1
|
Ye C, Shi D, Zhu Y, Shi P, Zhao N, Sun Z, Zhang Z, Zhang D, Lv Y, Wu W, Yu J, Karimi-Maleh H, Li H, Fu L, Jiang N, Liu J, Lin CT. Graphene electrochemical biosensors combining effervescent solid-phase extraction (ESPE) for rapid, ultrasensitive, and simultaneous determination of DA, AA, and UA. Biosens Bioelectron 2025; 268:116899. [PMID: 39499969 DOI: 10.1016/j.bios.2024.116899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/02/2024] [Accepted: 10/29/2024] [Indexed: 11/25/2024]
Abstract
Simultaneous monitoring of key metabolites like dopamine, ascorbic acid, and uric acid is essential for early disease diagnosis and evaluating treatment. Electrochemical techniques are increasingly used for precise, point-of-care testing (POCT) of these metabolites. Herein, a sample pretreatment method called effervescent solid-phase extraction (ESPE) was proposed for efficient enrichment of trace analytes for electrochemical detection. In an ESPE process, effervescent tablets made of gold nanoparticle-decorated graphene oxide (Au/GO) were first self-dispersed in a test solution to promote the enrichment of analytes on the Au/GO adsorbents, followed by the addition of flocculant effervescent tablets to cause Au/GO sheets to form self-assembled aggregates, which then can be efficiently collected by the foam electrodes. The entire sample pretreatment process operates without external power and takes only 5 min. With the assistance of the ESPE method, our electrochemical sensors achieve an ultralow detection limit for dopamine, ascorbic acid, and uric acid of 80 pM, 1.8 nM, and 460 pM, respectively, which are two to three orders of magnitude lower than the results obtained by the drop casting technique. The enhancement mechanism of our approach is based on increasing the contact probability with analytes through dynamic dispersion of the Au/GO adsorbents, in contrast to the static diffusion mechanism relied on Brownian motion. We also show that combining the ESPE solution kit with a portable micro-electrochemical workstation can attain the same detection level as HPLC in real urine samples. The proposed ESPE approach holds great promise for POCT applications in 2D material-based biosensors.
Collapse
Affiliation(s)
- Chen Ye
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Diwei Shi
- School of Naval Architecture and Maritime, Zhejiang Ocean University, Zhoushan, Zhejiang 316022, PR China
| | - Yangguang Zhu
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Peizheng Shi
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Ningbin Zhao
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Zhuang Sun
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Zhe Zhang
- Shenzhen Refresh Biosensing Technology Co., Ltd., Shenzhen, 518129, PR China
| | - Diming Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, PR China
| | - Yaokang Lv
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Wenqi Wu
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangdong, 510260, PR China; Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangdong, 511495, PR China
| | - Jiancheng Yu
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Ningbo University, Ningbo, 315211, PR China; Zhenhai Institute of Mass Spectrometry, Ningbo, 315211, PR China; Faculty of Electrical Engineering and Computer Science, Ningbo University, Ningbo, 315211, PR China
| | - Hassan Karimi-Maleh
- School of Resources and Environment, University of Electronic Science and Technology of China, Chengdu, 611731, PR China; School of Engineering, Lebanese American University, Byblos, 1102-2801, Lebanon
| | - He Li
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Li Fu
- College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou 310018, PR China.
| | - Nan Jiang
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China.
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Cheng-Te Lin
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China.
| |
Collapse
|
2
|
Li X, Xiong L, Li Y. The role of the prefrontal cortex in modulating aggression in humans and rodents. Behav Brain Res 2025; 476:115285. [PMID: 39369825 DOI: 10.1016/j.bbr.2024.115285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 09/15/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Accumulating evidence suggests that the prefrontal cortex (PFC) plays an important role in aggression. However, the findings regarding the key neural mechanisms and molecular pathways underlying the modulation of aggression by the PFC are relatively scattered, with many inconsistencies and areas that would benefit from exploration. Here, we highlight the relationship between the PFC and aggression in humans and rodents and describe the anatomy and function of the human PFC, along with homologous regions in rodents. At the molecular level, we detail how the major neuromodulators of the PFC impact aggression. At the circuit level, this review provides an overview of known and potential subcortical projections that regulate aggression in rodents. Finally, at the disease level, we review the correlation between PFC alterations and heightened aggression in specific human psychiatric disorders. Our review provides a framework for PFC modulation of aggression, resolves several intriguing paradoxes from previous studies, and illuminates new avenues for further study.
Collapse
Affiliation(s)
- Xinyang Li
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated with Tongji University School of Medicine, Shanghai, China.
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated with Tongji University School of Medicine, Shanghai, China.
| | - Yan Li
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Liu Y, Cao L, Zhou J, Li C, Du J, Xue Y, Li X, Mao L. Single-Vesicle Electrochemistry in Fresh Brain Slices Enables In Situ Quantification of Vesicular Monoamine. J Am Chem Soc 2024. [PMID: 39705330 DOI: 10.1021/jacs.4c06572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
The quantitative analysis of vesicular neurotransmitters in neurons in situ is paramount for investigating neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease (PD). Unfortunately, a direct approach for monitoring neurotransmitter chemistry in single vesicles in fresh brain tissue has remained inaccessible so far. Here, we introduce an innovative platform of single-vesicle electrochemistry (SVE) in fresh brain tissue, enabling the quantification of neurotransmitters at the single-vesicle level for both soma and varicosity. Utilizing this methodology, we investigated a PD animal model, which demonstrated a significant reduction in both vesicular dopamine (DA) storage and exocytotic release in DA neurons in the substantia nigra. Furthermore, SVE unveiled the heterogeneous nature of chemical neurotransmission among DA neurons across different brain regions. Importantly, this tissue-based SVE approach can be extended to quantify other monoamine neurotransmitters, such as norepinephrine and serotonin, at the single-vesicle level. The introduction of this methodology marks a significant advancement, offering a novel avenue to explore neurological and psychiatric disorders through the lens of neurotransmitter signaling in the mammalian brain. This breakthrough has the potential to deepen our understanding of the underlying mechanisms of these complex disorders and may pave the way for the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Yuying Liu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lijiao Cao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junlan Zhou
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chuqi Li
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jinchang Du
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yifei Xue
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xianchan Li
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
4
|
Bezerra TO, Roque AC. Dopamine facilitates the response to glutamatergic inputs in astrocyte cell models. PLoS Comput Biol 2024; 20:e1012688. [PMID: 39680609 DOI: 10.1371/journal.pcbi.1012688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Astrocytes respond to neurotransmitters by increasing their intracellular Ca2+ concentration (Ca2+ signals). While glutamate released by neurons trigger Ca2+ signals through IP3- and glutamate transporter-dependent mechanisms, dopamine released in distant sites activates astrocytes via dopaminergic receptors. However, little is known about the modulatory effects of dopamine on glutamate-evoked astrocytic activity. To investigate this question, we developed multi-compartment, conductance-based astrocyte models with three distinct morphologies: unipolar; bipolar; and bifurcated-terminal. Glutamate induced localized responses, while dopamine activated all compartments. In the unipolar model, global dopaminergic stimulation reduced the threshold frequency of glutamatergic stimulation required to activate Ca2+ signals. Phase-plane analysis of a simplified version of this model revealed that Ca2+ signals are influenced by compartment radius and neurotransmitter type. Morphology significantly influenced glutamate-dopamine interactions. In the bipolar model, glutamatergic stimulation in one process minimally affected the other. Conversely, in the bifurcated-terminal model, where a single process bifurcates into two secondary processes, high-frequency glutamatergic stimulation in one secondary process evoked Ca2+ signals in the other. Dopamine further facilitated this latter cross-process interaction by lowering the glutamatergic stimulation frequency needed to elicit Ca2+ signals in the adjacent secondary process. These findings suggest that dopamine enhances the initiation and propagation of glutamate-evoked Ca2+ signals, with the extent of propagation depending on astrocytic morphology and the spatial distribution of glutamatergic inputs.
Collapse
Affiliation(s)
- Thiago Ohno Bezerra
- Department of Physics, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Antonio C Roque
- Department of Physics, School of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
5
|
Enriquez-Traba J, Arenivar M, Yarur-Castillo HE, Noh C, Flores RJ, Weil T, Roy S, Usdin TB, LaGamma CT, Wang H, Tsai VS, Kerspern D, Moritz AE, Sibley DR, Lutas A, Moratalla R, Freyberg Z, Tejeda HA. Dissociable control of motivation and reinforcement by distinct ventral striatal dopamine receptors. Nat Neurosci 2024:10.1038/s41593-024-01819-9. [PMID: 39653808 DOI: 10.1038/s41593-024-01819-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/22/2024] [Indexed: 12/18/2024]
Abstract
Dopamine (DA) release in striatal circuits, including the nucleus accumbens medial shell (mNAcSh), tracks separable features of reward like motivation and reinforcement. However, the cellular and circuit mechanisms by which DA receptors transform DA release into distinct constructs of reward remain unclear. Here we show that DA D3 receptor (D3R) signaling in the mNAcSh drives motivated behavior in mice by regulating local microcircuits. Furthermore, D3Rs coexpress with DA D1 receptors, which regulate reinforcement, but not motivation. Paralleling dissociable roles in reward function, we report nonoverlapping physiological actions of D3R and DA D1 receptor signaling in mNAcSh neurons. Our results establish a fundamental framework wherein DA signaling within the same nucleus accumbens cell type is physiologically compartmentalized via actions on distinct DA receptors. This structural and functional organization provides neurons in a limbic circuit with the unique ability to orchestrate dissociable aspects of reward-related behaviors relevant to the etiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Juan Enriquez-Traba
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
- Department of Biochemistry, Universidad Autonoma de Madrid, Madrid, Spain
- Department of Functional and Systems Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - Miguel Arenivar
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Hector E Yarur-Castillo
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Chloe Noh
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Rodolfo J Flores
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Tenley Weil
- Section on Light and Circadian Rhythms, National Institute of Mental Health, Bethesda, MD, USA
| | - Snehashis Roy
- Systems Neuroscience Imaging Resource, National Institute of Mental Health, Bethesda, MD, USA
| | - Ted B Usdin
- Systems Neuroscience Imaging Resource, National Institute of Mental Health, Bethesda, MD, USA
| | - Christina T LaGamma
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Huikun Wang
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Valerie S Tsai
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA
| | - Damien Kerspern
- Neuromodulation and Motivation Section, Diabetes, Endocrinology, & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Amy E Moritz
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Andrew Lutas
- Neuromodulation and Motivation Section, Diabetes, Endocrinology, & Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Rosario Moratalla
- Department of Functional and Systems Neurobiology, Instituto Cajal-CSIC, Madrid, Spain
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Schuster BA, Lamm C. How dopamine shapes trust beliefs. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111206. [PMID: 39586370 DOI: 10.1016/j.pnpbp.2024.111206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Learning whom to trust is integral for healthy relationships and social cohesion, and atypicalities in trust learning are common across a range of clinical conditions, including schizophrenia spectrum disorders, Parkinson's disease, and depression. Persecutory delusions - rigid, unfounded beliefs that others are intending to harm oneself - significantly impact affected individuals' lives as they are associated with a range of negative health outcomes, including suicidal behaviour and relapse. Recent advances in computational modelling and psychopharmacology have significantly extended our understanding of the brain bases of dynamic trust learning, and the neuromodulator dopamine has been suggested to play a key role in this. However, the specifics of this role on a computational and neurobiological level remain to be fully established. The current review article provides a comprehensive summary of novel conceptual developments and empirical findings regarding the computational role of dopamine in social learning processes. Research findings strongly suggest a conceptual shift, from dopamine as a reward mechanism to a teaching signal indicating which information is relevant for learning, and shed light on the neurocomputational mechanisms via which antipsychotics may alleviate symptoms of aberrant social learning processes such as persecutory delusions. Knowledge gaps and inconsistencies in the extant literature are examined and the most pressing issues highlighted, laying the foundation for future research that will further advance our understanding of the neuromodulation of social belief updating processes.
Collapse
Affiliation(s)
- Bianca A Schuster
- Department of Cognition, Emotion, and Methods in Psychology, University of Vienna, Liebiggasse 5, 1010 Vienna, Austria.
| | - Claus Lamm
- Department of Cognition, Emotion, and Methods in Psychology, University of Vienna, Liebiggasse 5, 1010 Vienna, Austria
| |
Collapse
|
7
|
Muehlberg C, Goerg S, Rullmann M, Hesse S, Sabri O, Wawrzyniak M, Classen J, Fricke C, Rumpf JJ. Motor learning is modulated by dopamine availability in the sensorimotor putamen. Brain Commun 2024; 6:fcae409. [PMID: 39584157 PMCID: PMC11582004 DOI: 10.1093/braincomms/fcae409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/16/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Successful motor skill acquisition requires the dynamic interaction of multiple brain regions, with the striatum playing a critical role in this network. Animal studies suggest that dopaminergic mechanisms are involved in the regulation of motor learning-associated striatal plasticity. In humans, however, the contribution of nigrostriatal dopaminergic transmission to motor learning remains elusive beyond its well-characterized role in initiation and fluent execution of movements. In this prospective observational study, we investigated motor sequence learning in individuals who had undergone 123I-N-ω-fluoropropyl-2β-carbomethoxy-3β-(4-iodophenyl)nortropane single-photon emission computed tomography for the differential diagnosis of Parkinson's disease (n = 41) and age-matched healthy controls (n = 20). We found that striatal dopamine transporter depletion exhibited distinct spatial patterns that were associated with impairments in motor sequence learning and the manifestation of Parkinsonian motor symptoms, respectively. Specifically, significant associations between striatal dopamine transporter depletion and impairments in motor sequence learning were confined to posterior putaminal regions, whereas significant associations of striatal dopamine transporter depletion with Parkinsonian motor symptom severity showed a widespread spatial pattern across the entire striatal volume with an anterior maximum. Normative functional connectivity analysis revealed that both behavioural domains shared largely overlapping connectivity patterns with the basal ganglia and supplementary motor area. However, apart from connectivity with more posterior parts of the supplementary motor area, significant functional connectivity with primary motor cortical areas was only present for striatal dopamine transporter availability-related modulation of online motor learning. Our findings indicate that striatal dopaminergic signalling plays a specific role in motor sequence learning beyond its influence on mere motor execution, implicating learning-related sensorimotor striatum recruitment and cortico-striatal plasticity as dopamine-dependent mechanisms.
Collapse
Affiliation(s)
- Christoph Muehlberg
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Sophia Goerg
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Swen Hesse
- Department of Nuclear Medicine, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Max Wawrzyniak
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Joseph Classen
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Christopher Fricke
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Jost-Julian Rumpf
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| |
Collapse
|
8
|
D'Souza SP, Upton BA, Eldred KC, Glass I, Nayak G, Grover K, Ahmed A, Nguyen MT, Hu YC, Gamlin P, Lang RA. Developmental control of rod number via a light-dependent retrograde pathway from intrinsically photosensitive retinal ganglion cells. Dev Cell 2024; 59:2897-2911.e6. [PMID: 39142280 PMCID: PMC11537824 DOI: 10.1016/j.devcel.2024.07.018] [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: 02/27/2024] [Revised: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024]
Abstract
Photoreception is essential for the development of the visual system, shaping vision's first synapse to cortical development. Here, we find that the lighting environment controls developmental rod apoptosis via Opn4-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). Using genetics, sensory environment manipulations, and computational approaches, we establish a pathway where light-dependent glutamate released from ipRGCs is detected via a transiently expressed glutamate receptor (Grik3) on rod precursors within the inner retina. Communication between these cells is mediated by hybrid neurites on ipRGCs that sense light before eye opening. These structures span the ipRGC-rod precursor distance over development and contain the machinery for photoreception (Opn4) and neurotransmitter release (Vglut2 & Syp). Assessment of the human gestational retina identifies conserved hallmarks of an ipRGC-to-rod axis, including displaced rod precursors, transient GRIK3 expression, and ipRGCs with deep-projecting neurites. This analysis defines an adaptive retrograde pathway linking the sensory environment to rod precursors via ipRGCs prior to eye opening.
Collapse
Affiliation(s)
- Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Brian A Upton
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kiara C Eldred
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Ian Glass
- Birth Defects Research Laboratory, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98195, USA
| | - Gowri Nayak
- Transgenic Animal and Genome Editing Core, Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kassidy Grover
- Division of Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Abdulla Ahmed
- Medical Doctor (M.D.) Training Program, George Washington University School of Medicine, Washington, DC 20052, USA
| | - Minh-Thanh Nguyen
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yueh-Chiang Hu
- Transgenic Animal and Genome Editing Core, Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Richard A Lang
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Ophthalmology, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
9
|
Herborg F. Substance- and Cell-Specific Roles of Mesolimbic Dopamine D 3 Receptors. Biol Psychiatry 2024; 96:691-693. [PMID: 39357968 DOI: 10.1016/j.biopsych.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Freja Herborg
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
10
|
Millidge B, Song Y, Lak A, Walton ME, Bogacz R. Reward Bases: A simple mechanism for adaptive acquisition of multiple reward types. PLoS Comput Biol 2024; 20:e1012580. [PMID: 39561186 PMCID: PMC11614280 DOI: 10.1371/journal.pcbi.1012580] [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: 04/25/2024] [Revised: 12/03/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Animals can adapt their preferences for different types of reward according to physiological state, such as hunger or thirst. To explain this ability, we employ a simple multi-objective reinforcement learning model that learns multiple values according to different reward dimensions such as food or water. We show that by weighting these learned values according to the current needs, behaviour may be flexibly adapted to present preferences. This model predicts that individual dopamine neurons should encode the errors associated with some reward dimensions more than with others. To provide a preliminary test of this prediction, we reanalysed a small dataset obtained from a single primate in an experiment which to our knowledge is the only published study where the responses of dopamine neurons to stimuli predicting distinct types of rewards were recorded. We observed that in addition to subjective economic value, dopamine neurons encode a gradient of reward dimensions; some neurons respond most to stimuli predicting food rewards while the others respond more to stimuli predicting fluids. We also proposed a possible implementation of the model in the basal ganglia network, and demonstrated how the striatal system can learn values in multiple dimensions, even when dopamine neurons encode mixtures of prediction error from different dimensions. Additionally, the model reproduces the instant generalisation to new physiological states seen in dopamine responses and in behaviour. Our results demonstrate how a simple neural circuit can flexibly guide behaviour according to animals' needs.
Collapse
Affiliation(s)
- Beren Millidge
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, United Kingdom
| | - Yuhang Song
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, United Kingdom
| | - Armin Lak
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Mark E. Walton
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Rafal Bogacz
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, United Kingdom
- Theoretical Sciences Visiting Program (TSVP), Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| |
Collapse
|
11
|
Cai X, Liu C, Tsutsui-Kimura I, Lee JH, Guo C, Banerjee A, Lee J, Amo R, Xie Y, Patriarchi T, Li Y, Watabe-Uchida M, Uchida N, Kaeser PS. Dopamine dynamics are dispensable for movement but promote reward responses. Nature 2024; 635:406-414. [PMID: 39415006 DOI: 10.1038/s41586-024-08038-z] [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: 07/21/2022] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
Dopamine signalling modes differ in kinetics and spatial patterns of receptor activation1,2. How these modes contribute to motor function, motivation and learning has long been debated3-21. Here we show that action-potential-induced dopamine release is dispensable for movement initiation but supports reward-oriented behaviour. We generated mice with dopamine-neuron-specific knockout of the release site organizer protein RIM to disrupt action-potential-induced dopamine release. In these mice, rapid in vivo dopamine dynamics were strongly impaired, but baseline dopamine persisted and fully supported spontaneous movement. Conversely, reserpine-mediated dopamine depletion or blockade of dopamine receptors disrupted movement initiation. The dopamine precursor L-DOPA reversed reserpine-induced bradykinesia without restoring fast dopamine dynamics, a result that substantiated the conclusion that these dynamics are dispensable for movement initiation. In contrast to spontaneous movement, reward-oriented behaviour was impaired in dopamine-neuron-specific RIM knockout mice. In conditioned place preference and two-odour discrimination tasks, the mice effectively learned to distinguish the cues, which indicates that reward-based learning persists after RIM ablation. However, the performance vigour was reduced. During probabilistic cue-reward association, dopamine dynamics and conditioned responses assessed through anticipatory licking were disrupted. These results demonstrate that action-potential-induced dopamine release is dispensable for motor function and subsecond precision of movement initiation but promotes motivation and performance during reward-guided behaviours.
Collapse
Affiliation(s)
- Xintong Cai
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Iku Tsutsui-Kimura
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Joon-Hyuk Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Chong Guo
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Jinoh Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yudi Xie
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024; 29:3680-3693. [PMID: 38789677 PMCID: PMC11540752 DOI: 10.1038/s41380-024-02608-3] [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: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
13
|
Gao X, Wei H, Ma W, Wu W, Ji W, Mao J, Yu P, Mao L. Inflammation-free electrochemical in vivo sensing of dopamine with atomic-level engineered antioxidative single-atom catalyst. Nat Commun 2024; 15:7915. [PMID: 39256377 PMCID: PMC11387648 DOI: 10.1038/s41467-024-52279-5] [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: 02/07/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
Electrochemical methods with tissue-implantable microelectrodes provide an excellent platform for real-time monitoring the neurochemical dynamics in vivo due to their superior spatiotemporal resolution and high selectivity and sensitivity. Nevertheless, electrode implantation inevitably damages the brain tissue, upregulates reactive oxygen species level, and triggers neuroinflammatory response, resulting in unreliable quantification of neurochemical events. Herein, we report a multifunctional sensing platform for inflammation-free in vivo analysis with atomic-level engineered Fe single-atom catalyst that functions as both single-atom nanozyme with antioxidative activity and electrode material for dopamine oxidation. Through high-temperature pyrolysis and catalytic performance screening, we fabricate a series of Fe single-atom nanozymes with different coordination configurations and find that the Fe single-atom nanozyme with FeN4 exhibits the highest activity toward mimicking catalase and superoxide dismutase as well as eliminating hydroxyl radical, while also featuring high electrode reactivity toward dopamine oxidation. These dual functions endow the single-atom nanozyme-based sensor with anti-inflammatory capabilities, enabling accurate dopamine sensing in living male rat brain. This study provides an avenue for designing inflammation-free electrochemical sensing platforms with atomic-precision engineered single-atom catalysts.
Collapse
Affiliation(s)
- Xiaolong Gao
- College of Chemistry, Beijing Normal University, 100875, Beijing, China
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, The Chinese Academy of Sciences (CAS), 100190, Beijing, China
| | - Huan Wei
- College of Chemistry, Beijing Normal University, 100875, Beijing, China
| | - Wenjie Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, The Chinese Academy of Sciences (CAS), 100190, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Wenjie Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, The Chinese Academy of Sciences (CAS), 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Wenliang Ji
- College of Chemistry, Beijing Normal University, 100875, Beijing, China
| | - Junjie Mao
- College of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241002, China
| | - Ping Yu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, The Chinese Academy of Sciences (CAS), 100190, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, 100875, Beijing, China.
| |
Collapse
|
14
|
Kuiper LB, Dawes MH, West AM, DiMarco EK, Galante EV, Kishida KT, Jones SR. Comparison of dopamine release and uptake parameters across sex, species and striatal subregions. Eur J Neurosci 2024; 60:5113-5140. [PMID: 39161062 PMCID: PMC11632670 DOI: 10.1111/ejn.16495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 07/05/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
For over four decades, fast-scan cyclic voltammetry (FSCV) has been used to selectively measure neurotransmitters such as dopamine (DA) with high spatial and temporal resolution, providing detailed information about the regulation of DA in the extracellular space. FSCV is an optimal method for determining concentrations of stimulus-evoked DA in brain tissue. When modelling diseases involving disturbances in DA transmission, preclinical rodent models are especially useful because of the availability of specialized tools and techniques that serve as a foundation for translational research. There is known heterogeneity in DA dynamics between and within DA-innervated brain structures and between males and females. However, systematic evaluations of sex- and species-differences across multiple areas are lacking. Therefore, using FSCV, we captured a broad range of DA dynamics across five sub-regions of the dorsal and ventral striatum of males and females of both rats and mice that reflect the functional heterogeneity of DA kinetics and dynamics within these structures. While numerous differences were found, in particular, we documented a strong, consistent pattern of increased DA transporter activity in females in all of the regions surveyed. The data herein are intended to be used as a resource for further investigation of DA terminal function.
Collapse
Affiliation(s)
- Lindsey B. Kuiper
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Monica H. Dawes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alyssa M. West
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Emily K. DiMarco
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Emma V. Galante
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Kenneth T. Kishida
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
- Department of Neurosurgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Sara R. Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
15
|
Hou M, Herold F, Zhang Z, Ando S, Cheval B, Ludyga S, Erickson KI, Hillman CH, Yu Q, Liu-Ambrose T, Kuang J, Kramer AF, Chen Y, Costello JT, Chen C, Dupuy O, Pindus DM, McMorris T, Stiernman L, Zou L. Human dopaminergic system in the exercise-cognition link. Trends Mol Med 2024; 30:708-712. [PMID: 38719712 DOI: 10.1016/j.molmed.2024.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 08/17/2024]
Abstract
While the dopaminergic system is important for cognitive processes, it is also sensitive to the influence of physical activity (PA). We summarize current evidence on whether PA-related changes in the human dopaminergic system are associated with alterations in cognitive performance, discuss recent advances, and highlight challenges and opportunities for future research.
Collapse
Affiliation(s)
- Meijun Hou
- Body-Brain-Mind Laboratory, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Fabian Herold
- Body-Brain-Mind Laboratory, School of Psychology, Shenzhen University, Shenzhen 518060, China; Research Group Degenerative and Chronic Diseases, Movement, Faculty of Health Sciences Brandenburg, University of Potsdam, Potsdam 14476, Germany
| | - Zhihao Zhang
- Body-Brain-Mind Laboratory, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Soichi Ando
- Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan; Nuclear Medicine Laboratory, Research Center for Accelerator and Radioisotope Science, Tohoku University, Miyagi, Japan
| | - Boris Cheval
- Department of Sport Sciences and Physical Education, Ecole Normale Supérieure Rennes, Bruz, France; Laboratory VIPS2, University of Rennes, Rennes, France
| | - Sebastian Ludyga
- Department of Sport, Exercise, and Health, University of Basel, Basel 4052, Switzerland
| | - Kirk I Erickson
- AdventHealth Research Institute, Department of Neuroscience, Advent Health, Orlando, FL, USA; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles H Hillman
- Center for Cognitive and Brain Health, Northeastern University, Boston, MA 02115, USA; Department of Psychology, Northeastern University, Boston, MA 02115, USA; Department of Physical Therapy, Movement, and Rehabilitation Sciences, Northeastern University, Boston, MA 02115, USA
| | - Qian Yu
- Faculty of Education, University of Macau, Macau 999078, China
| | - Teresa Liu-Ambrose
- Aging, Mobility, and Cognitive Health Laboratory, Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, Vancouver Coastal Health Research Institute, University of British Columbia, Canada; Centre for Aging SMART at Vancouver Coastal Health, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Jin Kuang
- Body-Brain-Mind Laboratory, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Arthur F Kramer
- Center for Cognitive and Brain Health, Northeastern University, Boston, MA 02115, USA; Department of Psychology, Northeastern University, Boston, MA 02115, USA; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Yanxia Chen
- Body-Brain-Mind Laboratory, School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Joseph T Costello
- Extreme Environments Laboratory, School of Sport, Health, and Exercise Science, University of Portsmouth, Portsmouth, UK
| | - Chong Chen
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Olivier Dupuy
- Laboratory MOVE (UR 20296), Faculty of Sport Sciences, University of Poitiers, Poitiers, France; School of Kinesiology and Physical Activity Science (EKAPS), Faculty of Medicine, University of Montreal, Montreal, Canada
| | - Dominika M Pindus
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Terry McMorris
- Institue of Sport, Nursing and Allied Health, University of Chichester, College Lane, Chichester PO19 6PE, UK
| | - Lars Stiernman
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden; Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Liye Zou
- Body-Brain-Mind Laboratory, School of Psychology, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
16
|
Liu J, Jiang Y, Liu R, Jin J, Wei S, Ji W, He X, Wu F, Yu P, Mao L. Vitamin C Drives Reentrant Actin Phase Transition: Biphasic Exocytosis Regulation Revealed by Single-Vesicle Electrochemistry. J Am Chem Soc 2024; 146:17747-17756. [PMID: 38889317 DOI: 10.1021/jacs.4c02710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Unveiling molecular mechanisms that dominate protein phase dynamics has been a pressing need for deciphering the intricate intracellular modulation machinery. While ions and biomacromolecules have been widely recognized for modulating protein phase separations, effects of small molecules that essentially constitute the cytosolic chemical atmosphere on the protein phase behaviors are rarely understood. Herein, we report that vitamin C (VC), a key small molecule for maintaining a reductive intracellular atmosphere, drives reentrant phase transitions of myosin II/F-actin (actomyosin) cytoskeletons. The actomyosin bundle condensates dissemble in the low-VC regime and assemble in the high-VC regime in vitro or inside neuronal cells, through a concurrent myosin II protein aggregation-dissociation process with monotonic VC concentration increase. Based on this finding, we employ in situ single-cell and single-vesicle electrochemistry to demonstrate the quantitative modulation of catecholamine transmitter vesicle exocytosis by intracellular VC atmosphere, i.e., exocytotic release amount increases in the low-VC regime and decreases in the high-VC regime. Furthermore, we show how VC regulates cytomembrane-vesicle fusion pore dynamics through counteractive or synergistic effects of actomyosin phase transitions and the intracellular free calcium level on membrane tensions. Our work uncovers the small molecule-based reversive protein phase regulatory mechanism, paving a new way to chemical neuromodulation and therapeutic repertoire expansion.
Collapse
Affiliation(s)
- Jing Liu
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- College of Chemistry, Beijing Normal University, Beijing 100875, China
- Institute of Analysis and Testing, Beijing Academy of Science and Technology, Beijing 100089, China
| | - Ying Jiang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ran Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jing Jin
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Shiyi Wei
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wenliang Ji
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xiulan He
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Fei Wu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ping Yu
- Beijing National Laboratory for Molecular Science, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
17
|
Labouesse MA, Wilhelm M, Kagiampaki Z, Yee AG, Denis R, Harada M, Gresch A, Marinescu AM, Otomo K, Curreli S, Serratosa Capdevila L, Zhou X, Cola RB, Ravotto L, Glück C, Cherepanov S, Weber B, Zhou X, Katner J, Svensson KA, Fellin T, Trudeau LE, Ford CP, Sych Y, Patriarchi T. A chemogenetic approach for dopamine imaging with tunable sensitivity. Nat Commun 2024; 15:5551. [PMID: 38956067 PMCID: PMC11219860 DOI: 10.1038/s41467-024-49442-3] [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: 11/07/2023] [Accepted: 06/05/2024] [Indexed: 07/04/2024] Open
Abstract
Genetically-encoded dopamine (DA) sensors enable high-resolution imaging of DA release, but their ability to detect a wide range of extracellular DA levels, especially tonic versus phasic DA release, is limited by their intrinsic affinity. Here we show that a human-selective dopamine receptor positive allosteric modulator (PAM) can be used to boost sensor affinity on-demand. The PAM enhances DA detection sensitivity across experimental preparations (in vitro, ex vivo and in vivo) via one-photon or two-photon imaging. In vivo photometry-based detection of optogenetically-evoked DA release revealed that DETQ administration produces a stable 31 minutes window of potentiation without effects on animal behavior. The use of the PAM revealed region-specific and metabolic state-dependent differences in tonic DA levels and enhanced single-trial detection of behavior-evoked phasic DA release in cortex and striatum. Our chemogenetic strategy can potently and flexibly tune DA imaging sensitivity and reveal multi-modal (tonic/phasic) DA signaling across preparations and imaging approaches.
Collapse
Affiliation(s)
- Marie A Labouesse
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Institute for Neuroscience, ETH Zurich, Zurich, Switzerland
| | | | - Andrew G Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Raphaelle Denis
- Department of Pharmacology & Physiology, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
| | - Masaya Harada
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Andrea Gresch
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | | | - Kanako Otomo
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Sebastiano Curreli
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Xuehan Zhou
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Reto B Cola
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Stanislav Cherepanov
- Institute of Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Xin Zhou
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Louis-Eric Trudeau
- Department of Pharmacology & Physiology, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yaroslav Sych
- Institute of Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
18
|
Gooding SW, Lewis E, Chau C, Sandhu S, Glienke J, Whistler JL. Nucleus accumbens sub-regions experience distinct dopamine release responses following acute and chronic morphine exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601282. [PMID: 39005415 PMCID: PMC11244850 DOI: 10.1101/2024.06.28.601282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
It is well established that dopamine neurons of the ventral tegmental area (VTA) play a critical role in reward and aversion as well as pathologies including drug dependence and addiction. The distinct effects of acute and chronic opioid exposure have been previously characterized at VTA synapses. Recent work suggests that distinct VTA projections that target the medial and lateral shell of the nucleus accumbens (NAc), may play opposing roles in modulating behavior. It is possible that these two anatomically and functionally distinct pathways also have disparate roles in opioid reward, tolerance, and withdrawal in the brain. In this study we monitored dopamine release in the medial or lateral shell of the NAc of male mice during a week-long morphine treatment paradigm. We measured dopamine release in response to an intravenous morphine injection both acutely and following a week of repeated morphine. We also measured dopamine in response to a naloxone injection both prior to and following repeated morphine treatment. Morphine induced a transient increase in dopamine in the medial NAc shell that was much larger than the slower rise observed in the lateral shell. Surprisingly, chronic morphine treatment induced a sensitization of the medial dopamine response to morphine that opposed a diminished response observed in the saline-treated control group. This study expands on our current understanding of the medial NAc shell as hub of opioid-induced dopamine fluctuation. It also highlights the need for future opioid studies to appreciate the heterogeneity of dopamine neurons. Significance Statement The social and economic consequences of the opioid epidemic are tragic and far-reaching. Yet, opioids are indisputably necessary in clinical settings where they remain the most useful treatment for severe pain. To combat this crisis, we must improve our understanding of opioid function in the brain, particularly the neural mechanisms that underlie opioid dependence and addictive behaviors. This study uses fiber photometry to examine dopamine changes that occur in response to repeated morphine, and morphine withdrawal, at multiple stages of a longitudinal opioid-dependence paradigm. We reveal key differences in how dopamine levels respond to opioid administration in distinct sub-regions of the ventral striatum and lay a foundation for future opioid research that appreciates our contemporary understanding of the dopamine system.
Collapse
Affiliation(s)
| | - Elinor Lewis
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Christine Chau
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Suhail Sandhu
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Julianna Glienke
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
19
|
Tai MDS, Gamiz-Arco G, Martinez A. Dopamine synthesis and transport: current and novel therapeutics for parkinsonisms. Biochem Soc Trans 2024; 52:1275-1291. [PMID: 38813865 PMCID: PMC11346439 DOI: 10.1042/bst20231061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
Parkinsonism is the primary type of movement disorder in adults, encompassing a set of clinical symptoms, including rigidity, tremors, dystonia, bradykinesia, and postural instability. These symptoms are primarily caused by a deficiency in dopamine (DA), an essential neurotransmitter in the brain. Currently, the DA precursor levodopa (synthetic L-DOPA) is the standard medication to treat DA deficiency, but it only addresses symptoms rather than provides a cure. In this review, we provide an overview of disorders associated with DA dysregulation and deficiency, particularly Parkinson's disease and rare inherited disorders leading predominantly to dystonia and/or parkinsonism, even in childhood. Although levodopa is relatively effective for the management of motor dysfunctions, it is less effective for severe forms of parkinsonism and is also associated with side effects and a loss of efficacy over time. We present ongoing efforts to reinforce the effect of levodopa and to develop innovative therapies that target the underlying pathogenic mechanisms affecting DA synthesis and transport, increasing neurotransmission through disease-modifying approaches, such as cell-based therapies, nucleic acid- and protein-based biologics, and small molecules.
Collapse
Affiliation(s)
| | - Gloria Gamiz-Arco
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, 5020 Bergen, Norway
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
20
|
Qu L, Li Y, Liu F, Fang Y, He J, Ma J, Xu T, Wang L, Lei P, Dong H, Jin L, Yang Q, Wu W, Sun D. Microbiota-Gut-Brain Axis Dysregulation in Alzheimer's Disease: Multi-Pathway Effects and Therapeutic Potential. Aging Dis 2024; 15:1108-1131. [PMID: 37728579 PMCID: PMC11081173 DOI: 10.14336/ad.2023.0823-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
An essential regulator of neurodegenerative conditions like Alzheimer's disease (AD) is the gut microbiota. Alterations in intestinal permeability brought on by gut microbiota dysregulation encourage neuroinflammation, central immune dysregulation, and peripheral immunological dysregulation in AD, as well as hasten aberrant protein aggregation and neuronal death in the brain. However, it is unclear how the gut microbiota transmits information to the brain and how it influences brain cognition and function. In this review, we summarized the multiple pathways involved in the gut microbiome in AD and provided detailed treatment strategies based on the gut microbiome. Based on these observations, this review also discusses the problems, challenges, and strategies to address current therapeutic strategies.
Collapse
Affiliation(s)
- Linkai Qu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
- College of Veterinary Medicine, Jilin University, Changchun 130118, China.
| | - Yanwei Li
- Core Facilities, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Fan Liu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Ting Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China.
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| |
Collapse
|
21
|
Li H, Zhang Y, Deng Z, Lu B, Ma L, Wang R, Wang X, Jiao Z, Wang Y, Zhou K, Wei Q. Constructing a Hydrophilic Microsensor for High-Antifouling Neurotransmitter Dopamine Sensing. ACS Sens 2024; 9:1785-1798. [PMID: 38384144 DOI: 10.1021/acssensors.3c02042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Real-time sensing of dopamine is essential for understanding its physiological function and clarifying the pathophysiological mechanism of diseases caused by impaired dopamine systems. However, severe fouling from nonspecific protein adsorption, for a long time, limited conventional neural recording electrodes concerning recording stability. This study reported a high-antifouling nanocrystalline boron-doped diamond microsensor grown on a carbon fiber substrate. The antifouling properties of this diamond sensor were strongly related to the grain size (i.e., nanocrystalline and microcrystalline) and surface terminations (i.e., oxygen and hydrogen terminals). Experimental observations and molecular dynamics calculations demonstrated that the oxygen-terminated nanocrystalline boron-doped diamond microsensor exhibited enhanced antifouling characteristics against protein adsorption, which was attributed to the formation of a strong hydration layer as a physical and energetic barrier that prevents protein adsorption on the surface. This finally allowed for in vivo monitoring of dopamine in rat brains upon potassium chloride stimulation, thus presenting a potential solution for the design of next-generation antifouling neural recording sensors. Experimental observations and molecular dynamics calculations demonstrated that the oxygen-terminated nanocrystalline boron-doped diamond (O-NCBDD) microsensor exhibited ultrahydrophilic properties with a contact angle of 4.9°, which was prone to forming a strong hydration layer as a physical and energetic barrier to withstand the adsorption of proteins. The proposed O-NCBDD microsensor exhibited a high detection sensitivity of 5.14 μA μM-1 cm-2 and a low detection limit of 25.7 nM. This finally allowed for in vivo monitoring of dopamine with an average concentration of 1.3 μM in rat brains upon 2 μL of potassium chloride stimulation, thus presenting a potential solution for the design of next-generation antifouling neural recording sensors.
Collapse
Affiliation(s)
- Haichao Li
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Yening Zhang
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410000, P. R. China
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, P. R. China
| | - Zejun Deng
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Ben Lu
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410000, P. R. China
- Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, P. R. China
| | - Li Ma
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Run Wang
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Xiang Wang
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Zengkai Jiao
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Yijia Wang
- Institute for Advanced Study, Central South University, Changsha 410083, P. R. China
| | - Kechao Zhou
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| | - Qiuping Wei
- State Key Laboratory of Powder Metallurgy, School of Materials Science and Engineering, Central South University, Changsha 410083, P. R. China
| |
Collapse
|
22
|
Pan K, Jinnah HA, Hess EJ, Smith Y, Villalba RM. Ultrastructural analysis of nigrostriatal dopaminergic terminals in a knockin mouse model of DYT1 dystonia. Eur J Neurosci 2024; 59:1407-1427. [PMID: 38123503 DOI: 10.1111/ejn.16197] [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: 03/08/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 12/23/2023]
Abstract
DYT1 dystonia is associated with decreased striatal dopamine release. In this study, we examined the possibility that ultrastructural changes of nigrostriatal dopamine terminals could contribute to this neurochemical imbalance using a serial block face/scanning electron microscope (SBF/SEM) and three-dimensional reconstruction to analyse striatal tyrosine hydroxylase-immunoreactive (TH-IR) terminals and their synapses in a DYT1(ΔE) knockin (DYT1-KI) mouse model of DYT1 dystonia. Furthermore, to study possible changes in vesicle packaging capacity of dopamine, we used transmission electron microscopy to assess the synaptic vesicle size in striatal dopamine terminals. Quantitative comparative analysis of 80 fully reconstructed TH-IR terminals in the WT and DYT1-KI mice indicate (1) no significant difference in the volume of TH-IR terminals; (2) no major change in the proportion of axo-spinous versus axo-dendritic synapses; (3) no significant change in the post-synaptic density (PSD) area of axo-dendritic synapses, while the PSDs of axo-spinous synapses were significantly smaller in DYT1-KI mice; (4) no significant change in the contact area between TH-IR terminals and dendritic shafts or spines, while the ratio of PSD area/contact area decreased significantly for both axo-dendritic and axo-spinous synapses in DYT1-KI mice; (5) no significant difference in the mitochondria volume; and (6) no significant difference in the synaptic vesicle area between the two groups. Altogether, these findings suggest that abnormal morphometric changes of nigrostriatal dopamine terminals and their post-synaptic targets are unlikely to be a major source of reduced striatal dopamine release in DYT1 dystonia.
Collapse
Affiliation(s)
- Ke Pan
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Physical Therapy & Human Movement Sciences, Northwestern University, Chicago, Illinois, USA
| | - Hyder A Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Human Genetics and Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Ellen J Hess
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, Georgia, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Rosa M Villalba
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Patel JC, Sherpa AD, Melani R, Witkovsky P, Wiseman MR, O'Neill B, Aoki C, Tritsch NX, Rice ME. GABA co-released from striatal dopamine axons dampens phasic dopamine release through autoregulatory GABA A receptors. Cell Rep 2024; 43:113834. [PMID: 38431842 PMCID: PMC11089423 DOI: 10.1016/j.celrep.2024.113834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Striatal dopamine axons co-release dopamine and gamma-aminobutyric acid (GABA), using GABA provided by uptake via GABA transporter-1 (GAT1). Functions of GABA co-release are poorly understood. We asked whether co-released GABA autoinhibits dopamine release via axonal GABA type A receptors (GABAARs), complementing established inhibition by dopamine acting at axonal D2 autoreceptors. We show that dopamine axons express α3-GABAAR subunits in mouse striatum. Enhanced dopamine release evoked by single-pulse optical stimulation in striatal slices with GABAAR antagonism confirms that an endogenous GABA tone limits dopamine release. Strikingly, an additional inhibitory component is seen when multiple pulses are used to mimic phasic axonal activity, revealing the role of GABAAR-mediated autoinhibition of dopamine release. This autoregulation is lost in conditional GAT1-knockout mice lacking GABA co-release. Given the faster kinetics of ionotropic GABAARs than G-protein-coupled D2 autoreceptors, our data reveal a mechanism whereby co-released GABA acts as a first responder to dampen phasic-to-tonic dopamine signaling.
Collapse
Affiliation(s)
- Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | - Ang D Sherpa
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Riccardo Melani
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Paul Witkovsky
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Madeline R Wiseman
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Brian O'Neill
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Chiye Aoki
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; Center for Neural Science New York University, 4 Washington Place, New York, NY 10003, USA
| | - Nicolas X Tritsch
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA; NYU Neuroscience Institute, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
24
|
Stowell R, Wang KH. Dopaminergic signaling regulates microglial surveillance and adolescent plasticity in the frontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584167. [PMID: 38559264 PMCID: PMC10979918 DOI: 10.1101/2024.03.08.584167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Adolescence is a sensitive period for frontal cortical development and cognitive maturation. The dopaminergic (DA) mesofrontal circuit is particularly malleable in response to changes in adolescent experience and DA activity. However, the cellular mechanisms engaged in this plasticity remain unexplored. Here, we report that microglia, the innate immune cells of the brain, are uniquely sensitive to adolescent mesofrontal DA signaling. Longitudinal in vivo two-photon imaging in mice shows that frontal cortical microglia respond dynamically to plasticity-inducing behavioral or optogenetic DA axon stimulation with increased parenchymal and DA bouton surveillance. Microglial-axon contact precedes new bouton formation, and both D1 and D2-type DA receptors regulate microglial-bouton interactions and axonal plasticity. Moreover, D2 antagonism in adults reinstates adolescent plasticity, including increased microglial surveillance and new DA bouton formation. Our results reveal that DA signaling regulates microglial surveillance and axonal plasticity uniquely in the adolescent frontal cortex, presenting potential interventions for restoring plasticity in the adult brain.
Collapse
Affiliation(s)
- Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| | - Kuan Hong Wang
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| |
Collapse
|
25
|
Hamati R, Ahrens J, Shvetz C, Holahan MR, Tuominen L. 65 years of research on dopamine's role in classical fear conditioning and extinction: A systematic review. Eur J Neurosci 2024; 59:1099-1140. [PMID: 37848184 DOI: 10.1111/ejn.16157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023]
Abstract
Dopamine, a catecholamine neurotransmitter, has historically been associated with the encoding of reward, whereas its role in aversion has received less attention. Here, we systematically gathered the vast evidence of the role of dopamine in the simplest forms of aversive learning: classical fear conditioning and extinction. In the past, crude methods were used to augment or inhibit dopamine to study its relationship with fear conditioning and extinction. More advanced techniques such as conditional genetic, chemogenic and optogenetic approaches now provide causal evidence for dopamine's role in these learning processes. Dopamine neurons encode conditioned stimuli during fear conditioning and extinction and convey the signal via activation of D1-4 receptor sites particularly in the amygdala, prefrontal cortex and striatum. The coordinated activation of dopamine receptors allows for the continuous formation, consolidation, retrieval and updating of fear and extinction memory in a dynamic and reciprocal manner. Based on the reviewed literature, we conclude that dopamine is crucial for the encoding of classical fear conditioning and extinction and contributes in a way that is comparable to its role in encoding reward.
Collapse
Affiliation(s)
- Rami Hamati
- Neuroscience Graduate Program, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Institute of Mental Health Research, University of Ottawa, Ottawa, Ontario, Canada
| | - Jessica Ahrens
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Cecelia Shvetz
- University of Ottawa Institute of Mental Health Research, University of Ottawa, Ottawa, Ontario, Canada
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Matthew R Holahan
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Lauri Tuominen
- University of Ottawa Institute of Mental Health Research, University of Ottawa, Ottawa, Ontario, Canada
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Department of Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
26
|
Bellon A. Comparing stem cells, transdifferentiation and brain organoids as tools for psychiatric research. Transl Psychiatry 2024; 14:127. [PMID: 38418498 PMCID: PMC10901833 DOI: 10.1038/s41398-024-02780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 03/01/2024] Open
Abstract
The inaccessibility of neurons coming directly from patients has hindered our understanding of mental illnesses at the cellular level. To overcome this obstacle, six different cellular approaches that carry the genetic vulnerability to psychiatric disorders are currently available: Olfactory Neuroepithelial Cells, Mesenchymal Stem Cells, Pluripotent Monocytes, Induced Pluripotent Stem Cells, Induced Neuronal cells and more recently Brain Organoids. Here we contrast advantages and disadvantages of each of these six cell-based methodologies. Neuronal-like cells derived from pluripotent monocytes are presented in more detail as this technique was recently used in psychiatry for the first time. Among the parameters used for comparison are; accessibility, need for reprograming, time to deliver differentiated cells, differentiation efficiency, reproducibility of results and cost. We provide a timeline on the discovery of these cell-based methodologies, but, our main goal is to assist researchers selecting which cellular approach is best suited for any given project. This manuscript also aims to help readers better interpret results from the published literature. With this goal in mind, we end our work with a discussion about the differences and similarities between cell-based techniques and postmortem research, the only currently available tools that allow the study of mental illness in neurons or neuronal-like cells coming directly from patients.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA, USA.
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, USA.
| |
Collapse
|
27
|
Sadibolova R, DiMarco EK, Jiang A, Maas B, Tatter SB, Laxton A, Kishida KT, Terhune DB. Sub-second and multi-second dopamine dynamics underlie variability in human time perception. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.09.24302276. [PMID: 38370629 PMCID: PMC10871373 DOI: 10.1101/2024.02.09.24302276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Timing behaviour and the perception of time are fundamental to cognitive and emotional processes in humans. In non-human model organisms, the neuromodulator dopamine has been associated with variations in timing behaviour, but the connection between variations in dopamine levels and the human experience of time has not been directly assessed. Here, we report how dopamine levels in human striatum, measured with sub-second temporal resolution during awake deep brain stimulation surgery, relate to participants' perceptual judgements of time intervals. Fast, phasic, dopaminergic signals were associated with underestimation of temporal intervals, whereas slower, tonic, decreases in dopamine were associated with poorer temporal precision. Our findings suggest a delicate and complex role for the dynamics and tone of dopaminergic signals in the conscious experience of time in humans.
Collapse
Affiliation(s)
- Renata Sadibolova
- Department of Psychology, Goldsmiths, University of London; London SE14 6NW, UK
- Department of Psychology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London; London SE5 8AB, UK
- School of Psychology, University of Roehampton; London SW15 4JD, UK
| | - Emily K. DiMarco
- Neuroscience Graduate Program, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Department of Translational Neuroscience, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
| | - Angela Jiang
- Department of Translational Neuroscience, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
| | - Benjamin Maas
- Department of Translational Neuroscience, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Virginia Tech – Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Department of Biomedical Engineering, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
| | - Stephen B. Tatter
- Department of Neurosurgery, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
| | - Adrian Laxton
- Department of Neurosurgery, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
| | - Kenneth T. Kishida
- Neuroscience Graduate Program, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Department of Translational Neuroscience, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Virginia Tech – Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Department of Biomedical Engineering, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
- Department of Neurosurgery, Wake Forest School of Medicine; Winston-Salem, NC, 27157, USA
| | - Devin B. Terhune
- Department of Psychology, Goldsmiths, University of London; London SE14 6NW, UK
- Department of Psychology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London; London SE5 8AB, UK
| |
Collapse
|
28
|
Watanabe H, Dijkstra JM, Nagatsu T. Parkinson's Disease: Cells Succumbing to Lifelong Dopamine-Related Oxidative Stress and Other Bioenergetic Challenges. Int J Mol Sci 2024; 25:2009. [PMID: 38396687 PMCID: PMC10888576 DOI: 10.3390/ijms25042009] [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: 12/29/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
The core pathological event in Parkinson's disease (PD) is the specific dying of dopamine (DA) neurons of the substantia nigra pars compacta (SNc). The reasons why SNc DA neurons are especially vulnerable and why idiopathic PD has only been found in humans are still puzzling. The two main underlying factors of SNc DA neuron vulnerability appear related to high DA production, namely (i) the toxic effects of cytoplasmic DA metabolism and (ii) continuous cytosolic Ca2+ oscillations in the absence of the Ca2+-buffer protein calbindin. Both factors cause oxidative stress by producing highly reactive quinones and increasing intra-mitochondrial Ca2+ concentrations, respectively. High DA expression in human SNc DA neuron cell bodies is suggested by the abundant presence of the DA-derived pigment neuromelanin, which is not found in such abundance in other species and has been associated with toxicity at higher levels. The oxidative stress created by their DA production system, despite the fact that the SN does not use unusually high amounts of energy, explains why SNc DA neurons are sensitive to various genetic and environmental factors that create mitochondrial damage and thereby promote PD. Aging increases multiple risk factors for PD, and, to a large extent, PD is accelerated aging. To prevent PD neurodegeneration, possible approaches that are discussed here are (1) reducing cytoplasmic DA accumulation, (2) blocking cytoplasmic Ca2+ oscillations, and (3) providing bioenergetic support.
Collapse
Affiliation(s)
- Hirohisa Watanabe
- Department of Neurology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Johannes M. Dijkstra
- Center for Medical Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Toshiharu Nagatsu
- Center for Research Promotion and Support, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| |
Collapse
|
29
|
Sharp BM, Jiang Q, Kim P, Chen H. Inactivation of phosphodiesterase-4B gene in rat nucleus accumbens shell by CRISPR/Cas9 or positive allosteric modulation of the protein affects the motivation to chronically self-administer nicotine. Sci Rep 2024; 14:2562. [PMID: 38297069 PMCID: PMC10831042 DOI: 10.1038/s41598-024-53037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/27/2024] [Indexed: 02/02/2024] Open
Abstract
Large scale human genome wide association studies (GWAS) have identified a growing pool of genes associated with cigarette smoking. One of the most prominent, phosphodiesterase-4B (PDE4B), has been associated with multiple smoking phenotypes. Although PDE4B modulates the half-life of neuronal cAMP, its precise role in smoking behaviors is unknown. To address this knowledge gap, we used a reverse translational approach. We inactivated PDE4B in bilateral medial nucleus accumbens shell (NAcs) neurons by injecting AAV containing a specific gRNA in female transgenic Cas9+ Long Evans rats. These rats then were given 23-h chronic access to nicotine intravenous self-administration (IVSA) under a schedule of increasing fixed ratios (FR). With the increased effort required at FR7, nicotine SA (i.e. active presses and drug infusions) declined significantly in controls, whereas it was maintained in the mutagenized group. A progressive ratio (PR) study also showed significantly greater cumulative nicotine infusions in the PDE4B-edited group. Hence, we hypothesized that enhanced PDE4B protein activity would reduce nicotine IVSA. A positive allosteric modulator, 2-(3-(4-chloro-3-fluorophenyl)-5-ethyl-1H-1,2,4-triazol-1-yl)-N-(3,5-dichlorobenzyl)acetamide (MR-L2), was microinfused into NAcs bilaterally at FR3 or FR5; in both cohorts, MR-L2 acutely reduced nicotine IVSA. In summary, these studies show that the activity of PDE4B regulates the capacity of NAcs to maintain nicotine IVSA in face of the cost of increasing work. This finding and the results of the PR study indicate that PDE4B affects the motivation to obtain nicotine. These reverse translational studies in rats provide insight into the motivational effects of NAcs PDE4B that advance our understanding of the smoking behaviors mapped in human GWAS.
Collapse
Affiliation(s)
- Burt M Sharp
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Qin Jiang
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Panjun Kim
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
30
|
Li L, Rana AN, Li EM, Feng J, Li Y, Bruchas MR. Activity-dependent constraints on catecholamine signaling. Cell Rep 2023; 42:113566. [PMID: 38100349 PMCID: PMC11090260 DOI: 10.1016/j.celrep.2023.113566] [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: 06/11/2023] [Revised: 10/24/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Catecholamine signaling is thought to modulate cognition in an inverted-U relationship, but the mechanisms are unclear. We measured norepinephrine and dopamine release, postsynaptic calcium responses, and interactions between tonic and phasic firing modes under various stimuli and conditions. High tonic activity in vivo depleted catecholamine stores, desensitized postsynaptic responses, and decreased phasic transmission. Together, these findings provide a more complete understanding of the inverted-U relationship, offering insights into psychiatric disorders and neurodegenerative diseases with impaired catecholamine signaling.
Collapse
Affiliation(s)
- Li Li
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Seattle Children's Research Institute, Seattle, WA 98101, USA.
| | - Akshay N Rana
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Esther M Li
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Psychology, University of Washington, Seattle, WA 98105, USA
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Michael R Bruchas
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98105, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
31
|
Khan WU, Hussain MM, Ahmed F, Xiong H. A review of the growing trend towards heteroatoms-doped carbon dots based on dopamine acting as a hybrid agent and detected analyte. Talanta 2023; 265:124781. [PMID: 37348356 DOI: 10.1016/j.talanta.2023.124781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
Dopamine (DA) is a biomolecule that plays a critical part in the functioning of our brains by promoting motivation, maintaining focus, and altering mood. Excessive or low-level concentrations of DA in the human brain led to a dangerous neurological disorder. It is significantly important to trace the precise amount of DA to prevent such risky brain disease. Recently, heteroatoms-doped carbon dots (H-CDs) have attracted great attention for their capacity to detect biomolecules, metal ions, organic solvents, chemical dyes, etc. In this review, we have provided a comprehensive summary of the emerging trends in the heteroatom functional dopamine-doped carbon dots (DA-CDs), which are based on DA used as starting substances or functionalizing agents. Our analysis encompasses a detailed exploration of the synthetic methods, physical and chemical properties of carbon dots derived from dopamine, as well as their diverse range of applications. Additionally, we have also discussed the application of H-CDs in the dopmine detection by using various fluorescent, colorimetric, and electrochemical techniques.
Collapse
Affiliation(s)
- Waheed Ullah Khan
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China; School of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, PR China
| | | | - Farid Ahmed
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, PR China.
| |
Collapse
|
32
|
Blaess S, Krabbe S. Cell type specificity for circuit output in the midbrain dopaminergic system. Curr Opin Neurobiol 2023; 83:102811. [PMID: 37972537 DOI: 10.1016/j.conb.2023.102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/14/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Midbrain dopaminergic neurons are a relatively small group of neurons in the mammalian brain controlling a wide range of behaviors. In recent years, increasingly sophisticated tracing, imaging, transcriptomic, and machine learning approaches have provided substantial insights into the anatomical, molecular, and functional heterogeneity of dopaminergic neurons. Despite this wealth of new knowledge, it remains unclear whether and how the diverse features defining dopaminergic subclasses converge to delineate functional ensembles within the dopaminergic system. Here, we review recent studies investigating various aspects of dopaminergic heterogeneity and discuss how development, behavior, and disease influence subtype characteristics. We then outline what further approaches could be pursued to gain a more inclusive picture of dopaminergic diversity, which could be crucial to understanding the functional architecture of this system.
Collapse
Affiliation(s)
- Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Sabine Krabbe
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| |
Collapse
|
33
|
van Oostrum M, Blok TM, Giandomenico SL, Tom Dieck S, Tushev G, Fürst N, Langer JD, Schuman EM. The proteomic landscape of synaptic diversity across brain regions and cell types. Cell 2023; 186:5411-5427.e23. [PMID: 37918396 PMCID: PMC10686415 DOI: 10.1016/j.cell.2023.09.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/18/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023]
Abstract
Neurons build synaptic contacts using different protein combinations that define the specificity, function, and plasticity potential of synapses; however, the diversity of synaptic proteomes remains largely unexplored. We prepared synaptosomes from 7 different transgenic mouse lines with fluorescently labeled presynaptic terminals. Combining microdissection of 5 different brain regions with fluorescent-activated synaptosome sorting (FASS), we isolated and analyzed the proteomes of 18 different synapse types. We discovered ∼1,800 unique synapse-type-enriched proteins and allocated thousands of proteins to different types of synapses (https://syndive.org/). We identify shared synaptic protein modules and highlight the proteomic hotspots for synapse specialization. We reveal unique and common features of the striatal dopaminergic proteome and discover the proteome signatures that relate to the functional properties of different interneuron classes. This study provides a molecular systems-biology analysis of synapses and a framework to integrate proteomic information for synapse subtypes of interest with cellular or circuit-level experiments.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Thomas M Blok
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | | | | | - Georgi Tushev
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Nicole Fürst
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Julian D Langer
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
34
|
Luján MÁ, Covey DP, Young-Morrison R, Zhang L, Kim A, Morgado F, Patel S, Bass CE, Paladini C, Cheer JF. Mobilization of endocannabinoids by midbrain dopamine neurons is required for the encoding of reward prediction. Nat Commun 2023; 14:7545. [PMID: 37985770 PMCID: PMC10662422 DOI: 10.1038/s41467-023-43131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023] Open
Abstract
Brain levels of the endocannabinoid 2-arachidonoylglycerol (2-AG) shape motivated behavior and nucleus accumbens (NAc) dopamine release. However, it is not clear whether mobilization of 2-AG specifically from midbrain dopamine neurons is necessary for dopaminergic responses to external stimuli predicting forthcoming reward. Here, we use a viral-genetic strategy to prevent the expression of the 2-AG-synthesizing enzyme diacylglycerol lipase α (DGLα) from ventral tegmental area (VTA) dopamine cells in adult mice. We find that DGLα deletion from VTA dopamine neurons prevents depolarization-induced suppression of excitation (DSE), a form of 2-AG-mediated synaptic plasticity, in dopamine neurons. DGLα deletion also decreases effortful, cue-driven reward-seeking but has no effect on non-cued or low-effort operant tasks and other behaviors. Moreover, dopamine recording in the NAc reveals that deletion of DGLα impairs the transfer of accumbal dopamine signaling from a reward to its earliest predictors. These results demonstrate that 2-AG mobilization from VTA dopamine neurons is a necessary step for the generation of dopamine-based predictive associations that are required to direct and energize reward-oriented behavior.
Collapse
Affiliation(s)
- Miguel Á Luján
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dan P Covey
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, USA
| | - Reana Young-Morrison
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - LanYuan Zhang
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew Kim
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fiorella Morgado
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Caroline E Bass
- Department of Pharmacology and Toxicology, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Carlos Paladini
- UTSA Neuroscience Institute, University of Texas at San Antonio, San Antonio, TX, USA
| | - Joseph F Cheer
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
35
|
Kiani MM, Heidari Beni MH, Aghajan H. Aberrations in temporal dynamics of cognitive processing induced by Parkinson's disease and Levodopa. Sci Rep 2023; 13:20195. [PMID: 37980451 PMCID: PMC10657430 DOI: 10.1038/s41598-023-47410-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023] Open
Abstract
The motor symptoms of Parkinson's disease (PD) have been shown to significantly improve by Levodopa. However, despite the widespread adoption of Levodopa as a standard pharmaceutical drug for the treatment of PD, cognitive impairments linked to PD do not show visible improvement with Levodopa treatment. Furthermore, the neuronal and network mechanisms behind the PD-induced cognitive impairments are not clearly understood. In this work, we aim to explain these cognitive impairments, as well as the ones exacerbated by Levodopa, through examining the differential dynamic patterns of the phase-amplitude coupling (PAC) during cognitive functions. EEG data recorded in an auditory oddball task performed by a cohort consisting of controls and a group of PD patients during both on and off periods of Levodopa treatment were analyzed to derive the temporal dynamics of the PAC across the brain. We observed distinguishing patterns in the PAC dynamics, as an indicator of information binding, which can explain the slower cognitive processing associated with PD in the form of a latency in the PAC peak time. Thus, considering the high-level connections between the hippocampus, the posterior and prefrontal cortices established through the dorsal and ventral striatum acting as a modulatory system, we posit that the primary issue with cognitive impairments of PD, as well as Levodopa's cognitive deficit side effects, can be attributed to the changes in temporal dynamics of dopamine release influencing the modulatory function of the striatum.
Collapse
Affiliation(s)
- Mohammad Mahdi Kiani
- Department of Electrical Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Hamid Aghajan
- Department of Electrical Engineering, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
36
|
Rudibaugh TT, Stuppy SR, Keung AJ. Reactive Oxygen Species Mediate Transcriptional Responses to Dopamine and Cocaine in Human Cerebral Organoids. Int J Mol Sci 2023; 24:16474. [PMID: 38003664 PMCID: PMC10671319 DOI: 10.3390/ijms242216474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Dopamine signaling in the adult ventral forebrain regulates behavior, stress response, and memory formation and in neurodevelopment regulates neural differentiation and cell migration. Excessive dopamine levels, including those due to cocaine use in utero and in adults, could lead to long-term adverse consequences. The mechanisms underlying both homeostatic and pathological changes remain unclear, in part due to the diverse cellular responses elicited by dopamine and the reliance on animal models that exhibit species-specific differences in dopamine signaling. In this study, we use the human-derived ventral forebrain organoid model of Xiang-Tanaka and characterize their response to cocaine or dopamine. We explore dosing regimens of dopamine or cocaine to simulate acute or chronic exposure. We then use calcium imaging, cAMP imaging, and bulk RNA-sequencing to measure responses to cocaine or dopamine exposure. We observe an upregulation of inflammatory pathways in addition to indicators of oxidative stress following exposure. Using inhibitors of reactive oxygen species (ROS), we then show ROS to be necessary for multiple transcriptional responses of cocaine exposure. These results highlight novel response pathways and validate the potential of cerebral organoids as in vitro human models for studying complex biological processes in the brain.
Collapse
Affiliation(s)
| | | | - Albert J. Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, USA; (T.T.R.); (S.R.S.)
| |
Collapse
|
37
|
Wei H, Liu J, Wang X, Li Z, Ju L, Yao B, Zhou J, Zhao L, Zhou M, Zhang J, Yang S. Secondary metal doped cuprous-cyanoimidazole frameworks for triple-mode detection of dopamine. Anal Chim Acta 2023; 1279:341798. [PMID: 37827638 DOI: 10.1016/j.aca.2023.341798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUNDS Metal-organic framework-based nanozymes enable several opportunities for designing novel analysis methods for the detection of pesticides, heavy metal ions, and biomolecules; however, practical applications are still limited by a complicated synthesis route, lower catalytic activity, and single detection mode. Dopamine (DA) is a crucial catecholamine substance in the human body that acts as a neurotransmitter regulating a variety of physiological functions of the central nervous system. Therefore, it is highly significant to explore simple nanozymes synthesis methods for constructing a multiple analysis system to detection DA. RESULTS Herein, we elaborately selected cobalt ions as the secondary metal doping in cuprous-cyanoimidazole frameworks (CuCo-CIFs) with a mass-production strategy. CuCo-CIFs possess intrinsic peroxidase-like activity that can convert hydrogen peroxide into various reactive oxygen species (i.e., 1O2, OH·, O2·-) and thereby oxidize colorless 3,3',5,5'-tetramethylbenzidine (TMB) and DA to blue oxTMB and orange polydopamine (PDA), respectively. The absorption of the detection system increases at 460 nm while decreases at 652 nm as the concentration of DA increases under near-neutral pH (6.1), resulting in a color transition from blue to orange. Consequently, an unprecedented triple-mode analysis system of DA monitored by naked eyes, ratiometric-absorption, and scanometric was constructed. The limit of detection for the ratiometric-absorption and scanometric mode can reach 20 nM and 28 nM, respectively. CuCo-CIFs were successfully used for the rapid and accurate detection of DA in practical samples. SIGNIFICANCE As a simple, low-cost, multi-mode colorimetric platform, this kind of nanozyme detection with peroxidase-like activity exhibits significant potential for the detection of DA. Our work not only expands the applications of MOFs in analytical fields but also addresses the general challenges faced by nanozyme-based colorimetric detection systems of DA. This work provides valuable insights for the rational application of nanozyme and the design of new analysis systems.
Collapse
Affiliation(s)
- Hua Wei
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Jian Liu
- Institute of Advanced Materials, Jiangxi Normal University, Nanchang, 330022, China.
| | - Xin Wang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Zihan Li
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Lijuan Ju
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Boxuan Yao
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Jiarui Zhou
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Lei Zhao
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Mingyang Zhou
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Jie Zhang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Shenghong Yang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| |
Collapse
|
38
|
Mastwal S, Li X, Stowell R, Manion M, Zhang W, Kim NS, Yoon KJ, Song H, Ming GL, Wang KH. Adolescent neurostimulation of dopamine circuit reverses genetic deficits in frontal cortex function. eLife 2023; 12:RP87414. [PMID: 37830916 PMCID: PMC10575630 DOI: 10.7554/elife.87414] [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] [Indexed: 10/14/2023] Open
Abstract
Dopamine system dysfunction is implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive and novel paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in Arc or Disc1 to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc-/- mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in Disc1+/- mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Xinjian Li
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Matthew Manion
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| | - Nam-Shik Kim
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Ki-Jun Yoon
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Guo-Li Ming
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental HealthBethesdaUnited States
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical CenterRochesterUnited States
| |
Collapse
|
39
|
Rourk C. Comment on Albantakis et al. Computing the Integrated Information of a Quantum Mechanism. Entropy 2023, 25, 449. ENTROPY (BASEL, SWITZERLAND) 2023; 25:1436. [PMID: 37895557 PMCID: PMC10606108 DOI: 10.3390/e25101436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/20/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023]
Abstract
Integrated information theory (IIT) is a powerful tool that provides a framework for evaluating consciousness, whether in the human brain or in other systems. In Computing the Integrated Information of a Quantum Mechanism, the authors extend IIT from digital gates to a quantum CNOT logic gate, and while they explicitly distinguish the analysis from quantum theories of consciousness, they nonetheless provide an analytical road map for extending IIT not only to other quantum mechanisms but also to hybrid computing structures like the brain. This comment provides additional information relating to an adiabatic quantum mechanical energy routing mechanism that is part of a hybrid biological computer that provides an action selection mechanism, which has been hypothesized to exist in the human brain and for which predicted evidence has been subsequently observed, and it hopes to motivate the further evaluation and extension of IIT not only to that hypothesized mechanism but also to other hybrid biological computers.
Collapse
|
40
|
Srivastav S, Cui X, Varela RB, Kesby JP, Eyles D. Increasing dopamine synthesis in nigrostriatal circuits increases phasic dopamine release and alters dorsal striatal connectivity: implications for schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:69. [PMID: 37798312 PMCID: PMC10556015 DOI: 10.1038/s41537-023-00397-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023]
Abstract
One of the most robust neurochemical abnormalities reported in patients with schizophrenia is an increase in dopamine (DA) synthesis and release, restricted to the dorsal striatum (DS). This hyper functionality is strongly associated with psychotic symptoms and progresses in those who later transition to schizophrenia. To understand the implications of this progressive neurobiology on brain function, we have developed a model in rats which we refer to as EDiPs (Enhanced Dopamine in Prodromal schizophrenia). The EDiPs model features a virally mediated increase in dorsal striatal (DS) DA synthesis capacity across puberty and into adulthood. This protocol leads to progressive changes in behaviour and neurochemistry. Our aim in this study was to explore if increased DA synthesis capacity alters the physiology of DA release and DS connectivity. Using fast scan cyclic voltammetry to assess DA release we show that evoked/phasic DA release is increased in the DS of EDiPs rats, whereas tonic/background levels of DA remain unaffected. Using quantitative immunohistochemistry methods to quantify DS synaptic architecture we show a presynaptic marker for DA release sites (Bassoon) was elevated within TH axons specifically within the DS, consistent with the increased phasic DA release in this region. Alongside changes in DA systems, we also show increased density of vesicular glutamate transporter 1 (VGluT1) synapses in the EDiPs DS suggesting changes in cortical connectivity. Our data may prove relevant in understanding the long-term implications for DS function in response to the robust and prolonged increases in DA synthesis uptake and release reported in schizophrenia.
Collapse
Affiliation(s)
- Sunil Srivastav
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Queensland Centre for Mental Health Research, Brisbane, QLD, Australia
| | | | - James P Kesby
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Queensland Centre for Mental Health Research, Brisbane, QLD, Australia
| | - Darryl Eyles
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
- Queensland Centre for Mental Health Research, Brisbane, QLD, Australia.
| |
Collapse
|
41
|
Salinas AG, Lee JO, Augustin SM, Zhang S, Patriarchi T, Tian L, Morales M, Mateo Y, Lovinger DM. Distinct sub-second dopamine signaling in dorsolateral striatum measured by a genetically-encoded fluorescent sensor. Nat Commun 2023; 14:5915. [PMID: 37739964 PMCID: PMC10517008 DOI: 10.1038/s41467-023-41581-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 09/06/2023] [Indexed: 09/24/2023] Open
Abstract
The development of genetically encoded dopamine sensors such as dLight has provided a new approach to measuring slow and fast dopamine dynamics both in brain slices and in vivo, possibly enabling dopamine measurements in areas like the dorsolateral striatum (DLS) where previously such recordings with fast-scan cyclic voltammetry (FSCV) were difficult. To test this, we first evaluated dLight photometry in mouse brain slices with simultaneous FSCV and found that both techniques yielded comparable results, but notable differences in responses to dopamine transporter inhibitors, including cocaine. We then used in vivo fiber photometry with dLight in mice to examine responses to cocaine in DLS. We also compared dopamine responses during Pavlovian conditioning across the striatum. We show that dopamine increases were readily detectable in DLS and describe transient dopamine kinetics, as well as slowly developing signals during conditioning. Overall, our findings indicate that dLight photometry is well suited to measuring dopamine dynamics in DLS.
Collapse
Affiliation(s)
- Armando G Salinas
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA.
- Department of Bioengineering, George Mason University, Fairfax, VA, USA.
- Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.
| | - Jeong Oen Lee
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Shana M Augustin
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shiliang Zhang
- Confocal and Electron Microscopy Core, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Tommaso Patriarchi
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Davis, CA, USA
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Davis, CA, USA
| | - Marisela Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Yolanda Mateo
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - David M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
42
|
Kagiampaki Z, Rohner V, Kiss C, Curreli S, Dieter A, Wilhelm M, Harada M, Duss SN, Dernic J, Bhat MA, Zhou X, Ravotto L, Ziebarth T, Wasielewski LM, Sönmez L, Benke D, Weber B, Bohacek J, Reiner A, Wiegert JS, Fellin T, Patriarchi T. Sensitive multicolor indicators for monitoring norepinephrine in vivo. Nat Methods 2023; 20:1426-1436. [PMID: 37474807 PMCID: PMC7615053 DOI: 10.1038/s41592-023-01959-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 06/16/2023] [Indexed: 07/22/2023]
Abstract
Genetically encoded indicators engineered from G-protein-coupled receptors are important tools that enable high-resolution in vivo neuromodulator imaging. Here, we introduce a family of sensitive multicolor norepinephrine (NE) indicators, which includes nLightG (green) and nLightR (red). These tools report endogenous NE release in vitro, ex vivo and in vivo with improved sensitivity, ligand selectivity and kinetics, as well as a distinct pharmacological profile compared with previous state-of-the-art GRABNE indicators. Using in vivo multisite fiber photometry recordings of nLightG, we could simultaneously monitor optogenetically evoked NE release in the mouse locus coeruleus and hippocampus. Two-photon imaging of nLightG revealed locomotion and reward-related NE transients in the dorsal CA1 area of the hippocampus. Thus, the sensitive NE indicators introduced here represent an important addition to the current repertoire of indicators and provide the means for a thorough investigation of the NE system.
Collapse
Affiliation(s)
| | - Valentin Rohner
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Cedric Kiss
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Sebastiano Curreli
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Alexander Dieter
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Masaya Harada
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Sian N Duss
- Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Jan Dernic
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Musadiq A Bhat
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Xuehan Zhou
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Tim Ziebarth
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Laura Moreno Wasielewski
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Latife Sönmez
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Johannes Bohacek
- Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Andreas Reiner
- Cellular Neurobiology, Department of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - J Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
43
|
Krok AC, Maltese M, Mistry P, Miao X, Li Y, Tritsch NX. Intrinsic dopamine and acetylcholine dynamics in the striatum of mice. Nature 2023; 621:543-549. [PMID: 37558873 PMCID: PMC11577287 DOI: 10.1038/s41586-023-05995-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/22/2023] [Indexed: 08/11/2023]
Abstract
External rewards such as food and money are potent modifiers of behaviour1,2. Pioneering studies established that these salient sensory stimuli briefly interrupt the tonic discharge of neurons that produce the neuromodulators dopamine (DA) and acetylcholine (ACh): midbrain DA neurons (DANs) fire a burst of action potentials that broadly elevates DA in the striatum3,4 at the same time that striatal cholinergic interneurons (CINs) produce a characteristic pause in firing5,6. These phasic responses are thought to create unique, temporally limited conditions that motivate action and promote learning7-11. However, the dynamics of DA and ACh outside explicitly rewarded situations remain poorly understood. Here we show that extracellular DA and ACh levels fluctuate spontaneously and periodically at a frequency of approximately 2 Hz in the dorsal striatum of mice and maintain the same temporal relationship relative to one another as that evoked by reward. We show that this neuromodulatory coordination does not arise from direct interactions between DA and ACh within the striatum. Instead, we provide evidence that periodic fluctuations in striatal DA are inherited from midbrain DANs, while striatal ACh transients are driven by glutamatergic inputs, which act to locally synchronize the spiking of CINs. Together, our findings show that striatal neuromodulatory dynamics are autonomously organized by distributed extra-striatal afferents. The dominance of intrinsic rhythms in DA and ACh offers new insights for explaining how reward-associated neural dynamics emerge and how the brain motivates action and promotes learning from within.
Collapse
Affiliation(s)
- Anne C Krok
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Marta Maltese
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Pratik Mistry
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Xiaolei Miao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
44
|
Tam RW, Keung AJ. Profiling transcriptomic responses of human stem cell-derived medium spiny neuron-like cells to exogenous phasic and tonic neurotransmitters. Mol Cell Neurosci 2023; 126:103876. [PMID: 37385515 PMCID: PMC10528483 DOI: 10.1016/j.mcn.2023.103876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/06/2023] [Accepted: 06/24/2023] [Indexed: 07/01/2023] Open
Abstract
Transcriptomic responses to neurotransmitters contribute to the complex processes driving memory and addiction. Advances in both measurement methods and experimental models continue to improve our understanding of this regulatory layer. Here we focus on the experimental potential of stem cell derived neurons, currently the only ethical model that can be used in reductionist and experimentally perturbable studies of human cells. Prior work has focused on generating distinct cell types from human stem cells, and has also shown their utility in modeling development and cellular phenotypes related to neurodegeneration. Here we seek an understanding of how stem cell derived neural cultures respond to perturbations experienced during development and disease progression. This work profiles transcriptomic responses of human medium spiny neuron-like cells with three specific goals. We first characterize transcriptomic responses to dopamine and dopamine receptor agonists and antagonists presented in dosing patterns mimicking acute, chronic, and withdrawal regimens. We also assess transcriptomic responses to low and persistent tonic levels of dopamine, acetylcholine, and glutamate to better mimic the in vivo environment. Finally, we identify similar and distinct responses between hMSN-like cells derived from H9 and H1 stem cell lines, providing some context for the extent of variability these types of systems will likely pose for experimentalists. The results here suggest future optimizations of human stem cell derived neurons to increase their in vivo relevance and the biological insights that can be garnered from these models.
Collapse
Affiliation(s)
- Ryan W Tam
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, United States of America
| | - Albert J Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, United States of America.
| |
Collapse
|
45
|
Shrestha P, Kand D, Weinstain R, Winter AH. meso-Methyl BODIPY Photocages: Mechanisms, Photochemical Properties, and Applications. J Am Chem Soc 2023; 145:17497-17514. [PMID: 37535757 DOI: 10.1021/jacs.3c01682] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
meso-methyl BODIPY photocages have recently emerged as an exciting new class of photoremovable protecting groups (PPGs) that release leaving groups upon absorption of visible to near-infrared light. In this Perspective, we summarize the development of these PPGs and highlight their critical photochemical properties and applications. We discuss the absorption properties of the BODIPY PPGs, structure-photoreactivity studies, insights into the photoreaction mechanism, the scope of functional groups that can be caged, the chemical synthesis of these structures, and how substituents can alter the water solubility of the PPG and direct the PPG into specific subcellular compartments. Applications that exploit the unique optical and photochemical properties of BODIPY PPGs are also discussed, from wavelength-selective photoactivation to biological studies to photoresponsive organic materials and photomedicine.
Collapse
Affiliation(s)
- Pradeep Shrestha
- Department of Chemistry, Iowa State University, Ames, Iowa 50010, United States
| | - Dnyaneshwar Kand
- School of Plant Sciences and Food Security, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Roy Weinstain
- School of Plant Sciences and Food Security, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Arthur H Winter
- Department of Chemistry, Iowa State University, Ames, Iowa 50010, United States
| |
Collapse
|
46
|
Wärnberg E, Kumar A. Feasibility of dopamine as a vector-valued feedback signal in the basal ganglia. Proc Natl Acad Sci U S A 2023; 120:e2221994120. [PMID: 37527344 PMCID: PMC10410740 DOI: 10.1073/pnas.2221994120] [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: 12/29/2022] [Accepted: 06/08/2023] [Indexed: 08/03/2023] Open
Abstract
It is well established that midbrain dopaminergic neurons support reinforcement learning (RL) in the basal ganglia by transmitting a reward prediction error (RPE) to the striatum. In particular, different computational models and experiments have shown that a striatum-wide RPE signal can support RL over a small discrete set of actions (e.g., no/no-go, choose left/right). However, there is accumulating evidence that the basal ganglia functions not as a selector between predefined actions but rather as a dynamical system with graded, continuous outputs. To reconcile this view with RL, there is a need to explain how dopamine could support learning of continuous outputs, rather than discrete action values. Inspired by the recent observations that besides RPE, the firing rates of midbrain dopaminergic neurons correlate with motor and cognitive variables, we propose a model in which dopamine signal in the striatum carries a vector-valued error feedback signal (a loss gradient) instead of a homogeneous scalar error (a loss). We implement a local, "three-factor" corticostriatal plasticity rule involving the presynaptic firing rate, a postsynaptic factor, and the unique dopamine concentration perceived by each striatal neuron. With this learning rule, we show that such a vector-valued feedback signal results in an increased capacity to learn a multidimensional series of real-valued outputs. Crucially, we demonstrate that this plasticity rule does not require precise nigrostriatal synapses but remains compatible with experimental observations of random placement of varicosities and diffuse volume transmission of dopamine.
Collapse
Affiliation(s)
- Emil Wärnberg
- Department of Neuroscience, Karolinska Institutet, 171 77Stockholm, Sweden
- Division of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 114 28Stockholm, Sweden
| | - Arvind Kumar
- Division of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 114 28Stockholm, Sweden
| |
Collapse
|
47
|
Delignat-Lavaud B, Kano J, Ducrot C, Massé I, Mukherjee S, Giguère N, Moquin L, Lévesque C, Burke S, Denis R, Bourque MJ, Tchung A, Rosa-Neto P, Lévesque D, De Beaumont L, Trudeau LÉ. Synaptotagmin-1-dependent phasic axonal dopamine release is dispensable for basic motor behaviors in mice. Nat Commun 2023; 14:4120. [PMID: 37433762 PMCID: PMC10336101 DOI: 10.1038/s41467-023-39805-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 06/27/2023] [Indexed: 07/13/2023] Open
Abstract
In Parkinson's disease (PD), motor dysfunctions only become apparent after extensive loss of DA innervation. This resilience has been hypothesized to be due to the ability of many motor behaviors to be sustained through a diffuse basal tone of DA; but experimental evidence for this is limited. Here we show that conditional deletion of the calcium sensor synaptotagmin-1 (Syt1) in DA neurons (Syt1 cKODA mice) abrogates most activity-dependent axonal DA release in the striatum and mesencephalon, leaving somatodendritic (STD) DA release intact. Strikingly, Syt1 cKODA mice showed intact performance in multiple unconditioned DA-dependent motor tasks and even in a task evaluating conditioned motivation for food. Considering that basal extracellular DA levels in the striatum were unchanged, our findings suggest that activity-dependent DA release is dispensable for such tasks and that they can be sustained by a basal tone of extracellular DA. Taken together, our findings reveal the striking resilience of DA-dependent motor functions in the context of a near-abolition of phasic DA release, shedding new light on why extensive loss of DA innervation is required to reveal motor dysfunctions in PD.
Collapse
Affiliation(s)
- Benoît Delignat-Lavaud
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Jana Kano
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Charles Ducrot
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Ian Massé
- Hôpital du Sacré-Cœur-de-Montréal, CIUSSS NIM, Université de Montréal, Montreal, QC, Canada
| | - Sriparna Mukherjee
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Nicolas Giguère
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Luc Moquin
- Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | | | - Samuel Burke
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Raphaëlle Denis
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Marie-Josée Bourque
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Alex Tchung
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada
| | - Pedro Rosa-Neto
- Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Daniel Lévesque
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Louis De Beaumont
- Hôpital du Sacré-Cœur-de-Montréal, CIUSSS NIM, Université de Montréal, Montreal, QC, Canada
| | - Louis-Éric Trudeau
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
- SNC and CIRCA Research Groups, Université de Montréal, Montréal, QC, Canada.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
48
|
Abstract
Striosomes form neurochemically specialized compartments of the striatum embedded in a large matrix made up of modules called matrisomes. Striosome-matrix architecture is multiplexed with the canonical direct-indirect organization of the striatum. Striosomal functions remain to be fully clarified, but key information is emerging. First, striosomes powerfully innervate nigral dopamine-containing neurons and can completely shut down their activity, with a following rebound excitation. Second, striosomes receive limbic and cognition-related corticostriatal afferents and are dynamically modulated in relation to value-based actions. Third, striosomes are spatially interspersed among matrisomes and interneurons and are influenced by local and global neuromodulatory and oscillatory activities. Fourth, striosomes tune engagement and the motivation to perform reinforcement learning, to manifest stereotypical behaviors, and to navigate valence conflicts and valence discriminations. We suggest that, at an algorithmic level, striosomes could serve as distributed scaffolds to provide formats of the striatal computations generated through development and refined through learning. We propose that striosomes affect subjective states. By transforming corticothalamic and other inputs to the functional formats of the striatum, they could implement state transitions in nigro-striato-nigral circuits to affect bodily and cognitive actions according to internal motives whose functions are compromised in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
49
|
Sharp BM, Jiang Q, Kim P, Chen H. Inactivation of phosphodiesterase-4B gene in rat nucleus accumbens shell by CRISPR/Cas9 modulates the motivation to chronically self-administer nicotine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531588. [PMID: 37461457 PMCID: PMC10349965 DOI: 10.1101/2023.03.07.531588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Large scale human genome wide association studies (GWAS) have identified a growing pool of genes associated with cigarette smoking. One of the most prominent, phosphodiesterase-4B (PDE4B), has been associated with multiple smoking phenotypes. Although PDE4B modulates the half-life of neuronal cAMP, its precise role in smoking behaviors is unknown. To address this knowledge gap, we used a reverse translational approach. We inactivated PDE4B in bilateral medial nucleus accumbens shell (NAcs) neurons by injecting AAV containing a specific gRNA in female transgenic Cas9+ Long Evans rats. These rats then were given 23-hour chronic access to nicotine intravenous self-administration (IVSA) under a schedule of increasing fixed ratios (FR). With the increased effort required at FR7, nicotine SA (i.e. active presses and drug infusions) declined significantly in controls, whereas it was maintained in the mutagenized group. A progressive ratio (PR) study also showed significantly greater cumulative nicotine infusions in the mutant group. Hence, we hypothesized that enhanced PDE4B protein activity would reduce nicotine IVSA. A positive allosteric modulator,2-(3-(4-chloro-3-fluorophenyl)-5-ethyl-1H-1,2,4-triazol-1-yl)-N-(3,5-dichlorobenzyl)acetamide (MR-L2), was microinfused into NAcs bilaterally at FR3 or FR5; in both cohorts, MR-L2 acutely reduced nicotine IVSA. In summary, these studies show that the activity of PDE4B regulates the capacity of NAcs to maintain nicotine IVSA in face of the cost of increasing work. This finding and the results of the PR study indicate that PDE4B affects the motivation to obtain nicotine. These reverse translational studies in rats provide insight into the motivational effects of NAcs PDE4B that advance our understanding of the smoking behaviors mapped in human GWAS.
Collapse
Affiliation(s)
- Burt M Sharp
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Qin Jiang
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Panjun Kim
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
50
|
Zheng Y, Li Y. Past, present, and future of tools for dopamine detection. Neuroscience 2023:S0306-4522(23)00295-6. [PMID: 37419404 DOI: 10.1016/j.neuroscience.2023.06.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Dopamine (DA) is a critical neuromodulator involved in various brain functions. To understand how DA regulates neural circuits and behaviors in the physiological and pathological conditions, it is essential to have tools that enable the direct detection of DA dynamics in vivo. Recently, genetically encoded DA sensors based on G protein-coupled receptors revolutionized this field, as it allows us to track in vivo DA dynamic with unprecedented spatial-temporal resolution, high molecular specificity, and sub-second kinetics. In this review, we first summarize traditional DA detection methods. Then we focus on the development of genetically encoded DA sensors and feature its significance to understanding dopaminergic neuromodulation across diverse behaviors and species. Finally, we present our perspectives about the future direction of the next-generation DA sensors and extend their potential applications. Overall, this review offers a comprehensive perspective on the past, present, and future of DA detection tools, with important implications for the study of DA functions in health and disease.
Collapse
Affiliation(s)
- Yu Zheng
- Peking-Tsinghua Center for Life Sciences, 100871 Beijing, China
| | - Yulong Li
- Peking-Tsinghua Center for Life Sciences, 100871 Beijing, China; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871 Beijing, China; PKU-IDG/McGovern Institute for Brain Research, 100871 Beijing, China; National Biomedical Imaging Center, Peking University, 100871 Beijing, China.
| |
Collapse
|