1
|
Herzog N, Hartmann H, Janssen LK, Kanyamibwa A, Waltmann M, Kovacs P, Deserno L, Fallon S, Villringer A, Horstmann A. Working memory gating in obesity is moderated by striatal dopaminergic gene variants. eLife 2024; 13:RP93369. [PMID: 39431987 PMCID: PMC11493406 DOI: 10.7554/elife.93369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
Everyday life requires an adaptive balance between distraction-resistant maintenance of information and the flexibility to update this information when needed. These opposing mechanisms are proposed to be balanced through a working memory gating mechanism. Prior research indicates that obesity may elevate the risk of working memory deficits, yet the underlying mechanisms remain elusive. Dopaminergic alterations have emerged as a potential mediator. However, current models suggest these alterations should only shift the balance in working memory tasks, not produce overall deficits. The empirical support for this notion is currently lacking, however. To address this gap, we pooled data from three studies (N = 320) where participants performed a working memory gating task. Higher BMI was associated with overall poorer working memory, irrespective of whether there was a need to maintain or update information. However, when participants, in addition to BMI level, were categorized based on certain putative dopamine-signaling characteristics (single-nucleotide polymorphisms [SNPs]; specifically, Taq1A and DARPP-32), distinct working memory gating effects emerged. These SNPs, primarily associated with striatal dopamine transmission, appear to be linked with differences in updating, specifically, among high-BMI individuals. Moreover, blood amino acid ratio, which indicates central dopamine synthesis capacity, combined with BMI shifted the balance between distractor-resistant maintenance and updating. These findings suggest that both dopamine-dependent and dopamine-independent cognitive effects exist in obesity. Understanding these effects is crucial if we aim to modify maladaptive cognitive profiles in individuals with obesity.
Collapse
Affiliation(s)
- Nadine Herzog
- Department of Neurology, Max Planck Institute for Human Cognitive & Brain SciencesLeipzigGermany
- International Max Planck Research School NeuroComLeipzigGermany
| | - Hendrik Hartmann
- Department of Neurology, Max Planck Institute for Human Cognitive & Brain SciencesLeipzigGermany
- Collaborative Research Centre 1052, University of LeipzigLeipzigGermany
- Department of Psychology and Logopedics, Faculty of Medicine, University of HelsinkiHelsinkiFinland
| | - Lieneke Katharina Janssen
- Department of Neurology, Max Planck Institute for Human Cognitive & Brain SciencesLeipzigGermany
- Institute of Psychology, Otto von Guericke University MagdeburgMagdeburgGermany
| | - Arsene Kanyamibwa
- Department of Psychology and Logopedics, Faculty of Medicine, University of HelsinkiHelsinkiFinland
| | - Maria Waltmann
- Department of Neurology, Max Planck Institute for Human Cognitive & Brain SciencesLeipzigGermany
- Department of Child and Adolescent Psychiatry, University of WürzburgWürzburgGermany
| | - Peter Kovacs
- Medical Department III – Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical CenterLeipzigGermany
| | - Lorenz Deserno
- Department of Child and Adolescent Psychiatry, University of WürzburgWürzburgGermany
- Department of Psychiatry and Psychotherapy, Technische Universität DresdenDresdenGermany
| | - Sean Fallon
- School of Psychology, University of PlymouthPlymouthUnited Kingdom
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive & Brain SciencesLeipzigGermany
| | - Annette Horstmann
- Department of Neurology, Max Planck Institute for Human Cognitive & Brain SciencesLeipzigGermany
- Collaborative Research Centre 1052, University of LeipzigLeipzigGermany
- Department of Psychology and Logopedics, Faculty of Medicine, University of HelsinkiHelsinkiFinland
| |
Collapse
|
2
|
Lindsey J, Markowitz JE, Gillis WF, Datta SR, Litwin-Kumar A. Dynamics of striatal action selection and reinforcement learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580408. [PMID: 38464083 PMCID: PMC10925202 DOI: 10.1101/2024.02.14.580408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Spiny projection neurons (SPNs) in dorsal striatum are often proposed as a locus of reinforcement learning in the basal ganglia. Here, we identify and resolve a fundamental inconsistency between striatal reinforcement learning models and known SPN synaptic plasticity rules. Direct-pathway (dSPN) and indirect-pathway (iSPN) neurons, which promote and suppress actions, respectively, exhibit synaptic plasticity that reinforces activity associated with elevated or suppressed dopamine release. We show that iSPN plasticity prevents successful learning, as it reinforces activity patterns associated with negative outcomes. However, this pathological behavior is reversed if functionally opponent dSPNs and iSPNs, which promote and suppress the current behavior, are simultaneously activated by efferent input following action selection. This prediction is supported by striatal recordings and contrasts with prior models of SPN representations. In our model, learning and action selection signals can be multiplexed without interference, enabling learning algorithms beyond those of standard temporal difference models.
Collapse
|
3
|
Gupta S, Tielemans A, Guevara CA, Huntley GW, Benson DL. Parkinson's-linked LRRK2-G2019S derails AMPAR trafficking, mobility, and composition in striatum with cell-type and subunit specificity. Proc Natl Acad Sci U S A 2024; 121:e2317833121. [PMID: 38968112 PMCID: PMC11252801 DOI: 10.1073/pnas.2317833121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/27/2024] [Indexed: 07/07/2024] Open
Abstract
Parkinson's disease (PD) is a multifactorial disease that affects multiple brain systems and circuits. While defined by motor symptoms caused by degeneration of brainstem dopamine neurons, debilitating non-motor abnormalities in fronto-striatal-based cognitive function are common, appear early, and are initially independent of dopamine. Young adult mice expressing the PD-associated G2019S missense mutation in Lrrk2 also exhibit deficits in fronto-striatal-based cognitive tasks. In mice and humans, cognitive functions require dynamic adjustments in glutamatergic synapse strength through cell-surface trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs), but it is unknown how LRRK2 mutation impacts dynamic features of AMPAR trafficking in striatal projection neurons (SPNs). Here, we used Lrrk2G2019S knockin mice to show that surface AMPAR subunit stoichiometry is altered biochemically and functionally in mutant SPNs in dorsomedial striatum to favor the incorporation of GluA1 over GluA2. GluA1-containing AMPARs were resistant to internalization from the cell surface, leaving an excessive accumulation of GluA1 on the surface within and outside synapses. This negatively impacted trafficking dynamics that normally support synapse strengthening, as GluA1-containing AMPARs failed to increase at synapses in response to a potentiating stimulus and showed significantly reduced surface mobility. Surface GluA2-containing AMPARs were expressed at normal levels in synapses, indicating subunit-selective impairment. Abnormal surface accumulation of GluA1 was independent of PKA activity and was limited to D1R SPNs. Since LRRK2 mutation is thought to be part of a common PD pathogenic pathway, our data suggest that sustained, striatal cell-type specific changes in AMPAR composition and trafficking contribute to cognitive or other impairments associated with PD.
Collapse
Affiliation(s)
- Swati Gupta
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Alexander Tielemans
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Christopher A. Guevara
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - George W. Huntley
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Deanna L. Benson
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| |
Collapse
|
4
|
Khandelwal N, Kulkarni A, Ahmed NI, Harper M, Konopka G, Gibson JR. FOXP1 regulates the development of excitatory synaptic inputs onto striatal neurons and induces phenotypic reversal with reinstatement. SCIENCE ADVANCES 2024; 10:eadm7039. [PMID: 38701209 PMCID: PMC11068015 DOI: 10.1126/sciadv.adm7039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/01/2024] [Indexed: 05/05/2024]
Abstract
Long-range glutamatergic inputs originating from the cortex and thalamus are indispensable for striatal development, providing the foundation for motor and cognitive functions. Despite their significance, transcriptional regulation governing these inputs remains largely unknown. We investigated the role of a transcription factor encoded by a high-risk autism-associated gene, FOXP1, in sculpting glutamatergic inputs onto spiny projection neurons (SPNs) within the striatum. We find a neuron subtype-specific role of FOXP1 in strengthening and maturing glutamatergic inputs onto dopamine receptor 2-expressing SPNs (D2 SPNs). We also find that FOXP1 promotes synaptically driven excitability in these neurons. Using single-nuclei RNA sequencing, we identify candidate genes that mediate these cell-autonomous processes through postnatal FOXP1 function at the post-synapse. Last, we demonstrate that postnatal FOXP1 reinstatement rescues electrophysiological deficits, cell type-specific gene expression changes, and behavioral phenotypes. Together, this study enhances our understanding of striatal circuit development and provides proof of concept for a therapeutic approach for FOXP1 syndrome and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nitin Khandelwal
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Newaz I. Ahmed
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Matthew Harper
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | | | | |
Collapse
|
5
|
Bove F, Angeloni B, Sanginario P, Rossini PM, Calabresi P, Di Iorio R. Neuroplasticity in levodopa-induced dyskinesias: An overview on pathophysiology and therapeutic targets. Prog Neurobiol 2024; 232:102548. [PMID: 38040324 DOI: 10.1016/j.pneurobio.2023.102548] [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: 07/18/2023] [Revised: 10/29/2023] [Accepted: 11/26/2023] [Indexed: 12/03/2023]
Abstract
Levodopa-induced dyskinesias (LIDs) are a common complication in patients with Parkinson's disease (PD). A complex cascade of electrophysiological and molecular events that induce aberrant plasticity in the cortico-basal ganglia system plays a key role in the pathophysiology of LIDs. In the striatum, multiple neurotransmitters regulate the different forms of physiological synaptic plasticity to provide it in a bidirectional and Hebbian manner. In PD, impairment of both long-term potentiation (LTP) and long-term depression (LTD) progresses with disease and dopaminergic denervation of striatum. The altered balance between LTP and LTD processes leads to unidirectional changes in plasticity that cause network dysregulation and the development of involuntary movements. These alterations have been documented, in both experimental models and PD patients, not only in deep brain structures but also at motor cortex. Invasive and non-invasive neuromodulation treatments, as deep brain stimulation, transcranial magnetic stimulation, or transcranial direct current stimulation, may provide strategies to modulate the aberrant plasticity in the cortico-basal ganglia network of patients affected by LIDs, thus restoring normal neurophysiological functioning and treating dyskinesias. In this review, we discuss the evidence for neuroplasticity impairment in experimental PD models and in patients affected by LIDs, and potential neuromodulation strategies that may modulate aberrant plasticity.
Collapse
Affiliation(s)
- Francesco Bove
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Benedetta Angeloni
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pasquale Sanginario
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paolo Maria Rossini
- Brain Connectivity Laboratory, Department of Neuroscience and Neurorehabilitation, IRCCS San Raffaele Roma, Rome, Italy
| | - Paolo Calabresi
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Di Iorio
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
6
|
Parr AC, Riek HC, Coe BC, Pari G, Masellis M, Marras C, Munoz DP. Genetic variation in the dopamine system is associated with mixed-strategy decision-making in patients with Parkinson's disease. Eur J Neurosci 2023; 58:4523-4544. [PMID: 36453013 DOI: 10.1111/ejn.15875] [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: 07/15/2022] [Revised: 11/16/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022]
Abstract
Decision-making during mixed-strategy games requires flexibly adapting choice strategies in response to others' actions and dynamically tracking outcomes. Such decisions involve diverse cognitive processes, including reinforcement learning, which are affected by disruptions to the striatal dopamine system. We therefore investigated how genetic variation in dopamine function affected mixed-strategy decision-making in Parkinson's disease (PD), which involves striatal dopamine pathology. Sixty-six PD patients (ages 49-85, Hoehn and Yahr Stages 1-3) and 22 healthy controls (ages 54-75) competed in a mixed-strategy game where successful performance depended on minimizing choice biases (i.e., flexibly adapting choices trial by trial). Participants also completed a fixed-strategy task that was matched for sensory input, motor outputs and overall reward rate. Factor analyses were used to disentangle cognitive from motor aspects within both tasks. Using a within-subject, multi-centre design, patients were examined on and off dopaminergic therapy, and genetic variation was examined via a multilocus genetic profile score representing the additive effects of three single nucleotide polymorphisms (SNPs) that influence dopamine transmission: rs4680 (COMT Val158 Met), rs6277 (C957T) and rs907094 (encoding DARPP-32). PD and control participants displayed comparable mixed-strategy choice behaviour (overall); however, PD patients with genetic profile scores indicating higher dopamine transmission showed improved performance relative to those with low scores. Exploratory follow-up tests across individual SNPs revealed better performance in individuals with the C957T polymorphism, reflecting higher striatal D2/D3 receptor density. Importantly, genetic variation modulated cognitive aspects of performance, above and beyond motor function, suggesting that genetic variation in dopamine signalling may underlie individual differences in cognitive function in PD.
Collapse
Affiliation(s)
- Ashley C Parr
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Heidi C Riek
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Brian C Coe
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Giovanna Pari
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
- Movement Disorder Clinic, Kingston General Hospital, Kingston, Ontario, Canada
| | - Mario Masellis
- Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Connie Marras
- Movement Disorders Clinic, Krembil Neuroscience Centre, University Health Network, Toronto, Ontario, Canada
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
7
|
Khandelwal N, Kulkarni A, Ahmed NI, Harper M, Konopka G, Gibson J. FOXP1 regulates the development of excitatory synaptic inputs onto striatal neurons and induces phenotypic reversal with reinstatement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563675. [PMID: 37961477 PMCID: PMC10634768 DOI: 10.1101/2023.10.23.563675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Long-range glutamatergic inputs from the cortex and thalamus are critical for motor and cognitive processing in the striatum. Transcription factors that orchestrate the development of these inputs are largely unknown. We investigated the role of a transcription factor and high-risk autism-associated gene, FOXP1, in the development of glutamatergic inputs onto spiny projection neurons (SPNs) in the striatum. We find that FOXP1 robustly drives the strengthening and maturation of glutamatergic input onto dopamine receptor 2-expressing SPNs (D2 SPNs) but has a comparatively milder effect on D1 SPNs. This process is cell-autonomous and is likely mediated through postnatal FOXP1 function at the postsynapse. We identified postsynaptic FOXP1-regulated transcripts as potential candidates for mediating these effects. Postnatal reinstatement of FOXP1 rescues electrophysiological deficits, reverses gene expression alterations resulting from embryonic deletion, and mitigates behavioral phenotypes. These results provide support for a possible therapeutic approach for individuals with FOXP1 syndrome.
Collapse
|
8
|
Gupta S, Guevara CA, Tielemans A, Huntley GW, Benson DL. Parkinson's-linked LRRK2-G2019S derails AMPAR trafficking, mobility and composition in striatum with cell-type and subunit specificity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562231. [PMID: 37905106 PMCID: PMC10614818 DOI: 10.1101/2023.10.13.562231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Parkinson's (PD) is a multi-factorial disease that affects multiple brain systems and circuits. While defined by motor symptoms caused by degeneration of brainstem dopamine neurons, debilitating non-motor abnormalities in fronto-striatal based cognitive function are common, appear early and are initially independent of dopamine. Young adult mice expressing the PD-associated G2019S missense mutation in Lrrk2 also exhibit deficits in fronto-striatal-based cognitive tasks. In mice and humans, cognitive functions require dynamic adjustments in glutamatergic synapse strength through cell-surface trafficking of AMPA-type glutamate receptors (AMPARs), but it is unknown how LRRK2 mutation impacts dynamic features of AMPAR trafficking in striatal projection neurons (SPNs). Here, we used Lrrk2 G2019S knockin mice to show that surface AMPAR subunit stoichiometry is altered biochemically and functionally in mutant SPNs to favor incorporation of GluA1 over GluA2. GluA1-containing AMPARs were resistant to internalization from the cell surface, leaving an excessive accumulation of GluA1 on the surface within and outside synapses. This negatively impacted trafficking dynamics that normally support synapse strengthening, as GluA1-containing AMPARs failed to increase at synapses in response to a potentiating stimulus and showed significantly reduced surface mobility. Surface GluA2-containing AMPARs were expressed at normal levels in synapses, indicating subunit-selective impairment. Abnormal surface accumulation of GluA1 was independent of PKA activity and was limited to D 1 R SPNs. Since LRRK2 mutation is thought to be part of a common PD pathogenic pathway, our data suggest that sustained, striatal cell-type specific changes in AMPAR composition and trafficking contribute to cognitive or other impairments associated with PD. SIGNIFICANCE STATEMENT Mutations in LRRK2 are common genetic risks for PD. Lrrk2 G2019S mice fail to exhibit long-term potentiation at corticostriatal synapses and show significant deficits in frontal-striatal based cognitive tasks. While LRRK2 has been implicated generally in protein trafficking, whether G2019S derails AMPAR trafficking at synapses on striatal neurons (SPNs) is unknown. We show that surface GluA1-AMPARs fail to internalize and instead accumulate excessively within and outside synapses. This effect is selective to D 1 R SPNs and negatively impacts synapse strengthening as GluA1-AMPARs fail to increase at the surface in response to potentiation and show limited surface mobility. Thus, LRRK2-G2019S narrows the effective range of plasticity mechanisms, supporting the idea that cognitive symptoms reflect an imbalance in AMPAR trafficking mechanisms within cell-type specific projections.
Collapse
|
9
|
Lewitus VJ, Blackwell KT. Estradiol Receptors Inhibit Long-Term Potentiation in the Dorsomedial Striatum. eNeuro 2023; 10:ENEURO.0071-23.2023. [PMID: 37487741 PMCID: PMC10405883 DOI: 10.1523/eneuro.0071-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/25/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023] Open
Abstract
Estradiol, a female sex hormone and the predominant form of estrogen, has diverse effects throughout the brain including in learning and memory. Estradiol modulates several types of learning that depend on the dorsomedial striatum (DMS), a subregion of the basal ganglia involved in goal-directed learning, cued action-selection, and motor skills. A cellular basis of learning is synaptic plasticity, and the presence of extranuclear estradiol receptors ERα, ERβ, and G-protein-coupled estrogen receptor (GPER) throughout the DMS suggests that estradiol may influence rapid cellular actions including those involved in plasticity. To test whether estradiol affects synaptic plasticity in the DMS, corticostriatal long-term potentiation (LTP) was induced using theta-burst stimulation (TBS) in ex vivo brain slices from intact male and female C57BL/6 mice. Extracellular field recordings showed that female mice in the diestrous stage of the estrous cycle exhibited LTP similar to male mice, while female mice in estrus did not exhibit LTP. Furthermore, antagonists of ERα or GPER rescued LTP in estrous females and agonists of ERα or GPER reduced LTP in diestrous females. In males, activating ERα but not GPER reduced LTP. These results uncover an inhibitory action of estradiol receptors on cellular learning in the DMS and suggest a cellular mechanism underlying the impairment in certain types of DMS-based learning observed in the presence of high estradiol. Because of the dorsal striatum's role in substance use disorders, these findings may provide a mechanism underlying an estradiol-mediated progression from goal-directed to habitual drug use.
Collapse
Affiliation(s)
| | - Kim T Blackwell
- Interdisciplinary Neuroscience PhD Program
- Department of Bioengineering, George Mason University, Fairfax, VA 22030
| |
Collapse
|
10
|
Marino G, Campanelli F, Natale G, De Carluccio M, Servillo F, Ferrari E, Gardoni F, Caristo ME, Picconi B, Cardinale A, Loffredo V, Crupi F, De Leonibus E, Viscomi MT, Ghiglieri V, Calabresi P. Intensive exercise ameliorates motor and cognitive symptoms in experimental Parkinson's disease restoring striatal synaptic plasticity. SCIENCE ADVANCES 2023; 9:eadh1403. [PMID: 37450585 PMCID: PMC10348672 DOI: 10.1126/sciadv.adh1403] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Intensive physical activity improves motor functions in patients with Parkinson's disease (PD) at early stages. However, the mechanisms underlying the beneficial effects of exercise on PD-associated neuronal alterations have not been fully clarified yet. Here, we tested the hypothesis that an intensive treadmill training program rescues alterations in striatal plasticity and early motor and cognitive deficits in rats receiving an intrastriatal injection of alpha-synuclein (α-syn) preformed fibrils. Improved motor control and visuospatial learning in active animals were associated with a recovery of dendritic spine density alterations and a lasting rescue of a physiological corticostriatal long-term potentiation (LTP). Pharmacological analyses of LTP show that modulations of N-methyl-d-aspartate receptors bearing GluN2B subunits and tropomyosin receptor kinase B, the main brain-derived neurotrophic factor receptor, are involved in these beneficial effects. We demonstrate that intensive exercise training has effects on the early plastic alterations induced by α-syn aggregates and reduces the spread of toxic α-syn species to other vulnerable brain areas.
Collapse
Affiliation(s)
- Gioia Marino
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Campanelli
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppina Natale
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria De Carluccio
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Neurosciences and Neurorehabilitation IRCCS S.Raffaele-Roma, Rome, Italy
| | - Federica Servillo
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Ferrari
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milan, Italy
| | | | - Barbara Picconi
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, Rome, Italy
- IRCCS San Raffaele Roma, Lab. Neurofisiologia Sperimentale, Roma, Italy
| | - Antonella Cardinale
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
- IRCCS San Raffaele Roma, Lab. Neurofisiologia Sperimentale, Roma, Italy
| | - Vittorio Loffredo
- Institute of Biochemistry and Cell Biology, National Research Council, Monterotondo (Rome), Italy
| | - Francesco Crupi
- Institute of Biochemistry and Cell Biology, National Research Council, Monterotondo (Rome), Italy
| | - Elvira De Leonibus
- Institute of Biochemistry and Cell Biology, National Research Council, Monterotondo (Rome), Italy
- Telethon Institute of Genetics and Medicine, Telethon Foundation, Pozzuoli (NA), Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Veronica Ghiglieri
- Department of Human Sciences and Quality of Life Promotion, Università Telematica San Raffaele, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paolo Calabresi
- Sezione di Neurologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
11
|
Andreska T, Lüningschrör P, Wolf D, McFleder RL, Ayon-Olivas M, Rattka M, Drechsler C, Perschin V, Blum R, Aufmkolk S, Granado N, Moratalla R, Sauer M, Monoranu C, Volkmann J, Ip CW, Stigloher C, Sendtner M. DRD1 signaling modulates TrkB turnover and BDNF sensitivity in direct pathway striatal medium spiny neurons. Cell Rep 2023; 42:112575. [PMID: 37252844 DOI: 10.1016/j.celrep.2023.112575] [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: 05/12/2022] [Revised: 03/09/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
Disturbed motor control is a hallmark of Parkinson's disease (PD). Cortico-striatal synapses play a central role in motor learning and adaption, and brain-derived neurotrophic factor (BDNF) from cortico-striatal afferents modulates their plasticity via TrkB in striatal medium spiny projection neurons (SPNs). We studied the role of dopamine in modulating the sensitivity of direct pathway SPNs (dSPNs) to BDNF in cultures of fluorescence-activated cell sorting (FACS)-enriched D1-expressing SPNs and 6-hydroxydopamine (6-OHDA)-treated rats. DRD1 activation causes enhanced TrkB translocation to the cell surface and increased sensitivity for BDNF. In contrast, dopamine depletion in cultured dSPN neurons, 6-OHDA-treated rats, and postmortem brain of patients with PD reduces BDNF responsiveness and causes formation of intracellular TrkB clusters. These clusters associate with sortilin related VPS10 domain containing receptor 2 (SORCS-2) in multivesicular-like structures, which apparently protects them from lysosomal degradation. Thus, impaired TrkB processing might contribute to disturbed motor function in PD.
Collapse
Affiliation(s)
- Thomas Andreska
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Daniel Wolf
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Rhonda L McFleder
- Department of Neurology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Maurilyn Ayon-Olivas
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany
| | - Marta Rattka
- Department of Neurology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Christine Drechsler
- Department of Microbiology, Biocenter, Julius-Maximilians-University Wuerzburg, 97074 Wuerzburg, Germany
| | - Veronika Perschin
- Imaging Core Facility of the Biocenter, Julius-Maximilians-University Wuerzburg, 97074 Wuerzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Sarah Aufmkolk
- Department of Biotechnology and Biophysics, Julius-Maximilians-University Wuerzburg, 97074 Wuerzburg, Germany; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Noelia Granado
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; CIBERNED, Instituto de Salud Carlos III, 28002 Madrid, Spain
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Julius-Maximilians-University Wuerzburg, 97074 Wuerzburg, Germany
| | - Camelia Monoranu
- Department for Neuropathology, Julius-Maximilians-University Wuerzburg, 97080 Wuerzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Christian Stigloher
- Imaging Core Facility of the Biocenter, Julius-Maximilians-University Wuerzburg, 97074 Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany.
| |
Collapse
|
12
|
Yu J, Sesack SR, Huang Y, Schlüter OM, Grace AA, Dong Y. Contingent Amygdala Inputs Trigger Heterosynaptic LTP at Hippocampus-To-Accumbens Synapses. J Neurosci 2022; 42:6581-6592. [PMID: 35840324 PMCID: PMC9410749 DOI: 10.1523/jneurosci.0838-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/14/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
Abstract
The nucleus accumbens shell (NAcSh) is a key brain region where environmental cues acquire incentive salience to reinforce motivated behaviors. Principal medium spiny neurons (MSNs) in the NAcSh receive extensive glutamatergic projections from limbic regions, among which, the ventral hippocampus (vH) transmits information enriched in contextual cues, and the basolateral amygdala (BLA) encodes real-time arousing states. The vH and BLA project convergently to NAcSh MSNs, both activated in a time-locked manner on a cue-conditioned motivational action. In brain slices prepared from male and female mice, we show that co-activation of the two projections induces long-term potentiation (LTP) at vH-to-NAcSh synapses without affecting BLA-to-NAcSh synapses, revealing a heterosynaptic mechanism through which BLA signals persistently increase the temporally contingent vH-to-NAcSh transmission. Furthermore, this LTP is more prominent in dopamine D1 receptor-expressing (D1) MSNs than D2 MSNs and can be prevented by inhibition of either D1 receptors or dopaminergic terminals in NAcSh. This heterosynaptic LTP may provide a dopamine-guided mechanism through which vH-encoded cue inputs that are contingent to BLA activation acquire increased circuit representation to reinforce behavior.SIGNIFICANCE STATEMENT In motivated behaviors, environmental cues associated with arousing stimuli acquire increased incentive salience, processes mediated in part by the nucleus accumbens (NAc). NAc principal neurons receive glutamatergic projections from the ventral hippocampus (vH) and basolateral amygdala (BLA), which transmit information encoding contextual cues and affective states, respectively. Our results show that co-activation of the two projections induces long-term potentiation (LTP) at vH-to-NAc synapses without affecting BLA-to-NAc synapses, revealing a heterosynaptic mechanism through which BLA signals potentiate the temporally contingent vH-to-NAc transmission. Furthermore, this LTP is prevented by inhibition of either D1 receptors or dopaminergic axons. This heterosynaptic LTP may provide a dopamine-guided mechanism through which vH-encoded cue inputs that are contingent to BLA activation acquire increased circuit representation to reinforce behavior.
Collapse
Affiliation(s)
- Jun Yu
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Susan R Sesack
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Yanhua Huang
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Oliver M Schlüter
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| |
Collapse
|
13
|
Thomas R, Hernandez A, Benavides DR, Li W, Tan C, Umfress A, Plattner F, Chakraborti A, Pozzo-Miller L, Taylor SS, Bibb JA. Integrated regulation of PKA by fast and slow neurotransmission in the nucleus accumbens controls plasticity and stress responses. J Biol Chem 2022; 298:102245. [PMID: 35835216 PMCID: PMC9386499 DOI: 10.1016/j.jbc.2022.102245] [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: 04/19/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cortical glutamate and midbrain dopamine neurotransmission converge to mediate striatum-dependent behaviors, while maladaptations in striatal circuitry contribute to mental disorders. However, the crosstalk between glutamate and dopamine signaling has not been entirely elucidated. Here we uncover a molecular mechanism by which glutamatergic and dopaminergic signaling integrate to regulate cAMP-dependent protein kinase (PKA) via phosphorylation of the PKA regulatory subunit, RIIβ. Using a combination of biochemical, pharmacological, neurophysiological, and behavioral approaches, we find that glutamate-dependent reduction in cyclin-dependent kinase 5 (Cdk5)-dependent RIIβ phosphorylation alters the PKA holoenzyme autoinhibitory state to increase PKA signaling in response to dopamine. Furthermore, we show that disruption of RIIβ phosphorylation by Cdk5 enhances cortico-ventral striatal synaptic plasticity. In addition, we demonstrate that acute and chronic stress in rats inversely modulate RIIβ phosphorylation and ventral striatal infusion of a small interfering peptide that selectively targets RIIβ regulation by Cdk5 improves behavioral response to stress. We propose this new signaling mechanism integrating ventral striatal glutamate and dopamine neurotransmission is important to brain function, may contribute to neuropsychiatric conditions, and serves as a possible target for the development of novel therapeutics for stress-related disorders.
Collapse
Affiliation(s)
- Rachel Thomas
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adan Hernandez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, Santiago de Querétaro, Querétaro, México; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David R Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Li
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Chunfeng Tan
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alan Umfress
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Florian Plattner
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ayanabha Chakraborti
- Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA
| | - Susan S Taylor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - James A Bibb
- Department of Neurobiology, Civitan International Research Center, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Surgery, The University of Alabama Birmingham Medical Center, Birmingham, AL 35233, USA; Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
14
|
Li T, Xu G, Yi J, Huang Y. Intraoperative Hypothermia Induces Vascular Dysfunction in the CA1 Region of Rat Hippocampus. Brain Sci 2022; 12:brainsci12060692. [PMID: 35741578 PMCID: PMC9221322 DOI: 10.3390/brainsci12060692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Intraoperative hypothermia is very common and leads to memory decline. The hippocampus is responsible for memory formation. As a functional core area, the cornu ammonis 1 (CA1) region of the hippocampus contains abundant blood vessels and is susceptible to ischemia. The aim of the study was to explore vascular function and neuronal state in the CA1 region of rats undergoing intraoperative hypothermia. The neuronal morphological change and activity-regulated cytoskeleton-associated protein (Arc) expression were evaluated by haematoxylin-eosin staining and immunofluorescence respectively. Histology and immunohistochemistry were used to assess vascular function. Results showed that intraoperative hypothermia inhibited the expression of vascular endothelial growth factor and endothelial nitric oxide synthase, and caused reactive oxygen species accumulation. Additionally, the phenotype of vascular smooth muscle cells was transformed from contractile to synthetic, showing a decrease in smooth muscle myosin heavy chain and an increase in osteopontin. Ultimately, vascular dysfunction caused neuronal pyknosis in the CA1 region and reduced memory-related Arc expression. In conclusion, neuronal disorder in the CA1 region was caused by intraoperative hypothermia-related vascular dysfunction. This study could provide a novel understanding of the effect of intraoperative hypothermia in the hippocampus, which might identify a new research target and treatment strategy.
Collapse
|
15
|
Adaptive changes in striatal projection neurons explain the long duration response and the emergence of dyskinesias in patients with Parkinson's disease. J Neural Transm (Vienna) 2022; 129:497-503. [PMID: 35538324 PMCID: PMC9188504 DOI: 10.1007/s00702-022-02510-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/28/2022] [Indexed: 11/06/2022]
Abstract
Neuronal activity in the brain is tightly regulated. During operation in real time, for instance, feedback and feedforward loops limit excessive excitation. In addition, cell autonomous processes ensure that neurons’ average activity is restored to a setpoint in response to chronic perturbations. These processes are summarized as homeostatic plasticity (Turrigiano in Cold Spring Harb Perspect Biol 4:a005736–a005736, 2012). In the basal ganglia, information is mainly transmitted through disinhibition, which already constraints the possible range of neuronal activity. When this tightly adjusted system is challenged by the chronic decline in dopaminergic neurotransmission in Parkinson’s disease (PD), homeostatic plasticity aims to compensate for this perturbation. We here summarize recent experimental work from animals demonstrating that striatal projection neurons adapt excitability and morphology in response to chronic dopamine depletion and substitution. We relate these cellular processes to clinical observations in patients with PD that cannot be explained by the classical model of basal ganglia function. These include the long duration response to dopaminergic medication that takes weeks to develop and days to wear off. Moreover, dyskinesias are considered signs of excessive dopaminergic neurotransmission in Parkinson’s disease, but they are typically more severe on the body side that is more strongly affected by dopamine depletion. We hypothesize that these clinical observations can be explained by homeostatic plasticity in the basal ganglia, suggesting that plastic changes in response to chronic dopamine depletion and substitution need to be incorporated into models of basal ganglia function. In addition, better understanding the molecular mechanism of homeostatic plasticity might offer new treatment options to avoid motor complications in patients with PD.
Collapse
|
16
|
Striatal glutamatergic hyperactivity in Parkinson's disease. Neurobiol Dis 2022; 168:105697. [DOI: 10.1016/j.nbd.2022.105697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
|
17
|
Bove F, Calabresi P. Plasticity, genetics, and epigenetics in l-dopa-induced dyskinesias. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:167-184. [PMID: 35034732 DOI: 10.1016/b978-0-12-819410-2.00009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
l-Dopa-induced dyskinesias (LIDs) are a frequent complication in l-dopa-treated patients affected by Parkinson's disease (PD). In the last years, several progresses in the knowledge of LIDs mechanisms have led to the identification of several molecular and electrophysiologic events. A complex cascade of intracellular events underlies the pathophysiology of LIDs, and, among these, aberrant plasticity in the cortico-basal ganglia system, at striatal and cortical level, plays a key role. Furthermore, several recent studies have investigated genetic susceptibility and epigenetic modifications in LIDs pathophysiology that might have future relevance in clinical practice and pharmacologic research. These progresses might lead to the development of specific strategies not only to treat, but also to prevent or delay the development of LIDs in PD.
Collapse
Affiliation(s)
- Francesco Bove
- UOC Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paolo Calabresi
- UOC Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
18
|
Abstract
The last century was characterized by a significant scientific effort aimed at unveiling the neurobiological basis of learning and memory. Thanks to the characterization of the mechanisms regulating the long-term changes of neuronal synaptic connections, it was possible to understand how specific neural networks shape themselves during the acquisition of memory traces or complex motor tasks. In this chapter, we will summarize the mechanisms underlying the main forms of synaptic plasticity taking advantage of the studies performed in the hippocampus and in the nucleus striatum, key brain structures that play a crucial role in cognition. Moreover, we will discuss how the molecular pathways involved in the induction of physiologic synaptic long-term changes could be disrupted during neurodegenerative and neuroinflammatory disorders, highlighting the translational relevance of this intriguing research field.
Collapse
Affiliation(s)
- Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Antonio de Iure
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy
| | - Barbara Picconi
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy; University San Raffaele, Rome, Italy.
| |
Collapse
|
19
|
Vallés AS, Barrantes FJ. Nanoscale Sub-Compartmentalization of the Dendritic Spine Compartment. Biomolecules 2021; 11:1697. [PMID: 34827695 PMCID: PMC8615865 DOI: 10.3390/biom11111697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/04/2023] Open
Abstract
Compartmentalization of the membrane is essential for cells to perform highly specific tasks and spatially constrained biochemical functions in topographically defined areas. These membrane lateral heterogeneities range from nanoscopic dimensions, often involving only a few molecular constituents, to micron-sized mesoscopic domains resulting from the coalescence of nanodomains. Short-lived domains lasting for a few milliseconds coexist with more stable platforms lasting from minutes to days. This panoply of lateral domains subserves the great variety of demands of cell physiology, particularly high for those implicated in signaling. The dendritic spine, a subcellular structure of neurons at the receiving (postsynaptic) end of central nervous system excitatory synapses, exploits this compartmentalization principle. In its most frequent adult morphology, the mushroom-shaped spine harbors neurotransmitter receptors, enzymes, and scaffolding proteins tightly packed in a volume of a few femtoliters. In addition to constituting a mesoscopic lateral heterogeneity of the dendritic arborization, the dendritic spine postsynaptic membrane is further compartmentalized into spatially delimited nanodomains that execute separate functions in the synapse. This review discusses the functional relevance of compartmentalization and nanodomain organization in synaptic transmission and plasticity and exemplifies the importance of this parcelization in various neurotransmitter signaling systems operating at dendritic spines, using two fast ligand-gated ionotropic receptors, the nicotinic acetylcholine receptor and the glutamatergic receptor, and a second-messenger G-protein coupled receptor, the cannabinoid receptor, as paradigmatic examples.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca 8000, Argentina;
| | - Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AFF, Argentina
| |
Collapse
|
20
|
Sexually dimorphic prelimbic cortex mechanisms play a role in alcohol dependence: protection by endostatin. Neuropsychopharmacology 2021; 46:1937-1949. [PMID: 34253856 PMCID: PMC8429630 DOI: 10.1038/s41386-021-01075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 02/05/2023]
Abstract
Angiogenesis or proliferation of endothelial cells plays a role in brain microenvironment homeostasis. Previously we have shown enhanced expression of markers of angiogenesis in the medial prefrontal cortex during abstinence in an animal model of ethanol dependence induced by chronic intermittent ethanol vapor (CIE) and ethanol drinking (ED) procedure. Here we report that systemic injections of the angiogenesis inhibitor endostatin reduced relapse to drinking behavior in female CIE-ED rats without affecting relapse to drinking in male CIE-ED rats, and female and male nondependent ED rats. Endostatin did not alter relapse to sucrose drinking in both sexes. Endostatin reduced expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) in all groups; however, rescued expression of tight junction protein claudin-5 in the prelimbic cortex (PLC) of female CIE-ED rats. In both sexes, CIE-ED enhanced microglial activation in the PLC and this was selectively prevented by endostatin in female CIE-ED rats. Endostatin prevented CIE-ED-induced enhanced NF-kB activity and expression and Fos expression in females and did not alter reduced Fos expression in males. Analysis of synaptic processes within the PLC revealed sexually dimorphic adaptations, with CIE-ED reducing synaptic transmission and altering synaptic plasticity in the PLC in females, and increasing synaptic transmission in males. Endostatin prevented the neuroadaptations in the PLC in females via enhancing phosphorylation of CaMKII, without affecting the neuroadaptations in males. Our multifaceted approach is the first to link PLC endothelial cell damage to the behavioral, neuroimmune, and synaptic changes associated with relapse to ethanol drinking in female subjects, and provides a new therapeutic strategy to reduce relapse in dependent subjects.
Collapse
|
21
|
Caveolin-1 Expression in the Dorsal Striatum Drives Methamphetamine Addiction-Like Behavior. Int J Mol Sci 2021; 22:ijms22158219. [PMID: 34360984 PMCID: PMC8348638 DOI: 10.3390/ijms22158219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Dopamine D1 receptor (D1R) function is regulated by membrane/lipid raft-resident protein caveolin-1 (Cav1). We examined whether altered expression of Cav1 in the dorsal striatum would affect self-administration of methamphetamine, an indirect agonist at the D1Rs. A lentiviral construct expressing Cav1 (LV-Cav1) or containing a short hairpin RNA against Cav1 (LV-shCav1) was used to overexpress or knock down Cav1 expression respectively, in the dorsal striatum. Under a fixed-ratio schedule, LV-Cav1 enhanced and LV-shCav1 reduced responding for methamphetamine in an extended access paradigm compared to LV-GFP controls. LV-Cav1 and LV-shCav1 also produced an upward and downward shift in a dose–response paradigm, generating a drug vulnerable/resistant phenotype. LV-Cav1 and LV-shCav1 did not alter responding for sucrose. Under a progressive-ratio schedule, LV-shCav1 generally reduced positive-reinforcing effects of methamphetamine and sucrose as seen by reduced breakpoints. Western blotting confirmed enhanced Cav1 expression in LV-Cav1 rats and reduced Cav1 expression in LV-shCav1 rats. Electrophysiological findings in LV-GFP rats demonstrated an absence of high-frequency stimulation (HFS)-induced long-term potentiation (LTP) in the dorsal striatum after extended access methamphetamine self-administration, indicating methamphetamine-induced occlusion of plasticity. LV-Cav1 prevented methamphetamine-induced plasticity via increasing phosphorylation of calcium calmodulin kinase II, suggesting a mechanism for addiction vulnerability. LV-shCav1 produced a marked deficit in the ability of HFS to produce LTP and, therefore, extended access methamphetamine was unable to alter striatal plasticity, indicating a mechanism for resistance to addiction-like behavior. Our results demonstrate that Cav1 expression and knockdown driven striatal plasticity assist with modulating addiction to drug and nondrug rewards, and inspire new strategies to reduce psychostimulant addiction.
Collapse
|
22
|
Tozzi A, Sciaccaluga M, Loffredo V, Megaro A, Ledonne A, Cardinale A, Federici M, Bellingacci L, Paciotti S, Ferrari E, La Rocca A, Martini A, Mercuri NB, Gardoni F, Picconi B, Ghiglieri V, De Leonibus E, Calabresi P. Dopamine-dependent early synaptic and motor dysfunctions induced by α-synuclein in the nigrostriatal circuit. Brain 2021; 144:3477-3491. [PMID: 34297092 PMCID: PMC8677552 DOI: 10.1093/brain/awab242] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 05/26/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Misfolding and aggregation of α-synuclein are specific features of Parkinson’s disease and other neurodegenerative diseases defined as synucleinopathies. Parkinson’s disease progression has been correlated with the formation and extracellular release of α-synuclein aggregates, as well as with their spread from neuron to neuron. Therapeutic interventions in the initial stages of Parkinson’s disease require a clear understanding of the mechanisms by which α-synuclein disrupts the physiological synaptic and plastic activity of the basal ganglia. For this reason, we identified two early time points to clarify how the intrastriatal injection of α-synuclein-preformed fibrils in rodents via retrograde transmission induces time-dependent electrophysiological and behavioural alterations. We found that intrastriatal α-synuclein-preformed fibrils perturb the firing rate of dopaminergic neurons in the substantia nigra pars compacta, while the discharge of putative GABAergic cells of the substantia nigra pars reticulata is unchanged. The α-synuclein-induced dysregulation of nigrostriatal function also impairs, in a time-dependent manner, the two main forms of striatal synaptic plasticity, long-term potentiation and long-term depression. We also observed an increased glutamatergic transmission measured as an augmented frequency of spontaneous excitatory synaptic currents. These changes in neuronal function in the substantia nigra pars compacta and striatum were observed before overt neuronal death occurred. In an additional set of experiments, we were able to rescue α-synuclein-induced alterations of motor function, striatal synaptic plasticity and increased spontaneous excitatory synaptic currents by subchronic treatment with l-DOPA, a precursor of dopamine widely used in the therapy of Parkinson’s disease, clearly demonstrating that a dysfunctional dopamine system plays a critical role in the early phases of the disease.
Collapse
Affiliation(s)
- Alessandro Tozzi
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Miriam Sciaccaluga
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Vittorio Loffredo
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy.,Institute of Biochemistry and Cell Biology-CNR, 00015 Monterotondo scalo, Italy
| | - Alfredo Megaro
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Ada Ledonne
- Laboratory of Experimental Neuroscience, Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Antonella Cardinale
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Mauro Federici
- Laboratory of Experimental Neuroscience, Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Laura Bellingacci
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Silvia Paciotti
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Elena Ferrari
- University of Milan, Department of Pharmacological and Biomolecular Sciences, 20133 Milan, Italy
| | - Antonino La Rocca
- Institute of Biochemistry and Cell Biology-CNR, 00015 Monterotondo scalo, Italy
| | - Alessandro Martini
- Laboratory of Experimental Neuroscience, Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Nicola B Mercuri
- Laboratory of Experimental Neuroscience, Santa Lucia Foundation IRCCS, 00143 Rome, Italy.,Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Fabrizio Gardoni
- University of Milan, Department of Pharmacological and Biomolecular Sciences, 20133 Milan, Italy
| | - Barbara Picconi
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy.,Telematic University San Raffaele, 00166 Rome, Italy
| | | | - Elvira De Leonibus
- Institute of Biochemistry and Cell Biology-CNR, 00015 Monterotondo scalo, Italy.,Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | - Paolo Calabresi
- Neurological Clinic, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy.,Department of Neuroscience, Faculty of Medicine, Università Cattolica del "Sacro Cuore", 00168 Rome, Italy
| |
Collapse
|
23
|
Verduzco-Mendoza A, Carrillo-Mora P, Avila-Luna A, Gálvez-Rosas A, Olmos-Hernández A, Mota-Rojas D, Bueno-Nava A. Role of the Dopaminergic System in the Striatum and Its Association With Functional Recovery or Rehabilitation After Brain Injury. Front Neurosci 2021; 15:693404. [PMID: 34248494 PMCID: PMC8264205 DOI: 10.3389/fnins.2021.693404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Disabilities are estimated to occur in approximately 2% of survivors of traumatic brain injury (TBI) worldwide, and disability may persist even decades after brain injury. Facilitation or modulation of functional recovery is an important goal of rehabilitation in all patients who survive severe TBI. However, this recovery tends to vary among patients because it is affected by the biological and physical characteristics of the patients; the types, doses, and application regimens of the drugs used; and clinical indications. In clinical practice, diverse dopaminergic drugs with various dosing and application procedures are used for TBI. Previous studies have shown that dopamine (DA) neurotransmission is disrupted following moderate to severe TBI and have reported beneficial effects of drugs that affect the dopaminergic system. However, the mechanisms of action of dopaminergic drugs have not been completely clarified, partly because dopaminergic receptor activation can lead to restoration of the pathway of the corticobasal ganglia after injury in brain structures with high densities of these receptors. This review aims to provide an overview of the functionality of the dopaminergic system in the striatum and its roles in functional recovery or rehabilitation after TBI.
Collapse
Affiliation(s)
- Antonio Verduzco-Mendoza
- Ph.D. Program in Biological and Health Sciences, Universidad Autónoma Metropolitana, Mexico City, Mexico
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Paul Carrillo-Mora
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Alberto Avila-Luna
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Arturo Gálvez-Rosas
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Antonio Bueno-Nava
- Division of Neurosciences, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Mexico City, Mexico
| |
Collapse
|
24
|
Mancini A, Ghiglieri V, Parnetti L, Calabresi P, Di Filippo M. Neuro-Immune Cross-Talk in the Striatum: From Basal Ganglia Physiology to Circuit Dysfunction. Front Immunol 2021; 12:644294. [PMID: 33953715 PMCID: PMC8091963 DOI: 10.3389/fimmu.2021.644294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/16/2021] [Indexed: 01/02/2023] Open
Abstract
The basal ganglia network is represented by an interconnected group of subcortical nuclei traditionally thought to play a crucial role in motor learning and movement execution. During the last decades, knowledge about basal ganglia physiology significantly evolved and this network is now considered as a key regulator of important cognitive and emotional processes. Accordingly, the disruption of basal ganglia network dynamics represents a crucial pathogenic factor in many neurological and psychiatric disorders. The striatum is the input station of the circuit. Thanks to the synaptic properties of striatal medium spiny neurons (MSNs) and their ability to express synaptic plasticity, the striatum exerts a fundamental integrative and filtering role in the basal ganglia network, influencing the functional output of the whole circuit. Although it is currently established that the immune system is able to regulate neuronal transmission and plasticity in specific cortical areas, the role played by immune molecules and immune/glial cells in the modulation of intra-striatal connections and basal ganglia activity still needs to be clarified. In this manuscript, we review the available evidence of immune-based regulation of synaptic activity in the striatum, also discussing how an abnormal immune activation in this region could be involved in the pathogenesis of inflammatory and degenerative central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, Università degli Studi di Perugia, Perugia, Italy
| | | | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, Università degli Studi di Perugia, Perugia, Italy
| | - Paolo Calabresi
- Section of Neurology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
25
|
Toval A, Garrigos D, Kutsenko Y, Popović M, Do-Couto BR, Morales-Delgado N, Tseng KY, Ferran JL. Dopaminergic Modulation of Forced Running Performance in Adolescent Rats: Role of Striatal D1 and Extra-striatal D2 Dopamine Receptors. Mol Neurobiol 2021; 58:1782-1791. [PMID: 33394335 PMCID: PMC7932989 DOI: 10.1007/s12035-020-02252-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/04/2020] [Indexed: 12/24/2022]
Abstract
Improving exercise capacity during adolescence impacts positively on cognitive and motor functions. However, the neural mechanisms contributing to enhance physical performance during this sensitive period remain poorly understood. Such knowledge could help to optimize exercise programs and promote a healthy physical and cognitive development in youth athletes. The central dopamine system is of great interest because of its role in regulating motor behavior through the activation of D1 and D2 receptors. Thus, the aim of the present study is to determine whether D1 or D2 receptor signaling contributes to modulate the exercise capacity during adolescence and if this modulation takes place through the striatum. To test this, we used a rodent model of forced running wheel that we implemented recently to assess the exercise capacity. Briefly, rats were exposed to an 8-day period of habituation in the running wheel before assessing their locomotor performance in response to an incremental exercise test, in which the speed was gradually increased until exhaustion. We found that systemic administration of D1-like (SCH23390) and/or D2-like (raclopride) receptor antagonists prior to the incremental test reduced the duration of forced running in a dose-dependent manner. Similarly, locomotor activity in the open field was decreased by the dopamine antagonists. Interestingly, this was not the case following intrastriatal infusion of an effective dose of SCH23390, which decreased motor performance during the incremental test without disrupting the behavioral response in the open field. Surprisingly, intrastriatal delivery of raclopride failed to impact the duration of forced running. Altogether, these results indicate that the level of locomotor response to incremental loads of forced running in adolescent rats is dopamine dependent and mechanistically linked to the activation of striatal D1 and extra-striatal D2 receptors.
Collapse
Affiliation(s)
- Angel Toval
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia - IMIB, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Daniel Garrigos
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia - IMIB, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Yevheniy Kutsenko
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia - IMIB, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Miroljub Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia - IMIB, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Bruno Ribeiro Do-Couto
- Department of Human Anatomy and Psychobiology, Faculty of Psychology, University of Murcia, Murcia, Spain
| | - Nicanor Morales-Delgado
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain
- Institute of Biomedical Research of Murcia - IMIB, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
- Department of Histology and Anatomy, Faculty of Medicine, University Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.
- Institute of Biomedical Research of Murcia - IMIB, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.
| |
Collapse
|
26
|
Diekhof EK, Richter A, Brodmann K, Gruber O. Dopamine multilocus genetic profiles predict sex differences in reactivity of the human reward system. Brain Struct Funct 2021; 226:1099-1114. [PMID: 33580321 DOI: 10.1007/s00429-021-02227-6] [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: 11/20/2019] [Accepted: 01/21/2021] [Indexed: 11/30/2022]
Abstract
Sex differences in the neural processing of decision-making are of high interest as they may have pronounced effects on reward- and addiction-related processes. In these, the neurotransmitter dopamine plays a central role by modulating the responsiveness of the reward circuitry. The present functional magnetic resonance imaging study aimed to explore sex and dopamine transmission interactions in decision-making. 172 subjects (111 women) performed a behavioral self-control task assessing reward-related activation during acceptance and rejection of conditioned rewards. Participants were genotyped for six key genetic polymorphisms in the dopamine system that have previously been associated with individual differences in reward sensitivity or dopaminergic transmission in the human striatum, such as rs7118900 (dopamine receptor D2 (DRD2) Taq1A), rs1554929 (DRD2 C957T), rs907094 (DARPP-32), rs12364283 (DRD2), rs6278 (DRD2), and rs107656 (DRD2). The selected polymorphisms were combined in a so-called multilocus genetic composite (MGC) score reflecting the additive effect of different alleles conferring relative increased dopamine transmission in every individual. We successfully demonstrated that reward-related activation in the ventral striatum and ventral tegmental area (VTA) was significantly modulated by biologically informed MGC profiles and sex. When comparing men and women with low MGC profiles that may indicate lower dopamine transmission, only women displayed a reduced down-regulation of activation in the mesolimbic system during reward rejection and additionally, a significant non-linear u-shape relationship between MGC score and VTA activation. Taken together, by integrating neuroimaging and genetics, the present findings contribute to a better understanding of the effects of sex differences on the human brain.
Collapse
Affiliation(s)
- Esther K Diekhof
- Section for Neuroendocrinology, Department of Biology, University of Hamburg, Hamburg, Germany.,Center for Translational Research in Systems Neuroscience and Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Anja Richter
- Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University Hospital, Heidelberg, Germany. .,Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, SE5 8AF, UK.
| | - Katja Brodmann
- Center for Translational Research in Systems Neuroscience and Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Oliver Gruber
- Center for Translational Research in Systems Neuroscience and Psychiatry, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.,Section for Experimental Psychopathology and Neuroimaging, Department of General Psychiatry, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
27
|
Menniti FS, Chappie TA, Schmidt CJ. PDE10A Inhibitors-Clinical Failure or Window Into Antipsychotic Drug Action? Front Neurosci 2021; 14:600178. [PMID: 33551724 PMCID: PMC7855852 DOI: 10.3389/fnins.2020.600178] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/21/2020] [Indexed: 01/21/2023] Open
Abstract
PDE10A, a phosphodiesterase that inactivates both cAMP and cGMP, is a unique signaling molecule in being highly and nearly exclusively expressed in striatal medium spiny neurons. These neurons dynamically integrate cortical information with dopamine-signaled value to mediate action selection among available behavioral options. Medium spiny neurons are components of either the direct or indirect striatal output pathways. Selective activation of indirect pathway medium spiny neurons by dopamine D2 receptor antagonists is putatively a key element in the mechanism of their antipsychotic efficacy. While PDE10A is expressed in all medium spiny neurons, studies in rodents indicated that PDE10A inhibition has behavioral effects in several key assays that phenocopy dopamine D2 receptor inhibition. This finding gave rise to the hypothesis that PDE10A inhibition also preferentially activates indirect pathway medium spiny neurons, a hypothesis that is consistent with electrophysiological, neurochemical, and molecular effects of PDE10A inhibitors. These data underwrote industry-wide efforts to investigate and develop PDE10A inhibitors as novel antipsychotics. Disappointingly, PDE10A inhibitors from 3 companies failed to evidence antipsychotic activity in patients with schizophrenia to the same extent as standard-of-care D2 antagonists. Given the notable similarities between PDE10A inhibitors and D2 antagonists, gaining an understanding of why only the latter class is antipsychotic affords a unique window into the basis for this therapeutic efficacy. With this in mind, we review the data on PDE10A inhibition as a step toward back-translating the limited antipsychotic efficacy of PDE10A inhibitors, hopefully to inform new efforts to develop better therapeutics to treat psychosis and schizophrenia.
Collapse
Affiliation(s)
- Frank S Menniti
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
| | - Thomas A Chappie
- Internal Medicine Medicinal Chemistry, Pfizer Worldwide Research and Development, Cambridge, MA, United States
| | - Christopher J Schmidt
- Pfizer Innovation and Research Lab Unit, Pfizer Worldwide Research and Development, Cambridge, MA, United States
| |
Collapse
|
28
|
Abstract
DARPP-32 (dopamine- and cAMP-regulated phosphoprotein with an apparent Mr of 32,000), now also known as phosphoprotein phosphatase 1 regulatory subunit 1B (PPP1R1B), is a potent inhibitor of protein phosphatase 1 (PP1, also known as PPP1) when phosphorylated at Thr34 by cAMP-dependent protein kinase (PKA). DARPP-32 exhibits a remarkable regional distribution in brain, roughly similar to that of dopamine innervation. Its discovery was a culmination of the long-standing effort of Paul Greengard to understand the mechanisms through which neurotransmitters such as dopamine exert their effects on target neurons. DARPP-32 is particularly enriched in striatal projection neurons where it is regulated by numerous signals through which it integrates and amplifies responses to many stimuli. Molecular studies of DARPP-32 have revealed that its regulation and function are more complex than anticipated. It is phosphorylated on multiple sites by several protein kinases that modulate DARPP-32 properties. Primarily, when phosphorylated at Thr34 DARPP-32 is a potent inhibitor of PP1, whereas when phosphorylated at Thr75 by Cdk5 it inhibits PKA. Phosphorylation at serine residues by CK1 and CK2 modulates its intracellular localization and its sensitivity to kinases or phosphatases. Modeling studies provide evidence that the signaling pathways including DARPP-32 are endowed of strong robustness and bistable properties favoring switch-like responses. Thus DARPP-32 combined with a set of other distinct signaling molecules enriched in striatal projection neurons plays a key role in the characteristic properties and physiological function of these neurons.
Collapse
|
29
|
Platholi J, Hemmings HC. Modulation of dendritic spines by protein phosphatase-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:117-144. [PMID: 33706930 DOI: 10.1016/bs.apha.2020.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Protein phosphatase-1 (PP-1), a highly conserved multifunctional serine/threonine phosphatase, is enriched in dendritic spines where it plays a major role in modulating excitatory synaptic activity. In addition to established functions in spine maturation and development, multi-subunit holoenzyme forms of PP-1 modulate higher-order cognitive functions such learning and memory. Mechanisms involved in regulating PP-1 activity and localization in spines include interactions with neurabin and spinophilin, structurally related synaptic scaffolding proteins associated with the actin cytoskeleton. Since PP-1 is a critical element in synaptic development, signaling, and plasticity, alterations in PP-1 signaling in dendritic spines are implicated in various neurological and psychiatric disorders. The effects of PP-1 depend on its isoform-specific association with regulatory proteins and activation of downstream signaling pathways. Here we review the role of PP-1 and its binding proteins neurabin and spinophilin in both developing and established dendritic spines, as well as some of the disorders that result from its dysregulation.
Collapse
Affiliation(s)
- Jimcy Platholi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
30
|
Ketamine disrupted storage but not retrieval of information in male rats and apomorphine counteracted its impairing effect. Neurosci Lett 2020; 737:135321. [PMID: 32846219 DOI: 10.1016/j.neulet.2020.135321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/04/2020] [Accepted: 08/20/2020] [Indexed: 11/20/2022]
Abstract
Ketamine, a non-competitive NMDA receptor antagonist, has been reported to mimic the cognitive symptoms of schizophrenia in animals. It has been reported to produce learning and memory deficits in rodents. However, there have limited number of reports that investigated the specific components of memory process that are affected with ketamine. In the present study, we investigated the effects of ketamine [8 and 20 mg/kg, intraperitoneally, (i.p.)] on storage and retrieval of information in rats using an object recognition test. We examined also whether a low dose range of the D1/D2 dopamine receptor agonist apomorphine (0.05 and 0.1 mg/kg, i.p.) would counteract the effects of ketamine. The results show that ketamine dose-dependently impaired storage of information while it did not affect rats' retrieval abilities. Administration of apomorphine reversed the ketamine-induced performance deficits in the ORT. The current findings show a differential modulation of post-training memory components (storage and retrieval of information) by ketamine and suggest a functional interaction between dopamine and NMDA receptors in the control of memory storage which may be of relevance to cognitive deficits a core feature of schizophrenia.
Collapse
|
31
|
Jones-Tabah J, Mohammad H, Hadj-Youssef S, Kim LEH, Martin RD, Benaliouad F, Tanny JC, Clarke PBS, Hébert TE. Dopamine D1 receptor signalling in dyskinetic Parkinsonian rats revealed by fiber photometry using FRET-based biosensors. Sci Rep 2020; 10:14426. [PMID: 32879346 PMCID: PMC7468292 DOI: 10.1038/s41598-020-71121-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
As with many G protein-coupled receptors (GPCRs), the signalling pathways regulated by the dopamine D1 receptor (D1R) are dynamic, cell type-specific, and can change in the face of disease or drug exposures. In striatal neurons, the D1R activates cAMP/protein kinase A (PKA) signalling. However, in Parkinson's disease (PD), alterations in this pathway lead to functional upregulation of extracellular regulated kinases 1/2 (ERK1/2), contributing to L-DOPA-induced dyskinesia (LID). In order to detect D1R activation in vivo and to study the progressive dysregulation of D1R signalling in PD and LID, we developed ratiometric fiber-photometry with Förster resonance energy transfer (FRET) biosensors and optically detected PKA and ERK1/2 signalling in freely moving rats. We show that in Parkinsonian animals, D1R signalling through PKA and ERK1/2 is sensitized, but that following chronic treatment with L-DOPA, these pathways become partially desensitized while concurrently D1R activation leads to greater induction of dyskinesia.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Hanan Mohammad
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Shadi Hadj-Youssef
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Lucy E H Kim
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Faïza Benaliouad
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada.
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Room 1325, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
32
|
Calabrese V, Di Maio A, Marino G, Cardinale A, Natale G, De Rosa A, Campanelli F, Mancini M, Napolitano F, Avallone L, Calabresi P, Usiello A, Ghiglieri V, Picconi B. Rapamycin, by Inhibiting mTORC1 Signaling, Prevents the Loss of Striatal Bidirectional Synaptic Plasticity in a Rat Model of L-DOPA-Induced Dyskinesia. Front Aging Neurosci 2020; 12:230. [PMID: 32848709 PMCID: PMC7431470 DOI: 10.3389/fnagi.2020.00230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/30/2020] [Indexed: 12/16/2022] Open
Abstract
Levodopa (L-DOPA) treatment is the main gold-standard therapy for Parkinson disease (PD). Besides good antiparkinsonian effects, prolonged use of this drug is associated to the development of involuntary movements known as L-DOPA-induced dyskinesia (LID). L-DOPA-induced dyskinesia is linked to a sensitization of dopamine (DA) D1 receptors located on spiny projection neurons (SPNs) of the dorsal striatum. Several evidences have shown that the emergence of LID can be related to striatal D1/cAMP/PKA/DARPP-32 and extracellular signal-regulated kinases (ERK1/2) pathway overactivation associated to aberrant N-methyl-d-aspartate (NMDA) receptor function. In addition, within striatum, ERK1/2 is also able to modulate in a D1 receptor-dependent manner the activity of the mammalian target of rapamycin complex 1 (mTORC1) pathway under DA depletion and L-DOPA therapy. Consistently, increased mTORC1 signaling appears during chronic administration of L-DOPA and shows a high correlation with the severity of dyskinesia. Furthermore, the abnormal activation of the D1/PKA/DARPP-32 cascade is paralleled by increased phosphorylation of the GluA1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor at the PKA Ser845 site. The GluA1 promotes excitatory AMPA receptor-mediated transmission and may be implicated in the alterations found at the corticostriatal synapses of dyskinetic animals. In our study, we investigated the role of mTORC1 pathway activation in modulating bidirectional striatal synaptic plasticity in L-DOPA-treated parkinsonian rats. Inhibition of mTORC1 by coadministration of rapamycin to L-DOPA was able to limit the magnitude of LID expression, accounting for a therapeutic effect of this drug. In particular, behavioral data showed that, in L-DOPA-treated rats, rapamycin administration induced a selective decrease of distinct components of abnormal involuntary movements (i.e., axial and orolingual dyskinesia). Furthermore, ex vivo patch clamp and intracellular recordings of SPNs revealed that pharmacological inhibition of mTORC1 also resulted associated with a physiological bidirectional plasticity, when compared to dyskinetic rats treated with L-DOPA alone. This study uncovers the important role of mTORC1 inhibition to prevent the loss of striatal bidirectional plasticity under chronic L-DOPA treatment in rodent models of PD.
Collapse
Affiliation(s)
- Valeria Calabrese
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Gioia Marino
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Antonella Cardinale
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Department of Medicine, University of Perugia, Perugia, Italy
| | - Giuseppina Natale
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Arianna De Rosa
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Federica Campanelli
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Mancini
- Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Francesco Napolitano
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Paolo Calabresi
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DISTABIF), University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Veronica Ghiglieri
- Department of Medicine, University of Perugia, Perugia, Italy.,Laboratory of Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Barbara Picconi
- Laboratory of Experimental Neurophysiology, IRCCS San Raffaele Pisana, Rome, Italy.,Università Telematica San Raffaele, Rome, Italy
| |
Collapse
|
33
|
Castela I, Hernandez LF. Shedding light on dyskinesias. Eur J Neurosci 2020; 53:2398-2413. [DOI: 10.1111/ejn.14777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Ivan Castela
- HM‐CINAC Hospital Universitario HM Puerta del Sur Fundación de Investigación HM Hospitales Madrid Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED) Carlos III Health Institute Madrid Spain
| | - Ledia F. Hernandez
- HM‐CINAC Hospital Universitario HM Puerta del Sur Fundación de Investigación HM Hospitales Madrid Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED) Carlos III Health Institute Madrid Spain
| |
Collapse
|
34
|
Chakroun K, Mathar D, Wiehler A, Ganzer F, Peters J. Dopaminergic modulation of the exploration/exploitation trade-off in human decision-making. eLife 2020; 9:e51260. [PMID: 32484779 PMCID: PMC7266623 DOI: 10.7554/elife.51260] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/01/2020] [Indexed: 01/15/2023] Open
Abstract
Involvement of dopamine in regulating exploration during decision-making has long been hypothesized, but direct causal evidence in humans is still lacking. Here, we use a combination of computational modeling, pharmacological intervention and functional magnetic resonance imaging to address this issue. Thirty-one healthy male participants performed a restless four-armed bandit task in a within-subjects design under three drug conditions: 150 mg of the dopamine precursor L-dopa, 2 mg of the D2 receptor antagonist haloperidol, and placebo. Choices were best explained by an extension of an established Bayesian learning model accounting for perseveration, directed exploration and random exploration. Modeling revealed attenuated directed exploration under L-dopa, while neural signatures of exploration, exploitation and prediction error were unaffected. Instead, L-dopa attenuated neural representations of overall uncertainty in insula and dorsal anterior cingulate cortex. Our results highlight the computational role of these regions in exploration and suggest that dopamine modulates how this circuit tracks accumulating uncertainty during decision-making.
Collapse
Affiliation(s)
- Karima Chakroun
- Department of Systems Neuroscience, University Medical Center Hamburg-EppendorfHamburgGermany
| | - David Mathar
- Department of Psychology, Biological Psychology, University of CologneCologneGermany
| | - Antonius Wiehler
- Department of Systems Neuroscience, University Medical Center Hamburg-EppendorfHamburgGermany
- Institut du Cerveau et de la Moelle épinière - ICM, Centre de NeuroImagerie de Recherche - CENIR, Sorbonne Universités, Groupe Hospitalier Pitié-SalpêtrièreParisFrance
| | - Florian Ganzer
- German Center for Addiction Research in Childhood and Adolescence, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Jan Peters
- Department of Systems Neuroscience, University Medical Center Hamburg-EppendorfHamburgGermany
- Department of Psychology, Biological Psychology, University of CologneCologneGermany
| |
Collapse
|
35
|
Xiao Y, Luo H, Yang WZ, Zeng Y, Shen Y, Ni X, Shi Z, Zhong J, Liang Z, Fu X, Tu H, Sun W, Shen WL, Hu J, Yang J. A Brain Signaling Framework for Stress-Induced Depression and Ketamine Treatment Elucidated by Phosphoproteomics. Front Cell Neurosci 2020; 14:48. [PMID: 32317933 PMCID: PMC7156020 DOI: 10.3389/fncel.2020.00048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/20/2020] [Indexed: 12/25/2022] Open
Abstract
Depression is a common affective disorder characterized by significant and persistent low mood. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, is reported to have a rapid and durable antidepressant effect, but the mechanisms are unclear. Protein phosphorylation is a post-translational modification that plays a crucial role in cell signaling. Thus, we present a phosphoproteomics approach to investigate the mechanisms underlying stress-induced depression and the rapid antidepressant effect of ketamine in mice. We analyzed the phosphoprotein changes induced by chronic unpredictable mild stress (CUMS) and ketamine treatment in two known mood control centers, the medial prefrontal cortex (mPFC) and the nucleus accumbens (NAc). We initially obtained >8,000 phosphorylation sites. Quantitation revealed 3,988 sites from the mPFC and 3,196 sites from the NAc. Further analysis revealed that changes in synaptic transmission-related signaling are a common feature. Notably, CUMS-induced changes were reversed by ketamine treatment, as shown by the analysis of commonly altered sites. Ketamine also induced specific changes, such as alterations in synapse organization, synaptic transmission, and enzyme binding. Collectively, our findings establish a signaling framework for stress-induced depression and the rapid antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huoqing Luo
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wen Z Yang
- Shanghai Institute for Advanced Immunochemical Studies & School of Life Science and Technology, Shanghaitech University, Shanghai, China.,CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yeting Zeng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yinbo Shen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xinyan Ni
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Zhaomei Shi
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Jun Zhong
- Delta Omics Inc., Baltimore, MD, United States
| | - Ziqi Liang
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Xiaoyu Fu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongqing Tu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenzhi Sun
- Chinese Institute For Brain Research, Beijing, China
| | - Wei L Shen
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Ji Hu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Jiajun Yang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
36
|
Han QW, Yuan YH, Chen NH. The therapeutic role of cannabinoid receptors and its agonists or antagonists in Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109745. [PMID: 31442553 DOI: 10.1016/j.pnpbp.2019.109745] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease and its characteristic is the progressive degeneration of dopaminergic neurons within the substantia nigra (SN) of the midbrain. There is hardly any clinically proven efficient therapeutics for its cure in several recent preclinical advances proposed to treat PD. Recent studies have found that the endocannabinoid signaling system in particular the comprised two receptors, CB1 and CB2 receptors, has a significant regulatory function in basal ganglia and is involved in the pathogenesis of PD. Therefore, adding new insights into the biochemical interactions between cannabinoids and other signaling pathways may help develop new pharmacological strategies. Factors of the endocannabinoid system (ECS) are abundantly expressed in the neural circuits of basal ganglia, where they interact interactively with glutamatergic, γ-aminobutyric acid-ergic (GABAergic), and dopaminergic signaling systems. Although preclinical studies on PD are promising, the use of cannabinoids at the clinical level has not been thoroughly studied. In this review, we evaluated the available evidence and reviewed the involvement of ECS in etiologies, symptoms and treatments related to PD. Since CB1 and CB2 receptors are the two main receptors of endocannabinoids, we primarily put the focus on the therapeutic role of CB1 and CB2 receptors in PD. We will try to determine future research clues that will help understand the potential therapeutic benefits of the ECS in the treatment of PD, aiming to open up new strategies and ideas for the treatment of PD.
Collapse
Affiliation(s)
- Qi-Wen Han
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
37
|
Modified Glutamatergic Postsynapse in Neurodegenerative Disorders. Neuroscience 2019; 454:116-139. [PMID: 31887357 DOI: 10.1016/j.neuroscience.2019.12.002] [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: 06/10/2019] [Revised: 10/02/2019] [Accepted: 12/02/2019] [Indexed: 01/27/2023]
Abstract
The postsynaptic density (PSD) is a complex subcellular domain important for postsynaptic signaling, function, and plasticity. The PSD is present at excitatory synapses and specialized to allow for precise neuron-to-neuron transmission of information. The PSD is localized immediately underneath the postsynaptic membrane forming a major protein network that regulates postsynaptic signaling and synaptic plasticity. Glutamatergic synaptic dysfunction affecting PSD morphology and signaling events have been described in many neurodegenerative disorders, either sporadic or familial forms. Thus, in this review we describe the main protein players forming the PSD and their activity, as well as relevant modifications in key components of the postsynaptic architecture occurring in Huntington's, Parkinson's and Alzheimer's diseases.
Collapse
|
38
|
Li W, Pozzo-Miller L. Dysfunction of the corticostriatal pathway in autism spectrum disorders. J Neurosci Res 2019; 98:2130-2147. [PMID: 31758607 DOI: 10.1002/jnr.24560] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
The corticostriatal pathway that carries sensory, motor, and limbic information to the striatum plays a critical role in motor control, action selection, and reward. Dysfunction of this pathway is associated with many neurological and psychiatric disorders. Corticostriatal synapses have unique features in their cortical origins and striatal targets. In this review, we first describe axonal growth and synaptogenesis in the corticostriatal pathway during development, and then summarize the current understanding of the molecular bases of synaptic transmission and plasticity at mature corticostriatal synapses. Genes associated with autism spectrum disorder (ASD) have been implicated in axonal growth abnormalities, imbalance of the synaptic excitation/inhibition ratio, and altered long-term synaptic plasticity in the corticostriatal pathway. Here, we review a number of ASD-associated high-confidence genes, including FMR1, KMT2A, GRIN2B, SCN2A, NLGN1, NLGN3, MET, CNTNAP2, FOXP2, TSHZ3, SHANK3, PTEN, CHD8, MECP2, DYRK1A, RELN, FOXP1, SYNGAP1, and NRXN, and discuss their relevance to proper corticostriatal function.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
39
|
Neuromodulators and Long-Term Synaptic Plasticity in Learning and Memory: A Steered-Glutamatergic Perspective. Brain Sci 2019; 9:brainsci9110300. [PMID: 31683595 PMCID: PMC6896105 DOI: 10.3390/brainsci9110300] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/24/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The molecular pathways underlying the induction and maintenance of long-term synaptic plasticity have been extensively investigated revealing various mechanisms by which neurons control their synaptic strength. The dynamic nature of neuronal connections combined with plasticity-mediated long-lasting structural and functional alterations provide valuable insights into neuronal encoding processes as molecular substrates of not only learning and memory but potentially other sensory, motor and behavioural functions that reflect previous experience. However, one key element receiving little attention in the study of synaptic plasticity is the role of neuromodulators, which are known to orchestrate neuronal activity on brain-wide, network and synaptic scales. We aim to review current evidence on the mechanisms by which certain modulators, namely dopamine, acetylcholine, noradrenaline and serotonin, control synaptic plasticity induction through corresponding metabotropic receptors in a pathway-specific manner. Lastly, we propose that neuromodulators control plasticity outcomes through steering glutamatergic transmission, thereby gating its induction and maintenance.
Collapse
|
40
|
Mantas I, Yang Y, Mannoury-la-Cour C, Millan MJ, Zhang X, Svenningsson P. Genetic deletion of GPR88 enhances the locomotor response to L-DOPA in experimental parkinsonism while counteracting the induction of dyskinesia. Neuropharmacology 2019; 162:107829. [PMID: 31666199 DOI: 10.1016/j.neuropharm.2019.107829] [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: 08/24/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 11/27/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive loss of midbrain dopaminergic neurons and treated with the dopamine precursor, 3,4-dihydroxy-l-phenylalanine (L-DOPA). Prolonged L-DOPA treatment is however associated with waning efficacy and the induction of L-DOPA induced dyskinesia (LID). GPR88 is an orphan G-protein Coupled Receptor (GPCR) expressed in dopaminoceptive striatal medium spiny neurons (MSNs) and their afferent corticostriatal glutamatergic neurons. Here, we studied the role of GPR88 in experimental parkinsonism and LID. Chronic L-DOPA administration to male GPR88 KO mice, subjected to unilateral 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle, resulted in more rotations than in their WT counterparts. Conversely, GPR88 KO mice had a lower abnormal involuntary movements (AIMs) score. These behavioral responses were accompanied by altered transcription of L-DOPA upregulated genes in lesioned GPR88 KO compared to WT striata. In accordance with a role for serotonin neurons in LID development, WT but not GPR88 KO striata exhibited 5-hydroxytryptamine displacement upon repeated L-DOPA treatment. Intact male GPR88 KO mice showed diminished tacrine-induced PD-like tremor and spontaneous hyperlocomotion. Dopamine and its metabolites were not increased in male GPR88 KO mice, but biosensor recordings revealed increased spontaneous/basal and evoked glutamate release in striata of male GPR88 KO mice. In conclusion, genetic deletion of GPR88 promotes l-DOPA-induced rotation and spontaneous locomotion yet suppresses the induction of LIDs and also reduces tremor. These data provide behavioral, neurochemical and molecular support that GPR88 antagonism may favour motor relief in PD patients without aggravating the induction of motor side effects.
Collapse
Affiliation(s)
- Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Yunting Yang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Clotilde Mannoury-la-Cour
- Centre for Therapeutic Innovation-CNS, Institut de Recherches Servier, Centre de Recherches de Croissy, Paris, France
| | - Mark J Millan
- Centre for Therapeutic Innovation-CNS, Institut de Recherches Servier, Centre de Recherches de Croissy, Paris, France
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
Li KD, Yan K, Wang QS, Tian JS, Xu D, Zhang WY, Cui YL. Antidepressant-like effects of dietary gardenia blue pigment derived from genipin and tyrosine. Food Funct 2019; 10:4533-4545. [PMID: 31264676 DOI: 10.1039/c9fo00480g] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Gardenia blue pigments derived from genipin reacting with amino acids have been used as natural food colorants for nearly 30 years in East Asia. However, their pharmacological effects, especially antidepressant-like effects, have not been reported so far. In this study, one of the gardenia blue pigments, was obtained from the reaction of genipin with tyrosine (genipin-tyrosine derivant (GTD)), and its antidepressant-like effects were investigated in lipopolysaccharide (LPS) or chronic unpredictable mild stress (CUMS) models. The results showed that GTD could attenuate depressive-like behaviors in both animal models. GTD reversed the LPS-induced cytokine increase of TNF-α, IL-6, and corticosterone (CORT) in mice plasma and hippocampus. In CUMS rats, GTD treatment significantly reduced hypothalamic-pituitary-adrenal (HPA) axis-related stress hormone levels in plasma including those of CORT, adrenocorticotropic hormone (ACTH), and corticotropin-releasing hormone (CRH). Besides, GTD increased plasma testosterone and hippocampal brain-derived neurotrophic factor (BDNF) levels in CUMS rats. GTD increased serotonin (5-HT), dopamine (DA), and norepinephrine (NE) in rat hippocampus and corpus striatum. Consistently, hippocampal metabolomic analysis demonstrated that GTD restored monoamine neurotransmitter metabolism, mitochondrial oxidative function, and membrane structural integrity. Our data suggested that GTD produced antidepressant-like activity through the restoration of the HPA axis hormone balance and the regulation of neurotransmitter release.
Collapse
Affiliation(s)
- Kuang-Dai Li
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | | | | | | | | | | | | |
Collapse
|
42
|
The Beneficial Effect of Acute Exercise on Motor Memory Consolidation is Modulated by Dopaminergic Gene Profile. J Clin Med 2019; 8:jcm8050578. [PMID: 31035583 PMCID: PMC6572639 DOI: 10.3390/jcm8050578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/21/2019] [Accepted: 04/22/2019] [Indexed: 12/31/2022] Open
Abstract
When aerobic exercise is performed following skilled motor practice, it can enhance motor memory consolidation. Previous studies have suggested that dopamine may play a role in motor memory consolidation, but whether it is involved in the exercise effects on consolidation is unknown. Hence, we aimed to investigate the influence of dopaminergic pathways on the exercise-induced modulation of motor memory consolidation. We compared the effect of acute exercise on motor memory consolidation between the genotypes that are known to affect dopaminergic transmission and learning. By combining cluster analyses and fitting linear models with and without included polymorphisms, we provide preliminary evidence that exercise benefits the carriers of alleles that are associated with low synaptic dopamine content. In line with previous reports, our findings implicate dopamine as a modulator of the exercise-induced effects on motor memory consolidation, and suggest exercise as a potential clinical tool to counteract low endogenous dopamine bioavailability. Further experiments are needed to establish causal relations.
Collapse
|
43
|
Oliveira S, Oliveira M, Hipolide D. A1 adenosine receptors in the striatum play a role in the memory impairment caused by sleep deprivation through downregulation of the PKA pathway. Neurobiol Learn Mem 2019; 160:91-97. [DOI: 10.1016/j.nlm.2018.03.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/08/2018] [Accepted: 03/30/2018] [Indexed: 02/04/2023]
|
44
|
Mai HN, Sharma N, Jeong JH, Shin EJ, Pham DT, Trinh QD, Lee YJ, Jang CG, Nah SY, Bing G, Kim HC. P53 knockout mice are protected from cocaine-induced kindling behaviors via inhibiting mitochondrial oxidative burdens, mitochondrial dysfunction, and proapoptotic changes. Neurochem Int 2019; 124:68-81. [DOI: 10.1016/j.neuint.2018.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 11/30/2022]
|
45
|
Alsiö J, Phillips BU, Sala-Bayo J, Nilsson SRO, Calafat-Pla TC, Rizwand A, Plumbridge JM, López-Cruz L, Dalley JW, Cardinal RN, Mar AC, Robbins TW. Dopamine D2-like receptor stimulation blocks negative feedback in visual and spatial reversal learning in the rat: behavioural and computational evidence. Psychopharmacology (Berl) 2019; 236:2307-2323. [PMID: 31218428 PMCID: PMC6695374 DOI: 10.1007/s00213-019-05296-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/02/2019] [Indexed: 02/02/2023]
Abstract
RATIONALE Dopamine D2-like receptors (D2R) are important drug targets in schizophrenia and Parkinson's disease, but D2R ligands also cause cognitive inflexibility such as poor reversal learning. The specific role of D2R in reversal learning remains unclear. OBJECTIVES We tested the hypotheses that D2R agonism impairs reversal learning by blocking negative feedback and that antagonism of D1-like receptors (D1R) impairs learning from positive feedback. METHODS Male Lister Hooded rats were trained on a novel visual reversal learning task. Performance on "probe trials", during which the correct or incorrect stimulus was presented with a third, probabilistically rewarded (50% of trials) and therefore intermediate stimulus, revealed individual learning curves for the processes of positive and negative feedback. The effects of D2R and D1R agonists and antagonists were evaluated. A separate cohort was tested on a spatial probabilistic reversal learning (PRL) task after D2R agonism. Computational reinforcement learning modelling was applied to choice data from the PRL task to evaluate the contribution of latent factors. RESULTS D2R agonism with quinpirole dose-dependently impaired both visual reversal and PRL. Analysis of the probe trials on the visual task revealed a complete blockade of learning from negative feedback at the 0.25 mg/kg dose, while learning from positive feedback was intact. Estimated parameters from the model that best described the PRL choice data revealed a steep and selective decrease in learning rate from losses. D1R antagonism had a transient effect on the positive probe trials. CONCLUSIONS D2R stimulation impairs reversal learning by blocking the impact of negative feedback.
Collapse
Affiliation(s)
- Johan Alsiö
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
| | - Benjamin U Phillips
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Júlia Sala-Bayo
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Simon R O Nilsson
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Neuroscience Institute, New York University Medical Center, New York, NY, USA
- Department of Neuroscience and Physiology, School of Medicine, New York University, New York, NY, USA
| | - Teresa C Calafat-Pla
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Arazo Rizwand
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Jessica M Plumbridge
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Laura López-Cruz
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Jeffrey W Dalley
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Rudolf N Cardinal
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire & Peterborough NHS Foundation Trust, Box 190 (Liaison Psychiatry), Cambridge Biomedical Campus, Cambridge, UK
| | - Adam C Mar
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Neuroscience Institute, New York University Medical Center, New York, NY, USA
- Department of Neuroscience and Physiology, School of Medicine, New York University, New York, NY, USA
| | - Trevor W Robbins
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
46
|
Mahmoudi M, Pakpour S, Perry G. Drug-Abuse Nanotechnology: Opportunities and Challenges. ACS Chem Neurosci 2018; 9:2288-2298. [PMID: 29851334 DOI: 10.1021/acschemneuro.8b00127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Opioid drug abuse and dependence/addiction are complex disorders regulated by a wide range of interacting networks of genes and pathways that control a variety of phenotypes. Although the field has been extensively progressed since the birth of the National Institute on Drug Abuse in 1974, the fundamental knowledge and involved mechanisms that lead to drug dependence/addiction are poorly understood, and thus, there has been limited success in the prevention of drug addiction and development of therapeutics for definitive treatment and cure of addiction disease. The lack of success in both identification of addiction in at-risk populations and the development of efficient drugs has resulted in a serious social and economic burden from opioid drug abuse with global increasing rate of mortality from drug overdoses. This perspective aims to draw the attention of scientists to the potential role of nanotechnologies, which might pave the way for the development of more practical platforms for either drug development or identification and screening of patients who may be vulnerable to addiction after using opioid drugs.
Collapse
Affiliation(s)
- Morteza Mahmoudi
- Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sepideh Pakpour
- Infectious Disease & Microbiome, Broad Institute, Cambridge, Massachusetts 02142, United States
- School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - George Perry
- Neurosciences Institute and Department of Biology, College of Sciences, University of Texas, San Antonio, Texas 78249, United States
| |
Collapse
|
47
|
Cell type-specific activation of mitogen-activated protein kinase in D1 receptor-expressing neurons of the nucleus accumbens potentiates stimulus-reward learning in mice. Sci Rep 2018; 8:14413. [PMID: 30258218 PMCID: PMC6158283 DOI: 10.1038/s41598-018-32840-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 09/12/2018] [Indexed: 01/11/2023] Open
Abstract
Medium spiny neurons (MSN) in the nucleus accumbens (NAc) are a fundamental component of various aspects of motivated behavior. Although mitogen-activated protein kinase (MAPK) signaling plays a crucial role in several types of learning, the cell type-specific role of MAPK pathway in stimulus-reward learning and motivation remains unclear. We herein investigated the role of MAPK in accumbal MSNs in reward-associated learning and memory. During the acquisition of Pavlovian conditioning, the number of phosphorylated MAPK1/3-positive cells was increased significantly and exclusively in the NAc core by 7-days of extensive training. MAPK signaling in the respective D1R- and D2R-MSNs was manipulated by transfecting an adeno-associated virus (AAV) plasmid into the NAc of Drd1a-Cre and Drd2-Cre transgenic mice. Potentiation of MAPK signaling shifted the learning curve of Pavlovian conditioning to the left only in Drd1a-Cre mice, whereas such manipulation in D2R-MSNs had negligible effects. In contrast, MAPK manipulation in D2R-MSNs of the NAc core significantly increased motivation for food rewards as found in Drd1a-Cre mice. These results suggest that MAPK signaling in the D1R-MSNs of NAc core plays an important role in stimulus-reward learning, while MAPK signaling in both D1R- and D2R-MSNs is involved in motivation for natural rewards.
Collapse
|
48
|
Liu JJ, Sharma K, Zangrandi L, Chen C, Humphrey SJ, Chiu YT, Spetea M, Liu-Chen LY, Schwarzer C, Mann M. In vivo brain GPCR signaling elucidated by phosphoproteomics. Science 2018; 360:360/6395/eaao4927. [PMID: 29930108 DOI: 10.1126/science.aao4927] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 04/27/2018] [Indexed: 12/12/2022]
Abstract
A systems view of G protein-coupled receptor (GPCR) signaling in its native environment is central to the development of GPCR therapeutics with fewer side effects. Using the kappa opioid receptor (KOR) as a model, we employed high-throughput phosphoproteomics to investigate signaling induced by structurally diverse agonists in five mouse brain regions. Quantification of 50,000 different phosphosites provided a systems view of KOR in vivo signaling, revealing novel mechanisms of drug action. Thus, we discovered enrichment of the mechanistic target of rapamycin (mTOR) pathway by U-50,488H, an agonist causing aversion, which is a typical KOR-mediated side effect. Consequently, mTOR inhibition during KOR activation abolished aversion while preserving beneficial antinociceptive and anticonvulsant effects. Our results establish high-throughput phosphoproteomics as a general strategy to investigate GPCR in vivo signaling, enabling prediction and modulation of behavioral outcomes.
Collapse
Affiliation(s)
- Jeffrey J Liu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Kirti Sharma
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Luca Zangrandi
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Chongguang Chen
- Center for Substance Abuse Research and Department of Pharmacology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Sean J Humphrey
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Yi-Ting Chiu
- Center for Substance Abuse Research and Department of Pharmacology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020 Innsbruck, Austria
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research and Department of Pharmacology, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. .,Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
49
|
Gower A, Tiberi M. The Intersection of Central Dopamine System and Stroke: Potential Avenues Aiming at Enhancement of Motor Recovery. Front Synaptic Neurosci 2018; 10:18. [PMID: 30034335 PMCID: PMC6043669 DOI: 10.3389/fnsyn.2018.00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Dopamine, a major neurotransmitter, plays a role in a wide range of brain sensorimotor functions. Parkinson's disease and schizophrenia are two major human neuropsychiatric disorders typically associated with dysfunctional dopamine activity levels, which can be alleviated through the druggability of the dopaminergic systems. Meanwhile, several studies suggest that optimal brain dopamine activity levels are also significantly impacted in other serious neurological conditions, notably stroke, but this has yet to be fully appreciated at both basic and clinical research levels. This is of utmost importance as there is a need for better treatments to improve recovery from stroke. Here, we discuss the state of knowledge regarding the modulation of dopaminergic systems following stroke, and the use of dopamine boosting therapies in animal stroke models to improve stroke recovery. Indeed, studies in animals and humans show stroke leads to changes in dopamine functioning. Moreover, evidence from animal stroke models suggests stimulation of dopamine receptors may be a promising therapeutic approach for enhancing motor recovery from stroke. With respect to the latter, we discuss the evidence for several possible receptor-linked mechanisms by which improved motor recovery may be mediated. One avenue of particular promise is the subtype-selective stimulation of dopamine receptors in conjunction with physical therapy. However, results from clinical trials so far have been more mixed due to a number of potential reasons including, targeting of the wrong patient populations and use of drugs which modulate a wide array of receptors. Notwithstanding these issues, it is hoped that future research endeavors will assist in the development of more refined dopaminergic therapeutic approaches to enhance stroke recovery.
Collapse
Affiliation(s)
- Annette Gower
- Ottawa Hospital Research Institute (Neuroscience Program), Ottawa, ON, Canada.,University of Ottawa Brain and Mind Institute, Ottawa, ON, Canada.,Departments of Medicine, Cellular and Molecular Medicine, and Psychiatry, University of Ottawa, Ottawa, ON, Canada
| | - Mario Tiberi
- Ottawa Hospital Research Institute (Neuroscience Program), Ottawa, ON, Canada.,University of Ottawa Brain and Mind Institute, Ottawa, ON, Canada.,Departments of Medicine, Cellular and Molecular Medicine, and Psychiatry, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
50
|
Li D, Musante V, Zhou W, Picciotto MR, Nairn AC. Striatin-1 is a B subunit of protein phosphatase PP2A that regulates dendritic arborization and spine development in striatal neurons. J Biol Chem 2018; 293:11179-11194. [PMID: 29802198 DOI: 10.1074/jbc.ra117.001519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/06/2018] [Indexed: 01/09/2023] Open
Abstract
Striatin-1, a subunit of the serine/threonine phosphatase PP2A, is preferentially expressed in neurons in the striatum. As a member of the striatin family of B subunits, striatin-1 is a core component together with PP2A of a multiprotein complex called STRIPAK, the striatin-interacting phosphatase and kinase complex. Little is known about the function of striatin-1 or the STRIPAK complex in the mammalian striatum. Here, we identify a selective role for striatin-1 in striatal neuron maturation. Using a small hairpin RNA (shRNA) knockdown approach in primary striatal neuronal cultures, we determined that reduced expression of striatin-1 results in increased dendritic complexity and an increased density of dendritic spines, classified as stubby spines. The dendritic phenotype was rescued by co-expression of a striatin-1 mutant construct insensitive to the knockdown shRNA but was not rescued by co-expression of PP2A- or Mob3-binding deficient striatin-1 constructs. Reduction of striatin-1 did not result in deficits in neuronal connectivity in this knockdown model, as we observed no abnormalities in synapse formation or in spontaneous excitatory postsynaptic currents. Thus, this study suggests that striatin-1 is a regulator of neuronal development in striatal neurons.
Collapse
Affiliation(s)
- Daniel Li
- From the Department of Psychiatry, Yale University, New Haven, Connecticut 06520
| | - Veronica Musante
- From the Department of Psychiatry, Yale University, New Haven, Connecticut 06520
| | - Wenliang Zhou
- From the Department of Psychiatry, Yale University, New Haven, Connecticut 06520
| | - Marina R Picciotto
- From the Department of Psychiatry, Yale University, New Haven, Connecticut 06520
| | - Angus C Nairn
- From the Department of Psychiatry, Yale University, New Haven, Connecticut 06520
| |
Collapse
|