1
|
Marchionini DM, De Lombaerde S, van Rijswijk J, Zajicek F, Everix L, Miranda A, Aaltonen MJ, Kluger CM, Wild T, Kakoulidou A, Gundelach J, Fieblinger T, Fentz J, Rosinski J, Obenauer J, Greene JR, Liu L, Munoz-Sanjuan I, Verhoye M, Verhaeghe J, Bard J, Staelens S, Bertoglio D. Pharmacodynamic biomarkers responsive to mutant huntingtin lowering in a Huntington's disease mouse model. Neurobiol Dis 2025; 209:106906. [PMID: 40204170 DOI: 10.1016/j.nbd.2025.106906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/20/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025] Open
Affiliation(s)
- Deanna M Marchionini
- CHDI Management, Inc., The Company That Manages the Scientific Activities of CHDI Foundation, Inc., 350 7(th) Ave, Suite 200, New York, NY, 10001, USA.
| | - Stef De Lombaerde
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Campus Drie Eiken, Room D.UC.059, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Joëlle van Rijswijk
- Bio-Imaging Lab, University of Antwerp, Campus Drie Eiken, Building UC, Room 111, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Franziska Zajicek
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Campus Drie Eiken, Room D.UC.059, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Liesbeth Everix
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Campus Drie Eiken, Room D.UC.059, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Campus Drie Eiken, Room D.UC.059, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Mari J Aaltonen
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Carleen M Kluger
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Thomas Wild
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Aglaia Kakoulidou
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Jannis Gundelach
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Tim Fieblinger
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Joachim Fentz
- Evotec International GmbH, Anna-Sigmund-Str. 5, 82061, Neuried, Germany
| | - Jim Rosinski
- CHDI Management, Inc., The Company That Manages the Scientific Activities of CHDI Foundation, Inc., 350 7(th) Ave, Suite 200, New York, NY, 10001, USA
| | - John Obenauer
- Rancho Biosciences, 16955 Via Del Campo, Suite 200, San Diego, CA, 92127, USA
| | - Jonathan R Greene
- Rancho Biosciences, 16955 Via Del Campo, Suite 200, San Diego, CA, 92127, USA
| | - Longbin Liu
- CHDI Management, Inc., The Company That Manages the Scientific Activities of CHDI Foundation, Inc., 350 7(th) Ave, Suite 200, New York, NY, 10001, USA
| | - Ignacio Munoz-Sanjuan
- CHDI Management, Inc., The Company That Manages the Scientific Activities of CHDI Foundation, Inc., 350 7(th) Ave, Suite 200, New York, NY, 10001, USA
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Campus Drie Eiken, Building UC, Room 111, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Jeroen Verhaeghe
- Bio-Imaging Lab, University of Antwerp, Campus Drie Eiken, Building UC, Room 111, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Jonathan Bard
- CHDI Management, Inc., The Company That Manages the Scientific Activities of CHDI Foundation, Inc., 350 7(th) Ave, Suite 200, New York, NY, 10001, USA
| | - Steven Staelens
- Bio-Imaging Lab, University of Antwerp, Campus Drie Eiken, Building UC, Room 111, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Campus Drie Eiken, Room D.UC.059, Universiteitsplein 1, B-2610 Wilrijk, Belgium; Bio-Imaging Lab, University of Antwerp, Campus Drie Eiken, Building UC, Room 111, Universiteitsplein 1, B-2610 Wilrijk, Belgium; μNeuro Centre of Excellence, University of Antwerp, Campus Drie Eiken, Building N, D.N. 110, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| |
Collapse
|
2
|
Hamilos AE, Wijsman IC, Ding Q, Assawaphadungsit P, Ozcan Z, Assad JA. A mechanism linking dopamine's roles in reinforcement, movement and motivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647288. [PMID: 40236124 PMCID: PMC11996583 DOI: 10.1101/2025.04.04.647288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Dopamine neurons (DANs) play seemingly distinct roles in reinforcement, 1-3 motivation, 4,5 and movement, 6,7 and DA-modulating therapies relieve symptoms across a puzzling spectrum of neurologic and psychiatric symptoms. 8 Yet, the mechanistic relationship among these roles is unknown. Here, we show DA's tripartite roles are causally linked by a process in which phasic striatal DA rapidly and persistently recalibrates the propensity to move, a measure of vigor. Using a self-timed movement task, we found that single exposures to reward-related DA transients (both endogenous and exogenously-induced) exerted one-shot updates to movement timing-but in a surprising fashion. Rather than reinforce specific movement times, DA transients quantitatively changed movement timing on the next trial, with larger transients leading to earlier movements (and smaller to later), consistent with a stochastic search process that calibrates the frequency of movement. Both abrupt and gradual changes in external and internal contingencies-such as timing criterion, reward content, and satiety state-caused changes to the amplitude of DA transients that causally altered movement timing. The rapidity and bidirectionality of the one-shot effects are difficult to reconcile with gradual synaptic plasticity, and instead point to more flexible cellular mechanisms, such as DA-dependent modulation of neuronal excitability. Our findings shed light on how natural reinforcement, as well as DA-related disorders such as Parkinson's disease, could affect behavioral vigor.
Collapse
|
3
|
Cheng J, Koch ET, Ramandi D, Mackay JP, O'Leary TP, Rees-Jones W, Raymond LA. Synaptic modulation of glutamate in striatum of the YAC128 mouse model of Huntington disease. Neurobiol Dis 2025; 205:106774. [PMID: 39716682 DOI: 10.1016/j.nbd.2024.106774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Altered balance between striatal direct and indirect pathways contributes to early motor, cognitive and psychiatric symptoms in Huntington disease (HD). While degeneration of striatal D2-type dopamine receptor (D2)-expressing indirect pathway medium spiny neurons (iMSNs) occurs prior to that of D1-type dopamine receptor (D1)-expressing direct pathway neurons, altered corticostriatal synaptic function precedes degeneration. D2-mediated signaling on iMSNs reduces their excitability and promotes endocannabinoid (eCB) synthesis, suppressing glutamate release from cortical afferents. D2 receptors are also expressed on glutamatergic cortical terminals, cholinergic interneurons, and dopaminergic terminals from substantia nigra where they suppress release of glutamate, acetylcholine and dopamine, respectively, and these cell types may contribute to early striatal dysfunction in HD. Thus, we used corticostriatal brain slices and optogenetic probes to directly investigate neuromodulatory signaling in the transgenic YAC128 HD mouse model. RESULTS Low-dose D2 agonist quinpirole reduced cortically-evoked glutamate release in dorsal striatum of premanifest YAC128 slices but not WT, and blocking type 1 cannabinoid receptors mitigated this effect. YAC128 corticostriatal brain slices also showed increased evoked dopamine and reduced evoked eCB release compared to WT, while acetylcholine signaling patterns remained relatively intact. CONCLUSIONS These findings suggest that YAC128 corticostriatal slices show increased D2 sensitivity that is eCB-dependent, and that dopamine and eCB release are altered at an early disease stage. We provide evidence for impaired neuromodulatory signaling in early HD, guiding therapeutic efforts prior to the onset of overt motor symptoms later on.
Collapse
Affiliation(s)
- Judy Cheng
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Ellen T Koch
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Daniel Ramandi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Cell and Developmental Biology, University of British Columbia, Vancouver, BC, Canada
| | - James P Mackay
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Timothy P O'Leary
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - William Rees-Jones
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Lynn A Raymond
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Tan AM, Geva M, Goldberg YP, Schuring H, Sanson BJ, Rosser A, Raymond L, Reilmann R, Hayden MR, Anderson K. Antidopaminergic medications in Huntington's disease. J Huntingtons Dis 2025:18796397241304312. [PMID: 39973394 DOI: 10.1177/18796397241304312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder marked by motor, cognitive, and behavioral impairments. Antidopaminergic medications (ADMs), such as VMAT2 inhibitors and antipsychotics, are commonly used to manage HD motor disturbances and behavioral disorders. For patients and caregivers, ADMs are an important tool for managing symptoms that negatively affect daily life. However, the impact of ADM use in HD is not firmly understood due to a lack of robust, systematic studies that assessed their overall effect on HD disease. A mounting body of evidence suggests these medications may be associated with worse clinical measures of cognitive function and functional impairment. While regulatory guidelines highlight adverse effects like sedation, cognitive dysfunction, and extrapyramidal symptoms, it is unclear whether ADMs directly impact disease progression or if the side effects mimic or exacerbate measures of HD symptoms in clinical trials. Given ADM effects on the central nervous system and biological uncertainty within HD outcomes, clinical trial designs should recognize the impact of ADMs on key outcomes, as measured by acceptable scales including Total Functional Capacity, Stoop Word Reading, Symbol Digit Modality Test, and the composite Unified Huntington's Disease Rating Scale. The development of novel HD interventions requires consideration of concomitant ADM use that may influence measures of disease presentation. In this review, we highlight the role of ADMs in HD management, their symptomatic benefits and potential risks, especially with high dose associated side effects, interactions with CYP2D6 inhibitors, and the individualized need for careful dose monitoring for clinical care and trial design.
Collapse
Affiliation(s)
- Andrew M Tan
- Prilenia Therapeutics B.V., Naarden, The Netherlands
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Michal Geva
- Prilenia Therapeutics B.V., Naarden, The Netherlands
| | | | - Henk Schuring
- Prilenia Therapeutics B.V., Naarden, The Netherlands
| | | | - Anne Rosser
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- School of Biosciences Life Sciences Building, Cardiff University Brain Repair Group, Cardiff, UK
- Advanced Neurotherapeutics Centre, Neuroscience and Mental Health Innovation Institute, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Lynn Raymond
- Departments of Psychiatry and Medicine, University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, Canada
| | - Ralf Reilmann
- Section for Neurodegenerative Diseases, Department of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
- Institute of Clinical Radiology, University of Münster, Germany
| | - Michael R Hayden
- Prilenia Therapeutics B.V., Naarden, The Netherlands
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Karen Anderson
- Department of Psychiatry and Department of Neurology, Georgetown University, Washington, DC, USA
| |
Collapse
|
5
|
Wei F, Liu H, Zhang W, Wang J, Zhang Y. Drug inhibition and substrate transport mechanisms of human VMAT2. Nat Commun 2025; 16:323. [PMID: 39747030 PMCID: PMC11695631 DOI: 10.1038/s41467-024-55361-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Vesicular monoamine transporter 2 (VMAT2) is crucial for packaging monoamine neurotransmitters into synaptic vesicles, with their dysregulation linked to schizophrenia, mood disorders, and Parkinson's disease. Tetrabenazine (TBZ) and valbenazine (VBZ), both FDA-approved VMAT2 inhibitors, are employed to treat chorea and tardive dyskinesia (TD). Our study presents the structures of VMAT2 bound to substrates serotonin (5-HT) and dopamine (DA), as well as the inhibitors TBZ and VBZ. Utilizing cryo-electron microscopy (cryo-EM), mutagenesis functional assays, and molecular dynamics (MD) simulations, we elucidate the mechanisms of substrate transport and drug inhibition. Our MD simulations indicate potential binding poses of substrate (5-HT) in both cytosol-facing and lumen-facing states, emphasizing the significance of protonation of key acidic residues for substrate release. We demonstrate that TBZ locks VMAT2 in a lumen-facing occluded state, while VBZ stabilizes it in a lumen-facing conformation. These insights enhance our understanding of VMAT2 function and provide valuable insights for the development of novel therapeutic strategies for psychiatric disorders.
Collapse
Affiliation(s)
- Feiwen Wei
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Huihui Liu
- Arieh Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Wei Zhang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jufang Wang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanqing Zhang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Eddy CM. Basal ganglia contributions to social cognition: evidence from movement disorders. Cogn Neuropsychiatry 2025; 30:1-14. [PMID: 40213824 DOI: 10.1080/13546805.2025.2490054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/02/2025] [Indexed: 04/27/2025]
Abstract
Introduction and method: Over the last few decades many studies have demonstrated that various populations with movement disorder exhibit a variety of difficulties with social cognition. This brief review paper summarises the major findings of social cognitive research conducted in various hypo- and hyper-kinetic movement disorders (Parkinson's disease, Huntington's disease and Tourette syndrome), with a focus on findings that may shed new light on the functions of the basal ganglia.Results: Task impairments include more predictable difficulties with embodied processes involving the motor system, such as problems with facial expressions and other non-verbal gestures. The cognitive and emotional changes associated with broader frontostriatal dysfunction may also explain some deficits, particularly difficulties with verbal tasks or perspective taking, with impacts varying according to the type of disorder and disease stage. Most intriguingly, transdiagnostic comparisons reveal patterns implying that the role of the basal ganglia in social cognition may be more important than previously recognised.Conclusions: Movement disorders show remarkable overlap in terms of atypical intention attribution across various tasks, as well as relationships between social cognition and behavioural or motivational symptoms (e.g., apathy; anhedonia; impulsivity). The possible mechanisms underlying these similarities are explored with a view to guiding future research.
Collapse
Affiliation(s)
- Clare M Eddy
- College of Medicine and Health, University of Birmingham, Birmingham, UK
- BSMHFT Research and Development, National Centre for Mental Health, Birmingham, UK
| |
Collapse
|
7
|
Niebrügge N, Trovato O, Praschberger R, Lieb A. Disease-Associated Dopamine Receptor D2 Variants Exhibit Functional Consequences Depending on Different Heterotrimeric G-Protein Subunit Combinations. Biomedicines 2024; 13:46. [PMID: 39857630 PMCID: PMC11761627 DOI: 10.3390/biomedicines13010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Dopamine receptors (DRs) are G-protein-coupled receptors (GPCRs) found in the central nervous system (CNS). DRs are essential for mediating various downstream signaling cascades and play a critical role in regulating the dopaminergic nigrostriatal pathway, which is involved in motor control. Recently, mutations in DRD2 (WT), p.Ile212Phe (I212F), and p.Met345Arg (M345R) have been associated with hyperkinetic movement disorders and shown to alter heterotrimeric G-protein complex signaling and β-arrestin recruitment. Methods: To conduct a detailed investigation of the I212F and M345R functional phenotypes, we used the TRansdUcer PATHway (TRUPATH) assay to study heterotrimeric G-protein recruitment and the Parallel Receptorome Expression and Screening via Transcriptional Output (PRESTO-Tango) assay to evaluate transcriptional activation following arrestin translocation for β-arrestin recruitment. Results: In our study, we could confirm the reported mutant's loss-of-function phenotype in β-arrestin 2 recruitment (reduced agonist potency and decreased maximal signaling efficacy in comparison to the WT). However, a detailed analysis of basal/constitutive activity also revealed a gain-of-function phenotype for mutant M345R. For a more comprehensive investigation of heterotrimeric G-protein complex signaling, we investigated the impact of WT mutants in combination with (i) a specifically suggested assay, and (ii) the most abundantly expressed heterotrimeric G-protein complex combinations in WT receptor-enriched regions. We were able to confirm the reported gain-of-function phenotype by Rodriguez-Contreras et al. and extend it by the use of the most abundant heterotrimeric G-protein subunits, GαoA and Gαi1, β1 and β2, and γ3 and γ7, in mouse and human basal ganglia. Conclusions: Although our results indicate that the interaction of the two variants with the most highly expressed heterotrimeric G-protein complex subunit combinations also results in a gain-of-function phenotype, they also clearly demonstrate that the phenotype can be significantly altered, dependent on heterotrimeric G-protein complex expression.
Collapse
Affiliation(s)
- Nele Niebrügge
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Olga Trovato
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Roman Praschberger
- Institute of Human Genetic, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Andreas Lieb
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
8
|
Tod P, Varga A, Román V, Lendvai B, Pálkovács R, Sperlágh B, Vizi ES. Tetrabenazine, a vesicular monoamine transporter 2 inhibitor, inhibits vesicular storage capacity and release of monoamine transmitters in mouse brain tissue. Br J Pharmacol 2024; 181:5094-5109. [PMID: 39304979 DOI: 10.1111/bph.17348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 07/10/2024] [Accepted: 08/23/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Tetrabenazine (TBZ), used for treating hyperkinetic disorders, inhibits vesicular monoamine transporter-2 (VMAT-2), which sequesters monoamines into vesicles for exocytosis. However, our knowledge of the effect of TBZ on monoaminergic transmission is limited. Herein, we provide neurochemical evidence regarding the effect of VMAT-2 inhibition on vesicular neurotransmitter release from the prefrontal cortex (PFC) and striatum (STR) (brain regions involved in characteristic TBZ treatment side effects). The interaction between TBZ and MDMA was also assessed regarding motor behaviour in mice. EXPERIMENTAL APPROACH Vesicular storage capacity and release of [3H]-noradrenaline ([3H]-NA), [3H]-dopamine ([3H]-DA), [3H]-serotonin ([3H]-5-HT), and [3H]-acetylcholine ([3H]-ACh) was studied in mouse PFC and STR ex vivo slice preparations using electrical field stimulation. Additionally, locomotor activity was assessed in vehicle-treated mice and compared with that of MDMA, TBZ, and co-administered animals (n = 6) using the LABORAS system. KEY RESULTS TBZ lowered the storage capacity and inhibited the vesicular release of [3H]-NA and [3H]-DA from the PFC, and [3H]-DA and [3H]-5-HT from the STR in a concentration-dependent manner. Unlike vesamicol (vesicular ACh uptake inhibitor), TBZ failed to inhibit the vesicular release of [3H]-ACh from the PFC. When the vesicular storage of the investigated monoamines was inhibited by TBZ in the PFC and STR, MDMA induced the release of transmitters through transporter reversal; MDMA dose dependently increased locomotor activity in vivo. CONCLUSION AND IMPLICATIONS Our observations provide neurochemical evidence explaining the mechanism of VMAT-2 inhibitors in the brain and support the involvement of dopaminergic and noradrenergic transmission in hyperkinetic movement disorders.
Collapse
Affiliation(s)
- Pál Tod
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Anita Varga
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Viktor Román
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Balázs Lendvai
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Roland Pálkovács
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - E Sylvester Vizi
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
9
|
Subin JA, Shrestha RLS. Computational Assessment of the Phytochemicals of Panax ginseng C.A. Meyer Against Dopamine Receptor D1 for Early Huntington's Disease Prophylactics. Cell Biochem Biophys 2024; 82:3413-3423. [PMID: 39046621 DOI: 10.1007/s12013-024-01426-2] [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] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
A herb, Panax ginseng C.A. Meyer has been used traditionally for the treatment of various diseases. In this work, its chemical components have been explored by computational methods for the possibility of therapeutic potential against early Huntington's disease. The molecular docking calculations against dopamine receptor D1 (PDB ID: 7X2F) involved in pathogenesis of early Huntington's disease gave the binding affinities (kcal/mol) of schizandrin (-10.530), ergosterol (-10.124), protopanaxadiol (-9.650), panaxydol (-9.399), diphenhydramine (-9.358), and panasenoside (-9.358). The values for native ligand (-7.748) and some selected drugs, Nefazodone (-9.880), Risperidone (-9.752), and Haloperidol (-9.712) were higher revealing weaker interactions. The stability assessment of top protein-ligand adducts in terms of various geometrical and thermodynamical parameters extracted from 200 ns molecular dynamics simulations pointed to schizandrin, protopanaxadiol, and panasenoside as hit molecules. The minimal translational and rotational motion of the docked ligands at orthosteric pocket of the receptor at near physiological conditions hinted at the probability of it restricting or inhibiting over-activation of DRD1. The sustained thermodynamic spontaneity of complex formation reaction augmented the inferences derived from spatial results. The phytochemicals from Panax ginseng could be used in the prophylactics of early Huntington's disease and recommendation is made for further evaluation by experimental work.
Collapse
Affiliation(s)
- Jhashanath Adhikari Subin
- Bioinformatics and Cheminformatics Division, Scientific Research and Training Nepal P. Ltd., Kaushaltar, Bhaktapur, 44800, Nepal
| | - Ram Lal Swagat Shrestha
- Bioinformatics and Cheminformatics Division, Scientific Research and Training Nepal P. Ltd., Kaushaltar, Bhaktapur, 44800, Nepal.
- Department of Chemistry, Amrit Campus, Tribhuvan University, Thamel, Kathmandu, 44600, Nepal.
| |
Collapse
|
10
|
Mora I, Teixidó A, Vázquez-Manrique RP, Puiggròs F, Arola L. Docosahexaenoic Acid (DHA) Supplementation in a Triglyceride Form Prevents from Polyglutamine-Induced Dysfunctions in Caenorhabditis elegans. Int J Mol Sci 2024; 25:12594. [PMID: 39684306 DOI: 10.3390/ijms252312594] [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: 11/05/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
A common hallmark of neurodegenerative diseases is the accumulation of polypeptide aggregates in neurons. Despite the primary cause of these diseases being inherently genetic, their development can be delayed with proper preventive treatments. Long-chain polyunsaturated fatty acids (ω-3 LCPUFA) are promising bioactive nutrients that are beneficial for brain health. In this study, the impact of an oil rich in a structured form of docosahexaenoic acid (DHA) triglyceride (TG) was assessed in a Caenorhabditis elegans model expressing long poly-glutamine (polyQ) chains, which mimics the symptomatology of polyQ-related neurodegenerative diseases such as Huntington's disease (HD), among others. The lifespan, the motility, the number of polyQ aggregates, the oxidative stress resistance, and the cognitive performance associated with sensitive stimuli was measured in mutant nematodes with polyQ aggregates. Overall, DHA-TG at 0.5 µM improved the lifespan, the motility, the oxidative stress resistance, and the cognitive performance of the nematodes, emphasizing the protection against serotonergic synapse dysfunction. Furthermore, the treatment reduced the polyQ aggregates in the nematodes. The data described herein shed light on the connection between DHA and the cognitive performance in neurodegenerative diseases and demonstrated the potential of DHA-TG as nutritional co-adjuvant to prevent the development of polyQ-associated dysfunctions.
Collapse
Affiliation(s)
- Ignasi Mora
- Brudy Technology S.L., 08006 Barcelona, Spain
- Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Alex Teixidó
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health Unit, 43204 Reus, Spain
| | - Rafael P Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Joint Unit for Rare Diseases, Insituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Tarragona, Spain
| | - Lluís Arola
- Nutrigenomics Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| |
Collapse
|
11
|
Bhat AA, Moglad E, Afzal M, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Ali H, Pant K, Singh TG, Dureja H, Singh SK, Dua K, Gupta G, Subramaniyan V. Therapeutic approaches targeting aging and cellular senescence in Huntington's disease. CNS Neurosci Ther 2024; 30:e70053. [PMID: 39428700 PMCID: PMC11491556 DOI: 10.1111/cns.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disease that is manifested by a gradual loss of physical, cognitive, and mental abilities. As the disease advances, age has a major impact on the pathogenic signature of mutant huntingtin (mHTT) protein aggregation. This review aims to explore the intricate relationship between aging, mHTT toxicity, and cellular senescence in HD. Scientific data on the interplay between aging, mHTT, and cellular senescence in HD were collected from several academic databases, including PubMed, Google Scholar, Google, and ScienceDirect. The search terms employed were "AGING," "HUNTINGTON'S DISEASE," "MUTANT HUNTINGTIN," and "CELLULAR SENESCENCE." Additionally, to gather information on the molecular mechanisms and potential therapeutic targets, the search was extended to include relevant terms such as "DNA DAMAGE," "OXIDATIVE STRESS," and "AUTOPHAGY." According to research, aging leads to worsening HD pathophysiology through some processes. As a result of the mHTT accumulation, cellular senescence is promoted, which causes DNA damage, oxidative stress, decreased autophagy, and increased inflammatory responses. Pro-inflammatory cytokines and other substances are released by senescent cells, which may worsen the neuronal damage and the course of the disease. It has been shown that treatments directed at these pathways reduce some of the HD symptoms and enhance longevity in experimental animals, pointing to a new possibility of treating the condition. Through their amplification of the harmful effects of mHTT, aging and cellular senescence play crucial roles in the development of HD. Comprehending these interplays creates novel opportunities for therapeutic measures targeted at alleviating cellular aging and enhancing HD patients' quality of life.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical SciencesUttaranchal UniversityDehradunIndia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of PharmacyPrince Sattam Bin Abdulaziz UniversityAl KharjSaudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy ProgramBatterjee Medical CollegeJeddahSaudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical SciencesUttaranchal UniversityDehradunIndia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of PharmacyUmm Al‐Qura UniversityMakkahSaudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of PharmacyJouf UniversitySakakaAl‐JoufSaudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Department of PharmacologyKyrgyz State Medical CollegeBishkekKyrgyzstan
| | - Kumud Pant
- Graphic Era (Deemed to be University), Dehradun, India
| | | | - Harish Dureja
- Department of Pharmaceutical SciencesMaharshi Dayanand UniversityRohtakIndia
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraPunjabIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of PharmacyChitkara UniversityRajpuraPunjabIndia
- Centre of Medical and Bio‐Allied Health Sciences ResearchAjman UniversityAjmanUnited Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health SciencesMonash UniversityBandar SunwaySelangor Darul EhsanMalaysia
- Department of Medical SciencesSchool of Medical and Life Sciences Sunway UniversityBandar SunwaySelangor Darul EhsanMalaysia
| |
Collapse
|
12
|
Orsini A, Santangelo A, Costagliola G, Scacciati M, Massart F, Operto FF, D'Elios S, Consolini R, De Benedetti F, Maggio MC, Miniaci A, Ferretti A, Cordelli DM, Battini R, Bonuccelli A, Savasta S, Parisi P, Fazzi E, Ruggieri M, Striano P, Peroni DG, Foiadelli T. Management, treatment, and clinical approach of Sydenham's chorea in children: Italian survey on expert-based experience. Eur J Paediatr Neurol 2024; 52:103-108. [PMID: 39226700 DOI: 10.1016/j.ejpn.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
Sydenham's chorea (SC), an autoimmune disorder affecting the central nervous system, is a pivotal diagnostic criterion for acute rheumatic fever. Primarily prevalent in childhood, especially in developing countries, SC manifests with involuntary movements and neuropsychiatric symptoms. Predominantly occurring between ages 5 and 15, with a female bias, SC may recur, particularly during pregnancy or estrogen use. The autoimmune response affecting the basal ganglia, notably against dopamine, underlies the pathophysiology. Clinical management necessitates an integrated approach, potentially involving immunomodulatory therapies. To address discrepancies in SC management, a survey was conducted across Italy, targeting specialists in neurology, pediatrics, child neuropsychiatry, and rheumatology. Of the 51 responding physicians, consensus favored hospitalization for suspected SC, with broad support for laboratory tests and brain MRI. Treatment preferences showed agreement on oral prednisone and IVIG, while opinions varied on duration and plasmapheresis. Haloperidol emerged as the preferred symptomatic therapy. Post-SC penicillin prophylaxis and steroid therapy gained strong support, although opinions differed on duration. Follow-up recommendations included neuropsychological and cardiological assessments. Despite offering valuable insights, broader and more studies are needed in order to guide treatment decisions in this well-known yet challenging complication of acute rheumatic fever, which continues to warrant scientific attention and concerted clinical efforts.
Collapse
Affiliation(s)
- Alessandro Orsini
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - Andrea Santangelo
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.
| | | | | | - Francesco Massart
- Pediatric Endocrinology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | | | - Sofia D'Elios
- Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - Rita Consolini
- Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | | | - Maria Cristina Maggio
- University Department PROMISE "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Angela Miniaci
- Clinica Pediatrica, Policlinico Sant'Orsola Malpighi, Bologna, Italy
| | - Alessandro Ferretti
- UOC di Pediatria, Facoltà di Medicina e Psicologia, Dipartimento NESMOS, Azienda Ospedaliero Universitaria Sant'Andrea, Rome, Italy
| | - Duccio Maria Cordelli
- Unitá Operativa Complessa (UOC) Neuropsichiatria dell'età Pediatrica, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Calambrone, (Pisa), Italy; Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alice Bonuccelli
- Pediatric Neurology, Pediatric Department, AOUP Santa Chiara Hospital, Pisa, Italy
| | - Salvatore Savasta
- Pediatric Clinic and Rare Diseases, "Microcitemico Hospital", Department of Medical Sciences and Public Health University of Cagliari, Cagliari, Italy
| | - Pasquale Parisi
- UOC di Pediatria, Facoltà di Medicina e Psicologia, Dipartimento NESMOS, Azienda Ospedaliero Universitaria Sant'Andrea, Rome, Italy
| | - Elisa Fazzi
- Unit of Child Neurology and Psychiatry, Brescia Civil Hospital, Department of Clinical and Experimental Sciences University of Brescia, Italy
| | - Martino Ruggieri
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, IRCCS Istituto G. Gaslini, Genoa, Italy
| | | | - Thomas Foiadelli
- Clinica Pediatrica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
13
|
Galimberti M, Nucera MR, Bocchi VD, Conforti P, Vezzoli E, Cereda M, Maffezzini C, Iennaco R, Scolz A, Falqui A, Cordiglieri C, Cremona M, Espuny-Camacho I, Faedo A, Felsenfeld DP, Vogt TF, Ranzani V, Zuccato C, Besusso D, Cattaneo E. Huntington's disease cellular phenotypes are rescued non-cell autonomously by healthy cells in mosaic telencephalic organoids. Nat Commun 2024; 15:6534. [PMID: 39095390 PMCID: PMC11297310 DOI: 10.1038/s41467-024-50877-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Huntington's disease (HD) causes selective degeneration of striatal and cortical neurons, resulting in cell mosaicism of coexisting still functional and dysfunctional cells. The impact of non-cell autonomous mechanisms between these cellular states is poorly understood. Here we generated telencephalic organoids with healthy or HD cells, grown separately or as mosaics of the two genotypes. Single-cell RNA sequencing revealed neurodevelopmental abnormalities in the ventral fate acquisition of HD organoids, confirmed by cytoarchitectural and transcriptional defects leading to fewer GABAergic neurons, while dorsal populations showed milder phenotypes mainly in maturation trajectory. Healthy cells in mosaic organoids restored HD cell identity, trajectories, synaptic density, and communication pathways upon cell-cell contact, while showing no significant alterations when grown with HD cells. These findings highlight cell-type-specific alterations in HD and beneficial non-cell autonomous effects of healthy cells, emphasizing the therapeutic potential of modulating cell-cell communication in disease progression and treatment.
Collapse
Affiliation(s)
- Maura Galimberti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Maria R Nucera
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Stem Cell Biology Department; Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Vittoria D Bocchi
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Center for Stem Cell Biology and Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paola Conforti
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Vezzoli
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- ALEMBIC Advanced Light and Electron Microscopy BioImaging Center, San Raffaele Scientific Institute, DIBIT 1, Via Olgettina 58, 20132, Milan, Italy
| | - Matteo Cereda
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Camilla Maffezzini
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Raffaele Iennaco
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Scolz
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Falqui
- Department of Physics "Aldo Pontremoli", University of Milan, Via Celoria 16, 20133, Milan, Italy
| | - Chiara Cordiglieri
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Martina Cremona
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Swiss Stem Cell Foundation, Via Petrini 2, 6900, Lugano, Switzerland
| | - Ira Espuny-Camacho
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- GIGA-Neuroscience, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, 4000, Liège, Belgium
| | - Andrea Faedo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
- Axxam, OpenZone, Via Meucci 3, 20091, Bresso, Milan, Italy
| | | | | | - Valeria Ranzani
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Chiara Zuccato
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Dario Besusso
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elena Cattaneo
- Laboratory of Stem Cell Biology and Pharmacology of Neurodegenerative Diseases, Department of Biosciences, University of Milan, 20122, Milan, Italy.
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy.
| |
Collapse
|
14
|
Giménez S, Millan A, Mora-Morell A, Ayuso N, Gastaldo-Jordán I, Pardo M. Advances in Brain Stimulation, Nanomedicine and the Use of Magnetoelectric Nanoparticles: Dopaminergic Alterations and Their Role in Neurodegeneration and Drug Addiction. Molecules 2024; 29:3580. [PMID: 39124985 PMCID: PMC11314096 DOI: 10.3390/molecules29153580] [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: 06/30/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Recent advancements in brain stimulation and nanomedicine have ushered in a new era of therapeutic interventions for psychiatric and neurodegenerative disorders. This review explores the cutting-edge innovations in brain stimulation techniques, including their applications in alleviating symptoms of main neurodegenerative disorders and addiction. Deep Brain Stimulation (DBS) is an FDA-approved treatment for specific neurodegenerative disorders, including Parkinson's Disease (PD), and is currently under evaluation for other conditions, such as Alzheimer's Disease. This technique has facilitated significant advancements in understanding brain electrical circuitry by enabling targeted brain stimulation and providing insights into neural network function and dysfunction. In reviewing DBS studies, this review places particular emphasis on the underlying main neurotransmitter modifications and their specific brain area location, particularly focusing on the dopaminergic system, which plays a critical role in these conditions. Furthermore, this review delves into the groundbreaking developments in nanomedicine, highlighting how nanotechnology can be utilized to target aberrant signaling in neurodegenerative diseases, with a specific focus on the dopaminergic system. The discussion extends to emerging technologies such as magnetoelectric nanoparticles (MENPs), which represent a novel intersection between nanoformulation and brain stimulation approaches. These innovative technologies offer promising avenues for enhancing the precision and effectiveness of treatments by enabling the non-invasive, targeted delivery of therapeutic agents as well as on-site, on-demand stimulation. By integrating insights from recent research and technological advances, this review aims to provide a comprehensive understanding of how brain stimulation and nanomedicine can be synergistically applied to address complex neuropsychiatric and neurodegenerative disorders, paving the way for future therapeutic strategies.
Collapse
Affiliation(s)
- Silvia Giménez
- Department of Psychobiology, Universidad de Valencia, 46010 Valencia, Spain; (S.G.); (N.A.)
| | - Alexandra Millan
- Department of Neurobiology and Neurophysiology, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Alba Mora-Morell
- Faculty of Biological Sciences, Universidad de Valencia, 46100 Valencia, Spain;
| | - Noa Ayuso
- Department of Psychobiology, Universidad de Valencia, 46010 Valencia, Spain; (S.G.); (N.A.)
| | - Isis Gastaldo-Jordán
- Psychiatry Service, Doctor Peset University Hospital, FISABIO, 46017 Valencia, Spain;
| | - Marta Pardo
- Department of Psychobiology, Universidad de Valencia, 46010 Valencia, Spain; (S.G.); (N.A.)
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), 46022 Valencia, Spain
| |
Collapse
|
15
|
Lauretani F, Giallauria F, Testa C, Zinni C, Lorenzi B, Zucchini I, Salvi M, Napoli R, Maggio MG. Dopamine Pharmacodynamics: New Insights. Int J Mol Sci 2024; 25:5293. [PMID: 38791331 PMCID: PMC11121567 DOI: 10.3390/ijms25105293] [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: 02/25/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Dopamine is a key neurotransmitter involved in physiological processes such as motor control, motivation, reward, cognitive function, and maternal and reproductive behaviors. Therefore, dysfunctions of the dopaminergic system are related to a plethora of human diseases. Dopamine, via different circuitries implicated in compulsive behavior, reward, and habit formation, also represents a key player in substance use disorder and the formation and perpetuation of mechanisms leading to addiction. Here, we propose dopamine as a model not only of neurotransmission but also of neuromodulation capable of modifying neuronal architecture. Abuse of substances like methamphetamine, cocaine, and alcohol and their consumption over time can induce changes in neuronal activities. These modifications lead to synaptic plasticity and finally to morphological and functional changes, starting from maladaptive neuro-modulation and ending in neurodegeneration.
Collapse
Affiliation(s)
- Fulvio Lauretani
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
- Cognitive and Motor Center, Medicine and Geriatric-Rehabilitation Department of Parma, University-Hospital of Parma, 43126 Parma, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, “Federico II” University of Naples, via S. Pansini 5, 80131 Naples, Italy; (F.G.); (R.N.)
| | - Crescenzo Testa
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
| | - Claudia Zinni
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
| | - Beatrice Lorenzi
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
| | - Irene Zucchini
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
| | - Marco Salvi
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
| | - Raffaele Napoli
- Department of Translational Medical Sciences, “Federico II” University of Naples, via S. Pansini 5, 80131 Naples, Italy; (F.G.); (R.N.)
| | - Marcello Giuseppe Maggio
- Geriatric Clinic Unit, Geriatric-Rehabilitation Department, University Hospital, 43126 Parma, Italy; (C.T.); (C.Z.); (B.L.); (I.Z.); (M.S.); (M.G.M.)
- Cognitive and Motor Center, Medicine and Geriatric-Rehabilitation Department of Parma, University-Hospital of Parma, 43126 Parma, Italy
| |
Collapse
|
16
|
Hedreen JC, Berretta S, White III CL. Postmortem neuropathology in early Huntington disease. J Neuropathol Exp Neurol 2024; 83:294-306. [PMID: 38553027 PMCID: PMC11029463 DOI: 10.1093/jnen/nlae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Two aspects of the neuropathology of early Huntington disease (HD) are examined. Neurons of the neostriatum are counted to determine relative loss in striosomes versus matrix at early stages, including for the first time in preclinical cases. An immunohistochemical procedure is described that tentatively distinguishes early HD from HD mimic disorders in postmortem brains. Counts of striatal projection neurons (SPNs) in striosomes defined by calbindin immunohistochemistry versus counts in the surrounding matrix are reported for 8 Vonsattel grade 0 (including 5 premanifest), 8 grade 1, 2 grade 2 HD, and for 8 control postmortem brains. Mean counts of striosome and matrix SPNs were significantly lower in premanifest grade 0 versus controls, with striosome counts significantly lower than matrix. In 8 grade 1 and 2 grade 2 brains, no striosomes with higher SPN counts than in the surrounding matrix were observed. Comparing dorsal versus ventral neostriatum, SPNs in dorsal striosomes and matrix declined more than ventral, making clear the importance of the dorsoventral site of tissue selection for research studies. A characteristic pattern of expanded polyglutamine-immunopositive inclusions was seen in all HD cases. Inclusions were always present in some SPNs and some pontine nucleus neurons and were absent in Purkinje cells, which showed no obvious cell loss.
Collapse
Affiliation(s)
- John C Hedreen
- Harvard Brain Tissue Resource Center, McLean Hospital, Belmont, Massachusetts, USA
| | - Sabina Berretta
- McLean Hospital, Belmont, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Charles L White III
- Neuropathology Section, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
17
|
Elmers J, Colzato LS, Ziemssen F, Ziemssen T, Beste C. Optical coherence tomography as a potential surrogate marker of dopaminergic modulation across the life span. Ageing Res Rev 2024; 96:102280. [PMID: 38518921 DOI: 10.1016/j.arr.2024.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/02/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
The retina has been considered a "window to the brain" and shares similar innervation by the dopaminergic system with the cortex in terms of an unequal distribution of D1 and D2 receptors. Here, we provide a comprehensive overview that Optical Coherence Tomography (OCT), a non-invasive imaging technique, which provides an "in vivo" representation of the retina, shows promise to be used as a surrogate marker of dopaminergic neuromodulation in cognition. Overall, most evidence supports reduced retinal thickness in individuals with dopaminergic dysregulation (e.g., patients with Parkinson's Disease, non-demented older adults) and with poor cognitive functioning. By using the theoretical framework of metacontrol, we derive hypotheses that retinal thinning associated to decreased dopamine (DA) levels affecting D1 families, might lead to a decrease in the signal-to-noise ratio (SNR) affecting cognitive persistence (depending on D1-modulated DA activity) but not cognitive flexibility (depending on D2-modulated DA activity). We argue that the use of OCT parameters might not only be an insightful for cognitive neuroscience research, but also a potentially effective tool for individualized medicine with a focus on cognition. As our society progressively ages in the forthcoming years and decades, the preservation of cognitive abilities and promoting healthy aging will hold of crucial significance. OCT has the potential to function as a swift, non-invasive, and economical method for promptly recognizing individuals with a heightened vulnerability to cognitive deterioration throughout all stages of life.
Collapse
Affiliation(s)
- Julia Elmers
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Lorenza S Colzato
- Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| | - Focke Ziemssen
- Ophthalmological Clinic, University Clinic Leipzig, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Germany; Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China.
| |
Collapse
|
18
|
Hou G, Hao M, Duan J, Han MH. The Formation and Function of the VTA Dopamine System. Int J Mol Sci 2024; 25:3875. [PMID: 38612683 PMCID: PMC11011984 DOI: 10.3390/ijms25073875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The midbrain dopamine system is a sophisticated hub that integrates diverse inputs to control multiple physiological functions, including locomotion, motivation, cognition, reward, as well as maternal and reproductive behaviors. Dopamine is a neurotransmitter that binds to G-protein-coupled receptors. Dopamine also works together with other neurotransmitters and various neuropeptides to maintain the balance of synaptic functions. The dysfunction of the dopamine system leads to several conditions, including Parkinson's disease, Huntington's disease, major depression, schizophrenia, and drug addiction. The ventral tegmental area (VTA) has been identified as an important relay nucleus that modulates homeostatic plasticity in the midbrain dopamine system. Due to the complexity of synaptic transmissions and input-output connections in the VTA, the structure and function of this crucial brain region are still not fully understood. In this review article, we mainly focus on the cell types, neurotransmitters, neuropeptides, ion channels, receptors, and neural circuits of the VTA dopamine system, with the hope of obtaining new insight into the formation and function of this vital brain region.
Collapse
Affiliation(s)
- Guoqiang Hou
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mei Hao
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawen Duan
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ming-Hu Han
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
19
|
Adam H, Gopinath SCB, Arshad MKM, Adam T, Subramaniam S, Hashim U. An Update on Parkinson's Disease and its Neurodegenerative Counterparts. Curr Med Chem 2024; 31:2770-2787. [PMID: 37016529 DOI: 10.2174/0929867330666230403085733] [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: 11/23/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Neurodegenerative disorders are a group of diseases that cause nerve cell degeneration in the brain, resulting in a variety of symptoms and are not treatable with drugs. Parkinson's disease (PD), prion disease, motor neuron disease (MND), Huntington's disease (HD), spinal cerebral dyskinesia (SCA), spinal muscle atrophy (SMA), multiple system atrophy, Alzheimer's disease (AD), spinocerebellar ataxia (SCA) (ALS), pantothenate kinase-related neurodegeneration, and TDP-43 protein disorder are examples of neurodegenerative diseases. Dementia is caused by the loss of brain and spinal cord nerve cells in neurodegenerative diseases. BACKGROUND Even though environmental and genetic predispositions have also been involved in the process, redox metal abuse plays a crucial role in neurodegeneration since the preponderance of symptoms originates from abnormal metal metabolism. METHOD Hence, this review investigates several neurodegenerative diseases that may occur symptoms similar to Parkinson's disease to understand the differences and similarities between Parkinson's disease and other neurodegenerative disorders based on reviewing previously published papers. RESULTS Based on the findings, the aggregation of alpha-synuclein occurs in Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. Other neurodegenerative diseases occur with different protein aggregation or mutations. CONCLUSION We can conclude that Parkinson's disease, Multiple system atrophy, and Dementia with Lewy bodies are closely related. Therefore, researchers must distinguish among the three diseases to avoid misdiagnosis of Multiple System Atrophy and Dementia with Lewy bodies with Parkinson's disease symptoms.
Collapse
Affiliation(s)
- Hussaini Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
| | - Subash C B Gopinath
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600, Arau, Perlis, Malaysia
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600, Arau, Perlis, Malaysia
- Centre for Chemical Biology (CCB), Universiti Sains Malaysia, Bayan Lepas, 11900 Penang, Malaysia
| | - M K Md Arshad
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600 Arau, Perlis, Malaysia
| | - Tijjani Adam
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
- Faculty of Electronic Engineering & Technology, Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600 Arau, Perlis, Malaysia
- Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600, Arau, Perlis, Malaysia
| | - Sreeramanan Subramaniam
- School of Biological Sciences, Universiti Sains Malaysia, Georgetown, 11800 Penang, Malaysia
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600, Arau, Perlis, Malaysia
- Centre for Chemical Biology (CCB), Universiti Sains Malaysia, Bayan Lepas, 11900 Penang, Malaysia
- National Poison Centre, Universiti Sains Malaysia (USM), Georgetown, 11800, Penang, Malaysia
| | - Uda Hashim
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000, Kangar, Perlis, Malaysia
| |
Collapse
|
20
|
Csehi R, Molnar V, Fedor M, Zsumbera V, Palasti A, Acsai K, Grosz Z, Nemeth G, Molnar MJ. The improvement of motor symptoms in Huntington's disease during cariprazine treatment. Orphanet J Rare Dis 2023; 18:375. [PMID: 38041194 PMCID: PMC10690981 DOI: 10.1186/s13023-023-02930-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 09/24/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Huntington's disease (HD) is a progressive neurodegenerative disease, characterised by motor disturbances and non-motor (i.e., psychiatric) symptoms. Motor symptoms are the hallmark features of HD and take many forms. Their emergence is related to alterations in striatal dopaminergic neurotransmission: dopamine levels increase in the early stages of the disease, while more advanced stages are characterised by reduced dopamine levels. Such a biphasic change potentially explains the alterations in motor symptoms: increased dopamine-production induces hyperkinetic movements early in the disease course, while depleted dopamine storage leads to hypokinetic symptoms in the advanced phase. Dopamine D2-D3 partial agonists could be a promising treatment option in HD, as they have the potential to either elevate or lower the surrounding dopamine levels if the levels are too low or too high, respectively, potentially offering symptom-relief across the illness-course. Therefore, the present study aimed at exploring the effects of cariprazine, a dopamine D2-D3 partial agonist with high affinity to D3 receptors, on motor symptoms associated with HD. METHODS This was a single-centre, retrospective study where sixteen patients received off-label cariprazine treatment for 12 weeks (1.5-3 mg/day). Motor symptoms were evaluated using the Motor Assessment of the Unified Huntington's Disease Rating Scale. Least Square (LS) Mean Changes from Baseline (BL) to Week 8 and Week 12 in the Total Motor Score (TMS) were analysed using the Mixed Model for Repeated Measures method. In addition, improvement from BL to Week 8 and 12 was calculated for all motor items. RESULTS Data of 16 patients were collected, but data of only 15 patients were analysed as one patient dropped out due to non-compliance. Significant changes were observed from BL to Week 8 (LS Mean Change: -9.4, p < 0.0001) and to Week 12 (LS Mean Change: -12.8, p < 0.0001) in the TMS. The improvement was captured in the majority of motor functions, excluding bradykinesia and gait. Mild akathisia was the most commonly reported side-effect, affecting 3 patients. CONCLUSION This is the first study investigating the effectiveness of a D2-D3 partial agonist, cariprazine, in the treatment of HD. The findings of this study revealed that cariprazine was effective in the treatment of a wide range of motor symptoms associated with HD.
Collapse
Affiliation(s)
- Reka Csehi
- Global Medical Division, Richter Gedeon Plc., Budapest, Hungary
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
| | - Viktor Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
- Eotvos Lorand Research Network-Semmelweis University Multiomics Neurodegeneration Research Group, Budapest, Hungary
| | - Mariann Fedor
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
| | - Vivien Zsumbera
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
| | - Agnes Palasti
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
| | - Karoly Acsai
- Global Medical Division, Richter Gedeon Plc., Budapest, Hungary
| | - Zoltan Grosz
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
- Eotvos Lorand Research Network-Semmelweis University Multiomics Neurodegeneration Research Group, Budapest, Hungary
| | - Gyorgy Nemeth
- Global Medical Division, Richter Gedeon Plc., Budapest, Hungary
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University Budapest, Budapest, Hungary.
- Eotvos Lorand Research Network-Semmelweis University Multiomics Neurodegeneration Research Group, Budapest, Hungary.
- , 1428 Budapest Pf. 2, Üllői út 26., Budapest, 1085, Hungary.
| |
Collapse
|
21
|
Lee H, Kim H, Choi D, Ko EN, Choi J, Seo Y, Lee S, Kim S, Jung S, Kim M, Kang D, Im C, Bae G, Jung S, Kwon O. Dopaminergic cell protection and alleviation of neuropsychiatric disease symptoms by VMAT2 expression through the class I HDAC inhibitor TC-H 106. Pharmacol Res Perspect 2023; 11:e01135. [PMID: 37740715 PMCID: PMC10517640 DOI: 10.1002/prp2.1135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/25/2023] Open
Abstract
The importance of vesicular monoamine transporter 2 (VMAT2) in dopamine regulation, which is considered crucial for neuropsychiatric disorders, is currently being studied. Moreover, the development of disease treatments using histone deacetylase (HDAC) inhibitors (HDACi) is actively progressing in various fields. Recently, research on the possibility of regulating neuropsychiatric disorders has been conducted. In this study, we evaluated whether VMAT2 expression increased by an HDACi can fine-tune neuropsychotic behavior, such as attention deficit hyperactivity disorder (ADHD) and protect against the cell toxicity through oxidized dopamine. First, approximately 300 candidate HDACi compounds were added to the SH-SY5Y dopaminergic cell line to identify the possible changes in the VMAT2 expression levels, which were measured using quantitative polymerase chain reaction. The results demonstrated, that treatment with pimelic diphenylamide 106 (TC-H 106), a class I HDACi, increased VMAT2 expression in both the SH-SY5Y cells and mouse brain. The increased VMAT2 expression induced by TC-H 106 alleviated the cytotoxicity attributed to 6-hydroxydopamine (6-OHDA) or 1-methyl-4-phenylpyridinium (MPP+ ) and free dopamine treatment. Moreover, dopamine concentrations, both intracellularly and in the synaptosomes, were significantly elevated by increased VMAT2 expression. These results suggest that dopamine concentration regulation by VMAT2 expression induced by TC-H 106 could alter several related behavioral aspects that was confirmed by attenuation of hyperactivity and impulsivity, which were major characteristics of animal model showing ADHD-like behaviors. These results indicate that HDACi-increased VMAT2 expression offers sufficient protections against dopaminergic cell death induced by oxidative stress. Thus, the epigenetic approach could be considered as therapeutic candidate for neuropsychiatric disease regulation.
Collapse
Affiliation(s)
- Heejin Lee
- New Drug Development Center, K‐MedihubDaeguKorea
| | - Hye‐Ji Kim
- Department of Physiology, School of MedicineJeju National UniversityJejuKorea
| | | | - Eu n.‐A. Ko
- Department of Physiology, School of MedicineJeju National UniversityJejuKorea
| | | | - Yohan Seo
- New Drug Development Center, K‐MedihubDaeguKorea
| | - Sion Lee
- New Drug Development Center, K‐MedihubDaeguKorea
| | | | - Sejin Jung
- New Drug Development Center, K‐MedihubDaeguKorea
| | - Minwoo Kim
- New Drug Development Center, K‐MedihubDaeguKorea
| | - Dongwan Kang
- New Drug Development Center, K‐MedihubDaeguKorea
| | | | - Gi‐Hun Bae
- New Drug Development Center, K‐MedihubDaeguKorea
| | - Sung‐Cherl Jung
- Department of Physiology, School of MedicineJeju National UniversityJejuKorea
| | - Oh‐Bin Kwon
- New Drug Development Center, K‐MedihubDaeguKorea
| |
Collapse
|
22
|
Jiang A, Handley RR, Lehnert K, Snell RG. From Pathogenesis to Therapeutics: A Review of 150 Years of Huntington's Disease Research. Int J Mol Sci 2023; 24:13021. [PMID: 37629202 PMCID: PMC10455900 DOI: 10.3390/ijms241613021] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative genetic disorder caused by an expanded polyglutamine-coding (CAG) trinucleotide repeat in the huntingtin (HTT) gene. HD behaves as a highly penetrant dominant disorder likely acting through a toxic gain of function by the mutant huntingtin protein. Widespread cellular degeneration of the medium spiny neurons of the caudate nucleus and putamen are responsible for the onset of symptomology that encompasses motor, cognitive, and behavioural abnormalities. Over the past 150 years of HD research since George Huntington published his description, a plethora of pathogenic mechanisms have been proposed with key themes including excitotoxicity, dopaminergic imbalance, mitochondrial dysfunction, metabolic defects, disruption of proteostasis, transcriptional dysregulation, and neuroinflammation. Despite the identification and characterisation of the causative gene and mutation and significant advances in our understanding of the cellular pathology in recent years, a disease-modifying intervention has not yet been clinically approved. This review includes an overview of Huntington's disease, from its genetic aetiology to clinical presentation and its pathogenic manifestation. An updated view of molecular mechanisms and the latest therapeutic developments will also be discussed.
Collapse
Affiliation(s)
- Andrew Jiang
- Applied Translational Genetics Group, Centre for Brain Research, School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand; (R.R.H.); (K.L.); (R.G.S.)
| | | | | | | |
Collapse
|
23
|
Plácido E, Gomes Welter P, Wink A, Karasiak GD, Outeiro TF, Dafre AL, Gil-Mohapel J, Brocardo PS. Beyond Motor Deficits: Environmental Enrichment Mitigates Huntington's Disease Effects in YAC128 Mice. Int J Mol Sci 2023; 24:12607. [PMID: 37628801 PMCID: PMC10454852 DOI: 10.3390/ijms241612607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative genetic disorder characterized by motor, psychiatric, cognitive, and peripheral symptoms without effective therapy. Evidence suggests that lifestyle factors can modulate disease onset and progression, and environmental enrichment (EE) has emerged as a potential approach to mitigate the progression and severity of neurodegenerative processes. Wild-type (WT) and yeast artificial chromosome (YAC) 128 mice were exposed to different EE conditions. Animals from cohort 1 were exposed to EE between postnatal days 21 and 60, and animals from cohort 2 were exposed to EE between postnatal days 60 and 120. Motor and non-motor behavioral tests were employed to evaluate the effects of EE on HD progression. Monoamine levels, hippocampal cell proliferation, neuronal differentiation, and dendritic arborization were also assessed. Here we show that EE had an antidepressant-like effect and slowed the progression of motor deficits in HD mice. It also reduced monoamine levels, which correlated with better motor performance, particularly in the striatum. EE also modulated neuronal differentiation in the YAC128 hippocampus. These results confirm that EE can impact behavior, hippocampal neuroplasticity, and monoamine levels in YAC128 mice, suggesting this could be a therapeutic strategy to modulate neuroplasticity deficits in HD. However, further research is needed to fully understand EE's mechanisms and long-term effects as an adjuvant therapy for this debilitating condition.
Collapse
Affiliation(s)
- Evelini Plácido
- Neuroscience Graduate Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil (A.W.); (A.L.D.)
| | - Priscilla Gomes Welter
- Neuroscience Graduate Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil (A.W.); (A.L.D.)
| | - Ana Wink
- Neuroscience Graduate Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil (A.W.); (A.L.D.)
| | - Gabriela Duarte Karasiak
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil;
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany;
- Max Planck Institute for Natural Sciences, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE1 7RU, UK
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 18147 Göttingen, Germany
| | - Alcir Luiz Dafre
- Neuroscience Graduate Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil (A.W.); (A.L.D.)
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil;
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia and Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Patricia S. Brocardo
- Neuroscience Graduate Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil (A.W.); (A.L.D.)
- Department of Morphological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil
| |
Collapse
|
24
|
Arancibia-Opazo S, Contreras-Riquelme JS, Sánchez M, Cisternas-Olmedo M, Vidal RL, Martin AJM, Sáez MA. Transcriptional and Histone Acetylation Changes Associated with CRE Elements Expose Key Factors Governing the Regulatory Circuit in the Early Stage of Huntington's Disease Models. Int J Mol Sci 2023; 24:10848. [PMID: 37446028 DOI: 10.3390/ijms241310848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Huntington's disease (HD) is a disorder caused by an abnormal expansion of trinucleotide CAG repeats within the huntingtin (Htt) gene. Under normal conditions, the CREB Binding Protein interacts with CREB elements and acetylates Lysine 27 of Histone 3 to direct the expression of several genes. However, mutant Htt causes depletion of CBP, which in turn induces altered histone acetylation patterns and transcriptional deregulation. Here, we have studied a differential expression analysis and H3K27ac variation in 4- and 6-week-old R6/2 mice as a model of juvenile HD. The analysis of differential gene expression and acetylation levels were integrated into Gene Regulatory Networks revealing key regulators involved in the altered transcription cascade. Our results show changes in acetylation and gene expression levels that are related to impaired neuronal development, and key regulators clearly defined in 6-week-old mice are proposed to drive the downstream regulatory cascade in HD. Here, we describe the first approach to determine the relationship among epigenetic changes in the early stages of HD. We determined the existence of changes in pre-symptomatic stages of HD as a starting point for early onset indicators of the progression of this disease.
Collapse
Affiliation(s)
- Sandra Arancibia-Opazo
- Chromatin, Epigenetic, and Neuroscience Laboratory, Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
- Programa de Doctorado en Genómica Integrativa, Vicerrectoría de Investigación, Universidad Mayor, Santiago 8580745, Chile
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Universidad San Sebastián, Santiago 8580704, Chile
| | - J Sebastián Contreras-Riquelme
- Plant Genome Regulation Lab, Centro de Biotecnología Vegetal, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370186, Chile
| | - Mario Sánchez
- Chromatin, Epigenetic, and Neuroscience Laboratory, Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
| | - Marisol Cisternas-Olmedo
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago 8380455, Chile
- Center for Geroscience, Brain Health, and Metabolism, Santiago 8380453, Chile
| | - René L Vidal
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago 8380455, Chile
- Center for Geroscience, Brain Health, and Metabolism, Santiago 8380453, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Alberto J M Martin
- Laboratorio de Redes Biológicas, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Universidad San Sebastián, Santiago 8580704, Chile
- Escuela de Ingeniería, Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Santiago 7500000, Chile
| | - Mauricio A Sáez
- Chromatin, Epigenetic, and Neuroscience Laboratory, Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago 8580745, Chile
- Centro de Oncología de Precisión, Facultad de Medicina Universidad Mayor, Santiago 7560908, Chile
- Laboratorio de Investigación en Salud de Precisión, Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de la Salud, Universidad Católica de Temuco, Temuco 4813302, Chile
| |
Collapse
|
25
|
Razick DI, Akhtar M, Wen J, Alam M, Dean N, Karabala M, Ansari U, Ansari Z, Tabaie E, Siddiqui S. The Role of Sirtuin 1 (SIRT1) in Neurodegeneration. Cureus 2023; 15:e40463. [PMID: 37456463 PMCID: PMC10349546 DOI: 10.7759/cureus.40463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Sirtuins (SIRT) are a class of histone deacetylases that regulate important metabolic pathways and play a role in several disease processes. Of the seven mammalian homologs currently identified, sirtuin 1 (SIRT1) is the best understood and most studied. It has been associated with several neurodegenerative diseases and cancers. As such, it has been further investigated as a therapeutic target in the treatment of disorders such as Parkinson's disease (PD), Huntington's disease (HD), and Alzheimer's disease (AD). SIRT1 deacetylates histones such as H1 lysine 26, H3 lysine 9, H3 lysine 56, and H4 lysine 16 to regulate chromatin remodeling and gene transcription. The homolog has also been observed to express contradictory responses to tumor suppression and tumor promotion. Studies have shown that SIRT1 may have anti-inflammatory properties by inhibiting the effects of NF-κB, as well as stimulating upregulation of autophagy. The SIRT1 activators resveratrol and cilostazol have been shown to improve Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog) scores in AD patients. In this review, we aim to explore the various roles of SIRT1 with regard to neuroprotection and neurodegeneration.
Collapse
Affiliation(s)
- Daniel I Razick
- Surgery, California Northstate University College of Medicine, Elk Grove, USA
| | - Muzammil Akhtar
- Surgery, California Northstate University College of Medicine, Elk Grove, USA
| | - Jimmy Wen
- Physical Medicine and Rehabilitation, California Northstate University College of Medicine, Elk Grove, USA
| | - Meraj Alam
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Nabeal Dean
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Muhammad Karabala
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Ubaid Ansari
- Internal Medicine, California Northstate University College of Medicine, Elk Grove, USA
| | - Zaid Ansari
- Internal Medicine, University of California Berkeley, Berkeley, USA
| | - Ethan Tabaie
- Neurosurgery, California Northstate University College of Medicine, Elk Grove, USA
| | - Shakeel Siddiqui
- Anesthesiology, OrthoMed Staffing Anesthesiology Group, Dallas, USA
| |
Collapse
|
26
|
Li C, Saliba NB, Martin H, Losurdo NA, Kolahdouzan K, Siddiqui R, Medeiros D, Li W. Purkinje cell dopaminergic inputs to astrocytes regulate cerebellar-dependent behavior. Nat Commun 2023; 14:1613. [PMID: 36959176 PMCID: PMC10036610 DOI: 10.1038/s41467-023-37319-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/13/2023] [Indexed: 03/25/2023] Open
Abstract
Dopamine has a significant role in motor and cognitive function. The dopaminergic pathways originating from the midbrain have received the most attention; however, the relevance of the cerebellar dopaminergic system is largely undiscovered. Here, we show that the major cerebellar astrocyte type Bergmann glial cells express D1 receptors. Dopamine can be synthesized in Purkinje cells by cytochrome P450 and released in an activity-dependent fashion. We demonstrate that activation of D1 receptors induces membrane depolarization and Ca2+ release from the internal store. These astrocytic activities in turn modify Purkinje cell output by altering its excitatory and inhibitory synaptic input. Lastly, we show that conditional knockout of D1 receptors in Bergmann glial cells results in decreased locomotor activity and impaired social activity. These results contribute to the understanding of the molecular, cellular, and circuit mechanisms underlying dopamine function in the cerebellum, revealing a critical role for the cerebellar dopaminergic system in motor and social behavior.
Collapse
Affiliation(s)
- Chang Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Natalie B Saliba
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hannah Martin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Nicole A Losurdo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Neuroscience Program, The University of Utah, Salt Lake City, UT, USA
| | - Kian Kolahdouzan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Riyan Siddiqui
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Destynie Medeiros
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
27
|
Rodriguez-Contreras D, Gong S, Lebowitz JJ, Fedorov LM, Asad N, Dore TM, Phillips TJ, Ford CP, Williams JT, Neve KA. Gait Abnormalities and Aberrant D2 Receptor Expression and Signaling in Mice Carrying the Human Pathogenic Mutation DRD2I212F. Mol Pharmacol 2023; 103:188-198. [PMID: 36456191 PMCID: PMC11033946 DOI: 10.1124/molpharm.122.000606] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/21/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
A dopamine D2 receptor mutation was recently identified in a family with a novel hyperkinetic movement disorder. That allelic variant D2-I212F is a constitutively active and G protein-biased receptor. We now describe mice engineered using CRISPR-Cas9-mediated gene editing technology to carry the D2-I212F variant. Drd2I212F mice exhibited gait abnormalities resembling those in other mouse models of chorea and/or dystonia and had striatal D2 receptor expression that was decreased approximately 30% per Drd2I212F allele. Electrically evoked inhibitory postsynaptic conductances in midbrain dopamine neurons and striatum from Drd2I212F mice, caused by G protein activation of potassium channels, exhibited slow kinetics (e.g., approximately four- to sixfold slower decay) compared with Drd2 +/+ mice. Current decay initiated by photolytic release of the D2 antagonist sulpiride from CyHQ-sulpiride was also ∼fourfold slower in midbrain slices from Drd2I212F mice than Drd2 +/+ mice. Furthermore, in contrast to Drd2 +/+ mice, in which dopamine is several-fold more potent at neurons in the nucleus accumbens than in the dorsal striatum, reflecting activation of Gα o versus Gα i, dopamine had similar potencies in those two brain regions of Drd2I212F mice. Repeated cocaine treatment, which decreases dopamine potency in the nucleus accumbens of Drd2 +/+ mice, had no effect on dopamine potency in Drd2 I212F mice. The results demonstrate the pathogenicity of the D2-I212F mutation and the utility of this mouse model for investigating the role of pathogenic DRD2 variants in early-onset hyperkinetic movement disorders. SIGNIFICANCE STATEMENT: The first dopamine receptor mutation to cause a movement disorder, D2-I212F, was recently identified. The mutation makes receptor activation of G protein-mediated signaling more efficient. To confirm the pathogenesis of D2-I212F, this study reports that mice carrying this mutation have gait abnormalities consistent with the clinical phenotype. The mutation also profoundly alters D2 receptor expression and function in vivo. This mouse model will be useful for further characterization of the mutant receptor and for evaluation of potential therapeutic drugs.
Collapse
Affiliation(s)
- Dayana Rodriguez-Contreras
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Sheng Gong
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Joseph J Lebowitz
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Lev M Fedorov
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Naeem Asad
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Timothy M Dore
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Tamara J Phillips
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Christopher P Ford
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - John T Williams
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| | - Kim A Neve
- Research Service, VA Portland Health Care System, Portland, Oregon (D.R.-C., T.J.P., K.A.N.); Department of Behavioral Neuroscience (D.R.-C., T.J.P., K.A.N.), Transgenic Mouse Models Shared Resource (L.M.F.), and Vollum Institute (J.J.L., J.T.W.), Oregon Health & Science University, Portland, Oregon; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (S.G., C.P.F.); Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio (S.G., C.P.F.); and New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates (N.A., T.M.D.)
| |
Collapse
|
28
|
Pradhan SS, Rao KR, Manjunath M, Saiswaroop R, Patnana DP, Phalguna KS, Choudhary B, Sivaramakrishnan V. Vitamin B 6, B 12 and folate modulate deregulated pathways and protein aggregation in yeast model of Huntington disease. 3 Biotech 2023; 13:96. [PMID: 36852176 PMCID: PMC9958225 DOI: 10.1007/s13205-023-03525-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Huntington's disease (HD) is an incurable and progressive neurodegenerative disease affecting the basal ganglia of the brain. HD is caused due to expansion of the polyglutamine tract in the protein Huntingtin resulting in aggregates. The increased PolyQ length results in aggregation of protein Huntingtin leading to neuronal cell death. Vitamin B6, B12 and folate are deficient in many neurodegenerative diseases. We performed an integrated analysis of transcriptomic, metabolomic and cofactor-protein network of vitamin B6, B12 and folate was performed. Our results show considerable overlap of pathways modulated by Vitamin B6, B12 and folate with those obtained from transcriptomic and metabolomic data of HD patients and model systems. Further, in yeast model of HD we showed treatment of B6, B12 or folate either alone or in combination showed impaired aggregate formation. Transcriptomic analysis of yeast model treated with B6, B12 and folate showed upregulation of pathways like ubiquitin mediated proteolysis, autophagy, peroxisome, fatty acid, lipid and nitrogen metabolism. Metabolomic analysis of yeast model shows deregulation of pathways like aminoacyl-tRNA biosynthesis, metabolism of various amino acids, nitrogen metabolism and glutathione metabolism. Integrated transcriptomic and metabolomic analysis of yeast model showed concordance in the pathways obtained. Knockout of Peroxisomal (PXP1 and PEX7) and Autophagy (ATG5) genes in yeast increased aggregates which is mitigated by vitamin B6, B12 and folate treatment. Taken together our results show a role for Vitamin B6, B12 and folate mediated modulation of pathways important for preventing protein aggregation with potential implications for HD. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03525-y.
Collapse
Affiliation(s)
- Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - K. Raksha Rao
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka 560100 India
| | - Meghana Manjunath
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka 560100 India
| | - R. Saiswaroop
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - Durga Prasad Patnana
- Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - Kanikaram Sai Phalguna
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka 560100 India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515134 India
| |
Collapse
|
29
|
D’Egidio F, Castelli V, Cimini A, d’Angelo M. Cell Rearrangement and Oxidant/Antioxidant Imbalance in Huntington's Disease. Antioxidants (Basel) 2023; 12:571. [PMID: 36978821 PMCID: PMC10045781 DOI: 10.3390/antiox12030571] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Huntington's Disease (HD) is a hereditary neurodegenerative disorder caused by the expansion of a CAG triplet repeat in the HTT gene, resulting in the production of an aberrant huntingtin (Htt) protein. The mutant protein accumulation is responsible for neuronal dysfunction and cell death. This is due to the involvement of oxidative damage, excitotoxicity, inflammation, and mitochondrial impairment. Neurons naturally adapt to bioenergetic alteration and oxidative stress in physiological conditions. However, this dynamic system is compromised when a neurodegenerative disorder occurs, resulting in changes in metabolism, alteration in calcium signaling, and impaired substrates transport. Thus, the aim of this review is to provide an overview of the cell's answer to the stress induced by HD, focusing on the role of oxidative stress and its balance with the antioxidant system.
Collapse
Affiliation(s)
| | | | | | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
30
|
Correa BH, Moreira CR, Hildebrand ME, Vieira LB. The Role of Voltage-Gated Calcium Channels in Basal Ganglia Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:183-201. [PMID: 35339179 PMCID: PMC10190140 DOI: 10.2174/1570159x20666220327211156] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 03/14/2022] [Indexed: 11/22/2022] Open
Abstract
Calcium (Ca2+) plays a central role in regulating many cellular processes and influences cell survival. Several mechanisms can disrupt Ca2+ homeostasis to trigger cell death, including oxidative stress, mitochondrial damage, excitotoxicity, neuroinflammation, autophagy, and apoptosis. Voltage-gated Ca2+ channels (VGCCs) act as the main source of Ca2+ entry into electrically excitable cells, such as neurons, and they are also expressed in glial cells such as astrocytes and oligodendrocytes. The dysregulation of VGCC activity has been reported in both Parkinson's disease (PD) and Huntington's (HD). PD and HD are progressive neurodegenerative disorders (NDs) of the basal ganglia characterized by motor impairment as well as cognitive and psychiatric dysfunctions. This review will examine the putative role of neuronal VGCCs in the pathogenesis and treatment of central movement disorders, focusing on PD and HD. The link between basal ganglia disorders and VGCC physiology will provide a framework for understanding the neurodegenerative processes that occur in PD and HD, as well as a possible path towards identifying new therapeutic targets for the treatment of these debilitating disorders.
Collapse
Affiliation(s)
- Bernardo H.M. Correa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlos Roberto Moreira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
31
|
Albekairi TH, Kamra A, Bhardwaj S, Mehan S, Giri A, Suri M, Alshammari A, Alharbi M, Alasmari AF, Narula AS, Kalfin R. Beta-Boswellic Acid Reverses 3-Nitropropionic Acid-Induced Molecular, Mitochondrial, and Histopathological Defects in Experimental Rat Model of Huntington's Disease. Biomedicines 2022; 10:2866. [PMID: 36359390 PMCID: PMC9687177 DOI: 10.3390/biomedicines10112866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 10/01/2023] Open
Abstract
Huntington's disease (HD) is distinguished by a triple repeat of CAG in exon 1, an increase in poly Q in the Htt gene, and a loss of GABAergic medium spiny neurons (MSN) in the striatum and white matter of the cortex. Mitochondrial ETC-complex dysfunctions are involved in the pathogenesis of HD, including neuronal energy loss, synaptic neurotrophic decline, neuronal inflammation, apoptosis, and grey and white matter destruction. A previous study has demonstrated that beta Boswellic acid (β-BA), a naturally occurring phytochemical, has several neuroprotective properties that can reduce pathogenic factors associated with various neurological disorders. The current investigation aimed to investigate the neuroprotective potential of β-BA at oral doses of 5, 10, and 15 mg/kg alone, as well as in conjunction with the potent antioxidant vitamin E (8 mg/kg, orally) in 3-NP-induced experimental HD rats. Adult Wistar rats were separated into seven groups, and 3-NP, at a dose of 10 mg/kg, was orally administered to each group of adult Wistar rats beginning on day 1 and continuing through day 14. The neurotoxin 3-NP induces neurodegenerative, g, neurochemical, and pathological alterations in experimental animals. Continuous injection of 3-NP, according to our results, aggravated HD symptoms by suppressing ETC-complex-II, succinate dehydrogenase activity, and neurochemical alterations. β-BA, when taken with vitamin E, improved behavioural dysfunctions such as neuromuscular and motor impairments, as well as memory and cognitive abnormalities. Pharmacological treatments with β-BA improved and restored ETC complexes enzymes I, II, and V levels in brain homogenates. β-BA treatment also restored neurotransmitter levels in the brain while lowering inflammatory cytokines and oxidative stress biomarkers. β-BA's neuroprotective potential in reducing neuronal death was supported by histopathological findings in the striatum and cortex. As a result, the findings of this research contributed to a better understanding of the potential role of natural phytochemicals β-BA in preventing neurological illnesses such as HD.
Collapse
Affiliation(s)
- Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Arzoo Kamra
- Department of Pharmacology, Seth G.L. Bihani S.D. College of Technical Education, Institute of Pharmaceutical Sciences and Drug Research, Sri Ganganagar 335001, Rajasthan, India
| | - Sudeep Bhardwaj
- Department of Pharmacology, Seth G.L. Bihani S.D. College of Technical Education, Institute of Pharmaceutical Sciences and Drug Research, Sri Ganganagar 335001, Rajasthan, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Aditi Giri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Manisha Suri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, 1113 Sofia, Bulgaria
- Department of Healthcare, South-West University “NeofitRilski”, Ivan Mihailov St. 66, 2700 Blagoevgrad, Bulgaria
| |
Collapse
|
32
|
Weiss AR, Liguore WA, Brandon K, Wang X, Liu Z, Domire JS, Button D, Srinivasan S, Kroenke CD, McBride JL. A novel rhesus macaque model of Huntington's disease recapitulates key neuropathological changes along with motor and cognitive decline. eLife 2022; 11:e77568. [PMID: 36205397 PMCID: PMC9545527 DOI: 10.7554/elife.77568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
We created a new nonhuman primate model of the genetic neurodegenerative disorder Huntington's disease (HD) by injecting a mixture of recombinant adeno-associated viral vectors, serotypes AAV2 and AAV2.retro, each expressing a fragment of human mutant HTT (mHTT) into the caudate and putamen of adult rhesus macaques. This modeling strategy results in expression of mutant huntingtin protein (mHTT) and aggregate formation in the injected brain regions, as well as dozens of other cortical and subcortical brain regions affected in human HD patients. We queried the disruption of cortico-basal ganglia circuitry for 30 months post-surgery using a variety of behavioral and imaging readouts. Compared to controls, mHTT-treated macaques developed working memory decline and progressive motor impairment. Multimodal imaging revealed circuit-wide white and gray matter degenerative processes in several key brain regions affected in HD. Taken together, we have developed a novel macaque model of HD that may be used to develop disease biomarkers and screen promising therapeutics.
Collapse
Affiliation(s)
- Alison R Weiss
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - William A Liguore
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Kristin Brandon
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Xiaojie Wang
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
| | - Zheng Liu
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
| | - Jacqueline S Domire
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Dana Button
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Sathya Srinivasan
- Imaging and Morphology Support Core, Oregon National Primate Research CenterBeavertonUnited States
| | - Christopher D Kroenke
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
- Department of Behavioral Neuroscience, Oregon Health and Science UniversityPortlandUnited States
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Department of Behavioral Neuroscience, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
33
|
Insights into the Promising Prospect of G Protein and GPCR-Mediated Signaling in Neuropathophysiology and Its Therapeutic Regulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8425640. [PMID: 36187336 PMCID: PMC9519337 DOI: 10.1155/2022/8425640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
G protein-coupled receptors (GPCRs) are intricately involved in the conversion of extracellular feedback to intracellular responses. These specialized receptors possess a crucial role in neurological and psychiatric disorders. Most nonsensory GPCRs are active in almost 90% of complex brain functions. At the time of receptor phosphorylation, a GPCR pathway is essentially activated through a G protein signaling mechanism via a G protein-coupled receptor kinase (GRK). Dopamine, an important neurotransmitter, is primarily involved in the pathophysiology of several CNS disorders; for instance, bipolar disorder, schizophrenia, Parkinson's disease, and ADHD. Since dopamine, acetylcholine, and glutamate are potent neuropharmacological targets, dopamine itself has potential therapeutic effects in several CNS disorders. GPCRs essentially regulate brain functions by modulating downstream signaling pathways. GPR6, GPR52, and GPR8 are termed orphan GPCRs because they colocalize with dopamine D1 and D2 receptors in neurons of the basal ganglia, either alone or with both receptors. Among the orphan GPCRs, the GPR52 is recognized for being an effective psychiatric receptor. Various antipsychotics like aripiprazole and quetiapine mainly target GPCRs to exert their actions. One of the most important parts of signal transduction is the regulation of G protein signaling (RGS). These substances inhibit the activation of the G protein that initiates GPCR signaling. Developing a combination of RGS inhibitors with GPCR agonists may prove to have promising therapeutic potential. Indeed, several recent studies have suggested that GPCRs represent potentially valuable therapeutic targets for various psychiatric disorders. Molecular biology and genetically modified animal model studies recommend that these enriched GPCRs may also act as potential therapeutic psychoreceptors. Neurotransmitter and neuropeptide GPCR malfunction in the frontal cortex and limbic-related regions, including the hippocampus, hypothalamus, and brainstem, is likely responsible for the complex clinical picture that includes cognitive, perceptual, emotional, and motor symptoms. G protein and GPCR-mediated signaling play a critical role in developing new treatment options for mental health issues, and this study is aimed at offering a thorough picture of that involvement. For patients who are resistant to current therapies, the development of new drugs that target GPCR signaling cascades remains an interesting possibility. These discoveries might serve as a fresh foundation for the creation of creative methods for pharmacologically useful modulation of GPCR function.
Collapse
|
34
|
Zhang J, Liu Y, Liu Y, Liu W, Lu F, Yuan Z, Lu C. Gold Nanocluster-Encapsulated Hyperbranched Polyethyleneimine for Selective and Ratiometric Dopamine Analyses by Enhanced Self-Polymerization. Front Chem 2022; 10:928607. [PMID: 35873048 PMCID: PMC9307107 DOI: 10.3389/fchem.2022.928607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
The exploitation of selective and sensitive dopamine (DA) sensors is essential to more deeply understand its biological function and diagnosis of related diseases. In this study, gold nanocluster-encapsulated hyperbranched polyethyleneimine (hPEI-Au NCs) has been explored as the specific and ratiometric DA nanoprobe through hPEI-assisted DA self-polymerization reactions. The Au NCs encapsulation not only provides a fluorescent internal reference but also enhances the DA self-polymerization by weakening the proton sponge effect of the hPEI layer. Rapid and sensitive DA detection is realized through the proposed hPEI-Au NC nanoprobe with a limit of detection of 10 nM. The favorable selectivity over other possible interferents including amino acids, sugars, and salts is due to the specific self-polymerization reaction. The DA analysis in urine samples with small relative standard deviations has been accomplished with an hPEI-Au NC nanoprobe.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
| | - Ying Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
| | - Yang Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
| | - Wencai Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
| | - Fengniu Lu
- Department of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Zhiqin Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
- Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing, China
- *Correspondence: Zhiqin Yuan, ; Chao Lu,
| | - Chao Lu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhiqin Yuan, ; Chao Lu,
| |
Collapse
|
35
|
Potts Y, Bekkers JM. Dopamine Increases the Intrinsic Excitability of Parvalbumin-Expressing Fast-Spiking Cells in the Piriform Cortex. Front Cell Neurosci 2022; 16:919092. [PMID: 35755774 PMCID: PMC9218566 DOI: 10.3389/fncel.2022.919092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The piriform cortex (PCx) is essential for the adaptive processing of olfactory information. Neuromodulatory systems, including those utilizing serotonin, acetylcholine, noradrenaline, and dopamine, innervate and regulate neuronal activity in the PCx. Previous research has demonstrated the importance of acetylcholine, noradrenaline and serotonin in odor learning and memory. In contrast, the role of dopamine in the PCx remains under-explored. Here we examined how dopamine modulates the intrinsic electrical properties of identified classes of neurons in the PCx. We found that dopamine had no consistent effect on the intrinsic electrical properties of two types of glutamatergic neurons (semilunar and superficial pyramidal cells) or three types of GABAergic interneurons (horizontal, neurogliaform and somatastatin-expressing regular-spiking cells). However, dopamine had a striking effect on the intrinsic excitability of the parvalbumin-expressing fast-spiking (FS) class of GABAergic interneuron. Dopamine depolarized the resting potential, increased the input resistance and increased the firing frequency of FS cells. Co-application of dopamine with the D1-class dopamine receptor antagonist SCH 23390 blocked the effects of dopamine modulation on FS cells. Conversely, co-application of dopamine with the D2-class antagonist RS-(±)-sulpiride had no effect on dopamine modulation of these cells. Our results indicate that dopamine binds to D1-class dopamine receptors to increase the intrinsic excitability of FS cells. These findings suggest that dopamine has a highly targeted effect in the PCx and reveal how dopamine may modulate the balance between excitation and inhibition, with consequences for odor processing. In addition, our findings provide clues for understanding why neurodegenerative disorders that modify the dopamine system, such as Parkinson's disease, have a deleterious effect on the sense of smell, and may suggest novel diagnostics for the early detection of such disorders.
Collapse
Affiliation(s)
- Yasmin Potts
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - John M Bekkers
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
36
|
Liu Y, Liu Y, Zhang J, Zheng J, Yuan Z, Lu C. Catechin-inspired gold nanocluster nanoprobe for selective and ratiometric dopamine detection via forming azamonardine. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 274:121142. [PMID: 35305522 DOI: 10.1016/j.saa.2022.121142] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
The sensitive and selective perception of dopamine (DA, a typical neurotransmitter) is important to evaluate the biological environment. In this study, a catechin-functionalized gold nanocluster (C-Au NC) nanoprobe has been explored for the ratiometric DA sensing. The detection mechanism is based on the formation of azamonardine via selective DA-catechin chemical reaction and subsequent enhanced fluorescence emission. Using Au NC emission as the internal reference, ratiometric fluorescence variation is realized, which allows sensitive DA analysis with a limit of detection of 1.0 nM (S/N = 3) and linear response concentration range from 0 to 500 nM. The characteristic chemical reaction between catechin and DA affords favorable selectivity over other amino acids, metal ions and small molecules. In addition, the practical application of the proposed nanoprobe is validated by the accurate detection of DA content in urea and cell lysate samples.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yang Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jing Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiaojiao Zheng
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhiqin Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Key Laboratory of Plant Resources Research and Development, Beijing Technology and Business University, Beijing 100048, China.
| | - Chao Lu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, China; Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou 450001,China.
| |
Collapse
|
37
|
Wildridge B, Rozewicz S, Mohamed A, James J, Connolly G. Use of clozapine for psychosis and chorea in Huntington's disease systematic narrative review. PROGRESS IN NEUROLOGY AND PSYCHIATRY 2022. [DOI: 10.1002/pnp.750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Bethany Wildridge
- Dr Wildridge is FY2 within South Thames Foundation School, Dr Rozewicz is a Senior House Officer at Gold Coast University Hospital, Australia
| | - Samuel Rozewicz
- Dr Wildridge is FY2 within South Thames Foundation School, Dr Rozewicz is a Senior House Officer at Gold Coast University Hospital, Australia
| | - Ashma Mohamed
- Dr Mohamed is ST5 Psychiatrist within Surrey and Borders Partnership Trust (SABP), Dr James is a Senior House Officer, St Peter's Hospital, Chertsey and Dr Connolly is a Consultant Psychiatrist within SABP
| | - Joel James
- Dr Mohamed is ST5 Psychiatrist within Surrey and Borders Partnership Trust (SABP), Dr James is a Senior House Officer, St Peter's Hospital, Chertsey and Dr Connolly is a Consultant Psychiatrist within SABP
| | - Gerard Connolly
- Dr Mohamed is ST5 Psychiatrist within Surrey and Borders Partnership Trust (SABP), Dr James is a Senior House Officer, St Peter's Hospital, Chertsey and Dr Connolly is a Consultant Psychiatrist within SABP
| |
Collapse
|
38
|
Li DQ, Jiang F, Zhang HS, Zheng LJ, Wang QJ, Fu R, Liu XG, Gao PY. Network pharmacology-based approach to investigate the mechanisms of Zingiber officinale Roscoe in the treatment of neurodegenerative diseases. J Food Biochem 2022; 46:e14068. [PMID: 35128682 DOI: 10.1111/jfbc.14068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 11/28/2022]
Abstract
Neurodegenerative diseases (NDDs) are chronic neurological disorders associated with cognitive or motor dysfunction. As a common spice, Zingiber officinale Roscoe has been used as a medicine to treat a variety of NDDs. However, at the molecular level, the mechanisms of Z. officinale in treating of NDDs have not been deeply investigated. In this study, network pharmacology method, molecular docking, Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used to predict the mechanisms of Z. officinale in the treatment of NDDs. After a series of biological information analyses, five core targets were obtained, including heme oxygenase 1 (HMOX1), acetylcholinesterase (AChE), nitric oxide synthase (NOS), catechol-O-methyl-transferase (COMT), and metabotropic glutamate receptor 5 (mGluR5). Compounds 75, 68, 46, 67, 69, 49, 66, 50, 34, and 64 were identified as the main components of Z. officinale in the treatment of NDDs. The crucial pathways mainly include neuroactive ligand-receptor signaling pathways, cyclic adenosine monophosphate signaling pathways, dopamine synaptic signaling pathways, and so on. Besides, in vitro experiments by AChE inhibitory activities assay and neuroprotective activities against H2 O2 -induced injury in human neuroblastoma SH-SY5Y cells validated the reliability of the results of network analysis. PRACTICAL APPLICATIONS: Zingiber officinale Roscoe is widely used as a traditional spice and herbal medicine. It contains a number of active ingredients, which have shown activities on anti-neurodegenerative diseases (NDDs). In this paper, the potential mechanism of Z. officinale in the treatment of NDDs is explored through network pharmacology, and it was verified by in vitro experiments. The mechanism was not only clarified at the system level but also proved to be effective at the biological level. The results can be used as a reference for Z. officinale in the treating of NDDs.
Collapse
Affiliation(s)
- Dan-Qi Li
- Institute of Functional Molecules, Shenyang University of Chemical Technology, Shenyang, PR China
- Liaoning Province Key Laboratory of Green Functional Molecular Design and Development, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Fan Jiang
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Han-Shuo Zhang
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Lian-Jun Zheng
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Qing-Jie Wang
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Ran Fu
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Xue-Gui Liu
- Institute of Functional Molecules, Shenyang University of Chemical Technology, Shenyang, PR China
- National-Local Joint Engineering Laboratory for Development of Boron and Magnesium Resources and Fine Chemical Technology, Shenyang University of Chemical Technology, Shenyang, PR China
| | - Pin-Yi Gao
- Liaoning Province Key Laboratory of Green Functional Molecular Design and Development, Shenyang University of Chemical Technology, Shenyang, PR China
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, PR China
| |
Collapse
|
39
|
Feleus S, van Schaijk M, Roos RAC, de Bot ST. The Many Faces of Huntington’s Chorea Treatment: The Impact of Sudden Withdrawal of Tiapride after 40 Years of Use and a Systematic Review. J Pers Med 2022; 12:jpm12040589. [PMID: 35455705 PMCID: PMC9025785 DOI: 10.3390/jpm12040589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
Huntington’s Disease (HD) is a rare, neurodegenerative disorder characterized by chorea, cognitive decline, and behavioral changes. Despite wide clinical use since the mid-1980s, tiapride was recently withdrawn from the Dutch market without rationale. Although alternatives are available, many patients experienced dysregulation after this unwanted change. We provide insight into the impact of sudden tiapride withdrawal by reviewing medical records of HD patients who were using tiapride at the time of withdrawal. In addition, we performed a systematic search in five databases on tiapride efficacy and its safety profile in HD. Original research and expert opinions were included. In our patient group on tiapride, 50% required tiapride import from abroad. Regarding the review, 12 articles on original datasets and three expert opinions were included. The majority of studies showed an improvement in chorea while patients were on tiapride. Due to limited sample sizes, not all studies performed statistical tests on their results. Fifty percent of clinical experts prefer tiapride as initial chorea monotherapy, especially when comorbid behavioral symptoms are present. Side effects are often rare and mild. No safety concerns were reported. In conclusion, tiapride is almost irreplaceable for some patients and is an effective and safe chorea treatment in HD.
Collapse
Affiliation(s)
- Stephanie Feleus
- Department of Neurology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
- Department of Epidemiology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
- Correspondence:
| | - Malu van Schaijk
- Intellectual Disability Medicine, Department of General Practice, Erasmus Medical Center Rotterdam, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
- Advisium, ‘s Heeren Loo, P.O. Box 647, 3800 AP Amersfoort, The Netherlands
| | - Raymund A. C. Roos
- Department of Neurology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Susanne T. de Bot
- Department of Neurology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
40
|
Smoking, alcohol consumption, and age at onset of Huntington's disease: a Mendelian randomization study. Parkinsonism Relat Disord 2022; 97:34-38. [PMID: 35299068 DOI: 10.1016/j.parkreldis.2022.02.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Smoking and alcohol consumption have been associated with earlier age at onset (AAO) of Huntington's disease (HD) in observational studies. We conducted this Mendelian randomization (MR) study to evaluate whether these associations are causal. METHODS We selected genetic instruments for lifetime smoking (n = 462,690) and alcohol consumption (n = 941,280) based on two large genome-wide association studies (GWAS). The summary-level data for residual AAO of HD were derived from a GWAS meta-analysis carried out by the Genetic Modifiers of Huntington's disease Consortium (n = 9,064 HD patients). We conducted univariable and multivariable MR analyses to evaluate the independent impact of smoking and alcohol consumption on AAO of HD. RESULTS Genetically predicted lifetime smoking was causally related to an earlier AAO of HD in the univariable MR analyses (β = -2.16 years per standard deviation (SD) increase in lifetime smoking index, 95% confidence interval (CI) = -3.70 to -0.63, P = 0.006). This association persisted significant in the multivariable MR analyses after adjusting for alcohol consumption (β = -2.04 years per SD increase in lifetime smoking index, 95% CI = -3.85 to -0.22, P = 0.028). However, no significant association was found between alcohol consumption and AAO of HD. CONCLUSIONS This study suggests that genetically predicted smoking is causally related to an earlier AAO of HD.
Collapse
|
41
|
Motor cortex plasticity response to acute cardiorespiratory exercise and intermittent theta-burst stimulation is attenuated in premanifest and early Huntington’s disease. Sci Rep 2022; 12:1104. [PMID: 35058470 PMCID: PMC8776762 DOI: 10.1038/s41598-021-04378-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
AbstractHuntington’s disease (HD) mouse models suggest that cardiovascular exercise may enhance neuroplasticity and delay disease signs, however, the effects of exercise on neuroplasticity in people with HD are unknown. Using a repeated-measures experimental design, we compared the effects of a single bout of high-intensity exercise, moderate-intensity exercise, or rest, on motor cortex synaptic plasticity in 14 HD CAG-expanded participants (9 premanifest and 5 early manifest) and 20 CAG-healthy control participants, using transcranial magnetic stimulation. Measures of cortico-motor excitability, short-interval intracortical inhibition and intracortical facilitation were obtained before and after a 20-min bout of either high-intensity interval exercise, moderate-intensity continuous exercise, or rest, and again after intermittent theta burst stimulation (iTBS). HD participants showed less inhibition at baseline compared to controls. Whereas the control group showed increased excitability and facilitation following high-intensity exercise and iTBS, the HD group showed no differences in neuroplasticity responses following either exercise intensity or rest, with follow-up Bayesian analyses providing consistent evidence that these effects were absent in the HD group. These findings indicate that exercise-induced synaptic plasticity mechanisms in response to acute exercise may be attenuated in HD, and demonstrate the need for future research to further investigate exercise and plasticity mechanisms in people with HD.
Collapse
|
42
|
Harris KL, Mason SL, Vallin B, Barker RA. Reduced expression of dopamine D2 receptors on astrocytes in R6/1 HD mice and HD post-mortem tissue. Neurosci Lett 2022; 767:136289. [PMID: 34637857 PMCID: PMC9188264 DOI: 10.1016/j.neulet.2021.136289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022]
Abstract
Dysfunction of the central dopaminergic system is thought to contribute to some of the clinical features of Huntington's disease (HD), and dopamine (DA) receptor antagonists are commonly used to good effect in its treatment. It is well established that there is an early significant reduction in neuronal D2 receptors in HD, considered to be a compensatory response to increased dopaminergic activity. However, no studies have examined the expression of D2 receptors on astrocytes which is important given that these cells have been shown to play a role in the pathogenesis of HD, as well as express dopamine receptors and modulate DA homeostasis in the normal brain. We therefore sought to investigate the expression of D2 receptors on astrocytes in HD, and found them to be reduced in both the R6/1 HD mouse model, and in human post-mortem brain in comparison to controls, suggesting that astrocytes may be important in DA-dependent aspects of HD. Further studies are needed to determine the functional significance of this finding.
Collapse
Affiliation(s)
- Kate L Harris
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK.
| | - Sarah L Mason
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Benjamin Vallin
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK; MRC-WT Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
43
|
Barry J, Bui MTN, Levine MS, Cepeda C. Synaptic pathology in Huntington's disease: Beyond the corticostriatal pathway. Neurobiol Dis 2022; 162:105574. [PMID: 34848336 PMCID: PMC9328779 DOI: 10.1016/j.nbd.2021.105574] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is a heritable, fatal neurodegenerative disorder caused by a mutation in the Huntingtin gene. It is characterized by chorea, as well as cognitive and psychiatric symptoms. Histopathologically, there is a massive loss of striatal projection neurons and less but significant loss in other areas throughout the cortico-basal ganglia-thalamocortical (CBGTC) loop. The mutant huntingtin protein has been implicated in numerous functions, including an important role in synaptic transmission. Most studies on anatomical and physiological alterations in HD have focused on striatum and cerebral cortex. However, based on recent CBGTC projectome evidence, the need to study other pathways has become increasingly clear. In this review, we examine the current status of our knowledge of morphological and electrophysiological alterations of those pathways in animal models of HD. Based on recent studies, there is accumulating evidence that synaptic disconnection, particularly along excitatory pathways, is pervasive and almost universal in HD, thus supporting a critical role of the huntingtin protein in synaptic transmission.
Collapse
Affiliation(s)
- Joshua Barry
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Minh T N Bui
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael S Levine
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Carlos Cepeda
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
44
|
Voronin MV, Kadnikov IA, Zainullina LF, Logvinov IO, Verbovaya ER, Antipova TA, Vakhitova YV, Seredenin SB. Neuroprotective Properties of Quinone Reductase 2 Inhibitor M-11, a 2-Mercaptobenzimidazole Derivative. Int J Mol Sci 2021; 22:13061. [PMID: 34884863 PMCID: PMC8658107 DOI: 10.3390/ijms222313061] [Citation(s) in RCA: 2] [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: 11/19/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 01/03/2023] Open
Abstract
The ability of NQO2 to increase the production of free radicals under enhanced generation of quinone derivatives of catecholamines is considered to be a component of neurodegenerative disease pathogenesis. The present study aimed to investigate the neuroprotective mechanisms of original NQO2 inhibitor M-11 (2-[2-(3-oxomorpholin-4-il)-ethylthio]-5-ethoxybenzimidazole hydrochloride) in a cellular damage model using NQO2 endogenous substrate adrenochrome (125 µM) and co-substrate BNAH (100 µM). The effects of M-11 (10-100 µM) on the reactive oxygen species (ROS) generation, apoptosis and lesion of nuclear DNA were evaluated using flow cytometry and single-cell gel electrophoresis assay (comet assay). Results were compared with S29434, the reference inhibitor of NQO2. It was found that treatment of HT-22 cells with M-11 results in a decline of ROS production triggered by incubation of cells with NQO2 substrate and co-substrate. Pre-incubation of HT-22 cells with compounds M-11 or S29434 results in a decrease of DNA damage and late apoptotic cell percentage reduction. The obtained results provide a rationale for further development of the M-11 compound as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Mikhail V. Voronin
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| | - Ilya A. Kadnikov
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| | | | | | | | | | - Yulia V. Vakhitova
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| | - Sergei B. Seredenin
- Department of Pharmacogenetics, Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia; (L.F.Z.); (I.O.L.); (E.R.V.); (T.A.A.)
| |
Collapse
|
45
|
Akimov SS, Jiang M, Kedaigle AJ, Arbez N, Marque LO, Eddings CR, Ranum PT, Whelan E, Tang A, Wang R, DeVine LR, Talbot CC, Cole RN, Ratovitski T, Davidson BL, Fraenkel E, Ross CA. Immortalized striatal precursor neurons from Huntington's disease patient-derived iPS cells as a platform for target identification and screening for experimental therapeutics. Hum Mol Genet 2021; 30:2469-2487. [PMID: 34296279 PMCID: PMC8643509 DOI: 10.1093/hmg/ddab200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/12/2022] Open
Abstract
We have previously established induced pluripotent stem cell (iPSC) models of Huntington's disease (HD), demonstrating CAG-repeat-expansion-dependent cell biological changes and toxicity. However, the current differentiation protocols are cumbersome and time consuming, making preparation of large quantities of cells for biochemical or screening assays difficult. Here, we report the generation of immortalized striatal precursor neurons (ISPNs) with normal (33) and expanded (180) CAG repeats from HD iPSCs, differentiated to a phenotype resembling medium spiny neurons (MSN), as a proof of principle for a more tractable patient-derived cell model. For immortalization, we used co-expression of the enzymatic component of telomerase hTERT and conditional expression of c-Myc. ISPNs can be propagated as stable adherent cell lines, and rapidly differentiated into highly homogeneous MSN-like cultures within 2 weeks, as demonstrated by immunocytochemical criteria. Differentiated ISPNs recapitulate major HD-related phenotypes of the parental iPSC model, including brain-derived neurotrophic factor (BDNF)-withdrawal-induced cell death that can be rescued by small molecules previously validated in the parental iPSC model. Proteome and RNA-seq analyses demonstrate separation of HD versus control samples by principal component analysis. We identified several networks, pathways, and upstream regulators, also found altered in HD iPSCs, other HD models, and HD patient samples. HD ISPN lines may be useful for studying HD-related cellular pathogenesis, and for use as a platform for HD target identification and screening experimental therapeutics. The described approach for generation of ISPNs from differentiated patient-derived iPSCs could be applied to a larger allelic series of HD cell lines, and to comparable modeling of other genetic disorders.
Collapse
Affiliation(s)
- Sergey S Akimov
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Amanda J Kedaigle
- Department of Biological Engineering, Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Leonard O Marque
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Chelsy R Eddings
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul T Ranum
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, The Raymond G Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emma Whelan
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Anthony Tang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ronald Wang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lauren R DeVine
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Conover C Talbot
- The Johns Hopkins School of Medicine, Institute for Basic Biomedical Sciences, Baltimore, MD 21205, USA
| | - Robert N Cole
- Mass Spectrometry and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Beverly L Davidson
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, The Raymond G Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Department of Pathology and Laboratory Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Computational and Systems Biology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurology, Neuroscience and Pharmacology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
46
|
Repetitive Transcranial Magnetic Stimulation for Major Depressive Disorder Comorbid with Huntington’s Disease: A Case Report. NEUROSCI 2021. [DOI: 10.3390/neurosci2040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Huntington’s disease (HD) is a rare genetic disorder resulting in progressive neurodegeneration leading to motor, cognitive and psychiatric symptoms. A high percentage of HD patients suffer from comorbid major depressive disorder (MDD). We are not aware of any literature on the use of repetitive transcranial magnetic stimulation (rTMS) for treating comorbid MDD in HD. We present the case of a 57-year-old man suffering from HD in which comorbid MDD was successfully treated with rTMS. Further work is required to better characterize the safety, tolerability and effectiveness of rTMS to treat comorbid MDD in HD.
Collapse
|
47
|
Kaye J, Reisine T, Finkbeiner S. Huntington's disease mouse models: unraveling the pathology caused by CAG repeat expansion. Fac Rev 2021; 10:77. [PMID: 34746930 PMCID: PMC8546598 DOI: 10.12703/r/10-77] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease that results in motor and cognitive dysfunction, leading to early death. HD is caused by an expansion of CAG repeats in the huntingtin gene (HTT). Here, we review the mouse models of HD. They have been used extensively to better understand the molecular and cellular basis of disease pathogenesis as well as to provide non-human subjects to test the efficacy of potential therapeutics. The first and best-studied in vivo rodent model of HD is the R6/2 mouse, in which a transgene containing the promoter and exon 1 fragment of human HTT with 150 CAG repeats was inserted into the mouse genome. R6/2 mice express rapid, robust behavioral pathologies and display a number of degenerative abnormalities in neuronal populations most vulnerable in HD. The first conditional full-length mutant huntingtin (mHTT) mouse model of HD was the bacterial artificial chromosome (BAC) transgenic mouse model of HD (BACHD), which expresses human full-length mHTT with a mixture of 97 CAG-CAA repeats under the control of endogenous HTT regulatory machinery. It has been useful in identifying the role of mHTT in specific neuronal populations in degenerative processes. In the knock-in (KI) model of HD, the expanded human CAG repeats and human exon 1 are inserted into the mouse Htt locus, so a chimera of the full-length mouse protein with the N-terminal human portion is expressed. Many of aspects of the pathology and behavioral deficits in the KI model better mimic disease characteristics found in HD patients than other models. Accordingly, some have proposed that these mice may be preferable models of the disease over others. Indeed, as our understanding of HD advances, so will the design of animal models to test and develop HD therapies.
Collapse
Affiliation(s)
- Julia Kaye
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, CA, USA
| | - Steve Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA, USA
- Department of Neurology and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
48
|
Kumar V, Singh C, Singh A. Zebrafish an experimental model of Huntington's disease: molecular aspects, therapeutic targets and current challenges. Mol Biol Rep 2021; 48:8181-8194. [PMID: 34665402 DOI: 10.1007/s11033-021-06787-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
Huntington disease (HD) is a lethal autosomal dominant neurodegenerative disease whose exact causative mechanism is still unknown. It can transform from one generation to another generation. The CAG triplet expansion on polyglutamine (PolyQ) tract on Huntingtin protein primarily contributes in HD pathogenesis. Apart from this some another molecular mechanisms are also involved in HD pathology such as loss of Brain derived neurotrophic factor in medium spiny neurons, mitochondrial dysfunction, and alterations in synaptic plasticity are briefly discussed in this review. However, several chemicals (3-nitropropionic acid, and Quinolinic acid) and genetic (mHTT-ΔN17-97Q over expression) experimental models are used to explore the exact pathogenic mechanism and finding of new drug targets for the development of novel therapeutic approaches. The zebrafish (Danio rerio) is widely used in in-vivo screening of several central nervous system (CNS) diseases such as HD, Alzheimer's disease (AD), Parkinson's disease (PD), and in memory deficits. Thus, this makes zebrafish as an excellent animal model for the development of new therapeutic strategies against various CNS disorders. We had reviewed several publications utilizing zebrafish and rodents to explore the disease pathology. Studies suggested that zebrafish genes and their human homologues have conserved functions. Zebrafish advantages and their characteristics over the other experimental animals make it an excellent tool for the disease study. This review explains the possible pathogenic mechanism of HD and also discusses about possible treatment therapies, apart from this we also discussed about possible potential therapeutic targets which will helps in designing of novel therapeutic approaches to overcome the disease progression. Diagrammatic depiction shows prevention of HD pathogenesis through attenuation of various biochemical alterations.
Collapse
Affiliation(s)
- Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
49
|
Jabłońska M, Grzelakowska K, Wiśniewski B, Mazur E, Leis K, Gałązka P. Pridopidine in the treatment of Huntington's disease. Rev Neurosci 2021; 31:441-451. [PMID: 32083454 DOI: 10.1515/revneuro-2019-0085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/06/2019] [Indexed: 01/26/2023]
Abstract
Huntington's disease (HD) is a highly common inherited monogenic neurodegenerative disease, and the gene responsible for its development is located in the 4p16.3 chromosome. The product of that gene mutation is an abnormal huntingtin (Htt) protein that disrupts the neural conduction, thus leading to motor and cognitive disorders. The disease progresses to irreversible changes in the central nervous system (CNS). Although only a few drugs are available to symptomatic treatment, 'dopamine stabilizers' (as represented by the pridopidine) may be the new treatment options. The underlying causes of HD are dopaminergic conduction disorders. Initially, the disease is hyperkinetic (chorea) until it eventually reaches the hypokinetic phase. Studies confirmed a correlation between the amount of dopamine in the CNS and the stage of the disease. Pridopidine has the capacity to be a dopamine buffer, which could increase or decrease the dopamine content depending on the disease phase. A research carried out on animal models demonstrated the protective effect of pridopidine on nerve cells thanks to its ability to alter the cortical glutamatergic signaling through the N-methyl-D-aspartate (NMDA) receptors. Studies on dopamine stabilizers also reported that pridopidine has a 100-fold greater affinity for the sigma-1 receptor than for the D2 receptor. Disturbances in the activity of sigma-1 receptors occur in neurodegenerative diseases, including HD. Their interaction with pridopidine results in the neuroprotective effect, which is manifested as an increase in the plasticity of synaptic neurons and prevention of their atrophy within the striatum. To determine the effectiveness of pridopidine in the treatment of HD, large multicenter randomized studies such as HART, MermaiHD, and PRIDE-HD were carried out.
Collapse
Affiliation(s)
- Magdalena Jabłońska
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Klaudyna Grzelakowska
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Bartłomiej Wiśniewski
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Ewelina Mazur
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Kamil Leis
- Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| | - Przemysław Gałązka
- Department of General and Oncological Pediatric Surgery, Antoni Jurasz University Hospital No. 1 in Bydgoszcz, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-090 Bydgoszcz, Poland
| |
Collapse
|
50
|
Leroy A, Teixidor J, Bertsch A, Renaud P. In-flow electrochemical detection of chemicals in droplets with pyrolysed photoresist electrodes: application as a module for quantification of microsampled dopamine. LAB ON A CHIP 2021; 21:3328-3337. [PMID: 34250532 DOI: 10.1039/d1lc00116g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The electrochemical quantification of analytes in droplets of PBS separated by a fluorinated phase was investigated. PDMS-fused silica chips with pyrolysed photoresist electrodes were prepared using a simple fabrication technique and used to analyze droplets in flow. Potentiostatic chronoamperometry provided current readouts consistent with mass transport and the concentration inside the droplets. This paper highlights measurements of dopamine in droplets in T-junction microfluidic chips at unprecedently low concentrations, with a limit of detection of 207 nM and a linear range of 0.21-20 μM, giving results similar to continuous flow electrochemistry and allowing the analysis in the striatal extracellular range (<1 μM). The system was applied to the quick and reliable on-line detection of dopamine concentration steps in droplets collected with a microsampling probe in vitro, demonstrating the usefulness of the electrochemical device as a quantification module for microsampled chemicals in droplets.
Collapse
Affiliation(s)
- Albert Leroy
- EPFL-STI-IMT-LMIS4, École Polytechnique Fédérale de Lausanne, Station 17, CH-1015 Lausanne, Switzerland.
| | | | | | | |
Collapse
|