1
|
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
|
2
|
Oppman AM, Paradee WJ, Narayanan NS, Kim YC. Generation and validation of a D1 dopamine receptor Flpo knock-in mouse. J Neurosci Methods 2024:110345. [PMID: 39701542 DOI: 10.1016/j.jneumeth.2024.110345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Dopamine is a powerful neuromodulator of diverse brain functions, including movement, motivation, reward, and cognition. D1-type dopamine receptors (D1DRs) are the most prevalently expressed dopamine receptors in the brain. Neurons expressing D1DRs are heterogeneous and involve several subpopulations. Although these neurons can be studied with BAC-transgenic rodents, these models have some limitations especially when considering their integration with conditional or intersectional genetic tools. NEW METHOD We developed a novel Drd1-P2A-Flpo (Drd1-Flpo) mouse line in which the Flpo gene was knocked in immediately after the Drd1 gene using CRISPR-Cas9. We validated the Drd1-Flpo line by confirming Flp expression and functionality specific to D1DR+ neurons with immunohistochemistry and in situ hybridization. COMPARISON WITH EXISTING METHODS The Drd1-Flpo line is a useful resource for studying subpopulations of D1DR+ neurons with intersectional genetic tools. CONCLUSIONS We demonstrated brain-wide GFP expression driven by Drd1-Flpo, suggesting that this mouse line may be useful for comprehensive anatomical and functional studies in many brain regions. The Drd1-Flpo model will advance the study of dopaminergic signaling by providing a new tool for investigating the diverse roles of D1DR+ neurons and their subpopulations in brain disease.
Collapse
Affiliation(s)
| | | | | | - Young-Cho Kim
- Department of Neurology, University of Iowa; The Iowa Neuroscience Institute.
| |
Collapse
|
3
|
Tolooshams B, Matias S, Wu H, Temereanca S, Uchida N, Murthy VN, Masset P, Ba D. Interpretable deep learning for deconvolutional analysis of neural signals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574379. [PMID: 38260512 PMCID: PMC10802267 DOI: 10.1101/2024.01.05.574379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The widespread adoption of deep learning to build models that capture the dynamics of neural populations is typically based on "black-box" approaches that lack an interpretable link between neural activity and network parameters. Here, we propose to apply algorithm unrolling, a method for interpretable deep learning, to design the architecture of sparse deconvolutional neural networks and obtain a direct interpretation of network weights in relation to stimulus-driven single-neuron activity through a generative model. We characterize our method, referred to as deconvolutional unrolled neural learning (DUNL), and show its versatility by applying it to deconvolve single-trial local signals across multiple brain areas and recording modalities. To exemplify use cases of our decomposition method, we uncover multiplexed salience and reward prediction error signals from midbrain dopamine neurons in an unbiased manner, perform simultaneous event detection and characterization in somatosensory thalamus recordings, and characterize the heterogeneity of neural responses in the piriform cortex and in the striatum during unstructured, naturalistic experiments. Our work leverages the advances in interpretable deep learning to gain a mechanistic understanding of neural activity.
Collapse
Affiliation(s)
- Bahareh Tolooshams
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge MA, 02138
- Computing + Mathematical Sciences, California Institute of Technology, Pasadena, CA, 91125
| | - Sara Matias
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, 02138
| | - Hao Wu
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, 02138
| | - Simona Temereanca
- Carney Institute for Brain Science, Brown University, Providence, RI, 02906
| | - Naoshige Uchida
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, 02138
| | - Venkatesh N. Murthy
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, 02138
| | - Paul Masset
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, 02138
- Department of Psychology, McGill University, Montréal QC, H3A 1G1
| | - Demba Ba
- Center for Brain Science, Harvard University, Cambridge MA, 02138
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge MA, 02138
- Kempner Institute for the Study of Natural & Artificial Intelligence, Harvard University, Cambridge MA, 02138
| |
Collapse
|
4
|
Brandner DD, Mashal MA, Grissom NM, Rothwell PE. Sex differences in morphine sensitivity of neuroligin-3 knockout mice. Psychopharmacology (Berl) 2024; 241:2431-2440. [PMID: 39083079 DOI: 10.1007/s00213-024-06660-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Sex has a strong influence on the prevalence and course of brain conditions, including autism spectrum disorders. The mechanistic basis for these sex differences remains poorly understood, due in part to historical bias in biomedical research favoring analysis of male subjects, and the exclusion of female subjects. For example, studies of male mice carrying autism-associated mutations in neuroligin-3 are over-represented in the literature, including our own prior work showing diminished responses to chronic morphine exposure in male neuroligin-3 knockout mice. We therefore studied how constitutive and conditional genetic knockout of neuroligin-3 affects morphine sensitivity of female mice, using locomotor activity as a proxy for differences in opioid sensitivity that may be related to the pathophysiology and treatment of autism spectrum disorders. In contrast to male mice, female neuroligin-3 knockout mice showed normal psychomotor sensitization after chronic morphine exposure. However, in the absence of neuroligin-3 expression, both female and male mice show a similar change in the topography of locomotor stimulation produced by morphine. Conditional genetic deletion of neuroligin-3 from dopamine neurons increased the locomotor response of female mice to high doses of morphine, contrasting with the decrease in psychomotor sensitization caused by the same manipulation in male mice. Together, our data reveal that knockout of neuroligin-3 has both common and distinct effects on morphine sensitivity in female and male mice. These results also support the notion that female sex can confer resilience against the impact of autism-associated gene variants.
Collapse
Affiliation(s)
- Dieter D Brandner
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN, USA
| | - Mohammed A Mashal
- Department of Neuroscience, University of Minnesota, 4-142 Wallin Medical Biosciences Building, 2101 6 Street SE, Minneapolis, MN, 55455, USA
| | - Nicola M Grissom
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Patrick E Rothwell
- Department of Neuroscience, University of Minnesota, 4-142 Wallin Medical Biosciences Building, 2101 6 Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
5
|
López RC, Noble N, Özçete ÖD, Cai X, Handy GE, Andersen JW, Patriarchi T, Li Y, Kaeser PS. Innervation density governs crosstalk of GPCR-based norepinephrine and dopamine sensors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624963. [PMID: 39605389 PMCID: PMC11601633 DOI: 10.1101/2024.11.23.624963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
GPCR-based fluorescent sensors are widely used to correlate neuromodulatory signaling with brain function. While experiments in transfected cells often reveal selectivity for individual neurotransmitters, sensor specificity in the brain frequently remains uncertain. Pursuing experiments in brain slices and in vivo, we find that norepinephrine and dopamine cross-activate the respective sensors. Non-specific activation occurred when innervation of the cross-reacting transmitter was high, and silencing of specific innervation was indispensable for interpreting sensor fluorescence.
Collapse
Affiliation(s)
- Ricardo C. López
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Natalie Noble
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Özge D. Özçete
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Xintong Cai
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Gillian E. Handy
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | | | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH and University of Zürich, Zürich, Switzerland
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Pascal S. Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
6
|
Lazaridis I, Crittenden JR, Ahn G, Hirokane K, Wickersham IR, Yoshida T, Mahar A, Skara V, Loftus JH, Parvataneni K, Meletis K, Ting JT, Hueske E, Matsushima A, Graybiel AM. Striosomes control dopamine via dual pathways paralleling canonical basal ganglia circuits. Curr Biol 2024; 34:5263-5283.e8. [PMID: 39447573 DOI: 10.1016/j.cub.2024.09.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024]
Abstract
Balanced activity of canonical direct D1 and indirect D2 basal ganglia pathways is considered a core requirement for normal movement, and their imbalance is an etiologic factor in movement and neuropsychiatric disorders. We present evidence for a conceptually equivalent pair of direct D1 and indirect D2 pathways that arise from striatal projection neurons (SPNs) of the striosome compartment rather than from SPNs of the matrix, as do the canonical pathways. These striosomal D1 (S-D1) and D2 (S-D2) pathways target substantia nigra dopamine-containing neurons instead of basal ganglia motor output nuclei. They modulate movement with net effects opposite to those exerted by the canonical pathways: S-D1 is net inhibitory and S-D2 is net excitatory. The S-D1 and S-D2 circuits likely influence motivation for learning and action, complementing and reorienting canonical pathway modulation. A major conceptual reformulation of the classic direct-indirect pathway model of basal ganglia function is needed, as well as reconsideration of the effects of D2-targeting therapeutic drugs.
Collapse
Affiliation(s)
- Iakovos Lazaridis
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Jill R Crittenden
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gun Ahn
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kojiro Hirokane
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomoko Yoshida
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ara Mahar
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vasiliki Skara
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Johnny H Loftus
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Krishna Parvataneni
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Jonathan T Ting
- Human Cell Types Department, Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Emily Hueske
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ayano Matsushima
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
7
|
Clark DN, Brown SV, Xu L, Lee RL, Ragusa JV, Xu Z, Milner JD, Filiano AJ. Prolonged STAT1 signaling in neurons causes hyperactive behavior. Brain Behav Immun 2024; 124:1-8. [PMID: 39542073 DOI: 10.1016/j.bbi.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
The interferon (IFN)-induced STAT1 signaling pathway is a canonical immune pathway that has also been implicated in regulating neuronal activity. The pathway is enriched in brains of individuals with autism spectrum disorder (ASD) and schizophrenia (SZ). Over-activation of the STAT1 pathway causes pathological transcriptional responses, however it is unclear how these responses might translate into behavioral phenotypes. We hypothesized that prolonged STAT1 signaling in neurons would be sufficient to cause behavioral deficits associated with neurodevelopmental disorders. In this study, we developed a novel mouse model with the clinical STAT1 gain-of-function mutation, T385M, in neurons. These mice were hyperactive and displayed neural hypoactivity with less neuron counts in the caudate putamen. Driving the STAT1 gain-of-function mutation exclusively in dopaminergic neurons, which project to the caudate putamen of the dorsal striatum, mimicked some hyperactive behaviors without a reduction of neurons. Moreover, we demonstrated that this phenotype is neuron specific, as mice with prolonged STAT1 signaling in all excitatory or inhibitory neurons or in microglia were not hyperactive. Overall, these findings suggest that STAT1 signaling in neurons is a crucial player in regulating striatal neuron activity and aspects of motor behavior.
Collapse
Affiliation(s)
- Danielle N Clark
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Shelby V Brown
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Rae-Ling Lee
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Joey V Ragusa
- Department of Pathology, Duke University, Durham, NC, USA
| | - Zhenghao Xu
- Marcus Center for Cellular Cures, Duke University, Durham, NC, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Anthony J Filiano
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA; Marcus Center for Cellular Cures, Duke University, Durham, NC, USA; Department of Pathology, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Kamath T, Lodder B, Bilsel E, Green I, Dalangin R, Capelli P, Raghubardayal M, Legister J, Hulshof L, Wallace JB, Tian L, Uchida N, Watabe-Uchida M, Sabatini BL. Hunger modulates exploration through suppression of dopamine signaling in the tail of striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622990. [PMID: 39713287 PMCID: PMC11661229 DOI: 10.1101/2024.11.11.622990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Caloric depletion leads to behavioral changes that help an animal find food and restore its homeostatic balance. Hunger increases exploration and risk-taking behavior, allowing an animal to forage for food despite risks; however, the neural circuitry underlying this change is unknown. Here, we characterize how hunger restructures an animal's spontaneous behavior as well as its directed exploration of a novel object. We show that hunger-induced changes in exploration are accompanied by and result from modulation of dopamine signaling in the tail of the striatum (TOS). Dopamine signaling in the TOS is modulated by internal hunger state through the activity of agouti-related peptide (AgRP) neurons, putative "hunger neurons" in the arcuate nucleus of the hypothalamus. These AgRP neurons are poly-synaptically connected to TOS-projecting dopaminergic neurons through the lateral hypothalamus, the central amygdala, and the periaqueductal grey. We thus delineate a hypothalamic-midbrain circuit that coordinates changes in exploration behavior in the hungry state.
Collapse
|
9
|
Cai X, Liu C, Tsutsui-Kimura I, Lee JH, Guo C, Banerjee A, Lee J, Amo R, Xie Y, Patriarchi T, Li Y, Watabe-Uchida M, Uchida N, Kaeser PS. Dopamine dynamics are dispensable for movement but promote reward responses. Nature 2024; 635:406-414. [PMID: 39415006 DOI: 10.1038/s41586-024-08038-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
Dopamine signalling modes differ in kinetics and spatial patterns of receptor activation1,2. How these modes contribute to motor function, motivation and learning has long been debated3-21. Here we show that action-potential-induced dopamine release is dispensable for movement initiation but supports reward-oriented behaviour. We generated mice with dopamine-neuron-specific knockout of the release site organizer protein RIM to disrupt action-potential-induced dopamine release. In these mice, rapid in vivo dopamine dynamics were strongly impaired, but baseline dopamine persisted and fully supported spontaneous movement. Conversely, reserpine-mediated dopamine depletion or blockade of dopamine receptors disrupted movement initiation. The dopamine precursor L-DOPA reversed reserpine-induced bradykinesia without restoring fast dopamine dynamics, a result that substantiated the conclusion that these dynamics are dispensable for movement initiation. In contrast to spontaneous movement, reward-oriented behaviour was impaired in dopamine-neuron-specific RIM knockout mice. In conditioned place preference and two-odour discrimination tasks, the mice effectively learned to distinguish the cues, which indicates that reward-based learning persists after RIM ablation. However, the performance vigour was reduced. During probabilistic cue-reward association, dopamine dynamics and conditioned responses assessed through anticipatory licking were disrupted. These results demonstrate that action-potential-induced dopamine release is dispensable for motor function and subsecond precision of movement initiation but promotes motivation and performance during reward-guided behaviours.
Collapse
Affiliation(s)
- Xintong Cai
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Iku Tsutsui-Kimura
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Joon-Hyuk Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Chong Guo
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Jinoh Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Yudi Xie
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Pomrenze MB, Vaillancourt S, Salgado JS, Raymond KB, Llorach P, Touponse GC, Cardozo Pinto DF, Rastegar Z, Casey AB, Eshel N, Malenka RC, Heifets BD. 5-HT 2C receptors in the nucleus accumbens constrain the rewarding effects of MDMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619256. [PMID: 39484424 PMCID: PMC11527024 DOI: 10.1101/2024.10.20.619256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
MDMA is a promising adjunct to psychotherapy and has well-known abuse liability, although less than other amphetamine analogs. While the reinforcing dopamine (DA)-releasing properties of MDMA are on par with methamphetamine (METH), MDMA is a far more potent serotonin (5-HT) releaser, via the 5-HT transporter (SERT). MDMA-mediated 5-HT release in a major reward center, the nucleus accumbens (NAc), drives prosocial behaviors via 5-HT1BR activation. We hypothesized that this prosocial mechanism contributes to the reduced reinforcing properties of MDMA compared to METH and used a platform of assays to predict the balance of prosocial and abuse-linked effects of (R)-MDMA, a novel entactogen in clinical development. NAc DA release, measured by GRAB-DA photometry in vivo, increased in proportion to MDMA (7.5 and 15 mg/kg, i.p.) and METH (2 mg/kg i.p.)-conditioned place preference (CPP). Using conditional knockouts (cKOs) for DAT and SERT, microdialysis, and photometry, we found that MDMA-released 5-HT limited MDMA-released DA through actions in the NAc, rather than at ventral tegmental area DAergic cell bodies. SERT cKO reduced the MDMA dose required for CPP three-fold. This enhanced MDMA-CPP and increased DA release were replicated by intra-NAc infusion of either a 5-HT reuptake inhibitor (escitalopram) to prevent MDMA interaction with SERT, or a 5-HT2CR antagonist (SB242084), but not by the 5-HT1BR antagonist NAS-181. These data support separate mechanisms for the low abuse potential versus prosocial effect of MDMA. Using this platform of assays, (R)-MDMA is predicted to have prosocial effects and low abuse potential.
Collapse
Affiliation(s)
- Matthew B. Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Sam Vaillancourt
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Juliana S. Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kendall B. Raymond
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Pierre Llorach
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Gavin C. Touponse
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Daniel F. Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Zahra Rastegar
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Austen B. Casey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Neir Eshel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Robert C. Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Boris D. Heifets
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
11
|
Pollock TA, Margetts AV, Vilca SJ, Tuesta LM. Cocaine taking and craving produce distinct transcriptional profiles in dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617923. [PMID: 39416214 PMCID: PMC11482921 DOI: 10.1101/2024.10.11.617923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Dopamine (DA) signaling plays an essential role in reward valence attribution and in encoding the reinforcing properties of natural and artificial rewards. The adaptive responses from midbrain dopamine neurons to artificial rewards such as drugs of abuse are therefore important for understanding the development of substance use disorders. Drug-induced changes in gene expression are one such adaptation that can determine the activity of dopamine signaling in projection regions of the brain reward system. One of the major challenges to obtaining this understanding involves the complex cellular makeup of the brain, where each neuron population can be defined by a distinct transcriptional profile. To bridge this gap, we have adapted a virus-based method for labeling and capture of dopamine nuclei, coupled with nuclear RNA-sequencing, to study the transcriptional adaptations, specifically, of dopamine neurons in the ventral tegmental area (VTA) during cocaine taking and cocaine craving, using a mouse model of cocaine intravenous self-administration (IVSA). Our results show significant changes in gene expression across non-drug operant training, cocaine taking, and cocaine craving, highlighted by an enrichment of repressive epigenetic modifying enzyme gene expression during cocaine craving. Immunohistochemical validation further revealed an increase of H3K9me3 deposition in DA neurons during cocaine craving. These results demonstrate that cocaine-induced transcriptional adaptations in dopamine neurons vary by phase of self-administration and underscore the utility of this approach for identifying relevant phase-specific molecular targets to study the behavioral course of substance use disorders.
Collapse
Affiliation(s)
- Tate A. Pollock
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Alexander V. Margetts
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Samara J. Vilca
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Luis M. Tuesta
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
12
|
Geibl FF, Henrich MT, Xie Z, Zampese E, Ueda J, Tkatch T, Wokosin DL, Nasiri E, Grotmann CA, Dawson VL, Dawson TM, Chandel NS, Oertel WH, Surmeier DJ. α-Synuclein pathology disrupts mitochondrial function in dopaminergic and cholinergic neurons at-risk in Parkinson's disease. Mol Neurodegener 2024; 19:69. [PMID: 39379975 PMCID: PMC11462807 DOI: 10.1186/s13024-024-00756-2] [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: 12/15/2023] [Accepted: 09/18/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Pathological accumulation of aggregated α-synuclein (aSYN) is a common feature of Parkinson's disease (PD). However, the mechanisms by which intracellular aSYN pathology contributes to dysfunction and degeneration of neurons in the brain are still unclear. A potentially relevant target of aSYN is the mitochondrion. To test this hypothesis, genetic and physiological methods were used to monitor mitochondrial function in substantia nigra pars compacta (SNc) dopaminergic and pedunculopontine nucleus (PPN) cholinergic neurons after stereotaxic injection of aSYN pre-formed fibrils (PFFs) into the mouse brain. METHODS aSYN PFFs were stereotaxically injected into the SNc or PPN of mice. Twelve weeks later, mice were studied using a combination of approaches, including immunocytochemical analysis, cell-type specific transcriptomic profiling, electron microscopy, electrophysiology and two-photon-laser-scanning microscopy of genetically encoded sensors for bioenergetic and redox status. RESULTS In addition to inducing a significant neuronal loss, SNc injection of PFFs induced the formation of intracellular, phosphorylated aSYN aggregates selectively in dopaminergic neurons. In these neurons, PFF-exposure decreased mitochondrial gene expression, reduced the number of mitochondria, increased oxidant stress, and profoundly disrupted mitochondrial adenosine triphosphate production. Consistent with an aSYN-induced bioenergetic deficit, the autonomous spiking of dopaminergic neurons slowed or stopped. PFFs also up-regulated lysosomal gene expression and increased lysosomal abundance, leading to the formation of Lewy-like inclusions. Similar changes were observed in PPN cholinergic neurons following aSYN PFF exposure. CONCLUSIONS Taken together, our findings suggest that disruption of mitochondrial function, and the subsequent bioenergetic deficit, is a proximal step in the cascade of events induced by aSYN pathology leading to dysfunction and degeneration of neurons at-risk in PD.
Collapse
Affiliation(s)
- Fanni F Geibl
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35043, Marburg, Germany
| | - Martin T Henrich
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35043, Marburg, Germany
| | - Zhong Xie
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Enrico Zampese
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, US
| | - Jun Ueda
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Tatiana Tkatch
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, US
| | - David L Wokosin
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Elena Nasiri
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35043, Marburg, Germany
| | - Constantin A Grotmann
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35043, Marburg, Germany
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, US
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, US.
| |
Collapse
|
13
|
Wang W, Wang D, Zhao D, Xu L, Jiang S, Zhang Y, Cui M, Liu J, Meng F, Liu C, Liu D, Li W, Li C. Dorsal raphe dopaminergic neurons target CaMKII + neurons in dorsal bed nucleus of the stria terminalis for mediating depression-related behaviors. Transl Psychiatry 2024; 14:408. [PMID: 39358336 PMCID: PMC11447211 DOI: 10.1038/s41398-024-03093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Dopamine (DA) neurons play a crucial role in the development and manifestation of depression, as well as in response to antidepressant treatments. While the function of the predominantly distributed DA neurons in the ventral tegmental area (VTA) is well established, the contribution of a small fraction of DA neurons in the dorsal raphe nucleus (DRN) during depression remains unclear. In this study, we found that chronic unpredictable stress (CUS) induces depression-related behaviors and decreases spontaneous firing rates, excitatory and inhibitory postsynaptic currents of DA neurons in the DRN associated with reduced excitatory synaptic transmission in male and female mice. The chemogenetic inhibition of DA neurons in the DRN produces depressive phenotypes. Conversely, their activation completely reversed the anhedonic and despair behaviors induced by CUS. Furthermore, we showed that a DRN dopaminergic projecting to the dorsal bed nucleus of the stria terminalis (dBNST) selectively controls depressive behaviors by influencing the neural activity and N-methyl-D-aspartate receptor (NMDAR) mediating EPSC of calcium/calmodulin-dependent protein kinase II+ (CaMKII+) target neurons by regulating dopamine neurotransmitter and dopamine receptor 2 (DR2) in the dBNST. Overall, these findings highlight the essential role of the DRNDA → dBNSTCaMKII+ neural circuit in bi-directionally mediating stress-induced depression-related behaviors. Our findings indicate that DRN DA neurons are a key component of the neural circuitry involved in regulating depression-related behaviors, making them a potential therapeutic target for depression.
Collapse
Affiliation(s)
- Wentao Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Di Zhao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lihong Xu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Yu Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Nursing, Binzhou Medical University, Yantai, Shandong, China
| | - Minghu Cui
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Cuilan Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dunjiang Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wei Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Chen Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China.
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China.
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
14
|
Pajarillo E, Kim S, Digman A, Ajayi I, Nyarko-Danquah I, Son DS, Aschner M, Lee E. Dopaminergic REST/NRSF is protective against manganese-induced neurotoxicity in mice. J Biol Chem 2024; 300:107707. [PMID: 39178947 PMCID: PMC11421342 DOI: 10.1016/j.jbc.2024.107707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/23/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Chronic exposure to elevated levels of manganese (Mn) may cause a neurological disorder referred to as manganism. The transcription factor REST is dysregulated in several neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. REST upregulated tyrosine hydroxylase and induced protection against Mn toxicity in neuronal cultures. In the present study, we investigated if dopaminergic REST plays a critical role in protecting against Mn-induced toxicity in vivo using dopaminergic REST conditional knockout (REST-cKO) mice and REST loxP mice as wild-type (WT) controls. Restoration of REST in the substantia nigra (SN) with neuronal REST AAV vector infusion was performed to further support the role of REST in Mn toxicity. Mice were exposed to Mn (330 μg, intranasal, daily for 3 weeks), followed by behavioral tests and molecular biology experiments. Results showed that Mn decreased REST mRNA/protein levels in the SN-containing midbrain, as well as locomotor activity and motor coordination in WT mice, which were further decreased in REST-cKO mice. Mn-induced mitochondrial insults, such as impairment of fission/fusion and mitophagy, apoptosis, and oxidative stress, in the midbrain of WT mice were more pronounced in REST-cKO mice. However, REST restoration in the SN of REST-cKO mice attenuated Mn-induced neurotoxicity. REST's molecular target for its protection is unclear, but REST attenuated Mn-induced mitochondrial dysregulation, indicating that it is a primary intracellular target for both Mn and REST. These novel findings suggest that dopaminergic REST in the nigrostriatal pathway is critical in protecting against Mn toxicity, underscoring REST as a potential therapeutic target for treating manganism.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Itunu Ajayi
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, New York, USA
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| |
Collapse
|
15
|
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
|
16
|
Watamura N, Kakiya N, Fujioka R, Kamano N, Takahashi M, Nilsson P, Saito T, Iwata N, Fujisawa S, Saido TC. The dopaminergic system promotes neprilysin-mediated degradation of amyloid-β in the brain. Sci Signal 2024; 17:eadk1822. [PMID: 39106321 DOI: 10.1126/scisignal.adk1822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Deposition of amyloid-β (Aβ) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aβ degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aβ-degrading enzyme neprilysin and reduced the amount of Aβ deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aβ deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aβ, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 171 64, Solna, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nobuhisa Iwata
- Department of Genome-based Drug Discovery & Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8521, Japan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
17
|
Beaver ML, Evans RC. Muscarinic receptor activation preferentially inhibits rebound in vulnerable dopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605819. [PMID: 39131326 PMCID: PMC11312546 DOI: 10.1101/2024.07.30.605819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Dopaminergic subpopulations of the substantia nigra pars compacta (SNc) differentially degenerate in Parkinson's disease and are characterized by unique electrophysiological properties. The vulnerable population expresses a T-type calcium channel-mediated afterdepolarization (ADP) and shows rebound activity upon release from inhibition, whereas the resilient population does not have an ADP and is slower to fire after hyperpolarization. This rebound activity can trigger dopamine release in the striatum, an important component of basal ganglia function. Using whole-cell patch clamp electrophysiology on ex vivo slices from adult mice of both sexes, we find that muscarinic activation with the non-selective muscarinic agonist Oxotremorine inhibits rebound activity more strongly in vulnerable vs resilient SNc neurons. Here, we show that this effect depends on the direct activation of muscarinic receptors on the SNc dopaminergic neurons. Through a series of pharmacological and transgenic knock-out experiments, we tested whether the muscarinic inhibition of rebound was mediated through the canonical rebound-related ion channels: T-type calcium channels, hyperpolarization-activated cation channels (HCN), and A-type potassium channels. We find that muscarinic receptor activation inhibits HCN-mediated current (Ih) in vulnerable SNc neurons, but that Ih activity is not necessary for the muscarinic inhibition of rebound activity. Similarly, we find that Oxotremorine inhibits rebound activity independently of T-type calcium channels and A-type potassium channels. Together these findings reveal new principles governing acetylcholine and dopamine interactions, showing that muscarinic receptors directly affect SNc rebound activity in the midbrain at the somatodendritic level and differentially modify information processing in distinct SNc subpopulations.
Collapse
Affiliation(s)
- Megan L. Beaver
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, DC, USA 20007
| | - Rebekah C. Evans
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA 20007
| |
Collapse
|
18
|
Chuhma N, Rayport S. Regional heterogeneity in the membrane properties of mouse striatal neurons. Front Cell Neurosci 2024; 18:1412897. [PMID: 39144155 PMCID: PMC11321984 DOI: 10.3389/fncel.2024.1412897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/19/2024] [Indexed: 08/16/2024] Open
Abstract
The cytoarchitecture of the striatum is remarkably homogeneous, in contrast to the regional variation in striatal functions. Whether differences in the intrinsic membrane properties of striatal neurons contribute to regional heterogeneity has not been addressed systematically. We made recordings throughout the young adult mouse striatum under identical conditions, with synaptic input blocked, from four major striatal neuron types, namely, the two subtypes of spiny projection neurons (SPNs), cholinergic interneurons (ChIs), and fast-spiking GABAergic interneurons (FSIs), sampling at least 100 cells per cell type. Regional variation manifested across all cell types. All cell types in the nucleus accumbens (NAc) shell had higher input impedance and increased excitability. Cells in the NAc core were differentiated from the caudate-putamen (CPu) for both SPN subtypes by smaller action potentials and increased excitability. Similarity between the two SPN subtypes showed regional variation, differing more in the NAc than in the CPu. So, in the Str, both the intrinsic properties of interneurons and projection neurons are regionally heterogeneous, with the greatest difference between the NAc and CPu; greater excitability of NAc shell neurons may make the region more susceptible to activity-dependent plasticity.
Collapse
Affiliation(s)
- Nao Chuhma
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Stephen Rayport
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
| |
Collapse
|
19
|
Young PA, Waller O, Ball K, Williams CC, Nashmi R. Phasic Stimulation of Dopaminergic Neurons of the Lateral Substantia Nigra Increases Open Field Exploratory Behaviour and Reduces Habituation Over Time. Neuroscience 2024; 551:276-289. [PMID: 38838978 DOI: 10.1016/j.neuroscience.2024.05.025] [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: 10/05/2023] [Revised: 04/09/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024]
Abstract
Transient nigrostriatal dopaminergic signalling is well known for its role in reinforcement learning and increasingly so for its role in the initiation of voluntary movement. However, how transient bursts of dopamine modulate voluntary movement remains unclear, likely due to the heterogeneity of the nigrostriatal system, the focus of optogenetic studies on locomotion at sub-sec time intervals, and the overlapping roles of phasic dopamine in behaviour and novelty signalling. In this study we investigated how phasic activity in the lateral substantia nigra pars compacta (lateral SNc) over time affects voluntary behaviours during exploration. Using a transgenic mouse model of both sexes expressing channelrhodopsin (ChR2) in dopamine transporter-expressing cells, we stimulated the lateral SNc while mice explored an open field over two consecutive days. We found that phasic activation of the lateral SNc induced an increase in exploratory behaviours including horizontal movement activity, locomotion initiation, and rearing specifically on the first open field exposure, but not on the second day. In addition, stimulated animals did not habituate to the same extent as their ChR2-negative counterparts, as indicated by a lack of decrease in baseline activity. These findings suggest that rather than prompting voluntary movement in general, phasic nigrostriatal dopamine prompts context-appropriate behaviours. In addition, dopamine signalling that modulates movement acts over longer timescales than the transient signal, affecting behaviour even after the signal has ended.
Collapse
Affiliation(s)
- Penelope A Young
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada; Division of Medical Sciences, University of Victoria, British Columbia V8W 2Y2, Canada
| | - Olivia Waller
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada
| | - Katherine Ball
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada
| | - Chad C Williams
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Raad Nashmi
- Department of Biology, University of Victoria, British Columbia V8W 2Y2, Canada; Division of Medical Sciences, University of Victoria, British Columbia V8W 2Y2, Canada.
| |
Collapse
|
20
|
Labouesse MA, Wilhelm M, Kagiampaki Z, Yee AG, Denis R, Harada M, Gresch A, Marinescu AM, Otomo K, Curreli S, Serratosa Capdevila L, Zhou X, Cola RB, Ravotto L, Glück C, Cherepanov S, Weber B, Zhou X, Katner J, Svensson KA, Fellin T, Trudeau LE, Ford CP, Sych Y, Patriarchi T. A chemogenetic approach for dopamine imaging with tunable sensitivity. Nat Commun 2024; 15:5551. [PMID: 38956067 PMCID: PMC11219860 DOI: 10.1038/s41467-024-49442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 06/05/2024] [Indexed: 07/04/2024] Open
Abstract
Genetically-encoded dopamine (DA) sensors enable high-resolution imaging of DA release, but their ability to detect a wide range of extracellular DA levels, especially tonic versus phasic DA release, is limited by their intrinsic affinity. Here we show that a human-selective dopamine receptor positive allosteric modulator (PAM) can be used to boost sensor affinity on-demand. The PAM enhances DA detection sensitivity across experimental preparations (in vitro, ex vivo and in vivo) via one-photon or two-photon imaging. In vivo photometry-based detection of optogenetically-evoked DA release revealed that DETQ administration produces a stable 31 minutes window of potentiation without effects on animal behavior. The use of the PAM revealed region-specific and metabolic state-dependent differences in tonic DA levels and enhanced single-trial detection of behavior-evoked phasic DA release in cortex and striatum. Our chemogenetic strategy can potently and flexibly tune DA imaging sensitivity and reveal multi-modal (tonic/phasic) DA signaling across preparations and imaging approaches.
Collapse
Affiliation(s)
- Marie A Labouesse
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Maria Wilhelm
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Institute for Neuroscience, ETH Zurich, Zurich, Switzerland
| | | | - Andrew G Yee
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Raphaelle Denis
- Department of Pharmacology & Physiology, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
| | - Masaya Harada
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Andrea Gresch
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | | | - Kanako Otomo
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Sebastiano Curreli
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Xuehan Zhou
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Reto B Cola
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Stanislav Cherepanov
- Institute of Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Xin Zhou
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Louis-Eric Trudeau
- Department of Pharmacology & Physiology, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, SNC and CIRCA Research groups, Université de Montréal, Montréal, QC, Canada
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yaroslav Sych
- Institute of Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
21
|
Li J, Ng KW, Sung CC, Chung KKK. The role of age-associated alpha-synuclein aggregation in a conditional transgenic mouse model of Parkinson's disease: Implications for Lewy body formation. J Neurochem 2024; 168:1215-1236. [PMID: 38693066 DOI: 10.1111/jnc.16122] [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: 11/08/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that is affecting an increasing number of older adults. Although PD is mostly sporadic, genetic mutations have been found in cohorts of families with a history of familial PD (FPD). The first such mutation linked to FPD causes a point mutation (A53T) in α-synuclein (α-syn), a major component of Lewy bodies, which are a classical pathological hallmark of PD. These findings suggest that α-syn is an important contributor to the development of PD. In our previous study, we developed an adenoviral mouse model of PD and showed that the expression of wild-type (WT) α-syn or a mutant form with an increased propensity to aggregate, designated as WT-CL1 α-syn, could be used to study how α-syn aggregation contributes to PD. In this study, we established a transgenic mouse model that conditionally expresses WT or WT-CL1 α-syn in dopaminergic (DA) neurons and found that the expression of either WT or WT-CL1 α-syn was associated with an age-dependent degeneration of DA neurons and movement dysfunction. Using this model, we were able to monitor the process of α-syn aggregate formation and found a correlation between age and the number and sizes of α-syn aggregates formed. These results provide a potential mechanism by which age-dependent α-syn aggregation may lead to the formation of Lewy bodies in PD pathogenesis.
Collapse
Affiliation(s)
- Jiahua Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ka Wai Ng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Chun Chau Sung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kenny K K Chung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
22
|
Kang J, Huang G, Ma L, Tong Y, Shahapal A, Chen P, Shen J. Cell-autonomous role of leucine-rich repeat kinase in the protection of dopaminergic neuron survival. eLife 2024; 12:RP92673. [PMID: 38856715 PMCID: PMC11164531 DOI: 10.7554/elife.92673] [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: 06/11/2024] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease (PD). However, whether LRRK2 mutations cause PD and degeneration of dopaminergic (DA) neurons via a toxic gain-of-function or a loss-of-function mechanism is unresolved and has pivotal implications for LRRK2-based PD therapies. In this study, we investigate whether Lrrk2 and its functional homolog Lrrk1 play a cell-intrinsic role in DA neuron survival through the development of DA neuron-specific Lrrk conditional double knockout (cDKO) mice. Unlike Lrrk germline DKO mice, DA neuron-restricted Lrrk cDKO mice exhibit normal mortality but develop age-dependent loss of DA neurons, as shown by the progressive reduction of DA neurons in the substantia nigra pars compacta (SNpc) at the ages of 20 and 24 months. Moreover, DA neurodegeneration is accompanied with increases in apoptosis and elevated microgliosis in the SNpc as well as decreases in DA terminals in the striatum, and is preceded by impaired motor coordination. Taken together, these findings provide the unequivocal evidence for the cell-intrinsic requirement of LRRK in DA neurons and raise the possibility that LRRK2 mutations may impair its protection of DA neurons, leading to DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Jongkyun Kang
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Guodong Huang
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Long Ma
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Youren Tong
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Anu Shahapal
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Phoenix Chen
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
| | - Jie Shen
- Department of Neurology, Brigham and Women’s HospitalBostonUnited States
- Program in Neuroscience, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
23
|
Brandner DD, Mashal MA, Grissom NM, Rothwell PE. Sex Differences in Morphine Sensitivity of Neuroligin-3 Knockout Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596965. [PMID: 38854153 PMCID: PMC11160712 DOI: 10.1101/2024.06.01.596965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Sex has a strong influence on the prevalence and course of brain conditions, including autism spectrum disorders. The mechanistic basis for these sex differences remains poorly understood, due in part to historical bias in biomedical research favoring analysis of male subjects, and the exclusion of female subjects. For example, studies of male mice carrying autism-associated mutations in neuroligin-3 are over-represented in the literature, including our own prior work showing diminished responses to chronic morphine exposure in male neuroligin-3 knockout mice. We therefore studied how constitutive and conditional genetic knockout of neuroligin-3 affects morphine sensitivity of female mice. In contrast to male mice, female neuroligin-3 knockout mice showed normal psychomotor sensitization after chronic morphine exposure. However, in the absence of neuroligin-3 expression, both female and male mice show a similar change in the topography of locomotor stimulation produced by morphine. Conditional genetic deletion of neuroligin-3 from dopamine neurons increased the locomotor response of female mice to high doses of morphine, contrasting with the decrease in psychomotor sensitization caused by the same manipulation in male mice. Together, our data reveal that knockout of neuroligin-3 has both common and distinct effects on morphine sensitivity in female and male mice. These results also support the notion that female sex can confer resilience against the impact of autism-associated gene variants.
Collapse
|
24
|
Rademacher K, Doric Z, Haddad D, Mamaligas A, Liao SC, Creed RB, Kano K, Chatterton Z, Fu Y, Garcia JH, Vance V, Sei Y, Kreitzer A, Halliday GM, Nelson AB, Margolis EB, Nakamura K. Chronic hyperactivation of midbrain dopamine neurons causes preferential dopamine neuron degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588321. [PMID: 38645054 PMCID: PMC11030348 DOI: 10.1101/2024.04.05.588321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Parkinson's disease (PD) is characterized by the death of substantia nigra (SNc) dopamine (DA) neurons, but the pathophysiological mechanisms that precede and drive their death remain unknown. The activity of DA neurons is likely altered in PD, but we understand little about if or how chronic changes in activity may contribute to degeneration. To address this question, we developed a chemogenetic (DREADD) mouse model to chronically increase DA neuron activity, and confirmed this increase using ex vivo electrophysiology. Chronic hyperactivation of DA neurons resulted in prolonged increases in locomotor activity during the light cycle and decreases during the dark cycle, consistent with chronic changes in DA release and circadian disturbances. We also observed early, preferential degeneration of SNc projections, recapitulating the PD hallmarks of selective vulnerability of SNc axons and the comparative resilience of ventral tegmental area axons. This was followed by eventual loss of midbrain DA neurons. Continuous DREADD activation resulted in a sustained increase in baseline calcium levels, supporting an important role for increased calcium in the neurodegeneration process. Finally, spatial transcriptomics from DREADD mice examining midbrain DA neurons and striatal targets, and cross-validation with human patient samples, provided insights into potential mechanisms of hyperactivity-induced toxicity and PD. Our results thus reveal the preferential vulnerability of SNc DA neurons to increased neural activity, and support a potential role for increased neural activity in driving degeneration in PD.
Collapse
Affiliation(s)
- Katerina Rademacher
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Zak Doric
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
| | - Dominik Haddad
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Aphroditi Mamaligas
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Szu-Chi Liao
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, CA
| | - Rose B. Creed
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Kohei Kano
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - 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
| | - 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
| | - Joseph H. Garcia
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- School of Medicine, University of California, San Francisco, California, USA
| | - Victoria Vance
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- College of Science, Northeastern University, Boston, MA
| | - Yoshitaka Sei
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Anatol Kreitzer
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- UCSF Department of Physiology, University of California San Francisco, CA
| | - Glenda M 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
| | - Alexandra B. Nelson
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Elyssa B. Margolis
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Ken Nakamura
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
- Graduate Program in Biomedical Sciences, University of California San Francisco, San Francisco
| |
Collapse
|
25
|
Recinto SJ, Premachandran S, Mukherjee S, Allot A, MacDonald A, Yaqubi M, Gruenheid S, Trudeau LE, Stratton JA. Characterizing enteric neurons in dopamine transporter (DAT)-Cre reporter mice reveals dopaminergic subtypes with dual-transmitter content. Eur J Neurosci 2024; 59:2465-2482. [PMID: 38487941 DOI: 10.1111/ejn.16307] [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: 07/31/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 05/22/2024]
Abstract
The enteric nervous system (ENS) comprises a complex network of neurons whereby a subset appears to be dopaminergic although the characteristics, roles, and implications in disease are less understood. Most investigations relating to enteric dopamine (DA) neurons rely on immunoreactivity to tyrosine hydroxylase (TH)-the rate-limiting enzyme in the production of DA. However, TH immunoreactivity is likely to provide an incomplete picture. This study herein provides a comprehensive characterization of DA neurons in the gut using a reporter mouse line, expressing a fluorescent protein (tdTomato) under control of the DA transporter (DAT) promoter. Our findings confirm a unique localization of DA neurons in the gut and unveil the discrete subtypes of DA neurons in this organ, which we characterized using both immunofluorescence and single-cell transcriptomics, as well as validated using in situ hybridization. We observed distinct subtypes of DAT-tdTomato neurons expressing co-transmitters and modulators across both plexuses; some of them likely co-releasing acetylcholine, while others were positive for a slew of canonical DAergic markers (TH, VMAT2 and GIRK2). Interestingly, we uncovered a seemingly novel population of DA neurons unique to the ENS which was ChAT/DAT-tdTomato-immunoreactive and expressed Grp, Calcb, and Sst. Given the clear heterogeneity of DAergic gut neurons, further investigation is warranted to define their functional signatures and decipher their implication in disease.
Collapse
Affiliation(s)
- Sherilyn Junelle Recinto
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Shobina Premachandran
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Sriparna Mukherjee
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Pharmacology and Physiology, Department of Neurosciences, Université de Montreal, Faculty of Medicine, SNC and CIRCA Research Groups, Montreal, Quebec, Canada
| | - Alexis Allot
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Adam MacDonald
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Moein Yaqubi
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Samantha Gruenheid
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Louis-Eric Trudeau
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Pharmacology and Physiology, Department of Neurosciences, Université de Montreal, Faculty of Medicine, SNC and CIRCA Research Groups, Montreal, Quebec, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
26
|
Kapur M, Molumby MJ, Guzman C, Heinz S, Ackerman SL. Cell-type-specific expression of tRNAs in the brain regulates cellular homeostasis. Neuron 2024; 112:1397-1415.e6. [PMID: 38377989 PMCID: PMC11065635 DOI: 10.1016/j.neuron.2024.01.028] [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: 07/25/2023] [Revised: 12/23/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
Defects in tRNA biogenesis are associated with multiple neurological disorders, yet our understanding of these diseases has been hampered by an inability to determine tRNA expression in individual cell types within a complex tissue. Here, we developed a mouse model in which RNA polymerase III is conditionally epitope tagged in a Cre-dependent manner, allowing us to accurately profile tRNA expression in any cell type in vivo. We investigated tRNA expression in diverse nervous system cell types, revealing dramatic heterogeneity in the expression of tRNA genes between populations. We found that while maintenance of levels of tRNA isoacceptor families is critical for cellular homeostasis, neurons are differentially vulnerable to insults to distinct tRNA isoacceptor families. Cell-type-specific translatome analysis suggests that the balance between tRNA availability and codon demand may underlie such differential resilience. Our work provides a platform for investigating the complexities of mRNA translation and tRNA biology in the brain.
Collapse
Affiliation(s)
- Mridu Kapur
- Department of Cellular and Molecular Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; The Howard Hughes Medical Institute
| | - Michael J Molumby
- Department of Cellular and Molecular Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; The Howard Hughes Medical Institute
| | - Carlos Guzman
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, Bioinformatics & Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sven Heinz
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, Bioinformatics & Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Susan L Ackerman
- Department of Cellular and Molecular Medicine, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; The Howard Hughes Medical Institute; Department of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
27
|
Chaves T, Török B, Fazekas CL, Correia P, Sipos E, Várkonyi D, Tóth ZE, Dóra F, Dobolyi Á, Zelena D. The Dopaminergic Cells in the Median Raphe Region Regulate Social Behavior in Male Mice. Int J Mol Sci 2024; 25:4315. [PMID: 38673899 PMCID: PMC11050709 DOI: 10.3390/ijms25084315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
According to previous studies, the median raphe region (MRR) is known to contribute significantly to social behavior. Besides serotonin, there have also been reports of a small population of dopaminergic neurons in this region. Dopamine is linked to reward and locomotion, but very little is known about its role in the MRR. To address that, we first confirmed the presence of dopaminergic cells in the MRR of mice (immunohistochemistry, RT-PCR), and then also in humans (RT-PCR) using healthy donor samples to prove translational relevance. Next, we used chemogenetic technology in mice containing the Cre enzyme under the promoter of the dopamine transporter. With the help of an adeno-associated virus, designer receptors exclusively activated by designer drugs (DREADDs) were expressed in the dopaminergic cells of the MRR to manipulate their activity. Four weeks later, we performed an extensive behavioral characterization 30 min after the injection of the artificial ligand (Clozapine-N-Oxide). Stimulation of the dopaminergic cells in the MRR decreased social interest without influencing aggression and with an increase in social discrimination. Additionally, inhibition of the same cells increased the friendly social behavior during social interaction test. No behavioral changes were detected in anxiety, memory or locomotion. All in all, dopaminergic cells were present in both the mouse and human samples from the MRR, and the manipulation of the dopaminergic neurons in the MRR elicited a specific social response.
Collapse
Affiliation(s)
- Tiago Chaves
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Bibiána Török
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Eszter Sipos
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
| | - Dorottya Várkonyi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
| | - Zsuzsanna E. Tóth
- Laboratory of Neuroendocrinology and in Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary;
| | - Fanni Dóra
- Human Brain Tissue Bank, Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary;
| | - Árpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, H1117 Budapest, Hungary;
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
| |
Collapse
|
28
|
Yu M, Sun F, Xiang G, Zhang Y, Wang X, Liu X, Huang B, Li X, Zhang D. Liver kinase B-1 modulates the activity of dopamine neurons in the ventral tegmental area and regulates social memory formation. Front Mol Neurosci 2024; 17:1289476. [PMID: 38646099 PMCID: PMC11026561 DOI: 10.3389/fnmol.2024.1289476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 03/19/2024] [Indexed: 04/23/2024] Open
Abstract
Social memory is the ability to discriminate between familiar and unknown conspecifics. It is an important component of social cognition and is therefore essential for the establishment of social relationships. Although the neural circuit mechanisms underlying social memory encoding have been well investigated, little focus has been placed on the regulatory mechanisms of social memory processing. The dopaminergic system, originating from the midbrain ventral tegmental area (VTA), is a key modulator of cognitive function. This study aimed to illustrate its role in modulating social memory and explore the possible molecular mechanisms. Here, we show that the activation of VTA dopamine (DA) neurons is required for the formation, but not the retrieval, of social memory. Inhibition of VTA DA neurons before social interaction, but not 24 h after social interaction, significantly impaired social discrimination the following day. In addition, we showed that the activation of VTA DA neurons was regulated by the serine/threonine protein kinase liver kinase B1 (Lkb1). Deletion of Lkb1 in VTA DA neurons reduced the frequency of burst firing of dopaminergic neurons. Furthermore, Lkb1 plays an important role in regulating social behaviors. Both genetic and virus-mediated deletions of Lkb1 in the VTA of adult mice impaired social memory and subsequently attenuated social familiarization. Altogether, our results provide direct evidence linking social memory formation to the activation of VTA DA neurons in mice and illustrate the crucial role of Lkb1 in regulating VTA DA neuron function.
Collapse
Affiliation(s)
- Meng Yu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Fengjiao Sun
- Institute of Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Guo Xiang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Yuhan Zhang
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xuejun Wang
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xia Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| |
Collapse
|
29
|
Hose L, Langenhagen AK, Kefalakes E, Schweitzer T, Kubinski S, Barak S, Pich A, Grothe C. A dual-omics approach on the effects of fibroblast growth factor-2 (FGF-2) on ventral tegmental area dopaminergic neurons in response to alcohol consumption in mice. Eur J Neurosci 2024; 59:1519-1535. [PMID: 38185886 DOI: 10.1111/ejn.16234] [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: 07/31/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024]
Abstract
Harmful alcohol consumption is a major socioeconomic burden to the health system, as it can be the cause of mortality of heavy alcohol drinkers. The dopaminergic (DAergic) system is thought to play an important role in the pathogenesis of alcohol drinking behaviour; however, its exact role remains elusive. Fibroblast growth factor 2 (FGF-2), a neurotrophic factor, associated with both the DAergic system and alcohol consumption, may play an important role in DAergic neuroadaptations during alcohol abuse. Within this study, we aimed to clarify the role of endogenous FGF-2 on the DAergic system and whether there is a possible link to alcohol consumption. We found that lack of FGF-2 reduces the alcohol intake of mice. Transcriptome analysis of DAergic neurons revealed that FGF-2 knockout (FGF-2 KO) shifts the molecular fingerprint of midbrain dopaminergic (mDA) neurons to DA subtypes of the ventral tegmental area (VTA). In line with this, proteomic changes predominantly appear also in the VTA. Interestingly, these changes led to an altered regulation of the FGF-2 signalling cascades and DAergic pathways in a region-specific manner, which was only marginally affected by voluntary alcohol consumption. Thus, lack of FGF-2 not only affects the gene expression but also the proteome of specific brain regions of mDA neurons. Our study provides new insights into the neuroadaptations of the DAergic system during alcohol abuse and, therefore, comprises novel targets for future pharmacological interventions.
Collapse
Affiliation(s)
- Leonie Hose
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Alina Katharina Langenhagen
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Ekaterini Kefalakes
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
| | - Theresa Schweitzer
- Institute of Toxicology, Hannover, Germany
- Core Facility Proteomics, Institute of Toxicology, Hannover, Germany
| | - Sabrina Kubinski
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Andreas Pich
- Institute of Toxicology, Hannover, Germany
- Core Facility Proteomics, Institute of Toxicology, Hannover, Germany
| | - Claudia Grothe
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
30
|
Lau MYH, Gadiwalla S, Jones S, Galliano E. Different electrophysiological profiles of genetically labelled dopaminergic neurons in the mouse midbrain and olfactory bulb. Eur J Neurosci 2024; 59:1480-1499. [PMID: 38169095 DOI: 10.1111/ejn.16239] [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: 08/29/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024]
Abstract
Dopaminergic (DA) neurons play pivotal roles in diverse brain functions, spanning movement, reward processing and sensory perception. DA neurons are most abundant in the midbrain (Substantia Nigra pars compacta [SNC] and Ventral Tegmental Area [VTA]) and the olfactory bulb (OB) in the forebrain. Interestingly, a subtype of OB DA neurons is capable of regenerating throughout life, while a second class is exclusively born during embryonic development. Compelling evidence in SNC and VTA also indicates substantial heterogeneity in terms of morphology, connectivity and function. To further investigate this heterogeneity and directly compare form and function of midbrain and forebrain bulbar DA neurons, we performed immunohistochemistry and whole-cell patch-clamp recordings in ex vivo brain slices from juvenile DAT-tdTomato mice. After confirming the penetrance and specificity of the dopamine transporter (DAT) Cre line, we compared soma shape, passive membrane properties, voltage sags and action potential (AP) firing across midbrain and forebrain bulbar DA subtypes. We found that each DA subgroup within midbrain and OB was highly heterogeneous, and that DA neurons across the two brain areas are also substantially different. These findings complement previous work in rats as well as gene expression and in vivo datasets, further questioning the existence of a single "dopaminergic" neuronal phenotype.
Collapse
Affiliation(s)
- Maggy Yu Hei Lau
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Sana Gadiwalla
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Elisa Galliano
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| |
Collapse
|
31
|
Ellioff KJ, Osting SM, Lentine A, Welper AD, Burger C, Greenspan DS. Ablation of Mitochondrial RCC1-L Induces Nigral Dopaminergic Neurodegeneration and Parkinsonian-like Motor Symptoms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.01.567409. [PMID: 38585782 PMCID: PMC10996473 DOI: 10.1101/2023.12.01.567409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Mitochondrial dysfunction has been linked to both idiopathic and familial forms of Parkinson's disease (PD). We have previously identified RCC1-like (RCC1L) as a protein of the inner mitochondrial membrane important to mitochondrial fusion. Herein, to test whether deficits in RCC1L mitochondrial function might be involved in PD pathology, we have selectively ablated the Rcc1l gene in the dopaminergic (DA) neurons of mice. A PD-like phenotype resulted that includes progressive movement abnormalities, paralleled by progressive degeneration of the nigrostriatal tract. Experimental and control groups were examined at 2, 3-4, and 5-6 months of age. Animals were tested in the open field task to quantify anxiety, exploratory drive, locomotion, and immobility; and in the cylinder test to quantify rearing behavior. Beginning at 3-4 months, both female and male Rcc1l knockout mice show rigid muscles and resting tremor, kyphosis and a growth deficit compared with heterozygous or wild type littermate controls. Rcc1l knockout mice begin showing locomotor impairments at 3-4 months, which progress until 5-6 months of age, at which age the Rcc1l knockout mice die. The progressive motor impairments were associated with progressive and significantly reduced tyrosine hydroxylase immunoreactivity in the substantia nigra pars compacta (SNc), and dramatic loss of nigral DA projections in the striatum. Dystrophic spherical mitochondria are apparent in the soma of SNc neurons in Rcc1l knockout mice as early as 1.5-2.5 months of age and become progressively more pronounced until 5-6 months. Together, the results reveal the RCC1L protein to be essential to in vivo mitochondrial function in DA neurons. Further characterization of this mouse model will determine whether it represents a new model for in vivo study of PD, and the putative role of the human RCC1L gene as a risk factor that might increase PD occurrence and severity in humans.
Collapse
Affiliation(s)
- Kaylin J. Ellioff
- Department of Neurology, University of Wisconsin, Madison WI, 53706
- Present Address, Department of Pharmacology, University of Washington, Seattle WA, 98195
| | | | - Alyssa Lentine
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, 53705
| | - Ashley D. Welper
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, 53705
| | - Corinna Burger
- Department of Neurology, University of Wisconsin, Madison WI, 53706
| | - Daniel S. Greenspan
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, 53705
| |
Collapse
|
32
|
Kilfeather P, Khoo JH, Wagner K, Liang H, Caiazza MC, An Y, Zhang X, Chen X, Connor-Robson N, Shang Z, Wade-Martins R. Single-cell spatial transcriptomic and translatomic profiling of dopaminergic neurons in health, aging, and disease. Cell Rep 2024; 43:113784. [PMID: 38386560 DOI: 10.1016/j.celrep.2024.113784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 11/14/2023] [Accepted: 01/27/2024] [Indexed: 02/24/2024] Open
Abstract
The brain is spatially organized and contains unique cell types, each performing diverse functions and exhibiting differential susceptibility to neurodegeneration. This is exemplified in Parkinson's disease with the preferential loss of dopaminergic neurons of the substantia nigra pars compacta. Using a Parkinson's transgenic model, we conducted a single-cell spatial transcriptomic and dopaminergic neuron translatomic analysis of young and old mouse brains. Through the high resolving capacity of single-cell spatial transcriptomics, we provide a deep characterization of the expression features of dopaminergic neurons and 27 other cell types within their spatial context, identifying markers of healthy and aging cells, spanning Parkinson's relevant pathways. We integrate gene enrichment and genome-wide association study data to prioritize putative causative genes for disease investigation, identifying CASR as a regulator of dopaminergic calcium handling. These datasets represent the largest public resource for the investigation of spatial gene expression in brain cells in health, aging, and disease.
Collapse
Affiliation(s)
- Peter Kilfeather
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | - Katherina Wagner
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | | | - Maria Claudia Caiazza
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Yanru An
- BGI Research, 49276 Riga, Latvia
| | | | | | - Natalie Connor-Robson
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK
| | | | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre and Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford OX1 3QU, UK; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
33
|
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
|
34
|
Glykos V, Fujisawa S. Memory-specific encoding activities of the ventral tegmental area dopamine and GABA neurons. eLife 2024; 12:RP89743. [PMID: 38512339 PMCID: PMC10957172 DOI: 10.7554/elife.89743] [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: 03/22/2024] Open
Abstract
Although the midbrain dopamine (DA) system plays a crucial role in higher cognitive functions, including updating and maintaining short-term memory, the encoding properties of the somatic spiking activity of ventral tegmental area (VTA) DA neurons for short-term memory computations have not yet been identified. Here, we probed and analyzed the activity of optogenetically identified DA and GABA neurons while mice engaged in short-term memory-dependent behavior in a T-maze task. Single-neuron analysis revealed that significant subpopulations of DA and GABA neurons responded differently between left and right trials in the memory delay. With a series of control behavioral tasks and regression analysis tools, we show that firing rate differences are linked to short-term memory-dependent decisions and cannot be explained by reward-related processes, motivated behavior, or motor-related activities. This evidence provides novel insights into the mnemonic encoding activities of midbrain DA and GABA neurons.
Collapse
Affiliation(s)
- Vasileios Glykos
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain ScienceWakoJapan
- Synapse Biology Unit, Okinawa Institute of Science and TechnologyOkinawaJapan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain ScienceWakoJapan
| |
Collapse
|
35
|
Amo R, Uchida N, Watabe-Uchida M. Glutamate inputs send prediction error of reward, but not negative value of aversive stimuli, to dopamine neurons. Neuron 2024; 112:1001-1019.e6. [PMID: 38278147 PMCID: PMC10957320 DOI: 10.1016/j.neuron.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 11/10/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024]
Abstract
Midbrain dopamine neurons are thought to signal reward prediction errors (RPEs), but the mechanisms underlying RPE computation, particularly the contributions of different neurotransmitters, remain poorly understood. Here, we used a genetically encoded glutamate sensor to examine the pattern of glutamate inputs to dopamine neurons in mice. We found that glutamate inputs exhibit virtually all of the characteristics of RPE rather than conveying a specific component of RPE computation, such as reward or expectation. Notably, whereas glutamate inputs were transiently inhibited by reward omission, they were excited by aversive stimuli. Opioid analgesics altered dopamine negative responses to aversive stimuli into more positive responses, whereas excitatory responses of glutamate inputs remained unchanged. Our findings uncover previously unknown synaptic mechanisms underlying RPE computations; dopamine responses are shaped by both synergistic and competitive interactions between glutamatergic and GABAergic inputs to dopamine neurons depending on valences, with competitive interactions playing a role in responses to aversive stimuli.
Collapse
Affiliation(s)
- Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
36
|
Stowell R, Wang KH. Dopaminergic signaling regulates microglial surveillance and adolescent plasticity in the frontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584167. [PMID: 38559264 PMCID: PMC10979918 DOI: 10.1101/2024.03.08.584167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Adolescence is a sensitive period for frontal cortical development and cognitive maturation. The dopaminergic (DA) mesofrontal circuit is particularly malleable in response to changes in adolescent experience and DA activity. However, the cellular mechanisms engaged in this plasticity remain unexplored. Here, we report that microglia, the innate immune cells of the brain, are uniquely sensitive to adolescent mesofrontal DA signaling. Longitudinal in vivo two-photon imaging in mice shows that frontal cortical microglia respond dynamically to plasticity-inducing behavioral or optogenetic DA axon stimulation with increased parenchymal and DA bouton surveillance. Microglial-axon contact precedes new bouton formation, and both D1 and D2-type DA receptors regulate microglial-bouton interactions and axonal plasticity. Moreover, D2 antagonism in adults reinstates adolescent plasticity, including increased microglial surveillance and new DA bouton formation. Our results reveal that DA signaling regulates microglial surveillance and axonal plasticity uniquely in the adolescent frontal cortex, presenting potential interventions for restoring plasticity in the adult brain.
Collapse
Affiliation(s)
- Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| | - Kuan Hong Wang
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| |
Collapse
|
37
|
Kang J, Huang G, Ma L, Tong Y, Shahapal A, Chen P, Shen J. Cell autonomous role of leucine-rich repeat kinase in protection of dopaminergic neuron survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.06.561293. [PMID: 37873418 PMCID: PMC10592668 DOI: 10.1101/2023.10.06.561293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease (PD), which is the leading neurodegenerative movement disorder characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). However, whether LRRK2 mutations cause PD and degeneration of DA neurons via a toxic gain-of-function or a loss-of-function mechanism is unresolved and has pivotal implications for LRRK2-based PD therapies. In this study, we investigate whether LRRK2 and its functional homologue LRRK1 play an essential, intrinsic role in DA neuron survival through the development of DA neuron-specific LRRK conditional double knockout (cDKO) mice. We first generated and characterized floxed LRRK1 and LRRK2 mice and then confirmed that germline deletions of the floxed LRRK1 and LRRK2 alleles result in null mutations, as evidenced by the absence of LRRK1 and LRRK2 mRNA and protein in the respective homozygous deleted mutant mice. We further examined the specificity of Cre-mediated recombination driven by the dopamine transporter-Cre (DAT-Cre) knockin (KI) allele using a GFP reporter line and confirmed that DAT-Cre-mediated recombination is restricted to DA neurons in the SNpc. Crossing these validated floxed LRRK1 and LRRK2 mice with DAT-Cre KI mice, we then generated DA neuron-restricted LRRK cDKO mice and further showed that levels of LRRK1 and LRRK2 are reduced in dissected ventral midbrains of LRRK cDKO mice. While DA neuron-restricted LRRK cDKO mice of both sexes exhibit normal mortality and body weight, they develop age-dependent loss of DA neurons in the SNpc, as demonstrated by the progressive reduction of DA neurons in the SNpc of LRRK cDKO mice at the ages of 20 and 24 months but the unaffected number of DA neurons at the age of 15 months. Moreover, DA neurodegeneration is accompanied with increases of apoptosis and elevated microgliosis in the SNpc as well as decreases of DA terminals in the striatum, and is preceded by impaired motor coordination. Taken together, these findings provide the unequivocal evidence for the importance of LRRK in DA neurons and raise the possibility that LRRK2 mutations may impair its protection of DA neurons, leading to DA neurodegeneration in PD.
Collapse
Affiliation(s)
- Jongkyun Kang
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Guodong Huang
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Long Ma
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Youren Tong
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Anu Shahapal
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Phoenix Chen
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Jie Shen
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States of America
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, United States of America
| |
Collapse
|
38
|
Brimblecombe KR, Connor-Robson N, Bataille CJR, Roberts BM, Gracie C, O'Connor B, Te Water Naude R, Karthik G, Russell AJ, Wade-Martins R, Cragg SJ. Inhibition of striatal dopamine release by the L-type calcium channel inhibitor isradipine co-varies with risk factors for Parkinson's. Eur J Neurosci 2024; 59:1242-1259. [PMID: 37941514 PMCID: PMC11426196 DOI: 10.1111/ejn.16180] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/10/2023]
Abstract
Ca2+ entry into nigrostriatal dopamine (DA) neurons and axons via L-type voltage-gated Ca2+ channels (LTCCs) contributes, respectively, to pacemaker activity and DA release and has long been thought to contribute to vulnerability to degeneration in Parkinson's disease. LTCC function is greater in DA axons and neurons from substantia nigra pars compacta than from ventral tegmental area, but this is not explained by channel expression level. We tested the hypothesis that LTCC control of DA release is governed rather by local mechanisms, focussing on candidate biological factors known to operate differently between types of DA neurons and/or be associated with their differing vulnerability to parkinsonism, including biological sex, α-synuclein, DA transporters (DATs) and calbindin-D28k (Calb1). We detected evoked DA release ex vivo in mouse striatal slices using fast-scan cyclic voltammetry and assessed LTCC support of DA release by detecting the inhibition of DA release by the LTCC inhibitors isradipine or CP8. Using genetic knockouts or pharmacological manipulations, we identified that striatal LTCC support of DA release depended on multiple intersecting factors, in a regionally and sexually divergent manner. LTCC function was promoted by factors associated with Parkinsonian risk, including male sex, α-synuclein, DAT and a dorsolateral co-ordinate, but limited by factors associated with protection, that is, female sex, glucocerebrosidase activity, Calb1 and ventromedial co-ordinate. Together, these data show that LTCC function in DA axons and isradipine effect are locally governed and suggest they vary in a manner that in turn might impact on, or reflect, the cellular stress that leads to parkinsonian degeneration.
Collapse
Affiliation(s)
- Katherine R Brimblecombe
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Natalie Connor-Robson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Carole J R Bataille
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Bradley M Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Caitlin Gracie
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bethan O'Connor
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Gayathri Karthik
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, UK
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Stephanie J Cragg
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
39
|
Bandarabadi M, Li S, Aeschlimann L, Colombo G, Tzanoulinou S, Tafti M, Becchetti A, Boutrel B, Vassalli A. Inactivation of hypocretin receptor-2 signaling in dopaminergic neurons induces hyperarousal and enhanced cognition but impaired inhibitory control. Mol Psychiatry 2024; 29:327-341. [PMID: 38123729 PMCID: PMC11116111 DOI: 10.1038/s41380-023-02329-z] [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: 05/15/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023]
Abstract
Hypocretin/Orexin (HCRT/OX) and dopamine (DA) are both key effectors of salience processing, reward and stress-related behaviors and motivational states, yet their respective roles and interactions are poorly delineated. We inactivated HCRT-to-DA connectivity by genetic disruption of Hypocretin receptor-1 (Hcrtr1), Hypocretin receptor-2 (Hcrtr2), or both receptors (Hcrtr1&2) in DA neurons and analyzed the consequences on vigilance states, brain oscillations and cognitive performance in freely behaving mice. Unexpectedly, loss of Hcrtr2, but not Hcrtr1 or Hcrtr1&2, induced a dramatic increase in theta (7-11 Hz) electroencephalographic (EEG) activity in both wakefulness and rapid-eye-movement sleep (REMS). DAHcrtr2-deficient mice spent more time in an active (or theta activity-enriched) substate of wakefulness, and exhibited prolonged REMS. Additionally, both wake and REMS displayed enhanced theta-gamma phase-amplitude coupling. The baseline waking EEG of DAHcrtr2-deficient mice exhibited diminished infra-theta, but increased theta power, two hallmarks of EEG hyperarousal, that were however uncoupled from locomotor activity. Upon exposure to novel, either rewarding or stress-inducing environments, DAHcrtr2-deficient mice featured more pronounced waking theta and fast-gamma (52-80 Hz) EEG activity surges compared to littermate controls, further suggesting increased alertness. Cognitive performance was evaluated in an operant conditioning paradigm, which revealed that DAHcrtr2-ablated mice manifest faster task acquisition and higher choice accuracy under increasingly demanding task contingencies. However, the mice concurrently displayed maladaptive patterns of reward-seeking, with behavioral indices of enhanced impulsivity and compulsivity. None of the EEG changes observed in DAHcrtr2-deficient mice were seen in DAHcrtr1-ablated mice, which tended to show opposite EEG phenotypes. Our findings establish a clear genetically-defined link between monosynaptic HCRT-to-DA neurotransmission and theta oscillations, with a differential and novel role of HCRTR2 in theta-gamma cross-frequency coupling, attentional processes, and executive functions, relevant to disorders including narcolepsy, attention-deficit/hyperactivity disorder, and Parkinson's disease.
Collapse
Affiliation(s)
- Mojtaba Bandarabadi
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sha Li
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Lea Aeschlimann
- Centre for Psychiatric Neuroscience, Department of Psychiatry, The Lausanne University Hospital, Lausanne, Switzerland
| | - Giulia Colombo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | | | - Mehdi Tafti
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Benjamin Boutrel
- Centre for Psychiatric Neuroscience, Department of Psychiatry, The Lausanne University Hospital, Lausanne, Switzerland
| | - Anne Vassalli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
40
|
Everett T, Ten Eyck TW, Wu CH, Shelowitz AL, Stansbury SM, Firek A, Setlow B, McIntyre JC. Cilia loss on distinct neuron populations differentially alters cocaine-induced locomotion and reward. J Psychopharmacol 2024; 38:200-212. [PMID: 38151883 PMCID: PMC11078551 DOI: 10.1177/02698811231219058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuronal primary cilia are being recognized for their role in mediating signaling associated with a variety of neurobehaviors, including responses to drugs of abuse. They function as signaling hubs, enriched with a diverse array of G-protein coupled receptors (GPCRs), including several associated with motivation and drug-related behaviors. However, our understanding of how cilia regulate neuronal function and behavior is still limited. AIMS The objective of the current study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to cocaine. METHODS To test the consequences of cilia loss on cocaine-induced locomotion and reward-related behavior, we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. RESULTS Cilia ablation on either population of neurons failed to significantly alter acute locomotor responses to cocaine at a range of doses. With repeated administration, mice lacking cilia on GAD2-GABAergic neurons showed no difference in locomotor sensitization to cocaine compared to wild-type (WT) littermates, whereas mice lacking cilia on dopaminergic neurons exhibited reduced locomotor sensitization to cocaine at 10 and 30 mg/kg. Mice lacking cilia on GAD2-GABAergic neurons showed no difference in cocaine conditioned place preference (CPP), whereas mice lacking cilia on dopaminergic neurons exhibited reduced CPP compared to WT littermates. CONCLUSIONS Combined with previous findings using amphetamine, our results show that behavioral effects of cilia ablation are cell- and drug type-specific, and that neuronal cilia contribute to modulation of both the locomotor-inducing and rewarding properties of cocaine.
Collapse
Affiliation(s)
- Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Tyler W. Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Chang-Hung Wu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | | | - Sofia M. Stansbury
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Alexandra Firek
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| |
Collapse
|
41
|
Ike KGO, Lamers SJC, Kaim S, de Boer SF, Buwalda B, Billeter JC, Kas MJH. The human neuropsychiatric risk gene Drd2 is necessary for social functioning across evolutionary distant species. Mol Psychiatry 2024; 29:518-528. [PMID: 38114631 PMCID: PMC11116113 DOI: 10.1038/s41380-023-02345-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
The Drd2 gene, encoding the dopamine D2 receptor (D2R), was recently indicated as a potential target in the etiology of lowered sociability (i.e., social withdrawal), a symptom of several neuropsychiatric disorders such as Schizophrenia and Major Depression. Many animal species show social withdrawal in response to stimuli, including the vinegar fly Drosophila melanogaster and mice, which also share most human disease-related genes. Here we will test for causality between Drd2 and sociability and for its evolutionary conserved function in these two distant species, as well as assess its mechanism as a potential therapeutic target. During behavioral observations in groups of freely interacting D. melanogaster, Drd2 homologue mutant showed decreased social interactions and locomotor activity. After confirming Drd2's social effects in flies, conditional transgenic mice lacking Drd2 in dopaminergic cells (autoreceptor KO) or in serotonergic cells (heteroreceptor KO) were studied in semi-natural environments, where they could freely interact. Autoreceptor KOs showed increased sociability, but reduced activity, while no overall effect of Drd2 deletion was observed in heteroreceptor KOs. To determine acute effects of D2R signaling on sociability, we also showed that a direct intervention with the D2R agonist Sumanirole decreased sociability in wild type mice, while the antagonist showed no effects. Using a computational ethological approach, this study demonstrates that Drd2 regulates sociability across evolutionary distant species, and that activation of the mammalian D2R autoreceptor, in particular, is necessary for social functioning.
Collapse
Affiliation(s)
- Kevin G O Ike
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Sanne J C Lamers
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Soumya Kaim
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Sietse F de Boer
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Bauke Buwalda
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jean-Christophe Billeter
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
42
|
Jin L, Sullivan HA, Zhu M, Lavin TK, Matsuyama M, Fu X, Lea NE, Xu R, Hou Y, Rutigliani L, Pruner M, Babcock KR, Ip JPK, Hu M, Daigle TL, Zeng H, Sur M, Feng G, Wickersham IR. Long-term labeling and imaging of synaptically connected neuronal networks in vivo using double-deletion-mutant rabies viruses. Nat Neurosci 2024; 27:373-383. [PMID: 38212587 PMCID: PMC10849964 DOI: 10.1038/s41593-023-01545-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/05/2023] [Indexed: 01/13/2024]
Abstract
Rabies-virus-based monosynaptic tracing is a widely used technique for mapping neural circuitry, but its cytotoxicity has confined it primarily to anatomical applications. Here we present a second-generation system for labeling direct inputs to targeted neuronal populations with minimal toxicity, using double-deletion-mutant rabies viruses. Viral spread requires expression of both deleted viral genes in trans in postsynaptic source cells. Suppressing this expression with doxycycline following an initial period of viral replication reduces toxicity to postsynaptic cells. Longitudinal two-photon imaging in vivo indicated that over 90% of both presynaptic and source cells survived for the full 12-week course of imaging. Ex vivo whole-cell recordings at 5 weeks postinfection showed that the second-generation system perturbs input and source cells much less than the first-generation system. Finally, two-photon calcium imaging of labeled networks of visual cortex neurons showed that their visual response properties appeared normal for 10 weeks, the longest we followed them.
Collapse
Affiliation(s)
- Lei Jin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Lingang Laboratory, Shanghai, China
| | - Heather A Sullivan
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mulangma Zhu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas K Lavin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Makoto Matsuyama
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas E Lea
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ran Xu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - YuanYuan Hou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luca Rutigliani
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Maxwell Pruner
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kelsey R Babcock
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacque Pak Kan Ip
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming Hu
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
43
|
Wu M, Zhang X, Feng S, Freda SN, Kumari P, Dumrongprechachan V, Kozorovitskiy Y. Dopamine pathways mediating affective state transitions after sleep loss. Neuron 2024; 112:141-154.e8. [PMID: 37922904 PMCID: PMC10841919 DOI: 10.1016/j.neuron.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/25/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023]
Abstract
The pathophysiology of affective disorders-particularly circuit-level mechanisms underlying bidirectional, periodic affective state transitions-remains poorly understood. In patients, disruptions of sleep and circadian rhythm can trigger transitions to manic episodes, whereas depressive states are reversed. Here, we introduce a hybrid automated sleep deprivation platform to induce transitions of affective states in mice. Acute sleep loss causes mixed behavioral states, featuring hyperactivity, elevated social and sexual behaviors, and diminished depressive-like behaviors, where transitions depend on dopamine (DA). Using DA sensor photometry and projection-targeted chemogenetics, we reveal that elevated DA release in specific brain regions mediates distinct behavioral changes in affective state transitions. Acute sleep loss induces DA-dependent enhancement in dendritic spine density and uncaging-evoked dendritic spinogenesis in the medial prefrontal cortex, whereas optically mediated disassembly of enhanced plasticity reverses the antidepressant effects of sleep deprivation on learned helplessness. These findings demonstrate that brain-wide dopaminergic pathways control sleep-loss-induced polymodal affective state transitions.
Collapse
Affiliation(s)
- Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL 60208, USA
| | - Xin Zhang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sihan Feng
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Pushpa Kumari
- 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
|
44
|
van der Merwe R, Nadel J, Copes-Finke D, Pawelko S, Scott J, Ghanem M, Fox M, Morehouse C, McLaughlin R, Maddox C, Albert-Lyons R, Malaki G, Groce V, Turocy A, Aggadi N, Jin X, Howard C. Characterization of striatal dopamine projections across striatal subregions in behavioral flexibility. Eur J Neurosci 2023; 58:4466-4486. [PMID: 36617434 PMCID: PMC10329096 DOI: 10.1111/ejn.15910] [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: 09/17/2021] [Revised: 12/13/2022] [Accepted: 12/30/2022] [Indexed: 01/10/2023]
Abstract
Behavioural flexibility is key to survival in a dynamic environmentWhile flexible, goal-directed behaviours are initially dependent on dorsomedial striatum, they become dependent on lateral striatum as behaviours become inflexible. Similarly, lesions of dopamine terminals in lateral striatum disrupt the development of inflexible habits. This work suggests that dopamine release in lateral striatum may drive inflexible behaviours, though few studies have investigated a causative role of subpopulations of striatal dopamine terminals in reversal learning, a measure of flexibility. Here, we performed two optogenetic experiments to activate dopamine terminals in dorsomedial (DMS), dorsolateral (DLS) or ventral (nucleus accumbens [NAc]) striatum in DAT-Cre mice that expressed channelrhodopsin-2 via viral injection (Experiment I) or through transgenic breeding with an Ai32 reporter line (Experiment II) to determine how specific dopamine subpopulations impact reversal learning. Mice performed a reversal task in which they self-stimulated DMS, DLS, or NAc dopamine terminals by pressing one of two levers before action-outcome lever contingencies were reversed. Largely consistent with presumed ventromedial/lateral striatal function, we found that mice self-stimulating medial dopamine terminals reversed lever preference following contingency reversal, while mice self-stimulating NAc showed parial flexibility, and DLS self-stimulation resulted in impaired reversal. Impairments in DLS mice were characterized by more regressive errors and reliance on lose-stay strategies following reversal, as well as reduced within-session learning, suggesting reward insensitivity and overreliance on previously learned actions. This study supports a model of striatal function in which DMS and ventral dopamine facilitate goal-directed responding, and DLS dopamine supports more inflexible responding.
Collapse
Affiliation(s)
- R.K. van der Merwe
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - J.A. Nadel
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
- Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA
| | - D. Copes-Finke
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - S. Pawelko
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - J.S. Scott
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - M. Ghanem
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - M. Fox
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - C. Morehouse
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - R. McLaughlin
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - C. Maddox
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - R. Albert-Lyons
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - G. Malaki
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - V. Groce
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - A. Turocy
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - N. Aggadi
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| | - X. Jin
- Center for Motor Control and Disease, Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
- NYU–ECNU Institute of Brain and Cognitive Science, New York University Shanghai, Shanghai 200062, China
| | - C.D. Howard
- Neuroscience Department, Oberlin College, 173 Lorain St., Oberlin, OH, USA
| |
Collapse
|
45
|
So WL, Hu J, Jeffs L, Dempsey H, Lockie SH, Zigman JM, Stark R, Reichenbach A, Andrews ZB. Ghrelin signalling in AgRP neurons links metabolic state to the sensory regulation of AgRP neural activity. Mol Metab 2023; 78:101826. [PMID: 37898450 PMCID: PMC10643323 DOI: 10.1016/j.molmet.2023.101826] [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: 06/06/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/30/2023] Open
Abstract
OBJECTIVE The sensory detection of food and food cues suppresses Agouti related peptide (AgRP) neuronal activity prior to consumption with greatest suppression occurring in response to highly caloric food or interoceptive energy need. However, the interoceptive mechanisms priming an appropriate AgRP neural response to external sensory information of food availability remain unexplored. Since hunger increases plasma ghrelin, we hypothesized that ghrelin receptor (GHSR) signalling on AgRP neurons is a key interoceptive mechanism integrating energy need with external sensory cues predicting caloric availability. METHODS We used in vivo photometry to measure the effects of ghrelin administration or fasting on AgRP neural activity with GCaMP6s and dopamine release in the nucleus accumbens with GRAB-DA in mice lacking ghrelin receptors in AgRP neurons. RESULTS The deletion of GHSR on AgRP neurons prevented ghrelin-induced food intake, motivation and AgRP activity. The presentation of food (peanut butter pellet) or a wooden dowel suppressed AgRP activity in fasted WT but not mice lacking GHSRs in AgRP neurons. Similarly, peanut butter and a wooden dowel increased dopamine release in the nucleus accumbens after ip ghrelin injection in WT but not mice lacking GHSRs in AgRP neurons. No difference in dopamine release was observed in fasted mice. Finally, ip ghrelin administration did not directly increase dopamine neural activity in the ventral tegmental area. CONCLUSIONS Our results suggest that AgRP GHSRs integrate an interoceptive state of energy need with external sensory information to produce an optimal change in AgRP neural activity. Thus, ghrelin signalling on AgRP neurons is more than just a feedback signal to increase AgRP activity during hunger.
Collapse
Affiliation(s)
- Wang Lok So
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Jiachen Hu
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Lotus Jeffs
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Harry Dempsey
- The Florey Institute of Neuroscience and Mental Health, Mental Health Division, Parkville, Melbourne, Australia
| | - Sarah H Lockie
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Romana Stark
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Alex Reichenbach
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton 3800, Victoria, Australia.
| |
Collapse
|
46
|
van Oostrum M, Blok TM, Giandomenico SL, Tom Dieck S, Tushev G, Fürst N, Langer JD, Schuman EM. The proteomic landscape of synaptic diversity across brain regions and cell types. Cell 2023; 186:5411-5427.e23. [PMID: 37918396 PMCID: PMC10686415 DOI: 10.1016/j.cell.2023.09.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/18/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023]
Abstract
Neurons build synaptic contacts using different protein combinations that define the specificity, function, and plasticity potential of synapses; however, the diversity of synaptic proteomes remains largely unexplored. We prepared synaptosomes from 7 different transgenic mouse lines with fluorescently labeled presynaptic terminals. Combining microdissection of 5 different brain regions with fluorescent-activated synaptosome sorting (FASS), we isolated and analyzed the proteomes of 18 different synapse types. We discovered ∼1,800 unique synapse-type-enriched proteins and allocated thousands of proteins to different types of synapses (https://syndive.org/). We identify shared synaptic protein modules and highlight the proteomic hotspots for synapse specialization. We reveal unique and common features of the striatal dopaminergic proteome and discover the proteome signatures that relate to the functional properties of different interneuron classes. This study provides a molecular systems-biology analysis of synapses and a framework to integrate proteomic information for synapse subtypes of interest with cellular or circuit-level experiments.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Thomas M Blok
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | | | | | - Georgi Tushev
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Nicole Fürst
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Julian D Langer
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
47
|
Martianova E, Sadretdinova R, Pageau A, Pausic N, Gentiletti TD, Leblanc D, Rivera AM, Labonté B, Proulx CD. Hypothalamic neuronal outputs transmit sensorimotor signals at the onset of locomotor initiation. iScience 2023; 26:108328. [PMID: 38026162 PMCID: PMC10665817 DOI: 10.1016/j.isci.2023.108328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/27/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
The lateral hypothalamus (LH) plays a critical role in sensory integration to organize behavior responses. However, how projection-defined LH neuronal outputs dynamically transmit sensorimotor signals to major downstream targets to organize behavior is unknown. Here, using multi-fiber photometry, we show that three major LH neuronal outputs projecting to the dorsal raphe nucleus (DRN), ventral tegmental area (VTA), and lateral habenula (LHb) exhibit significant coherent activity in mice engaging sensory-evoked or self-initiated motor responses. Increased activity at LH axon terminals precedes movement initiation during active coping responses and the activity of serotonin neurons and dopamine neurons. The optogenetic activation of LH axon terminals in either of the DRN, VTA, or LHb was sufficient to increase motor initiation but had different effects on passive avoidance and sucrose consumption. Our findings support the complementary role of three projection-defined LH neuronal outputs in the transmission of sensorimotor signals to major downstream regions at movement onset.
Collapse
Affiliation(s)
- Ekaterina Martianova
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Renata Sadretdinova
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Alicia Pageau
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Nikola Pausic
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Tommy Doucet Gentiletti
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Danahé Leblanc
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Arturo Marroquin Rivera
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Benoît Labonté
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Christophe D. Proulx
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| |
Collapse
|
48
|
Amo R, Uchida N, Watabe-Uchida M. Glutamate inputs send prediction error of reward but not negative value of aversive stimuli to dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566472. [PMID: 37986868 PMCID: PMC10659341 DOI: 10.1101/2023.11.09.566472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Midbrain dopamine neurons are thought to signal reward prediction errors (RPEs) but the mechanisms underlying RPE computation, particularly contributions of different neurotransmitters, remain poorly understood. Here we used a genetically-encoded glutamate sensor to examine the pattern of glutamate inputs to dopamine neurons. We found that glutamate inputs exhibit virtually all of the characteristics of RPE, rather than conveying a specific component of RPE computation such as reward or expectation. Notably, while glutamate inputs were transiently inhibited by reward omission, they were excited by aversive stimuli. Opioid analgesics altered dopamine negative responses to aversive stimuli toward more positive responses, while excitatory responses of glutamate inputs remained unchanged. Our findings uncover previously unknown synaptic mechanisms underlying RPE computations; dopamine responses are shaped by both synergistic and competitive interactions between glutamatergic and GABAergic inputs to dopamine neurons depending on valences, with competitive interactions playing a role in responses to aversive stimuli.
Collapse
Affiliation(s)
- Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
49
|
Kim HY, Lee J, Kim HJ, Lee BE, Jeong J, Cho EJ, Jang HJ, Shin KJ, Kim MJ, Chae YC, Lee SE, Myung K, Baik JH, Suh PG, Kim JI. PLCγ1 in dopamine neurons critically regulates striatal dopamine release via VMAT2 and synapsin III. Exp Mol Med 2023; 55:2357-2375. [PMID: 37907739 PMCID: PMC10689754 DOI: 10.1038/s12276-023-01104-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: 01/13/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 11/02/2023] Open
Abstract
Dopamine neurons are essential for voluntary movement, reward learning, and motivation, and their dysfunction is closely linked to various psychological and neurodegenerative diseases. Hence, understanding the detailed signaling mechanisms that functionally modulate dopamine neurons is crucial for the development of better therapeutic strategies against dopamine-related disorders. Phospholipase Cγ1 (PLCγ1) is a key enzyme in intracellular signaling that regulates diverse neuronal functions in the brain. It was proposed that PLCγ1 is implicated in the development of dopaminergic neurons, while the physiological function of PLCγ1 remains to be determined. In this study, we investigated the physiological role of PLCγ1, one of the key effector enzymes in intracellular signaling, in regulating dopaminergic function in vivo. We found that cell type-specific deletion of PLCγ1 does not adversely affect the development and cellular morphology of midbrain dopamine neurons but does facilitate dopamine release from dopaminergic axon terminals in the striatum. The enhancement of dopamine release was accompanied by increased colocalization of vesicular monoamine transporter 2 (VMAT2) at dopaminergic axon terminals. Notably, dopamine neuron-specific knockout of PLCγ1 also led to heightened expression and colocalization of synapsin III, which controls the trafficking of synaptic vesicles. Furthermore, the knockdown of VMAT2 and synapsin III in dopamine neurons resulted in a significant attenuation of dopamine release, while this attenuation was less severe in PLCγ1 cKO mice. Our findings suggest that PLCγ1 in dopamine neurons could critically modulate dopamine release at axon terminals by directly or indirectly interacting with synaptic machinery, including VMAT2 and synapsin III.
Collapse
Affiliation(s)
- Hye Yun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jieun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hyun-Jin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Byeong Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jaewook Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Eun Jeong Cho
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
| | - Kyeong Jin Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Min Ji Kim
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Pann-Ghill Suh
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
50
|
Heo JY, Park AH, Lee MJ, Ryu MJ, Kim YK, Jang YS, Kim SJ, Shin SY, Son HJ, Stein TD, Huh YH, Chung SK, Choi SY, Kim JM, Hwang O, Shong M, Hyeon SJ, Lee J, Ryu H, Kim D, Kweon GR. Crif1 deficiency in dopamine neurons triggers early-onset parkinsonism. Mol Psychiatry 2023; 28:4474-4484. [PMID: 37648779 DOI: 10.1038/s41380-023-02234-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.
Collapse
Affiliation(s)
- Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Ah Hyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Yun Seon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - So Yeon Shin
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jin Son
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
- VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Yang Hoon Huh
- Electron Microscopy Research center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Sookja K Chung
- Faculty of Medicine & Dr Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Song Yi Choi
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jin Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Minho Shong
- Graduate School of Medical Science and Education, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Junghee Lee
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|