1
|
Srinivasan S, Limani F, Hanzlova M, La Batide-Alanore S, Klotz S, Hnasko TS, Steinkellner T. Evidence for low affinity of GABA at the vesicular monoamine transporter VMAT2 - Implications for transmitter co-release from dopamine neurons. Neuropharmacology 2025; 270:110367. [PMID: 39961377 DOI: 10.1016/j.neuropharm.2025.110367] [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: 01/07/2025] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
Midbrain dopamine (DA) neurons comprise a heterogeneous population of cells. For instance, some DA neurons express the vesicular glutamate transporter VGLUT2 allowing these cells to co-release DA and glutamate. Additionally, GABA may be co-released from DA neurons. However, most cells do not express the canonical machinery to synthesize GABA or the vesicular GABA transporter VGAT. Instead, GABA seems to be taken up into DA neurons by a plasmalemmal GABA transporter (GAT1) and stored in synaptic vesicles via the vesicular monoamine transporter VMAT2. Yet, it remains unclear whether GABA indeed interacts with VMAT2. Here, we used radiotracer flux measurements in VMAT2 expressing HEK-293 cells and synaptic vesicles from male and female mice to determine whether GABA qualifies as substrate at VMAT2. We found that GABA reduced uptake of VMAT2 substrates in mouse synaptic vesicle preparations from striatum and cerebellum at millimolar concentrations but had no effect in VMAT2-expressing HEK-293 cells. Interestingly, while the closely related amino acid glycine did not affect substrate uptake at VMAT2 in mouse synaptic vesicles, the amino sulfonic acid taurine reduced uptake similar to GABA. Lastly, we discovered that the majority of mouse and human midbrain DA neurons in the substantia nigra of either sex expressed VMAT2 and GAT1 suggesting that most of them could be capable of co-releasing DA and GABA. Together, our findings suggest that GABA is a low-affinity substrate at VMAT2 with potential implications for basal ganglia physiology and disease.
Collapse
Affiliation(s)
- Sivakumar Srinivasan
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Fabian Limani
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Michaela Hanzlova
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Ségolène La Batide-Alanore
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | - Sigrid Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Austria
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Department of Veterans Affairs, San Diego Veterans Affairs Healthcare System, San Diego, CA, USA
| | - Thomas Steinkellner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
2
|
Xiao Y, Liu C, Wang X, Li H, Wang L, Gou K, Liu X, Guan X, Zhou X, He X, Zhao Y, Tao L, Pan X, Jiang L, Chen Y, Liu H, Dai Y, Bu Q, Qin M, Zhu R, Chen B, Flores AD, Zhao Y, Cen X. Dysregulated glycerophospholipid metabolism in amygdala may mediate favipiravir-induced anxiety-like behaviors in mice. Front Pharmacol 2025; 16:1491150. [PMID: 40103591 PMCID: PMC11913839 DOI: 10.3389/fphar.2025.1491150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/20/2025] [Indexed: 03/20/2025] Open
Abstract
Favipiravir, the first RNA polymerase inhibitor approved to treat resistant influenza, has been reported to be associated with central nervous system (CNS) side effects, particularly anxiety-like behavior; nevertheless, the underlying mechanism remains largely unknown. In this study, we investigated the effect of favipiravir on the neurobehavior of mice, and combined lipidomics and transcriptomics analysis to explore the mechanism underlying this effect. In behavioral tests, the mice displayed anxiety-like behaviors after oral favipiravir administration (200 mg/kg) for 7 days continuously. By lipidomics analysis, we observed that favipiravir induced a dysregulation of glycerophospholipid metabolism in the amygdala. Moreover, favipiravir significantly reduced the mRNA level of glycerol-3-phosphate acyltransferase 2 (Gpat2), the rate-limiting enzyme of glycerophospholipid synthesis. Notably, favipiravir markedly reduced the levels of docosahexaenoic acid-enriched phosphatidylethanolamine or phosphatidylcholine (DHA-PE/PC) and arachidonic acid-enriched phosphatidylethanolamine or phosphatidylcholine (AA-PE/PC), two components of glycerophospholipids, in the amygdala. The increased expression of phospholipase A2 (Pla2) may attribute to the enhanced release of arachidonic acid (AA) from AA-PE/PC. Furthermore, favipiravir altered neurite morphology and reduced neurophysiological activity in amygdala neurons in vitro. Collectively, dysregulated glycerophospholipid metabolism in the amygdala may contribute to the adverse effect of favipiravir.
Collapse
Affiliation(s)
- Yuzhou Xiao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunqi Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojie Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Gou
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xingchen Liu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xinqi Guan
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhou
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiumei He
- School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Tao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaodan Pan
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiming Zhu
- Chengdu Westchina Frontier Pharmatech, Co., Ltd., Chengdu, China
| | - Bo Chen
- Chengdu Westchina Frontier Pharmatech, Co., Ltd., Chengdu, China
| | - Angelo D Flores
- Department of Neuroscience, City University of Hong Kong, Kowloon, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Boumhaouad S, Makowicz EA, Choi S, Bouhaddou N, Balla J, Taghzouti K, Sulzer D, Mosharov EV. Regulation of Dopamine Release by Tonic Activity Patterns in the Striatal Brain Slice. ACS Chem Neurosci 2025; 16:303-310. [PMID: 39798080 PMCID: PMC11804867 DOI: 10.1021/acschemneuro.4c00323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Voluntary movement, motivation, and reinforcement learning depend on the activity of ventral midbrain neurons, which extend axons to release dopamine (DA) in the striatum. These neurons exhibit two patterns of action potential activity: low-frequency tonic activity that is intrinsically generated and superimposed high-frequency phasic bursts that are driven by synaptic inputs. Ex vivo acute striatal brain preparations are widely employed to study the regulation of evoked DA release but exhibit very different DA release kinetics than in vivo recordings. To investigate the relationship between phasic and tonic neuronal activity, we stimulated the slice in patterns intended to mimic tonic activity, which were interrupted by a series of burst stimuli. Conditioning the striatal slice with low-frequency activity altered DA release triggered by high-frequency bursts and produced kinetic parameters that resemble those in vivo. In the absence of applied tonic activity, nicotinic acetylcholine receptor and D2 DA receptor antagonists had no significant effect on neurotransmitter release, driven by repeated burst activity in the striatal brain slice. In contrast, in tonically stimulated slices, the D2 receptor blockade decreased the amount of DA released during a single-burst and facilitated DA release in subsequent bursts. This experimental system provides a means to reconcile the difference in the kinetics of DA release ex vivo and in vivo and provides a novel approach to more accurately emulate pre- and postsynaptic mechanisms that control axonal DA release in vivo.
Collapse
Affiliation(s)
- Siham Boumhaouad
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - Emily A Makowicz
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| | - Sejoon Choi
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| | - Nezha Bouhaddou
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - Jihane Balla
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - Khalid Taghzouti
- Physiology
and Physiopathology Team, Genomics of Human Pathologies Research Center,
Faculty of Sciences, Mohammed V University
in Rabat, Rabat 10000, Morocco
| | - David Sulzer
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| | - Eugene V. Mosharov
- Departments
of Psychiatry and Neurology, Division of Molecular Therapeutics, New
York State Psychiatric Institute, Columbia
University Medical Center, New York, New York 10032, United States
| |
Collapse
|
4
|
Flores-García M, Flores Á, Aso E, Otero-López P, Ciruela F, Videla S, Grau-Sánchez J, Rodríguez-Fornells A, Bonaventura J, Fernández-Dueñas V. Dopamine dynamics in chronic pain: music-induced, sex-dependent, behavioral effects in mice. Pain Rep 2025; 10:e1205. [PMID: 39664710 PMCID: PMC11631031 DOI: 10.1097/pr9.0000000000001205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction Chronic pain is a debilitating disease that is usually comorbid to anxiety and depression. Current treatment approaches mainly rely on analgesics but often neglect emotional aspects. Nonpharmacological interventions, such as listening to music, have been incorporated into clinics to provide a more comprehensive management of chronic pain. However, the underlying mechanisms of music-mediated pain relief are not fully understood. Objectives Our aim was to evaluate the effects and mechanisms of music exposure in an animal model of chronic pain. Methods We injected mice with the complete Freund adjuvant (CFA) inflammatory agent into the hind paw and housed them for 14 days with background music, or ambient noise, during their active period (Mozart K.205, overnight). The effect of music exposure on nociception, anxiety-like behaviors, and depression-like behaviors was evaluated through different paradigms, including the hot plate, Von Frey, elevated plus maze, splash, and tail suspension tests. In addition, we conducted fiber photometry experiments to investigate whether music influences dopamine dynamics in the nucleus accumbens (NAcc), a crucial region involved in pain processing, anhedonia, and reward. Results Our findings indicate that music exposure prevents the decrease in NAcc activity observed in CFA-injected mice, linking with a sex-dependent reduction in allodynia, anxiety-like behaviors, and depression-like behaviors. Accordingly, female mice were more sensitive to music exposure than male mice. Conclusion Collectively, our findings provide compelling evidence for the integration of music as a nonpharmacological intervention in chronic pain conditions. Moreover, the observed effect on NAcc suggests its potential as a therapeutic target for addressing chronic pain and its associated symptoms.
Collapse
Affiliation(s)
- Montse Flores-García
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - Paloma Otero-López
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - Sebastià Videla
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - Jennifer Grau-Sánchez
- Research Group on Complex Health Diagnoses and Interventions from Occupation and Care (OCCARE), University School of Nursing and Occupational Therapy of Terrassa, Autonomous University of Barcelona, Terrassa, Spain
| | - Antoni Rodríguez-Fornells
- Cognition and Brain Plasticity Unit, Department of Cognition, Development and Educational Psychology, Faculty of Psychology, University of Barcelona and Bellvitge Institute for Biomedical Research, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Jordi Bonaventura
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| | - Víctor Fernández-Dueñas
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, IDIBELL-Bellvitge Institute for Biomedical Research, Barcelona, Spain
| |
Collapse
|
5
|
Lopez GC, Lerner TN. How Dopamine Enables Learning from Aversion. Curr Opin Behav Sci 2025; 61:101476. [PMID: 39719969 PMCID: PMC11666190 DOI: 10.1016/j.cobeha.2024.101476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Dopamine is heavily studied for its role in reward learning, but it is becoming increasingly appreciated that dopamine can also enable learning from aversion. Dopamine neurons modulate their firing and neurotransmitter release patterns in response to aversive outcomes. However, there is considerable heterogeneity in the timing and directionality of the modulation. Open questions remain as to the factors that determine this heterogeneity and how varying patterns of responses to aversion in different dopamine-receptive brain regions contribute to value learning, decision-making, and avoidance. Here, we review recent progress in this area and highlight important future directions.
Collapse
Affiliation(s)
- Gabriela C. Lopez
- Feinberg School of Medicine, Department of Neuroscience, Northwestern University, Chicago, IL, USA
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA
| | - Talia N. Lerner
- Feinberg School of Medicine, Department of Neuroscience, Northwestern University, Chicago, IL, USA
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA
| |
Collapse
|
6
|
Buck SA, Mabry SJ, Glausier JR, Banks-Tibbs T, Ward C, Kozel J, Fu C, Fish KN, Lewis DA, Logan RW, Freyberg Z. Aging disrupts the coordination between mRNA and protein expression in mouse and human midbrain. Mol Psychiatry 2025:10.1038/s41380-025-02909-1. [PMID: 39875589 DOI: 10.1038/s41380-025-02909-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/27/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Age-related dopamine (DA) neuron loss is a primary feature of Parkinson's disease. However, whether similar biological processes occur during healthy aging, but to a lesser degree, remains unclear. We therefore determined whether midbrain DA neurons degenerate during aging in mice and humans. In mice, we identified no difference in midbrain neuron numbers throughout aging. Despite this, we found age-related decreases in midbrain mRNA expression of tyrosine hydroxylase (Th), the rate limiting enzyme of DA synthesis. Among midbrain glutamatergic cells, we similarly identified age-related declines in vesicular glutamate transporter 2 (Vglut2) mRNA expression. In co-transmitting Th+/Vglut2+ neurons, Th and Vglut2 transcripts decreased with aging. However, Th and Vglut2 protein levels in striatal synaptic release sites (e.g., terminals and axonal projections) did not differ throughout aging. Similar to the mouse, an initial study of human brain showed no effect of aging on midbrain neuron number with a concomitant decrease in TH and VGLUT2 mRNA expression. Unlike in mice, the density of striatal TH+ dopaminergic terminals was lower in aged human subjects. However, TH and VGLUT2 protein levels were unaffected in the remaining striatal boutons. Finally, in contrast to Th and Vglut2 mRNA, expression of most ribosomal genes in Th+ neurons was either maintained or even upregulated during aging. This suggests a homeostatic mechanism where age-related declines in transcriptional efficiency are overcome by ongoing ribosomal translation. Overall, we demonstrate species-conserved transcriptional effects of aging in midbrain dopaminergic and glutamatergic neurons that are not accompanied by marked cell death or lower striatal protein expression. This opens the door to novel therapeutic approaches to maintain neurotransmission and bolster neuronal resilience.
Collapse
Affiliation(s)
- Silas A Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Samuel J Mabry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jill R Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tabitha Banks-Tibbs
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Caroline Ward
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenesis Kozel
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chen Fu
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Fujise K, Mishra J, Rosenfeld MS, Rafiq NM. Synaptic vesicle characterization of iPSC-derived dopaminergic neurons provides insight into distinct secretory vesicle pools. NPJ Parkinsons Dis 2025; 11:16. [PMID: 39788994 PMCID: PMC11718109 DOI: 10.1038/s41531-024-00862-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
The dysfunction of dopaminergic (DA) neurons is central to Parkinson's disease. Distinct synaptic vesicle (SV) populations, differing in neurotransmitter content (dopamine vs. glutamate), may vary due to differences in trafficking and exocytosis. However, the structural organization of these vesicles remains unclear. In this study, we examined axonal varicosities in human iPSC-derived DA and glutamatergic neurons (i3Neurons). i3Neurons primarily contained small, clear SVs (40-50 nm), whereas DA neurons contained larger, pleiomorphic vesicles including dense core and empty vesicles, in addition to the classical SVs. VMAT2-positive vesicles in DA neurons, which load dopamine, were spatially segregated from VGLUT1/2-positive vesicles in an SV-like reconstitution system. These vesicles also colocalized with SV markers (e.g., VAMP2, SV2C), and can be clustered by synapsin. Moreover, DA axonal terminals in mouse striata showed similar vesicle pool diversity. These findings reveal structural differences in DA neurons' vesicles, highlighting iPSC-derived neurons as effective models for studying presynaptic structures.
Collapse
Affiliation(s)
- Kenshiro Fujise
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Jaya Mishra
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | | | - Nisha Mohd Rafiq
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
8
|
Hirai H, Konno K, Yamasaki M, Watanabe M, Sakaba T, Hashimotodani Y. Distinct release properties of glutamate/GABA co-transmission serve as a frequency-dependent filtering of supramammillary inputs. eLife 2024; 13:RP99711. [PMID: 39680436 DOI: 10.7554/elife.99711] [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: 12/17/2024] Open
Abstract
Glutamate and GABA co-transmitting neurons exist in several brain regions; however, the mechanism by which these two neurotransmitters are co-released from the same synaptic terminals remains unclear. Here, we show that the supramammillary nucleus (SuM) to dentate granule cell synapses, which co-release glutamate and GABA, exhibit differences between glutamate and GABA release properties in paired-pulse ratio, Ca2+-sensitivity, presynaptic receptor modulation, and Ca2+ channel-vesicle coupling configuration. Moreover, uniquantal synaptic responses show independent glutamatergic and GABAergic responses. Morphological analysis reveals that most SuM terminals form distinct glutamatergic and GABAergic synapses in proximity, each characterized by GluN1 and GABAAα1 labeling, respectively. Notably, glutamate/GABA co-transmission exhibits distinct short-term plasticities, with frequency-dependent depression of glutamate and frequency-independent stable depression of GABA. Our findings suggest that glutamate and GABA are co-released from different synaptic vesicles within the SuM terminals, and reveal that distinct transmission modes of glutamate/GABA co-release serve as frequency-dependent filters of SuM inputs.
Collapse
Affiliation(s)
- Himawari Hirai
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | | |
Collapse
|
9
|
Ceballos CC, Ma L, Qin M, Zhong H. Widespread co-release of glutamate and GABA throughout the mouse brain. Commun Biol 2024; 7:1502. [PMID: 39537846 PMCID: PMC11560972 DOI: 10.1038/s42003-024-07198-y] [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: 06/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Several brain neuronal populations transmit both the excitatory and inhibitory neurotransmitters, glutamate, and GABA. However, it remains largely unknown whether these opposing neurotransmitters are co-released simultaneously or are independently transmitted at different times and locations. By recording from acute mouse brain slices, we observed biphasic miniature postsynaptic currents, i.e., minis with time-locked excitatory and inhibitory currents, in striatal spiny projection neurons. This observation cannot be explained by accidental coincidence of monophasic excitatory and inhibitory minis. Interestingly, these biphasic minis could either be an excitatory current leading an inhibitory current or vice versa. Deletion of dopaminergic neurons did not eliminate biphasic minis, indicating that they originate from another source. Importantly, we found that both types of biphasic minis were present in multiple striatal neuronal types and in nine out of ten other brain regions. Overall, co-release of glutamate and GABA appears to be a widespread mode of neurotransmission in the brain.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Kim DI, Kang SJ, Jhang J, Jo YS, Park S, Ye M, Pyeon GH, Im GH, Kim SG, Han S. Encoding opposing valences through frequency-dependent transmitter switching in single peptidergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.09.622790. [PMID: 39574736 PMCID: PMC11581014 DOI: 10.1101/2024.11.09.622790] [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: 11/29/2024]
Abstract
Peptidergic neurons often co-express fast transmitters and neuropeptides in separate vesicles with distinct release properties. However, the release dynamics of each transmitter in various contexts have not been fully understood in behaving animals. Here, we demonstrate that calcitonin gene-related peptide (CGRP) neurons in the external lateral subdivision of the parabrachial nucleus (CGRPPBel) encode opposing valence via differential release, rather than co-release, of glutamate and neuropeptides, according to firing rate. Glutamate is released preferentially at lower firing rates with minimal release at higher firing rates, whereas neuropeptides are released at higher firing rates, resulting in frequency-dependent switching of transmitters. Aversive stimuli evoke high frequency responses with accompanying neuropeptide release to encode negative valence, whereas appetitive stimuli evoke low frequency responses with glutamate release to encode positive valence. Our study reveals a previously unknown capability of single CGRPPBel neurons to bidirectionally encode valence via frequency-dependent differential release of transmitters in vivo.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Sukjae J. Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Yong S. Jo
- School of Psychology, Korea University; Seoul, Republic of Korea
| | - Seahyung Park
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Gyeong Hee Pyeon
- School of Psychology, Korea University; Seoul, Republic of Korea
| | - Geun-Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University; Suwon, Republic of Korea
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University; Suwon, Republic of Korea
| |
Collapse
|
11
|
Kim DI, Park S, Park S, Ye M, Chen JY, Kang SJ, Jhang J, Hunker AC, Zweifel LS, Caron KM, Vaughan JM, Saghatelian A, Palmiter RD, Han S. Presynaptic sensor and silencer of peptidergic transmission reveal neuropeptides as primary transmitters in pontine fear circuit. Cell 2024; 187:5102-5117.e16. [PMID: 39043179 PMCID: PMC11380597 DOI: 10.1016/j.cell.2024.06.035] [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: 01/13/2023] [Revised: 11/17/2023] [Accepted: 06/25/2024] [Indexed: 07/25/2024]
Abstract
Neurons produce and release neuropeptides to communicate with one another. Despite their importance in brain function, circuit-based mechanisms of peptidergic transmission are poorly understood, primarily due to the lack of tools for monitoring and manipulating neuropeptide release in vivo. Here, we report the development of two genetically encoded tools for investigating peptidergic transmission in behaving mice: a genetically encoded large dense core vesicle (LDCV) sensor that detects presynaptic neuropeptide release and a genetically encoded silencer that specifically degrades neuropeptides inside LDCVs. Using these tools, we show that neuropeptides, not glutamate, encode the unconditioned stimulus in the parabrachial-to-amygdalar threat pathway during Pavlovian threat learning. We also show that neuropeptides play important roles in encoding positive valence and suppressing conditioned threat response in the amygdala-to-parabrachial endogenous opioidergic circuit. These results show that our sensor and silencer for presynaptic peptidergic transmission are reliable tools to investigate neuropeptidergic systems in awake, behaving animals.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sekun Park
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Seahyung Park
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jane Y Chen
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sukjae J Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Avery C Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan M Vaughan
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alan Saghatelian
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon 16419, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
12
|
Zhao M, Zhou Y, Sheng R, Zhang H, Xiang J, Wang J, Li P, Ma T, Liu P, Chen Q, Wen W, Xu S. Gastrodin relieves Parkinson's disease-related motor deficits by facilitating the MEK-dependent VMAT2 to maintain dopamine homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155819. [PMID: 38885579 DOI: 10.1016/j.phymed.2024.155819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Dysfunction of dopamine homeostasis (DAH), which is regulated by vesicular monoamine transporter 2 (VMAT2), is a vital cause of dopamine (DA) neurotoxicity and motor deficits in Parkinson's disease (PD). Gastrodin (4-hydroxybenzyl alcohol 4-O-β-D-glucoside; GTD), a natural active compound derived from Gastrodia elata Blume, can be used to treat multiple neurological disorders, including PD. However, whether GTD regulates VMAT2-mediated DAH dysfunction in PD models remains unclear. PURPOSE To explore whether GTD confers dopaminergic neuroprotection by facilitating DA vesicle storage and maintaining DAH in PD models. METHODS Mice were treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and PC12 cells with 1-methyl-4-phenyl-pyridinium (MPP+) to induce PD characteristics. Multiple behavioural tests were performed to evaluate the motor functions of the mice. HPLC was used to measure DA and 3,4-dihydroxyphenylacetic acid (DOPAC) levels. Transmission electron microscopy was used to observe synaptic vesicles. Molecular docking and molecular dynamics were used to determine the binding affinity of GTD to the target protein. Reserpine (Res, a VMAT2 inhibitor) and PD0325901 (901, a MEK inhibitor) were employed to investigate the mechanism of GTD. Western blotting and immunohistochemistry were used to assess the expression of the target proteins. RESULTS GTD attenuated motor deficits and dopaminergic neuronal injury, reversed the imbalance of DAH, and increased VMAT2 levels and vesicle volume in MPTP-induced mice. GTD ameliorated cell damage, ROS release, and dysfunction of DAH in MPP+-induced PC12 cells. Moreover, the neuroprotective effects of GTD were reversed by Res in vitro and in vivo. Furthermore, GTD can activate the MEK/ERK/CREB pathway to upregulate VMAT2 in vitro and in vivo. Interestingly, 901 reversed the effects of GTD on VMAT2 and dopaminergic neuronal impairment. CONCLUSION GTD relieved PD-related motor deficits and dopaminergic neuronal impairment by facilitating MEK-depended VMAT2 to regulate DAH, which offers new insights into its therapeutic potential.
Collapse
Affiliation(s)
- Meihuan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Yongtao Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Ruilin Sheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Haijun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Junbao Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Jie Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Ping Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Tengyun Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Panwang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Qi Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China
| | - Wen Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China.
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University traditional Chinese medicine, Chengdu, 611137, PR China.
| |
Collapse
|
13
|
Asmerian H, Alberts J, Sanetra AM, Diaz AJ, Silm K. Role of adaptor protein complexes in generating functionally distinct synaptic vesicle pools. J Physiol 2024. [PMID: 39034608 DOI: 10.1113/jp286179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
The synaptic vesicle (SV) cycle ensures the release of neurotransmitters and the replenishment of SVs to sustain neuronal activity. Multiple endocytosis and sorting pathways contribute to the recapture of the SV membrane and proteins after fusion. Adaptor protein (AP) complexes are among the critical components of the SV retrieval machinery. The canonical clathrin adaptor AP2 ensures the replenishment of most SVs across many neuronal populations. An alternative AP1/AP3-dependent process mediates the formation of a subset of SVs that differ from AP2 vesicles in molecular composition and respond preferentially during higher frequency firing. Furthermore, recent studies show that vesicular transporters for different neurotransmitters depend to a different extent on the AP3 pathway and this affects the release properties of the respective neurotransmitters. This review focuses on the current understanding of the AP-dependent molecular and functional diversity among SVs. We also discuss the contribution of these pathways to the regulation of neurotransmitter release across neuronal populations.
Collapse
Affiliation(s)
- Hrach Asmerian
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jacob Alberts
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anna M Sanetra
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexia J Diaz
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Katlin Silm
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
14
|
Abela L, Gianfrancesco L, Tagliatti E, Rossignoli G, Barwick K, Zourray C, Reid KM, Budinger D, Ng J, Counsell J, Simpson A, Pearson TS, Edvardson S, Elpeleg O, Brodsky FM, Lignani G, Barral S, Kurian MA. Neurodevelopmental and synaptic defects in DNAJC6 parkinsonism, amenable to gene therapy. Brain 2024; 147:2023-2037. [PMID: 38242634 PMCID: PMC11146427 DOI: 10.1093/brain/awae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/10/2023] [Accepted: 12/16/2023] [Indexed: 01/21/2024] Open
Abstract
DNAJC6 encodes auxilin, a co-chaperone protein involved in clathrin-mediated endocytosis (CME) at the presynaptic terminal. Biallelic mutations in DNAJC6 cause a complex, early-onset neurodegenerative disorder characterized by rapidly progressive parkinsonism-dystonia in childhood. The disease is commonly associated with additional neurodevelopmental, neurological and neuropsychiatric features. Currently, there are no disease-modifying treatments for this condition, resulting in significant morbidity and risk of premature mortality. To investigate the underlying disease mechanisms in childhood-onset DNAJC6 parkinsonism, we generated induced pluripotent stem cells (iPSC) from three patients harbouring pathogenic loss-of-function DNAJC6 mutations and subsequently developed a midbrain dopaminergic neuronal model of disease. When compared to age-matched and CRISPR-corrected isogenic controls, the neuronal cell model revealed disease-specific auxilin deficiency as well as disturbance of synaptic vesicle recycling and homeostasis. We also observed neurodevelopmental dysregulation affecting ventral midbrain patterning and neuronal maturation. To explore the feasibility of a viral vector-mediated gene therapy approach, iPSC-derived neuronal cultures were treated with lentiviral DNAJC6 gene transfer, which restored auxilin expression and rescued CME. Our patient-derived neuronal model provides deeper insights into the molecular mechanisms of auxilin deficiency as well as a robust platform for the development of targeted precision therapy approaches.
Collapse
Affiliation(s)
- Lucia Abela
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Lorita Gianfrancesco
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Erica Tagliatti
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, 20089 Milano, Italy
| | - Giada Rossignoli
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Katy Barwick
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Clara Zourray
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kimberley M Reid
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Dimitri Budinger
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Joanne Ng
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Genetic Therapy Accelerator Centre, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - John Counsell
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Arlo Simpson
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Toni S Pearson
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032-3784, USA
- Department of Pediatrics, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43210, USA
- Department of Neurology, Nationwide Children’s Hospital, Ohio State University, Columbus, OH 43210, USA
| | - Simon Edvardson
- Department of Genetics, Hadassah, Hebrew University Medical Center, 9574869 Jerusalem, Israel
| | - Orly Elpeleg
- Department of Genetics, Hadassah, Hebrew University Medical Center, 9574869 Jerusalem, Israel
| | - Frances M Brodsky
- Research Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
| | - Gabriele Lignani
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Serena Barral
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
| | - Manju A Kurian
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, WC1N 1DZ, UK
- Department of Neurology, Great Ormond Street Hospital, London, WC1N 3JH, UK
| |
Collapse
|
15
|
Buck SA, Mabry SJ, Glausier JR, Banks-Tibbs T, Ward C, Kozel JG, Fu C, Fish KN, Lewis DA, Logan RW, Freyberg Z. Aging disrupts the coordination between mRNA and protein expression in mouse and human midbrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596950. [PMID: 38854057 PMCID: PMC11160743 DOI: 10.1101/2024.06.01.596950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Age-related dopamine (DA) neuron loss is a primary feature of Parkinson's disease. However, it remains unclear whether similar biological processes occur during healthy aging, albeit to a lesser degree. We therefore determined whether midbrain DA neurons degenerate during aging in mice and humans. In mice, we identified no changes in midbrain neuron numbers throughout aging. Despite this, we found age-related decreases in midbrain mRNA expression of tyrosine hydroxylase (Th), the rate limiting enzyme of DA synthesis. Among midbrain glutamatergic cells, we similarly identified age-related declines in vesicular glutamate transporter 2 (Vglut2) mRNA expression. In co-transmitting Th +/Vglut2 + neurons, Th and Vglut2 transcripts decreased with aging. Importantly, striatal Th and Vglut2 protein expression remained unchanged. In translating our findings to humans, we found no midbrain neurodegeneration during aging and identified age-related decreases in TH and VGLUT2 mRNA expression similar to mouse. Unlike mice, we discovered diminished density of striatal TH+ dopaminergic terminals in aged human subjects. However, TH and VGLUT2 protein expression were unchanged in the remaining striatal boutons. Finally, in contrast to Th and Vglut2 mRNA, expression of most ribosomal genes in Th + neurons was either maintained or even upregulated during aging. This suggests a homeostatic mechanism where age-related declines in transcriptional efficiency are overcome by ongoing ribosomal translation. Overall, we demonstrate species-conserved transcriptional effects of aging in midbrain dopaminergic and glutamatergic neurons that are not accompanied by marked cell death or lower striatal protein expression. This opens the door to novel therapeutic approaches to maintain neurotransmission and bolster neuronal resilience.
Collapse
Affiliation(s)
- Silas A. Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Samuel J. Mabry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jill R. Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tabitha Banks-Tibbs
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Caroline Ward
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenesis Gayden Kozel
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chen Fu
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kenneth N. Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Ceballos CC, Ma L, Qin M, Zhong H. Prevalent co-release of glutamate and GABA throughout the mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587069. [PMID: 38585864 PMCID: PMC10996720 DOI: 10.1101/2024.03.27.587069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Several neuronal populations in the brain transmit both the excitatory and inhibitory neurotransmitters, glutamate, and GABA, to downstream neurons. However, it remains largely unknown whether these opposing neurotransmitters are co-released onto the same postsynaptic neuron simultaneously or are independently transmitted at different time and locations (called co-transmission). Here, using whole-cell patch-clamp recording on acute mouse brain slices, we observed biphasic miniature postsynaptic currents, i.e., minis with time-locked excitatory and inhibitory currents, in striatal spiny projection neurons (SPNs). This observation cannot be explained by accidental coincidence of monophasic miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs, respectively), arguing for the co-release of glutamate and GABA. Interestingly, these biphasic minis could either be an mEPSC leading an mIPSC or vice versa. Although dopaminergic axons release both glutamate and GABA in the striatum, deletion of dopamine neurons did not eliminate biphasic minis, indicating that the co-release originates from another neuronal type. Importantly, we found that both types of biphasic minis were detected in other neuronal subtypes in the striatum as well as in nine out of ten additionally tested brain regions. Our results suggest that co-release of glutamate and GABA is a prevalent mode of neurotransmission in the brain.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
17
|
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
|
18
|
Guzikowski NJ, Kavalali ET. Synaptic vesicle pool heterogeneity drives an anomalous form of synaptic plasticity. Proc Natl Acad Sci U S A 2024; 121:e2401734121. [PMID: 38422062 PMCID: PMC10945817 DOI: 10.1073/pnas.2401734121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Affiliation(s)
- Natalie J. Guzikowski
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN37240-7933
| | - Ege T. Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN37240-7933
| |
Collapse
|
19
|
Martínez-Aguirre C, Márquez LA, Santiago-Castañeda CL, Carmona-Cruz F, Nuñez-Lumbreras MDLA, Martínez-Rojas VA, Alonso-Vanegas M, Aguado-Carrillo G, Gómez-Víquez NL, Galván EJ, Cuéllar-Herrera M, Rocha L. Cannabidiol Modifies the Glutamate Over-Release in Brain Tissue of Patients and Rats with Epilepsy: A Pilot Study. Biomedicines 2023; 11:3237. [PMID: 38137458 PMCID: PMC10741033 DOI: 10.3390/biomedicines11123237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Drug-resistant epilepsy (DRE) is associated with high extracellular levels of glutamate. Studies support the idea that cannabidiol (CBD) decreases glutamate over-release. This study focused on investigating whether CBD reduces the evoked glutamate release in cortical synaptic terminals obtained from patients with DRE as well as in a preclinical model of epilepsy. Synaptic terminals (synaptosomes) were obtained from the epileptic neocortex of patients with drug-resistant temporal lobe epilepsy (DR-TLE, n = 10) or drug-resistant extratemporal lobe epilepsy (DR-ETLE, n = 10) submitted to epilepsy surgery. Synaptosomes highly purified by Percoll-sucrose density gradient were characterized by confocal microscopy and Western blot. Synaptosomes were used to estimate the high KCl (33 mM)-evoked glutamate release in the presence of CBD at different concentrations. Our results revealed responsive tissue obtained from seven patients with DR-TLE and seven patients with DR-ETLE. Responsive tissue showed lower glutamate release (p < 0.05) when incubated with CBD at low concentrations (less than 100 µM) but not at higher concentrations. Tissue that was non-responsive to CBD (DR-TLE, n = 3 and DR-ELTE, n = 3) showed high glutamate release despite CBD exposure at different concentrations. Simultaneously, a block of the human epileptic neocortex was used to determine its viability through whole-cell and extracellular electrophysiological recordings. The electrophysiological evaluations supported that the responsive and non-responsive human epileptic neocortices used in the present study exhibited proper neuronal viability and stability to acquire electrophysiological responses. We also investigated whether the subchronic administration of CBD could reduce glutamate over-release in a preclinical model of temporal lobe epilepsy. Administration of CBD (200 mg/kg, p.o. every 24 h for 7 days) to rats with lithium-pilocarpine-evoked spontaneous recurrent seizures reduced glutamate over-release in the hippocampus. The present study revealed that acute exposure to low concentrations of CBD can reduce the glutamate over-release in synaptic terminals obtained from some patients with DRE. This effect is also evident when applied subchronically in rats with spontaneous recurrent seizures. An important finding was the identification of a group of patients that were non-responsive to CBD effects. Future studies are essential to identify biomarkers of responsiveness to CBD to control DRE.
Collapse
Affiliation(s)
- Christopher Martínez-Aguirre
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Luis Alfredo Márquez
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Cindy Lizbeth Santiago-Castañeda
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Francia Carmona-Cruz
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Maria de los Angeles Nuñez-Lumbreras
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Vladimir A. Martínez-Rojas
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
- Center for Research on Aging, Center for Research and Advanced Studies, Mexico City 14330, Mexico
| | - Mario Alonso-Vanegas
- International Center for Epilepsy Surgery, HMG-Coyoacán Hospital, Mexico City 04380, Mexico;
| | - Gustavo Aguado-Carrillo
- Clinic of Epilepsy, General Hospital of México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Norma L. Gómez-Víquez
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| | - Emilio J. Galván
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
- Center for Research on Aging, Center for Research and Advanced Studies, Mexico City 14330, Mexico
| | - Manola Cuéllar-Herrera
- Clinic of Epilepsy, General Hospital of México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City 14330, Mexico; (C.M.-A.); (L.A.M.); (C.L.S.-C.); (F.C.-C.); (M.d.l.A.N.-L.); (V.A.M.-R.); (N.L.G.-V.); (E.J.G.)
| |
Collapse
|
20
|
Barcomb K, Ford CP. Alterations in neurotransmitter co-release in Parkinson's disease. Exp Neurol 2023; 370:114562. [PMID: 37802381 PMCID: PMC10842357 DOI: 10.1016/j.expneurol.2023.114562] [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: 05/05/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Parkinson's disease is a neurological disorder characterized by degeneration of midbrain dopamine neurons, which results in numerous adaptations in basal ganglia circuits. Research over the past twenty-five years has identified that midbrain dopamine neurons of the substantia nigra pars compacta (SNc) and ventral tegmental area (VTA) co-release multiple other transmitters including glutamate and GABA, in addition to their canonical transmitter, dopamine. This review summarizes previous work characterizing neurotransmitter co-release from dopamine neurons, work examining potential changes in co-release dynamics that result in animal models of Parkinson's disease, and future opportunities for determining how dysfunction in co-release may contribute to circuit dysfunction in Parkinson's disease.
Collapse
Affiliation(s)
- Kelsey Barcomb
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
21
|
Chu F, Lu C, Jiao Z, Yang W, Yang X, Ma H, Yu H, Wang S, Li Y, Sun D, Sun H. Unveiling the LncRNA-miRNA-mRNA Regulatory Network in Arsenic-Induced Nerve Injury in Rats through High-Throughput Sequencing. TOXICS 2023; 11:953. [PMID: 38133354 PMCID: PMC10747658 DOI: 10.3390/toxics11120953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023]
Abstract
Arsenic is a natural toxin which is widely distributed in the environment, incurring diverse toxicities and health problems. Previous studies have shown that long non-coding RNAs (LncRNAs) are also reported to contribute to As-induced adverse effects. LncRNAs are involved in the development of nerve injury, generally acting as sponges for microRNAs (miRNAs). This study aimed to investigate the competitive endogenous RNA (ceRNA) regulatory networks associated with arsenic-induced nerve damage. A total of 40 male Wistar rats were exposed to different doses of arsenic for 12 weeks, and samples were collected for pathological observation and high-throughput sequencing. The ceRNA network was constructed using Cytoscape, and key genes were identified through the PPI network and CytoHubba methods. A real-time quantitative PCR assay was performed to validate gene expression levels. The results showed that subchronic exposure to arsenic in drinking water resulted in pathological and ultrastructural damage to the hippocampal tissue, including changes in neuron morphology, mitochondria, and synapses. Exposure to arsenic results in the dysregulation of LncRNA and mRNA expression in the hippocampal tissues of rats. These molecules participated in multiple ceRNA axes and formed a network of ceRNAs associated with nerve injury. This study also verified key molecules within the ceRNA network and provided preliminary evidence implicating the ENRNOT-00000022622-miR-206-3p-Bdnf axis in the mechanism of neural damage induced by arsenic in rats. These findings provide novel insights into the underlying mechanism of nervous system damage induced by arsenic exposure.
Collapse
Affiliation(s)
- Fang Chu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Chunqing Lu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Zhe Jiao
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
- Institute for Kashin-Beck Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Wenjing Yang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Xiyue Yang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Hao Ma
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Hao Yu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Sheng Wang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Yang Li
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Dianjun Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| | - Hongna Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China; (F.C.); (C.L.); (W.Y.); (X.Y.); (H.M.); (H.Y.); (S.W.); (Y.L.)
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China;
| |
Collapse
|
22
|
Camacho-Hernandez GA, Jahan K, Newman AH. Illuminating the monoamine transporters: Fluorescently labelled ligands to study dopamine, serotonin and norepinephrine transporters. Basic Clin Pharmacol Toxicol 2023; 133:473-484. [PMID: 36527444 PMCID: PMC11309735 DOI: 10.1111/bcpt.13827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Fluorescence microscopy has revolutionized the visualization of physiological processes in live-cell systems. With the recent innovations in super resolution microscopy, these events can be examined with high precision and accuracy. The development of fluorescently labelled small molecules has provided a significant advance in understanding the physiological relevance of targeted proteins that can now be visualized at the cellular level. One set of physiologically important target proteins are the monoamine transporters (MATs) that play an instrumental role in maintaining monoamine signalling homeostasis. Understanding the mechanisms underlying their regulation and dysregulation is fundamental to treating several neuropsychiatric conditions such as attention deficit hyperactivity disorder (ADHD), anxiety, depression and substance use disorders. Herein, we describe the rationale behind the small molecule design of fluorescently labelled ligands (FLL) either as MAT substrates or inhibitors as well as their applications to advance our understanding of this class of transporters in health and disease.
Collapse
Affiliation(s)
- Gisela Andrea Camacho-Hernandez
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes on Drug Abuse - Intramural Research Program, Baltimore, Maryland, USA
| | - Khorshada Jahan
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes on Drug Abuse - Intramural Research Program, Baltimore, Maryland, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes on Drug Abuse - Intramural Research Program, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Jain S, Yee AG, Maas J, Gierok S, Xu H, Stansil J, Eriksen J, Nelson AB, Silm K, Ford CP, Edwards RH. Adaptor protein-3 produces synaptic vesicles that release phasic dopamine. Proc Natl Acad Sci U S A 2023; 120:e2309843120. [PMID: 37812725 PMCID: PMC10589613 DOI: 10.1073/pnas.2309843120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/06/2023] [Indexed: 10/11/2023] Open
Abstract
The burst firing of midbrain dopamine neurons releases a phasic dopamine signal that mediates reinforcement learning. At many synapses, however, high firing rates deplete synaptic vesicles (SVs), resulting in synaptic depression that limits release. What accounts for the increased release of dopamine by stimulation at high frequency? We find that adaptor protein-3 (AP-3) and its coat protein VPS41 promote axonal dopamine release by targeting vesicular monoamine transporter VMAT2 to the axon rather than dendrites. AP-3 and VPS41 also produce SVs that respond preferentially to high-frequency stimulation, independent of their role in axonal polarity. In addition, conditional inactivation of VPS41 in dopamine neurons impairs reinforcement learning, and this involves a defect in the frequency dependence of release rather than the amount of dopamine released. Thus, AP-3 and VPS41 promote the axonal polarity of dopamine release but enable learning by producing a distinct population of SVs tuned specifically to high firing frequency that confers the phasic release of dopamine.
Collapse
Affiliation(s)
- Shweta Jain
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Andrew G. Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO80045
| | - James Maas
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Sarah Gierok
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Hongfei Xu
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Jasmine Stansil
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Jacob Eriksen
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Alexandra B. Nelson
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Katlin Silm
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO80045
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Robert H. Edwards
- Department of Physiology, University of California School of Medicine, San Francisco, CA94143
- Department of Neurology, University of California School of Medicine, San Francisco, CA94143
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| |
Collapse
|
24
|
Wallace ML, Sabatini BL. Synaptic and circuit functions of multitransmitter neurons in the mammalian brain. Neuron 2023; 111:2969-2983. [PMID: 37463580 PMCID: PMC10592565 DOI: 10.1016/j.neuron.2023.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
Neurons in the mammalian brain are not limited to releasing a single neurotransmitter but often release multiple neurotransmitters onto postsynaptic cells. Here, we review recent findings of multitransmitter neurons found throughout the mammalian central nervous system. We highlight recent technological innovations that have made the identification of new multitransmitter neurons and the study of their synaptic properties possible. We also focus on mechanisms and molecular constituents required for neurotransmitter corelease at the axon terminal and synaptic vesicle, as well as some possible functions of multitransmitter neurons in diverse brain circuits. We expect that these approaches will lead to new insights into the mechanism and function of multitransmitter neurons, their role in circuits, and their contribution to normal and pathological brain function.
Collapse
Affiliation(s)
- Michael L Wallace
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Bernardo L Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Xu H, Oses-Prieto JA, Khvotchev M, Jain S, Liang J, Burlingame A, Edwards RH. Adaptor protein AP-3 produces synaptic vesicles that release at high frequency by recruiting phospholipid flippase ATP8A1. Nat Neurosci 2023; 26:1685-1700. [PMID: 37723322 DOI: 10.1038/s41593-023-01434-0] [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: 01/04/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023]
Abstract
Neural systems encode information in the frequency of action potentials, which is then decoded by synaptic transmission. However, the rapid, synchronous release of neurotransmitters depletes synaptic vesicles (SVs), limiting release at high firing rates. How then do synapses convey information about frequency? Here, we show in mouse hippocampal neurons and slices that the adaptor protein AP-3 makes a subset of SVs that respond specifically to high-frequency stimulation. Neurotransmitter transporters slot onto these SVs in different proportions, contributing to the distinct properties of release observed at different excitatory synapses. Proteomics reveals that AP-3 targets the phospholipid flippase ATP8A1 to SVs; loss of ATP8A1 recapitulates the defect in SV mobilization at high frequency observed with loss of AP-3. The mechanism involves recruitment of synapsin by the cytoplasmically oriented phosphatidylserine translocated by ATP8A1. Thus, ATP8A1 enables the subset of SVs made by AP-3 to release at high frequency.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Mikhail Khvotchev
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Shweta Jain
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Jocelyn Liang
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Alma Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Robert H Edwards
- Departments of Physiology and Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, USA.
| |
Collapse
|
26
|
Jain S, Yee AG, Maas J, Gierok S, Xu H, Stansil J, Eriksen J, Nelson A, Silm K, Ford CP, Edwards RH. Adaptor Protein-3 Produces Synaptic Vesicles that Release Phasic Dopamine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.07.552338. [PMID: 37609166 PMCID: PMC10441354 DOI: 10.1101/2023.08.07.552338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The burst firing of midbrain dopamine neurons releases a phasic dopamine signal that mediates reinforcement learning. At many synapses, however, high firing rates deplete synaptic vesicles (SVs), resulting in synaptic depression that limits release. What accounts for the increased release of dopamine by stimulation at high frequency? We find that adaptor protein-3 (AP-3) and its coat protein VPS41 promote axonal dopamine release by targeting vesicular monoamine transporter VMAT2 to the axon rather than dendrites. AP-3 and VPS41 also produce SVs that respond preferentially to high frequency stimulation, independent of their role in axonal polarity. In addition, conditional inactivation of VPS41 in dopamine neurons impairs reinforcement learning, and this involves a defect in the frequency dependence of release rather than the amount of dopamine released. Thus, AP-3 and VPS41 promote the axonal polarity of dopamine release but enable learning by producing a novel population of SVs tuned specifically to high firing frequency that confers the phasic release of dopamine.
Collapse
Affiliation(s)
- Shweta Jain
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Andrew G. Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora USA
| | - James Maas
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Sarah Gierok
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Hongfei Xu
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Jasmine Stansil
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Jacob Eriksen
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Alexandra Nelson
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Katlin Silm
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora USA
| | - Robert H. Edwards
- Department of Physiology, UCSF School of Medicine, San Francisco USA
- Department of Neurology, UCSF School of Medicine, San Francisco USA
| |
Collapse
|
27
|
Chen N, Zhang Y, Rivera-Rodriguez EJ, Yu AD, Hobin M, Rosbash M, Griffith LC. Widespread posttranscriptional regulation of cotransmission. SCIENCE ADVANCES 2023; 9:eadg9836. [PMID: 37267358 PMCID: PMC10413644 DOI: 10.1126/sciadv.adg9836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/01/2023] [Indexed: 06/04/2023]
Abstract
While neurotransmitter identity was once considered singular and immutable for mature neurons, it is now appreciated that one neuron can release multiple neuroactive substances (cotransmission) whose identities can even change over time. To explore the mechanisms that tune the suite of transmitters a neuron releases, we developed transcriptional and translational reporters for cholinergic, glutamatergic, and GABAergic signaling in Drosophila. We show that many glutamatergic and GABAergic cells also transcribe cholinergic genes, but fail to accumulate cholinergic effector proteins. Suppression of cholinergic signaling involves posttranscriptional regulation of cholinergic transcripts by the microRNA miR-190; chronic loss of miR-190 function allows expression of cholinergic machinery, reducing and fragmenting sleep. Using a "translation-trap" strategy, we show that neurons in these populations have episodes of transient translation of cholinergic proteins, demonstrating that suppression of cotransmission is actively modulated. Posttranscriptional restriction of fast transmitter cotransmission provides a mechanism allowing reversible tuning of neuronal output.
Collapse
Affiliation(s)
- Nannan Chen
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Yunpeng Zhang
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Emmanuel J. Rivera-Rodriguez
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Albert D. Yu
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
- Howard Hughes Medical Institute, Brandeis University, Waltham, MA 02454-9110, USA
| | - Michael Hobin
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Michael Rosbash
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
- Howard Hughes Medical Institute, Brandeis University, Waltham, MA 02454-9110, USA
| | - Leslie C. Griffith
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| |
Collapse
|
28
|
Malagon G, Myeong J, Klyachko VA. Two forms of asynchronous release with distinctive spatiotemporal dynamics in central synapses. eLife 2023; 12:e84041. [PMID: 37166282 PMCID: PMC10174687 DOI: 10.7554/elife.84041] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/24/2023] [Indexed: 05/12/2023] Open
Abstract
Asynchronous release is a ubiquitous form of neurotransmitter release that persists for tens to hundreds of milliseconds after an action potential. How asynchronous release is organized and regulated at the synaptic active zone (AZ) remains debatable. Using nanoscale-precision imaging of individual release events in rat hippocampal synapses, we observed two spatially distinct subpopulations of asynchronous events, ~75% of which occurred inside the AZ and with a bias towards the AZ center, while ~25% occurred outside of the functionally defined AZ, that is, ectopically. The two asynchronous event subpopulations also differed from each other in temporal properties, with ectopic events occurring at significantly longer time intervals from synchronous events than the asynchronous events inside the AZ. Both forms of asynchronous release did not, to a large extent, utilize the same release sites as synchronous events. The two asynchronous event subpopulations also differ from synchronous events in some aspects of exo-endocytosis coupling, particularly in the contribution from the fast calcium-dependent endocytosis. These results identify two subpopulations of asynchronous release events with distinctive organization and spatiotemporal dynamics.
Collapse
Affiliation(s)
- Gerardo Malagon
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| | - Jongyun Myeong
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of MedicineSt LouisUnited States
| |
Collapse
|
29
|
Chuhma N, Oh SJ, Rayport S. The dopamine neuron synaptic map in the striatum. Cell Rep 2023; 42:112204. [PMID: 36867530 PMCID: PMC10657204 DOI: 10.1016/j.celrep.2023.112204] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/21/2022] [Accepted: 02/16/2023] [Indexed: 03/04/2023] Open
Abstract
Dopamine neurons project to the striatum to control movement, cognition, and motivation via slower volume transmission as well as faster dopamine, glutamate, and GABA synaptic actions capable of conveying the temporal information in dopamine neuron firing. To define the scope of these synaptic actions, recordings of dopamine-neuron-evoked synaptic currents were made in four major striatal neuron types, spanning the entire striatum. This revealed that inhibitory postsynaptic currents are widespread, while excitatory postsynaptic currents are localized to the medial nucleus accumbens and the anterolateral-dorsal striatum, and that all synaptic actions are weak in the posterior striatum. Synaptic actions in cholinergic interneurons are the strongest, variably mediating inhibition throughout the striatum and excitation in the medial accumbens, capable of controlling their activity. This mapping shows that dopamine neuron synaptic actions extend throughout the striatum, preferentially target cholinergic interneurons, and define distinct striatal subregions.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Soo Jung Oh
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Stephen Rayport
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
30
|
Chen N, Zhang Y, Rivera-Rodriguez EJ, Yu AD, Hobin M, Rosbash M, Griffith LC. Widespread post-transcriptional regulation of co-transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530653. [PMID: 36909471 PMCID: PMC10002718 DOI: 10.1101/2023.03.01.530653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
While neurotransmitter identity was once considered singular and immutable for mature neurons, it is now appreciated that one neuron can release multiple neuroactive substances (co-transmission) whose identities can even change over time. To explore the mechanisms that tune the suite of transmitters a neuron releases, we developed transcriptional and translational reporters for cholinergic, glutamatergic, and GABAergic signaling in Drosophila . We show that many glutamatergic and GABAergic cells also transcribe cholinergic genes, but fail to accumulate cholinergic effector proteins. Suppression of cholinergic signaling involves posttranscriptional regulation of cholinergic transcripts by the microRNA miR-190; chronic loss of miR-190 function allows expression of cholinergic machinery, reducing and fragmenting sleep. Using a "translation-trap" strategy we show that neurons in these populations have episodes of transient translation of cholinergic proteins, demonstrating that suppression of co-transmission is actively modulated. Posttranscriptional restriction of fast transmitter co-transmission provides a mechanism allowing reversible tuning of neuronal output. One-Sentence Summary Cholinergic co-transmission in large populations of glutamatergic and GABAergic neurons in the Drosophila adult brain is controlled by miR-190.
Collapse
Affiliation(s)
- Nannan Chen
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Yunpeng Zhang
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Emmanuel J. Rivera-Rodriguez
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Albert D. Yu
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
- Howard Hughes Medical Institute, Brandeis University, Waltham, MA 02454-9110, USA
| | - Michael Hobin
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Michael Rosbash
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
- Howard Hughes Medical Institute, Brandeis University, Waltham, MA 02454-9110, USA
| | - Leslie C. Griffith
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| |
Collapse
|
31
|
Margolis EB, Moulton MG, Lambeth PS, O'Meara MJ. The life and times of endogenous opioid peptides: Updated understanding of synthesis, spatiotemporal dynamics, and the clinical impact in alcohol use disorder. Neuropharmacology 2023; 225:109376. [PMID: 36516892 PMCID: PMC10548835 DOI: 10.1016/j.neuropharm.2022.109376] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The opioid G-protein coupled receptors (GPCRs) strongly modulate many of the central nervous system structures that contribute to neurological and psychiatric disorders including pain, major depressive disorder, and substance use disorders. To better treat these and related diseases, it is essential to understand the signaling of their endogenous ligands. In this review, we focus on what is known and unknown about the regulation of the over two dozen endogenous peptides with high affinity for one or more of the opioid receptors. We briefly describe which peptides are produced, with a particular focus on the recently proposed possible synthesis pathways for the endomorphins. Next, we describe examples of endogenous opioid peptide expression organization in several neural circuits and how they appear to be released from specific neural compartments that vary across brain regions. We discuss current knowledge regarding the strength of neural activity required to drive endogenous opioid peptide release, clues about how far peptides diffuse from release sites, and their extracellular lifetime after release. Finally, as a translational example, we discuss the mechanisms of action of naltrexone (NTX), which is used clinically to treat alcohol use disorder. NTX is a synthetic morphine analog that non-specifically antagonizes the action of most endogenous opioid peptides developed in the 1960s and FDA approved in the 1980s. We review recent studies clarifying the precise endogenous activity that NTX prevents. Together, the works described here highlight the challenges and opportunities the complex opioid system presents as a therapeutic target.
Collapse
Affiliation(s)
- Elyssa B Margolis
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
| | - Madelyn G Moulton
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Philip S Lambeth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
32
|
Kim DI, Park S, Ye M, Chen JY, Jhang J, Hunker AC, Zweifel LS, Palmiter RD, Han S. Novel genetically encoded tools for imaging or silencing neuropeptide release from presynaptic terminals in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524797. [PMID: 36712060 PMCID: PMC9882317 DOI: 10.1101/2023.01.19.524797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neurons produce and release neuropeptides to communicate with one another. Despite their profound impact on critical brain functions, circuit-based mechanisms of peptidergic transmission are poorly understood, primarily due to the lack of tools for monitoring and manipulating neuropeptide release in vivo. Here, we report the development of two genetically encoded tools for investigating peptidergic transmission in behaving mice: a genetically encoded large dense core vesicle (LDCV) sensor that detects the neuropeptides release presynaptically, and a genetically encoded silencer that specifically degrades neuropeptides inside the LDCV. Monitoring and silencing peptidergic and glutamatergic transmissions from presynaptic terminals using our newly developed tools and existing genetic tools, respectively, reveal that neuropeptides, not glutamate, are the primary transmitter in encoding unconditioned stimulus during Pavlovian threat learning. These results show that our sensor and silencer for peptidergic transmission are reliable tools to investigate neuropeptidergic systems in awake behaving animals.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sekun Park
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jane Y. Chen
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Avery C. Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Richard D. Palmiter
- Howard Hughes Medical Institute, Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
33
|
Kim S, Sabatini BL. Analytical approaches to examine gamma-aminobutyric acid and glutamate vesicular co-packaging. Front Synaptic Neurosci 2023; 14:1076616. [PMID: 36685083 PMCID: PMC9846491 DOI: 10.3389/fnsyn.2022.1076616] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
Multi-transmitter neurons, i.e., those that release more than one type of neurotransmitter, have been found in many organisms and brain areas. Given the peculiar biology of these cells, as well as the potential for diverse effects of each of the transmitters released, new tools, and approaches are necessary to parse the mechanisms and functions of synaptic co-transmission. Recently, we and others have studied neurons that project to the lateral habenula and release both gamma-aminobutyric acid (GABA) and glutamate, in some cases by packaging both transmitters in the same synaptic vesicles. Here, we discuss the main challenges with current electrophysiological approaches to studying the mechanisms of glutamate/GABA co-release, a novel statistical analysis that can identify co-packaging of neurotransmitters versus release from separate vesicle, and the implications of glutamate/GABA co-release for synapse function and plasticity.
Collapse
Affiliation(s)
| | - Bernardo L. Sabatini
- Department of Neurobiology and Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, United States
| |
Collapse
|
34
|
Kershberg L, Banerjee A, Kaeser PS. Protein composition of axonal dopamine release sites in the striatum. eLife 2022; 11:e83018. [PMID: 36579890 PMCID: PMC9937654 DOI: 10.7554/elife.83018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
Dopamine is an important modulator of cognition and movement. We recently found that evoked dopamine secretion is fast and relies on active zone-like release sites. Here, we used in vivo biotin identification (iBioID) proximity proteomics in mouse striatum to assess which proteins are present at these sites. Using three release site baits, we identified proteins that are enriched over the general dopamine axonal protein content, and they fell into several categories, including active zone, Ca2+ regulatory, and synaptic vesicle proteins. We also detected many proteins not previously associated with vesicular exocytosis. Knockout of the presynaptic organizer protein RIM strongly decreased the hit number obtained with iBioID, while Synaptotagmin-1 knockout did not. α-Synuclein, a protein linked to Parkinson's disease, was enriched at release sites, and its enrichment was lost in both tested mutants. We conclude that RIM organizes scaffolded dopamine release sites and provide a proteomic assessment of the composition of these sites.
Collapse
Affiliation(s)
- Lauren Kershberg
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
35
|
Myeong J, Klyachko VA. Rapid astrocyte-dependent facilitation amplifies multi-vesicular release in hippocampal synapses. Cell Rep 2022; 41:111820. [PMID: 36516768 PMCID: PMC9805313 DOI: 10.1016/j.celrep.2022.111820] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Synaptic facilitation is a major form of short-term plasticity typically driven by an increase in residual presynaptic calcium. Using near-total internal reflection fluorescence (near-TIRF) imaging of single vesicle release in cultured hippocampal synapses, we demonstrate a distinctive, release-dependent form of facilitation in which probability of vesicle release is higher following a successful glutamate release event than following a failure. This phenomenon has an onset of ≤500 ms and lasts several seconds, resulting in clusters of successful release events. The release-dependent facilitation requires neuronal contact with astrocytes and astrocytic glutamate uptake by EAAT1. It is not observed in neurons grown alone or in the presence of astrocyte-conditioned media. This form of facilitation dynamically amplifies multi-vesicular release. Facilitation-evoked release events exhibit spatial clustering and have a preferential localization toward the active zone center. These results uncover a rapid astrocyte-dependent form of facilitation acting via modulation of multi-vesicular release and displaying distinctive spatiotemporal properties.
Collapse
Affiliation(s)
- Jongyun Myeong
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63132, USA
| | - Vitaly A. Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63132, USA,Lead contact,Correspondence:
| |
Collapse
|
36
|
Buck SA, Quincy Erickson-Oberg M, Logan RW, Freyberg Z. Relevance of interactions between dopamine and glutamate neurotransmission in schizophrenia. Mol Psychiatry 2022; 27:3583-3591. [PMID: 35681081 PMCID: PMC9712151 DOI: 10.1038/s41380-022-01649-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 02/08/2023]
Abstract
Dopamine (DA) and glutamate neurotransmission are strongly implicated in schizophrenia pathophysiology. While most studies focus on contributions of neurons that release only DA or glutamate, neither DA nor glutamate models alone recapitulate the full spectrum of schizophrenia pathophysiology. Similarly, therapeutic strategies limited to either system cannot effectively treat all three major symptom domains of schizophrenia: positive, negative, and cognitive symptoms. Increasing evidence suggests extensive interactions between the DA and glutamate systems and more effective treatments may therefore require the targeting of both DA and glutamate signaling. This offers the possibility that disrupting DA-glutamate circuitry between these two systems, particularly in the striatum and forebrain, culminate in schizophrenia pathophysiology. Yet, the mechanisms behind these interactions and their contributions to schizophrenia remain unclear. In addition to circuit- or system-level interactions between neurons that solely release either DA or glutamate, here we posit that functional alterations involving a subpopulation of neurons that co-release both DA and glutamate provide a novel point of integration between DA and glutamate systems, offering a key missing link in our understanding of schizophrenia pathophysiology. Better understanding of mechanisms underlying DA/glutamate co-release from these neurons may therefore shed new light on schizophrenia pathophysiology and lead to more effective therapeutics.
Collapse
Affiliation(s)
- Silas A Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - M Quincy Erickson-Oberg
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ryan W Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02118, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
37
|
Zhao F, Cheng Z, Piao J, Cui R, Li B. Dopamine Receptors: Is It Possible to Become a Therapeutic Target for Depression? Front Pharmacol 2022; 13:947785. [PMID: 36059987 PMCID: PMC9428607 DOI: 10.3389/fphar.2022.947785] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Dopamine and its receptors are currently recognized targets for the treatment of several neuropsychiatric disorders, including Parkinson’s disease, schizophrenia, some drug use addictions, as well as depression. Dopamine receptors are widely distributed in various regions of the brain, but their role and exact contribution to neuropsychiatric diseases has not yet been thoroughly studied. Based on the types of dopamine receptors and their distribution in different brain regions, this paper reviews the current research status of the molecular, cellular and circuit mechanisms of dopamine and its receptors involved in depression. Multiple lines of investigation of these mechanisms provide a new future direction for understanding the etiology and treatment of depression and potential new targets for antidepressant treatments.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Jingjing Piao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- Engineering Laboratory for Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
- *Correspondence: Bingjin Li,
| |
Collapse
|
38
|
Li H, Datunashvili M, Reyes RC, Voglmaier SM. Inositol hexakisphosphate kinases differentially regulate trafficking of vesicular glutamate transporters 1 and 2. Front Cell Neurosci 2022; 16:926794. [PMID: 35936490 PMCID: PMC9355605 DOI: 10.3389/fncel.2022.926794] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Inositol pyrophosphates have been implicated in cellular signaling and membrane trafficking, including synaptic vesicle (SV) recycling. Inositol hexakisphosphate kinases (IP6Ks) and their product, diphosphoinositol pentakisphosphate (PP-IP5 or IP7), directly and indirectly regulate proteins important in vesicle recycling by the activity-dependent bulk endocytosis pathway (ADBE). In the present study, we show that two isoforms, IP6K1 and IP6K3, are expressed in axons. The role of the kinases in SV recycling are investigated using pharmacologic inhibition, shRNA knockdown, and IP6K1 and IP6K3 knockout mice. Live-cell imaging experiments use optical reporters of SV recycling based on vesicular glutamate transporter isoforms, VGLUT1- and VGLUT2-pHluorins (pH), which recycle differently. VGLUT1-pH recycles by classical AP-2 dependent endocytosis under moderate stimulation conditions, while VGLUT2-pH recycles using AP-1 and AP-3 adaptor proteins as well. Using a short stimulus to release the readily releasable pool (RRP), we show that IP6K1 KO increases exocytosis of both VGLUT1-and VGLUT2-pH, while IP6K3 KO decreases the amount of both transporters in the RRP. In electrophysiological experiments we measure glutamate signaling with short stimuli and under the intense stimulation conditions that trigger bulk endocytosis. IP6K1 KO increases synaptic facilitation and IP6K3 KO decreases facilitation compared to wild type in CA1 hippocampal Schaffer collateral synapses. After intense stimulation, the rate of endocytosis of VGLUT2-pH, but not VGLUT1-pH, is increased by knockout, knockdown, and pharmacologic inhibition of IP6Ks. Thus IP6Ks differentially affect the endocytosis of two SV protein cargos that use different endocytic pathways. However, while IP6K1 KO and IP6K3 KO exert similar effects on endocytosis after stimulation, the isoforms exert different effects on exocytosis earlier in the stimulus and on the early phase of glutamate release. Taken together, the data indicate a role for IP6Ks both in exocytosis early in the stimulation period and in endocytosis, particularly under conditions that may utilize AP-1/3 adaptors.
Collapse
|
39
|
Burlingham SR, Wong NF, Peterkin L, Lubow L, Dos Santos Passos C, Benner O, Ghebrial M, Cast TP, Xu-Friedman MA, Südhof TC, Chanda S. Induction of synapse formation by de novo neurotransmitter synthesis. Nat Commun 2022; 13:3060. [PMID: 35650274 PMCID: PMC9160008 DOI: 10.1038/s41467-022-30756-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 05/17/2022] [Indexed: 11/09/2022] Open
Abstract
A vital question in neuroscience is how neurons align their postsynaptic structures with presynaptic release sites. Although synaptic adhesion proteins are known to contribute in this process, the role of neurotransmitters remains unclear. Here we inquire whether de novo biosynthesis and vesicular release of a noncanonical transmitter can facilitate the assembly of its corresponding postsynapses. We demonstrate that, in both stem cell-derived human neurons as well as in vivo mouse neurons of purely glutamatergic identity, ectopic expression of GABA-synthesis enzymes and vesicular transporters is sufficient to both produce GABA from ambient glutamate and transmit it from presynaptic terminals. This enables efficient accumulation and consistent activation of postsynaptic GABAA receptors, and generates fully functional GABAergic synapses that operate in parallel but independently of their glutamatergic counterparts. These findings suggest that presynaptic release of a neurotransmitter itself can signal the organization of relevant postsynaptic apparatus, which could be directly modified to reprogram the synapse identity of neurons.
Collapse
Affiliation(s)
- Scott R Burlingham
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Nicole F Wong
- Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lindsay Peterkin
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Lily Lubow
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | | | - Orion Benner
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Michael Ghebrial
- Biological Science, California State University Fullerton, Fullerton, CA, USA
| | - Thomas P Cast
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | | | - Thomas C Südhof
- Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Soham Chanda
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA.
- Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
40
|
Zych SM, Ford CP. Divergent properties and independent regulation of striatal dopamine and GABA co-transmission. Cell Rep 2022; 39:110823. [PMID: 35584679 PMCID: PMC9134867 DOI: 10.1016/j.celrep.2022.110823] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Accepted: 04/24/2022] [Indexed: 01/11/2023] Open
Abstract
Substantia nigra pars compacta (SNc) dopamine neurons play a key role in regulating the activity of striatal circuits within the basal ganglia. In addition to dopamine, these neurons release several other transmitters, including the major inhibitory neurotransmitter γ-aminobutyric acid (GABA). Both dopamine and GABA are loaded into SNc synaptic vesicles by the vesicular monoamine transporter 2 (VMAT2), and co-release of GABA provides strong inhibition to the striatum by directly inhibiting striatal medium spiny projection neurons (MSNs) through activation of GABAA receptors. Here, we found that despite both dopamine and GABA being co-packaged by VMAT2, the properties of transmission, including Ca2+ sensitivity, release probability, and requirement of active zone scaffolding proteins, differ between the two transmitters. Moreover, the extent by which presynaptic neuromodulators inhibit co-transmission also varied. Differences in modulation and the mechanisms controlling release allow for independent regulation of dopamine and GABA signals despite both being loaded via similar mechanisms.
Collapse
Affiliation(s)
- Sarah M Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, USA.
| |
Collapse
|
41
|
Xu H, Chang F, Jain S, Heller BA, Han X, Liu Y, Edwards RH. SNX5 targets a monoamine transporter to the TGN for assembly into dense core vesicles by AP-3. J Cell Biol 2022; 221:e202106083. [PMID: 35426896 PMCID: PMC9016777 DOI: 10.1083/jcb.202106083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/06/2021] [Accepted: 02/16/2022] [Indexed: 11/22/2022] Open
Abstract
The time course of signaling by peptide hormones, neural peptides, and other neuromodulators depends on their storage inside dense core vesicles (DCVs). Adaptor protein 3 (AP-3) assembles the membrane proteins that confer regulated release of DCVs and is thought to promote their trafficking from endosomes directly to maturing DCVs. We now find that regulated monoamine release from DCVs requires sorting nexin 5 (SNX5). Loss of SNX5 disrupts trafficking of the vesicular monoamine transporter (VMAT) to DCVs. The mechanism involves a role for SNX5 in retrograde transport of VMAT from endosomes to the TGN. However, this role for SNX5 conflicts with the proposed function of AP-3 in trafficking from endosomes directly to DCVs. We now identify a transient role for AP-3 at the TGN, where it associates with DCV cargo. Thus, retrograde transport from endosomes by SNX5 enables DCV assembly at the TGN by AP-3, resolving the apparent antagonism. A novel role for AP-3 at the TGN has implications for other organelles that also depend on this adaptor.
Collapse
Affiliation(s)
- Hongfei Xu
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Fei Chang
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Shweta Jain
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| | - Bradley Austin Heller
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| | - Xu Han
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yongjian Liu
- Jiangsu Key Laboratory of Xenotransplantation, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Departments of Pharmacology and Biological Chemistry, University of Pittsburgh, Pittsburgh, PA
| | - Robert H. Edwards
- Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
42
|
Melani R, Tritsch NX. Inhibitory co-transmission from midbrain dopamine neurons relies on presynaptic GABA uptake. Cell Rep 2022; 39:110716. [PMID: 35443174 PMCID: PMC9097974 DOI: 10.1016/j.celrep.2022.110716] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/18/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
Dopamine (DA)-releasing neurons in the substantia nigra pars compacta (SNcDA) inhibit target cells in the striatum through postsynaptic activation of γ-aminobutyric acid (GABA) receptors. However, the molecular mechanisms responsible for GABAergic signaling remain unclear, as SNcDA neurons lack enzymes typically required to produce GABA or package it into synaptic vesicles. Here, we show that aldehyde dehydrogenase 1a1 (Aldh1a1), an enzyme proposed to function as a GABA synthetic enzyme in SNcDA neurons, does not produce GABA for synaptic transmission. Instead, we demonstrate that SNcDA axons obtain GABA exclusively through presynaptic uptake using the membrane GABA transporter Gat1 (encoded by Slc6a1). GABA is then packaged for vesicular release using the vesicular monoamine transporter Vmat2. Our data therefore show that presynaptic transmitter recycling can substitute for de novo GABA synthesis and that Vmat2 contributes to vesicular GABA transport, expanding the range of molecular mechanisms available to neurons to support inhibitory synaptic communication. Melani and Tritsch demonstrate that inhibitory co-transmission from midbrain dopaminergic neurons does not depend on cell-autonomous GABA synthesis but instead on presynaptic import from the extracellular space through the membrane transporter Gat1 and that GABA loading into synaptic vesicles relies on the vesicular monoamine transporter Vmat2.
Collapse
Affiliation(s)
- Riccardo Melani
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA; Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY 10016, USA
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA; Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
43
|
Upmanyu N, Jin J, Emde HVD, Ganzella M, Bösche L, Malviya VN, Zhuleku E, Politi AZ, Ninov M, Silbern I, Leutenegger M, Urlaub H, Riedel D, Preobraschenski J, Milosevic I, Hell SW, Jahn R, Sambandan S. Colocalization of different neurotransmitter transporters on synaptic vesicles is sparse except for VGLUT1 and ZnT3. Neuron 2022; 110:1483-1497.e7. [PMID: 35263617 DOI: 10.1016/j.neuron.2022.02.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/08/2022] [Accepted: 02/10/2022] [Indexed: 12/26/2022]
Abstract
Vesicular transporters (VTs) define the type of neurotransmitter that synaptic vesicles (SVs) store and release. While certain mammalian neurons release multiple transmitters, it is not clear whether the release occurs from the same or distinct vesicle pools at the synapse. Using quantitative single-vesicle imaging, we show that a vast majority of SVs in the rodent brain contain only one type of VT, indicating specificity for a single neurotransmitter. Interestingly, SVs containing dual transporters are highly diverse (27 types) but small in proportion (2% of all SVs), excluding the largest pool that carries VGLUT1 and ZnT3 (34%). Using VGLUT1-ZnT3 SVs, we demonstrate that the transporter colocalization influences the SV content and synaptic quantal size. Thus, the presence of diverse transporters on the same vesicle is bona fide, and depending on the VT types, this may act to regulate neurotransmitter type, content, and release in space and time.
Collapse
Affiliation(s)
- Neha Upmanyu
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Jialin Jin
- European Neurosciences Institute, A Joint Initiative of the University Medical Center Göttingen and the Max Planck Society, Göttingen 37077, Germany
| | - Henrik von der Emde
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Marcelo Ganzella
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Leon Bösche
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Viveka Nand Malviya
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Evi Zhuleku
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Antonio Zaccaria Politi
- Live-Cell Imaging Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Momchil Ninov
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Marcel Leutenegger
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Dietmar Riedel
- Department of Structural Dynamics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Julia Preobraschenski
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen 37075, Germany
| | - Ira Milosevic
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 7BN, UK; Multidisciplinary Institute of Ageing, MIA-Portugal, University of Coimbra, Coimbra 3000-370, Portugal
| | - Stefan W Hell
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg 69028, Germany
| | - Reinhard Jahn
- Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Sivakumar Sambandan
- Synaptic Metal Ion Dynamics and Signaling, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Laboratory of Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany; Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany.
| |
Collapse
|
44
|
Clyburn C, Andresen MC, Ingram SL, Habecker BA. Untangling Peripheral Sympathetic Neurocircuits. Front Cardiovasc Med 2022; 9:842656. [PMID: 35224065 PMCID: PMC8866570 DOI: 10.3389/fcvm.2022.842656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
The sympathetic nervous system plays a critical role in regulating many autonomic functions, including cardiac rhythm. The postganglionic neurons in the sympathetic chain ganglia are essential components that relay sympathetic signals to target tissues and disruption of their activity leads to poor health outcomes. Despite this importance, the neurocircuitry within sympathetic ganglia is poorly understood. Canonically, postganglionic sympathetic neurons are thought to simply be activated by monosynaptic inputs from preganglionic cholinergic neurons of the intermediolateral cell columns of the spinal cord. Early electrophysiological studies of sympathetic ganglia where the peripheral nerve trunks were electrically stimulated identified excitatory cholinergic synaptic events in addition to retrograde action potentials, leading some to speculate that excitatory collateral projections are present. However, this seemed unlikely since sympathetic postganglionic neurons were known to synthesize and release norepinephrine and expression of dual neurochemical phenotypes had not been well recognized. In vitro studies clearly established the capacity of cultured sympathetic neurons to express and release acetylcholine and norepinephrine throughout development and even in pathophysiological conditions. Given this insight, we believe that the canonical view of ganglionic transmission needs to be reevaluated and may provide a mechanistic understanding of autonomic imbalance in disease. Further studies likely will require genetic models manipulating neurochemical phenotypes within sympathetic ganglia to resolve the function of cholinergic collateral projections between postganglionic neurons. In this perspective article, we will discuss the evidence for collateral projections in sympathetic ganglia, determine if current laboratory techniques could address these questions, and discuss potential obstacles and caveats.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - Michael C. Andresen
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, United States
| | - Beth A. Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
- *Correspondence: Beth A. Habecker
| |
Collapse
|
45
|
Banerjee A, Imig C, Balakrishnan K, Kershberg L, Lipstein N, Uronen RL, Wang J, Cai X, Benseler F, Rhee JS, Cooper BH, Liu C, Wojcik SM, Brose N, Kaeser PS. Molecular and functional architecture of striatal dopamine release sites. Neuron 2022; 110:248-265.e9. [PMID: 34767769 PMCID: PMC8859508 DOI: 10.1016/j.neuron.2021.10.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 09/22/2021] [Accepted: 10/19/2021] [Indexed: 01/21/2023]
Abstract
Despite the importance of dopamine for striatal circuit function, mechanistic understanding of dopamine transmission remains incomplete. We recently showed that dopamine secretion relies on the presynaptic scaffolding protein RIM, indicating that it occurs at active zone-like sites similar to classical synaptic vesicle exocytosis. Here, we establish using a systematic gene knockout approach that Munc13 and Liprin-α, active zone proteins for vesicle priming and release site organization, are important for dopamine secretion. Furthermore, RIM zinc finger and C2B domains, which bind to Munc13 and Liprin-α, respectively, are needed to restore dopamine release after RIM ablation. In contrast, and different from typical synapses, the active zone scaffolds RIM-BP and ELKS, and RIM domains that bind to them, are expendable. Hence, dopamine release necessitates priming and release site scaffolding by RIM, Munc13, and Liprin-α, but other active zone proteins are dispensable. Our work establishes that efficient release site architecture mediates fast dopamine exocytosis.
Collapse
Affiliation(s)
- Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | | | - Lauren Kershberg
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Noa Lipstein
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Riikka-Liisa Uronen
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Jiexin Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Xintong Cai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Jeong Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Benjamin H Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Sonja M Wojcik
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Prichard KL, O'Brien NS, Murcia SR, Baker JR, McCluskey A. Role of Clathrin and Dynamin in Clathrin Mediated Endocytosis/Synaptic Vesicle Recycling and Implications in Neurological Diseases. Front Cell Neurosci 2022; 15:754110. [PMID: 35115907 PMCID: PMC8805674 DOI: 10.3389/fncel.2021.754110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Endocytosis is a process essential to the health and well-being of cell. It is required for the internalisation and sorting of “cargo”—the macromolecules, proteins, receptors and lipids of cell signalling. Clathrin mediated endocytosis (CME) is one of the key processes required for cellular well-being and signalling pathway activation. CME is key role to the recycling of synaptic vesicles [synaptic vesicle recycling (SVR)] in the brain, it is pivotal to signalling across synapses enabling intracellular communication in the sensory and nervous systems. In this review we provide an overview of the general process of CME with a particular focus on two key proteins: clathrin and dynamin that have a central role to play in ensuing successful completion of CME. We examine these two proteins as they are the two endocytotic proteins for which small molecule inhibitors, often of known mechanism of action, have been identified. Inhibition of CME offers the potential to develop therapeutic interventions into conditions involving defects in CME. This review will discuss the roles and the current scope of inhibitors of clathrin and dynamin, providing an insight into how further developments could affect neurological disease treatments.
Collapse
|
47
|
Kljakic O, Janíčková H, Skirzewski M, Reichelt A, Memar S, El Mestikawy S, Li Y, Saksida LM, Bussey TJ, Prado VF, Prado MAM. Functional dissociation of behavioral effects from acetylcholine and glutamate released from cholinergic striatal interneurons. FASEB J 2022; 36:e22135. [PMID: 35032355 PMCID: PMC9303754 DOI: 10.1096/fj.202101425r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/03/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
In the striatum, cholinergic interneurons (CINs) have the ability to release both acetylcholine and glutamate, due to the expression of the vesicular acetylcholine transporter (VAChT) and the vesicular glutamate transporter 3 (VGLUT3). However, the relationship these neurotransmitters have in the regulation of behavior is not fully understood. Here we used reward‐based touchscreen tests in mice to assess the individual and combined contributions of acetylcholine/glutamate co‐transmission in behavior. We found that reduced levels of the VAChT from CINs negatively impacted dopamine signalling in response to reward, and disrupted complex responses in a sequential chain of events. In contrast, diminished VGLUT3 levels had somewhat opposite effects. When mutant mice were treated with haloperidol in a cue‐based task, the drug did not affect the performance of VAChT mutant mice, whereas VGLUT3 mutant mice were highly sensitive to haloperidol. In mice where both vesicular transporters were deleted from CINs, we observed altered reward‐evoked dopaminergic signalling and behavioral deficits that resemble, but were worse, than those in mice with specific loss of VAChT alone. These results demonstrate that the ability to secrete two different neurotransmitters allows CINs to exert complex modulation of a wide range of behaviors.
Collapse
Affiliation(s)
- Ornela Kljakic
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Helena Janíčková
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Miguel Skirzewski
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada
| | - Amy Reichelt
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sara Memar
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada
| | - Salah El Mestikawy
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada.,INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université, Paris, France
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Lisa M Saksida
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Timothy J Bussey
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Vania F Prado
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Marco A M Prado
- Translational Neuroscience Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.,Brain and Mind Institute, The University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
48
|
Pierson J, Shin YK. Stabilization of the SNARE Core by Complexin-1 Facilitates Fusion Pore Expansion. Front Mol Biosci 2022; 8:805000. [PMID: 34970598 PMCID: PMC8712692 DOI: 10.3389/fmolb.2021.805000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
In the neuron, neurotransmitter release is an essential function that must be both consistent and tightly regulated. The continuity of neurotransmitter release is dependent in large part on vesicle recycling. However, the protein factors that dictate the vesicle recycling pathway are elusive. Here, we use a single vesicle-to-supported bilayer fusion assay to investigate complexin-1 (cpx1)’s influence on SNARE-dependent fusion pore expansion. With total internal reflection (TIR) microscopy using a 10 kDa polymer fluorescence probe, we are able to detect the presence of large fusion pores. With cpx1, however, we observe a significant increase of the probability of the formation of large fusion pores. The domain deletion analysis reveals that the SNARE-binding core domain of cpx1 is mainly responsible for its ability to promote the fusion pore expansion. In addition, the results show that cpx1 helps the pore to expand larger, which results in faster release of the polymer probe. Thus, the results demonstrate a reciprocal relationship between event duration and the size of the fusion pore. Based on the data, a hypothetical mechanistic model can be deduced. In this mechanistic model, the cpx1 binding stabilizes the four-helix bundle structure of the SNARE core throughout the fusion pore expansion, whereby the highly curved bilayer within the fusion pore is stabilized by the SNARE pins.
Collapse
Affiliation(s)
- Josh Pierson
- Professor Yeon-Kyun Shin Lab, Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Yeon-Kyun Shin
- Professor Yeon-Kyun Shin Lab, Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| |
Collapse
|
49
|
Pignalosa FC, Desiderio A, Mirra P, Nigro C, Perruolo G, Ulianich L, Formisano P, Beguinot F, Miele C, Napoli R, Fiory F. Diabetes and Cognitive Impairment: A Role for Glucotoxicity and Dopaminergic Dysfunction. Int J Mol Sci 2021; 22:ijms222212366. [PMID: 34830246 PMCID: PMC8619146 DOI: 10.3390/ijms222212366] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/09/2021] [Accepted: 11/13/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by hyperglycemia, responsible for the onset of several long-term complications. Recent evidence suggests that cognitive dysfunction represents an emerging complication of DM, but the underlying molecular mechanisms are still obscure. Dopamine (DA), a neurotransmitter essentially known for its relevance in the regulation of behavior and movement, modulates cognitive function, too. Interestingly, alterations of the dopaminergic system have been observed in DM. This review aims to offer a comprehensive overview of the most relevant experimental results assessing DA’s role in cognitive function, highlighting the presence of dopaminergic dysfunction in DM and supporting a role for glucotoxicity in DM-associated dopaminergic dysfunction and cognitive impairment. Several studies confirm a role for DA in cognition both in animal models and in humans. Similarly, significant alterations of the dopaminergic system have been observed in animal models of experimental diabetes and in diabetic patients, too. Evidence is accumulating that advanced glycation end products (AGEs) and their precursor methylglyoxal (MGO) are associated with cognitive impairment and alterations of the dopaminergic system. Further research is needed to clarify the molecular mechanisms linking DM-associated dopaminergic dysfunction and cognitive impairment and to assess the deleterious impact of glucotoxicity.
Collapse
Affiliation(s)
- Francesca Chiara Pignalosa
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Antonella Desiderio
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Paola Mirra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Cecilia Nigro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Giuseppe Perruolo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Luca Ulianich
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Claudia Miele
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-746-3248
| | - Raffaele Napoli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
| | - Francesca Fiory
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| |
Collapse
|
50
|
Cifuentes F, Morales MA. Functional Implications of Neurotransmitter Segregation. Front Neural Circuits 2021; 15:738516. [PMID: 34720888 PMCID: PMC8548464 DOI: 10.3389/fncir.2021.738516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Here, we present and discuss the characteristics and properties of neurotransmitter segregation, a subtype of neurotransmitter cotransmission. We review early evidence of segregation and discuss its properties, such as plasticity, while placing special emphasis on its probable functional implications, either in the central nervous system (CNS) or the autonomic nervous system. Neurotransmitter segregation is a process by which neurons separately route transmitters to independent and distant or to neighboring neuronal processes; it is a plastic phenomenon that changes according to synaptic transmission requirements and is regulated by target-derived signals. Distant neurotransmitter segregation in the CNS has been shown to be related to an autocrine/paracrine function of some neurotransmitters. In retinal amacrine cells, segregation of acetylcholine (ACh) and GABA, and glycine and glutamate to neighboring terminals has been related to the regulation of the firing rate of direction-selective ganglion cells. In the rat superior cervical ganglion, segregation of ACh and GABA to neighboring varicosities shows a heterogeneous regional distribution, which is correlated to a similar regional distribution in transmission strength. We propose that greater segregation of ACh and GABA produces less GABAergic inhibition, strengthening ganglionic transmission. Segregation of ACh and GABA varies in different physiopathological conditions; specifically, segregation increases in acute sympathetic hyperactivity that occurs in cold stress, does not vary in chronic hyperactivity that occurs in hypertension, and rises in early ages of normotensive and hypertensive rats. Given this, we propose that variations in the extent of transmitter segregation may contribute to the alteration of neural activity that occurs in some physiopathological conditions and with age.
Collapse
Affiliation(s)
- Fredy Cifuentes
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Angel Morales
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|