1
|
Littlepage-Saunders M, Hochstein MJ, Chang DS, Johnson KA. G protein-coupled receptor modulation of striatal dopamine transmission: Implications for psychoactive drug effects. Br J Pharmacol 2024; 181:4399-4413. [PMID: 37258878 PMCID: PMC10687321 DOI: 10.1111/bph.16151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Dopamine transmission in the striatum is a critical mediator of the rewarding and reinforcing effects of commonly misused psychoactive drugs. G protein-coupled receptors (GPCRs) that bind a variety of neuromodulators including dopamine, endocannabinoids, acetylcholine and endogenous opioid peptides regulate dopamine release by acting on several components of dopaminergic circuitry. Striatal dopamine release can be driven by both somatic action potential firing and local mechanisms that depend on acetylcholine released from striatal cholinergic interneurons. GPCRs that primarily regulate somatic firing of dopamine neurons via direct effects or modulation of synaptic inputs are likely to affect distinct aspects of behaviour and psychoactive drug actions compared with those GPCRs that primarily regulate local acetylcholine-dependent dopamine release in striatal regions. This review will highlight mechanisms by which GPCRs modulate dopaminergic transmission and the relevance of these findings to psychoactive drug effects on physiology and behaviour.
Collapse
Affiliation(s)
- Mydirah Littlepage-Saunders
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Michael J Hochstein
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Doris S Chang
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Kari A Johnson
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Neuroscience Graduate Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Spicer MM, Weber MA, Luo Z, Yang J, Narayanan NS, Fisher RA. Regulator of G protein signaling 6 (RGS6) in dopamine neurons promotes EtOH seeking, behavioral reward, and susceptibility to relapse. Psychopharmacology (Berl) 2024; 241:2255-2269. [PMID: 38856764 PMCID: PMC11518640 DOI: 10.1007/s00213-024-06631-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Mesolimbic dopamine (DA) transmission is believed to play a critical role in mediating reward responses to drugs of abuse, including alcohol (EtOH). The neurobiological mechanisms underlying EtOH-seeking behavior and dependence are not fully understood, and abstinence remains the only effective way to prevent alcohol use disorders (AUDs). Here, we developed novel RGS6fl/fl; DAT-iCreER mice to determine the role of RGS6 in DA neurons on EtOH consumption, reward, and relapse behaviors. We found that RGS6 is expressed in DA neurons in both human and mouse ventral tegmental area (VTA), and that RGS6 loss in mice upregulates DA transporter (DAT) expression in VTA DA neuron synaptic terminals. Remarkably, loss of RGS6 in DA neurons significantly reduced EtOH consumption, preference, and reward in a manner indistinguishable from that seen in RGS6-/- mice. Strikingly, RGS6 loss from DA neurons before or after EtOH behavioral reward is established significantly reduced (~ 50%) re-instatement of reward following extinguishment, demonstrating distinct roles of RGS6 in promoting reward and relapse susceptibility to EtOH. These studies identify DA neurons as the locus of RGS6 action in promoting EtOH consumption, preference, reward, and relapse. RGS6 is unique among R7 RGS proteins in promoting rather than suppressing behavioral responses to drugs of abuse and to modulate EtOH behavioral reward. This is a result of RGS6's pre-synaptic actions that we hypothesize promote VTA DA transmission by suppressing GPCR-Gαi/o-DAT signaling in VTA DA neurons. These studies identify RGS6 as a potential therapeutic target for behavioral reward and relapse to EtOH.
Collapse
Affiliation(s)
- Mackenzie M Spicer
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Matthew A Weber
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Zili Luo
- Department of Pediatrics, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Jianqi Yang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
| | - Nandakumar S Narayanan
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA
- Department of Neurology, University of Iowa Carver College of Medicine, 200 Hawkins Dr., Iowa City, IA, 52242, USA
| | - Rory A Fisher
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 51 Newton Rd. BSB 2-512, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 169 Newton Rd., Iowa City, IA, 52242, USA.
| |
Collapse
|
3
|
Tiwari H, Kumar A, Barik MR, Kaur H, Mahajan S, Shukla MK, Gupta M, Yadav G, Nargotra A. Repositioning the existing drugs for neuroinflammation: a fusion of computational approach and biological validation to counter the Parkinson's disease progression. Mol Divers 2024; 28:2759-2770. [PMID: 37542020 DOI: 10.1007/s11030-023-10708-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Parkinson's disease is caused by the deficiency of striatal dopamine and the accumulation of aggregated α-synuclein in the substantia nigra pars compacta (SNpc). Neuroinflammation associated with oxidative stress is a key factor contributing to the death of dopaminergic neurons in SNpc and advancement of Parkinson's disease. Two molecular targets, i.e., nuclear factor kappa-light-chain-enhancer (NF-kB) and α-synuclein play a substantial role in neuroinflammation progression. Therefore, the compounds targeting these neuroinflammatory targets hold a great potential to combat Parkinson's disease. Thereby, in this study, molecular docking and Connectivity Map (CMap) based gene expression profiling was utilized to reposition the approved drugs as neuroprotective agents for Parkinson's disease. With in silico screening, two drugs namely theophylline and propylthiouracil were selected for anti-neuroinflammatory activity evaluation in in vivo models of chronic neuroinflammation. The neuroinflammatory effect of the identified compounds was confirmed by quantifying the expression of three important neuroinflammatory mediators, i.e. IL-6, TNF-alpha, and IL-1 beta on brain tissue using ELISA assay. The ELISA experiment demonstrated that both compounds significantly decreased the expression of neuroinflammatory mediators, highlighting the compounds' potential in neuroinflammation management. Furthermore, the drug and disease interaction network of the two identified drugs and diseases (neuroinflammation and Parkinson's disease) suggested that the two drugs might interact with various targets namely adenosine receptors, Poly [ADP-ribose] polymerase-1, myeloperoxidase (MPO) and thyroid peroxidase through multiple pathways associated with neuroinflammation and Parkinson's disease. Computational studies suggest that a particular drug may be effective in managing Parkinson's disease associated with neuroinflammation. However, further research is needed to confirm this in biological experiments.
Collapse
Affiliation(s)
- Harshita Tiwari
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Amit Kumar
- Mutagenicity Laboratory, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manas Ranjan Barik
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Harjot Kaur
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Shubham Mahajan
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Monu Kumar Shukla
- PK-PD Toxicology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Monika Gupta
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Govind Yadav
- Mutagenicity Laboratory, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Amit Nargotra
- Discovery Informatics, NPMC Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.
| |
Collapse
|
4
|
Saenz J, Khezerlou E, Aggarwal M, Shaikh A, Ganti N, Herborg F, Pan PY. Parkinson's disease gene, Synaptojanin1, dysregulates the surface maintenance of the dopamine transporter. NPJ Parkinsons Dis 2024; 10:148. [PMID: 39117637 PMCID: PMC11310474 DOI: 10.1038/s41531-024-00769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Missense mutations of PARK20/SYNJ1 (synaptojanin1/Synj1) were found in complex forms of familial Parkinsonism. However, the Synj1-regulated molecular and cellular changes associated with dopaminergic dysfunction remain unknown. We now report a fast depletion of evoked dopamine and impaired maintenance of the axonal dopamine transporter (DAT) in the Synj1 haploinsufficient (Synj1+/-) neurons. While Synj1 has been traditionally known to facilitate the endocytosis of synaptic vesicles, we provide in vitro and in vivo evidence demonstrating that Synj1 haploinsufficiency results in an increase of total DAT but a reduction of the surface DAT. Synj1+/- neurons exhibit maladaptive DAT trafficking, which could contribute to the altered DA release. We showed that the loss of surface DAT is associated with the impaired 5'-phosphatase activity and the hyperactive PI(4,5)P2-PKCβ pathway downstream of Synj1 deficiency. Thus, our findings provided important mechanistic insight for Synj1-regulated DAT trafficking integral to dysfunctional DA signaling, which might be relevant to early Parkinsonism.
Collapse
Affiliation(s)
- Jacqueline Saenz
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
- Rutgers Graduate School of Biomedical Sciences, Molecular Biosciences Graduate Program, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Elnaz Khezerlou
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Meha Aggarwal
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Amina Shaikh
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Naga Ganti
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Freja Herborg
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ, 08854, USA.
| |
Collapse
|
5
|
Mayer FP, Stewart A, Varman DR, Moritz AE, Foster JD, Owens AW, Areal LB, Gowrishankar R, Velez M, Wickham K, Phelps H, Katamish R, Rabil M, Jayanthi LD, Vaughan RA, Daws LC, Blakely RD, Ramamoorthy S. Kappa Opioid Receptor Antagonism Restores Phosphorylation, Trafficking and Behavior induced by a Disease Associated Dopamine Transporter Variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.03.539310. [PMID: 37205452 PMCID: PMC10187322 DOI: 10.1101/2023.05.03.539310] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Aberrant dopamine (DA) signaling is implicated in schizophrenia, bipolar disorder (BPD), autism spectrum disorder (ASD), substance use disorder, and attention-deficit/hyperactivity disorder (ADHD). Treatment of these disorders remains inadequate, as exemplified by the therapeutic use of d-amphetamine and methylphenidate for the treatment of ADHD, agents with high abuse liability. In search for an improved and non-addictive therapeutic approach for the treatment of DA-linked disorders, we utilized a preclinical mouse model expressing the human DA transporter (DAT) coding variant DAT Val559, previously identified in individuals with ADHD, ASD, or BPD. DAT Val559, like several other disease-associated variants of DAT, exhibits anomalous DA efflux (ADE) that can be blocked by d-amphetamine and methylphenidate. Kappa opioid receptors (KORs) are expressed by DA neurons and modulate DA release and clearance, suggesting that targeting KORs might also provide an alternative approach to normalizing DA-signaling disrupted by perturbed DAT function. Here we demonstrate that KOR stimulation leads to enhanced surface trafficking and phosphorylation of Thr53 in wildtype DAT, effects achieved constitutively by the Val559 mutant. Moreover, these effects can be rescued by KOR antagonism of DAT Val559 in ex vivo preparations. Importantly, KOR antagonism also corrected in vivo DA release as well as sex-dependent behavioral abnormalities observed in DAT Val559 mice. Given their low abuse liability, our studies with a construct valid model of human DA associated disorders reinforce considerations of KOR antagonism as a pharmacological strategy to treat DA associated brain disorders.
Collapse
Affiliation(s)
- Felix P. Mayer
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy E. Moritz
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Anthony W. Owens
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Lorena B. Areal
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Raajaram Gowrishankar
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Michelle Velez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Kyria Wickham
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Hannah Phelps
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Rania Katamish
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian Rabil
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Lankupalle D. Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
6
|
Zhang AQ, Ralph MR, Stinchcombe AR. A mathematical model for the role of dopamine-D2 self-regulation in the production of ultradian rhythms. PLoS Comput Biol 2024; 20:e1012082. [PMID: 38701077 PMCID: PMC11095719 DOI: 10.1371/journal.pcbi.1012082] [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: 11/09/2023] [Revised: 05/15/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Many self-motivated and goal-directed behaviours display highly flexible, approximately 4 hour ultradian (shorter than a day) oscillations. Despite lacking direct correspondence to physical cycles in the environment, these ultradian rhythms may be involved in optimizing functional interactions with the environment and reflect intrinsic neural dynamics. Current evidence supports a role of mesostriatal dopamine (DA) in the expression and propagation of ultradian rhythmicity, however, the biochemical processes underpinning these oscillations remain to be identified. Here, we use a mathematical model to investigate D2 autoreceptor-dependent DA self-regulation as the source of ultradian behavioural rhythms. DA concentration at the midbrain-striatal synapses is governed through a dual-negative feedback-loop structure, which naturally gives rise to rhythmicity. This model shows the propensity of striatal DA to produce an ultradian oscillation characterized by a flexible period that is highly sensitive to parameter variations. Circadian (approximately 24 hour) regulation consolidates the ultradian oscillations and alters their response to the phase-dependent, rapid-resetting effect of a transient excitatory stimulus. Within a circadian framework, the ultradian rhythm orchestrates behavioural activity and enhances responsiveness to an external stimulus. This suggests a role for the circadian-ultradian timekeeping hierarchy in governing organized behaviour and shaping daily experience through coordinating the motivation to engage in recurring, albeit not highly predictable events, such as social interactions.
Collapse
Affiliation(s)
- An Qi Zhang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Martin R. Ralph
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
7
|
Saenz J, Khezerlou E, Aggarwal M, Shaikh A, Ganti N, Herborg F, Pan PY. Parkinson's disease gene, Synaptojanin1, dysregulates the surface maintenance of the dopamine transporter. RESEARCH SQUARE 2024:rs.3.rs-4021466. [PMID: 38559229 PMCID: PMC10980101 DOI: 10.21203/rs.3.rs-4021466/v1] [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
Missense mutations of PARK20/SYNJ1 (synaptojanin1/Synj1) have been linked to complex forms of familial parkinsonism, however, the molecular and cellular changes associated with dopaminergic dysfunction remains unknown. We now report fast depletion of evoked dopamine (DA) and altered maintenance of the axonal dopamine transporter (DAT) in the Synj1+/- neurons. While Synj1 has been traditionally known to facilitate the endocytosis of synaptic vesicles, we demonstrated that axons of cultured Synj1+/- neurons exhibit an increase of total DAT but a reduction of the surface DAT, which could be exacerbated by neuronal activity. We revealed that the loss of surface DAT is specifically associated with the impaired 5'-phosphatase activity of Synj1 and the hyperactive downstream PI(4,5)P2-PKCβ pathway. Thus, our findings provided important mechanistic insight for Synj1-regulated DAT trafficking integral to dysfunctional DA signaling in early parkinsonism.
Collapse
Affiliation(s)
- Jacqueline Saenz
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
- Rutgers Graduate School of Biomedical Sciences, Molecular Biosciences Graduate Program, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Elnaz Khezerlou
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Meha Aggarwal
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Amina Shaikh
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Naga Ganti
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Freja Herborg
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, 675 Hoes Lane West, Piscataway, NJ 08854, USA
| |
Collapse
|
8
|
Suzuki N, Hiraide S, Shikanai H, Isshiki T, Yamaguchi T, Izumi T, Iizuka K. Impaired monoamine neural system in the mPFC of SHRSP/Ezo as an animal model of attention-deficit/hyperactivity disorder. J Pharmacol Sci 2024; 154:61-71. [PMID: 38246729 DOI: 10.1016/j.jphs.2023.12.002] [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: 09/24/2023] [Revised: 11/16/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is the most common childhood-onset psychiatric disorder. We investigated the effects of systemic administration of monoamine reuptake inhibitors on long-term potentiation (LTP) formation and monoamine release in the medial prefrontal cortex (mPFC) of the stroke-prone spontaneously hypertensive rat (SHRSP)/Ezo, an animal model of ADHD, and its genetic control, Wistar Kyoto (WKY)/Ezo, to elucidate the functional changes in the mPFC monoamine neural system. Methylphenidate (dopamine (DA) and noradrenaline (NA) reuptake inhibitor) and desipramine (NA reuptake inhibitor) improved LTP formation defects in the mPFC of SHRSP/Ezo, suggesting that NA or both DA and NA are required for improvement of impaired LTP. Methylphenidate increased mPFC DA in both WKY/Ezo and SHRSP/Ezo, but the increase was greater in the former. GBR-12909 (DA reuptake inhibitor) increased mPFC DA in WKY/Ezo but had no effect in SHRSP/Ezo. This may be because DA transporter in SHRSP/Ezo is functionally impaired and contributes less to DA reuptake, so its inhibition did not increase DA level. Meanwhile, basal DA levels in the mPFC of SHRSP/Ezo were paradoxically decreased. These results suggest that functional changes in the DA and NA neural system in the frontal lobe are involved in the pathology of ADHD.
Collapse
Affiliation(s)
- Naoya Suzuki
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Sachiko Hiraide
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Hiroki Shikanai
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan; Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan.
| | - Takeru Isshiki
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Taku Yamaguchi
- Department of Pharmacotherapeutics and Neuropsychopharmacology, Faculty of Pharmaceutical Sciences, Nagasaki International University, Nagasaki, 2825-7 Huis Ten Bosch Sasebo, Nagasaki, 859-3298, Japan
| | - Takeshi Izumi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan; Advanced Research Promotion Center, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| | - Kenji Iizuka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| |
Collapse
|
9
|
Li A, Li W, Ali T, Yang C, Liu Z, Gao R, He K, Liu XA, Chen Z, Yu ZJ, Li T, Li S. A novel dopamine D2 receptor-NR2B protein complex might contribute to morphine use disorders. Eur J Pharmacol 2023; 961:176174. [PMID: 37939993 DOI: 10.1016/j.ejphar.2023.176174] [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: 09/20/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Dopamine receptors can form heteromeric interactions with other receptors, including glutamate receptors, and present a novel pharmacological target because it contribute to dopamine-dysregulated brain disorders such as addiction and other motor-related diseases. In addition, dopamine receptors D2 (D2Rs) and glutamate NMDA receptors subtype-NR2B have been implicated in morphine use disorders; however, the molecular mechanism underlying the heteromeric complex of these two receptors in morphine use disorders is unclear. Herein, we focus on interactions between D2R and NR2B in morphine-induced conditioned place preference (CPP) and hyperlocomotion mice models. We found that the D2R-NR2B complex significantly increases in morphine-induced mice models, accompanied by ERK signaling impairment, implying the complex could contribute to the morphine addiction pathophysiological process. Further, we design a brain-penetrant interfering peptide (TAT-D2-KT), which could disrupt interactions of D2R-NR2B and decrease addictive-like behaviors concurrent to ERK signaling improvement. In summary, our data provided the first evidence for a D2R-NMDAR complex formation in morphine use disorders and its underlying mechanism of ERK signaling, which could present a novel therapeutic target with direct implications for morphine acquisition and relapse treatment.
Collapse
Affiliation(s)
- Axiang Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China; State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Weifen Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Tahir Ali
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China; Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| | - Canyu Yang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China; State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Zizhen Liu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Ruyan Gao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Kaiwu He
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Zuxin Chen
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), China.
| | - Zhi-Jian Yu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, The 6th Affiliated Hospital of Shenzhen University Health Science Center, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| | - Tao Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China; NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
| | - Shupeng Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China; Shenzhen Bay Laboratory, Shenzhen, 518055, China; Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
10
|
Zald DH. The influence of dopamine autoreceptors on temperament and addiction risk. Neurosci Biobehav Rev 2023; 155:105456. [PMID: 37926241 PMCID: PMC11330662 DOI: 10.1016/j.neubiorev.2023.105456] [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: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
As a major regulator of dopamine (DA), DA autoreceptors (DAARs) exert substantial influence over DA-mediated behaviors. This paper reviews the physiological and behavioral impact of DAARs. Individual differences in DAAR functioning influences temperamental traits such as novelty responsivity and impulsivity, both of which are associated with vulnerability to addictive behavior in animal models and a broad array of externalizing behaviors in humans. DAARs additionally impact the response to psychostimulants and other drugs of abuse. Human PET studies of D2-like receptors in the midbrain provide evidence for parallels to the animal literature. These data lead to the proposal that weak DAAR regulation is a risk factor for addiction and externalizing problems. The review highlights the potential to build translational models of the functional role of DAARs in behavior. It also draws attention to key limitations in the current literature that would need to be addressed to further advance a weak DAAR regulation model of addiction and externalizing risk.
Collapse
Affiliation(s)
- David H Zald
- Center for Advanced Human Brain Imaging and Department of Psychiatry, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
11
|
Davis SE, Cirincione AB, Jimenez-Torres AC, Zhu J. The Impact of Neurotransmitters on the Neurobiology of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:15340. [PMID: 37895020 PMCID: PMC10607327 DOI: 10.3390/ijms242015340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide. Neurodegenerative diseases result from progressive damage to nerve cells in the brain or peripheral nervous system connections that are essential for cognition, coordination, strength, sensation, and mobility. Dysfunction of these brain and nerve functions is associated with Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and motor neuron disease. In addition to these, 50% of people living with HIV develop a spectrum of cognitive, motor, and/or mood problems collectively referred to as HIV-Associated Neurocognitive Disorders (HAND) despite the widespread use of a combination of antiretroviral therapies. Neuroinflammation and neurotransmitter systems have a pathological correlation and play a critical role in developing neurodegenerative diseases. Each of these diseases has a unique pattern of dysregulation of the neurotransmitter system, which has been attributed to different forms of cell-specific neuronal loss. In this review, we will focus on a discussion of the regulation of dopaminergic and cholinergic systems, which are more commonly disturbed in neurodegenerative disorders. Additionally, we will provide evidence for the hypothesis that disturbances in neurotransmission contribute to the neuronal loss observed in neurodegenerative disorders. Further, we will highlight the critical role of dopamine as a mediator of neuronal injury and loss in the context of NeuroHIV. This review will highlight the need to further investigate neurotransmission systems for their role in the etiology of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (S.E.D.); (A.B.C.); (A.C.J.-T.)
| |
Collapse
|
12
|
Shetty M, Bolland DE, Morrell J, Grove BD, Foster JD, Vaughan RA. Dopamine transporter membrane mobility is bidirectionally regulated by phosphorylation and palmitoylation. Curr Res Physiol 2023; 6:100106. [PMID: 38107792 PMCID: PMC10724222 DOI: 10.1016/j.crphys.2023.100106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023] Open
Abstract
The primary regulator of dopamine availability in the brain is the dopamine transporter (DAT), a plasma membrane protein that drives reuptake of released dopamine from the extracellular space into the presynaptic neuron. DAT activity is regulated by post-translational modifications that establish clearance capacity through impacts on transport kinetics, and dysregulation of these events may underlie dopaminergic imbalances in mood and psychiatric disorders. Here, using fluorescence recovery after photobleaching, we show that phosphorylation and palmitoylation induce opposing effects on DAT lateral membrane mobility, which may influence functional outcomes by regulating subcellular localization and binding partner interactions. Membrane mobility was also impacted by amphetamine and in polymorphic variant A559V in directions consistent with enhanced phosphorylation. These findings grow the list of DAT properties controlled by these post-translational modifications and highlight their role in establishment of dopaminergic tone in physiological and pathophysiological states.
Collapse
Affiliation(s)
- Madhur Shetty
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | | | - Joshua Morrell
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Bryon D. Grove
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| |
Collapse
|
13
|
Santiago NA, He B, Howard SL, Beaudin S, Strupp BJ, Smith DR. Developmental Manganese Exposure Causes Lasting Attention Deficits Accompanied by Dysregulation of mTOR Signaling and Catecholaminergic Gene Expression in Brain Prefrontal Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549215. [PMID: 37503220 PMCID: PMC10370122 DOI: 10.1101/2023.07.16.549215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Elevated manganese (Mn) exposure is associated with attentional deficits in children, and is an environmental risk factor for attention deficit hyperactivity disorder (ADHD). We have shown that developmental Mn exposure causes lasting attention and sensorimotor deficits in a rat model of early childhood Mn exposure, and that these deficits are associated with a hypofunctioning catecholaminergic system in the prefrontal cortex (PFC), though the mechanistic basis for these deficits is not well understood. To address this, male Long-Evans rats were exposed orally to Mn (50 mg/kg/d) over PND 1-21 and attentional function was assessed in adulthood using the 5-Choice Serial Reaction Time Task. Targeted catecholaminergic system and epigenetic gene expression, followed by unbiased differential DNA methylation and gene regulation expression transcriptomics in the PFC, were performed in young adult littermates. Results show that developmental Mn exposure causes lasting focused attention deficits that are associated with reduced gene expression of tyrosine hydroxylase, dopamine transporter, and DNA methyltransferase 3a. Further, developmental Mn exposure causes broader lasting methylation and gene expression dysregulation associated with epigenetic regulation, inflammation, cell development, and hypofunctioning catecholaminergic neuronal systems. Pathway enrichment analyses uncovered mTOR and Wnt signaling pathway genes as significant transcriptomic regulators of the Mn altered transcriptome, and Western blot of total, C1 and C2 phospho-mTOR confirmed mTOR pathway dysregulation. Our findings deepen our understanding of the mechanistic basis of how developmental Mn exposure leads to lasting catecholaminergic dysfunction and attention deficits, which may aid future therapeutic interventions of environmental exposure associated disorders. Significance Statement Attention deficit hyperactivity disorder (ADHD) is associated with environmental risk factors, including exposure to neurotoxic agents. Here we used a rodent model of developmental manganese (Mn) exposure producing lasting attention deficits to show broad epigenetic and gene expression changes in the prefrontal cortex, and to identify disrupted mTOR and Wnt signaling pathways as a novel mechanism for how developmental Mn exposure may induce lasting attention and catecholaminergic system impairments. Importantly, our findings establish early development as a critical period of susceptibility to lasting deficits in attentional function caused by elevated environmental toxicant exposure. Given that environmental health threats disproportionately impact communities of color and low socioeconomic status, our findings can aid future studies to assess therapeutic interventions for vulnerable populations.
Collapse
|
14
|
Caridade-Silva R, Araújo B, Martins-Macedo J, Teixeira FG. N-Acetylcysteine Treatment May Compensate Motor Impairments through Dopaminergic Transmission Modulation in a Striatal 6-Hydroxydopamine Parkinson's Disease Rat Model. Antioxidants (Basel) 2023; 12:1257. [PMID: 37371987 DOI: 10.3390/antiox12061257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Preventing degeneration and the loss of dopaminergic neurons (DAn) in the brain while mitigating motor symptoms remains a challenge in Parkinson's Disease (PD) treatment development. In light of this, developing or repositioning potential disease-modifying approaches is imperative to achieve meaningful translational gains in PD research. Under this concept, N-acetylcysteine (NAC) has revealed promising perspectives in preserving the dopaminergic system capability and modulating PD mechanisms. Although NAC has been shown to act as an antioxidant and (neuro)protector of the brain, it has yet to be acknowledged how this repurposed drug can improve motor symptomatology and provide disease-modifying properties in PD. Therefore, in the present work, we assessed the impact of NAC on motor and histological deficits in a striatal 6-hydroxydopamine (6-OHDA) rat model of PD. The results revealed that NAC enhanced DAn viability, as we found that it could restore dopamine transporter (DAT) levels compared to the untreated 6-OHDA group. Such findings were positively correlated with a significant amelioration in the motor outcomes of the 6-OHDA-treated animals, demonstrating that NAC may, somehow, be a modulator of PD degenerative mechanisms. Overall, we postulated a proof-of-concept milestone concerning the therapeutic application of NAC. Nevertheless, it is extremely important to understand the complexity of this drug and how its therapeutical properties interact with the cellular and molecular PD mechanisms.
Collapse
Affiliation(s)
- Rita Caridade-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| | - Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| | - Joana Martins-Macedo
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| | - Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research, School of Health, Polytechnic University of Porto, 4200-465 Porto, Portugal
| |
Collapse
|
15
|
Su P, Yan S, Yang J, Tong J, Samsom J, You F, Li Y, Chen Q, Jiang A, Zhai D, Chen J, Sun Z, Zhou J, Liu M, Lee FJS, Xu ZQD, Wang X, Vasdev N, Wong AHC, Liu F. Serum amyloid P component (SAP) modulates antidepressant effects through promoting membrane insertion of the serotonin transporter. Neuropsychopharmacology 2023; 48:508-517. [PMID: 36076020 PMCID: PMC9852251 DOI: 10.1038/s41386-022-01449-4] [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: 03/31/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 02/02/2023]
Abstract
Serum amyloid P component (SAP) is a universal constituent of human amyloid deposits including those in Alzheimer's disease. SAP has been observed to be elevated in patients with depression, and higher SAP levels are associated with better response to the antidepressant escitalopram. The mechanisms underlying these clinical observations remain unclear. We examined the effect of SAP on serotonin transporter (SERT) expression and localization using Western blot, confocal microscopy, and positron emission tomography with the radioligand [11C]DASB. We also investigated the effect of SAP on treatment response to escitalopram in mice with the forced swim test (FST), a classical behaviour paradigm to assess antidepressant effects. SAP reduced [11C]DASB binding as an index of SERT levels, consistent with Western blots showing decreased total SAP protein because of increased protein degradation. In conjunction with the global decrease in SERT levels, SAP also promotes VAMP-2 mediated SERT membrane insertion. SAP levels are correlated with behavioural despair and SSRI treatment response in mice with FST. In MDD patients, the SAP and membrane SERT levels are correlated with response to SSRI treatment. SAP has complex effects on SERT levels and localization, thereby modulating the effect of SSRIs, which could partially explain clinical variability in antidepressant treatment response. These results add to our understanding of the mechanism for antidepressant drug action, and with further work could be of clinical utility.
Collapse
Affiliation(s)
- Ping Su
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, Beijing, China
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Shuxin Yan
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, Beijing, China
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jian Yang
- Beijing AnDing Hospital, Capital Medical University, Beijing, China, Beijing, China
| | - Junchao Tong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - James Samsom
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Fan You
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, Beijing, China
| | - Yun Li
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, Beijing, China
| | - Qiuyue Chen
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, Beijing, China
| | - Anlong Jiang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Dongxu Zhai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jiahao Chen
- Departments of Neurobiology and Pathology, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Zuoli Sun
- Beijing AnDing Hospital, Capital Medical University, Beijing, China, Beijing, China
| | - Jingjing Zhou
- Beijing AnDing Hospital, Capital Medical University, Beijing, China, Beijing, China
| | - Min Liu
- Beijing AnDing Hospital, Capital Medical University, Beijing, China, Beijing, China
| | - Frank J S Lee
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Zhi-Qing David Xu
- Departments of Neurobiology and Pathology, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Neil Vasdev
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Departments of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Albert H C Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Departments of Psychiatry, University of Toronto, Toronto, ON, Canada
- Departments of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Fang Liu
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, Beijing, China.
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Departments of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Departments of Physiology, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
16
|
Nepal B, Das S, Reith ME, Kortagere S. Overview of the structure and function of the dopamine transporter and its protein interactions. Front Physiol 2023; 14:1150355. [PMID: 36935752 PMCID: PMC10020207 DOI: 10.3389/fphys.2023.1150355] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The dopamine transporter (DAT) plays an integral role in dopamine neurotransmission through the clearance of dopamine from the extracellular space. Dysregulation of DAT is central to the pathophysiology of numerous neuropsychiatric disorders and as such is an attractive therapeutic target. DAT belongs to the solute carrier family 6 (SLC6) class of Na+/Cl- dependent transporters that move various cargo into neurons against their concentration gradient. This review focuses on DAT (SCL6A3 protein) while extending the narrative to the closely related transporters for serotonin and norepinephrine where needed for comparison or functional relevance. Cloning and site-directed mutagenesis experiments provided early structural knowledge of DAT but our contemporary understanding was achieved through a combination of crystallization of the related bacterial transporter LeuT, homology modeling, and subsequently the crystallization of drosophila DAT. These seminal findings enabled a better understanding of the conformational states involved in the transport of substrate, subsequently aiding state-specific drug design. Post-translational modifications to DAT such as phosphorylation, palmitoylation, ubiquitination also influence the plasma membrane localization and kinetics. Substrates and drugs can interact with multiple sites within DAT including the primary S1 and S2 sites involved in dopamine binding and novel allosteric sites. Major research has centered around the question what determines the substrate and inhibitor selectivity of DAT in comparison to serotonin and norepinephrine transporters. DAT has been implicated in many neurological disorders and may play a role in the pathology of HIV and Parkinson's disease via direct physical interaction with HIV-1 Tat and α-synuclein proteins respectively.
Collapse
Affiliation(s)
- Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Sanjay Das
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Maarten E. Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- *Correspondence: Sandhya Kortagere,
| |
Collapse
|
17
|
Rosikon KD, Bone MC, Lawal HO. Regulation and modulation of biogenic amine neurotransmission in Drosophila and Caenorhabditis elegans. Front Physiol 2023; 14:970405. [PMID: 36875033 PMCID: PMC9978017 DOI: 10.3389/fphys.2023.970405] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
Neurotransmitters are crucial for the relay of signals between neurons and their target. Monoamine neurotransmitters dopamine (DA), serotonin (5-HT), and histamine are found in both invertebrates and mammals and are known to control key physiological aspects in health and disease. Others, such as octopamine (OA) and tyramine (TA), are abundant in invertebrates. TA is expressed in both Caenorhabditis elegans and Drosophila melanogaster and plays important roles in the regulation of essential life functions in each organism. OA and TA are thought to act as the mammalian homologs of epinephrine and norepinephrine respectively, and when triggered, they act in response to the various stressors in the fight-or-flight response. 5-HT regulates a wide range of behaviors in C. elegans including egg-laying, male mating, locomotion, and pharyngeal pumping. 5-HT acts predominantly through its receptors, of which various classes have been described in both flies and worms. The adult brain of Drosophila is composed of approximately 80 serotonergic neurons, which are involved in modulation of circadian rhythm, feeding, aggression, and long-term memory formation. DA is a major monoamine neurotransmitter that mediates a variety of critical organismal functions and is essential for synaptic transmission in invertebrates as it is in mammals, in which it is also a precursor for the synthesis of adrenaline and noradrenaline. In C. elegans and Drosophila as in mammals, DA receptors play critical roles and are generally grouped into two classes, D1-like and D2-like based on their predicted coupling to downstream G proteins. Drosophila uses histamine as a neurotransmitter in photoreceptors as well as a small number of neurons in the CNS. C. elegans does not use histamine as a neurotransmitter. Here, we review the comprehensive set of known amine neurotransmitters found in invertebrates, and discuss their biological and modulatory functions using the vast literature on both Drosophila and C. elegans. We also suggest the potential interactions between aminergic neurotransmitters systems in the modulation of neurophysiological activity and behavior.
Collapse
Affiliation(s)
- Katarzyna D Rosikon
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, United States
| | - Megan C Bone
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, United States
| | - Hakeem O Lawal
- Neuroscience Program, Department of Biological Sciences, Delaware State University, Dover, DE, United States
| |
Collapse
|
18
|
Stewart A, Mayer FP, Gowrishankar R, Davis GL, Areal LB, Gresch PJ, Katamish RM, Peart R, Stilley SE, Spiess K, Rabil MJ, Diljohn FA, Wiggins AE, Vaughan RA, Hahn MK, Blakely RD. Behaviorally penetrant, anomalous dopamine efflux exposes sex and circuit dependent regulation of dopamine transporters. Mol Psychiatry 2022; 27:4869-4880. [PMID: 36117213 DOI: 10.1038/s41380-022-01773-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 08/18/2022] [Accepted: 08/31/2022] [Indexed: 01/19/2023]
Abstract
Virtually all neuropsychiatric disorders display sex differences in prevalence, age of onset, and/or clinical symptomology. Although altered dopamine (DA) signaling is a feature of many of these disorders, sex-dependent mechanisms uniquely responsive to DA that drive sex-dependent behaviors remain unelucidated. Previously, we established that anomalous DA efflux (ADE) is a prominent feature of the DA transporter (DAT) variant Val559, a coding substitution identified in two male-biased disorders: attention-deficit/hyperactivity disorder and autism spectrum disorder. In vivo, Val559 ADE induces activation of nigrostriatal D2-type DA autoreceptors (D2ARs) that magnifies inappropriate, nonvesicular DA release by elevating phosphorylation and surface trafficking of ADE-prone DAT proteins. Here we demonstrate that DAT Val559 mice exhibit sex-dependent alterations in psychostimulant responses, social behavior, and cognitive performance. In a search for underlying mechanisms, we discovered that the ability of ADE to elicit D2AR regulation of DAT is both sex and circuit-dependent, with dorsal striatum D2AR/DAT coupling evident only in males, whereas D2AR/DAT coupling in the ventral striatum is exclusive to females. Moreover, systemic administration of the D2R antagonist sulpiride, which precludes ADE-driven DAT trafficking, can normalize DAT Val559 behavioral changes unique to each sex and without effects on the opposite sex or wildtype mice. Our studies support the sex- and circuit dependent capacity of D2ARs to regulate DAT as a critical determinant of the sex-biased effects of perturbed DA signaling in neurobehavioral disorders. Moreover, our work provides a cogent example of how a shared biological insult drives alternative physiological and behavioral trajectories as opposed to resilience.
Collapse
Affiliation(s)
- Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Gwynne L Davis
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Lorena B Areal
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Paul J Gresch
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Rania M Katamish
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Rodeania Peart
- Wilkes Honors College, Florida Atlantic University, Jupiter, FL, USA
| | - Samantha E Stilley
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Keeley Spiess
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian J Rabil
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | | | - Angelica E Wiggins
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Maureen K Hahn
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA.,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, USA. .,Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
19
|
Huang L, Zhang W, Han Y, Tang Y, Zhou W, Liu G, Shi W. Anti-Depressant Fluoxetine Hampers Olfaction of Goldfish by Interfering with the Initiation, Transmission, and Processing of Olfactory Signals. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:15848-15859. [PMID: 36260920 DOI: 10.1021/acs.est.2c02987] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The ubiquitous presence of fluoxetine (FLX) in aquatic environments poses great threat to fish species. However, little is known about its deleterious impacts on fish olfaction. In this study, the olfactory toxicity of FLX at environmentally realistic levels was assessed by monitoring the behavioral and electroolfactogram (EOG) responses to olfactory stimuli with goldfish (Carassius auratus), and the toxification mechanisms underlying the observed olfaction dysfunction were also investigated. Our results showed that the behavioral and EOG responses of goldfish to olfactory stimuli were significantly weakened by FLX, indicating an evident toxicity of FLX to olfaction. Moreover, FLX exposure led to significant alterations in olfactory initiation-related genes, suppression of ion pumps (Ca2+-ATPase and Na+/K+-ATPase), tissue lesions, and fewer olfactory sensory neurons in olfactory epithelium. In addition to altering the expression of olfactory transmission-related genes, comparative metabolomic analysis found that olfaction-related neurotransmitters (i.e., l-glutamate and acetylcholine) and the olfactory transduction pathway were significantly affected by FLX. Furthermore, evident tissue lesions, aggravated lipid peroxidation and apoptosis, and less neuropeptide Y were observed in the olfactory bulbs of FLX-exposed goldfish. Our findings indicate that FLX may hamper goldfish olfaction by interfering with the initiation, transmission, and processing of olfactory signals.
Collapse
Affiliation(s)
- Lin Huang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Weixia Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yu Han
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yu Tang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Weishang Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| |
Collapse
|
20
|
Hettiarachchi P, Johnson MA. Characterization of D3 Autoreceptor Function in Whole Zebrafish Brain with Fast-Scan Cyclic Voltammetry. ACS Chem Neurosci 2022; 13:2863-2873. [PMID: 36099546 PMCID: PMC10105970 DOI: 10.1021/acschemneuro.2c00280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Zebrafish (Danio rerio) are ideal model organisms for investigating nervous system function, both in health and disease. Nevertheless, functional characteristics of dopamine (DA) release and uptake regulation are still not well-understood in zebrafish. In this study, we assessed D3 autoreceptor function in the telencephalon of whole zebrafish brains ex vivo by measuring the electrically stimulated DA release ([DA]max) and uptake at carbon fiber microelectrodes with fast-scan cyclic voltammetry. Treatment with pramipexole and 7-OH-DPAT, selective D3 autoreceptor agonists, sharply decreased [DA]max. Conversely, SB277011A, a selective D3 antagonist, nearly doubled [DA]max and decreased k, the first-order rate constant for the DA uptake, to about 20% of its original value. Treatment with desipramine, a selective norepinephrine transporter blocker, failed to increase current, suggesting that our electrochemical signal arises solely from the release of DA. Furthermore, blockage of DA uptake with nomifensine-reversed 7-OH-DPAT induced decreases in [DA]max. Collectively, our data show that, as in mammals, D3 autoreceptors regulate DA release, likely by inhibiting uptake. The results of this study are useful in the further development of zebrafish as a model organism for DA-related neurological disorders such as Parkinson's disease, schizophrenia, and drug addiction.
Collapse
Affiliation(s)
- Piyanka Hettiarachchi
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Michael A Johnson
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
21
|
Torres DJ, Yorgason JT, Andres MA, Bellinger FP. Methamphetamine Exposure During Development Causes Lasting Changes to Mesolimbic Dopamine Signaling in Mice. Cell Mol Neurobiol 2022; 42:2433-2438. [PMID: 34138411 PMCID: PMC8678368 DOI: 10.1007/s10571-021-01120-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 06/11/2021] [Indexed: 10/21/2022]
Abstract
Methamphetamine (MA) abuse remains a public health issue. Prenatal MA exposure (PME) poses a significant health problem, as we know very little about the drug's long-term physiological impact on the developing human brain. We investigated the long-term consequences of early MA exposure using a mouse model that targets the brain growth spurt, which occurs during human third-trimester. Adult mice previously subjected to acute MA during post-natal days 4-9 exhibited hyperactivity during the Open-Field Test, while exhibiting no motor coordination changes during the Rotarod Test. Neonatal MA exposure reduced basal dopamine (DA) uptake rates in adult nucleus accumbens slices compared with saline-injected controls. Although slices from neonatal MA-exposed mice showed no change in evoked DA signals in the presence of MA, they exhibited potentiated non-evoked DA release through DA efflux in response to MA. These data suggest that developmental MA exposure alters brain development to produce long-lasting physiological changes to the adult mesolimbic DA system, as well as altering responses to acute MA exposure in adulthood. This study provides new insights into an important, under-investigated area in drugs of abuse research.
Collapse
Affiliation(s)
- Daniel J Torres
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA.
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| | - Jordan T Yorgason
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT, 84602, USA
| | - Marilou A Andres
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Frederick P Bellinger
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, 96813, USA
| |
Collapse
|
22
|
Lycas MD, Ejdrup AL, Sørensen AT, Haahr NO, Jørgensen SH, Guthrie DA, Støier JF, Werner C, Newman AH, Sauer M, Herborg F, Gether U. Nanoscopic dopamine transporter distribution and conformation are inversely regulated by excitatory drive and D2 autoreceptor activity. Cell Rep 2022; 40:111431. [PMID: 36170827 PMCID: PMC9617621 DOI: 10.1016/j.celrep.2022.111431] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/22/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
The nanoscopic organization and regulation of individual molecular components in presynaptic varicosities of neurons releasing modulatory volume neurotransmitters like dopamine (DA) remain largely elusive. Here we show, by application of several super-resolution microscopy techniques to cultured neurons and mouse striatal slices, that the DA transporter (DAT), a key protein in varicosities of dopaminergic neurons, exists in the membrane in dynamic equilibrium between an inward-facing nanodomain-localized and outward-facing unclustered configuration. The balance between these configurations is inversely regulated by excitatory drive and DA D2 autoreceptor activation in a manner dependent on Ca2+ influx via N-type voltage-gated Ca2+ channels. The DAT nanodomains contain tens of transporters molecules and overlap with nanodomains of PIP2 (phosphatidylinositol-4,5-bisphosphate) but show little overlap with D2 autoreceptor, syntaxin-1, and clathrin nanodomains. The data reveal a mechanism for rapid alterations of nanoscopic DAT distribution and show a striking link of this to the conformational state of the transporter.
Collapse
Affiliation(s)
- Matthew D Lycas
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Aske L Ejdrup
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Andreas T Sørensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Nicolai O Haahr
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Søren H Jørgensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Daryl A Guthrie
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jonatan F Støier
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Christian Werner
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-Universität Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Freja Herborg
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower 7.5, 2200 Copenhagen, Denmark.
| |
Collapse
|
23
|
Zhu F, Liu L, Li J, Liu B, Wang Q, Jiao R, Xu Y, Wang L, Sun S, Sun X, Younus M, Wang C, Hokfelt T, Zhang B, Gu H, Xu ZQD, Zhou Z. Cocaine increases quantal norepinephrine secretion through NET-dependent PKC activation in locus coeruleus neurons. Cell Rep 2022; 40:111199. [PMID: 35977516 DOI: 10.1016/j.celrep.2022.111199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/20/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022] Open
Abstract
The norepinephrine neurons in locus coeruleus (LC-NE neurons) are essential for sleep arousal, pain sensation, and cocaine addiction. According to previous studies, cocaine increases NE overflow (the profile of extracellular NE level in response to stimulation) by blocking the NE reuptake. NE overflow is determined by NE release via exocytosis and reuptake through NE transporter (NET). However, whether cocaine directly affects vesicular NE release has not been directly tested. By recording quantal NE release from LC-NE neurons, we report that cocaine directly increases the frequency of quantal NE release through regulation of NET and downstream protein kinase C (PKC) signaling, and this facilitation of NE release modulates the activity of LC-NE neurons and cocaine-induced stimulant behavior. Thus, these findings expand the repertoire of mechanisms underlying the effects of cocaine on NE (pro-release and anti-reuptake), demonstrate NET as a release enhancer in LC-NE neurons, and provide potential sites for treatment of cocaine addiction.
Collapse
Affiliation(s)
- Feipeng Zhu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lina Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China; Core Facilities Center, Departments of Neurobiology and Pathology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jie Li
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Bing Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Qinglong Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Ruiying Jiao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Yongxin Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lun Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Suhua Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Xiaoxuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Muhammad Younus
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Changhe Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Tomas Hokfelt
- Department of Neuroscience, Karolinska Institute, 171 71 Stockholm, Sweden
| | - Bo Zhang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China; Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Howard Gu
- Department of Biological Chemistry and Pharmacology, Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | - Zhi-Qing David Xu
- Core Facilities Center, Departments of Neurobiology and Pathology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
24
|
Su P, Zhai D, Wong AHC, Liu F. Development of a novel peptide to prevent entry of SARS-CoV-2 into lung and olfactory bulb cells of hACE2 expressing mice. Mol Brain 2022; 15:71. [PMID: 35945596 PMCID: PMC9361269 DOI: 10.1186/s13041-022-00956-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/23/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that has caused a global pandemic Coronavirus Disease 2019 (COVID-19). Currently, there are no effective treatments specifically for COVID-19 infection. The initial step in SARS-CoV-2 infection is attachment to the angiotensin-converting enzyme 2 (ACE2) on the cell surface. We have developed a protein peptide that effectively disrupts the binding between the SARS-CoV-2 spike protein and ACE2. When delivered by nasal spray, our peptide prevents SARS-CoV-2 spike protein from entering lung and olfactory bulb cells of mice expressing human ACE2. Our peptide represents a potential novel treatment and prophylaxis against COVID-19.
Collapse
Affiliation(s)
- Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Dongxu Zhai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Albert H C Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, M5T 1R8, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, M5T 1R8, Canada. .,Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
25
|
Formisano R, Rosikon KD, Singh A, Dhillon HS. The dopamine membrane transporter plays an active modulatory role in synaptic dopamine homeostasis. J Neurosci Res 2022; 100:1551-1559. [PMID: 34747520 PMCID: PMC9079189 DOI: 10.1002/jnr.24965] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/17/2021] [Accepted: 08/29/2021] [Indexed: 11/11/2022]
Abstract
Modulatory mechanisms of neurotransmitter release and clearance are highly controlled processes whose finely tuned regulation is critical for functioning of the nervous system. Dysregulation of the monoamine neurotransmitter dopamine can lead to several neuropathies. Synaptic modulation of dopamine is known to involve pre-synaptic D2 auto-receptors and acid sensing ion channels. In addition, the dopamine membrane transporter (DAT), which is responsible for clearance of dopamine from the synaptic cleft, is suspected to play an active role in modulating release of dopamine. Using functional imaging on the Caenorhabditis elegans model system, we show that DAT-1 acts as a negative feedback modulator to neurotransmitter vesicle fusion. Results from our fluorescence recovery after photo-bleaching (FRAP) based experiments were followed up with and reaffirmed using swimming-induced paralysis behavioral assays. Utilizing our numerical FRAP data we have developed a mechanistic model to dissect the dynamics of synaptic vesicle fusion, and compare the feedback effects of DAT-1 with the dopamine auto-receptor. Our experimental results and the mechanistic model are of potential broader significance, as similar dynamics are likely to be used by other synaptic modulators including membrane transporters for other neurotransmitters across species.
Collapse
Affiliation(s)
- Rosaria Formisano
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Katarzyna D. Rosikon
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, Biomedical Engineering, Data Sciences Institute, University of Delaware, Newark, DE, USA
| | - Harbinder S. Dhillon
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| |
Collapse
|
26
|
Festucci F, Annunzi E, Pepe M, Curcio G, D'Addario C, Adriani W. Dopamine-transporter heterozygous rats carrying maternal wild-type allele are more vulnerable to the development of compulsive behavior. Synapse 2022; 76:31-44. [PMID: 35772468 DOI: 10.1002/syn.22244] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 06/07/2022] [Accepted: 06/21/2022] [Indexed: 11/10/2022]
Abstract
Compulsivity is defined as an unstoppable tendency towards repetitive and habitual actions, which are reiterated despite negative consequences. Polydipsia is induced preclinically by intermittent reward leading rodents to ingest large amounts of fluids. We focused on the role of dopamine transporter (DAT) and inheritance factors in compulsive behavior. Our sample consisted of DAT heterozygous (HET) rats with different genetic inheritance (MAT-HET, born from WT-dams x KO-fathers; MIX-HET, born from HET-dams x KO-fathers). As controls, we used both wild-type (WT) rats and their socially-isolated (WTi) siblings. We ran the schedule-induced polydipsia (SIP) protocol, to induce compulsive behavior; then the Y-maze and marble-burying tests, to verify its actual development. Only MAT-HET (who inherited the functional DAT allele from the WT mother) is vulnerable to developing compulsive behavior. MAT-HET rats drank increasingly more water during SIP and showed significant perseverance in the Y-maze test and exhibited compulsive actions in the marble-burying test. Interestingly, compulsive behaviors of MAT-HET rats correlate with expression ex-vivo of different genes in different areas. Regarding the prefrontal cortex (PFC), D2R correlates with Y-maze "perseverance" in addition to BDNF; considering the amygdala (AMY), both D3R and OXTR correlate with SIP "licks". Indeed, compulsivity may be linked to D2R and BDNF in PFC, while extreme anxiety in MAT-HET rats may be associated with D3R and OXTR in the amygdala. These results confirm some similarities between MAT-HET and DAT-KO subjects and link the epigenetic context of the DAT gene to the development of compulsive behavior. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Fabiana Festucci
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Eugenia Annunzi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d' Annunzio" of Chieti-Pescara, Italy
| | - Martina Pepe
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Curcio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Walter Adriani
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
27
|
Dysfunctional Heteroreceptor Complexes as Novel Targets for the Treatment of Major Depressive and Anxiety Disorders. Cells 2022; 11:cells11111826. [PMID: 35681521 PMCID: PMC9180493 DOI: 10.3390/cells11111826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Among mental diseases, major depressive disorder (MDD) and anxiety deserve a special place due to their high prevalence and their negative impact both on society and patients suffering from these disorders. Consequently, the development of novel strategies designed to treat them quickly and efficiently, without or at least having limited side effects, is considered a highly important goal. Growing evidence indicates that emerging properties are developed on recognition, trafficking, and signaling of G-protein coupled receptors (GPCRs) upon their heteromerization with other types of GPCRs, receptor tyrosine kinases, and ionotropic receptors such as N-methyl-D-aspartate (NMDA) receptors. Therefore, to develop new treatments for MDD and anxiety, it will be important to identify the most vulnerable heteroreceptor complexes involved in MDD and anxiety. This review focuses on how GPCRs, especially serotonin, dopamine, galanin, and opioid heteroreceptor complexes, modulate synaptic and volume transmission in the limbic networks of the brain. We attempt to provide information showing how these emerging concepts can contribute to finding new ways to treat both MDD and anxiety disorders.
Collapse
|
28
|
Extrastriatal dopamine D2/3 receptor binding, functional connectivity, and autism socio-communicational deficits: a PET and fMRI study. Mol Psychiatry 2022; 27:2106-2113. [PMID: 35181754 DOI: 10.1038/s41380-022-01464-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 11/08/2022]
Abstract
The social motivation hypothesis of autism proposes that social communication symptoms in autism-spectrum disorder (ASD) stem from atypical social attention and reward networks, where dopamine acts as a crucial mediator. However, despite evidence indicating that individuals with ASD show atypical activation in extrastriatal regions while processing reward and social stimuli, no previous studies have measured extrastriatal dopamine D2/3 receptor (D2/3R) availability in ASD. Here, we investigated extrastriatal D2/3R availability in individuals with ASD and its association with ASD social communication symptoms using positron emission tomography (PET). Moreover, we employed a whole-brain multivariate pattern analysis of resting-state functional magnetic resonance imaging (fMRI) to identify regions where functional connectivity atypically correlates with D2/3R availability depending on ASD diagnosis. Twenty-two psychotropic-free males with ASD and 24 age- and intelligence quotient-matched typically developing males underwent [11C]FLB457 PET, fMRI, and clinical symptom assessment. Participants with ASD showed lower D2/3R availability throughout the D2/3R-rich extrastriatal regions of the dopaminergic pathways. Among these, the posterior region of the thalamus, which primarily comprises the pulvinar, displayed the largest effect size for the lower D2/3R availability, which correlated with a higher score on the Social Affect domain of the Autism Diagnostic Observation Schedule-2 in participants with ASD. Moreover, lower D2/3R availability was correlated with lower functional connectivity of the thalamus-superior temporal sulcus and cerebellum-medial occipital cortex, specifically in individuals with ASD. The current findings provide novel molecular evidence for the social motivation theory of autism and offer a novel therapeutic target.
Collapse
|
29
|
Wang J, Su P, Yang J, Xu L, Yuan A, Li C, Zhang T, Dong F, Zhou J, Samsom J, Wong AH, Liu F. The D2R-DISC1 protein complex and associated proteins are altered in schizophrenia and normalized with antipsychotic treatment. J Psychiatry Neurosci 2022; 47:E134-E147. [PMID: 35361701 PMCID: PMC8979657 DOI: 10.1503/jpn.210145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/30/2021] [Accepted: 01/24/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND For decades, the dopamine D2 receptor (D2R) has been known as the main target of antipsychotic medications, but the mechanism for antipsychotic effects beyond this pharmacological target remains unclear. Disrupted-in-schizophrenia 1 (DISC1) is a gene implicated in the etiology of schizophrenia, and we have found elevated levels of the D2R-DISC1 complex in the postmortem brain tissue of patients with schizophrenia. METHODS We used coimmunoprecipitation to measure D2R-DISC1 complex levels in peripheral blood samples from patients with schizophrenia and unaffected controls in 3 cohorts (including males and females) from different hospitals. We also used label-free mass spectrometry to conduct proteomic analysis of these samples. RESULTS Levels of the D2R-DISC1 complex were elevated in the peripheral blood samples of patients with schizophrenia from 3 independent cohorts, and were normalized with antipsychotic treatment. Proteomic analysis of the blood samples from patients with high D2R-DISC1 complex levels that were normalized with antipsychotic treatment revealed a number of altered proteins and pathways associated with D2R, DISC1 and the D2R-DISC1 complex. We identified additional proteins and pathways that were associated with antipsychotic treatment in schizophrenia, and that may also be novel targets for schizophrenia treatment. LIMITATIONS Sample sizes were relatively small, but were sufficient to detect associations between D2R-DISC1 levels, schizophrenia and treatment response. The relevance of leukocyte changes to the symptoms of schizophrenia is unknown. The coimmunoprecipitation lanes included several nonspecific bands. CONCLUSION Levels of the D2R-DISC1 complex were elevated in patients with schizophrenia and reduced with antipsychotic treatment. This finding reinforces the independent role of each protein in schizophrenia. Our results enhanced our understanding of the molecular pathways involved in schizophrenia and in antipsychotic medications, and identified novel potential molecular targets for treating schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fang Liu
- From the Shanghai Mental Health Centre, Shanghai Jiaotong University, School of Medicine, Shanghai, China (Wang, Xu, Yuan, Li, Zhang, Liu); the Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ont. (Su, Samsom, Wong, Liu); the National Clinical Research Centre for Mental Disorders, Beijing AnDing Hospital, Capital Medical University, Beijing, China (Yang, Dong, Zhou); the Department of Pharmacology, University of Toronto, Toronto, Ont. (Samsom, Wong); the Institute of Medical Science, University of Toronto, Toronto, Ont. (Wong, Liu); the Department of Psychiatry, University of Toronto, Toronto, Ont. (Wong, Liu); the Department of Physiology, University of Toronto, Toronto, Ont. (Liu)
| |
Collapse
|
30
|
A New Method for the Visualization of Living Dopaminergic Neurons and Prospects for Using It to Develop Targeted Drug Delivery to These Cells. Int J Mol Sci 2022; 23:ijms23073678. [PMID: 35409040 PMCID: PMC8998426 DOI: 10.3390/ijms23073678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
This is the first study aiming to develop a method for the long-term visualization of living nigrostriatal dopaminergic neurons using 1-(2-(bis(4-fluorophenyl)methoxy)ethyl)-4-(3-phenylpropyl)piperazine-BODIPY (GBR-BP), the original fluorescent substance, which is a derivative of GBR-12909, a dopamine uptake inhibitor. This method is based on the authors’ hypothesis about the possibility of specifically internalizing into dopaminergic neurons substances with a high affinity for the dopamine transporter (DAT). Using a culture of mouse embryonic mesencephalic and LUHMES cells (human embryonic mesencephalic cells), as well as slices of the substantia nigra of adult mice, we have obtained evidence that GBR-BP is internalized specifically into dopaminergic neurons in association with DAT via a clathrin-dependent mechanism. Moreover, GBR-BP has been proven to be nontoxic. As we have shown in a primary culture of mouse metencephalon, GBR-BP is also specifically internalized into some noradrenergic and serotonergic neurons, but is not delivered to nonmonoaminergic neurons. Our data hold great promise for visualization of dopaminergic neurons in a mixed cell population to study their functioning, and can also be considered a new approach for the development of targeted drug delivery to dopaminergic neurons in pathology, including Parkinson’s disease.
Collapse
|
31
|
Palumbo S, Mariotti V, Vellucci S, Antonelli K, Anderson N, Harenski C, Pietrini P, Kiehl KA, Pellegrini S. ANKK1 and TH gene variants in combination with paternal maltreatment increase susceptibility to both cognitive and attentive impulsivity. Front Psychiatry 2022; 13:868804. [PMID: 35935430 PMCID: PMC9352854 DOI: 10.3389/fpsyt.2022.868804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Recent scientific findings suggest that dopamine exerts a central role on impulsivity, as well as that aversive life experiences may promote the high levels of impulsivity that often underlie violent behavior. To deepen our understanding of the complex gene by environment interplay on impulsive behavior, we genotyped six dopaminergic allelic variants (ANKK1-rs1800497, TH-rs6356, DRD4-rs1800955, DRD4-exonIII-VNTR, SLC6A3-VNTR and COMT-rs4680) in 655 US White male inmates convicted for violent crimes, whose impulsivity was assessed by BIS-11 (Barratt Impulsiveness Scale). Furthermore, in a subsample of 216 inmates from the whole group, we also explored the potential interplay between the genotyped dopaminergic variants and parental maltreatment measured by MOPS (Measure of Parental Style) in promoting impulsivity. We found a significant interaction among paternal MOPS scores, ANKK1-rs1800497-T allele and TH-rs6356-A allele, which increased the variance of BIS-11 cognitive/attentive scores explained by paternal maltreatment from 1.8 up to 20.5%. No direct association between any of the individual genetic variants and impulsivity was observed. Our data suggest that paternal maltreatment increases the risk of attentive/cognitive impulsivity and that this risk is higher in carriers of specific dopaminergic alleles that potentiate the dopaminergic neurotransmission. These findings add further evidence to the mutual role that genetics and early environmental factors exert in modulating human behavior and highlight the importance of childhood care interventions.
Collapse
Affiliation(s)
- Sara Palumbo
- Department of Surgical, Medical and Molecular Pathology and Critical Care, University of Pisa, Pisa, Italy
| | - Veronica Mariotti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Vellucci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Klizia Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nathaniel Anderson
- The Mind Research Network and Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
| | - Carla Harenski
- The Mind Research Network and Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
| | - Pietro Pietrini
- Molecular Mind Lab, IMT School for Advanced Studies Lucca, Lucca, Italy
| | - Kent A Kiehl
- The Mind Research Network and Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States.,Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | - Silvia Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
32
|
Piniella D, Martínez-Blanco E, Bartolomé-Martín D, Sanz-Martos AB, Zafra F. Identification by proximity labeling of novel lipidic and proteinaceous potential partners of the dopamine transporter. Cell Mol Life Sci 2021; 78:7733-7756. [PMID: 34709416 PMCID: PMC8629785 DOI: 10.1007/s00018-021-03998-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/07/2021] [Accepted: 10/17/2021] [Indexed: 12/05/2022]
Abstract
Dopamine (DA) transporters (DATs) are regulated by trafficking and modulatory processes that probably rely on stable and transient interactions with neighboring proteins and lipids. Using proximity-dependent biotin identification (BioID), we found novel potential partners for DAT, including several membrane proteins, such as the transmembrane chaperone 4F2hc, the proteolipid M6a and a potential membrane receptor for progesterone (PGRMC2). We also detected two cytoplasmic proteins: a component of the Cullin1-dependent ubiquitination machinery termed F-box/LRR-repeat protein 2 (FBXL2), and the enzyme inositol 5-phosphatase 2 (SHIP2). Immunoprecipitation (IP) and immunofluorescence studies confirmed either a physical association or a close spatial proximity between these proteins and DAT. M6a, SHIP2 and the Cullin1 system were shown to increase DAT activity in coexpression experiments, suggesting a functional role for their association. Deeper analysis revealed that M6a, which is enriched in neuronal protrusions (filopodia or dendritic spines), colocalized with DAT in these structures. In addition, the product of SHIP2 enzymatic activity (phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2]) was tightly associated with DAT, as shown by co-IP and by colocalization of mCherry-DAT with a specific biosensor for this phospholipid. PI(3,4)P2 strongly stimulated transport activity in electrophysiological recordings, and conversely, inhibition of SHIP2 reduced DA uptake in several experimental systems including striatal synaptosomes and the dopaminergic cell line SH-SY5Y. In summary, here we report several potential new partners for DAT and a novel regulatory lipid, which may represent new pharmacological targets for DAT, a pivotal protein in dopaminergic function of the brain.
Collapse
Affiliation(s)
- Dolores Piniella
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Martínez-Blanco
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
| | - David Bartolomé-Martín
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Universidad de La Laguna, Tenerife, Spain
| | - Ana B Sanz-Martos
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, 28925, Madrid, Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa and Departamento de Biología Molecular, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, C / Nicolás Cabrera 1, 28049, Madrid, Spain.
- IdiPAZ, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
33
|
Covey DP, Yocky AG. Endocannabinoid Modulation of Nucleus Accumbens Microcircuitry and Terminal Dopamine Release. Front Synaptic Neurosci 2021; 13:734975. [PMID: 34497503 PMCID: PMC8419321 DOI: 10.3389/fnsyn.2021.734975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/05/2021] [Indexed: 12/20/2022] Open
Abstract
The nucleus accumbens (NAc) is located in the ventromedial portion of the striatum and is vital to valence-based predictions and motivated action. The neural architecture of the NAc allows for complex interactions between various cell types that filter incoming and outgoing information. Dopamine (DA) input serves a crucial role in modulating NAc function, but the mechanisms that control terminal DA release and its effect on NAc neurons continues to be elucidated. The endocannabinoid (eCB) system has emerged as an important filter of neural circuitry within the NAc that locally shapes terminal DA release through various cell type- and site-specific actions. Here, we will discuss how eCB signaling modulates terminal DA release by shaping the activity patterns of NAc neurons and their afferent inputs. We then discuss recent technological advancements that are capable of dissecting how distinct cell types, their afferent projections, and local neuromodulators influence valence-based actions.
Collapse
Affiliation(s)
- Dan P Covey
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Alyssa G Yocky
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| |
Collapse
|
34
|
Nawaratne V, McLaughlin SP, Mayer FP, Gichi Z, Mastriano A, Carvelli L. Prolonged Amphetamine Exposures Increase the Endogenous Human Dopamine Receptors 2 at the Cellular Membrane in Cells Lacking the Dopamine Transporter. Front Cell Neurosci 2021; 15:681539. [PMID: 34512264 PMCID: PMC8427050 DOI: 10.3389/fncel.2021.681539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/03/2021] [Indexed: 11/30/2022] Open
Abstract
The dopamine 2 receptors (D2R) are G-protein coupled receptors expressed both in pre- and post-synaptic terminals that play an important role in mediating the physiological and behavioral effects of amphetamine (Amph). Previous studies have indicated that the effects of Amph at the D2R mainly rely on the ability of Amph to robustly increase extracellular dopamine through the dopamine transporter (DAT). This implies that the effects of Amph on D2R require the neurotransmitter dopamine. However, because of its lipophilic nature, Amph can cross the cellular membrane and thus potentially affect D2R expression independently of dopamine and DAT, e.g., in post-synaptic terminals. Here we used an in vitro system to study whether Amph affects total expression, cellular distribution, and function of the human D2R (hD2R), endogenously expressed in HEK293 cells. By performing Western blot experiments, we found that prolonged treatments with 1 or 50 μM Amph cause a significant decrease of the endogenous hD2R in cells transfected with human DAT (hDAT). On the other hand, in cells lacking expression of DAT, quantification of the hD2R-mediated changes in cAMP, biotinylation assays, Western blots and imaging experiments demonstrated an increase of hD2R at the cellular membrane after 15-h treatments with Amph. Moreover, imaging data suggested that barbadin, a specific inhibitor of the βarrestin-βadaptin interaction, blocked the Amph-induced increase of hD2R. Taken together our data suggest that prolonged exposures to Amph decrease or increase the endogenous hD2R at the cellular membrane in HEK293 cells expressing or lacking hDAT, respectively. Considering that this drug is often consumed for prolonged periods, during which tolerance develops, our data suggest that even in absence of DAT or dopamine, Amph can still alter D2R distribution and function.
Collapse
Affiliation(s)
- Vindhya Nawaratne
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, United States
| | - Sean P. McLaughlin
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Felix P. Mayer
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Zayna Gichi
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Alyssa Mastriano
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, United States
| | - Lucia Carvelli
- Department of Biology, Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, United States
- Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| |
Collapse
|
35
|
Kovtun O, Torres R, Bellocchio LG, Rosenthal SJ. Membrane Nanoscopic Organization of D2L Dopamine Receptor Probed by Quantum Dot Tracking. MEMBRANES 2021; 11:578. [PMID: 34436341 PMCID: PMC8401772 DOI: 10.3390/membranes11080578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 01/11/2023]
Abstract
The role of lateral mobility and nanodomain organization of G protein-coupled receptors in modulating subcellular signaling has been under increasing scrutiny. Investigation of D2 dopamine receptor diffusion dynamics is of particular interest, as these receptors have been linked to altered neurotransmission in affective disorders and represent the primary target for commonly prescribed antipsychotics. Here, we applied our single quantum dot tracking approach to decipher intrinsic diffusion patterns of the wild-type long isoform of the D2 dopamine receptor and its genetic variants previously identified in several cohorts of schizophrenia patients. We identified a subtle decrease in the diffusion rate of the Val96Ala mutant that parallels its previously reported reduced affinity for potent neuroleptics clozapine and chlorpromazine. Slower Val96Ala variant diffusion was not accompanied by a change in receptor-receptor transient interactions as defined by the diffraction-limited quantum dot colocalization events. In addition, we implemented a Voronoї tessellation-based algorithm to compare nanoclustering of the D2 dopamine receptor to the dominant anionic phospholipid phosphatidylinositol 4,5-bisphosphate in the plasma membrane of live cells.
Collapse
Affiliation(s)
- Oleg Kovtun
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (R.T.); (L.G.B.)
| | - Ruben Torres
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (R.T.); (L.G.B.)
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Laurel G. Bellocchio
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (R.T.); (L.G.B.)
| | - Sandra Jean Rosenthal
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; (R.T.); (L.G.B.)
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
36
|
Knockout or Knock-in? A Truncated D2 Receptor Protein Is Expressed in the Brain of Functional D2 Receptor Knockout Mice. NEUROSCI 2021. [DOI: 10.3390/neurosci2020014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Null mice for the dopamine D2 receptor (D2R) have been instrumental in understanding the function of this protein. For our research, we obtained the functional D2R knockout mouse strain described initially in 1997. Surprisingly, our biochemical characterization showed that this mouse strain is not a true knockout. We determined by sequence analysis of the rapid 3′ amplification of cDNA ends that functional D2R knockout mice express transcripts that lack only the eighth exon. Furthermore, immunofluorescence assays showed a D2R-like protein in the brain of functional D2R knockout mice. We verified by immunofluorescence that the recombinant truncated D2R is expressed in HEK293T cells, showing intracellular localization, colocalizing in the Golgi apparatus and the endoplasmic reticulum, but with less presence in the Golgi apparatus compared to the native D2R. As previously reported, functional D2R knockout mice are hypoactive and insensitive to the D2R agonist quinpirole. Concordantly, microdialysis studies confirmed that functional D2R knockout mice have lower extracellular dopamine levels in the striatum than the native mice. In conclusion, functional D2R knockout mice express transcripts that lead to a truncated D2R protein lacking from the sixth transmembrane domain to the C-terminus. We share these findings to avoid future confusion and the community considers this mouse strain in D2R traffic and protein–protein interaction studies.
Collapse
|
37
|
Kurose S, Kubota M, Takahata K, Yamamoto Y, Fujiwara H, Kimura Y, Ito H, Takeuchi H, Mimura M, Suhara T, Higuchi M. Relationship between regional gray matter volumes and dopamine D 2 receptor and transporter in living human brains. Hum Brain Mapp 2021; 42:4048-4058. [PMID: 34014611 PMCID: PMC8288088 DOI: 10.1002/hbm.25538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/24/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Although striatal dopamine neurotransmission is believed to be functionally linked to the formation of the corticostriatal network, there has been little evidence for this regulatory process in the human brain and its disruptions in neuropsychiatric disorders. Here, we aimed to investigate associations of striatal dopamine transporter (DAT) and D2 receptor availabilities with gray matter (GM) volumes in healthy humans. Positron emission tomography images of D2 receptor (n = 34) and DAT (n = 17) captured with the specific radioligands [11C]raclopride and [18F]FE‐PE2I, respectively, were acquired along with T1‐weighted magnetic resonance imaging data in our previous studies, and were re‐analyzed in this work. We quantified the binding potentials (BPND) of these radioligands in the limbic, executive, and sensorimotor functional subregions of the striatum. Correlations between the radioligand BPND and regional GM volume were then examined by voxel‐based morphometry. In line with the functional and anatomical connectivity, [11C]raclopride BPND in the limbic striatum was positively correlated with volumes of the uncal/parahippocampal gyrus and adjacent temporal areas. Similarly, we found positive correlations between the BPND of this radioligand in the executive striatum and volumes of the prefrontal cortices and their adjacent areas as well as between the BPND in the sensorimotor striatum and volumes of the somatosensory and supplementary motor areas. By contrast, no significant correlation was found between [18F]FE‐PE2I BPND and regional GM volumes. Our results suggest unique structural and functional corticostriatal associations involving D2 receptor in healthy humans, which might be partially independent of the nigrostriatal pathway reflected by striatal DAT.
Collapse
Affiliation(s)
- Shin Kurose
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Kubota
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hironobu Fujiwara
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hiroshi Ito
- Department of Radiology and Nuclear Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hiroyoshi Takeuchi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
38
|
Ryan RM, Ingram SL, Scimemi A. Regulation of Glutamate, GABA and Dopamine Transporter Uptake, Surface Mobility and Expression. Front Cell Neurosci 2021; 15:670346. [PMID: 33927596 PMCID: PMC8076567 DOI: 10.3389/fncel.2021.670346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 01/31/2023] Open
Abstract
Neurotransmitter transporters limit spillover between synapses and maintain the extracellular neurotransmitter concentration at low yet physiologically meaningful levels. They also exert a key role in providing precursors for neurotransmitter biosynthesis. In many cases, neurons and astrocytes contain a large intracellular pool of transporters that can be redistributed and stabilized in the plasma membrane following activation of different signaling pathways. This means that the uptake capacity of the brain neuropil for different neurotransmitters can be dynamically regulated over the course of minutes, as an indirect consequence of changes in neuronal activity, blood flow, cell-to-cell interactions, etc. Here we discuss recent advances in the mechanisms that control the cell membrane trafficking and biophysical properties of transporters for the excitatory, inhibitory and modulatory neurotransmitters glutamate, GABA, and dopamine.
Collapse
Affiliation(s)
- Renae M. Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
39
|
Yang J, Villar VAM, Jose PA, Zeng C. Renal Dopamine Receptors and Oxidative Stress: Role in Hypertension. Antioxid Redox Signal 2021; 34:716-735. [PMID: 32349533 PMCID: PMC7910420 DOI: 10.1089/ars.2020.8106] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: The kidney plays an important role in the long-term control of blood pressure. Oxidative stress is one of the fundamental mechanisms responsible for the development of hypertension. Dopamine, via five subtypes of receptors, plays an important role in the control of blood pressure by various mechanisms, including the inhibition of oxidative stress. Recent Advances: Dopamine receptors exert their regulatory function to decrease the oxidative stress in the kidney and ultimately maintain normal sodium balance and blood pressure homeostasis. An aberration of this regulation may be involved in the pathogenesis of hypertension. Critical Issues: Our present article reviews the important role of oxidative stress and intrarenal dopaminergic system in the regulation of blood pressure, summarizes the current knowledge on renal dopamine receptor-mediated antioxidation, including decreasing reactive oxygen species production, inhibiting pro-oxidant enzyme nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase, and stimulating antioxidative enzymes, and also discusses its underlying mechanisms, including the increased activity of G protein-coupled receptor kinase 4 (GRK4) and abnormal trafficking of renal dopamine receptors in hypertensive status. Future Directions: Identifying the mechanisms of renal dopamine receptors in the regulation of oxidative stress and their contribution to the pathogenesis of hypertension remains an important research focus. Increased understanding of the role of reciprocal regulation between renal dopamine receptors and oxidative stress in the regulation of blood pressure may give us novel insights into the pathogenesis of hypertension and provide a new treatment strategy for hypertension.
Collapse
Affiliation(s)
- Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Van Anthony M Villar
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Chunyu Zeng
- Department of Cardiology, Fujian Heart Medical Center, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
40
|
Pandey P, Singh A, Kaur H, Ghosh-Roy A, Babu K. Increased dopaminergic neurotransmission results in ethanol dependent sedative behaviors in Caenorhabditis elegans. PLoS Genet 2021; 17:e1009346. [PMID: 33524034 PMCID: PMC7877767 DOI: 10.1371/journal.pgen.1009346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/11/2021] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Ethanol is a widely used drug, excessive consumption of which could lead to medical conditions with diverse symptoms. Ethanol abuse causes dysfunction of memory, attention, speech and locomotion across species. Dopamine signaling plays an essential role in ethanol dependent behaviors in animals ranging from C. elegans to humans. We devised an ethanol dependent assay in which mutants in the dopamine autoreceptor, dop-2, displayed a unique sedative locomotory behavior causing the animals to move in circles while dragging the posterior half of their body. Here, we identify the posterior dopaminergic sensory neuron as being essential to modulate this behavior. We further demonstrate that in dop-2 mutants, ethanol exposure increases dopamine secretion and functions in a DVA interneuron dependent manner. DVA releases the neuropeptide NLP-12 that is known to function through cholinergic motor neurons and affect movement. Thus, DOP-2 modulates dopamine levels at the synapse and regulates alcohol induced movement through NLP-12.
Collapse
Affiliation(s)
- Pratima Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Anuradha Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
| | - Harjot Kaur
- National Brain Research Centre, Gurgaon, India
| | | | - Kavita Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Mohali, India
- Centre for Neuroscience, Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
41
|
Luis-Ravelo D, Fumagallo-Reading F, Castro-Hernandez J, Barroso-Chinea P, Afonso-Oramas D, Febles-Casquero A, Cruz-Muros I, Salas-Hernandez J, Mesa-Infante V, Rodriguez-Nuñez J, Gonzalez-Hernandez T. Prolonged dopamine D 3 receptor stimulation promotes dopamine transporter ubiquitination and degradation through a PKC-dependent mechanism. Pharmacol Res 2021; 165:105434. [PMID: 33484816 DOI: 10.1016/j.phrs.2021.105434] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/17/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
The dopamine transporter (DAT) is a membrane glycoprotein in dopaminergic neurons, which modulates extracellular and intracellular dopamine levels. DAT is regulated by different presynaptic proteins, including dopamine D2 (D2R) and D3 (D3R) receptors. While D2R signalling enhances DAT activity, some data suggest that D3R has a biphasic effect. However, despite the extensive therapeutic use of D2R/D3R agonists in neuropsychiatric disorders, this phenomenon has been little studied. In order to shed light on this issue, DAT activity, expression and posttranslational modifications were studied in mice and DAT-D3R-transfected HEK cells. Consistent with previous reports, acute treatment with D2R/D3R agonists promoted DAT recruitment to the plasma membrane and an increase in DA uptake. However, when the treatment was prolonged, DA uptake and total striatal DAT protein declined below basal levels. These effects were inhibited in mice by genetic and pharmacological inactivation of D3R, but not D2R, indicating that they are D3R-dependent. No changes were detected in mesostriatal tyrosine hydroxylase (TH) protein expression and midbrain TH and DAT mRNAs, suggesting that the dopaminergic system is intact and DAT is posttranslationally regulated. The use of immunoprecipitation and cell surface biotinylation revealed that DAT is phosphorylated at serine residues, ubiquitinated and released into late endosomes through a PKCβ-dependent mechanism. In sum, the results indicate that long-term D3R activation promotes DAT down-regulation, an effect that may underlie neuroprotective and antidepressant actions described for some D2R/D3R agonists.
Collapse
Affiliation(s)
- Diego Luis-Ravelo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Felipe Fumagallo-Reading
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Javier Castro-Hernandez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Pedro Barroso-Chinea
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Domingo Afonso-Oramas
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Alejandro Febles-Casquero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Ignacio Cruz-Muros
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Josmar Salas-Hernandez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Virginia Mesa-Infante
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Julia Rodriguez-Nuñez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Tomas Gonzalez-Hernandez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
42
|
De Micco R, Siciliano M, Sant'Elia V, Giordano A, Russo A, Tedeschi G, Tessitore A. Correlates of Psychological Distress in Patients with Parkinson's Disease During the COVID-19 Outbreak. Mov Disord Clin Pract 2021; 8:60-68. [PMID: 33426160 PMCID: PMC7780948 DOI: 10.1002/mdc3.13108] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Following the severe consequences of the COVID-19 outbreak, on March 9, 2020, the Italian government implemented extraordinary measures to limit viral transmission, including restrictive quarantine measures. This resulted in a rapid and profound change of people's daily lives. OBJECTIVE We assessed the psychological impact of the 40-day quarantine in a large cohort of patients with Parkinson's disease (PD) and caregivers. Moreover, we analyzed whether prelockdown clinical features may be associated with subjective response of patients with PD to this traumatic event. METHODS A total of 94 patients with PD were enrolled in the study. The Impact of Event Scale-Revised, the Kessler Psychological Distress Scale, and the 12-item Zarit Burden Inventory were obtained from patients and caregivers by email. A multivariate regression analysis was performed to determine whether prelockdown clinical motor and nonmotor features were associated with the psychological impact of lockdown. RESULTS Regression analyses showed that prelockdown levels of anxiety, treatment-related motor complications, patients' quality of life, and lockdown hours per day were significantly associated with psychological impact measures of the 40-day quarantine. In addition, we showed that caregiver burden was correlated with overall patient autonomy and attention/memory impairment. CONCLUSIONS We identified specific PD motor and nonmotor features potentially predisposing to higher psychological impact of stressful situations, such as quarantine. This may help guide postpandemic interventions and preventive strategies to avoid further impairment of psychological well-being in patients with PD.
Collapse
Affiliation(s)
- Rosa De Micco
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Mattia Siciliano
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
- Department of PsychologyUniversity of Campania “Luigi Vanvitelli”CasertaItaly
| | - Valeria Sant'Elia
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
- Department of PsychologyUniversity of Campania “Luigi Vanvitelli”CasertaItaly
| | - Alfonso Giordano
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Antonio Russo
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Gioacchino Tedeschi
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Alessandro Tessitore
- Department of Advanced Medical and Surgical SciencesUniversity of Campania “Luigi Vanvitelli”NaplesItaly
| |
Collapse
|
43
|
Morales-Mulia S, Magdaleno-Madrigal VM, Nicolini H, Genis-Mendoza A, Morales-Mulia M. Orexin-A up-regulates dopamine D2 receptor and mRNA in the nucleus accumbens Shell. Mol Biol Rep 2020; 47:9689-9697. [PMID: 33170427 DOI: 10.1007/s11033-020-05979-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/03/2020] [Indexed: 01/11/2023]
Abstract
Orexins-A (OrxA) and -B (OrxB) neuropeptides are synthesized by a group of neurons located in the lateral hypothalamus and adjacent perifornical area, which send their projections to the mesolimbic dopaminergic (DAergic) system including ventral tegmental area and nucleus accumbens (NAc), where orexin receptors are expressed. NAc plays a central role in reward-seeking behavior and drug abuse. NAc-neurons express dopamine-1 (D1R) and dopamine-2 (D2R) receptors. Orexins bind to their two cognate G-protein-coupled receptors, orexin-receptor type-1 (Orx1R) and type-2 (Orx2R). Orexin receptor signaling is involved in behaviors such as motivation and addiction. Orexin-containing neurons modulate DAergic activity that is key in synaptic plasticity induced by addictive drugs. However, the effect of OrxA on expression and content of DAergic receptors in NAc is unknown. The purpose of this study was to investigate whether OrxA can alter gene expression and protein levels of D1R/D2R in NAc. Gene expression was evaluated by real-time PCR analysis and protein levels by western blot in rats. The results show that intracerebroventricular (i.c.v.) injection of OrxA increases both gene transcription and protein content of D2R but fails to modify D1R. This effect was also confirmed with OrxA infusion in NAc/Shell. Our results demonstrate for the first time that OrxA induces up-regulation of gene and protein of D2R in NAc. These findings support the hypothesis that OrxA modulates the DAergic transmission and this may serve to understand how orexin signaling enhances DA responses at baseline conditions and in response to psychostimulants.
Collapse
Affiliation(s)
- Sandra Morales-Mulia
- Departamento de Biología Celular, Facultad de Ciencias, UNAM, Mexico, DF, Mexico
| | | | - Humberto Nicolini
- Laboratory of Genomics of Psychiatric Diseases, Neurodegenerative and Addictions, National Institute of Genomic Medicine, Mexico, Mexico
| | - Alma Genis-Mendoza
- Laboratory of Genomics of Psychiatric Diseases, Neurodegenerative and Addictions, National Institute of Genomic Medicine, Mexico, Mexico.,Hospital Psiquiátrico Infantil "Dr. Juan N. Navarro" Psychiatric Attention Services, Mexico, Mexico
| | - Marcela Morales-Mulia
- Bases Moleculares de las Adicciones, Subdirección de Investigaciones Clínicas, INPRFM, Calzada México-Xochimilco 101, San Lorenzo Huipulco, Tlalpan, 14370, Mexico, DF, Mexico.
| |
Collapse
|
44
|
Dopamine transporter is downregulated and its association with chaperone protein Hsc70 is enhanced by activation of dopamine D 3 receptor. Brain Res Bull 2020; 165:263-271. [PMID: 33049353 DOI: 10.1016/j.brainresbull.2020.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 01/11/2023]
Abstract
Synaptic dopamine (DA) concentrations are largely determined by the activities of presynaptic D2 and D3 autoreceptors (D2R and D3R) and DA transporter (DAT). Furthermore, the activity of DAT is regulated by phosphorylation events and protein interactions that affect its surface expression. Because DA autoreceptors and DAT coordinately maintain synaptic DA homeostasis, we hypothesized that D3R might crosstalk with DAT to fine-tune synaptic DA concentrations. To test this hypothesis, we established [3H]DA uptake and DAT surface expression assays in hD3/rDAT-double-transfected HEK-293 cells or limbic forebrain synaptosomal preparations. Ropinirole, a preferential D3R agonist, reduced [3H]DA uptake in HEK-hD3/rDAT cells in a dose-dependent manner, an effect which could be blocked by the D2R/D3R antagonist, raclopride. Furthermore, ropinirole also reduced DAT surface expression in limbic forebrain synaptosomes, and this effect could be blocked by raclopride or the internalization inhibitor, concanavalin A. To identify potential mediators of this apparent D3R-DAT crosstalk, DAT-associated proteins were co-immunoprecipitated from limbic forebrain synaptosomes after D3R activation and identified by MALDI-TOF. From this analysis, the Hsc70 chaperone was identified as a DAT-associated protein. Interestingly, ropinirole induced the association of Hsc70/Hsp70 with DAT, and the Hsc70/Hsp70 inhibitor, apoptozole, prevented the ropinirole-induced reduction of DAT surface expression. Together, these results suggest that D3R negatively regulates DAT activity by promoting the association of DAT and Hsc70/Hsp70.
Collapse
|
45
|
Blagotinšek Cokan K, Mavri M, Rutland CS, Glišić S, Senćanski M, Vrecl M, Kubale V. Critical Impact of Different Conserved Endoplasmic Retention Motifs and Dopamine Receptor Interacting Proteins (DRIPs) on Intracellular Localization and Trafficking of the D 2 Dopamine Receptor (D 2-R) Isoforms. Biomolecules 2020; 10:biom10101355. [PMID: 32977535 PMCID: PMC7598153 DOI: 10.3390/biom10101355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 01/13/2023] Open
Abstract
The type 2 dopamine receptor D2 (D2-R), member of the G protein-coupled receptor (GPCR) superfamily, exists in two isoforms, short (D2S-R) and long (D2L-R). They differ by an additional 29 amino acids (AA) in the third cytoplasmic loop (ICL3) of the D2L-R. These isoforms differ in their intracellular localization and trafficking functionality, as D2L-R possesses a larger intracellular pool, mostly in the endoplasmic reticulum (ER). This review focuses on the evolutionarily conserved motifs in the ICL3 of the D2-R and proteins interacting with the ICL3 of both isoforms, specifically with the 29 AA insert. These motifs might be involved in D2-R exit from the ER and have an impact on cell-surface and intracellular localization and, therefore, also play a role in the function of dopamine receptor signaling, ligand binding and possible homo/heterodimerization. Our recent bioinformatic data on potential new interaction partners for the ICL3 of D2-Rs are also presented. Both are highly relevant, and have clinical impacts on the pathophysiology of several diseases such as Parkinson’s disease, schizophrenia, Tourette’s syndrome, Huntington’s disease, manic depression, and others, as they are connected to a variety of essential motifs and differences in communication with interaction partners.
Collapse
Affiliation(s)
- Kaja Blagotinšek Cokan
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
| | - Maša Mavri
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
| | - Catrin Sian Rutland
- School of Veterinary Medicine and Science, Medical Faculty, University of Nottingham, Sutton, Bonington Campus, Loughborough LE12 5RD, UK;
| | - Sanja Glišić
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Vinča, Belgrade, Serbia; (S.G.); (M.S.)
| | - Milan Senćanski
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Vinča, Belgrade, Serbia; (S.G.); (M.S.)
| | - Milka Vrecl
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
| | - Valentina Kubale
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
- Correspondence:
| |
Collapse
|
46
|
Li H, Su P, Lai TK, Jiang A, Liu J, Zhai D, Campbell CT, Lee FH, Yong W, Pasricha S, Li S, Wong AH, Ressler KJ, Liu F. The glucocorticoid receptor-FKBP51 complex contributes to fear conditioning and posttraumatic stress disorder. J Clin Invest 2020; 130:877-889. [PMID: 31929189 DOI: 10.1172/jci130363] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/30/2019] [Indexed: 02/01/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) can develop after exposure to severe psychological trauma, leaving patients with disabling anxiety, nightmares, and flashbacks. Current treatments are only partially effective, and development of better treatments is hampered by limited knowledge of molecular mechanisms underlying PTSD. We have discovered that the glucocorticoid receptor (GR) and FK506 binding protein 51 (FKBP51) form a protein complex that is elevated in PTSD patients compared with unaffected control subjects, subjects exposed to trauma without PTSD, and patients with major depressive disorder (MDD). The GR-FKBP51 complex is also elevated in fear-conditioned mice, an aversive learning paradigm that models some aspects of PTSD. Both PTSD patients and fear-conditioned mice had decreased GR phosphorylation, decreased nuclear GR, and lower expression of 14-3-3ε, a gene regulated by GR. We created a peptide that disrupts GR-FKBP51 binding and reverses behavioral and molecular changes induced by fear conditioning. This peptide reduces freezing time and increases GR phosphorylation, GR-FKBP52 binding, GR nuclear translocation, and 14-3-3ε expression in fear-conditioned mice. These experiments demonstrate a molecular mechanism contributing to PTSD and suggest that the GR-FKBP51 complex may be a diagnostic biomarker and a potential therapeutic target for preventing or treating PTSD.
Collapse
Affiliation(s)
- Haiyin Li
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Terence Ky Lai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Anlong Jiang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jing Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Dongxu Zhai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Charlie Tg Campbell
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Frankie Hf Lee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - WeiDong Yong
- Comparative Medical Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Suvercha Pasricha
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry and
| | - Shupeng Li
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry and
| | - Albert Hc Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry and.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Kerry J Ressler
- McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry and.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Chen R, Ferris MJ, Wang S. Dopamine D2 autoreceptor interactome: Targeting the receptor complex as a strategy for treatment of substance use disorder. Pharmacol Ther 2020; 213:107583. [PMID: 32473160 PMCID: PMC7434700 DOI: 10.1016/j.pharmthera.2020.107583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Dopamine D2 autoreceptors (D2ARs), located in somatodendritic and axon terminal compartments of dopamine (DA) neurons, function to provide a negative feedback regulatory control on DA neuron firing, DA synthesis, reuptake and release. Dysregulation of D2AR-mediated DA signaling is implicated in vulnerability to substance use disorder (SUD). Due to the extreme low abundance of D2ARs compared to postsynaptic D2 receptors (D2PRs) and the lack of experimental tools to differentiate the signaling of D2ARs from D2PRs, the regulation of D2ARs by drugs of abuse is poorly understood. The recent availability of conditional D2AR knockout mice and newly developed virus-mediated gene delivery approaches have provided means to specifically study the function of D2ARs at the molecular, cellular and behavioral levels. There is a growing revelation of novel mechanisms and new proteins that mediate D2AR activity, suggesting that D2ARs act cooperatively with an array of membrane and intracellular proteins to tightly control DA transmission. This review highlights D2AR-interacting partners including transporters, G-protein-coupled receptors, ion channels, intracellular signaling modulators, and protein kinases. The complexity of the D2AR interaction network illustrates the functional divergence of D2ARs. Pharmacological targeting of multiple D2AR-interacting partners may be more effective to restore disrupted DA homeostasis by drugs of abuse.
Collapse
Affiliation(s)
- Rong Chen
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America.
| | - Mark J Ferris
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| | - Shiyu Wang
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| |
Collapse
|
48
|
Nolan SO, Zachry JE, Johnson AR, Brady LJ, Siciliano CA, Calipari ES. Direct dopamine terminal regulation by local striatal microcircuitry. J Neurochem 2020; 155:475-493. [PMID: 32356315 DOI: 10.1111/jnc.15034] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
Regulation of axonal dopamine release by local microcircuitry is at the hub of several biological processes that govern the timing and magnitude of signaling events in reward-related brain regions. An important characteristic of dopamine release from axon terminals in the striatum is that it is rapidly modulated by local regulatory mechanisms. These processes can occur via homosynaptic mechanisms-such as presynaptic dopamine autoreceptors and dopamine transporters - as well heterosynaptic mechanisms such as retrograde signaling from postsynaptic cholinergic and dynorphin systems, among others. Additionally, modulation of dopamine release via diffusible messengers, such as nitric oxide and hydrogen peroxide, allows for various metabolic factors to quickly and efficiently regulate dopamine release and subsequent signaling. Here we review how these mechanisms work in concert to influence the timing and magnitude of striatal dopamine signaling, independent of action potential activity at the level of dopaminergic cell bodies in the midbrain, thereby providing a parallel pathway by which dopamine can be modulated. Understanding the complexities of local regulation of dopamine signaling is required for building comprehensive frameworks of how activity throughout the dopamine system is integrated to drive signaling and control behavior.
Collapse
Affiliation(s)
- Suzanne O Nolan
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jennifer E Zachry
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Amy R Johnson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Lillian J Brady
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN TN, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.,Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
49
|
Fagan RR, Kearney PJ, Melikian HE. In Situ Regulated Dopamine Transporter Trafficking: There's No Place Like Home. Neurochem Res 2020; 45:1335-1343. [PMID: 32146647 DOI: 10.1007/s11064-020-03001-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Dopamine (DA) is critical for motivation, reward, movement initiation, and learning. Mechanisms that control DA signaling have a profound impact on these important behaviors, and additionally play a role in DA-related neuropathologies. The presynaptic SLC6 DA transporter (DAT) limits extracellular DA levels by clearing released DA, and is potently inhibited by addictive and therapeutic psychostimulants. Decades of evidence support that the DAT is subject to acute regulation by a number of signaling pathways, and that endocytic trafficking strongly regulates DAT availability and function. DAT trafficking studies have been performed in a variety of model systems, including both in vitro and ex vivo preparations. In this review, we focus on the breadth of DAT trafficking studies, with specific attention to, and comparison of, how context may influence DAT's response to different stimuli. In particular, this overview highlights that stimulated DAT trafficking not only differs between in vitro and ex vivo environments, but also is influenced by both sex and anatomical subregions.
Collapse
Affiliation(s)
- Rita R Fagan
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Patrick J Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Haley E Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
50
|
Lebowitz JJ, Khoshbouei H. Heterogeneity of dopamine release sites in health and degeneration. Neurobiol Dis 2019; 134:104633. [PMID: 31698055 DOI: 10.1016/j.nbd.2019.104633] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/12/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
Despite comprising only ~ 0.001% of all neurons in the human brain, ventral midbrain dopamine neurons exert a profound influence on human behavior and cognition. As a neuromodulator, dopamine selectively inhibits or enhances synaptic signaling to coordinate neural output for action, attention, and affect. Humans invariably lose brain dopamine during aging, and this can be exacerbated in disease states such as Parkinson's Disease. Further, it is well established in multiple disease states that cell loss is selective for a subset of highly sensitive neurons within the nigrostriatal dopamine tract. Regional differences in dopamine tone are regulated pre-synaptically, with subcircuits of projecting dopamine neurons exhibiting distinct molecular and physiological signatures. Specifically, proteins at dopamine release sites that synthesize and package cytosolic dopamine, modulate its release and reuptake, and alter neuronal excitability show regional differences that provide linkages to the observed sensitivity to neurodegeneration. The aim of this review is to outline the major components of dopamine homeostasis at neurotransmitter release sites and describe the regional differences most relevant to understanding why some, but not all, dopamine neurons exhibit heightened vulnerability to neurodegeneration.
Collapse
Affiliation(s)
- Joseph J Lebowitz
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|