 antagonist or diazoxide, as well as dispersed α-cells, indicating an α-cell intrinsic effect. The effect of leucine was matched by KIC and the glutamate dehydrogenase activator BCH, but not methyl-succinate, indicating a dependence on mitochondrial anaplerosis. Glucose, which stimulates anaplerosis via pyruvate carboxylase, had the same suppressive effect on α-cell cAMP but with lower potency. Similarly to mouse islets, leucine suppressed glucagon secretion from human islets under hypoglycemic conditions. Conclusions These findings highlight an important role for physiological levels of leucine in the metabolic regulation of α-cell cAMP and glucagon secretion. Leucine functions primarily through an α-cell intrinsic effect that is dependent on glutamate dehydrogenase, in addition to the well-established α-cell regulation by β/δ-cell paracrine signaling. Our results suggest that mitochondrial anaplerosis-cataplerosis facilitates the glucagonostatic effect of both leucine and glucose, which cooperatively suppress α-cell tone by reducing cAMP.
Collapse
Affiliation(s)
- Emily R. Knuth
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hannah R. Foster
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Erli Jin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Matthew J. Merrins
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| |
Collapse
|
13
|
Manchanda Y, Bitsi S, Chen S, Broichhagen J, Bernardino de la Serna J, Jones B, Tomas A. Enhanced Endosomal Signaling and Desensitization of GLP-1R vs GIPR in Pancreatic Beta Cells. Endocrinology 2023; 164:7034684. [PMID: 36774542 PMCID: PMC10016038 DOI: 10.1210/endocr/bqad028] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
The incretin receptors, glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic polypeptide receptor (GIPR), are prime therapeutic targets for the treatment of type 2 diabetes (T2D) and obesity. They are expressed in pancreatic beta cells where they potentiate insulin release in response to food intake. Despite GIP being the main incretin in healthy individuals, GLP-1R has been favored as a therapeutic target due to blunted GIPR responses in T2D patients and conflicting effects of GIPR agonists and antagonists in improving glucose tolerance and preventing weight gain. There is, however, a recently renewed interest in GIPR biology, following the realization that GIPR responses can be restored after an initial period of blood glucose normalization and the recent development of dual GLP-1R/GIPR agonists with superior capacity for controlling blood glucose levels and weight. The importance of GLP-1R trafficking and subcellular signaling in the control of receptor outputs is well established, but little is known about the pattern of spatiotemporal signaling from the GIPR in beta cells. Here, we have directly compared surface expression, trafficking, and signaling characteristics of both incretin receptors in pancreatic beta cells to identify potential differences that might underlie distinct pharmacological responses associated with each receptor. Our results indicate increased cell surface levels, internalization, degradation, and endosomal vs plasma membrane activity for the GLP-1R, while the GIPR is instead associated with increased plasma membrane recycling, reduced desensitization, and enhanced downstream signal amplification. These differences might have potential implications for the capacity of each incretin receptor to control beta cell function.
Collapse
Affiliation(s)
- Yusman Manchanda
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Stavroula Bitsi
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Shiqian Chen
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Johannes Broichhagen
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin 13125, Germany
| | | | - Ben Jones
- Correspondence: Alejandra Tomas, PhD, Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK, ; or Ben Jones, MD, PhD, Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK,
| | - Alejandra Tomas
- Correspondence: Alejandra Tomas, PhD, Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK, ; or Ben Jones, MD, PhD, Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK,
| |
Collapse
|
14
|
Day-Cooney J, Dalangin R, Zhong H, Mao T. Genetically encoded fluorescent sensors for imaging neuronal dynamics in vivo. J Neurochem 2023; 164:284-308. [PMID: 35285522 PMCID: PMC11322610 DOI: 10.1111/jnc.15608] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Abstract
The brain relies on many forms of dynamic activities in individual neurons, from synaptic transmission to electrical activity and intracellular signaling events. Monitoring these neuronal activities with high spatiotemporal resolution in the context of animal behavior is a necessary step to achieve a mechanistic understanding of brain function. With the rapid development and dissemination of highly optimized genetically encoded fluorescent sensors, a growing number of brain activities can now be visualized in vivo. To date, cellular calcium imaging, which has been largely used as a proxy for electrical activity, has become a mainstay in systems neuroscience. While challenges remain, voltage imaging of neural populations is now possible. In addition, it is becoming increasingly practical to image over half a dozen neurotransmitters, as well as certain intracellular signaling and metabolic activities. These new capabilities enable neuroscientists to test previously unattainable hypotheses and questions. This review summarizes recent progress in the development and delivery of genetically encoded fluorescent sensors, and highlights example applications in the context of in vivo imaging.
Collapse
Affiliation(s)
- Julian Day-Cooney
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rochelin Dalangin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
15
|
Multiple potassium channel tetramerization domain (KCTD) family members interact with Gβγ, with effects on cAMP signaling. J Biol Chem 2023; 299:102924. [PMID: 36736897 PMCID: PMC9976452 DOI: 10.1016/j.jbc.2023.102924] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) initiate an array of intracellular signaling programs by activating heterotrimeric G proteins (Gα and Gβγ subunits). Therefore, G protein modifiers are well positioned to shape GPCR pharmacology. A few members of the potassium channel tetramerization domain (KCTD) protein family have been found to adjust G protein signaling through interaction with Gβγ. However, comprehensive details on the KCTD interaction with Gβγ remain unresolved. Here, we report that nearly all the 25 KCTD proteins interact with Gβγ. In this study, we screened Gβγ interaction capacity across the entire KCTD family using two parallel approaches. In a live cell bioluminescence resonance energy transfer-based assay, we find that roughly half of KCTD proteins interact with Gβγ in an agonist-induced fashion, whereas all KCTD proteins except two were found to interact through coimmunoprecipitation. We observed that the interaction was dependent on an amino acid hot spot in the C terminus of KCTD2, KCTD5, and KCTD17. While KCTD2 and KCTD5 require both the Bric-à-brac, Tramtrack, Broad complex domain and C-terminal regions for Gβγ interaction, we uncovered that the KCTD17 C terminus is sufficient for Gβγ interaction. Finally, we demonstrated the functional consequence of the KCTD-Gβγ interaction by examining sensitization of the adenylyl cyclase-cAMP pathway in live cells. We found that Gβγ-mediated sensitization of adenylyl cyclase 5 was blunted by KCTD. We conclude that the KCTD family broadly engages Gβγ to shape GPCR signal transmission.
Collapse
|
16
|
Lewis JE, Woodward OR, Nuzzaci D, Smith CA, Adriaenssens AE, Billing L, Brighton C, Phillips BU, Tadross JA, Kinston SJ, Ciabatti E, Göttgens B, Tripodi M, Hornigold D, Baker D, Gribble FM, Reimann F. Relaxin/insulin-like family peptide receptor 4 (Rxfp4) expressing hypothalamic neurons modulate food intake and preference in mice. Mol Metab 2022; 66:101604. [PMID: 36184065 PMCID: PMC9579047 DOI: 10.1016/j.molmet.2022.101604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Insulin-like peptide 5 (INSL5) signalling, through its cognate receptor relaxin/insulin-like family peptide receptor 4 (RXFP4), has been reported to be orexigenic, and the high fat diet (HFD) preference observed in wildtype mice is altered in Rxfp4 knock-out mice. In this study, we used a new Rxfp4-Cre mouse model to investigate the mechanisms underlying these observations. METHODS We generated transgenic Rxfp4-Cre mice and investigated central expression of Rxfp4 by RT-qPCR, RNAscope and intraparenchymal infusion of INSL5. Rxfp4-expressing cells were chemogenetically manipulated in global Cre-reporter mice using designer receptors exclusively activated by designer drugs (DREADDs) or after stereotactic injection of a Cre-dependent AAV-DIO-Dq-DREADD targeting a population located in the ventromedial hypothalamus (RXFP4VMH). Food intake and feeding motivation were assessed in the presence and absence of a DREADD agonist. Rxfp4-expressing cells in the hypothalamus were characterised by single-cell RNA-sequencing (scRNAseq) and the connectivity of RXFP4VMH cells was investigated using viral tracing. RESULTS Rxfp4-Cre mice displayed Cre-reporter expression in the hypothalamus. Active expression of Rxfp4 in the adult mouse brain was confirmed by RT-qPCR and RNAscope. Functional receptor expression was supported by cyclic AMP-responses to INSL5 application in ex vivo brain slices and increased HFD and highly palatable liquid meal (HPM), but not chow, intake after intra-VMH INSL5 infusion. scRNAseq of hypothalamic RXFP4 neurons defined a cluster expressing VMH markers, alongside known appetite-modulating neuropeptide receptors (Mc4r, Cckar and Nmur2). Viral tracing demonstrated RXFP4VMH neural projections to nuclei implicated in hedonic feeding behaviour. Whole body chemogenetic inhibition (Di-DREADD) of Rxfp4-expressing cells, mimicking physiological INSL5-RXFP4 Gi-signalling, increased intake of the HFD and HPM, but not chow, whilst activation (Dq-DREADD), either at whole body level or specifically within the VMH, reduced HFD and HPM intake and motivation to work for the HPM. CONCLUSION These findings identify RXFP4VMH neurons as regulators of food intake and preference, and hypothalamic RXFP4 signalling as a target for feeding behaviour manipulation.
Collapse
Affiliation(s)
- Jo E Lewis
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Orla Rm Woodward
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Danaé Nuzzaci
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Christopher A Smith
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Alice E Adriaenssens
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Lawrence Billing
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Cheryl Brighton
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Benjamin U Phillips
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - John A Tadross
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK; Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Sarah J Kinston
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Ernesto Ciabatti
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Marco Tripodi
- MRC Laboratory of Molecular Biology, Neurobiology Division, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - David Hornigold
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca Ltd, Cambridge, UK
| | - David Baker
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca Ltd, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
17
|
Sensitive genetically encoded sensors for population and subcellular imaging of cAMP in vivo. Nat Methods 2022; 19:1461-1471. [PMID: 36303019 PMCID: PMC10171401 DOI: 10.1038/s41592-022-01646-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) signaling integrates information from diverse G-protein-coupled receptors, such as neuromodulator receptors, to regulate pivotal biological processes in a cellular-specific and subcellular-specific manner. However, in vivo cellular-resolution imaging of cAMP dynamics remains challenging. Here, we screen existing genetically encoded cAMP sensors and further develop the best performer to derive three improved variants, called cAMPFIREs. Compared with their parental sensor, these sensors exhibit up to 10-fold increased sensitivity to cAMP and a cytosolic distribution. cAMPFIREs are compatible with both ratiometric and fluorescence lifetime imaging and can detect cAMP dynamics elicited by norepinephrine at physiologically relevant, nanomolar concentrations. Imaging of cAMPFIREs in awake mice reveals tonic levels of cAMP in cortical neurons that are associated with wakefulness, modulated by opioids, and differentially regulated across subcellular compartments. Furthermore, enforced locomotion elicits neuron-specific, bidirectional cAMP dynamics. cAMPFIREs also function in Drosophila. Overall, cAMPFIREs may have broad applicability for studying intracellular signaling in vivo.
Collapse
|
18
|
Pittolo S, Yokoyama S, Willoughby DD, Taylor CR, Reitman ME, Tse V, Wu Z, Etchenique R, Li Y, Poskanzer KE. Dopamine activates astrocytes in prefrontal cortex via α1-adrenergic receptors. Cell Rep 2022; 40:111426. [PMID: 36170823 PMCID: PMC9555850 DOI: 10.1016/j.celrep.2022.111426] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 07/19/2022] [Accepted: 09/08/2022] [Indexed: 12/31/2022] Open
Abstract
The prefrontal cortex (PFC) is a hub for cognitive control, and dopamine profoundly influences its functions. In other brain regions, astrocytes sense diverse neurotransmitters and neuromodulators and, in turn, orchestrate regulation of neuroactive substances. However, basic physiology of PFC astrocytes, including which neuromodulatory signals they respond to and how they contribute to PFC function, is unclear. Here, we characterize divergent signaling signatures in mouse astrocytes of the PFC and primary sensory cortex, which show differential responsiveness to locomotion. We find that PFC astrocytes express receptors for dopamine but are unresponsive through the Gs/Gi-cAMP pathway. Instead, fast calcium signals in PFC astrocytes are time locked to dopamine release and are mediated by α1-adrenergic receptors both ex vivo and in vivo. Further, we describe dopamine-triggered regulation of extracellular ATP at PFC astrocyte territories. Thus, we identify astrocytes as active players in dopaminergic signaling in the PFC, contributing to PFC function though neuromodulator receptor crosstalk. Pittolo et al. demonstrate that the neuromodulator dopamine targets astrocytes, a type of brain cell, via receptors specific to another neuromodulator—norepinephrine. This study provides groundwork on how dopamine affects non-neuronal brain cells and suggests that crosstalk between neuromodulatory pathways occurs in vivo, with possible clinical implications.
Collapse
Affiliation(s)
- Silvia Pittolo
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Sae Yokoyama
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Drew D Willoughby
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Charlotte R Taylor
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Vincent Tse
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Roberto Etchenique
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CONICET, Intendente Güiraldes 2160, Ciudad Universitaria, Pabellón 2, C1428EGA, Buenos Aires, Argentina
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
19
|
Kawata S, Mukai Y, Nishimura Y, Takahashi T, Saitoh N. Green fluorescent cAMP indicator of high speed and specificity suitable for neuronal live-cell imaging. Proc Natl Acad Sci U S A 2022; 119:e2122618119. [PMID: 35867738 PMCID: PMC9282276 DOI: 10.1073/pnas.2122618119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/29/2022] [Indexed: 11/18/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a canonical intracellular messenger playing diverse roles in cell functions. In neurons, cAMP promotes axonal growth during early development, and mediates sensory transduction and synaptic plasticity after maturation. The molecular cascades of cAMP are well documented, but its spatiotemporal profiles associated with neuronal functions remain hidden. Hence, we developed a genetically encoded cAMP indicator based on a bacterial cAMP-binding protein. This indicator "gCarvi" monitors [cAMP]i at 0.2 to 20 µM with a subsecond time resolution and a high specificity over cyclic guanosine monophosphate (cGMP). gCarvi can be converted to a ratiometric probe for [cAMP]i quantification and its expression can be specifically targeted to various subcellular compartments. Monomeric gCarvi also enables simultaneous multisignal monitoring in combination with other indicators. As a proof of concept, simultaneous cAMP/Ca2+ imaging in hippocampal neurons revealed a tight linkage of cAMP to Ca2+ signals. In cerebellar presynaptic boutons, forskolin induced nonuniform cAMP elevations among boutons, which positively correlated with subsequent increases in the size of the recycling pool of synaptic vesicles assayed using FM dye. Thus, the cAMP domain in presynaptic boutons is an important determinant of the synaptic strength.
Collapse
Affiliation(s)
- Seiko Kawata
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yuki Mukai
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yumi Nishimura
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| | - Naoto Saitoh
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| |
Collapse
|
20
|
Kasatkina LA, Verkhusha VV. Transgenic mice encoding modern imaging probes: Properties and applications. Cell Rep 2022; 39:110845. [PMID: 35613592 PMCID: PMC9183799 DOI: 10.1016/j.celrep.2022.110845] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 12/04/2022] Open
Abstract
Modern biology is increasingly reliant on optical technologies, including visualization and longitudinal monitoring of cellular processes. The major limitation here is the availability of animal models to track the molecules and cells in their natural environment in vivo. Owing to the integrity of the studied tissue and the high stability of transgene expression throughout life, transgenic mice encoding fluorescent proteins and biosensors represent unique tools for in vivo studies in norm and pathology. We review the strategies for targeting probe expression in specific tissues, cell subtypes, or cellular compartments. We describe the application of transgenic mice expressing fluorescent proteins for tracking protein expression patterns, apoptotic events, tissue differentiation and regeneration, neurogenesis, tumorigenesis, and cell fate mapping. We overview the possibilities of functional imaging of secondary messengers, neurotransmitters, and ion fluxes. Finally, we provide the rationale and perspectives for the use of transgenic imaging probes in translational research and drug discovery.
Collapse
Affiliation(s)
- Ludmila A Kasatkina
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Vladislav V Verkhusha
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland.
| |
Collapse
|
21
|
Reddy GR, Ren L, Thai PN, Caldwell JL, Zaccolo M, Bossuyt J, Ripplinger CM, Xiang YK, Nieves-Cintrón M, Chiamvimonvat N, Navedo MF. Deciphering cellular signals in adult mouse sinoatrial node cells. iScience 2022; 25:103693. [PMID: 35036877 PMCID: PMC8749457 DOI: 10.1016/j.isci.2021.103693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/30/2021] [Accepted: 12/22/2021] [Indexed: 01/27/2023] Open
Abstract
Sinoatrial node (SAN) cells are the pacemakers of the heart. This study describes a method for culturing and infection of adult mouse SAN cells with FRET-based biosensors that can be exploited to examine signaling events. SAN cells cultured in media with blebbistatin or (S)-nitro-blebbistatin retain their morphology, protein distribution, action potential (AP) waveform, and cAMP dynamics for at least 40 h. SAN cells expressing targeted cAMP sensors show distinct β-adrenergic-mediated cAMP pools. Cyclic GMP, protein kinase A, Ca2+/CaM kinase II, and protein kinase D in SAN cells also show unique dynamics to different stimuli. Heart failure SAN cells show a decrease in cAMP and cGMP levels. In summary, a reliable method for maintaining adult mouse SAN cells in culture is presented, which facilitates studies of signaling networks and regulatory mechanisms during physiological and pathological conditions.
Collapse
Affiliation(s)
- Gopireddy R. Reddy
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Lu Ren
- Department of Internal Medicine, University of California Davis, 451 Health Science Drive, GBSF 6315, Davis, CA 95616, USA
| | - Phung N. Thai
- Department of Internal Medicine, University of California Davis, 451 Health Science Drive, GBSF 6315, Davis, CA 95616, USA
| | - Jessica L. Caldwell
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Crystal M. Ripplinger
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
- VA Northern California Healthcare System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Madeline Nieves-Cintrón
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, University of California Davis, 451 Health Science Drive, GBSF 6315, Davis, CA 95616, USA
- VA Northern California Healthcare System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, One Shields Avenue MED: PHARM Tupper 242, Davis, CA 95616, USA
| |
Collapse
|
22
|
Members of the KCTD family are major regulators of cAMP signaling. Proc Natl Acad Sci U S A 2022; 119:2119237119. [PMID: 34934014 PMCID: PMC8740737 DOI: 10.1073/pnas.2119237119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Neuromodulation is pivotal for brain function. One of the key pathways engaged by neuromodulators is signaling via second messenger cAMP, which controls a myriad of fundamental reactions. This study identifies KCTD5, a ubiquitin ligase adapter, as a regulatory element in this pathway and determines that it works by an unusual dual mode controlling the activity of cAMP-generating enzyme in neurons through both zinc transport and G protein signaling. Cyclic adenosine monophosphate (cAMP) is a pivotal second messenger with an essential role in neuronal function. cAMP synthesis by adenylyl cyclases (AC) is controlled by G protein–coupled receptor (GPCR) signaling systems. However, the network of molecular players involved in the process is incompletely defined. Here, we used CRISPR/Cas9–based screening to identify that members of the potassium channel tetradimerization domain (KCTD) family are major regulators of cAMP signaling. Focusing on striatal neurons, we show that the dominant isoform KCTD5 exerts its effects through an unusual mechanism that modulates the influx of Zn2+ via the Zip14 transporter to exert unique allosteric effects on AC. We further show that KCTD5 controls the amplitude and sensitivity of stimulatory GPCR inputs to cAMP production by Gβγ-mediated AC regulation. Finally, we report that KCTD5 haploinsufficiency in mice leads to motor deficits that can be reversed by chelating Zn2+. Together, our findings uncover KCTD proteins as major regulators of neuronal cAMP signaling via diverse mechanisms.
Collapse
|
23
|
Qiao H, Wu J, Zhang X, Luo J, Wang H, Ming D. The Advance of CRISPR-Cas9-Based and NIR/CRISPR-Cas9-Based Imaging System. Front Chem 2021; 9:786354. [PMID: 34976954 PMCID: PMC8716450 DOI: 10.3389/fchem.2021.786354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
The study of different genes, chromosomes and the spatiotemporal relationship between them is of great significance in the field of biomedicine. CRISPR-Cas9 has become the most widely used gene editing tool due to its excellent targeting ability. In recent years, a series of advanced imaging technologies based on Cas9 have been reported, providing fast and convenient tools for studying the sites location of genome, RNA, and chromatin. At the same time, a variety of CRISPR-Cas9-based imaging systems have been developed, which are widely used in real-time multi-site imaging in vivo. In this review, we summarized the component and mechanism of CRISPR-Cas9 system, overviewed the NIR imaging and the application of NIR fluorophores in the delivery of CRISPR-Cas9, and highlighted advances of the CRISPR-Cas9-based imaging system. In addition, we also discussed the challenges and potential solutions of CRISPR-Cas9-based imaging methods, and looked forward to the development trend of the field.
Collapse
Affiliation(s)
- Huanhuan Qiao
- Functional Materials Laboratory, Institute of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Jieting Wu
- Functional Materials Laboratory, Institute of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xiaodong Zhang
- Functional Materials Laboratory, Institute of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| | - Hao Wang
- Functional Materials Laboratory, Institute of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- *Correspondence: Hao Wang, ; Dong Ming,
| | - Dong Ming
- Functional Materials Laboratory, Institute of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- *Correspondence: Hao Wang, ; Dong Ming,
| |
Collapse
|
24
|
Jones-Tabah J, Martin RD, Tanny JC, Clarke PBS, Hébert TE. High-Content Single-Cell Förster Resonance Energy Transfer Imaging of Cultured Striatal Neurons Reveals Novel Cross-Talk in the Regulation of Nuclear Signaling by Protein Kinase A and Extracellular Signal-Regulated Kinase 1/2. Mol Pharmacol 2021; 100:526-539. [PMID: 34503973 DOI: 10.1124/molpharm.121.000290] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Genetically encoded biosensors can be used to track signaling events in living cells by measuring changes in fluorescence emitted by one or more fluorescent proteins. Here, we describe the use of genetically encoded biosensors based on Förster resonance energy transfer (FRET), combined with high-content microscopy, to image dynamic signaling events simultaneously in thousands of neurons in response to drug treatments. We first applied this approach to examine intercellular variation in signaling responses among cultured striatal neurons stimulated with multiple drugs. Using high-content FRET imaging and immunofluorescence, we identified neuronal subpopulations with unique responses to pharmacological manipulation and used nuclear morphology to identify medium spiny neurons within these heterogeneous striatal cultures. Focusing on protein kinase A (PKA) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in the cytoplasm and nucleus, we noted pronounced intercellular differences among putative medium spiny neurons, in both the magnitude and kinetics of signaling responses to drug application. Importantly, a conventional "bulk" analysis that pooled all cells in culture yielded a different rank order of drug potency than that revealed by single-cell analysis. Using a single-cell analytical approach, we dissected the relative contributions of PKA and ERK1/2 signaling in striatal neurons and unexpectedly identified a novel role for ERK1/2 in promoting nuclear activation of PKA in striatal neurons. This finding adds a new dimension of signaling crosstalk between PKA and ERK1/2 with relevance to dopamine D1 receptor signaling in striatal neurons. In conclusion, high-content single-cell imaging can complement and extend traditional population-level analyses and provides a novel vantage point from which to study cellular signaling. SIGNIFICANCE STATEMENT: High-content imaging revealed substantial intercellular variation in the magnitude and pattern of intracellular signaling events driven by receptor stimulation. Since individual neurons within the same population can respond differently to a given agonist, interpreting measures of intracellular signaling derived from the averaged response of entire neuronal populations may not always reflect what happened at the single-cell level. This study uses this approach to identify a new form of cross-talk between PKA and ERK1/2 signaling in the nucleus of striatal neurons.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| |
Collapse
|
25
|
Glycine Release Is Potentiated by cAMP via EPAC2 and Ca 2+ Stores in a Retinal Interneuron. J Neurosci 2021; 41:9503-9520. [PMID: 34620721 PMCID: PMC8612479 DOI: 10.1523/jneurosci.0670-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
Neuromodulation via the intracellular second messenger cAMP is ubiquitous at presynaptic nerve terminals. This modulation of synaptic transmission allows exocytosis to adapt to stimulus levels and reliably encode information. The AII amacrine cell (AII-AC) is a central hub for signal processing in the mammalian retina. The main apical dendrite of the AII-AC is connected to several lobular appendages that release glycine onto OFF cone bipolar cells and ganglion cells. However, the influence of cAMP on glycine release is not well understood. Using membrane capacitance measurements from mouse AII-ACs to directly measure exocytosis, we observe that intracellular dialysis of 1 mm cAMP enhances exocytosis without affecting the L-type Ca2+ current. Responses to depolarizing pulses of various durations show that the size of the readily releasable pool of vesicles nearly doubles with cAMP, while paired-pulse depression experiments suggest that release probability does not change. Specific agonists and antagonists for exchange protein activated by cAMP 2 (EPAC2) revealed that the cAMP-induced enhancement of exocytosis requires EPAC2 activation. Furthermore, intact Ca2+ stores were also necessary for the cAMP potentiation of exocytosis. Postsynaptic recordings from OFF cone bipolar cells showed that increasing cAMP with forskolin potentiated the frequency of glycinergic spontaneous IPSCs. We propose that cAMP elevations in the AII-AC lead to a robust enhancement of glycine release through an EPAC2 and Ca2+ store signaling pathway. Our results thus contribute to a better understanding of how AII-AC crossover inhibitory circuits adapt to changes in ambient luminance.SIGNIFICANCE STATEMENT The mammalian retina operates over a wide dynamic range of light intensities and contrast levels. To optimize the signal-to-noise ratio of processed visual information, both excitatory and inhibitory synapses within the retina must modulate their gain in synaptic transmission to adapt to different levels of ambient light. Here we show that increases of cAMP concentration within AII amacrine cells produce enhanced exocytosis from these glycinergic interneurons. Therefore, we propose that light-sensitive neuromodulators may change the output of glycine release from AII amacrine cells. This novel mechanism may fine-tune the amount of tonic and phasic synaptic inhibition received by bipolar cell terminals and, consequently, the spiking patterns that ganglion cells send to the upstream visual areas of the brain.
Collapse
|
26
|
Vollmer KM, Doncheck EM, Grant RI, Winston KT, Romanova EV, Bowen CW, Siegler PN, Green LM, Bobadilla AC, Trujillo-Pisanty I, Kalivas PW, Otis JM. A Novel Assay Allowing Drug Self-Administration, Extinction, and Reinstatement Testing in Head-Restrained Mice. Front Behav Neurosci 2021; 15:744715. [PMID: 34776891 PMCID: PMC8585999 DOI: 10.3389/fnbeh.2021.744715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Multiphoton microscopy is one of several new technologies providing unprecedented insight into the activity dynamics and function of neural circuits. Unfortunately, some of these technologies require experimentation in head-restrained animals, limiting the behavioral repertoire that can be integrated and studied. This issue is especially evident in drug addiction research, as no laboratories have coupled multiphoton microscopy with simultaneous intravenous drug self-administration, a behavioral paradigm that has predictive validity for treatment outcomes and abuse liability. Here, we describe a new experimental assay wherein head-restrained mice will press an active lever, but not inactive lever, for intravenous delivery of heroin or cocaine. Similar to freely moving animals, we find that lever pressing is suppressed through daily extinction training and subsequently reinstated through the presentation of relapse-provoking triggers (drug-associative cues, the drug itself, and stressors). Finally, we show that head-restrained mice will show similar patterns of behavior for oral delivery of a sucrose reward, a common control used for drug self-administration experiments. Overall, these data demonstrate the feasibility of combining drug self-administration experiments with technologies that require head-restraint, such as multiphoton imaging. The assay described could be replicated by interested labs with readily available materials to aid in identifying the neural underpinnings of substance use disorder.
Collapse
Affiliation(s)
- Kelsey M. Vollmer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Elizabeth M. Doncheck
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Roger I. Grant
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Kion T. Winston
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Elizaveta V. Romanova
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Christopher W. Bowen
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Preston N. Siegler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Lisa M. Green
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | | | | | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - James M. Otis
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
27
|
Massengill CI, Day-Cooney J, Mao T, Zhong H. Genetically encoded sensors towards imaging cAMP and PKA activity in vivo. J Neurosci Methods 2021; 362:109298. [PMID: 34339753 PMCID: PMC8659126 DOI: 10.1016/j.jneumeth.2021.109298] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 12/26/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) is a universal second messenger that plays a crucial role in diverse biological functions, ranging from transcription to neuronal plasticity, and from development to learning and memory. In the nervous system, cAMP integrates inputs from many neuromodulators across a wide range of timescales - from seconds to hours - to modulate neuronal excitability and plasticity in brain circuits during different animal behavioral states. cAMP signaling events are both cell-specific and subcellularly compartmentalized. The same stimulus may result in different, sometimes opposite, cAMP dynamics in different cells or subcellular compartments. Additionally, the activity of protein kinase A (PKA), a major cAMP effector, is also spatiotemporally regulated. For these reasons, many laboratories have made great strides toward visualizing the intracellular dynamics of cAMP and PKA. To date, more than 80 genetically encoded sensors, including original and improved variants, have been published. It is starting to become possible to visualize cAMP and PKA signaling events in vivo, which is required to study behaviorally relevant cAMP/PKA signaling mechanisms. Despite significant progress, further developments are needed to enhance the signal-to-noise ratio and practical utility of these sensors. This review summarizes the recent advances and challenges in genetically encoded cAMP and PKA sensors with an emphasis on in vivo imaging in the brain during behavior.
Collapse
Affiliation(s)
| | - Julian Day-Cooney
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
28
|
Cellular context shapes cyclic nucleotide signaling in neurons through multiple levels of integration. J Neurosci Methods 2021; 362:109305. [PMID: 34343574 DOI: 10.1016/j.jneumeth.2021.109305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023]
Abstract
Intracellular signaling with cyclic nucleotides are ubiquitous signaling pathways, yet the dynamics of these signals profoundly differ in different cell types. Biosensor imaging experiments, by providing direct measurements in intact cellular environment, reveal which receptors are activated by neuromodulators and how the coincidence of different neuromodulators is integrated at various levels in the signaling cascade. Phosphodiesterases appear as one important determinant of cross-talk between different signaling pathways. Finally, analysis of signal dynamics reveal that striatal medium-sized spiny neuron obey a different logic than other brain regions such as cortex, probably in relation with the function of this brain region which efficiently detects transient dopamine.
Collapse
|
29
|
Jones-Tabah J, Mohammad H, Clarke PBS, Hébert TE. In vivo detection of GPCR-dependent signaling using fiber photometry and FRET-based biosensors. Methods 2021; 203:422-430. [PMID: 34022351 DOI: 10.1016/j.ymeth.2021.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 12/27/2022] Open
Abstract
Genetically encoded fluorescent biosensors allow intracellular signaling dynamics to be tracked in live cells and tissues using optical detection. Many such biosensors are based on the principle of Förster resonance energy transfer (FRET), and we have recently developed a simple approach for in vivo detection of FRET-based biosensor signals using fiber photometry. By combining fiber photometry with FRET-based biosensors, we were able to track GPCR-dependent signaling pathways over time, and in response to drug treatments in freely-moving adult rats. Recording from specific neuronal populations, we can quantify intracellular signaling while simultaneously measuring behavioral responses. Our approach, described in detail here, uses adeno-associated viruses infused intracerebrally in order to express genetically-encoded FRET-based biosensors. After several weeks to allow biosensor expression, fiber photometry is used in order to record drug responses in real time from freely-moving adult rats. This methodology would be compatible with other mammalian species and with many biosensors. Hence, it has wide applicability across a spectrum of neuroscience research, ranging from the study of neural circuits and behavior, to preclinical drug development and screening.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Hanan Mohammad
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
30
|
Diversity of the Gβγ complexes defines spatial and temporal bias of GPCR signaling. Cell Syst 2021; 12:324-337.e5. [PMID: 33667409 DOI: 10.1016/j.cels.2021.02.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 12/09/2020] [Accepted: 02/04/2021] [Indexed: 01/04/2023]
Abstract
The signal transduction by G-protein-coupled receptors (GPCRs) is mediated by heterotrimeric G proteins composed from one of the 16 Gα subunits and the inseparable Gβγ complex assembled from a repertoire of 5 Gβ and 12 Gγ subunits. However, the functional role of compositional diversity in Gβγ complexes has been elusive. Using optical biosensors, we examined the function of all Gβγ combinations in living cells and uncovered two major roles of Gβγ diversity. First, we demonstrate that the identity of Gβγ subunits greatly influences the kinetics and efficacy of GPCR responses at the plasma membrane. Second, we show that different Gβγ combinations are selectively dispatched from the plasma membrane to various cellular organelles on a timescale from milliseconds to minutes. We describe the mechanisms regulating these processes and document their implications for GPCR signaling via various Gα subunits, thereby illustrating a role for the compositional diversity of G protein heterotrimers.
Collapse
|
31
|
El K, Gray SM, Capozzi ME, Knuth ER, Jin E, Svendsen B, Clifford A, Brown JL, Encisco SE, Chazotte BM, Sloop KW, Nunez DJ, Merrins MJ, D'Alessio DA, Campbell JE. GIP mediates the incretin effect and glucose tolerance by dual actions on α cells and β cells. SCIENCE ADVANCES 2021; 7:7/11/eabf1948. [PMID: 33712466 PMCID: PMC7954443 DOI: 10.1126/sciadv.abf1948] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/26/2021] [Indexed: 05/23/2023]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) communicates nutrient intake from the gut to islets, enabling optimal levels of insulin secretion via the GIP receptor (GIPR) on β cells. The GIPR is also expressed in α cells, and GIP stimulates glucagon secretion; however, the role of this action in the postprandial state is unknown. Here, we demonstrate that GIP potentiates amino acid-stimulated glucagon secretion, documenting a similar nutrient-dependent action to that described in β cells. Moreover, we demonstrate that GIP activity in α cells contributes to insulin secretion by invoking paracrine α to β cell communication. Last, specific loss of GIPR activity in α cells prevents glucagon secretion in response to a meal stimulus, limiting insulin secretion and driving glucose intolerance. Together, these data uncover an important axis by which GIPR activity in α cells is necessary to coordinate the optimal level of both glucagon and insulin secretion to maintain postprandial homeostasis.
Collapse
Affiliation(s)
- K El
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - S M Gray
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - M E Capozzi
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - E R Knuth
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - E Jin
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - B Svendsen
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - A Clifford
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - J L Brown
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - S E Encisco
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - B M Chazotte
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - K W Sloop
- Diabetes and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - D J Nunez
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - M J Merrins
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - D A D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC, USA
| | - J E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| |
Collapse
|
32
|
Gαo is a major determinant of cAMP signaling in the pathophysiology of movement disorders. Cell Rep 2021; 34:108718. [PMID: 33535037 PMCID: PMC7903328 DOI: 10.1016/j.celrep.2021.108718] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/07/2020] [Accepted: 01/11/2021] [Indexed: 01/20/2023] Open
Abstract
The G protein alpha subunit o (Gαo) is one of the most abundant proteins in the nervous system, and pathogenic mutations in its gene (GNAO1) cause movement disorder. However, the function of Gαo is ill defined mechanistically. Here, we show that Gαo dictates neuromodulatory responsiveness of striatal neurons and is required for movement control. Using in vivo optical sensors and enzymatic assays, we determine that Gαo provides a separate transduction channel that modulates coupling of both inhibitory and stimulatory dopamine receptors to the cyclic AMP (cAMP)-generating enzyme adenylyl cyclase. Through a combination of cell-based assays and rodent models, we demonstrate that GNAO1-associated mutations alter Gαo function in a neuron-type-specific fashion via a combination of a dominant-negative and loss-of-function mechanisms. Overall, our findings suggest that Gαo and its pathological variants function in specific circuits to regulate neuromodulatory signals essential for executing motor programs. Muntean et al. describe biochemical, cellular, and physiological mechanisms by which the heterotrimeric G protein subunit Gαo controls neuromodulatory signaling in the striatum and elucidate mechanisms by which Gαo mutations compromise movements in GNAO1 disorder.
Collapse
|
33
|
Kim N, Shin S, Bae SW. cAMP Biosensors Based on Genetically Encoded Fluorescent/Luminescent Proteins. BIOSENSORS-BASEL 2021; 11:bios11020039. [PMID: 33572585 PMCID: PMC7911721 DOI: 10.3390/bios11020039] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) plays a key role in signal transduction pathways as a second messenger. Studies on the cAMP dynamics provided useful scientific insights for drug development and treatment of cAMP-related diseases such as some cancers and prefrontal cortex disorders. For example, modulation of cAMP-mediated intracellular signaling pathways by anti-tumor drugs could reduce tumor growth. However, most early stage tools used for measuring the cAMP level in living organisms require cell disruption, which is not appropriate for live cell imaging or animal imaging. Thus, in the last decades, tools were developed for real-time monitoring of cAMP distribution or signaling dynamics in a non-invasive manner. Genetically-encoded sensors based on fluorescent proteins and luciferases could be powerful tools to overcome these drawbacks. In this review, we discuss the recent genetically-encoded cAMP sensors advances, based on single fluorescent protein (FP), Föster resonance energy transfer (FRET), single luciferase, and bioluminescence resonance energy transfer (BRET) for real-time non-invasive imaging.
Collapse
Affiliation(s)
- Namdoo Kim
- Department of Chemistry, Kongju National University, Gongju 32588, Korea;
| | - Seunghan Shin
- Green Chemistry & Materials Group, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Korea;
| | - Se Won Bae
- Green Chemistry & Materials Group, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Korea;
- Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Korea
- Correspondence: ; Tel.: +82-64-754-3543
| |
Collapse
|
34
|
Ehinger Y, Morisot N, Phamluong K, Sakhai SA, Soneja D, Adrover MF, Alvarez VA, Ron D. cAMP-Fyn signaling in the dorsomedial striatum direct pathway drives excessive alcohol use. Neuropsychopharmacology 2021; 46:334-342. [PMID: 32417851 PMCID: PMC7852539 DOI: 10.1038/s41386-020-0712-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
Fyn kinase in the dorsomedial striatum (DMS) of rodents plays a central role in mechanisms underlying excessive alcohol intake. The DMS is comprised of medium spiny neurons (MSNs) that project directly (dMSNs) or indirectly (iMSNs) to the substantia nigra. Here, we examined the cell-type specificity of Fyn's actions in alcohol use. First, we knocked down Fyn selectively in DMS dMSNs or iMSNs of mice and measured the level of alcohol consumption. We found that downregulation of Fyn in dMSNs, but not in iMSNs, reduces excessive alcohol but not saccharin intake. D1Rs are coupled to Gαs/olf, which activate cAMP signaling. To examine whether Fyn's actions are mediated through cAMP signaling, DMS dMSNs were infected with GαsDREADD, and the activation of Fyn signaling was measured following CNO treatment. We found that remote stimulation of cAMP signaling in DMS dMSNs activates Fyn and promotes the phosphorylation of the Fyn substrate, GluN2B. In contract, remote activation of GαsDREADD in DLS dMSNs did not alter Fyn signaling. We then tested whether activation of GαsDREADD in DMS dMSNs or iMSNs alters alcohol intake and observed that CNO-dependent activation of GαsDREADD in DMS dMSNs but not iMSNs increases alcohol but not saccharin intake. Finally, we examined the contribution of Fyn to GαsDREADD-dependent increase in alcohol intake, and found that systemic administration of the Fyn inhibitor, AZD0503 blocks GαsDREADD-dependent increase in alcohol consumption. Our results suggest that the cAMP-Fyn axis in the DMS dMSNs is a molecular transducer of mechanisms underlying the development of excessive alcohol consumption.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Nadege Morisot
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Nkarta Therapeutics, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Sage Therapeutics, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Martin F Adrover
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- INGEBI, CONICET, Buenos Aires, Argentina
| | - Veronica A Alvarez
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health, Bethesda, MD, 20892, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA.
| |
Collapse
|
35
|
Paton KF, Atigari DV, Kaska S, Prisinzano T, Kivell BM. Strategies for Developing κ Opioid Receptor Agonists for the Treatment of Pain with Fewer Side Effects. J Pharmacol Exp Ther 2020; 375:332-348. [PMID: 32913006 PMCID: PMC7589957 DOI: 10.1124/jpet.120.000134] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/27/2020] [Indexed: 12/21/2022] Open
Abstract
There is significant need to find effective, nonaddictive pain medications. κ Opioid receptor (KOPr) agonists have been studied for decades but have recently received increased attention because of their analgesic effects and lack of abuse potential. However, a range of side effects have limited the clinical development of these drugs. There are several strategies currently used to develop safer and more effective KOPr agonists. These strategies include identifying G-protein-biased agonists, developing peripherally restricted KOPr agonists without centrally mediated side effects, and developing mixed opioid agonists, which target multiple receptors at specific ratios to balance side-effect profiles and reduce tolerance. Here, we review the latest developments in research related to KOPr agonists for the treatment of pain. SIGNIFICANCE STATEMENT: This review discusses strategies for developing safer κ opioid receptor (KOPr) agonists with therapeutic potential for the treatment of pain. Although one strategy is to modify selective KOPr agonists to create peripherally restricted or G-protein-biased structures, another approach is to combine KOPr agonists with μ, δ, or nociceptin opioid receptor activation to obtain mixed opioid receptor agonists, therefore negating the adverse effects and retaining the therapeutic effect.
Collapse
Affiliation(s)
- Kelly F Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Diana V Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Sophia Kaska
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Thomas Prisinzano
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| |
Collapse
|
36
|
Masuho I, Balaji S, Muntean BS, Skamangas NK, Chavali S, Tesmer JJG, Babu MM, Martemyanov KA. A Global Map of G Protein Signaling Regulation by RGS Proteins. Cell 2020; 183:503-521.e19. [PMID: 33007266 PMCID: PMC7572916 DOI: 10.1016/j.cell.2020.08.052] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 07/03/2020] [Accepted: 08/29/2020] [Indexed: 12/24/2022]
Abstract
The control over the extent and timing of G protein signaling is provided by the regulator of G protein signaling (RGS) proteins that deactivate G protein α subunits (Gα). Mammalian genomes encode 20 canonical RGS and 16 Gα genes with key roles in physiology and disease. To understand the principles governing the selectivity of Gα regulation by RGS, we examine the catalytic activity of all canonical human RGS proteins and their selectivity for a complete set of Gα substrates using real-time kinetic measurements in living cells. The data reveal rules governing RGS-Gα recognition, the structural basis of its selectivity, and provide principles for engineering RGS proteins with defined selectivity. The study also explores the evolution of RGS-Gα selectivity through ancestral reconstruction and demonstrates how naturally occurring non-synonymous variants in RGS alter signaling. These results provide a blueprint for decoding signaling selectivity and advance our understanding of molecular recognition principles. Systematic analysis reveals G protein selectivity of all canonical RGS proteins RGS proteins rely on selectivity bar codes for selective G protein recognition Transplantation of bar codes across RGS proteins switches their G protein preferences Natural variants, mutations, and evolution shape RGS selectivity
Collapse
Affiliation(s)
- Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Santhanam Balaji
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Departments of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Nickolas K Skamangas
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Sreenivas Chavali
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Tirupati 517 507, India
| | - John J G Tesmer
- Departments of Biological Sciences and Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907-2054, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Departments of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
37
|
Allostatic Changes in the cAMP System Drive Opioid-Induced Adaptation in Striatal Dopamine Signaling. Cell Rep 2020; 29:946-960.e2. [PMID: 31644915 PMCID: PMC6871051 DOI: 10.1016/j.celrep.2019.09.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/29/2019] [Accepted: 09/12/2019] [Indexed: 01/06/2023] Open
Abstract
Opioids are powerful addictive agents that alter dopaminergic influence
on reward signaling in medium spiny neurons (MSNs) of the nucleus accumbens.
Repeated opioid exposure triggers adaptive changes, shifting reward valuation to
the allostatic state underlying tolerance. However, the cellular substrates and
molecular logic underlying such allostatic changes are not well understood.
Here, we report that the plasticity of dopamine-induced cyclic AMP (cAMP)
signaling in MSNs serves as a cellular substrate for drug-induced allostatic
adjustments. By recording cAMP responses to optically evoked dopamine in brain
slices from mice subjected to various opioid exposure paradigms, we define
profound neuronal-type-specific adaptations. We find that opioid exposure pivots
the initial hyper-responsiveness of D1-MSNs toward D2-MSN dominance as
dependence escalates. Presynaptic dopamine transporters and postsynaptic
phosphodiesterases critically enable cell-specific adjustments of cAMP that
control the balance between opponent D1-MSN and D2-MSN channels. We propose a
quantitative model of opioid-induced allostatic adjustments in cAMP signal
strength that balances circuit activity. Muntean et al. examine how opioid exposure influences cyclic AMP (cAMP)
responses to dopamine in striatal medium spiny neurons (MSNs). They describe
allostatic adaptations in the processing of dopaminergic signals by D1-MSN and
D2-MSN populations as opioid administration progresses from acute exposure to
chronic use, and they define molecular elements contributing to the process.
Collapse
|
38
|
Zhang KX, D'Souza S, Upton BA, Kernodle S, Vemaraju S, Nayak G, Gaitonde KD, Holt AL, Linne CD, Smith AN, Petts NT, Batie M, Mukherjee R, Tiwari D, Buhr ED, Van Gelder RN, Gross C, Sweeney A, Sanchez-Gurmaches J, Seeley RJ, Lang RA. Violet-light suppression of thermogenesis by opsin 5 hypothalamic neurons. Nature 2020; 585:420-425. [PMID: 32879486 PMCID: PMC8130195 DOI: 10.1038/s41586-020-2683-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022]
Abstract
The opsin family of G-protein-coupled receptors are used as light detectors in animals. Opsin 5 (also known as neuropsin or OPN5) is a highly conserved opsin that is sensitive to visible violet light1,2. In mice, OPN5 is a known photoreceptor in the retina3 and skin4 but is also expressed in the hypothalamic preoptic area (POA)5. Here we describe a light-sensing pathway in which POA neurons that express Opn5 regulate thermogenesis in brown adipose tissue (BAT). We show that Opn5 is expressed in glutamatergic warm-sensing POA neurons that receive synaptic input from several thermoregulatory nuclei. We further show that Opn5 POA neurons project to BAT and decrease its activity under chemogenetic stimulation. Opn5-null mice show overactive BAT, increased body temperature, and exaggerated thermogenesis when cold-challenged. Moreover, violet photostimulation during cold exposure acutely suppresses BAT temperature in wild-type mice but not in Opn5-null mice. Direct measurements of intracellular cAMP ex vivo show that Opn5 POA neurons increase cAMP when stimulated with violet light. This analysis thus identifies a violet light-sensitive deep brain photoreceptor that normally suppresses BAT thermogenesis.
Collapse
Affiliation(s)
- Kevin X Zhang
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Shane D'Souza
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Brian A Upton
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Stace Kernodle
- Department of Surgery, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Shruti Vemaraju
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gowri Nayak
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kevin D Gaitonde
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Amanda L Holt
- Department of Physics, Yale University, New Haven, CT, USA
| | - Courtney D Linne
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - April N Smith
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nathan T Petts
- Division of Clinical Engineering, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew Batie
- Division of Clinical Engineering, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ethan D Buhr
- Department of Ophthalmology, University of Washington Medical School, Seattle, WA, USA
| | - Russell N Van Gelder
- Department of Ophthalmology, University of Washington Medical School, Seattle, WA, USA
- Department of Biological Structure, University of Washington Medical School, Seattle, WA, USA
- Department of Pathology, University of Washington Medical School, Seattle, WA, USA
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Alison Sweeney
- Department of Physics, Yale University, New Haven, CT, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, School of Public Health, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Richard A Lang
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Ophthalmology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
39
|
Huey J, Keutler K, Schultz C. Chemical Biology Toolbox for Studying Pancreatic Islet Function - A Perspective. Cell Chem Biol 2020; 27:1015-1031. [PMID: 32822616 DOI: 10.1016/j.chembiol.2020.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/10/2020] [Accepted: 07/28/2020] [Indexed: 01/14/2023]
Abstract
The islets of Langerhans represent one of the many complex endocrine organs in mammals. Traditionally, islet function is studied by a mixture of physiological, cell biological, and molecular biological methods. Recently, novel techniques stemming from the ever-increasing toolbox provided by chemical laboratories have been added to the repertoire. Many emerging techniques will soon be available to manipulate and monitor islet function at the single-cell level and potentially in intact model animals, as well as in isolated human islets. Here, we review the most current small-molecule-based and genetically encoded molecular tool sets available to study islet function. We provide an outlook regarding future tool developments that will impact islet research, with a special focus on the interplay between different islet cell types.
Collapse
Affiliation(s)
- Julia Huey
- Program in Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97210, USA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97210, USA
| | - Kaya Keutler
- Program in Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97210, USA; Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97210, USA
| | - Carsten Schultz
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97210, USA.
| |
Collapse
|
40
|
Kelly MP, Heckman PRA, Havekes R. Genetic manipulation of cyclic nucleotide signaling during hippocampal neuroplasticity and memory formation. Prog Neurobiol 2020; 190:101799. [PMID: 32360536 DOI: 10.1016/j.pneurobio.2020.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Decades of research have underscored the importance of cyclic nucleotide signaling in memory formation and synaptic plasticity. In recent years, several new genetic techniques have expanded the neuroscience toolbox, allowing researchers to measure and modulate cyclic nucleotide gradients with high spatiotemporal resolution. Here, we will provide an overview of studies using genetic approaches to interrogate the role cyclic nucleotide signaling plays in hippocampus-dependent memory processes and synaptic plasticity. Particular attention is given to genetic techniques that measure real-time changes in cyclic nucleotide levels as well as newly-developed genetic strategies to transiently manipulate cyclic nucleotide signaling in a subcellular compartment-specific manner with high temporal resolution.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, VA Bldg1, 3(rd) Fl, D-12, Columbia, 29209, SC, USA.
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
41
|
Zych S, Ford CP. Opioid-Induced Adaptations of cAMP Dynamics in the Nucleus Accumbens. Trends Pharmacol Sci 2020; 41:230-232. [PMID: 31964511 DOI: 10.1016/j.tips.2020.01.004] [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: 01/07/2020] [Accepted: 01/07/2020] [Indexed: 10/25/2022]
Abstract
To investigate how opioid exposure alters dopamine (DA) responses in medium spiny neurons (MSNs), Muntean et al. used a novel cAMP sensor to track cAMP dynamics and report a coordinated effort of adaptations in D1- and D2-MSNs to integrate DA inputs and shift signaling strengths in various states of opioid dependence.
Collapse
Affiliation(s)
- Sarah Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
42
|
Masuho I, Chavali S, Muntean BS, Skamangas NK, Simonyan K, Patil DN, Kramer GM, Ozelius L, Babu MM, Martemyanov KA. Molecular Deconvolution Platform to Establish Disease Mechanisms by Surveying GPCR Signaling. Cell Rep 2019; 24:557-568.e5. [PMID: 30021154 PMCID: PMC6077248 DOI: 10.1016/j.celrep.2018.06.080] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/10/2018] [Accepted: 06/19/2018] [Indexed: 01/01/2023] Open
Abstract
Despite the wealth of genetic information available, mechanisms underlying pathological effects of disease-associated mutations in components of G protein-coupled receptor (GPCR) signaling cascades remain elusive. In this study, we developed a scalable approach for the functional analysis of clinical variants in GPCR pathways along with a complete analytical framework. We applied the strategy to evaluate an extensive set of dystonia-causing mutations in G protein Gαolf. Our quantitative analysis revealed diverse mechanisms by which pathogenic variants disrupt GPCR signaling, leading to a mechanism-based classification of dystonia. In light of significant clinical heterogeneity, the mechanistic analysis of individual disease-associated variants permits tailoring personalized intervention strategies, which makes it superior to the current phenotype-based approach. We propose that the platform developed in this study can be universally applied to evaluate disease mechanisms for conditions associated with genetic variation in all components of GPCR signaling. A scalable platform allows multidimensional analysis of GPCR signaling The approach is applied to dystonia-causing mutations in G protein Gαolf Pathogenic variants in Gαolf disrupt GPCR signaling by diverse mechanisms Mechanism-based disease classification could allow targeted therapies
Collapse
Affiliation(s)
- Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Sreenivas Chavali
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Nickolas K Skamangas
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Kristina Simonyan
- Department of Otolaryngology, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Dipak N Patil
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Grant M Kramer
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA; Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Laurie Ozelius
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
43
|
Identification of Novel Adenylyl Cyclase 5 (AC5) Signaling Networks in D 1 and D 2 Medium Spiny Neurons using Bimolecular Fluorescence Complementation Screening. Cells 2019; 8:cells8111468. [PMID: 31752385 PMCID: PMC6912275 DOI: 10.3390/cells8111468] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 11/17/2022] Open
Abstract
Adenylyl cyclase type 5 (AC5), as the principal isoform expressed in striatal medium spiny neurons (MSNs), is essential for the integration of both stimulatory and inhibitory midbrain signals that initiate from dopaminergic G protein-coupled receptor (GPCR) activation. The spatial and temporal control of cAMP signaling is dependent upon the composition of local regulatory protein networks. However, there is little understanding of how adenylyl cyclase protein interaction networks adapt to the multifarious pressures of integrating acute versus chronic and inhibitory vs. stimulatory receptor signaling in striatal MSNs. Here, we presented the development of a novel bimolecular fluorescence complementation (BiFC)-based protein-protein interaction screening methodology to further identify and characterize elements important for homeostatic control of dopamine-modulated AC5 signaling in a neuronal model cell line and striatal MSNs. We identified two novel AC5 modulators: the protein phosphatase 2A (PP2A) catalytic subunit (PPP2CB) and the intracellular trafficking associated protein-NSF (N-ethylmaleimide-sensitive factor) attachment protein alpha (NAPA). The effects of genetic knockdown (KD) of each gene were evaluated in several cellular models, including D1- and D2-dopamine receptor-expressing MSNs from CAMPER mice. The knockdown of PPP2CB was associated with a reduction in acute and sensitized adenylyl cyclase activity, implicating PP2A is an important and persistent regulator of adenylyl cyclase activity. In contrast, the effects of NAPA knockdown were more nuanced and appeared to involve an activity-dependent protein interaction network. Taken together, these data represent a novel screening method and workflow for the identification and validation of adenylyl cyclase protein-protein interaction networks under diverse cAMP signaling paradigms.
Collapse
|
44
|
Sutton LP, Muntean BS, Ostrovskaya O, Zucca S, Dao M, Orlandi C, Song C, Xie K, Martemyanov KA. NF1-cAMP signaling dissociates cell type-specific contributions of striatal medium spiny neurons to reward valuation and motor control. PLoS Biol 2019; 17:e3000477. [PMID: 31600280 PMCID: PMC6805008 DOI: 10.1371/journal.pbio.3000477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/22/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
The striatum plays a fundamental role in motor learning and reward-related behaviors that are synergistically shaped by populations of D1 dopamine receptor (D1R)- and D2 dopamine receptor (D2R)-expressing medium spiny neurons (MSNs). How various neurotransmitter inputs converging on common intracellular pathways are parsed out to regulate distinct behavioral outcomes in a neuron-specific manner is poorly understood. Here, we reveal that distinct contributions of D1R-MSNs and D2R-MSNs towards reward and motor behaviors are delineated by the multifaceted signaling protein neurofibromin 1 (NF1). Using genetic mouse models, we show that NF1 in D1R-MSN modulates opioid reward, whereas loss of NF1 in D2R-MSNs delays motor learning by impeding the formation and consolidation of repetitive motor sequences. We found that motor learning deficits upon NF1 loss were associated with the disruption in dopamine signaling to cAMP in D2R-MSN. Restoration of cAMP levels pharmacologically or chemogenetically rescued the motor learning deficits seen upon NF1 loss in D2R-MSN. Our findings illustrate that multiplex signaling capabilities of MSNs are deployed at the level of intracellular pathways to achieve cell-specific control over behavioral outcomes. A mouse genetic study reveals that the multifaceted signaling protein neurofibromin (known for its role in the human genetic disease neurofibromatosis type 1) plays a key role in differential routing of motor and reward signals in populations of striatal medium spiny neurons.
Collapse
Affiliation(s)
- Laurie P. Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Brian S. Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Stefano Zucca
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Maria Dao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- * E-mail:
| |
Collapse
|
45
|
French A, Tesmer AL, Tantama M. Dual-Mode FRET and BRET Sensors for Detecting cAMP Dynamics. ACS OMEGA 2019; 4:15504-15511. [PMID: 31572851 PMCID: PMC6761615 DOI: 10.1021/acsomega.9b01770] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/03/2019] [Indexed: 05/08/2023]
Abstract
Genetically encoded fluorescent and luminescent indicators have revolutionized our ability to monitor physiology in real time, but the separate development of new sensors for each of these imaging modalities involves substantial effort and resources. Methods to rapidly engineer multimodal sensors would, therefore, significantly accelerate the diversification of sensors for simultaneous use in different systems and applications. We hypothesized that the enhanced Nano-lanterns could be incorporated into modular ratiometric sensors as an efficient approach to creating dual-mode fluorescent-luminescent sensors. As a proof-of-concept, we engineered an Epac1-based sensor that responds to cyclic adenosine monophosphate binding with a greater than 80% change in both Förster Resonance Energy Transfer and bioluminescent resonance energy transfer (BRET) modes. We also demonstrate that our new sensor reports cellular changes in G-protein-coupled signaling, and that the ratiometric BRET mode is bright enough for subcutaneous measurements in mice.
Collapse
Affiliation(s)
- Alexander
R. French
- Department
of Chemistry and Institute for Integrated Neuroscience, Purdue University, West Lafayette, Indiana 47907, United States
| | - Alexander L. Tesmer
- Department
of Chemistry and Institute for Integrated Neuroscience, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mathew Tantama
- Department
of Chemistry and Institute for Integrated Neuroscience, Purdue University, West Lafayette, Indiana 47907, United States
- Department
of Chemistry, Wellesley College, Wellesley, Massachusetts 02481, United States
- E-mail:
| |
Collapse
|
46
|
Localised GPCR signalling as revealed by FRET biosensors. Curr Opin Cell Biol 2019; 57:48-56. [DOI: 10.1016/j.ceb.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/20/2022]
|