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
|
Duan Z, Zhao W, Tong Y, Coenen VA, Döbrössy MD. Acute and chronic gene expression activation following medial forebrain bundle DBS and selective dopamine pathway stimulation. Sci Rep 2025; 15:7131. [PMID: 40021746 PMCID: PMC11871370 DOI: 10.1038/s41598-025-91994-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
Deep brain stimulation (DBS) of the medial forebrain bundle (mfb) demonstrated anti-depressant effects both clinically and experimentally. Modulation of mesocorticolimbic dopaminergic (DA) activity could contribute-in part-to the therapeutic effects. By comparing selective and pathway specific midbrain DA optogenetic stimulation with the global, non-pathway specific mfb-DBS, the study explored changes in gene-expression of key biomarkers associated with neurocircuitry of depression. Rats received either optogenetic DAergic or mfb-DBS, delivered as acute/single or chronic/repeated stimulation. Micro-dissected regions were prepared for in situ hybridization targeting biomarkers of GABAergic, glutamatergic, and dopaminergic systems. Mfb-DBS mediated DA independent pathway increased GABAergic biomarkers (GABAA, GAD1) in frontal and accumbal regions, not in midbrain. The combinations of low frequency/high pulse width and high frequency/low pulse width stimulation generally increased biomarker expression similarly, but chronic/repetitive stimulation had no accumulative effect. Interestingly, unilateral stimulation had bilateral effects, but stimulation modalities had little impact on DAT and Vglut2 expression. In conclusion, both low and high frequency, acute/single and chronic/repetitive mfb-DBS-but not selective optogenetic stimulation -activated gene expression of biomarkers associated with GABAergic transmission. The increased expression was transitory and less chronic than predicted. Importantly, the study provides evidence that the anti-depressant therapeutic effects of clinical medial forebrain bundle DBS occurs-in part-be via modulation of GABAergic signalling which in turn could regulate the release of dopamine in frontal and accumbal regions. In addition, clinical implication of the data is that unilateral stimulation had bilateral consequences on the gene expression, although the physiological and functional sequelae of this are yet unknown.
Collapse
Affiliation(s)
- Zhuo Duan
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wen Zhao
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yixin Tong
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Volker A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany.
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany.
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Varga AG, Reid BT, Maletz SN, Dossat AM, Levitt ES. Opposing control of the respiratory brainstem on multiple timescales achieved by transmitter co-release from the locus coeruleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639476. [PMID: 40027822 PMCID: PMC11870594 DOI: 10.1101/2025.02.21.639476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The locus coeruleus (LC) provides widespread noradrenergic (NAergic) modulation throughout the brain to influence a wide range of functions, including breathing. Although both anatomical and physiological evidence supports the involvement of the LC in both the upstream integration and the downstream modulation of breathing, the circuitry behind the latter is unknown. Here, we show that NAergic LC neurons send projections to the Kӧlliker-Fuse nucleus (KF), a critical site in the control of breathing. Long duration activation of NAergic LC neuron terminals in pontine slices induces persistent inhibitory and excitatory NA currents or increases firing rate in postsynaptic KF neurons. Short stimulation on the other hand leads to the VGluT2-dependent release of glutamate that may be co-released with NA in a monosynaptic circuit. Together these results demonstrate that LC neurons can exert flexible, opposing effects on different timescales via glutamatergic and NAergic signaling onto a key respiratory brainstem nucleus.
Collapse
|
4
|
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
|
5
|
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
|
6
|
Johnson NL, Cotelo-Larrea A, Stetzik LA, Akkaya UM, Zhang Z, Gadziola MA, Varga AG, Ma M, Wesson DW. Dopaminergic signaling to ventral striatum neurons initiates sniffing behavior. Nat Commun 2025; 16:336. [PMID: 39747223 PMCID: PMC11696867 DOI: 10.1038/s41467-024-55644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Sniffing is a motivated behavior displayed by nearly all terrestrial vertebrates. While sniffing is associated with acquiring and processing odors, sniffing is also intertwined with affective and motivated states. The systems which influence the display of sniffing are unclear. Here, we report that dopamine release into the ventral striatum in mice is coupled with bouts of sniffing and that stimulation of dopaminergic terminals in these regions drives increases in respiratory rate to initiate sniffing whereas inhibition of these terminals reduces respiratory rate. Both the firing of individual neurons and the activity of post-synaptic D1 and D2 dopamine receptor-expressing neurons are coupled with sniffing and local antagonism of D1 and D2 receptors squelches sniffing. Together, these results support a model whereby sniffing can be initiated by dopamine's actions upon ventral striatum neurons. The nature of sniffing being integral to both olfaction and motivated behaviors implicates this circuit in a wide array of functions.
Collapse
Affiliation(s)
- Natalie L Johnson
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Anamaria Cotelo-Larrea
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Lucas A Stetzik
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Umit M Akkaya
- Department of Computer Engineering, Gebze Technical University, Kocaeli, Turkey
| | - Zihao Zhang
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA
| | - Marie A Gadziola
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Adrienn G Varga
- Department of Neuroscience, Breathing Research and Therapeutics Center, McKnight Brain Institute; University of Florida College of Medicine, Gainesville, FL, USA
| | - Minghong Ma
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel W Wesson
- Department of Pharmacology and Therapeutics, Florida Chemical Senses Institute, Center for Addiction Research and Education; University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
7
|
Kitchigina VF. Colocalization of Neurotransmitters in Hippocampus and Afferent Systems: Possible Functional Role. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:61-78. [PMID: 40058974 DOI: 10.1134/s0006297924603915] [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: 10/30/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 05/13/2025]
Abstract
In neurophysiology, the transmitter phenotype is considered as an indicator of neuronal identity. It has become known at the end of last century that a nerve cell can produce and use several different molecules to communicate with other neurons. These could be "classical" transmitters: glutamate or gamma-aminobutyric acid (or acetylcholine, serotonin, norepinephrine), as well as secondary messengers, mainly neuropeptides released from the same neurons. In the case, when classical neurotransmitters are released together from the same nerve cell, this event is called cotransmission or corelease (release from the same vesicles). In this review article, the term "cotransmission" is used in a broad sense, denoting neurons that can release more than one classical mediator. Since transmitters are often intermediate products of metabolism and are found in many cells, the neuron classification is currently based on the carrier proteins (transporters) that "pack" neurotransmitters synthesized in the cytoplasm into vesicles. Here, we limit the issue of colocalization of the main neurotransmitters in mammals to the neurons of hippocampus and those structures that send their pathways to it. The review considers problems concerning the mechanisms of multitransmitter signaling, as well as probable functional role of mediator colocalization in the work of hippocampus, which yet has been poorly understood. It has been suggested that co-expression of different mediator phenotypes is involved in maintaining the balance of excitation and inhibition in different regions of hippocampus, facilitates rapid selection of information processing mode, induction of long-term potentiation, maintenance of spatial coding by place cells, as well as ensuring flexibility of learning and formation of working memory. However, the functional role of mediator colocalization, as well as the mechanisms of release of "dual" transmitters, have not been fully elucidated. The solution of these problems will advance some areas of fundamental neuroscience and help in the treatment of those diseases, where disruption of the balance between excitation and inhibition is detected, such as, for example, in epilepsy, Alzheimer's disease, and many others.
Collapse
Affiliation(s)
- Valentina F Kitchigina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
8
|
Jerzemowska G, Podlacha M, Orzeł-Gryglewska J. Amphetamine Injection into the Nucleus Accumbens and Electrical Stimulation of the Ventral Tegmental Area in Rats After Novelty Test-Behavioral and Neurochemical Correlates. Int J Mol Sci 2024; 26:182. [PMID: 39796042 PMCID: PMC11720036 DOI: 10.3390/ijms26010182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Amphetamine abuse is a global health epidemic that is difficult to treat due to individual differences in response to environmental factors, including stress reactivity and anxiety levels, as well as individual neuronal differences, which may result in increased/decreased vulnerability to addiction. In the present study, we investigated whether the Wistar rats behavioral traits of high (HR) and low (LR) locomotor activity to novelty influence motivational behavior (induced feeding model; iFR by electrical stimulation of the ventral tegmental area; Es-VTA) supported by amphetamine injection into the nucleus accumbens shell (AcbSh) (HRAmph, n = 5; LRAmph, n = 5). A correlation was found between the novelty test's locomotor activity score and the frequency threshold percentage change (p < 0.001, Rs = -0.867). In HRAmph, there was a shortening (-24.16%), while in LRAmph, there was a lengthening (+51.84%) of iFR latency. Immunofluorescence studies showed differential neuronal density (activity of tyrosine hydroxylase, choline acetyltransferase, and cFos protein) in the selected brain structures in HRAmph and LRAmph animals as well as in comparison to a control group (HRACSF, n = 5; LRACSF, n = 5). These results contribute to expanding the state of knowledge of the behavioral and neuronal propensity to take drug abuse.
Collapse
Affiliation(s)
- Grażyna Jerzemowska
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, 59 Wita Stwosza Str., 80-308 Gdansk, Poland;
| | - Magdalena Podlacha
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, 59 Wita Stwosza Str., 80-308 Gdansk, Poland;
| | - Jolanta Orzeł-Gryglewska
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, 59 Wita Stwosza Str., 80-308 Gdansk, Poland;
| |
Collapse
|
9
|
Parker KE, Kuo CC, Buckley AR, Patterson AP, Duong V, Hunter SC, McCall JG. Monosynaptic ventral tegmental area glutamate projections to the locus coeruleus enhance aversive processing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.615025. [PMID: 39713345 PMCID: PMC11661122 DOI: 10.1101/2024.10.04.615025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Distinct excitatory synaptic inputs to the locus coeruleus (LC) modulate behavioral flexibility. Here we identify a novel monosynaptic glutamatergic input to the LC from the ventral tegmental area (VTA). We show robust VTA axonal projections provide direct glutamatergic transmission to LC. Despite weak synaptic summation, optogenetic activation of these axons enhances LC tonic firing and facilitates real-time and conditioned aversive behaviors. We hypothesized this projection may modulate synaptic integration with other excitatory inputs. We then used coincident VTA-LC photostimulation with local electrical stimulation and observed enhanced LC burst induction. To determine whether this integration also occurs in vivo, we took an analogous approach measuring reward-seeking behavior during unpredictable probabilistic punishment. Here, glutamatergic VTA-LC photostimulation during a concurrent noxious stimulus did not delay reward-seeking behavior, but increased probability of task failure. Together, we identified a novel VTA-LC glutamatergic projection that drives concurrent synaptic summation during salient stimuli to promote behavioral avoidance.
Collapse
Affiliation(s)
- Kyle E. Parker
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Alex R. Buckley
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Abigail P. Patterson
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Vincent Duong
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah C. Hunter
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G. McCall
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
10
|
Lu MQ, Shi ZG, Shang J, Gao L, Gao L, Gao WJ. ChangPu YuJin Tang improves Tourette disorder symptoms by modulating amino acid neurotransmitters in IDPN model rats. Metab Brain Dis 2024; 39:1543-1558. [PMID: 39312065 DOI: 10.1007/s11011-024-01411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 08/09/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Changpu Yujin Tang(CPYJT), a Chinese herbal compound, is an effective therapeutic strategy for pediatric patients with Tourette disorder (TD). Therefore, this work aims to investigate the therapeutic mechanisms of CPYJT. METHODS Behavioral and cellular ultrastructural evaluation of the therapeutic effects of CPYJT in TD model rats. Colorimetric methods, reverse transcription‑quantitative PCR, and Western Blot were used to measure the altered levels of GLU, GABA, and the levels of VGLUT1, GLUD1, GABRA3, and GAD65 in the cortex, striatum, and thalamus of the TD model rats after 7, 14, 21, and 28 days of CPYJT administration. RESULTS CPYJT significantly reduced stereotypic behavior and motor behavior scores in TD model rats. CPYJT ameliorates myelin structural damage in TD model rat neuronal cells. CPYJT decreased GLU content, elevated GABA content, decreased GLUD1 and VGLUT1 levels, and elevated GAD65 and GABRA3 levels in TD model rats' cortex, striatum, and thalamus. CPYJT has different regulatory time points in the cortex, striatum, and thalamus for critical factors of amino acid-based neurotransmission. CONCLUSION CPYJT protects behavioral and structural damage of neuronal cells in multiple brain regions in TD model rats.
Collapse
Affiliation(s)
- Man-Qi Lu
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, shanghai, 200000, China
| | - Zheng-Gang Shi
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China.
| | - Jing Shang
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| | - Lü Gao
- Shanxi University Of Chinese Medicine Third Clinical Medical College Pediatric Teaching and Research Department, Taiyuan, 140100, Shanxi, China
| | - Lei Gao
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| | - Wei-Jiao Gao
- Clinical College of Chinese Medicine, Gansu University Of Chinese Medicine, Lanzhou, 730000, Gansu, P.R. China
| |
Collapse
|
11
|
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
|
12
|
Conrad WS, Oriol L, Kollman GJ, Faget L, Hnasko TS. Proportion and distribution of neurotransmitter-defined cell types in the ventral tegmental area and substantia nigra pars compacta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582356. [PMID: 38464250 PMCID: PMC10925288 DOI: 10.1101/2024.02.28.582356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Most studies on the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) have focused on dopamine neurons and their role in processes such as motivation, learning, movement, and associated disorders such as addiction and Parkinson's disease. However there has been increasing attention on other VTA and SNc cell types that release GABA, glutamate, or a combination of neurotransmitters. Yet the relative distributions and proportions of neurotransmitter-defined cell types across VTA and SNc has remained unclear. Here, we used fluorescent in situ hybridization in male and female mice to label VTA and SNc neurons that expressed mRNA encoding the canonical vesicular transporters for dopamine, GABA, or glutamate: vesicular monoamine transporter (VMAT2), vesicular GABA transporter (VGAT), and vesicular glutamate transporter (VGLUT2). Within VTA, we found that no one type was particularly more abundant, instead we observed similar numbers of VMAT2+ (44%), VGAT+ (37%) and VGLUT2+ (41%) neurons. In SNc we found that a slight majority of neurons expressed VMAT2 (54%), fewer were VGAT+ (42%), and VGLUT2+ neurons were least abundant (16%). Moreover, 20% of VTA neurons and 10% of SNc neurons expressed more than one vesicular transporter, including 45% of VGLUT2+ neurons. We also assessed within VTA and SNc subregions and found remarkable heterogeneity in cell-type composition. And by quantifying density across both anterior-posterior and medial-lateral axes we generated heatmaps to visualize the distribution of each cell type. Our data complement recent single-cell RNAseq studies and support a more diverse landscape of neurotransmitter-defined cell types in VTA and SNc than is typically appreciated.
Collapse
Affiliation(s)
- William S Conrad
- University of California, San Diego, Department of Neurosciences, La Jolla CA, USA
| | - Lucie Oriol
- University of California, San Diego, Department of Neurosciences, La Jolla CA, USA
| | - Grace J Kollman
- University of California, San Diego, Department of Neurosciences, La Jolla CA, USA
| | - Lauren Faget
- University of California, San Diego, Department of Neurosciences, La Jolla CA, USA
| | - Thomas S Hnasko
- University of California, San Diego, Department of Neurosciences, La Jolla CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase MD 20815, USA
| |
Collapse
|
13
|
Muir J, Anguiano M, Kim CK. Neuromodulator and neuropeptide sensors and probes for precise circuit interrogation in vivo. Science 2024; 385:eadn6671. [PMID: 39325905 PMCID: PMC11488521 DOI: 10.1126/science.adn6671] [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: 03/27/2024] [Accepted: 07/01/2024] [Indexed: 09/28/2024]
Abstract
To determine how neuronal circuits encode and drive behavior, it is often necessary to measure and manipulate different aspects of neurochemical signaling in awake animals. Optogenetics and calcium sensors have paved the way for these types of studies, allowing for the perturbation and readout of spiking activity within genetically defined cell types. However, these methods lack the ability to further disentangle the roles of individual neuromodulator and neuropeptides on circuits and behavior. We review recent advances in chemical biology tools that enable precise spatiotemporal monitoring and control over individual neuroeffectors and their receptors in vivo. We also highlight discoveries enabled by such tools, revealing how these molecules signal across different timescales to drive learning, orchestrate behavioral changes, and modulate circuit activity.
Collapse
Affiliation(s)
- J. Muir
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - M. Anguiano
- Neuroscience Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - C. K. Kim
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
14
|
Chang Y, Lusk S, Chang A, Ward CS, Ray RS. Vglut2-based glutamatergic signaling in central noradrenergic neurons is dispensable for normal breathing and chemosensory reflexes. eLife 2024; 12:RP88673. [PMID: 39287624 PMCID: PMC11407767 DOI: 10.7554/elife.88673] [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: 09/19/2024] Open
Abstract
Central noradrenergic (NA) neurons are key constituents of the respiratory homeostatic network. NA dysfunction is implicated in several developmental respiratory disorders including Congenital Central Hyperventilation Syndrome (CCHS), Sudden Infant Death Syndrome (SIDS), and Rett Syndrome. The current unchallenged paradigm in the field, supported by multiple studies, is that glutamate co-transmission in subsets of central NA neurons plays a role in breathing control. If true, NA-glutamate co-transmission may also be mechanistically important in respiratory disorders. However, the requirement of NA-derived glutamate in breathing has not been directly tested and the extent of glutamate co-transmission in the central NA system remains uncharacterized. Therefore, we fully characterized the cumulative fate maps and acute adult expression patterns of all three vesicular glutamate transporters (Slc17a7 (Vglut1), Slc17a6 (Vglut2), and Slc17a8 (Vglut3)) in NA neurons, identifying a novel, dynamic expression pattern for Vglut2 and an undescribed co-expression domain for Vglut3 in the NA system. In contrast to our initial hypothesis that NA-derived glutamate is required to breathing, our functional studies showed that loss of Vglut2 throughout the NA system failed to alter breathing or metabolism under room air, hypercapnia, or hypoxia in unrestrained and unanesthetized mice. These data demonstrate that Vglut2-based glutamatergic signaling within the central NA system is not required for normal baseline breathing and hypercapnic, hypoxic chemosensory reflexes. These outcomes challenge the current understanding of central NA neurons in the control of breathing and suggests that glutamate may not be a critical target to understand NA neuron dysfunction in respiratory diseases.
Collapse
Affiliation(s)
- Yuan Chang
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Department of Integrative Physiology, Baylor College of MedicineHoustonUnited States
| | - Savannah Lusk
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Andersen Chang
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Christopher S Ward
- Department of Integrative Physiology, Baylor College of MedicineHoustonUnited States
| | - Russell S Ray
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Department of Integrative Physiology, Baylor College of MedicineHoustonUnited States
- McNair Medical InstituteHoustonUnited States
| |
Collapse
|
15
|
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
|
16
|
Liao SC, Kano K, Phanse S, Nguyen M, Margolis E, Fu Y, Meng J, Moutaoufik MT, Chatterton Z, Aoki H, Simms J, Hsieh I, Suteja F, Sei Y, Huang EJ, McAvoy K, Manfredi G, Halliday G, Babu M, Nakamura K. CHCHD2 mutant mice display mitochondrial protein accumulation and disrupted energy metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610586. [PMID: 39257750 PMCID: PMC11384018 DOI: 10.1101/2024.08.30.610586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Mutations in the mitochondrial cristae protein CHCHD2 lead to a late-onset autosomal dominant form of Parkinson's disease (PD) which closely resembles idiopathic PD, providing the opportunity to gain new insights into the mechanisms of mitochondrial dysfunction contributing to PD. To begin to address this, we used CRISPR genome-editing to generate CHCHD2 T61I point mutant mice. CHCHD2 T61I mice had normal viability, and had only subtle motor deficits with no signs of premature dopaminergic (DA) neuron degeneration. Nonetheless, CHCHD2 T61I mice exhibited robust molecular changes in the brain including increased CHCHD2 insolubility, accumulation of CHCHD2 protein preferentially in the substantia nigra (SN), and elevated levels of α-synuclein. Metabolic analyses revealed an increase in glucose metabolism through glycolysis relative to the TCA cycle with increased respiratory exchange ratio, and immune-electron microscopy revelated disrupted mitochondria in DA neurons. Moreover, spatial genomics revealed decreased expression of mitochondrial complex I and III respiratory chain proteins, while proteomics revealed increased respiratory chain and other mitochondrial protein-protein interactions. As such, the CHCHD2 T61I point-mutation mice exhibit robust mitochondrial disruption and a consequent metabolic shift towards glycolysis. These findings thus establish CHCHD2 T61I mice as a new model for mitochondrial-based PD, and implicate disrupted respiratory chain function as a likely causative driver.
Collapse
Affiliation(s)
- Szu-Chi Liao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Kohei Kano
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Sadhna Phanse
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Mai Nguyen
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Elyssa Margolis
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA
| | - YuHong Fu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jonathan Meng
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA
| | | | - Zac Chatterton
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Hiroyuki Aoki
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Jeffrey Simms
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Ivy Hsieh
- Department of Pathology, University of California San Francisco, San Francisco, CA
| | - Felecia Suteja
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Yoshitaka Sei
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Eric J. Huang
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA
- Department of Pathology, University of California San Francisco, San Francisco, CA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA
| | - Kevin McAvoy
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Glenda Halliday
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Ken Nakamura
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA
| |
Collapse
|
17
|
Johnson NL, Cotelo-Larrea A, Stetzik LA, Akkaya UM, Zhang Z, Gadziola MA, Varga AG, Ma M, Wesson DW. Sniffing can be initiated by dopamine's actions on ventral striatum neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.19.581052. [PMID: 39229099 PMCID: PMC11370338 DOI: 10.1101/2024.02.19.581052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Sniffing is a motivated behavior displayed by nearly all terrestrial vertebrates. While sniffing is associated with acquiring and processing odors, sniffing is also intertwined with affective and motivated states. The neuromodulatory systems which influence the display of sniffing are unclear. Here, we report that dopamine release into the ventral striatum is coupled with bouts of sniffing and that stimulation of dopaminergic terminals in these regions drives increases in respiratory rate to initiate sniffing whereas inhibition of these terminals reduces respiratory rate. Both the firing of individual neurons and the activity of post-synaptic D1 and D2 receptor-expressing neurons in the ventral striatum are also coupled with sniffing and local antagonism of D1 and D2 receptors squelches sniffing. Together, these results support a model whereby sniffing can be initiated by dopamine's actions upon ventral striatum neurons. The nature of sniffing being integral to both olfaction and motivated behaviors implicates this circuit in a wide array of functions.
Collapse
|
18
|
Favier M, Martin Garcia E, Icick R, de Almeida C, Jehl J, Desplanque M, Zimmermann J, Henrion A, Mansouri-Guilani N, Mounier C, Ribeiro S, Henderson F, Geoffroy A, Mella S, Poirel O, Bernard V, Fabre V, Li Y, Rosenmund C, Jamain S, Vorspan F, Mourot A, Duriez P, Pinhas L, Maldonado R, Pietrancosta N, Daumas S, El Mestikawy S. The human VGLUT3-pT8I mutation elicits uneven striatal DA signaling, food or drug maladaptive consumption in male mice. Nat Commun 2024; 15:5691. [PMID: 38971801 PMCID: PMC11227582 DOI: 10.1038/s41467-024-49371-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/07/2024] [Indexed: 07/08/2024] Open
Abstract
Cholinergic striatal interneurons (ChIs) express the vesicular glutamate transporter 3 (VGLUT3) which allows them to regulate the striatal network with glutamate and acetylcholine (ACh). In addition, VGLUT3-dependent glutamate increases ACh vesicular stores through vesicular synergy. A missense polymorphism, VGLUT3-p.T8I, was identified in patients with substance use disorders (SUDs) and eating disorders (EDs). A mouse line was generated to understand the neurochemical and behavioral impact of the p.T8I variant. In VGLUT3T8I/T8I male mice, glutamate signaling was unchanged but vesicular synergy and ACh release were blunted. Mutant male mice exhibited a reduced DA release in the dorsomedial striatum but not in the dorsolateral striatum, facilitating habit formation and exacerbating maladaptive use of drug or food. Increasing ACh tone with donepezil reversed the self-starvation phenotype observed in VGLUT3T8I/T8I male mice. Our study suggests that unbalanced dopaminergic transmission in the dorsal striatum could be a common mechanism between SUDs and EDs.
Collapse
Affiliation(s)
- Mathieu Favier
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada.
| | - Elena Martin Garcia
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Romain Icick
- Département de Psychiatrie et de Médecine Addictologique, DMU Neurosciences, APHP.Nord, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, F-75010, France
- INSERM U1144, "Therapeutic optimization in neuropsychopharmacology", Paris, F-75006, France
- Université Paris Cité, Inserm UMR-S1144, Paris, F-75006, France
- Neurobiologie Intégrative des Systèmes Cholinergiques, Département de Neurosciences, Institut Pasteur, Paris, F-75015, France
| | - Camille de Almeida
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Joachim Jehl
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
- Brain Plasticity Unit, CNRS UMR 8249, ESPCI Paris, PSL Research University, 75005, Paris, France
| | - Mazarine Desplanque
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Johannes Zimmermann
- Neurocure NWFZ, Charite Universitaetsmedizin, Institut für Neurophysiologie, Charitéplatz 1, 10117, Berlin, Germany
| | - Annabelle Henrion
- Fondation FondaMental, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Translational Neuropsychiatry, F-94010, Créteil, France
| | - Nina Mansouri-Guilani
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Coline Mounier
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Svethna Ribeiro
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Fiona Henderson
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Andrea Geoffroy
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Sebastien Mella
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Odile Poirel
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Véronique Bernard
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Véronique Fabre
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Christian Rosenmund
- Neurocure NWFZ, Charite Universitaetsmedizin, Institut für Neurophysiologie, Charitéplatz 1, 10117, Berlin, Germany
| | - Stéphane Jamain
- Fondation FondaMental, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Translational Neuropsychiatry, F-94010, Créteil, France
| | - Florence Vorspan
- Département de Psychiatrie et de Médecine Addictologique, DMU Neurosciences, APHP.Nord, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, F-75010, France
- INSERM U1144, "Therapeutic optimization in neuropsychopharmacology", Paris, F-75006, France
- Université Paris Cité, Inserm UMR-S1144, Paris, F-75006, France
| | - Alexandre Mourot
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
- Brain Plasticity Unit, CNRS UMR 8249, ESPCI Paris, PSL Research University, 75005, Paris, France
| | - Philibert Duriez
- GHU Paris Psychiatrie et Neurosciences (CMME, Hospital Sainte-Anne), Institute of Psychiatry and Neuroscience of Paris (INSERM UMR1266), Paris, France
| | - Leora Pinhas
- PHLIP Mental Health and Painless Medicine clinic, Toronto, Canada
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Nicolas Pietrancosta
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
- Sorbonne Université, École normale supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005, Paris, France
- LCBPT, Université Paris Descartes, Sorbonne Paris Cité, UMR 8601, CNRS, Paris, 75006, France
| | - Stéphanie Daumas
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Salah El Mestikawy
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada.
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France.
| |
Collapse
|
19
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 PMCID: PMC11694735 DOI: 10.1016/j.neuro.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
20
|
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
|
21
|
Fox ME, Montemarano A, Ostman AE, Basu M, Herb B, Ament SA, Fox LD. Transcriptional signatures of fentanyl use in the mouse ventral tegmental area. Addict Biol 2024; 29:e13403. [PMID: 38735880 PMCID: PMC11089014 DOI: 10.1111/adb.13403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024]
Abstract
Synthetic opioids such as fentanyl contribute to the vast majority of opioid-related overdose deaths, but fentanyl use remains broadly understudied. Like other substances with misuse potential, opioids cause lasting molecular adaptations to brain reward circuits, including neurons in the ventral tegmental area (VTA). The VTA contains numerous cell types that play diverse roles in opioid use and relapse; however, it is unknown how fentanyl experience alters the transcriptional landscape in specific subtypes. Here, we performed single nuclei RNA sequencing to study transcriptional programs in fentanyl-experienced mice. Male and female C57/BL6 mice self-administered intravenous fentanyl (1.5 μg/kg/infusion) or saline for 10 days. After 24 h abstinence, VTA nuclei were isolated and prepared for sequencing on the 10× platform. We identified different patterns of gene expression across cell types. In dopamine neurons, we found enrichment of genes involved in growth hormone signalling. In dopamine-glutamate-GABA combinatorial neurons, and some GABA neurons, we found enrichment of genes involved in Pi3k-Akt signalling. In glutamate neurons, we found enrichment of genes involved in cholinergic signalling. We identified transcriptional regulators for the differentially expressed genes in each neuron cluster, including downregulated transcriptional repressor Bcl6, and upregulated transcription factor Tcf4. We also compared the fentanyl-induced gene expression changes identified in mouse VTA with a published rat dataset in bulk VTA, and found overlap in genes related to GABAergic signalling and extracellular matrix interaction. Together, we provide a comprehensive picture of how fentanyl self-administration alters the transcriptional landscape of the mouse VTA that serves as the foundation for future mechanistic studies.
Collapse
Affiliation(s)
- Megan E. Fox
- Department of Anesthesiology and Perioperative MedicineThe Pennsylvania State University College of MedicineHersheyPennsylvaniaUSA
- Department of PharmacologyThe Pennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Annalisa Montemarano
- Department of Anesthesiology and Perioperative MedicineThe Pennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Alexandria E. Ostman
- Department of Anesthesiology and Perioperative MedicineThe Pennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| | - Mahashweta Basu
- Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Brian Herb
- Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Seth A. Ament
- Institute for Genome SciencesUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Logan D. Fox
- Department of Anesthesiology and Perioperative MedicineThe Pennsylvania State University College of MedicineHersheyPennsylvaniaUSA
| |
Collapse
|
22
|
Bertho M, Caldeira V, Hsu LJ, Löw P, Borgius L, Kiehn O. Excitatory Spinal Lhx9-Derived Interneurons Modulate Locomotor Frequency in Mice. J Neurosci 2024; 44:e1607232024. [PMID: 38438260 PMCID: PMC11063822 DOI: 10.1523/jneurosci.1607-23.2024] [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: 08/09/2023] [Revised: 01/18/2024] [Accepted: 02/15/2024] [Indexed: 03/06/2024] Open
Abstract
Locomotion allows us to move and interact with our surroundings. Spinal networks that control locomotion produce rhythm and left-right and flexor-extensor coordination. Several glutamatergic populations, Shox2 non-V2a, Hb9-derived interneurons, and, recently, spinocerebellar neurons have been proposed to be involved in the mouse rhythm generating networks. These cells make up only a smaller fraction of the excitatory cells in the ventral spinal cord. Here, we set out to identify additional populations of excitatory spinal neurons that may be involved in rhythm generation or other functions in the locomotor network. We use RNA sequencing from glutamatergic, non-glutamatergic, and Shox2 cells in the neonatal mice from both sexes followed by differential gene expression analyses. These analyses identified transcription factors that are highly expressed by glutamatergic spinal neurons and differentially expressed between Shox2 neurons and glutamatergic neurons. From this latter category, we identified the Lhx9-derived neurons as having a restricted spinal expression pattern with no Shox2 neuron overlap. They are purely glutamatergic and ipsilaterally projecting. Ablation of the glutamatergic transmission or acute inactivation of the neuronal activity of Lhx9-derived neurons leads to a decrease in the frequency of locomotor-like activity without change in coordination pattern. Optogenetic activation of Lhx9-derived neurons promotes locomotor-like activity and modulates the frequency of the locomotor activity. Calcium activities of Lhx9-derived neurons show strong left-right out-of-phase rhythmicity during locomotor-like activity. Our study identifies a distinct population of spinal excitatory neurons that regulates the frequency of locomotor output with a suggested role in rhythm-generation in the mouse alongside other spinal populations.
Collapse
Affiliation(s)
- Maëlle Bertho
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Vanessa Caldeira
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Li-Ju Hsu
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Peter Löw
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Lotta Borgius
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ole Kiehn
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
23
|
Kampmann M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat Rev Neurosci 2024; 25:351-371. [PMID: 38575768 DOI: 10.1038/s41583-024-00806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 04/06/2024]
Abstract
The selective vulnerability of specific neuronal subtypes is a hallmark of neurodegenerative diseases. In this Review, I summarize our current understanding of the brain regions and cell types that are selectively vulnerable in different neurodegenerative diseases and describe the proposed underlying cell-autonomous and non-cell-autonomous mechanisms. I highlight how recent methodological innovations - including single-cell transcriptomics, CRISPR-based screens and human cell-based models of disease - are enabling new breakthroughs in our understanding of selective vulnerability. An understanding of the molecular mechanisms that determine selective vulnerability and resilience would shed light on the key processes that drive neurodegeneration and point to potential therapeutic strategies to protect vulnerable cell populations.
Collapse
Affiliation(s)
- Martin Kampmann
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Valyear MD, Brown A, Deyab G, Villaruel FR, Lahlou S, Caporicci-Dinucci N, Chaudhri N. Augmenting glutamatergic, but not dopaminergic, activity in the nucleus accumbens shell disrupts responding to a discrete alcohol cue in an alcohol context. Eur J Neurosci 2024; 59:1500-1518. [PMID: 38185906 DOI: 10.1111/ejn.16231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024]
Abstract
Discrete alcohol cues and contexts are relapse triggers for people with alcohol use disorder exerting particularly powerful control over behaviour when they co-occur. Here, we investigated the neural substrates subserving the capacity for alcohol-associated contexts to elevate responding to an alcohol-predictive conditioned stimulus (CS). Specifically, rats were trained in a distinct 'alcohol context' to respond by entering a fluid port during a discrete auditory CS that predicted the delivery of alcohol and were familiarized with a 'neutral context' wherein alcohol was never available. When conditioned CS responding was tested by presenting the CS without alcohol, we found that augmenting glutamatergic activity in the nucleus accumbens (NAc) shell by microinfusing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) reduced responding to an alcohol CS in an alcohol, but not neutral, context. Further, AMPA microinfusion robustly affected behaviour, attenuating the number, duration and latency of CS responses selectively in the alcohol context. Although dopaminergic inputs to the NAc shell were previously shown to be necessary for CS responding in an alcohol context, here, chemogenetic excitation of ventral tegmental area (VTA) dopamine neurons and their inputs to the NAc shell did not affect CS responding. Critically, chemogenetic excitation of VTA dopamine neurons affected feeding behaviour and elevated c-fos immunoreactivity in the VTA and NAc shell, validating the chemogenetic approach. These findings enrich our understanding of the substrates underlying Pavlovian responding for alcohol and reveal that the capacity for contexts to modulate responding to discrete alcohol cues is delicately underpinned by the NAc shell.
Collapse
Affiliation(s)
- Milan D Valyear
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Alexa Brown
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Ghislaine Deyab
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Franz R Villaruel
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Soraya Lahlou
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nina Caporicci-Dinucci
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Nadia Chaudhri
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Izowit G, Walczak M, Drwięga G, Solecki W, Błasiak T. Brain state-dependent responses of midbrain dopaminergic neurons to footshock under urethane anaesthesia. Eur J Neurosci 2024; 59:1536-1557. [PMID: 38233998 DOI: 10.1111/ejn.16252] [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: 07/21/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
For a long time, it has been assumed that dopaminergic (DA) neurons in both the ventral tegmental area (VTA) and the substantia nigra pars compacta (SNc) uniformly respond to rewarding and aversive stimuli by either increasing or decreasing their activity, respectively. This response was believed to signal information about the perceived stimuli's values. The identification of VTA&SNc DA neurons that are excited by both rewarding and aversive stimuli has led to the categorisation of VTA&SNc DA neurons into two subpopulations: one signalling the value and the other signalling the salience of the stimuli. It has been shown that the general state of the brain can modulate the electrical activity of VTA&SNc DA neurons, but it remains unknown whether this factor may also influence responses to aversive stimuli, such as a footshock (FS). To address this question, we have recorded the responses of VTA&SNc DA neurons to FSs across cortical activation and slow wave activity brain states in urethane-anaesthetised rats. Adding to the knowledge of aversion signalling by midbrain DA neurons, we report that significant proportion of VTA&SNc DA neurons can change their responses to an aversive stimulus in a brain state-dependent manner. The majority of these neurons decreased their activity in response to FS during cortical activation but switched to increasing it during slow wave activity. It can be hypothesised that this subpopulation of DA neurons may be involved in the 'dual signalling' of both the value and the salience of the stimuli, depending on the general state of the brain.
Collapse
Affiliation(s)
- Gabriela Izowit
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Cracow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Cracow, Poland
| | - Magdalena Walczak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Cracow, Poland
| | - Gniewosz Drwięga
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Cracow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Cracow, Poland
| | - Wojciech Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Cracow, Poland
| | - Tomasz Błasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Cracow, Poland
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Warlow SM, Singhal SM, Hollon NG, Faget L, Dowlat DS, Zell V, Hunker AC, Zweifel LS, Hnasko TS. Mesoaccumbal glutamate neurons drive reward via glutamate release but aversion via dopamine co-release. Neuron 2024; 112:488-499.e5. [PMID: 38086374 PMCID: PMC10922836 DOI: 10.1016/j.neuron.2023.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/22/2023] [Accepted: 11/06/2023] [Indexed: 02/10/2024]
Abstract
Ventral tegmental area (VTA) projections to the nucleus accumbens (NAc) drive reward-related motivation. Although dopamine neurons are predominant, a substantial glutamatergic projection is also present, and a subset of these co-release both dopamine and glutamate. Optogenetic stimulation of VTA glutamate neurons not only supports self-stimulation but can also induce avoidance behavior, even in the same assay. Here, we parsed the selective contribution of glutamate or dopamine co-release from VTA glutamate neurons to reinforcement and avoidance. We expressed channelrhodopsin-2 (ChR2) in mouse VTA glutamate neurons in combination with CRISPR-Cas9 to disrupt either the gene encoding vesicular glutamate transporter 2 (VGLUT2) or tyrosine hydroxylase (Th). Selective disruption of VGLUT2 abolished optogenetic self-stimulation but left real-time place avoidance intact, whereas CRISPR-Cas9 deletion of Th preserved self-stimulation but abolished place avoidance. Our results demonstrate that glutamate release from VTA glutamate neurons is positively reinforcing but that dopamine release from VTA glutamate neurons can induce avoidance behavior.
Collapse
Affiliation(s)
- Shelley M Warlow
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Sarthak M Singhal
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Nick G Hollon
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Lauren Faget
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Dina S Dowlat
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Vivien Zell
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Avery C Hunker
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
29
|
Li M, Yang G. A mesocortical glutamatergic pathway modulates neuropathic pain independent of dopamine co-release. Nat Commun 2024; 15:643. [PMID: 38245542 PMCID: PMC10799877 DOI: 10.1038/s41467-024-45035-2] [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: 07/03/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
Dysfunction in the mesocortical pathway, connecting the ventral tegmental area (VTA) to the prefrontal cortex, has been implicated in chronic pain. While extensive research has focused on the role of dopamine, the contribution of glutamatergic signaling in pain modulation remains unknown. Using in vivo calcium imaging, we observe diminished VTA glutamatergic activity targeting the prelimbic cortex (PL) in a mouse model of neuropathic pain. Optogenetic activation of VTA glutamatergic terminals in the PL alleviates neuropathic pain, whereas inhibiting these terminals in naïve mice induces pain-like responses. Importantly, this pain-modulating effect is independent of dopamine co-release, as demonstrated by CRISPR/Cas9-mediated gene deletion. Furthermore, we show that VTA neurons primarily project to excitatory neurons in the PL, and their activation restores PL outputs to the anterior cingulate cortex, a key region involved in pain processing. These findings reveal a distinct mesocortical glutamatergic pathway that critically modulates neuropathic pain independent of dopamine signaling.
Collapse
Affiliation(s)
- Miao Li
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
30
|
Fox ME, Montemarano A, Ostman AE, Basu M, Herb B, Ament SA, Fox LD. Transcriptional signatures of fentanyl use in the mouse ventral tegmental area. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572172. [PMID: 38187661 PMCID: PMC10769205 DOI: 10.1101/2023.12.18.572172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Synthetic opioids such as fentanyl contribute to the vast majority of opioid-related overdose deaths, but fentanyl use remains broadly understudied. Like other substances with misuse potential, opioids cause lasting molecular adaptations to brain reward circuits, including neurons in the ventral tegmental area (VTA). The VTA contains numerous cell types that play diverse roles in opioid use and relapse, however it is unknown how fentanyl experience alters the transcriptional landscape in specific subtypes. Here, we performed single nuclei RNA sequencing to study transcriptional programs in fentanyl experienced mice. Male and female C57/BL6 mice self-administered intravenous fentanyl (1.5 µg/kg/infusion) or saline for 10 days. After 24 hr abstinence, VTA nuclei were isolated and prepared for sequencing on the 10X platform. We identified different patterns of gene expression across cell types. In dopamine neurons, we found enrichment of genes involved in growth hormone signaling. In dopamine-glutamate-GABA combinatorial neurons, and some GABA neurons, we found enrichment of genes involved in Pi3k-Akt signaling. In glutamate neurons, we found enrichment of genes involved in cholinergic signaling. We identified transcriptional regulators for the differentially expressed genes in each neuron cluster, including downregulation of transcriptional repressor Bcl6, and upregulation of Wnt signaling partner Tcf4. We also compared the fentanyl-induced gene expression changes identified in mouse VTA with a published rat dataset in bulk VTA, and found overlap in genes related to GABAergic signaling and extracellular matrix interaction. Together, we provide a comprehensive picture of how fentanyl self-administration alters the transcriptional landscape of the mouse VTA, that serves for the foundation for future mechanistic studies.
Collapse
|
31
|
Elhadi K, Daiwile AP, Cadet JL. Modeling methamphetamine use disorder and relapse in animals: short- and long-term epigenetic, transcriptional., and biochemical consequences in the rat brain. Neurosci Biobehav Rev 2023; 155:105440. [PMID: 38707245 PMCID: PMC11068368 DOI: 10.1016/j.neubiorev.2023.105440] [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: 07/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 05/07/2024]
Abstract
Methamphetamine use disorder (MUD) is a neuropsychiatric disorder characterized by binge drug taking episodes, intervals of abstinence, and relapses to drug use even during treatment. MUD has been modeled in rodents and investigators are attempting to identify its molecular bases. Preclinical experiments have shown that different schedules of methamphetamine self-administration can cause diverse transcriptional changes in the dorsal striatum of Sprague-Dawley rats. In the present review, we present data on differentially expressed genes (DEGs) identified in the rat striatum following methamphetamine intake. These include genes involved in transcription regulation, potassium channel function, and neuroinflammation. We then use the striatal data to discuss the potential significance of the molecular changes induced by methamphetamine by reviewing concordant or discordant data from the literature. This review identified potential molecular targets for pharmacological interventions. Nevertheless, there is a need for more research on methamphetamine-induced transcriptional consequences in various brain regions. These data should provide a more detailed neuroanatomical map of methamphetamine-induced changes and should better inform therapeutic interventions against MUD.
Collapse
Affiliation(s)
- Khalid Elhadi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Atul P. Daiwile
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| |
Collapse
|
32
|
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
|
33
|
Kimble L, Twiddy JS, Berger JM, Forderhase AG, McCarty GS, Meitzen J, Sombers LA. Simultaneous, Real-Time Detection of Glutamate and Dopamine in Rat Striatum Using Fast-Scan Cyclic Voltammetry. ACS Sens 2023; 8:4091-4100. [PMID: 37962541 PMCID: PMC10683757 DOI: 10.1021/acssensors.3c01267] [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: 06/23/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023]
Abstract
Glutamate and dopamine (DA) represent two key contributors to striatal functioning, a region of the brain that is essential to motor coordination and motivated behavior. While electroanalytical techniques can be utilized for rapid, spatially resolved detection of DA in the interferent-rich brain environment, glutamate, a nonelectroactive analyte, cannot be directly detected using electroanalytical techniques. However, it can be probed using enzyme-based sensors, which generate an electroactive reporter in the presence of glutamate. The vast majority of glutamate biosensors have relied on amperometric sensing, which is an inherently nonselective detection technique. This approach necessitates the use of complex and performance-limiting modifications to ensure the desired single-analyte specificity. Here, we present a novel glutamate microbiosensor fabricated on a carbon-fiber microelectrode substrate and coupled with fast-scan cyclic voltammetry (FSCV) to enable the simultaneous quantification of glutamate and DA at single recording sites in the brain, which is impossible when using typical amperometric approaches. The glutamate microbiosensors were characterized for sensitivity, stability, and selectivity by using a voltammetric waveform optimized for the simultaneous detection of both species. The applicability of these sensors for the investigation of neural circuits was validated in the rat ventral striatum. Electrically evoked glutamate and DA release were recorded at single-micrometer-scale locations before and after pharmacological manipulation of glutamatergic signaling. Our novel glutamate microbiosensor advances the state of the art by providing a powerful tool for probing coordination between these two species in a way that has previously not been possible.
Collapse
Affiliation(s)
- Laney
C. Kimble
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Jack S. Twiddy
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
- Joint
Department of Biomedical Engineering, North
Carolina State University and University of North Carolina at Chapel
Hill, Raleigh, North Carolina 27695, United States
| | - Jenna M. Berger
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Alexandra G. Forderhase
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Gregory S. McCarty
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - John Meitzen
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Leslie A. Sombers
- Department
of Chemistry, Department of Biological Sciences, and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
34
|
Zell V, Teuns G, Needham AS, Mukherjee S, Roscoe N, Le M, Fourgeaud L, Woodruff G, Bhattacharya A, Marella M, Bonaventure P, Drevets WC, Balana B. Characterization of Selective M 5 Acetylcholine Muscarinic Receptor Modulators on Dopamine Signaling in the Striatum. J Pharmacol Exp Ther 2023; 387:226-234. [PMID: 37679045 DOI: 10.1124/jpet.123.001737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The type-5 muscarinic acetylcholine receptor (mAChR, M5) is almost exclusively expressed in dopamine (DA) neurons of the ventral tegmental area and substantia nigra pars compacta; therefore, they are ideally located to modulate DA signaling and underlying behaviors. However, the role of M5 in shaping DA release is still poorly characterized. In this study, we first quantitatively mapped the expression of M5 in different neurons of the mouse midbrain, then used voltammetry in mouse striatum to evaluate the effect of M5-selective modulators on DA release. The M5 negative allosteric modulator ML375 significantly decreased electrically evoked DA release and blocked the effect of Oxotremorine-M (Oxo-M; nonselective mAChR agonist) on DA release in the presence of an acetylcholine nicotinic receptor blocker. Conversely, the M5 positive allosteric modulator VU 0365114 significantly increased electrically evoked DA release and the Oxo-M effect on DA release. We then assessed M5's impact on mesolimbic circuit function in vivo. Although psychostimulant-induced locomotor activity models in knockout mice have previously been used to characterize the role of M5 in DA transmission, the results of these studies conflict, leading us to select a different in vivo model, namely a cocaine self-administration paradigm. In contrast to a previous study that also used this model, in the current study, administration of ML375 did not decrease cocaine self-administration in rats (using fixed and progressive ratio). These conflicting results illustrate the complexity of M5 modulation and the need to further characterize its involvement in the regulation of dopamine signaling, central to multiple neuropsychiatric diseases. SIGNIFICANCE STATEMENT: This work describes the type-5 muscarinic receptor (M5) pattern of expression within the midbrain as well as its physiological modulation by selective compounds at the axon terminal level in the striatum, where M5 directly shapes dopamine transmission. It offers the first direct readout of mesolimbic dopamine release modulation by M5, highlighting its role in regulating neurocircuits implicated in the pathophysiology of neuropsychiatric disorders such as substance use disorders, major depressive disorder, and schizophrenia.
Collapse
Affiliation(s)
- Vivien Zell
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Greetje Teuns
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Alexandra Stormy Needham
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Sruti Mukherjee
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Nathaniel Roscoe
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Michelle Le
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Lawrence Fourgeaud
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Grace Woodruff
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Anindya Bhattacharya
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Mathieu Marella
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Pascal Bonaventure
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Wayne C Drevets
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Bartosz Balana
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| |
Collapse
|
35
|
Matityahu L, Gilin N, Sarpong GA, Atamna Y, Tiroshi L, Tritsch NX, Wickens JR, Goldberg JA. Acetylcholine waves and dopamine release in the striatum. Nat Commun 2023; 14:6852. [PMID: 37891198 PMCID: PMC10611775 DOI: 10.1038/s41467-023-42311-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Striatal dopamine encodes reward, with recent work showing that dopamine release occurs in spatiotemporal waves. However, the mechanism of dopamine waves is unknown. Here we report that acetylcholine release in mouse striatum also exhibits wave activity, and that the spatial scale of striatal dopamine release is extended by nicotinic acetylcholine receptors. Based on these findings, and on our demonstration that single cholinergic interneurons can induce dopamine release, we hypothesized that the local reciprocal interaction between cholinergic interneurons and dopamine axons suffices to drive endogenous traveling waves. We show that the morphological and physiological properties of cholinergic interneuron - dopamine axon interactions can be modeled as a reaction-diffusion system that gives rise to traveling waves. Analytically-tractable versions of the model show that the structure and the nature of propagation of acetylcholine and dopamine traveling waves depend on their coupling, and that traveling waves can give rise to empirically observed correlations between these signals. Thus, our study provides evidence for striatal acetylcholine waves in vivo, and proposes a testable theoretical framework that predicts that the observed dopamine and acetylcholine waves are strongly coupled phenomena.
Collapse
Affiliation(s)
- Lior Matityahu
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Naomi Gilin
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Gideon A Sarpong
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yara Atamna
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Lior Tiroshi
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jeffery R Wickens
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Joshua A Goldberg
- Department of Medical Neurobiology, Institute of Medical Research Israel - Canada, The Faculty of Medicine, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel.
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
de Ceglia R, Ledonne A, Litvin DG, Lind BL, Carriero G, Latagliata EC, Bindocci E, Di Castro MA, Savtchouk I, Vitali I, Ranjak A, Congiu M, Canonica T, Wisden W, Harris K, Mameli M, Mercuri N, Telley L, Volterra A. Specialized astrocytes mediate glutamatergic gliotransmission in the CNS. Nature 2023; 622:120-129. [PMID: 37674083 PMCID: PMC10550825 DOI: 10.1038/s41586-023-06502-w] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/31/2023] [Indexed: 09/08/2023]
Abstract
Multimodal astrocyte-neuron communications govern brain circuitry assembly and function1. For example, through rapid glutamate release, astrocytes can control excitability, plasticity and synchronous activity2,3 of synaptic networks, while also contributing to their dysregulation in neuropsychiatric conditions4-7. For astrocytes to communicate through fast focal glutamate release, they should possess an apparatus for Ca2+-dependent exocytosis similar to neurons8-10. However, the existence of this mechanism has been questioned11-13 owing to inconsistent data14-17 and a lack of direct supporting evidence. Here we revisited the astrocyte glutamate exocytosis hypothesis by considering the emerging molecular heterogeneity of astrocytes18-21 and using molecular, bioinformatic and imaging approaches, together with cell-specific genetic tools that interfere with glutamate exocytosis in vivo. By analysing existing single-cell RNA-sequencing databases and our patch-seq data, we identified nine molecularly distinct clusters of hippocampal astrocytes, among which we found a notable subpopulation that selectively expressed synaptic-like glutamate-release machinery and localized to discrete hippocampal sites. Using GluSnFR-based glutamate imaging22 in situ and in vivo, we identified a corresponding astrocyte subgroup that responds reliably to astrocyte-selective stimulations with subsecond glutamate release events at spatially precise hotspots, which were suppressed by astrocyte-targeted deletion of vesicular glutamate transporter 1 (VGLUT1). Furthermore, deletion of this transporter or its isoform VGLUT2 revealed specific contributions of glutamatergic astrocytes in cortico-hippocampal and nigrostriatal circuits during normal behaviour and pathological processes. By uncovering this atypical subpopulation of specialized astrocytes in the adult brain, we provide insights into the complex roles of astrocytes in central nervous system (CNS) physiology and diseases, and identify a potential therapeutic target.
Collapse
Affiliation(s)
- Roberta de Ceglia
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Ada Ledonne
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - David Gregory Litvin
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Wyss Center for Bio and Neuro Engineering, Campus Biotech, Geneva, Switzerland
| | - Barbara Lykke Lind
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Giovanni Carriero
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | | | - Erika Bindocci
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | | | - Iaroslav Savtchouk
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Ilaria Vitali
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Anurag Ranjak
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Mauro Congiu
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Tara Canonica
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - William Wisden
- Department of Life Sciences and UK Dementia Research Institute, Imperial College London, London, UK
| | - Kenneth Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Manuel Mameli
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Nicola Mercuri
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Ludovic Telley
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland.
| | - Andrea Volterra
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland.
- Wyss Center for Bio and Neuro Engineering, Campus Biotech, Geneva, Switzerland.
| |
Collapse
|
39
|
Vrettou M, Thalhammer SB, Svensson AL, Dumas S, Nilsson KW, Wallén-Mackenzie Å, Fredriksson R, Nylander I, Comasco E. Vesicular glutamate transporter 2 expression in the ventral tegmental area of outbred male rats following exposure to nicotine and alcohol. DRUG AND ALCOHOL DEPENDENCE REPORTS 2023; 8:100180. [PMID: 37533815 PMCID: PMC10391930 DOI: 10.1016/j.dadr.2023.100180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023]
Abstract
Background Initiation of use/co-use of nicotine and alcohol, commonly occurring in an episodic manner during adolescence, can imprint vulnerability to the developing brain and lead to addiction. The ventral tegmental area (VTA) is a key heterogeneous region of the mesocorticolimbic circuit involved in the binge-drinking and intoxication step of the addiction circuit. Higher human post-mortem VTA expression of vesicular glutamate transporter 2 (VGLUT2), a marker of the glutamatergic phenotype also expressed in dopaminergic [Tyrosine Hydroxylase (Th)-positive] neurons, has been associated with chronic nicotine use and co-use with alcohol. Methods The present study aimed to map and characterize the Vglut2- and Th-expressing neurons in the VTA of adolescent male rats exposed or not to prolonged (six-weeks) episodic (three consecutive days/week) nicotine and/or alcohol administration. Nicotine (0.35 mg/kg free base) was injected subcutaneously, whereas alcohol (2 g/kg 20%) was administrated via gavage. Vglut2 and Th mRNA was assessed in the anterior and posterior VTA by use of in situ hybridization. Results The profile of neurons varied with substance-exposure among VTA subregions. Th-only expressing neurons were more abundant in the posterior VTA of the group exposed to nicotine-only, compared to controls. The same neurons were, on the contrary, less present in the anterior VTA of animals exposed to alcohol-only, who also displayed a higher number of Vglut2-expressing neurons in the lateral anterior VTA. Conclusions VTA Vglut2- and Th-only neurons seem differentially involved in the effects of adolescent episodic nicotine and alcohol exposure in the anterior and posterior VTA.
Collapse
Affiliation(s)
- Maria Vrettou
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Stefan Bernhard Thalhammer
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anne-Lie Svensson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - Kent W Nilsson
- Centre for Clinical Research Västerås, Uppsala University, Västmanland County Hospital Västerås, Sweden
| | | | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ingrid Nylander
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
40
|
Krok AC, Maltese M, Mistry P, Miao X, Li Y, Tritsch NX. Intrinsic dopamine and acetylcholine dynamics in the striatum of mice. Nature 2023; 621:543-549. [PMID: 37558873 PMCID: PMC11577287 DOI: 10.1038/s41586-023-05995-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/22/2023] [Indexed: 08/11/2023]
Abstract
External rewards such as food and money are potent modifiers of behaviour1,2. Pioneering studies established that these salient sensory stimuli briefly interrupt the tonic discharge of neurons that produce the neuromodulators dopamine (DA) and acetylcholine (ACh): midbrain DA neurons (DANs) fire a burst of action potentials that broadly elevates DA in the striatum3,4 at the same time that striatal cholinergic interneurons (CINs) produce a characteristic pause in firing5,6. These phasic responses are thought to create unique, temporally limited conditions that motivate action and promote learning7-11. However, the dynamics of DA and ACh outside explicitly rewarded situations remain poorly understood. Here we show that extracellular DA and ACh levels fluctuate spontaneously and periodically at a frequency of approximately 2 Hz in the dorsal striatum of mice and maintain the same temporal relationship relative to one another as that evoked by reward. We show that this neuromodulatory coordination does not arise from direct interactions between DA and ACh within the striatum. Instead, we provide evidence that periodic fluctuations in striatal DA are inherited from midbrain DANs, while striatal ACh transients are driven by glutamatergic inputs, which act to locally synchronize the spiking of CINs. Together, our findings show that striatal neuromodulatory dynamics are autonomously organized by distributed extra-striatal afferents. The dominance of intrinsic rhythms in DA and ACh offers new insights for explaining how reward-associated neural dynamics emerge and how the brain motivates action and promotes learning from within.
Collapse
Affiliation(s)
- Anne C Krok
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Marta Maltese
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Pratik Mistry
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Xiaolei Miao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
41
|
Priest MF, Freda SN, Rieth IJ, Badong D, Dumrongprechachan V, Kozorovitskiy Y. Peptidergic and functional delineation of the Edinger-Westphal nucleus. Cell Rep 2023; 42:112992. [PMID: 37594894 PMCID: PMC10512657 DOI: 10.1016/j.celrep.2023.112992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 06/15/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023] Open
Abstract
Many neuronal populations that release fast-acting excitatory and inhibitory neurotransmitters in the brain also contain slower-acting neuropeptides. These facultative peptidergic cell types are common, but it remains uncertain whether neurons that solely release peptides exist. Our fluorescence in situ hybridization, genetically targeted electron microscopy, and electrophysiological characterization suggest that most neurons of the non-cholinergic, centrally projecting Edinger-Westphal nucleus in mice are obligately peptidergic. We further show, using anterograde projection mapping, monosynaptic retrograde tracing, angled-tip fiber photometry, and chemogenetic modulation and genetically targeted ablation in conjunction with canonical assays for anxiety, that this peptidergic population activates in response to loss of motor control and promotes anxiety responses. Together, these findings elucidate an integrative, ethologically relevant role for the Edinger-Westphal nucleus and functionally align the nucleus with the periaqueductal gray, where it resides. This work advances our understanding of peptidergic modulation of anxiety and provides a framework for future investigations of peptidergic systems.
Collapse
Affiliation(s)
- Michael F Priest
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Isabelle J Rieth
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanna Badong
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
42
|
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
|
43
|
Ma S, Zhong H, Liu X, Wang L. Spatial Distribution of Neurons Expressing Single, Double, and Triple Molecular Characteristics of Glutamatergic, Dopaminergic, or GABAergic Neurons in the Mouse Ventral Tegmental Area. J Mol Neurosci 2023; 73:345-362. [PMID: 37243808 DOI: 10.1007/s12031-023-02121-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
The ventral tegmental area (VTA) is a heterogeneous midbrain area that plays a significant role in diverse neural processes such as reward, aversion, and motivation. The VTA contains three main neuronal populations, namely, dopamine (DA), γ-aminobutyric acid (GABA), and glutamate neurons, but some neurons exhibit combinatorial molecular characteristics of dopaminergic, GABAergic, or glutamatergic neurons. However, little information is available regarding detailed distribution of neurons with single, double, and triple molecular characteristics of glutamatergic, dopaminergic, or GABAergic neurons in mice. We present a topographical distribution map of three main neuronal populations expressing a single molecular characteristic of dopaminergic, GABAergic, or glutamatergic neurons, and four neuronal populations co-expressing double or triple molecular characteristics in combinatorial manners, in the mouse VTA, following analysis of triple fluorescent in situ hybridization for the simultaneous detection of tyrosine hydroxylase (TH, marker for dopaminergic neurons), vesicular glutamate transporter 2 (VGLUT2, marker for glutamatergic neurons), and glutamic acid decarboxylase 2 (GAD2, marker for GABAergic neurons) mRNA. We found that the vast majority of neurons expressed a single type of mRNA, and these neurons were intermingled with neurons co-expressing double or triple combinations of VGLUT2, TH, or GAD2 in the VTA. These seven neuronal populations were differentially distributed in the VTA sub-nuclei across the rostro-caudal and latero-medial axes. This histochemical study will lead to a deeper understanding of the complexity of neuronal molecular characteristics in different VTA sub-nuclei, and potentially facilitate clarification of diverse functions of the VTA.
Collapse
Affiliation(s)
- Shaohua Ma
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hao Zhong
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xuemei Liu
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Liping Wang
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
44
|
Zhao C, Wang C, Zhang H, Yan W. A mini-review of the role of vesicular glutamate transporters in Parkinson's disease. Front Mol Neurosci 2023; 16:1118078. [PMID: 37251642 PMCID: PMC10211467 DOI: 10.3389/fnmol.2023.1118078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/06/2023] [Indexed: 05/31/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease implicated in multiple interacting neurotransmitter pathways. Glutamate is the central excitatory neurotransmitter in the brain and plays critical influence in the control of neuronal activity. Impaired Glutamate homeostasis has been shown to be closely associated with PD. Glutamate is synthesized in the cytoplasm and stored in synaptic vesicles by vesicular glutamate transporters (VGLUTs). Following its exocytotic release, Glutamate activates Glutamate receptors (GluRs) and mediates excitatory neurotransmission. While Glutamate is quickly removed by excitatory amino acid transporters (EAATs) to maintain its relatively low extracellular concentration and prevent excitotoxicity. The involvement of GluRs and EAATs in the pathophysiology of PD has been widely studied, but little is known about the role of VGLUTs in the PD. In this review, we highlight the role of VGLUTs in neurotransmitter and synaptic communication, as well as the massive alterations in Glutamate transmission and VGLUTs levels in PD. Among them, adaptive changes in the expression level and function of VGLUTs may exert a crucial role in excitatory damage in PD, and VGLUTs are considered as novel potential therapeutic targets for PD.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Chunyu Wang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hainan Zhang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Weiqian Yan
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
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
|
46
|
Ma D, Herndon N, Le JQ, Abruzzi KC, Zinn K, Rosbash M. Neural connectivity molecules best identify the heterogeneous clock and dopaminergic cell types in the Drosophila adult brain. SCIENCE ADVANCES 2023; 9:eade8500. [PMID: 36812309 PMCID: PMC9946362 DOI: 10.1126/sciadv.ade8500] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/26/2023] [Indexed: 05/25/2023]
Abstract
Our recent single-cell sequencing of most adult Drosophila circadian neurons indicated notable and unexpected heterogeneity. To address whether other populations are similar, we sequenced a large subset of adult brain dopaminergic neurons. Their gene expression heterogeneity is similar to that of clock neurons, i.e., both populations have two to three cells per neuron group. There was also unexpected cell-specific expression of neuron communication molecule messenger RNAs: G protein-coupled receptor or cell surface molecule (CSM) transcripts alone can define adult brain dopaminergic and circadian neuron cell type. Moreover, the adult expression of the CSM DIP-beta in a small group of clock neurons is important for sleep. We suggest that the common features of circadian and dopaminergic neurons are general, essential for neuronal identity and connectivity of the adult brain, and that these features underlie the complex behavioral repertoire of Drosophila.
Collapse
Affiliation(s)
- Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Nicholas Herndon
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Katharine C. Abruzzi
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| |
Collapse
|
47
|
Hughes AC, Pollard BG, Xu B, Gammons JW, Chapman P, Bikoff JB, Schwarz LA. A Novel Single Vector Intersectional AAV Strategy for Interrogating Cellular Diversity and Brain Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527312. [PMID: 36798174 PMCID: PMC9934562 DOI: 10.1101/2023.02.07.527312] [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: 02/11/2023]
Abstract
As the discovery of cellular diversity in the brain accelerates, so does the need for functional tools that target cells based on multiple features, such as gene expression and projection target. By selectively driving recombinase expression in a feature-specific manner, one can utilize intersectional strategies to conditionally promote payload expression only where multiple features overlap. We developed Conditional Viral Expression by Ribozyme Guided Degradation (ConVERGD), a single-construct intersectional targeting strategy that combines a self-cleaving ribozyme with traditional FLEx switches. ConVERGD offers benefits over existing platforms, such as expanded intersectionality, the ability to accommodate larger and more complex payloads, and a vector design that is easily modified to better facilitate rapid toolkit expansion. To demonstrate its utility for interrogating neural circuitry, we employed ConVERGD to target an unexplored subpopulation of norepinephrine (NE)-producing neurons within the rodent locus coeruleus (LC) identified via single-cell transcriptomic profiling to co-express the stress-related endogenous opioid gene prodynorphin (Pdyn). These studies showcase ConVERGD as a versatile tool for targeting diverse cell types and reveal Pdyn-expressing NE+ LC neurons as a small neuronal subpopulation capable of driving anxiogenic behavioral responses in rodents.
Collapse
Affiliation(s)
- Alex C. Hughes
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Brittany G. Pollard
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Jesse W. Gammons
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
- Present address: Department of Pediatrics, Stanford University, Stanford, CA, 94305
| | - Phillip Chapman
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Jay B. Bikoff
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
| | - Lindsay A. Schwarz
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, 38105
- Lead contact
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Adamson A, Buck SA, Freyberg Z, De Miranda BR. Sex Differences in Dopaminergic Vulnerability to Environmental Toxicants - Implications for Parkinson's Disease. Curr Environ Health Rep 2022; 9:563-573. [PMID: 36201109 PMCID: PMC10201647 DOI: 10.1007/s40572-022-00380-6] [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] [Accepted: 08/24/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Sex dimorphism in Parkinson's disease (PD) is an ostensible feature of the neurological disorder, particularly as men are 1.5-2 times more likely to develop PD than women. Clinical features of the disease, such as presentation at onset, most prevalent symptoms, and response to treatment, are also affected by sex. Despite these well-known sex differences in PD risk and phenotype, the mechanisms that impart sex dimorphisms in PD remain poorly understood. RECENT FINDINGS As PD incidence is influenced by environmental factors, an intriguing pattern has recently emerged in research studies suggesting a male-specific vulnerability to dopaminergic neurodegeneration caused by neurotoxicant exposure, with relative protection in females. These new experimental data have uncovered potential mechanisms that provide clues to the source of sex differences in dopaminergic neurodegeneration and other PD pathology such as alpha-synuclein toxicity. In this review, we discuss the emerging evidence of increased male sensitivity to neurodegeneration from environmental exposures. We examine mechanisms underlying dopaminergic neurodegeneration and PD-related pathologies with evidence supporting the roles of estrogen, SRY expression, the vesicular glutamate transporter VGLUT2, and the microbiome as prospective catalysts for male vulnerability. We also highlight the importance of including sex as a biological variable, particularly when evaluating dopaminergic neurotoxicity in the context of PD.
Collapse
Affiliation(s)
- Ashley Adamson
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA
| | - Silas A Buck
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Center for Neuroscience, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, 1719 6th Ave South, CIRC 560, Birmingham, AL, 35294, USA.
| |
Collapse
|