1
|
Serra M, Marongiu J, Simola N, Costa G. Emission of 50-kHz ultrasonic vocalizations stimulated by antiparkinsonian dopaminomimetic drugs in hemiparkinsonian rats is associated with neuronal activation in subcortical regions that regulate the affective state. Exp Neurol 2024; 381:114939. [PMID: 39191345 DOI: 10.1016/j.expneurol.2024.114939] [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: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/24/2024] [Indexed: 08/29/2024]
Abstract
Dopamine replacement therapy (DRT) of Parkinson's disease (PD) may trigger non-motor complications, some of which affect hedonic homeostatic regulation. Management of iatrogenic alterations in the affective state in PD is unsatisfactory, partly because of the limitations in the experimental models that are used in the preclinical investigation of the neurobiology and therapy of these alterations. In this connection, we recently employed a new experimental approach consisting in measuring the emission of 50-kHz ultrasonic vocalizations (USVs), a marker of positive affect, in hemiparkinsonian rats treated with drugs used in the DRT of PD. To further strengthen our approach, we here evaluated how the acute and repeated (× 5, on alternate days) administration of apomorphine (2 mg/kg, i.p.) or L-3,4-dihydroxyphenilalanine (L-DOPA, 12 mg/kg, i.p.) modified the immunoreactivity for Zif-268, a marker of neuronal activation, in the nucleus accumbens (NAc), caudate-putamen (CPu) and medial prefrontal cortex (mPFC), which are brain regions that regulate emotional states and drugs' affective properties. Acute and repeated treatment with either apomorphine or L-DOPA stimulated the emission of 50-kHz USVs in hemiparkinsonian rats, and this effect was paired with increased Zif-268 immunoreactivity in the NAc and CPu, but not mPFC. These findings indicate that subcortical and cortical regions may differently regulate the emission of 50-kHz USVs in hemiparkinsonian rats treated with dopaminergic drugs used in the DRT of PD. Moreover, they provide further evidence that measuring 50-kHz USV emissions in hemiparkinsonian rats may be a relevant approach to investigate at the preclinical level the affective properties of antiparkinsonian drugs.
Collapse
Affiliation(s)
- Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| | - Giulia Costa
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
2
|
Roman KM, Dinasarapu AR, Cherian S, Fan X, Donsante Y, Aravind N, Chan CS, Jinnah H, Hess EJ. Striatal cell-type-specific molecular signatures reveal therapeutic targets in a model of dystonia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617010. [PMID: 39415987 PMCID: PMC11482807 DOI: 10.1101/2024.10.07.617010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Striatal dysfunction is implicated in many forms of dystonia, including idiopathic, inherited and iatrogenic dystonias. The striatum is comprised largely of GABAergic spiny projection neurons (SPNs) that are defined by their long-range efferents. Direct SPNs (dSPNs) project to the internal globus pallidus/substantia nigra reticulata whereas indirect pathway SPNs (iSPNs) project to the external pallidum; the concerted activity of both SPN subtypes modulates movement. Convergent results from genetic, imaging and physiological studies in patients suggest that abnormalities of both dSPNs and iSPNs contribute to the expression of dystonia, but the molecular adaptations underlying these abnormalities are not known. Here we provide a comprehensive analysis of SPN cell-type-specific molecular signatures in a model of DOPA-responsive dystonia (DRD mice), which is caused by gene defects that reduce dopamine neurotransmission, resulting in dystonia that is specifically associated with striatal dysfunction. Individually profiling the translatome of dSPNs and iSPNs using translating ribosome affinity purification with RNA-seq revealed hundreds of differentially translating mRNAs in each SPN subtype in DRD mice, yet there was little overlap between the dysregulated genes in dSPNs and iSPNs. Despite the paucity of shared adaptations, a disruption in glutamatergic signaling was predicted for both dSPNs and iSPNs. Indeed, we found that both AMPA and NMDA receptor-mediated currents were enhanced in dSPNs but diminished in iSPNs in DRD mice. The pattern of mRNA dysregulation was specific to dystonia as the adaptations in DRD mice were distinct from those in parkinsonian mice where the dopamine deficit occurs in adults, suggesting that the phenotypic outcome is dependent on both the timing of the dopaminergic deficit and the SPN-specific adaptions. We leveraged the unique molecular signatures of dSPNs and iSPNs in DRD mice to identify biochemical mechanisms that may be targets for therapeutics, including LRRK2 inhibition. Administration of the LRRK2 inhibitor MLi-2 ameliorated the dystonia in DRD mice suggesting a novel target for therapeutics and demonstrating that the delineation of cell-type-specific molecular signatures provides a powerful approach to revealing both CNS dysfunction and therapeutic targets in dystonia.
Collapse
Affiliation(s)
- Kaitlyn M. Roman
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - Suraj Cherian
- Department of Neuroscience, Northwestern University, Chicago, Illinois, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Nivetha Aravind
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - C. Savio Chan
- Department of Neuroscience, Northwestern University, Chicago, Illinois, USA
| | - H.A. Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Ellen J. Hess
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Liang G, Lee YZ, Kow ASF, Lee QL, Cheng Lim LW, Yusof R, Tham CL, Ho YC, Lee MT. Neuroprotective effects of Gypenosides: A review on preclinical studies in neuropsychiatric disorders. Eur J Pharmacol 2024; 978:176766. [PMID: 38908668 DOI: 10.1016/j.ejphar.2024.176766] [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: 02/12/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Gynostemma pentaphyllum (Thunb.) Makino is a perennial creeping herb belonging to the Cucurbitaceae family that has a long history of usage in traditional oriental medicine. Gypenosides are the primary bioactive compounds in Gynostemma pentaphyllum. Because of the medicinal value of gypenosides, functional food and supplements containing gypenosides have been promoted and consumed with popularity, especially among Asian communities. This review presented the progress made in the research of pharmacological properties of gypenosides on diseases of the nervous system and their possible mechanism of action. To date, preclinical studies have demonstrated the therapeutic effects of gypenosides in alleviating neuropsychiatric disorders like depression, Parkinson's disease, Alzheimer's disease, secondary dementia, stroke, optic neuritis, etc. Pharmacological studies have discovered that gypenosides can modulate various major signaling pathways like NF-κB, Nrf2, AKT, ERK1/2, contributing to the neuroprotective properties. However, there is a dearth of clinical research on gypenosides, with current investigations on the compounds being mainly conducted in vitro and on animals. Future studies focusing on isolating and purifying novel gypenosides and investigations on exploring the potential molecular mechanism underlying their biological activities are warranted, which may serve as a foundation for further clinical trials for the betterment of human health.
Collapse
Affiliation(s)
- Gengfan Liang
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| | - Yu Zhao Lee
- Office of Postgraduate Studies, UCSI University, Kuala Lumpur, 56000, Malaysia; Faculty of Applied Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| | | | - Qi Long Lee
- School of Health Sciences, International Medical University, Kuala Lumpur, 57000, Malaysia.
| | - Luis Wei Cheng Lim
- School of Health Sciences, International Medical University, Kuala Lumpur, 57000, Malaysia.
| | - Rohana Yusof
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia.
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang , 43400, Selangor, Malaysia; Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, Serdang , 43400, Selangor, Malaysia.
| | - Yu-Cheng Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City, 82445, Taiwan.
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia; Centre of Research for Mental Health and Well-being, UCSI University, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
4
|
Ryan MB, Girasole AE, Flores AJ, Twedell EL, McGregor MM, Brakaj R, Paletzki RF, Hnasko TS, Gerfen CR, Nelson AB. Excessive firing of dyskinesia-associated striatal direct pathway neurons is gated by dopamine and excitatory synaptic input. Cell Rep 2024; 43:114483. [PMID: 39024096 DOI: 10.1016/j.celrep.2024.114483] [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: 11/28/2022] [Revised: 04/19/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
The striatum integrates dopaminergic and glutamatergic inputs to select preferred versus alternative actions. However, the precise mechanisms underlying this process remain unclear. One way to study action selection is to understand how it breaks down in pathological states. Here, we explored the cellular and synaptic mechanisms of levodopa-induced dyskinesia (LID), a complication of Parkinson's disease therapy characterized by involuntary movements. We used an activity-dependent tool (FosTRAP) in conjunction with a mouse model of LID to investigate functionally distinct subsets of striatal direct pathway medium spiny neurons (dMSNs). In vivo, levodopa differentially activates dyskinesia-associated (TRAPed) dMSNs compared to other dMSNs. We found this differential activation of TRAPed dMSNs is likely to be driven by higher dopamine receptor expression, dopamine-dependent excitability, and excitatory input from the motor cortex and thalamus. Together, these findings suggest how the intrinsic and synaptic properties of heterogeneous dMSN subpopulations integrate to support action selection.
Collapse
Affiliation(s)
- Michael B Ryan
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
| | - Allison E Girasole
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA
| | - Andrew J Flores
- Department of Neurosciences, UCSD, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Emily L Twedell
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Matthew M McGregor
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Rea Brakaj
- Department of Neurology, UCSF, San Francisco, CA 94158, USA
| | - Ronald F Paletzki
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Thomas S Hnasko
- Department of Neurosciences, UCSD, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Charles R Gerfen
- Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Alexandra B Nelson
- Neuroscience Graduate Program, UCSF, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, UCSF, San Francisco, CA 94158, USA; Weill Institute for Neurosciences, UCSF, San Francisco, CA 94158, USA; Department of Neurology, UCSF, San Francisco, CA 94158, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
5
|
Ballardin D, Makrini-Maleville L, Seper A, Valjent E, Rebholz H. 5-HT4R agonism reduces L-DOPA-induced dyskinesia via striatopallidal neurons in unilaterally 6-OHDA lesioned mice. Neurobiol Dis 2024; 198:106559. [PMID: 38852753 DOI: 10.1016/j.nbd.2024.106559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Parkinson's disease is caused by a selective vulnerability and cell loss of dopaminergic neurons of the Substantia Nigra pars compacta and, consequently, striatal dopamine depletion. In Parkinson's disease therapy, dopamine loss is counteracted by the administration of L-DOPA, which is initially effective in ameliorating motor symptoms, but over time leads to a burdening side effect of uncontrollable jerky movements, termed L-DOPA-induced dyskinesia. To date, no efficient treatment for dyskinesia exists. The dopaminergic and serotonergic systems are intrinsically linked, and in recent years, a role has been established for pre-synaptic 5-HT1a/b receptors in L-DOPA-induced dyskinesia. We hypothesized that post-synaptic serotonin receptors may have a role and investigated the effect of modulation of 5-HT4 receptor on motor symptoms and L-DOPA-induced dyskinesia in the unilateral 6-OHDA mouse model of Parkinson's disease. Administration of RS 67333, a 5-HT4 receptor partial agonist, reduces L-DOPA-induced dyskinesia without altering L-DOPA's pro-kinetic effect. In the dorsolateral striatum, we find 5-HT4 receptor to be predominantly expressed in D2R-containing medium spiny neurons, and its expression is altered by dopamine depletion and L-DOPA treatment. We further show that 5-HT4 receptor agonism not only reduces L-DOPA-induced dyskinesia, but also enhances the activation of the cAMP-PKA pathway in striatopallidal medium spiny neurons. Taken together, our findings suggest that agonism of the post-synaptic serotonin receptor 5-HT4 may be a novel therapeutic approach to reduce L-DOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Demetra Ballardin
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014 Paris, France
| | | | - Alexander Seper
- Center of Neurodegeneration, Faculty of Medicine, Danube Private University, Krems, Austria
| | - Emmanuel Valjent
- IGF, Univ. Montpellier, CNRS, INSERM, F-34094 Montpellier, France
| | - Heike Rebholz
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Laboratory of Signaling mechanisms in neurological disorders, 75014 Paris, France; Center of Neurodegeneration, Faculty of Medicine, Danube Private University, Krems, Austria; GHU-Paris Psychiatrie et Neuroscience, Hôpital Sainte Anne, F-75014 Paris, France.
| |
Collapse
|
6
|
El-Araby RE, Wasif K, Johnson R, Tu Q, Aboushousha T, Zhu ZX, Chen J. Establishment of a novel cellular model for Alzheimer's disease in vitro studies. Exp Neurol 2024; 378:114820. [PMID: 38789025 DOI: 10.1016/j.expneurol.2024.114820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/07/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory loss, cognitive impairment, and behavioral and psychological symptoms of dementia. The limited efficacy of drugs for the treatment of neurodegenerative diseases reflects their complex etiology and pathogenesis. A novel in vitro model may help to bridge the gap between existing preclinical animal models and human clinical trials, thus identifying promising therapeutic targets that can be explored in upcoming clinical trials. By assisting in the identification of the mechanism of action and potential dangers, in vitro testing can also shorten the time and expense of translation. AIM As a result of these factors, our objective is to develop a powerful and informative cellular model of AD within a short period of time. Through triggering the MAPK and NF-κβ signaling pathways with the aid of small chemical compounds (PAF C-16 and BetA), respectively, in mouse microglial (SIM-A9) and neuroblast Neuro-2a (N2a) cell lines. RESULTS PAF C-16, initiated an activation effect at a concentration of 3.12 nM to 25 nM in the SIM-A9 and N2a cell lines after 72 h. BetA, activated the NF-κβ pathway with a concentration of 12.5 nM to 25 nM in the SIM-A9 and N2a cell lines after 72 h. The combination of the activator chemicals provided suitable activation for MEK1/2-ERK and NF-κβ in more than three subcultures. Activators significantly initiate APP and MAPT gene expression, as well as the expression of proteins APP, β. Amyloid, tau, and p-tau. The activation of the targeted pathways leads to significant morphological changes. CONCLUSION We can infer that the MEK1/2-ERK and NF-κβ pathways, respectively, are directly activated by the PAF C-16 and BetA chemicals. The activation of MEK1/2-ERK pathway results in the activation of the APP gene, which in turn activates the β. Amyloid protein, which in turn results in plaque. Furthermore, NF-κβ activation results in the activation of the MAPT gene, which leads to Tau and p-Tau protein activation, which ultimately results in tangles. This can be put into practice in just three days, with a high level of activity and stability that is passed down to the next three generations (subculture), with significant morphological changes. In microglial and neuroblast cell lines, we were successful in creating a novel AD-cell model.
Collapse
Affiliation(s)
- Rady E El-Araby
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Komal Wasif
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Department of Human Physiology, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA 02215, USA
| | - Rebecca Johnson
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Department of Human Physiology, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA 02215, USA
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Tarek Aboushousha
- Theodor Bilharz Research Institute, Ministry of scientific Research, Cairo, Egypt
| | - Zoe Xiaofang Zhu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA; Department of Genetics, Molecular and Cell Biology, Tufts University School of Medicine, and Graduate School of Biomedical Sciences. 136 Harrison Ave, M&V 830, Boston, MA 02111, USA.
| |
Collapse
|
7
|
Ahmed MR, Zheng C, Dunning JL, Ahmed MS, Ge C, Pair FS, Gurevich VV, Gurevich EV. Arrestin-3-assisted activation of JNK3 mediates dopaminergic behavioral sensitization. Cell Rep Med 2024; 5:101623. [PMID: 38936368 PMCID: PMC11293330 DOI: 10.1016/j.xcrm.2024.101623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/15/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
In rodents with unilateral ablation of neurons supplying dopamine to the striatum, chronic treatment with the dopamine precursor L-DOPA induces a progressive increase of behavioral responses, a process known as behavioral sensitization. This sensitization is blunted in arrestin-3 knockout mice. Using virus-mediated gene delivery to the dopamine-depleted striatum of these mice, we find that the restoration of arrestin-3 fully rescues behavioral sensitization, whereas its mutant defective in c-Jun N-terminal kinase (JNK) activation does not. A 25-residue arrestin-3-derived peptide that facilitates JNK3 activation in cells, expressed ubiquitously or selectively in direct pathway striatal neurons, also fully rescues sensitization, whereas an inactive homologous arrestin-2-derived peptide does not. Behavioral rescue is accompanied by the restoration of JNK3 activity, as reflected by JNK-dependent phosphorylation of the transcription factor c-Jun in the dopamine-depleted striatum. Thus, arrestin-3-assisted JNK3 activation in direct pathway neurons is a critical element of the molecular mechanism underlying sensitization upon dopamine depletion and chronic L-DOPA treatment.
Collapse
Affiliation(s)
- Mohamed R Ahmed
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA; University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA; The University of Alabama at Birmingham, SHEL 121, 1825 University Boulevard, Birmingham, AL 35294-2182, USA
| | - Chen Zheng
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA
| | - Jeffery L Dunning
- Contet Laboratory, Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Mohamed S Ahmed
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA
| | - Connie Ge
- University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | - F Sanders Pair
- The University of Alabama at Birmingham, SHEL 121, 1825 University Boulevard, Birmingham, AL 35294-2182, USA
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, PRB422, Nashville, TN 37232, USA.
| |
Collapse
|
8
|
Matsuda T, Morigaki R, Hayasawa H, Koyama H, Oda T, Miyake K, Takagi Y. Striatal parvalbumin interneurons are activated in a mouse model of cerebellar dystonia. Dis Model Mech 2024; 17:dmm050338. [PMID: 38616770 PMCID: PMC11128288 DOI: 10.1242/dmm.050338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 04/09/2024] [Indexed: 04/16/2024] Open
Abstract
Dystonia is thought to arise from abnormalities in the motor loop of the basal ganglia; however, there is an ongoing debate regarding cerebellar involvement. We adopted an established cerebellar dystonia mouse model by injecting ouabain to examine the contribution of the cerebellum. Initially, we examined whether the entopeduncular nucleus (EPN), substantia nigra pars reticulata (SNr), globus pallidus externus (GPe) and striatal neurons were activated in the model. Next, we examined whether administration of a dopamine D1 receptor agonist and dopamine D2 receptor antagonist or selective ablation of striatal parvalbumin (PV, encoded by Pvalb)-expressing interneurons could modulate the involuntary movements of the mice. The cerebellar dystonia mice had a higher number of cells positive for c-fos (encoded by Fos) in the EPN, SNr and GPe, as well as a higher positive ratio of c-fos in striatal PV interneurons, than those in control mice. Furthermore, systemic administration of combined D1 receptor agonist and D2 receptor antagonist and selective ablation of striatal PV interneurons relieved the involuntary movements of the mice. Abnormalities in the motor loop of the basal ganglia could be crucially involved in cerebellar dystonia, and modulating PV interneurons might provide a novel treatment strategy.
Collapse
Affiliation(s)
- Taku Matsuda
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Ryoma Morigaki
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
- Department of Advanced Brain Research, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
- Parkinson's Disease and Dystonia Research Center, Tokushima University Hospital, Tokushima 770-8503, Japan
| | - Hiroaki Hayasawa
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Hiroshi Koyama
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Teruo Oda
- Department of Advanced Brain Research, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Kazuhisa Miyake
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
- Department of Advanced Brain Research, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| |
Collapse
|
9
|
Park J, Kang S, Lee Y, Choi JW, Oh YS. Continuous long-range measurement of tonic dopamine with advanced FSCV for pharmacodynamic analysis of levodopa-induced dyskinesia in Parkinson's disease. Front Bioeng Biotechnol 2024; 12:1335474. [PMID: 38328444 PMCID: PMC10847580 DOI: 10.3389/fbioe.2024.1335474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024] Open
Abstract
Levodopa, a dopamine prodrug, alleviates the motor symptoms of Parkinson's disease (PD), but its chronic use gives rise to levodopa-induced dyskinesia (LID). However, it remains unclear whether levodopa pharmacodynamics is altered during the progressive onset of LID. Using in vivo fast-scan cyclic voltammetry and second-derivative-based background drift removal, we continuously measured tonic dopamine levels using high temporal resolution recording over 1-h. Increases to tonic dopamine levels following acute levodopa administration were slow and marginal within the naïve PD model. However, these levels increased faster and higher in the LID model. Furthermore, we identified a strong positive correlation of dyskinetic behavior with the rate of dopamine increase, but much less with its cumulative level, at each time point. Here, we identified the altered signature of striatal DA dynamics underlying LID in PD using an advanced FSCV technique that demonstrates the long-range dynamics of tonic dopamine following drug administration.
Collapse
Affiliation(s)
- Jeongrak Park
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Seongtak Kang
- Department of Electrical Engineering and Computer Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Yaebin Lee
- Department of Electrical Engineering and Computer Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Ji-Woong Choi
- Department of Electrical Engineering and Computer Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Yong-Seok Oh
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| |
Collapse
|
10
|
Ahmed MR, Zheng C, Dunning JL, Ahmed MS, Ge C, Sanders Pair F, Gurevich VV, Gurevich EV. Arrestin-3-assisted activation of JNK3 mediates dopaminergic behavioral and signaling plasticity in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564447. [PMID: 37961199 PMCID: PMC10634923 DOI: 10.1101/2023.10.27.564447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In rodents with unilateral ablation of the substantia nigra neurons supplying dopamine to the striatum, chronic treatment with the dopamine precursor L-DOPA or dopamine agonists induces a progressive increase of behavioral responses, a process known as behavioral sensitization. The sensitization is blunted in arrestin-3 knockout mice. Using virus-mediated gene delivery to the dopamine-depleted striatum of arrestin-3 knockout mice, we found that the restoration of arrestin-3 fully rescued behavioral sensitization, whereas its mutant defective in JNK activation did not. A 25-residue arrestin-3-derived peptide that facilitates JNK3 activation in cells, expressed ubiquitously or selectively in the direct pathway striatal neurons, fully rescued sensitization, whereas an inactive homologous arrestin-2-derived peptide did not. Behavioral rescue was accompanied by the restoration of JNK3 activity and of JNK-dependent phosphorylation of the transcription factor c-Jun in the dopamine-depleted striatum. Thus, arrestin-3-dependent JNK3 activation in direct pathway neurons is a critical element of the molecular mechanism underlying sensitization.
Collapse
Affiliation(s)
- Mohamed R. Ahmed
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | - Chen Zheng
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | | | - Mohamed S. Ahmed
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | | | | | | | | |
Collapse
|
11
|
Lopez-Lopez A, Valenzuela R, Rodriguez-Perez AI, Guerra MJ, Labandeira-Garcia JL, Muñoz A. Interactions between Angiotensin Type-1 Antagonists, Statins, and ROCK Inhibitors in a Rat Model of L-DOPA-Induced Dyskinesia. Antioxidants (Basel) 2023; 12:1454. [PMID: 37507992 PMCID: PMC10376833 DOI: 10.3390/antiox12071454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/06/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Statins have been proposed for L-DOPA-induced dyskinesia (LID) treatment. Statin anti-dyskinetic effects were related to the inhibition of the Ras-ERK pathway. However, the mechanisms responsible for the anti-LID effect are unclear. Changes in cholesterol homeostasis and oxidative stress- and inflammation-related mechanisms such as angiotensin II and Rho-kinase (ROCK) inhibition may be involved. The nigra and striatum of dyskinetic rats showed increased levels of cholesterol, ROCK, and the inflammatory marker IL-1β, which were reduced by the angiotensin type-1 receptor (AT1) antagonist candesartan, simvastatin, and the ROCK inhibitor fasudil. As observed for LID, angiotensin II-induced, via AT1, increased levels of cholesterol and ROCK in the rat nigra and striatum. In cultured dopaminergic neurons, angiotensin II increased cholesterol biosynthesis and cholesterol efflux without changes in cholesterol uptake. In astrocytes, angiotensin induced an increase in cholesterol uptake, decrease in biosynthesis, and no change in cholesterol efflux, suggesting a neuronal accumulation of cholesterol that is reduced via transfer to astrocytes. Our data suggest mutual interactions between angiotensin/AT1, cholesterol, and ROCK pathways in LID, which are attenuated by the corresponding inhibitors. Interestingly, these three drugs have also been suggested as neuroprotective treatments against Parkinson's disease. Therefore, they may reduce dyskinesia and the progression of the disease using common mechanisms.
Collapse
Affiliation(s)
- Andrea Lopez-Lopez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Rita Valenzuela
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana Isabel Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - María J Guerra
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Jose Luis Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana Muñoz
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
12
|
Muraleva NA, Kolosova NG. P38 MAPK Signaling in the Retina: Effects of Aging and Age-Related Macular Degeneration. Int J Mol Sci 2023; 24:11586. [PMID: 37511345 PMCID: PMC10380409 DOI: 10.3390/ijms241411586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of irreversible visual impairment worldwide. Age is the greatest risk factor for AMD but the underlying mechanism remains unascertained, resulting in a lack of effective therapies. Growing evidence shows that dysregulation of the p38 MAPK signaling pathway (SP) contributes to aging and neurodegenerative diseases; however, information about its alteration in the retina with age and during AMD development is limited. To assess the contribution of alterations in p38 MAPK signaling to AMD, we compared age-associated changes in p38 MAPK SP activity in the retina between Wistar rats (control) and OXYS rats, which develop AMD-like retinopathy spontaneously. We analyzed changes in the mRNA levels of genes of this SP in the retina (data of RNA-seq) and evaluated the phosphorylation/activation of key kinases using Western blotting at different stages of AMD-like pathology including the preclinical stage. p38 MAPK SP activity increased in the retinas of healthy Wistar rats with age. The manifestation and dramatic progression of AMD-like pathology in OXYS rats was accompanied by hyperphosphorylation of p38 MAPK and MK2 as key p38 MAPK SP kinases. Retinopathy progression co-occurred with the enhancement of p38 MAPK-dependent phosphorylation of CryaB at Ser59 in the retina.
Collapse
Affiliation(s)
- Natalia A. Muraleva
- Institute of Cytology and Genetics (ICG), Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentieva Avenue, 630090 Novosibirsk, Russia;
| | | |
Collapse
|
13
|
Nakamura T, Nishijima H, Mori F, Kinoshita I, Kon T, Suzuki C, Wakabayashi K, Tomiyama M. Axon terminal hypertrophy of striatal projection neurons with levodopa-induced dyskinesia priming. Front Neurosci 2023; 17:1169336. [PMID: 37351424 PMCID: PMC10282195 DOI: 10.3389/fnins.2023.1169336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Background A rat model of levodopa-induced dyskinesia (LID) showed enlarged axon terminals of striatal direct pathway neurons in the internal segment of the globus pallidus (GPi) with excessive gamma-aminobutyric acid (GABA) storage in them. Massive GABA release to GPi upon levodopa administration determines the emergence of LID. Objectives We examined whether LID and axon terminal hypertrophy gradually develop with repeated levodopa treatment in Parkinsonian rats to examine if the hypertrophy reflects dyskinesia priming. Methods 6-hydroxydopamine-lesioned hemiparkinsonian rats were randomly allocated to receive saline injections (placebo group, 14 days; n = 4), injections of 6 mg/kg levodopa methyl ester combined with 12.5 mg/kg benserazide (levodopa-treated groups, 3-day-treatment; n = 4, 7-day-treatment; n = 4, 14-day-treatment; n = 4), or injections of 6 mg/kg levodopa methyl ester with 12.5 mg/kg benserazide and 1 mg/kg 8-hydroxy-2-(di-n-propylamino)tetralin for 14 days (8-OH-DPAT-treated group; n = 4). We evaluated abnormal involuntary movement (AIM) scores and axon terminals in the GPi. Results The AIM score increased with levodopa treatment, as did the hypertrophy of axon terminals in the GPi, showing an increased number of synaptic vesicles in hypertrophied terminals. Conclusion Increased GABA storage in axon terminals of the direct pathway neurons represents the priming process of LID.
Collapse
Affiliation(s)
- Takashi Nakamura
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Haruo Nishijima
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Fumiaki Mori
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Iku Kinoshita
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tomoya Kon
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Chieko Suzuki
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Masahiko Tomiyama
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
14
|
Roman KM, Briscione MA, Donsante Y, Ingram J, Fan X, Bernhard D, Campbell SA, Downs AM, Gutman D, Sardar TA, Bonno SQ, Sutcliffe DJ, Jinnah HA, Hess EJ. Striatal Subregion-selective Dysregulated Dopamine Receptor-mediated Intracellular Signaling in a Model of DOPA-responsive Dystonia. Neuroscience 2023; 517:37-49. [PMID: 36871883 PMCID: PMC10085842 DOI: 10.1016/j.neuroscience.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Although the mechanisms underlying dystonia are largely unknown, dystonia is often associated with abnormal dopamine neurotransmission. DOPA-responsive dystonia (DRD) is a prototype disorder for understanding dopamine dysfunction in dystonia because it is caused by mutations in genes necessary for the synthesis of dopamine and alleviated by the indirect-acting dopamine agonist l-DOPA. Although adaptations in striatal dopamine receptor-mediated intracellular signaling have been studied extensively in models of Parkinson's disease, another movement disorders associated with dopamine deficiency, little is known about dopaminergic adaptations in dystonia. To identify the dopamine receptor-mediated intracellular signaling associated with dystonia, we used immunohistochemistry to quantify striatal protein kinase A activity and extracellular signal-related kinase (ERK) phosphorylation after dopaminergic challenges in a knockin mouse model of DRD. l-DOPA treatment induced the phosphorylation of both protein kinase A substrates and ERK largely in D1 dopamine receptor-expressing striatal neurons. As expected, this response was blocked by pretreatment with the D1 dopamine receptor antagonist SCH23390. The D2 dopamine receptor antagonist raclopride also significantly reduced the phosphorylation of ERK; this contrasts with models of parkinsonism in which l-DOPA-induced ERK phosphorylation is not mediated by D2 dopamine receptors. Further, the dysregulated signaling was dependent on striatal subdomains whereby ERK phosphorylation was largely confined to dorsomedial (associative) striatum while the dorsolateral (sensorimotor) striatum was unresponsive. This complex interaction between striatal functional domains and dysregulated dopamine-receptor mediated responses has not been observed in other models of dopamine deficiency, such as parkinsonism, suggesting that regional variation in dopamine-mediated neurotransmission may be a hallmark of dystonia.
Collapse
Affiliation(s)
- Kaitlyn M Roman
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Maria A Briscione
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Jordan Ingram
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - Simone A Campbell
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Anthony M Downs
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - David Gutman
- Department of Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Tejas A Sardar
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Sofia Q Bonno
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - H A Jinnah
- Department of Neurology, Emory University, Atlanta, GA, USA; Department of Human Genetics, Emory University, Atlanta, GA, USA; Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Ellen J Hess
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
15
|
Castner SA, Zhang L, Yang CR, Hao J, Cramer JW, Wang X, Bruns RF, Marston H, Svensson KA, Williams GV. Effects of DPTQ, a novel positive allosteric modulator of the dopamine D1 receptor, on spontaneous eye blink rate and spatial working memory in the nonhuman primate. Psychopharmacology (Berl) 2023; 240:1033-1048. [PMID: 36961560 PMCID: PMC10102062 DOI: 10.1007/s00213-022-06282-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/23/2022] [Indexed: 03/25/2023]
Abstract
RATIONALE Dopamine (DA) signaling through the D1 receptor has been shown to be integral to multiple aspects of cognition, including the core process of working memory. The discovery of positive allosteric modulators (PAMs) of the D1 receptor has enabled treatment modalities that may have alternative benefits to orthosteric D1 agonists arising from a synergism of action with functional D1 receptor signaling. OBJECTIVES To investigate this potential, we have studied the effects of the novel D1 PAM DPTQ on a spatial delayed response working memory task in the rhesus monkey. Initial studies indicated that DPTQ binds to primate D1R with high affinity and selectivity and elevates spontaneous eye blink rate in rhesus monkeys in a dose-dependent manner consistent with plasma ligand exposures and central D1activation. RESULTS Based on those results, DPTQ was tested at 2.5 mg/kg IM in the working memory task. No acute effect was observed 1 h after dosing, but performance was impaired 48 h later. Remarkably, this deficit was immediately followed by a significant enhancement in cognition over the next 3 days. In a second experiment in which DPTQ was administered on days 1 and 5, the early impairment was smaller and did not reach statistical significance, but statistically significant enhancement of performance was observed over the following week. Lower doses of 0.1 and 1.0 mg/kg were also capable of producing this protracted enhancement without inducing any transient impairment. CONCLUSIONS DPTQ exemplifies a class of D1PAMs that may be capable of providing long-term improvements in working memory.
Collapse
Affiliation(s)
- Stacy A Castner
- Department of Comparative Medicine, Yale University, 310 Cedar St, New Haven, CT, 06520, USA
| | - Linli Zhang
- ChemPartner, 99 Lian He North Road, Zhe Lin Town, Fengxian Area, Shanghai, China
| | - Charles R Yang
- ChemPartner, 99 Lian He North Road, Zhe Lin Town, Fengxian Area, Shanghai, China
| | - Junliang Hao
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Jeffrey W Cramer
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Xushan Wang
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Robert F Bruns
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | | | - Kjell A Svensson
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Graham V Williams
- Department of Comparative Medicine, Yale University, 310 Cedar St, New Haven, CT, 06520, USA.
| |
Collapse
|
16
|
Gerfen CR. Segregation of D1 and D2 dopamine receptors in the striatal direct and indirect pathways: An historical perspective. Front Synaptic Neurosci 2023; 14:1002960. [PMID: 36741471 PMCID: PMC9892636 DOI: 10.3389/fnsyn.2022.1002960] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023] Open
Abstract
The direct and indirect striatal pathways form a cornerstone of the circuits of the basal ganglia. Dopamine has opponent affects on the function of these pathways due to the segregation of the D1- and D2-dopamine receptors in the spiny projection neurons giving rise to the direct and indirect pathways. An historical perspective is provided on the discovery of dopamine receptor segregation leading to models of how the direct and indirect affect motor behavior.
Collapse
|
17
|
Argyrofthalmidou M, Polissidis A, Karaliota S, Papapanagiotou I, Sotiriou E, Manousaki M, Papadopoulou-Daifoti Z, Spillantini MG, Stefanis L, Vassilatis DK. Functional Interaction Between α-Synuclein and Nurr1 in Dopaminergic Neurons. Neuroscience 2022; 506:114-126. [PMID: 36270413 DOI: 10.1016/j.neuroscience.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/24/2022]
Abstract
Increased expression of alpha-synuclein (ASYN) and decreased expression of Nurr1 are associated with Parkinson's disease (PD) pathogenesis. These two proteins interact functionally and ASYN overexpression suppresses Nurr1 levels. ASYN pan-neuronal overexpression coupled with Nurr1 hemizygosity followed by Nurr1 repression in aging mice results in the manifestation of a typical PD-related phenotype and pathology. Here we investigated in mice the effects of C-terminally truncated ASYN(120) overexpression in dopaminergic (DA-ergic) neurons compounded with Nurr1 hemizygosity ('2-hit-DA'). We report that '2-hit-DA' animals did not manifest a characteristic PD-related phenotype, despite further substantia nigra ASYN-overexpression-dependent and age dependent Nurr1 protein downregulation. However, they displayed increased energy expenditure, reduced striatal dopamine (DA) and prolonged hyperactivity to a novel environment indicating impaired habituation. This DA-ergic dysfunction was observed in young adult '2-hit-DA' mice, persisted throughout life and it was associated with ASYN and Nurr1 synergistic alterations of DAT levels and function. Our experiments indicate that the expression levels of ASYN and Nurr1 are critical in the dysregulation of the nigrostriatal DA system and may be involved in neuropsychiatric aspects of PD.
Collapse
Affiliation(s)
- Maria Argyrofthalmidou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Alexia Polissidis
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Sevasti Karaliota
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Basic Science Program, Frederick National Laboratory for Cancer Research, NCI/NIH, Frederick, MD 21702-1201, USA
| | - Ioanna Papapanagiotou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Evangelos Sotiriou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Maria Manousaki
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | | | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK
| | - Leonidas Stefanis
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Second Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens 11527, Greece
| | - Demetrios K Vassilatis
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece.
| |
Collapse
|
18
|
Potential Effects of Nrf2 in Exercise Intervention of Neurotoxicity Caused by Methamphetamine Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4445734. [PMID: 35480870 PMCID: PMC9038420 DOI: 10.1155/2022/4445734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/01/2022] [Indexed: 12/15/2022]
Abstract
Methamphetamine can cause oxidative stress-centered lipid peroxidation, endoplasmic reticulum stress, mitochondrial dysfunction, excitatory neurotoxicity, and neuroinflammation and ultimately lead to nerve cell apoptosis, abnormal glial cell activation, and dysfunction of blood-brain barrier. Protecting nerve cells from oxidative destroy is a hopeful strategy for treating METH use disorder. Nrf2 is a major transcriptional regulator that activates the antioxidant, anti-inflammatory, and cell-protective gene expression through endogenous pathways that maintains cell REDOX homeostasis and is conducive to the survival of neurons. The Nrf2-mediated endogenous antioxidant pathway can also prevent neurodegenerative effects and functional defects caused by METH oxidative stress. Moderate exercise activates this endogenous antioxidant system, which involves in many diseases, including neurodegenerative diseases. Based on evidence from existing literature, we argue that appropriate exercise can play an endogenous antioxidant regulatory role in the Nrf2 signaling pathway to reduce a number of issues caused by METH-induced oxidative stress. However, more experimental evidence is needed to support this idea. In addition, further exploration is necessary about the different effects of various parameters of exercise intervention (such as exercise mode, time, and intensity) on the Nrf2 signaling pathway intervention. Whether there are synergistic effects between exercise and plant-derived Nrf2 activators is worth further investigation.
Collapse
|
19
|
Scarduzio M, Hess EJ, Standaert DG, Eskow Jaunarajs KL. Striatal synaptic dysfunction in dystonia and levodopa-induced dyskinesia. Neurobiol Dis 2022; 166:105650. [DOI: 10.1016/j.nbd.2022.105650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
|
20
|
Jones-Tabah J, Mohammad H, Paulus EG, Clarke PBS, Hébert TE. The Signaling and Pharmacology of the Dopamine D1 Receptor. Front Cell Neurosci 2022; 15:806618. [PMID: 35110997 PMCID: PMC8801442 DOI: 10.3389/fncel.2021.806618] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
The dopamine D1 receptor (D1R) is a Gαs/olf-coupled GPCR that is expressed in the midbrain and forebrain, regulating motor behavior, reward, motivational states, and cognitive processes. Although the D1R was initially identified as a promising drug target almost 40 years ago, the development of clinically useful ligands has until recently been hampered by a lack of suitable candidate molecules. The emergence of new non-catechol D1R agonists, biased agonists, and allosteric modulators has renewed clinical interest in drugs targeting this receptor, specifically for the treatment of motor impairment in Parkinson's Disease, and cognitive impairment in neuropsychiatric disorders. To develop better therapeutics, advances in ligand chemistry must be matched by an expanded understanding of D1R signaling across cell populations in the brain, and in disease states. Depending on the brain region, the D1R couples primarily to either Gαs or Gαolf through which it activates a cAMP/PKA-dependent signaling cascade that can regulate neuronal excitability, stimulate gene expression, and facilitate synaptic plasticity. However, like many GPCRs, the D1R can signal through multiple downstream pathways, and specific signaling signatures may differ between cell types or be altered in disease. To guide development of improved D1R ligands, it is important to understand how signaling unfolds in specific target cells, and how this signaling affects circuit function and behavior. In this review, we provide a summary of D1R-directed signaling in various neuronal populations and describe how specific pathways have been linked to physiological and behavioral outcomes. In addition, we address the current state of D1R drug development, including the pharmacology of newly developed non-catecholamine ligands, and discuss the potential utility of D1R-agonists in Parkinson's Disease and cognitive impairment.
Collapse
|
21
|
Beeler JA, Burghardt NS. The Rise and Fall of Dopamine: A Two-Stage Model of the Development and Entrenchment of Anorexia Nervosa. Front Psychiatry 2022; 12:799548. [PMID: 35087433 PMCID: PMC8787068 DOI: 10.3389/fpsyt.2021.799548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Dopamine has long been implicated as a critical neural substrate mediating anorexia nervosa (AN). Despite nearly 50 years of research, the putative direction of change in dopamine function remains unclear and no consensus on the mechanistic role of dopamine in AN has been achieved. We hypothesize two stages in AN- corresponding to initial development and entrenchment- characterized by opposite changes in dopamine. First, caloric restriction, particularly when combined with exercise, triggers an escalating spiral of increasing dopamine that facilitates the behavioral plasticity necessary to establish and reinforce weight-loss behaviors. Second, chronic self-starvation reverses this escalation to reduce or impair dopamine which, in turn, confers behavioral inflexibility and entrenchment of now established AN behaviors. This pattern of enhanced, followed by impaired dopamine might be a common path to many behavioral disorders characterized by reinforcement learning and subsequent behavioral inflexibility. If correct, our hypothesis has significant clinical and research implications for AN and other disorders, such as addiction and obesity.
Collapse
Affiliation(s)
- Jeff A. Beeler
- Department of Psychology, Queens College, City University of New York, Flushing, NY, United States
- Psychology Program, The Graduate Center, CUNY, New York, NY, United States
- Biology Program, The Graduate Center, City University of New York, New York, NY, United States
| | - Nesha S. Burghardt
- Psychology Program, The Graduate Center, CUNY, New York, NY, United States
- Department of Psychology, Hunter College, CUNY, New York, NY, United States
| |
Collapse
|
22
|
Jones-Tabah J, Martin RD, Tanny JC, Clarke PBS, Hébert TE. High-Content Single-Cell Förster Resonance Energy Transfer Imaging of Cultured Striatal Neurons Reveals Novel Cross-Talk in the Regulation of Nuclear Signaling by Protein Kinase A and Extracellular Signal-Regulated Kinase 1/2. Mol Pharmacol 2021; 100:526-539. [PMID: 34503973 DOI: 10.1124/molpharm.121.000290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Genetically encoded biosensors can be used to track signaling events in living cells by measuring changes in fluorescence emitted by one or more fluorescent proteins. Here, we describe the use of genetically encoded biosensors based on Förster resonance energy transfer (FRET), combined with high-content microscopy, to image dynamic signaling events simultaneously in thousands of neurons in response to drug treatments. We first applied this approach to examine intercellular variation in signaling responses among cultured striatal neurons stimulated with multiple drugs. Using high-content FRET imaging and immunofluorescence, we identified neuronal subpopulations with unique responses to pharmacological manipulation and used nuclear morphology to identify medium spiny neurons within these heterogeneous striatal cultures. Focusing on protein kinase A (PKA) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in the cytoplasm and nucleus, we noted pronounced intercellular differences among putative medium spiny neurons, in both the magnitude and kinetics of signaling responses to drug application. Importantly, a conventional "bulk" analysis that pooled all cells in culture yielded a different rank order of drug potency than that revealed by single-cell analysis. Using a single-cell analytical approach, we dissected the relative contributions of PKA and ERK1/2 signaling in striatal neurons and unexpectedly identified a novel role for ERK1/2 in promoting nuclear activation of PKA in striatal neurons. This finding adds a new dimension of signaling crosstalk between PKA and ERK1/2 with relevance to dopamine D1 receptor signaling in striatal neurons. In conclusion, high-content single-cell imaging can complement and extend traditional population-level analyses and provides a novel vantage point from which to study cellular signaling. SIGNIFICANCE STATEMENT: High-content imaging revealed substantial intercellular variation in the magnitude and pattern of intracellular signaling events driven by receptor stimulation. Since individual neurons within the same population can respond differently to a given agonist, interpreting measures of intracellular signaling derived from the averaged response of entire neuronal populations may not always reflect what happened at the single-cell level. This study uses this approach to identify a new form of cross-talk between PKA and ERK1/2 signaling in the nucleus of striatal neurons.
Collapse
Affiliation(s)
- Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Paul B S Clarke
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| |
Collapse
|
23
|
MEK1/2-ERK Pathway Alterations as a Therapeutic Target in Sporadic Alzheimer's Disease: A Study in Senescence-Accelerated OXYS Rats. Antioxidants (Basel) 2021; 10:antiox10071058. [PMID: 34208998 PMCID: PMC8300733 DOI: 10.3390/antiox10071058] [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: 06/02/2021] [Revised: 06/21/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia worldwide, with no cure. There is growing interest in mitogen-activated protein kinases (MAPKs) as possible pathogenesis-related therapeutic targets in AD. Previously, using senescence-accelerated OXYS rats, which simulate key characteristics of the sporadic AD type, we have shown that prolonged treatment with mitochondria-targeted antioxidant plastoquinonyl-decyltriphenylphosphonium (SkQ1) during active progression of AD-like pathology improves the activity of many signaling pathways (SPs) including the p38 MAPK SP. In this study, we continued to investigate the mechanisms behind anti-AD effects of SkQ1 in OXYS rats and focused on hippocampal extracellular regulated kinases’ (ERK1 and -2) activity alterations. According to high-throughput RNA sequencing results, SkQ1 eliminated differences in the expression of eight out of nine genes involved in the ERK1/2 SP, compared to untreated control (Wistar) rats. Western blotting and immunofluorescent staining revealed that SkQ1 suppressed ERK1/2 activity via reductions in the phosphorylation of kinases ERK1/2, MEK1, and MEK2. SkQ1 decreased hyperphosphorylation of tau protein, which is present in pathological aggregates in AD. Thus, SkQ1 alleviates AD pathology by suppressing MEK1/2-ERK1/2 SP activity in the OXYS rat hippocampus and may be a promising candidate drug for human AD.
Collapse
|
24
|
Simola N, Serra M, Marongiu J, Costa G, Morelli M. Increased emissions of 50-kHz ultrasonic vocalizations in hemiparkinsonian rats repeatedly treated with dopaminomimetic drugs: A potential preclinical model for studying the affective properties of dopamine replacement therapy in Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2021; 108:110184. [PMID: 33242502 DOI: 10.1016/j.pnpbp.2020.110184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Dopamine replacement therapy used in Parkinson's disease (PD) may induce alterations in the emotional state that can underlie the manifestation of iatrogenic psychiatric-like disturbances. The preclinical investigation of these disturbances is limited, also because few reliable paradigms are available to study the affective properties of dopaminomimetic drugs in parkinsonian animals. To provide a relevant experimental tool in this respect, we evaluated whether dopaminomimetic drugs modified the emission of 50-kHz ultrasonic vocalizations (USVs), a behavioral marker of positive affect, in rats bearing a unilateral lesion with 6-hydroxydopamine in the medial forebrain bundle. Apomorphine (2 or 4 mg/kg, i.p.), L-3,4-dihydroxyphenilalanine (L-DOPA, 6 or 12 mg/kg, i.p.), or pramipexole (2 or 4 mg/kg, i.p.) were administered in a test cage (× 5 administrations) on alternate days. Seven days after treatment discontinuation, rats were re-exposed to the test cage to measure conditioned calling behavior and thereafter received a drug challenge. Hemiparkinsonian rats treated with either apomorphine or L-DOPA, but not pramipexole, markedly vocalized during repeated treatment and after challenge, and showed conditioned calling behavior. Moreover, apomorphine, L-DOPA and pramipexole elicited different patterns of 50-kHz USV emissions and rotational behavior, indicating that calling behavior in hemiparkinsonian rats treated with dopaminomimetic drugs is not a byproduct of motor activation. Taken together, these results suggest that measuring 50-kHz USV emissions may be a relevant experimental tool for studying how dopaminomimetic drugs modify the affective state in parkinsonian rats, with possible implications for the preclinical investigation of iatrogenic psychiatric-like disturbances in PD.
Collapse
Affiliation(s)
- Nicola Simola
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy.
| | - Marcello Serra
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giulia Costa
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy; CNR, National Research Council of Italy, Neuroscience Institute, Cagliari, Italy
| |
Collapse
|
25
|
Ferré S, Guitart X, Quiroz C, Rea W, García-Malo C, Garcia-Borreguero D, Allen RP, Earley CJ. Akathisia and Restless Legs Syndrome: Solving the Dopaminergic Paradox. Sleep Med Clin 2021; 16:249-267. [PMID: 33985651 DOI: 10.1016/j.jsmc.2021.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Akathisia is an urgent need to move that is associated with treatment with dopamine receptor blocking agents (DRBAs) and with restless legs syndrome (RLS). The pathogenetic mechanism of akathisia has not been resolved. This article proposes that it involves an increased presynaptic dopaminergic transmission in the ventral striatum and concomitant strong activation of postsynaptic dopamine D1 receptors, which form complexes (heteromers) with dopamine D3 and adenosine A1 receptors. It also proposes that in DRBA-induced akathisia, increased dopamine release depends on inactivation of autoreceptors, whereas in RLS it depends on a brain iron deficiency-induced down-regulation of striatal presynaptic A1 receptors.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Xavier Guitart
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - César Quiroz
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - William Rea
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Celia García-Malo
- Sleep Research Institute, Paseo de la Habana 151, Madrid 28036, Spain
| | | | - Richard P Allen
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Christopher J Earley
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| |
Collapse
|
26
|
Dyavar SR, Potts LF, Beck G, Dyavar Shetty BL, Lawson B, Podany AT, Fletcher CV, Amara RR, Papa SM. Transcriptomic approach predicts a major role for transforming growth factor beta type 1 pathway in L-Dopa-induced dyskinesia in parkinsonian rats. GENES BRAIN AND BEHAVIOR 2020; 19:e12690. [PMID: 32741046 DOI: 10.1111/gbb.12690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 01/21/2023]
Abstract
Dyskinesia induced by long-term L-Dopa (LID) therapy in Parkinson disease is associated with altered striatal function whose molecular bases remain unclear. Here, a transcriptomic approach was applied for comprehensive analysis of distinctively regulated genes in striatal tissue, their specific pathways, and functional- and disease-associated networks in a rodent model of LID. This approach has identified transforming growth factor beta type 1 (TGFβ1) as a highly upregulated gene in dyskinetic animals. TGFβ1 pathway is a top aberrantly regulated pathway in the striatum following LID development based on differentially expressed genes (> 1.5 fold change and P < 0.05). The induction of TGFβ1 pathway specific genes, TGFβ1, INHBA, AMHR2 and PMEPA1 was also associated with regulation of NPTX2, PDP1, SCG2, SYNPR, TAC1, TH, TNNT1 genes. Transcriptional network and upstream regulator analyses have identified AKT-centered functional and ERK-centered disease networks revealing the association of TGFβ1, IL-1β and TNFα with LID development. Therefore, results support that TGFβ1 pathway is a major contributor to the pathogenic mechanisms of LID.
Collapse
Affiliation(s)
- Shetty Ravi Dyavar
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Lisa F Potts
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Goichi Beck
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | | | - Benton Lawson
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Anthony T Podany
- Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Courtney V Fletcher
- Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rama Rao Amara
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
27
|
Pascale E, Divisato G, Palladino R, Auriemma M, Ngalya EF, Caiazzo M. Noncoding RNAs and Midbrain DA Neurons: Novel Molecular Mechanisms and Therapeutic Targets in Health and Disease. Biomolecules 2020; 10:E1269. [PMID: 32899172 PMCID: PMC7563414 DOI: 10.3390/biom10091269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Midbrain dopamine neurons have crucial functions in motor and emotional control and their degeneration leads to several neurological dysfunctions such as Parkinson's disease, addiction, depression, schizophrenia, and others. Despite advances in the understanding of specific altered proteins and coding genes, little is known about cumulative changes in the transcriptional landscape of noncoding genes in midbrain dopamine neurons. Noncoding RNAs-specifically microRNAs and long noncoding RNAs-are emerging as crucial post-transcriptional regulators of gene expression in the brain. The identification of noncoding RNA networks underlying all stages of dopamine neuron development and plasticity is an essential step to deeply understand their physiological role and also their involvement in the etiology of dopaminergic diseases. Here, we provide an update about noncoding RNAs involved in dopaminergic development and metabolism, and the related evidence of these biomolecules for applications in potential treatments for dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Emilia Pascale
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (G.D.); (R.P.); (M.A.); (E.F.N.)
| | - Giuseppina Divisato
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (G.D.); (R.P.); (M.A.); (E.F.N.)
| | - Renata Palladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (G.D.); (R.P.); (M.A.); (E.F.N.)
| | - Margherita Auriemma
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (G.D.); (R.P.); (M.A.); (E.F.N.)
| | - Edward Faustine Ngalya
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (G.D.); (R.P.); (M.A.); (E.F.N.)
| | - Massimiliano Caiazzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy; (E.P.); (G.D.); (R.P.); (M.A.); (E.F.N.)
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
28
|
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: 3.6] [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
|
29
|
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: 2.2] [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
|
30
|
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: 1.2] [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
|
31
|
Nishijima H, Mori F, Arai A, Zhu G, Wakabayashi K, Okada M, Ueno S, Ichinohe N, Suzuki C, Kon T, Tomiyama M. GABA storage and release in the medial globus pallidus in L-DOPA-induced dyskinesia priming. Neurobiol Dis 2020; 143:104979. [PMID: 32590036 DOI: 10.1016/j.nbd.2020.104979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 01/13/2023] Open
Abstract
Levo-dihydroxyphenylalanine (L-DOPA) is the most effective treatment for Parkinson's disease; however, most patients develop uncontrollable abnormal involuntary movements known as L-DOPA-induced dyskinesia. L-DOPA-induced dyskinesia can be reduced by pallidotomy of the medial globus pallidus or pallidal deep brain stimulation, suggesting that the medial globus pallidus plays a significant role in the development of L-DOPA-induced dyskinesia. In the present study, the pathological changes of the medial globus pallidus in L-DOPA-induced dyskinesia were studied in rat models of Parkinson's disease (unilateral 6-hydroxydopamine lesioning) and L-DOPA-induced dyskinesia (L-DOPA injection in Parkinson's disease-model rats twice daily for 2 weeks, confirmed by display of dyskinesia-like abnormal involuntary movements). L-DOPA-induced dyskinesia-model rats displayed medial globus pallidus hypertrophy, enlarged axon terminals surrounding the dendrites of medial globus pallidus neurons, and increased density of synaptic vesicles in enlarged axon terminals on the lesioned side. Synaptic terminal enlargement reversed after discontinuation of L-DOPA. Histological studies revealed the enlarged synaptic terminals were those of GABAergic striatal (direct pathway) neurons. A single injection of L-DOPA enhanced GABA release in the medial globus pallidus on the lesioned side in L-DOPA-induced dyskinesia-model rats compared to Parkinson's disease-model rats. In addition, microinjection of muscimol, a GABAA receptor agonist, into the medial globus pallidus on the lesioned side of Parkinson's disease-model rats induced dyskinesia-like abnormal involuntary movements. Microinjection of bicuculline, a GABAA receptor antagonist, into the medial globus pallidus on the lesioned side alleviated L-DOPA-induced dyskinesia in Parkinson's disease-model rats that had received L-DOPA prior to the microinjection. These results indicate that priming for L-DOPA-induced dyskinesia comprises excessive GABA storage in axon terminals of the direct pathway and that expression of L-DOPA-induced dyskinesia is associated with enhanced GABA release into the medial globus pallidus after L-DOPA dosing and the resultant excessive stimulation of GABAA receptors.
Collapse
Affiliation(s)
- Haruo Nishijima
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan.
| | - Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan
| | - Akira Arai
- Department of Neurology, Aomori Prefectural Central Hospital, 2-1-1 Higashi-Tsukurimichi, Aomori 030-8551, Japan
| | - Gang Zhu
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan
| | - Motohiro Okada
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Shinya Ueno
- Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan
| | - Noritaka Ichinohe
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawahigashi-cho, Kodaira, Tokyo 187-8502, Japan; Ichinohe Neural System Group, Laboratory for Molecular Analysis of Higher Brain Functions, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Chieko Suzuki
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan
| | - Tomoya Kon
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan
| | - Masahiko Tomiyama
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zifu-cho, Hirosaki 036-8562, Japan
| |
Collapse
|
32
|
Tubert C, Murer MG. What’s wrong with the striatal cholinergic interneurons in Parkinson’s disease? Focus on intrinsic excitability. Eur J Neurosci 2020; 53:2100-2116. [DOI: 10.1111/ejn.14742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilia Tubert
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| | - Mario Gustavo Murer
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| |
Collapse
|
33
|
Babenko VN, Galyamina AG, Rogozin IB, Smagin DA, Kudryavtseva NN. Dopamine response gene pathways in dorsal striatum MSNs from a gene expression viewpoint: cAMP-mediated gene networks. BMC Neurosci 2020; 21:12. [PMID: 32216748 PMCID: PMC7099774 DOI: 10.1186/s12868-020-00560-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/18/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Medium spiny neurons (MSNs) comprise the main body (95% in mouse) of the dorsal striatum neurons and represent dopaminoceptive GABAergic neurons. The cAMP (cyclic Adenosine MonoPhosphate)-mediated cascade of excitation and inhibition responses observed in MSN intracellular signal transduction is crucial for neuroscience research due to its involvement in the motor and behavioral functions. In particular, all types of addictions are related to MSNs. Shedding the light on the mechanics of the above-mentioned cascade is of primary importance for this research domain. RESULTS A mouse model of chronic social conflicts in daily agonistic interactions was used to analyze dorsal striatum neurons genes implicated in cAMP-mediated phosphorylation activation pathways specific for MSNs. Based on expression correlation analysis, we succeeded in dissecting Drd1- and Drd2-dopaminoceptive neurons (D1 and D2, correspondingly) gene pathways. We also found that D1 neurons genes clustering are split into two oppositely correlated states, passive and active ones, the latter apparently corresponding to D1 firing stage upon protein kinase A (PKA) activation. We observed that under defeat stress in chronic social conflicts the loser mice manifest overall depression of dopamine-mediated MSNs activity resulting in previously reported reduced motor activity, while the aggressive mice with positive fighting experience (aggressive mice) feature an increase in both D1-active phase and D2 MSNs genes expression leading to hyperactive behavior pattern corresponded by us before. Based on the alternative transcript isoforms expression analysis, it was assumed that many genes (Drd1, Adora1, Pde10, Ppp1r1b, Gnal), specifically those in D1 neurons, apparently remain transcriptionally repressed via the reversible mechanism of promoter CpG island silencing, resulting in alternative promoter usage following profound reduction in their expression rate. CONCLUSION Based on the animal stress model dorsal striatum pooled tissue RNA-Seq data restricted to cAMP related genes subset we elucidated MSNs steady states exhaustive projection for the first time. We correspond the existence of D1 active state not explicitly outlined before, and connected with dynamic dopamine neurotransmission cycles. Consequently, we were also able to indicate an oscillated postsynaptic dopamine vs glutamate action pattern in the course of the neurotransmission cycles.
Collapse
Affiliation(s)
- Vladimir N Babenko
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.
- Novosibirsk State University, Novosibirsk, Russia.
| | | | - Igor B Rogozin
- National Institutes of Health, Rockville Pike, Bethesda, MD, USA
| | - Dmitry A Smagin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | | |
Collapse
|
34
|
Castello J, Cortés M, Malave L, Kottmann A, Sibley DR, Friedman E, Rebholz H. The Dopamine D5 receptor contributes to activation of cholinergic interneurons during L-DOPA induced dyskinesia. Sci Rep 2020; 10:2542. [PMID: 32054879 PMCID: PMC7018760 DOI: 10.1038/s41598-020-59011-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 12/30/2019] [Indexed: 01/28/2023] Open
Abstract
The dopamine D5 receptor (D5R) is a Gαs-coupled dopamine receptor belonging to the dopamine D1-like receptor family. Together with the dopamine D2 receptor it is highly expressed in striatal cholinergic interneurons and therefore is poised to be a positive regulator of cholinergic activity in response to L-DOPA in the dopamine-depleted parkinsonian brain. Tonically active cholinergic interneurons become dysregulated during chronic L-DOPA administration and participate in the expression of L-DOPA induced dyskinesia. The molecular mechanisms involved in this process have not been elucidated, however a correlation between dyskinesia severity and pERK expression in cholinergic cells has been described. To better understand the function of the D5 receptor and how it affects cholinergic interneurons in L-DOPA induced dyskinesia, we used D5R knockout mice that were rendered parkinsonian by unilateral 6-OHDA injection. In the KO mice, expression of pERK was strongly reduced indicating that activation of these cells is at least in part driven by the D5 receptor. Similarly, pS6, another marker for the activity status of cholinergic interneurons was also reduced. However, mice lacking D5R exhibited slightly worsened locomotor performance in response to L-DOPA and enhanced LID scores. Our findings suggest that D5R can modulate L-DOPA induced dyskinesia and is a critical activator of CINs via pERK and pS6.
Collapse
Affiliation(s)
- Julia Castello
- Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine, New York, NY, USA
- Ph.D. Programs in Biochemistry and Biology, The Graduate Center, CUNY, New York, USA
| | - Marisol Cortés
- Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine, New York, NY, USA
| | - Lauren Malave
- Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine, New York, NY, USA
- Ph.D. Programs in Biochemistry and Biology, The Graduate Center, CUNY, New York, USA
| | - Andreas Kottmann
- Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine, New York, NY, USA
- Ph.D. Programs in Biochemistry and Biology, The Graduate Center, CUNY, New York, USA
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurologic Disorders and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland, USA
| | - Eitan Friedman
- Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine, New York, NY, USA
- Ph.D. Programs in Biochemistry and Biology, The Graduate Center, CUNY, New York, USA
| | - Heike Rebholz
- Department of Molecular, Cellular & Biomedical Sciences, CUNY School of Medicine, New York, NY, USA.
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR_S1266, INSERM, Université de Paris, 102-108 rue de la Santé, F-75014, Paris, France.
- GHU PARIS psychiatrie et neurosciences, Paris, France.
- Danube Private University (DPU), Krems, Austria.
| |
Collapse
|
35
|
Padovan-Neto FE, Patterson S, F Voelkner NM, Altwal F, Beverley JA, West AR, Steiner H. Selective Regulation of 5-HT1B Serotonin Receptor Expression in the Striatum by Dopamine Depletion and Repeated L-DOPA Treatment: Relationship to L-DOPA-Induced Dyskinesias. Mol Neurobiol 2020; 57:736-751. [PMID: 31468338 PMCID: PMC7035192 DOI: 10.1007/s12035-019-01739-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/16/2019] [Indexed: 11/28/2022]
Abstract
Dopamine and serotonin in the basal ganglia interact in a bidirectional manner. On the one hand, serotonin (5-HT) receptors regulate the effects of dopamine agonists on several levels, ranging from molecular signaling to behavior. These interactions include 5-HT receptor-mediated facilitation of dopamine receptor-induced gene regulation in striatal output pathways, which involves the 5-HT1B receptor and others. Conversely, there is evidence that dopamine action by psychostimulants regulates 5-HT1B receptor expression in the striatum. To further investigate the effects of dopamine and agonists on 5-HT receptors, we assessed the expression of 5-HT1B and other serotonin receptor subtypes in the striatum after unilateral dopamine depletion by 6-OHDA and subsequent treatment with L-DOPA (5 mg/kg; 4 weeks). Neither dopamine depletion nor L-DOPA treatment produced significant changes in 5-HT2C, 5-HT4, or 5-HT6 receptor expression in the striatum. In contrast, the 6-OHDA lesion caused a (modest) increase in 5-HT1B mRNA levels throughout the striatum. Moreover, repeated L-DOPA treatment markedly further elevated 5-HT1B expression in the dopamine-depleted striatum, an effect that was most robust in the sensorimotor striatum. A minor L-DOPA-induced increase in 5-HT1B expression was also seen in the intact striatum. These changes in 5-HT1B expression mimicked changes in the expression of neuropeptide markers (dynorphin, enkephalin mRNA) in striatal projection neurons. After repeated L-DOPA treatment, the severity of L-DOPA-induced dyskinesias and turning behavior was positively correlated with the increase in 5-HT1B expression in the associative, but not sensorimotor, striatum ipsilateral to the lesion, suggesting that associative striatal 5-HT1B receptors may play a role in L-DOPA-induced behavioral abnormalities.
Collapse
Affiliation(s)
- Fernando E Padovan-Neto
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Avenida dos Bandeirantes, 3900, Ribeirão Preto, 14040-901, SP, Brazil
| | - Santanna Patterson
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Nivea M F Voelkner
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Feras Altwal
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Joel A Beverley
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Anthony R West
- Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Heinz Steiner
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
36
|
Striatal overexpression of β-arrestin2 counteracts L-dopa-induced dyskinesia in 6-hydroxydopamine lesioned Parkinson's disease rats. Neurochem Int 2019; 131:104543. [DOI: 10.1016/j.neuint.2019.104543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/16/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
|
37
|
Bocarsly ME, da Silva E Silva D, Kolb V, Luderman KD, Shashikiran S, Rubinstein M, Sibley DR, Dobbs LK, Alvarez VA. A Mechanism Linking Two Known Vulnerability Factors for Alcohol Abuse: Heightened Alcohol Stimulation and Low Striatal Dopamine D2 Receptors. Cell Rep 2019; 29:1147-1163.e5. [PMID: 31665630 PMCID: PMC6880649 DOI: 10.1016/j.celrep.2019.09.059] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/10/2019] [Accepted: 09/18/2019] [Indexed: 12/21/2022] Open
Abstract
Alcohol produces both stimulant and sedative effects in humans and rodents. In humans, alcohol abuse disorder is associated with a higher stimulant and lower sedative responses to alcohol. Here, we show that this association is conserved in mice and demonstrate a causal link with another liability factor: low expression of striatal dopamine D2 receptors (D2Rs). Using transgenic mouse lines, we find that the selective loss of D2Rs on striatal medium spiny neurons enhances sensitivity to ethanol stimulation and generates resilience to ethanol sedation. These mice also display higher preference and escalation of ethanol drinking, which continues despite adverse outcomes. We find that striatal D1R activation is required for ethanol stimulation and that this signaling is enhanced in mice with low striatal D2Rs. These data demonstrate a link between two vulnerability factors for alcohol abuse and offer evidence for a mechanism in which low striatal D2Rs trigger D1R hypersensitivity, ultimately leading to compulsive-like drinking.
Collapse
Affiliation(s)
- Miriam E Bocarsly
- Laboratory on the Neurobiology of Compulsive Behaviors, NIAAA, NIH, Bethesda, MD, USA; NIGMS, IRP, NIH, Bethesda, MD, USA
| | | | - Vanessa Kolb
- Laboratory on the Neurobiology of Compulsive Behaviors, NIAAA, NIH, Bethesda, MD, USA
| | | | - Sannidhi Shashikiran
- Laboratory on the Neurobiology of Compulsive Behaviors, NIAAA, NIH, Bethesda, MD, USA
| | - Marcelo Rubinstein
- INGEBI, CONICET, and FCEN, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - David R Sibley
- Molecular Neuropharmacology Section, NINDS, IRP, NIH, Bethesda, MD, USA
| | - Lauren K Dobbs
- Laboratory on the Neurobiology of Compulsive Behaviors, NIAAA, NIH, Bethesda, MD, USA; Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, USA
| | - Veronica A Alvarez
- Laboratory on the Neurobiology of Compulsive Behaviors, NIAAA, NIH, Bethesda, MD, USA; Center on Compulsive Behaviors, IRP, NIH, Bethesda, MD, USA; NIDA, IRP, NIH, Bethesda, MD, USA.
| |
Collapse
|
38
|
Gangarossa G, Castell L, Castro L, Tarot P, Veyrunes F, Vincent P, Bertaso F, Valjent E. Contrasting patterns of ERK activation in the tail of the striatum in response to aversive and rewarding signals. J Neurochem 2019; 151:204-226. [PMID: 31245856 DOI: 10.1111/jnc.14804] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/13/2019] [Accepted: 06/19/2019] [Indexed: 01/08/2023]
Abstract
The caudal part of the striatum, also named the tail of the striatum (TS), defines a fourth striatal domain. Determining whether rewarding, aversive and salient stimuli regulate the activity of striatal spiny projections neurons (SPNs) of the TS is therefore of paramount importance to understand its functions, which remain largely elusive. Taking advantage of genetically encoded biosensors (A-kinase activity reporter 3) to record protein kinase A signals and by analyzing the distribution of dopamine D1R- and D2R-SPNs in the TS, we characterized three subterritories: a D2R/A2aR-lacking, a D1R/D2R-intermingled and a D1R/D2R-SPNs-enriched area (corresponding to the amygdalostriatal transition). In addition, we provide evidence that the distribution of D1R- and D2R-SPNs in the TS is evolutionarily conserved (mouse, rat, gerbil). The in vivo analysis of extracellular signal-regulated kinase (ERK) phosphorylation in these TS subterritories in response to distinct appetitive, aversive and pharmacological stimuli revealed that SPNs of the TS are not recruited by stimuli triggering innate aversive responses, fasting, satiety, or palatable signals whereas a reduction in ERK phosphorylation occurred following learned avoidance. In contrast, D1R-SPNs of the intermingled and D2R/A2aR-lacking areas were strongly activated by both D1R agonists and psychostimulant drugs (d-amphetamine, cocaine, 3,4-methyl enedioxy methamphetamine, or methylphenidate), but not by hallucinogens. Finally, a similar pattern of ERK activation was observed by blocking selectively dopamine reuptake. Together, our results reveal that the caudal TS might participate in the processing of specific reward signals and discrete aversive stimuli. Cover Image for this issue: doi: 10.1111/jnc.14526. Open Science: This manuscript was awarded with the Open Materials Badge For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.,Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | - Laia Castell
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Liliana Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Pauline Tarot
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frederic Veyrunes
- Institut des Sciences de l'Evolution de Montpellier, ISEM, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France
| | - Federica Bertaso
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| |
Collapse
|
39
|
Ahmed MR, Jayakumar M, Ahmed MS, Zamaleeva AI, Tao J, Li EH, Job JK, Pittenger C, Ohtsu H, Rajadas J. Pharmacological antagonism of histamine H2R ameliorated L-DOPA–induced dyskinesia via normalization of GRK3 and by suppressing FosB and ERK in PD. Neurobiol Aging 2019; 81:177-189. [DOI: 10.1016/j.neurobiolaging.2019.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023]
|
40
|
Methamphetamine exacerbates neuroinflammatory response to lipopolysaccharide by activating dopamine D1-like receptors. Int Immunopharmacol 2019; 73:1-9. [DOI: 10.1016/j.intimp.2019.04.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/03/2019] [Accepted: 04/25/2019] [Indexed: 01/11/2023]
|
41
|
Koch ET, Raymond LA. Dysfunctional striatal dopamine signaling in Huntington's disease. J Neurosci Res 2019; 97:1636-1654. [PMID: 31304622 DOI: 10.1002/jnr.24495] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/06/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022]
Abstract
Dopamine signaling in the striatum is critical for a variety of behaviors including movement, behavioral flexibility, response to reward and many forms of learning. Alterations to dopamine transmission contribute to pathological features of many neurological diseases, including Huntington's disease (HD). HD is an autosomal dominant genetic disorder caused by a CAG repeat expansion in the Huntingtin gene. The striatum is preferentially degenerated in HD, and this region receives dopaminergic input from the substantia nigra. Studies of HD patients and genetic rodent models have shown changes to levels of dopamine and its receptors in the striatum, and alterations in dopamine receptor signaling and modulation of other neurotransmitters, notably glutamate. Throughout his career, Dr. Michael Levine's research has furthered our understanding of dopamine signaling in the striatum of healthy rodents and HD mouse models. This review will focus on the work of his group and others in elucidating alterations to striatal dopamine signaling that contribute to pathophysiology in HD mouse models, and how these findings relate to human HD studies. We will also discuss current and potential therapeutic interventions for HD that target the dopamine system, and future research directions for this field.
Collapse
Affiliation(s)
- Ellen T Koch
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
42
|
Pagliaroli L, Widomska J, Nespoli E, Hildebrandt T, Barta C, Glennon J, Hengerer B, Poelmans G. Riluzole Attenuates L-DOPA-Induced Abnormal Involuntary Movements Through Decreasing CREB1 Activity: Insights from a Rat Model. Mol Neurobiol 2019; 56:5111-5121. [PMID: 30484112 PMCID: PMC6647536 DOI: 10.1007/s12035-018-1433-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/15/2018] [Indexed: 12/17/2022]
Abstract
Chronic administration of L-DOPA, the first-line treatment of dystonic symptoms in childhood or in Parkinson's disease, often leads to the development of abnormal involuntary movements (AIMs), which represent an important clinical problem. Although it is known that Riluzole attenuates L-DOPA-induced AIMs, the molecular mechanisms underlying this effect are not understood. Therefore, we studied the behavior and performed RNA sequencing of the striatum in three groups of rats that all received a unilateral lesion with 6-hydroxydopamine in their medial forebrain bundle, followed by the administration of saline, L-DOPA, or L-DOPA combined with Riluzole. First, we provide evidence that Riluzole attenuates AIMs in this rat model. Subsequently, analysis of the transcriptomics data revealed that Riluzole is predicted to reduce the activity of CREB1, a transcription factor that regulates the expression of multiple proteins that interact in a molecular landscape involved in apoptosis. Although this mechanism underlying the beneficial effect of Riluzole on AIMs needs to be confirmed, it provides clues towards novel or existing compounds for the treatment of AIMs that modulate the activity of CREB1 and, hence, its downstream targets.
Collapse
Affiliation(s)
- Luca Pagliaroli
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Joanna Widomska
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ester Nespoli
- CNS Department, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- Department of Child and Adolescent Psychiatry/Psychotherapy, University of Ulm, Ulm, Germany
| | - Tobias Hildebrandt
- Target Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Csaba Barta
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Jeffrey Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastian Hengerer
- CNS Department, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Geert Poelmans
- Department of Human Genetics, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
43
|
Mariani LL, Longueville S, Girault JA, Hervé D, Gervasi N. Differential enhancement of ERK, PKA and Ca 2+ signaling in direct and indirect striatal neurons of Parkinsonian mice. Neurobiol Dis 2019; 130:104506. [PMID: 31220556 DOI: 10.1016/j.nbd.2019.104506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/06/2019] [Accepted: 06/13/2019] [Indexed: 12/29/2022] Open
Abstract
Parkinson's disease (PD) is characterized by severe locomotor deficits due to the disappearance of dopamine (DA) from the dorsal striatum. The development of PD symptoms and treatment-related complications such as dyskinesia have been proposed to result from complex alterations in intracellular signaling in both direct and indirect pathway striatal projection neurons (dSPNs and iSPNs, respectively) following loss of DA afferents. To identify cell-specific and dynamical modifications of signaling pathways associated with PD, we used a hemiparkinsonian mouse model with 6-hydroxydopamine (6-OHDA) lesion combined with two-photon fluorescence biosensors imaging in adult corticostriatal slices. After DA lesion, extracellular signal-regulated kinase (ERK) activation was increased in response to DA D1 receptor (D1R) or α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) stimulation. The cAMP-dependent protein kinase (PKA) pathway contributing to ERK activation displayed supersensitive responses to D1R stimulation after 6-OHDA lesion. This cAMP/PKA supersensitivity was specific of D1R-responding SPNs and resulted from Gαolf upregulation and deficient phosphodiesterase activity. In lesioned striatum, the number of D1R-SPNs with spontaneous Ca2+ transients augmented while Ca2+ response to AMPA receptor stimulation specifically increased in iSPNs. Our work reveals distinct cell type-specific signaling alterations in the striatum after DA denervation. It suggests that over-activation of ERK pathway, observed in PD striatum, known to contribute to dyskinesia, may be linked to the combined dysregulation of DA and glutamate signaling pathways in the two populations of SPNs. These findings bring new insights into the implication of these respective neuronal populations in PD motor symptoms and the occurrence of PD treatment complications.
Collapse
Affiliation(s)
- Louise-Laure Mariani
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Sophie Longueville
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Denis Hervé
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France.
| | - Nicolas Gervasi
- Inserm UMR-S 1270, Paris, France; Sorbonne Université, Science and Engineering Faculty, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
44
|
Biased G Protein-Independent Signaling of Dopamine D 1-D 3 Receptor Heteromers in the Nucleus Accumbens. Mol Neurobiol 2019; 56:6756-6769. [PMID: 30919214 DOI: 10.1007/s12035-019-1564-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/13/2019] [Indexed: 10/27/2022]
Abstract
Several studies found in vitro evidence for heteromerization of dopamine D1 receptors (D1R) and D3 receptors (D3R), and it has been postulated that functional D1R-D3R heteromers that are normally present in the ventral striatum mediate synergistic locomotor-activating effects of D1R and D3R agonists in rodents. Based also on results obtained in vitro, with mammalian transfected cells, it has been hypothesized that those behavioral effects depend on a D1R-D3R heteromer-mediated G protein-independent signaling. Here, we demonstrate the presence on D1R-D3R heteromers in the mouse ventral striatum by using a synthetic peptide that selectively destabilizes D1R-D3R heteromers. Parallel locomotor activity and ex vivo experiments in reserpinized mice and in vitro experiments in D1R-D3R mammalian transfected cells were performed to dissect the signaling mechanisms of D1R-D3R heteromers. Co-administration of D1R and D3R agonists in reserpinized mice produced synergistic locomotor activation and a selective synergistic AKT phosphorylation in the most ventromedial region of the striatum in the shell of the nucleus accumbens. Application of the destabilizing peptide in transfected cells and in the shell of the nucleus accumbens allowed demonstrating that both in vitro and in vivo co-activation of D3R induces a switch from G protein-dependent to G protein-independent D1R-mediated signaling determined by D1R-D3R heteromerization. The results therefore demonstrate that a biased G protein-independent signaling of D1R-D3R heteromers localized in the shell of the nucleus accumbens mediate the locomotor synergistic effects of D1R and D3R agonists in reserpinized mice.
Collapse
|
45
|
Dobbs LK, Kaplan AR, Bock R, Phamluong K, Shin JH, Bocarsly ME, Eberhart L, Ron D, Alvarez VA. D1 receptor hypersensitivity in mice with low striatal D2 receptors facilitates select cocaine behaviors. Neuropsychopharmacology 2019; 44:805-816. [PMID: 30504927 PMCID: PMC6372593 DOI: 10.1038/s41386-018-0286-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/16/2018] [Accepted: 11/23/2018] [Indexed: 11/17/2022]
Abstract
Vulnerability for cocaine abuse in humans is associated with low dopamine D2 receptor (D2R) availability in the striatum. The mechanisms driving this vulnerability are poorly understood. In this study, we found that downregulating D2R expression selectively in striatal indirect-pathway neurons triggers a multitude of changes in D1 receptor (D1R)-expressing direct-pathway neurons, which comprise the other main subpopulation of striatal projection neurons. These changes include a leftward shift in the dose-response to a D1-like agonist that indicates a behavioral D1R hypersensitivity, a shift from PKA to ERK intracellular signaling cascades upon D1R activation, and a reduction in the density of bridging collaterals from D1R-expressing neurons to pallidal areas. We hypothesize that the D1R hypersensitivity underlies abuse vulnerability by facilitating the behavioral responses to repeated cocaine, such as locomotor sensitization and drug self-administration. We found evidence that littermate control mice develop D1R hypersensitivity after they are sensitized to cocaine. Indeed, D1-like agonist and cocaine cross-sensitize in control littermates and this effect was potentiated in mice lacking striatal D2Rs from indirect-pathway neurons. To our surprise, mice with low striatal D2Rs acquired cocaine self-administration similarly to littermate controls and showed no significant change in motivation to take cocaine but lower seeking. These findings indicate that downregulation of striatal D2Rs triggers D1R hypersensitivity to facilitate cocaine locomotor sensitization, which by itself was not associated with greater cocaine taking or seeking under the conditions tested.
Collapse
Affiliation(s)
- Lauren K Dobbs
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Alanna R Kaplan
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Roland Bock
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - J Hoon Shin
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Miriam E Bocarsly
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
- Postdoctoral Research Associate Program, National Institute of General Medical Sciences, NIH, Bethesda, MD, USA
| | - Lindsay Eberhart
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Veronica A Alvarez
- Laboratory on the Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, NIH, Bethesda, MD, USA.
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA.
- National Institute on Drug Abuse, Intramural Research Program, NIH, Baltimore, MD, USA.
| |
Collapse
|
46
|
Valjent E, Biever A, Gangarossa G, Puighermanal E. Dopamine signaling in the striatum. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 116:375-396. [PMID: 31036297 DOI: 10.1016/bs.apcsb.2019.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The striatum integrates dopamine-mediated reward signals to generate appropriate behavior in response to glutamate-mediated sensory cues. Such associative learning relies on enduring neural plasticity in striatal GABAergic spiny projection neurons which, when altered, can lead to the development of a wide variety of pathological states. Considerable progress has been made in our understanding of the intracellular signaling mechanisms in dopamine-related behaviors and pathologies. Through the prism of the regulation of histone H3 and ribosomal protein S6 phosphorylation, we review how dopamine-mediated signaling events regulate gene transcription and mRNA translation. Particularly, we focus on the intracellular cascades controlling these phosphorylations downstream of the modulation of dopamine receptors by psychostimulants, antipsychotics and l-DOPA. Finally, we highlight the importance to precisely determine in which neuronal populations these signaling events occur in order to understand how they participate in remodeling neural circuits and altering dopamine-related behaviors.
Collapse
Affiliation(s)
- Emmanuel Valjent
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.
| | - Anne Biever
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Giuseppe Gangarossa
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Emma Puighermanal
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
47
|
Rai SN, Dilnashin H, Birla H, Singh SS, Zahra W, Rathore AS, Singh BK, Singh SP. The Role of PI3K/Akt and ERK in Neurodegenerative Disorders. Neurotox Res 2019; 35:775-795. [PMID: 30707354 DOI: 10.1007/s12640-019-0003-y] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/05/2019] [Accepted: 01/15/2019] [Indexed: 12/27/2022]
Abstract
Disruption of Akt and Erk-mediated signal transduction significantly contributes in the pathogenesis of various neurodegenerative diseases (NDs), such as Parkinson's disease, Alzheimer's diseases, Huntington's disease, and many others. These regulatory proteins serve as the regulator of cell survival, motility, transcription, metabolism, and progression of the cell cycle. Therefore, targeting Akt and Erk pathway has been proposed as a reasonable approach to suppress ND progression. This review has emphasized on involvement of Akt/Erk cascade in the neurodegeneration. Akt has been reported to regulate neuronal toxicity through its various substrates like FOXos, GSK3β, and caspase-9 etc. Akt is also involved with PI3K in signaling pathway to mediate neuronal survival. ERK is another kinase which also regulates proliferation, differentiation, and survival of the neural cell. There has also been much progress in developing a therapeutic molecule targeting Akt and Erk signaling. Therefore, improved understanding of the molecular mechanism behind the regulatory aspect of Akt and Erk networks can make strong impact on exploration of the neurodegenerative disease pathogenesis.
Collapse
Key Words
- 6-OHDA, 6-hydroxydopamine
- BDNF, brain-derived neurotrophic factor
- HD, Huntington disease
- MAPK, mitogen-activated protein-extracellular kinase
- MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- NDs, neurodegenerative disorders
- Nrf2, nuclear factor erythroid 2 p45-related factor 2
- PD, Parkinson’s disease
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Hagera Dilnashin
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Hareram Birla
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Saumitra Sen Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Walia Zahra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Aaina Singh Rathore
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Brijesh Kumar Singh
- Department of Pathology and Cell Biology, Columbia University Medical Centre, Columbia University, New York, NY, 10032, USA
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
48
|
Abstract
β-Arrestin function has largely been investigated in cell culture-based systems. Here we describe methods to investigate β-arrestin2 signaling in vivo by developing novel mouse models and viral methods to overexpress β-arrestin2 in the mouse brain. The methods and concepts described here are in the context of Parkinson's disease (PD) and developing automated in vivo drug screening platforms.
Collapse
Affiliation(s)
- Nikhil M Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
49
|
Genetic enhancement of Ras-ERK pathway does not aggravate L-DOPA-induced dyskinesia in mice but prevents the decrease induced by lovastatin. Sci Rep 2018; 8:15381. [PMID: 30337665 PMCID: PMC6194127 DOI: 10.1038/s41598-018-33713-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/03/2018] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence supports a close relationship between Ras-ERK1/2 activation in the striatum and L-DOPA-induced dyskinesia (LID). ERK1/2 activation by L-DOPA takes place through the crosstalk between D1R/AC/PKA/DARPP-32 pathway and NMDA/Ras pathway. Compelling genetic and pharmacological evidence indicates that Ras-ERK1/2 inhibition prevents LID onset and may even revert already established dyskinetic symptoms. However, it is currently unclear whether exacerbation of Ras-ERK1/2 activity in the striatum may further aggravate dyskinesia in experimental animal models. Here we took advantage of two genetic models in which Ras-ERK1/2 signaling is hyperactivated, the Nf1+/− mice, in which the Ras inhibitor neurofibromin is reduced, and the Ras-GRF1 overexpressing (Ras-GRF1 OE) transgenic mice in which a specific neuronal activator of Ras is enhanced. Nf1+/− and Ras-GRF1 OE mice were unilaterally lesioned with 6-OHDA and treated with an escalating L-DOPA dosing regimen. In addition, a subset of Nf1+/− hemi-parkinsonian animals was also co-treated with the Ras inhibitor lovastatin. Our results revealed that Nf1+/− and Ras-GRF1 OE mice displayed similar dyskinetic symptoms to their wild-type counterparts. This observation was confirmed by the lack of differences between mutant and wild-type mice in striatal molecular changes associated to LID (i.e., FosB, and pERK1/2 expression). Interestingly, attenuation of Ras activity with lovastatin does not weaken dyskinetic symptoms in Nf1+/− mice. Altogether, these data suggest that ERK1/2-signaling activation in dyskinetic animals is maximal and does not require further genetic enhancement in the upstream Ras pathway. However, our data also demonstrate that such a genetic enhancement may reduce the efficacy of anti-dyskinetic drugs like lovastatin.
Collapse
|
50
|
Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4982453. [PMID: 30140365 PMCID: PMC6081569 DOI: 10.1155/2018/4982453] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/10/2018] [Indexed: 12/21/2022]
Abstract
Methamphetamine is a widely abused drug, which possesses neurotoxic activity and powerful addictive effects. Understanding methamphetamine toxicity is key beyond the field of drug abuse since it allows getting an insight into the molecular mechanisms which operate in a variety of neuropsychiatric disorders. In fact, key alterations produced by methamphetamine involve dopamine neurotransmission in a way, which is reminiscent of spontaneous neurodegeneration and psychiatric schizophrenia. Thus, understanding the molecular mechanisms operated by methamphetamine represents a wide window to understand both the addicted brain and a variety of neuropsychiatric disorders. This overlapping, which is already present when looking at the molecular and cellular events promoted immediately after methamphetamine intake, becomes impressive when plastic changes induced in the brain of methamphetamine-addicted patients are considered. Thus, the present manuscript is an attempt to encompass all the molecular events starting at the presynaptic dopamine terminals to reach the nucleus of postsynaptic neurons to explain how specific neurotransmitters and signaling cascades produce persistent genetic modifications, which shift neuronal phenotype and induce behavioral alterations. A special emphasis is posed on disclosing those early and delayed molecular events, which translate an altered neurotransmitter function into epigenetic events, which are derived from the translation of postsynaptic noncanonical signaling into altered gene regulation. All epigenetic effects are considered in light of their persistent changes induced in the postsynaptic neurons including sensitization and desensitization, priming, and shift of neuronal phenotype.
Collapse
|