1
|
Isono J, Nakagawasai O, Takahashi K, Nemoto W, Tan-No K. Microinjection of brain-derived neurotrophic factor into the dorsal hippocampus alleviates cognitive dysfunction and increases hippocampal cell proliferation in olfactory bulbectomized mice. Neurosci Lett 2025; 859-861:138257. [PMID: 40381880 DOI: 10.1016/j.neulet.2025.138257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/07/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Patients with depression show cognitive dysfunction and experience decreased motivation, which interfere with their daily lives. However, whether brain-derived neurotrophic factor (BDNF) in the dorsal hippocampus is involved in depression-associated cognitive deficits is unclear. In this study, olfactory bulbectomized mice, used as a model of depression, showed cognitive dysfunction in the Y-maze and passive avoidance tests and reduction of cell proliferation in the hippocampal dentate gyrus. Microinjection of BDNF into the dorsal hippocampus alleviated the cognitive dysfunction induced by the olfactory bulbectomy and increased hippocampal cell proliferation. This study suggests that BDNF injection in the dorsal hippocampus modulates cognitive functions with cell proliferation in the hippocampal dentate gyrus, thus supporting the importance of hippocampal BDNF in cognitive impairment accompanying depression.
Collapse
Affiliation(s)
- Jinichi Isono
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan
| | - Osamu Nakagawasai
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan.
| | - Kohei Takahashi
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan; Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, Ohtawara, Tochigi 324-8501, Japan
| | - Wataru Nemoto
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan
| | - Koichi Tan-No
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan
| |
Collapse
|
2
|
Dalto JF, Medina JH, Pastor V. Molecular Underpinnings of Memory Persistence and Forgetting. J Neurochem 2025; 169:e70089. [PMID: 40411122 DOI: 10.1111/jnc.70089] [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: 02/19/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025]
Abstract
The fate of memories depends mainly on external and internal factors affecting cellular and systems consolidation on the one hand and the decay or weakening of the memory trace on the other hand. Over the past 40 years, research has focused on the mechanisms of memory consolidation, retrieval, and its consequences: extinction and reconsolidation. In contrast, much less is known about the molecular mechanisms required for the maintenance of memory storage and forgetting. These opposing forces are both activity- and time-dependent. Here, we summarize the molecular signatures and inherent mechanisms involved in memory persistence and active forgetting, highlighting recent findings on the role of dopamine neurotransmission, intracellular signaling cascades, and actin cytoskeleton dynamics in these processes.
Collapse
Affiliation(s)
- Juliana F Dalto
- Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo De Robertis", CONICET-UBA, Buenos Aires, Argentina
| | - Verónica Pastor
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo De Robertis", CONICET-UBA, Buenos Aires, Argentina
| |
Collapse
|
3
|
De Paolis ML, Loffredo G, Krashia P, La Barbera L, Nobili A, Cauzzi E, Babicola L, Di Segni M, Coccurello R, Puglisi-Allegra S, Latagliata EC, D'Amelio M. Repetitive prefrontal tDCS activates VTA dopaminergic neurons, resulting in attenuation of Alzheimer's Disease-like deficits in Tg2576 mice. Alzheimers Res Ther 2025; 17:94. [PMID: 40301905 PMCID: PMC12039073 DOI: 10.1186/s13195-025-01736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Emerging evidence implicates early dysfunction of dopaminergic neurons in the Ventral Tegmental Area (VTA) as a key contributor to Alzheimer's Disease (AD) pathophysiology. Specifically, the VTA dopaminergic neurodegeneration and the consequent reduction of dopamine (DA) in mesocorticolimbic targets are associated with the onset of cognitive impairments and neuropsychiatric-like manifestations in AD animal models. Moreover, decreased midbrain volume and functional VTA disconnection are identified as predictors of accelerated progression from Mild Cognitive Impairment to AD-dementia in clinical populations. Given these findings, interventions capable of directly modulating VTA activity and augmenting DA release, despite the ongoing neurodegeneration, may hold therapeutic potential for mitigating DA-related deficits in AD. This study aims at evaluating the therapeutic potential of prefrontal transcranial Direct Current Stimulation (tDCS) in the Tg2576 mouse model of AD, exhibiting early VTA dopaminergic neurodegeneration. METHODS Repeated tDCS was applied to assess its ability to activate VTA DA neurons. We also evaluated tDCS effects on synaptic plasticity, cognitive and non-cognitive behaviours and AD-related pathology. Hippocampal DA release and Nucleus Accumbens (NAc) DA transporter (DAT) expression were measured. With immunohistochemistry we examined microglial density and morphological complexity at different disease stages. Additionally, intracellular amyloid-β (Aβ) levels and plaque burden were evaluated to determine the impact of tDCS on AD pathology. RESULTS Prefrontal tDCS enhanced the activity of VTA dopaminergic neurons, leading to increased hippocampal DA release and higher DAT levels in the NAc. The enhanced DA outflow is associated with restored CA3-CA1 synaptic plasticity and improvements in recognition memory and motivational behaviours. tDCS reduced microglial numbers and morphological complexity in Tg2576 mice at both pre-plaque stage (7-months) and at an advanced stage characterized by plaque accumulation (12-months). Notably, tDCS also decreased Aβ plaque burden, although no changes in intracellular Aβ levels were observed in younger Tg2576 mice. CONCLUSIONS These findings highlight the multifaceted therapeutic potential of prefrontal tDCS in targeting key AD pathophysiological hallmarks, including dopaminergic dysfunction, synaptic impairments, neuroinflammation and plaque deposition. As a non-invasive neuromodulatory approach, prefrontal tDCS emerges as a promising early intervention strategy to complement existing AD treatments, with the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Gilda Loffredo
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Paraskevi Krashia
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Emma Cauzzi
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Lucy Babicola
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- Department of Psychology, Sapienza University of Rome, P.Le Aldo Moro, 5, 00185, Rome, Italy
| | - Matteo Di Segni
- Child Psychopathology Unit, IRCCS Eugenio Medea, Via Don Luigi Monza, 20, 23842, Bosisio Parini, Italy
| | - Roberto Coccurello
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- National Research Council (CNR), Institute for Complex System (ISC), Via Dei Taurini, 19, 00185, Rome, Italy
| | - Stefano Puglisi-Allegra
- Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Neuromed, Via Atinense, 18, 86077, Pozzilli, Italy
| | - Emanuele Claudio Latagliata
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
- Department of Psychology, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
4
|
Tezanos P, Trejo JL. Why are threatening experiences remembered so well? Insights into memory strengthening from protocols of gradual aversive learning. Neurosci Biobehav Rev 2025; 174:106145. [PMID: 40250543 DOI: 10.1016/j.neubiorev.2025.106145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Aversive experiences often result in strong and persistent memory traces, which can sometimes lead to conditions such as Post-Traumatic Stress Disorder or phobias. Aversive stimulation tests are key tools in psychology and neuroscience for studying learning and memory. These tests typically use electric shocks as the unconditioned stimulus, allowing for precise control over the aversive content of the learning event. This feature has led to extensive research applying these tests with varying shock intensities to examine differences in learning, behavior, and memory formation between low- and high-aversive experiences. This line of research is particularly valuable for understanding the neurobiology underlying memory strengthening, but, to our knowledge, no review has yet compiled and organized the findings from this specific methodology. In this comprehensive review, we focus primarily on animal studies that have employed the same aversive test (i.e. Fear Conditioning, Passive Avoidance, Active Avoidance or Operant boxes) at different intensities. We will first outline and briefly describe the main aversive learning paradigms used in this field. Next, we will examine the relationship between aversiveness and memory strength. Finally, we will explore the neurobiological insights these studies have revealed over the years. Our aim is to gain a better understanding of how the nervous system gradually strengthens memory, while also addressing the remaining gaps and challenges in this area of research.
Collapse
Affiliation(s)
- Patricia Tezanos
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain; PhD Program in Neuroscience, Universidad Autónoma de Madrid-Instituto Cajal, Madrid 28002, Spain
| | - José Luis Trejo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain.
| |
Collapse
|
5
|
Kleinman MR, Foster DJ. Spatial localization of hippocampal replay requires dopamine signaling. eLife 2025; 13:RP99678. [PMID: 40126538 PMCID: PMC11932692 DOI: 10.7554/elife.99678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
Sequenced reactivations of hippocampal neurons called replays, concomitant with sharp-wave ripples in the local field potential, are critical for the consolidation of episodic memory, but whether replays depend on the brain's reward or novelty signals is unknown. Here, we combined chemogenetic silencing of dopamine neurons in ventral tegmental area (VTA) and simultaneous electrophysiological recordings in dorsal hippocampal CA1, in freely behaving male rats experiencing changes to reward magnitude and environmental novelty. Surprisingly, VTA silencing did not prevent ripple increases where reward was increased, but caused dramatic, aberrant ripple increases where reward was unchanged. These increases were associated with increased reverse-ordered replays. On familiar tracks this effect disappeared, and ripples tracked reward prediction error (RPE), indicating that non-VTA reward signals were sufficient to direct replay. Our results reveal a novel dependence of hippocampal replay on dopamine, and a role for a VTA-independent RPE signal that is reliable only in familiar environments.
Collapse
Affiliation(s)
- Matthew R Kleinman
- Helen Wills Neuroscience Institute and Department of Psychology, University of California, BerkeleyBerkeleyUnited States
| | - David J Foster
- Helen Wills Neuroscience Institute and Department of Psychology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
6
|
Fuchsberger T, Stockwell I, Woods M, Brzosko Z, Greger IH, Paulsen O. Dopamine increases protein synthesis in hippocampal neurons enabling dopamine-dependent LTP. eLife 2025; 13:RP100822. [PMID: 40063079 PMCID: PMC11893101 DOI: 10.7554/elife.100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
The reward and novelty-related neuromodulator dopamine plays an important role in hippocampal long-term memory, which is thought to involve protein-synthesis-dependent synaptic plasticity. However, the direct effects of dopamine on protein synthesis, and the functional implications of newly synthesised proteins for synaptic plasticity, have not yet been investigated. We have previously reported that timing-dependent synaptic depression (t-LTD) can be converted into potentiation by dopamine application during synaptic stimulation (Brzosko et al., 2015) or postsynaptic burst activation (Fuchsberger et al., 2022). Here, we show that dopamine increases protein synthesis in mouse hippocampal CA1 neurons, enabling dopamine-dependent long-term potentiation (DA-LTP), which is mediated via the Ca2+-sensitive adenylate cyclase (AC) subtypes 1/8, cAMP, and cAMP-dependent protein kinase (PKA). We found that neuronal activity is required for the dopamine-induced increase in protein synthesis. Furthermore, dopamine induced a protein-synthesis-dependent increase in the AMPA receptor subunit GluA1, but not GluA2. We found that DA-LTP is absent in GluA1 knock-out mice and that it requires calcium-permeable AMPA receptors. Taken together, our results suggest that dopamine together with neuronal activity controls synthesis of plasticity-related proteins, including GluA1, which enable DA-LTP via a signalling pathway distinct from that of conventional LTP.
Collapse
Affiliation(s)
- Tanja Fuchsberger
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Matty Woods
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Zuzanna Brzosko
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
7
|
Ghanavati E, Salehinejad MA, Beaupain MC, Melo L, Frese A, Kuo M, Nitsche MA. Contribution of Glutamatergic and GABAergic Mechanisms to the Plasticity-Modulating Effects of Dopamine in the Human Motor Cortex. Hum Brain Mapp 2025; 46:e70162. [PMID: 39945316 PMCID: PMC11822652 DOI: 10.1002/hbm.70162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Dopamine, a key neuromodulator in the central nervous system, regulates cortical excitability and plasticity by interacting with glutamate and GABA receptors, which are affected by dopamine receptor subtypes (D1- and D2-like). Non-invasive brain stimulation techniques can induce plasticity and monitor cortical facilitation and inhibition in humans. In a randomized, placebo-controlled, double-blinded study, we investigated how dopamine and D1- and D2-like receptors impact transcranial direct current stimulation (tDCS)-induced plasticity concerning glutamatergic and GABAergic mechanisms. Eighteen healthy volunteers received 1 mA anodal (13 min) and cathodal tDCS (9 min) over the left motor cortex combined with the dopaminergic agents l-dopa (general dopamine activation), bromocriptine (D2-like receptor agonist), combined D2 antagonism via sulpiride and general dopaminergic activation via l-dopa to activate D1-like receptors, and placebo medication. Glutamate-related cortical facilitation and GABA-related cortical inhibition were monitored using transcranial magnetic stimulation techniques, including I-O curve, intracortical facilitation (ICF), short-interval intracortical inhibition (SICI), and I-wave facilitation protocols. Our results indicate that anodal tDCS alone enhanced the I-O curve and ICF while decreasing SICI. Conversely, cathodal tDCS decreased the I-O curve and ICF while increasing SICI. General dopamine and D2 receptor activation combined with anodal tDCS decreased the I-O curve and ICF, but enhanced SICI compared to tDCS alone. When paired with cathodal tDCS, general Dopamine and D2-like receptor activity enhancement prolonged the cathodal tDCS effect on excitability. After anodal tDCS, D1-like receptor activation increased the I-O curve and ICF while reducing SICI. These effects were abolished with cathodal tDCS. Dopaminergic substances combined with anodal and cathodal tDCS did not have a significant effect on I-wave facilitation. These results suggest that D1-like receptor activation enhanced LTP-like plasticity and abolished LTD-like plasticity via glutamatergic NMDA receptor enhancement, while global dopaminergic and D2-like receptor enhancement weakened LTP-like but strengthened LTD-like plasticity primarily via glutamatergic NMDA receptor activity diminution.
Collapse
Affiliation(s)
- Elham Ghanavati
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
- Department of PsychologyRuhr University BochumBochumGermany
| | - Mohammad Ali Salehinejad
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
- School of Cognitive SciencesInstitute for Research in Fundamental SciencesTehranIran
| | - Marie C. Beaupain
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
- Department of PsychologyRuhr University BochumBochumGermany
| | - Lorena Melo
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
| | - Amba Frese
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
- International Graduate School of Neuroscience, Ruhr University BochumBochumGermany
| | - Min‐Fang Kuo
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
| | - Michael A. Nitsche
- Department of Psychology and NeurosciencesLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
- Bielefeld University, University Hospital OWL, Protestant Hospital of Bethel FoundationUniversity Clinic of Psychiatry PsychotherapyBielefeldGermany
- German Center for Mental Health (DZPG)BochumGermany
| |
Collapse
|
8
|
Pilotto A, Galli A, Sala A, Caminiti SP, Presotto L, Liguori C, Mercuri NB, Premi E, Garibotto V, Frisoni G, Chiaravalloti A, Schillaci O, D'Amelio M, Paghera B, Lucchini S, Bertagna F, Perani D, Padovani A. Dopaminergic deficits along the spectrum of Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-025-02913-5. [PMID: 39890920 DOI: 10.1038/s41380-025-02913-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Both post-mortem and in vivo data argue for dopamine dysfunction in patients with Alzheimer's Disease (AD). However, the timing and regional progression of dopaminergic systems alterations in AD are still debated. The aim of the study was to investigate in vivo the pattern of dopaminergic changes and connectivity using DAT-SPECT imaging in patients across the AD spectrum. Fifty-nine AD patients (n = 21 AD-MCI; n = 38 AD-DEM) and a control group (CG) of n = 45 age- and sex-matched individuals entered the study and underwent 123I-FP-CIT dopaminergic imaging. The occipital binding was used as reference region to obtain single-subject binding in different brain regions. Between-group differences in 123I-FP-CIT binding in both mesolimbic and nigrostriatal dopaminergic pathways were assessed using an ANCOVA test, adjusting for the effect of center of imaging acquisition, age, and sex. Regions resulting from the voxel-wise direct comparison between AD-MCI and AD-DEM were considered as a seed of interest for a voxel-wise interregional correlation analysis. Both AD-MCI and AD-DEM patients showed dopaminergic depletion within the basal ganglia, whereas cortico-limbic regions (namely hippocampus, amygdala, anterior and middle cingulate, frontal cortex and thalamus) resulted impaired only in the dementia phase. The brain voxel-wise interregional correlation analysis showed a progressive pattern of disruption of caudate/thalamus dopaminergic connectivity to hippocampus and amygdala from AD-MCI to AD-DEM stages. This study indicates basal ganglia dopaminergic alterations and connectivity disruption in the nigrostriatal and mesolimbic systems already in early stage AD, extending to several cortico-limbic regions in dementia phases.
Collapse
Affiliation(s)
- Andrea Pilotto
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy.
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy.
- Neurology Unit, Department of continuity of care and frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy.
- Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy.
| | - Alice Galli
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
| | - Arianna Sala
- Coma Science group- University of Liege, Liège, Belgium
| | | | - Luca Presotto
- Department of Physics "G. Occhialini", University of Milano-Bicocca, Milan, Italy
| | - Claudio Liguori
- Neurophysiology Unit, Sleep and Epilepsy Center- University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurophysiology Unit, Sleep and Epilepsy Center- University of Rome Tor Vergata, Rome, Italy
| | - Enrico Premi
- Stroke Unit, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Valentina Garibotto
- Department of Radiology and Medical Informatics- Geneva University Hospital, Geneva, Switzerland
| | - Giovanni Frisoni
- Department of Psychiatry- Geneva University Hospital, Geneva, Switzerland
| | | | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Barbara Paghera
- Nuclear Medicine Unit- University of Brescia, Brescia, Italy
| | - Silvia Lucchini
- Nuclear Medicine Unit- University of Brescia, Brescia, Italy
| | | | - Daniela Perani
- University Vita-Salute San Raffaele, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of continuity of care and frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| |
Collapse
|
9
|
Jäger R, Abou Sawan S, Orrú M, Tinlsey GM, Purpura M, Wells SD, Liao K, Godavarthi A. 1-Methylxanthine enhances memory and neurotransmitter levels. PLoS One 2025; 20:e0313486. [PMID: 39820866 PMCID: PMC11737676 DOI: 10.1371/journal.pone.0313486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/25/2024] [Indexed: 01/19/2025] Open
Abstract
1-Methylxanthine (1-MX) is the major metabolite of caffeine and paraxanthine and might contribute to their activity. 1-MX is an adenosine receptor antagonist and increases the release and survivability of neurotransmitters; however, no study has addressed the potential physiological effects of 1-MX ingestion. The aim of this study was to compare the effect of 1-MX on memory and related biomarkers in rats compared to control. Memory (escape latency in the Morris water maze test), neurotransmitters (acetylcholine, dopamine, gamma-amino butyric acid (GABA)), and neurochemicals (BDNF, catalase, glutathione, Amyloid Beta and cyclic GMP) were analyzed from whole brain samples in young (8-weeks-old) and aged (16-months-old) rats following 12 days of supplementation (100 mg/d HED of 1-MX [UPLEVEL®, Ingenious Ingredients L.P., Lewisville, TX, USA]) via oral gavage. 1-MX supplementation reduced escape latency by 39% in young animals and 27% in aged animals compared to controls (both p<0.001). Additionally, 1-MX increased the levels of acetylcholine, dopamine, GABA, and cyclic GMP (all p<0.001). Furthermore, 1-MX supplementation led to reduced amyloid beta and higher catalase, BDNF and glutathione concentrations (p<0.001). Collectively, our findings suggest that 1-MX may have cognitive-enhancing and neuroprotective properties.
Collapse
Affiliation(s)
- Ralf Jäger
- Ingenious Ingredients L.P., Lewisville, TX, United States of America
- Increnovo LLC, Whitefish Bay, WI, United States of America
| | | | - Marco Orrú
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States of America
| | - Grant M. Tinlsey
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States of America
| | - Martin Purpura
- Ingenious Ingredients L.P., Lewisville, TX, United States of America
- Increnovo LLC, Whitefish Bay, WI, United States of America
| | - Shawn D. Wells
- Ingenious Ingredients L.P., Lewisville, TX, United States of America
| | - Kylin Liao
- Ingenious Ingredients L.P., Lewisville, TX, United States of America
| | | |
Collapse
|
10
|
Merle L, Rastelli M, Datiche F, Véjux A, Jacquin-Piques A, Bouret SG, Benani A. Maternal Diet and Vulnerability to Cognitive Impairment in Adulthood: Possible Link with Alzheimer's Disease? Neuroendocrinology 2025; 115:242-266. [PMID: 39799941 DOI: 10.1159/000543499] [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: 09/13/2024] [Accepted: 12/15/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Aging is the main risk factor for developing cognitive impairments and associated neurodegenerative diseases. However, environmental factors, including nutritional health, are likely to promote or reduce cognitive impairments and neurodegenerative pathologies. An intricate relationship exists between maternal nutrition and adult eating behavior, metabolic phenotype, and cognitive abilities. SUMMARY The objective of the present review was to collect available data, suggesting a link between maternal overnutrition and the latter impairment of cognitive functions in the progeny, and to relate this relationship with Alzheimer's disease (AD). Indeed, cognitive impairments are major behavioral signs of AD. We first reviewed studies showing an association between unbalanced maternal diet and cognitive impairments in the progeny in humans and rodent models. Then we looked for cellular and molecular hallmarks which could constitute a breeding ground for AD in those models. With this end, we focused on synaptic dysfunction, altered neurogenesis, neuroinflammation, oxidative stress, and pathological protein aggregation. Finally, we proposed an indirect mechanism linking maternal unbalanced diet and progeny's vulnerability to cognitive impairments and neurodegeneration through promoting metabolic diseases. We also discussed the involvement of progeny's gut microbiota in the maternal diet-induced vulnerability to metabolic and neurodegenerative diseases. KEY MESSAGES Further investigations are needed to fully decipher how maternal diet programs the fetus and infant brain. Addressing this knowledge gap would pave the way to precise nutrition and personalized medicine to better handle cognitive impairments in adulthood.
Collapse
Affiliation(s)
- Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Marialetizia Rastelli
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, Inserm UMR-S1172, CHU Lille, University of Lille, Lille, France
| | - Frédérique Datiche
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Anne Véjux
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Agnès Jacquin-Piques
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Department of Clinical Neurophysiology, INRAE, Institut Agro, Université de Bourgogne, CHU Dijon, Dijon, France
| | - Sébastien G Bouret
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, Inserm UMR-S1172, CHU Lille, University of Lille, Lille, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| |
Collapse
|
11
|
Chikermane M, Weerdmeester L, Rajamani N, Köhler RM, Merk T, Vanhoecke J, Horn A, Neumann WJ. Cortical beta oscillations map to shared brain networks modulated by dopamine. eLife 2024; 13:RP97184. [PMID: 39630501 PMCID: PMC11616991 DOI: 10.7554/elife.97184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Brain rhythms can facilitate neural communication for the maintenance of brain function. Beta rhythms (13-35 Hz) have been proposed to serve multiple domains of human ability, including motor control, cognition, memory, and emotion, but the overarching organisational principles remain unknown. To uncover the circuit architecture of beta oscillations, we leverage normative brain data, analysing over 30 hr of invasive brain signals from 1772 channels from cortical areas in epilepsy patients, to demonstrate that beta is the most distributed cortical brain rhythm. Next, we identify a shared brain network from beta-dominant areas with deeper brain structures, like the basal ganglia, by mapping parametrised oscillatory peaks to whole-brain functional and structural MRI connectomes. Finally, we show that these networks share significant overlap with dopamine uptake as indicated by positron emission tomography. Our study suggests that beta oscillations emerge in cortico-subcortical brain networks that are modulated by dopamine. It provides the foundation for a unifying circuit-based conceptualisation of the functional role of beta activity beyond the motor domain and may inspire an extended investigation of beta activity as a feedback signal for closed-loop neurotherapies for dopaminergic disorders.
Collapse
Affiliation(s)
- Meera Chikermane
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Liz Weerdmeester
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Nanditha Rajamani
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Richard M Köhler
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Timon Merk
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Jojo Vanhoecke
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Andreas Horn
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
- Department of Neurology, Center for Brain Circuit Therapeutics, Brigham and Women’s HospitalBostonUnited States
- Departments of Neurology and Neurosurgery, Massachusetts General HospitalBostonUnited States
| | - Wolf Julian Neumann
- Department of Neurology, Movement Disorders and Neuromodulation Unit, Charité – Universitätsmedizin BerlinBerlinGermany
- Einstein Center for Neurosciences Berlin, Humboldt UniversitatBerlinGermany
| |
Collapse
|
12
|
Jäger R, Abou Sawan S, Orrú M, Tinsley GM, Purpura M, Wells SD, Liao K, Godavarthi A. Paraxanthine enhances memory and neuroplasticity more than caffeine in rats. Exp Brain Res 2024; 243:8. [PMID: 39617850 PMCID: PMC11609120 DOI: 10.1007/s00221-024-06954-0] [Citation(s) in RCA: 1] [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: 12/29/2023] [Accepted: 10/30/2024] [Indexed: 02/05/2025]
Abstract
Paraxanthine (PXN) is the main metabolite of caffeine (CAF). PXN supplementation has been shown to increase measures of cognition, memory, reasoning, response time, and sustained attention; however, no preclinical study has compared the effects of PXN with those of CAF. The aim of this study was to compare the effects of PXN and CAF on memory and related biomarkers in rats. The effects of two different doses of PXN (PXN LOW, PXN HIGH), CAF (CAF HIGH), and a control group on cognition (escape latency in the Morris water maze test), neurotransmitters (acetylcholine, dopamine, and gamma-aminobutyric acid), and neurochemicals (BDNF, catalase, glutathione, and cyclic GMP) were analyzed from whole brain samples in young (8 weeks old) and aged (16 months old) rats. Compared to the control group, escape latency improved in PXN LOW, PXN HIGH, and CAF HIGH (all P < 0.05) in young animals, and in PXN HIGH and CAF HIGH in older animals (P < 0.001). PXN HIGH improved escape latency compared to CAF HIGH in both young (P < 0.001) and old animals (P = 0.003). BDNF levels increased in PXN LOW, PXN HIGH, and CAF HIGH (all P < 0.001), with PXN HIGH increasing BDNF to a greater extent compared to CAF HIGH (P = 0.03). PXN HIGH also significantly increased BDNF levels compared to PXN LOW (P < 0.001). All other neurotransmitters and neurochemicals significantly increased in the PXN HIGH and CAF HIGH groups compared to the control. In conclusion, PXN showed greater improvements in cognition and BDNF levels compared to CAF, further substantiating PXN as a nootropic with greater benefits compared to CAF.
Collapse
Affiliation(s)
- Ralf Jäger
- Ingenious Ingredients L.P, Lewisville, TX, 75056, USA.
- Increnovo LLC, Whitefish Bay, WI, 53217, USA.
| | | | - Marco Orrú
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Grant M Tinsley
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, USA
| | - Martin Purpura
- Ingenious Ingredients L.P, Lewisville, TX, 75056, USA
- Increnovo LLC, Whitefish Bay, WI, 53217, USA
| | - Shawn D Wells
- Ingenious Ingredients L.P, Lewisville, TX, 75056, USA
| | - Kylin Liao
- Ingenious Ingredients L.P, Lewisville, TX, 75056, USA
| | | |
Collapse
|
13
|
Lima KR, Rosa ACDSD, Gomes GCM, Sigaran GJ, Perretto AC, Mello-Carpes PB. Acute exercise performed during the late consolidation phase improves memory persistence by hippocampal protein synthesis and catecholamine modulation. Pharmacol Biochem Behav 2024; 245:173893. [PMID: 39419355 DOI: 10.1016/j.pbb.2024.173893] [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: 04/22/2024] [Revised: 08/13/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Memory persistence is a crucial aspect of long-term memory (LTM) and involves late consolidation processes that modulate memory stability over time. Acute physical exercise (PE) has emerged as a potential strategy to modulate memory consolidation and enhance memory persistence. While its effects have been extensively explored in the early consolidation phase, its impact on the late phase remains unexplored. In this study, we investigated the effects and mechanisms of an acute PE on the late consolidation window of novel object recognition (NOR) memory in rats. A 30-minute running session applied 11 h after NOR memory acquisition significantly increased memory persistence for at least 7 days. The inhibition of hippocampal protein synthesis immediately after acute PE using anisomycin (a ribosomal inhibitor) or rapamycin (an mTOR pathway inhibitor) impaired the effect of PE on memory persistence. Animals only presented memory 1 day after acquisition. The same effect was observed with the inhibition of beta-adrenergic receptors by timolol. Although there were no differences between the groups' comparison, blocking D1/D5 receptors after acute PE resulted in a lack of memory persistence in the dichotomous testing (remember/non-remember). Therefore, our exploration of the mechanisms underlying this enhancement revealed the involvement of protein synthesis and the requirement of beta-adrenergic and dopaminergic D1/D5 receptors in the dorsal hippocampus. These findings provide valuable insights into PE as a potential memory modulator, contributing to expanding our understanding of memory consolidation dynamics and acute PE effects.
Collapse
Affiliation(s)
- Karine Ramires Lima
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | | | | | - Gabriela Jaques Sigaran
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Anna Cecilia Perretto
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil.
| |
Collapse
|
14
|
Kleinman MR, Foster DJ. Spatial localization of hippocampal replay requires dopamine signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597435. [PMID: 38895442 PMCID: PMC11185723 DOI: 10.1101/2024.06.04.597435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Sequenced reactivations of hippocampal neurons called replays, concomitant with sharp-wave ripples in the local field potential, are critical for the consolidation of episodic memory, but whether replays depend on the brain's reward or novelty signals is unknown. Here we combined chemogenetic silencing of dopamine neurons in ventral tegmental area (VTA) and simultaneous electrophysiological recordings in dorsal hippocampal CA1, in freely behaving male rats experiencing changes to reward magnitude and environmental novelty. Surprisingly, VTA silencing did not prevent ripple increases where reward was increased, but caused dramatic, aberrant ripple increases where reward was unchanged. These increases were associated with increased reverse-ordered replays. On familiar tracks this effect disappeared, and ripples tracked reward prediction error, indicating that non-VTA reward signals were sufficient to direct replay. Our results reveal a novel dependence of hippocampal replay on dopamine, and a role for a VTA-independent reward prediction error signal that is reliable only in familiar environments.
Collapse
Affiliation(s)
- Matthew R Kleinman
- Helen Wills Neuroscience Institute and Department of Psychology, University of California, Berkeley, CA 94720, USA
| | - David J Foster
- Helen Wills Neuroscience Institute and Department of Psychology, University of California, Berkeley, CA 94720, USA
- Lead contact
| |
Collapse
|
15
|
Sulaman BA, Zhang Y, Matosevich N, Kjærby C, Foustoukos G, Andersen M, Eban-Rothschild A. Emerging Functions of Neuromodulation during Sleep. J Neurosci 2024; 44:e1277242024. [PMID: 39358018 PMCID: PMC11450531 DOI: 10.1523/jneurosci.1277-24.2024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 10/04/2024] Open
Abstract
Neuromodulators act on multiple timescales to affect neuronal activity and behavior. They function as synaptic fine-tuners and master coordinators of neuronal activity across distant brain regions and body organs. While much research on neuromodulation has focused on roles in promoting features of wakefulness and transitions between sleep and wake states, the precise dynamics and functions of neuromodulatory signaling during sleep have received less attention. This review discusses research presented at our minisymposium at the 2024 Society for Neuroscience meeting, highlighting how norepinephrine, dopamine, and acetylcholine orchestrate brain oscillatory activity, control sleep architecture and microarchitecture, regulate responsiveness to sensory stimuli, and facilitate memory consolidation. The potential of each neuromodulator to influence neuronal activity is shaped by the state of the synaptic milieu, which in turn is influenced by the organismal or systemic state. Investigating the effects of neuromodulator release across different sleep substates and synaptic environments offers unique opportunities to deepen our understanding of neuromodulation and explore the distinct computational opportunities that arise during sleep. Moreover, since alterations in neuromodulatory signaling and sleep are implicated in various neuropsychiatric disorders and because existing pharmacological treatments affect neuromodulatory signaling, gaining a deeper understanding of the less-studied aspects of neuromodulators during sleep is of high importance.
Collapse
Affiliation(s)
- Bibi Alika Sulaman
- Department of Psychology, University of Michigan, Ann Arbor, Michigan 48109
| | - Yiyao Zhang
- Neuroscience Institute, New York University, New York, New York 10016
| | - Noa Matosevich
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Celia Kjærby
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Georgios Foustoukos
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne 1005, Switzerland
| | - Mie Andersen
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | | |
Collapse
|
16
|
Dubinsky JM, Hamid AA. The neuroscience of active learning and direct instruction. Neurosci Biobehav Rev 2024; 163:105737. [PMID: 38796122 DOI: 10.1016/j.neubiorev.2024.105737] [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: 12/19/2023] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Throughout the educational system, students experiencing active learning pedagogy perform better and fail less than those taught through direct instruction. Can this be ascribed to differences in learning from a neuroscientific perspective? This review examines mechanistic, neuroscientific evidence that might explain differences in cognitive engagement contributing to learning outcomes between these instructional approaches. In classrooms, direct instruction comprehensively describes academic content, while active learning provides structured opportunities for learners to explore, apply, and manipulate content. Synaptic plasticity and its modulation by arousal or novelty are central to all learning and both approaches. As a form of social learning, direct instruction relies upon working memory. The reinforcement learning circuit, associated agency, curiosity, and peer-to-peer social interactions combine to enhance motivation, improve retention, and build higher-order-thinking skills in active learning environments. When working memory becomes overwhelmed, additionally engaging the reinforcement learning circuit improves retention, providing an explanation for the benefits of active learning. This analysis provides a mechanistic examination of how emerging neuroscience principles might inform pedagogical choices at all educational levels.
Collapse
Affiliation(s)
- Janet M Dubinsky
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| | - Arif A Hamid
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
17
|
Faress I, Khalil V, Hou WH, Moreno A, Andersen N, Fonseca R, Piriz J, Capogna M, Nabavi S. Non-Hebbian plasticity transforms transient experiences into lasting memories. eLife 2024; 12:RP91421. [PMID: 39023519 PMCID: PMC11257676 DOI: 10.7554/elife.91421] [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: 07/20/2024] Open
Abstract
The dominant models of learning and memory, such as Hebbian plasticity, propose that experiences are transformed into memories through input-specific synaptic plasticity at the time of learning. However, synaptic plasticity is neither strictly input-specific nor restricted to the time of its induction. The impact of such forms of non-Hebbian plasticity on memory has been difficult to test, and hence poorly understood. Here, we demonstrate that synaptic manipulations can deviate from the Hebbian model of learning, yet produce a lasting memory. First, we established a weak associative conditioning protocol in mice, where optogenetic stimulation of sensory thalamic input to the amygdala was paired with a footshock, but no detectable memory was formed. However, when the same input was potentiated minutes before or after, or even 24 hr later, the associative experience was converted into a lasting memory. Importantly, potentiating an independent input to the amygdala minutes but not 24 hr after the pairing produced a lasting memory. Thus, our findings suggest that the process of transformation of a transient experience into a memory is neither restricted to the time of the experience nor to the synapses triggered by it; instead, it can be influenced by past and future events.
Collapse
Affiliation(s)
- Islam Faress
- Department of Molecular Biology and Genetics, Aarhus UniversityAahrusDenmark
- Department of Biomedicine, Aarhus UniversityAarhusDenmark
- DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus UniversityAahrusDenmark
- Center for Proteins in Memory – PROMEMO, Danish National Research Foundation, Aarhus UniversityAahrusDenmark
| | - Valentina Khalil
- Department of Molecular Biology and Genetics, Aarhus UniversityAahrusDenmark
- DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus UniversityAahrusDenmark
- Center for Proteins in Memory – PROMEMO, Danish National Research Foundation, Aarhus UniversityAahrusDenmark
| | - Wen-Hsien Hou
- Department of Biomedicine, Aarhus UniversityAarhusDenmark
| | - Andrea Moreno
- Department of Molecular Biology and Genetics, Aarhus UniversityAahrusDenmark
- DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus UniversityAahrusDenmark
- Center for Proteins in Memory – PROMEMO, Danish National Research Foundation, Aarhus UniversityAahrusDenmark
| | - Niels Andersen
- Department of Molecular Biology and Genetics, Aarhus UniversityAahrusDenmark
- DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus UniversityAahrusDenmark
- Center for Proteins in Memory – PROMEMO, Danish National Research Foundation, Aarhus UniversityAahrusDenmark
| | - Rosalina Fonseca
- Cellular and Systems Neurobiology, Universidade Nova de LisboaLisbonPortugal
| | - Joaquin Piriz
- Instituto de Fisiología Biología Molecular y Neurociencias (IFIBYNE), Universidad de Buenos AiresBuenos AiresArgentina
| | - Marco Capogna
- Department of Biomedicine, Aarhus UniversityAarhusDenmark
- Center for Proteins in Memory – PROMEMO, Danish National Research Foundation, Aarhus UniversityAahrusDenmark
| | - Sadegh Nabavi
- Department of Molecular Biology and Genetics, Aarhus UniversityAahrusDenmark
- DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus UniversityAahrusDenmark
- Center for Proteins in Memory – PROMEMO, Danish National Research Foundation, Aarhus UniversityAahrusDenmark
| |
Collapse
|
18
|
Brito DVC, Kupke J, Sokolov R, Cambridge S, Both M, Bengtson CP, Rozov A, Oliveira AMM. Biphasic Npas4 expression promotes inhibitory plasticity and suppression of fear memory consolidation in mice. Mol Psychiatry 2024; 29:1929-1940. [PMID: 38347124 PMCID: PMC11408256 DOI: 10.1038/s41380-024-02454-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/19/2024]
Abstract
Long-term memories are believed to be encoded by unique transcriptional signatures in the brain. The expression of immediate early genes (IEG) promotes structural and molecular changes required for memory consolidation. Recent evidence has shown that the brain is equipped with mechanisms that not only promote, but actively constrict memory formation. However, it remains unknown whether IEG expression may play a role in memory suppression. Here we uncovered a novel function of the IEG neuronal PAS domain protein 4 (Npas4), as an inducible memory suppressor gene of highly salient aversive experiences. Using a contextual fear conditioning paradigm, we found that low stimulus salience leads to monophasic Npas4 expression, while highly salient learning induces a biphasic expression of Npas4 in the hippocampus. The later phase requires N-methyl-D-aspartate (NMDA) receptor activity and is independent of dopaminergic neurotransmission. Our in vivo pharmacological and genetic manipulation experiments suggested that the later phase of Npas4 expression restricts the consolidation of a fear memory and promote behavioral flexibility, by facilitating fear extinction and the contextual specificity of fear responses. Moreover, immunofluorescence and electrophysiological analysis revealed a concomitant increase in synaptic input from cholecystokinin (CCK)-expressing interneurons. Our results demonstrate how salient experiences evoke unique temporal patterns of IEG expression that fine-tune memory consolidation. Moreover, our study provides evidence for inducible gene expression associated with memory suppression as a possible mechanism to balance the consolidation of highly salient memories, and thereby to evade the formation of maladaptive behavior.
Collapse
Affiliation(s)
- David V C Brito
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139, Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve, 8005-139, Faro, Portugal
| | - Janina Kupke
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, the Netherlands
| | - Rostilav Sokolov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia
- Federal Center of Brain Research and Neurotechnology, 117513, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny, Novgorod, Russia
| | - Sidney Cambridge
- Anatomy II, Dr. Senckenberg Anatomy, Goethe-University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Martin Both
- Institute of Physiology and Pathophysiology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - Andrei Rozov
- Federal Center of Brain Research and Neurotechnology, 117513, Moscow, Russia
- Institute of Physiology and Pathophysiology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
- OpenLab of Neurobiology, Kazan Federal University, 420008, Kazan, Russia
| | - Ana M M Oliveira
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany.
- Department of Molecular and Cellular Cognition Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.
| |
Collapse
|
19
|
Schroyens N, Vercammen L, Özcan B, Salazar VAO, Zaman J, De Bundel D, Beckers T, Luyten L. No evidence that post-training dopamine D2 receptor agonism affects fear generalization in male rats. J Psychopharmacol 2024; 38:672-682. [PMID: 39068641 PMCID: PMC7616352 DOI: 10.1177/02698811241261375] [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] [Indexed: 07/30/2024]
Abstract
BACKGROUND The neurotransmitter dopamine plays an important role in the processing of emotional memories, and prior research suggests that dopaminergic manipulations immediately after fear learning can affect the retention and generalization of acquired fear. AIMS The current study focuses specifically on the role of dopamine D2 receptors (D2Rs) regarding fear generalization in adult, male Wistar rats, and aims to replicate previous findings in mice. METHODS In a series of five experiments, D2R (ant)agonists were injected systemically, immediately after differential cued fear conditioning (CS+ followed by shock, CS- without shock). All five experiments involved the administration of the D2R agonist quinpirole at different doses versus saline (n = 12, 16, or 44 rats/group). In addition, one of the studies administered the D2R antagonist raclopride (n = 12). One day later, freezing during the CS+ and CS- was assessed. RESULTS We found no indications for an effect of quinpirole or raclopride on fear generalization during this drug-free test. Importantly, and contradicting earlier research in mice, the evidence for the absence of an effect of D2R agonist quinpirole (1 mg/kg) on fear generalization was substantial according to Bayesian analyses and was observed in a highly powered experiment (N = 87). We did find acute behavioral effects in line with the literature, for both quinpirole and raclopride in a locomotor activity test. CONCLUSION In contrast with prior studies in mice, we have obtained evidence against a preventative effect of post-training D2R agonist quinpirole administration on subsequent fear generalization in rats.
Collapse
Affiliation(s)
- Natalie Schroyens
- KU Leuven, Centre for the Psychology of Learning and Experimental Psychopathology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3712, 3000Leuven, Belgium
- KU Leuven, Leuven Brain Institute, O&N V Herestraat 49 box 1020, 3000Leuven, Belgium
| | - Laura Vercammen
- KU Leuven, Centre for the Psychology of Learning and Experimental Psychopathology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3712, 3000Leuven, Belgium
- KU Leuven, Leuven Brain Institute, O&N V Herestraat 49 box 1020, 3000Leuven, Belgium
- KU Leuven, Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3714, 3000Leuven, Belgium
| | - Burcu Özcan
- KU Leuven, Centre for the Psychology of Learning and Experimental Psychopathology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3712, 3000Leuven, Belgium
| | - Victoria Aurora Ossorio Salazar
- KU Leuven, Leuven Brain Institute, O&N V Herestraat 49 box 1020, 3000Leuven, Belgium
- KU Leuven, Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3714, 3000Leuven, Belgium
| | - Jonas Zaman
- KU Leuven, Health Psychology, Tiensestraat 102 box 3726, 3000Leuven, Belgium
| | - Dimitri De Bundel
- Research Group Experimental Pharmacology, Department of Pharmaceutical Sciences, Center for Neurosciences, Vrije Universiteit Brussel, 1090Brussel, Belgium
| | - Tom Beckers
- KU Leuven, Centre for the Psychology of Learning and Experimental Psychopathology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3712, 3000Leuven, Belgium
- KU Leuven, Leuven Brain Institute, O&N V Herestraat 49 box 1020, 3000Leuven, Belgium
| | - Laura Luyten
- KU Leuven, Centre for the Psychology of Learning and Experimental Psychopathology, Faculty of Psychology and Educational Sciences, Tiensestraat 102 box 3712, 3000Leuven, Belgium
- KU Leuven, Leuven Brain Institute, O&N V Herestraat 49 box 1020, 3000Leuven, Belgium
| |
Collapse
|
20
|
Rappeneau V, Castillo Díaz F. Convergence of oxytocin and dopamine signalling in neuronal circuits: Insights into the neurobiology of social interactions across species. Neurosci Biobehav Rev 2024; 161:105675. [PMID: 38608828 DOI: 10.1016/j.neubiorev.2024.105675] [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: 01/31/2024] [Revised: 03/21/2024] [Accepted: 04/10/2024] [Indexed: 04/14/2024]
Abstract
Social behaviour is essential for animal survival, and the hypothalamic neuropeptide oxytocin (OXT) critically impacts bonding, parenting, and decision-making. Dopamine (DA), is released by ventral tegmental area (VTA) dopaminergic neurons, regulating social cues in the mesolimbic system. Despite extensive exploration of OXT and DA roles in social behaviour independently, limited studies investigate their interplay. This narrative review integrates insights from human and animal studies, particularly rodents, emphasising recent research on pharmacological manipulations of OXT or DA systems in social behaviour. Additionally, we review studies correlating social behaviour with blood/cerebral OXT and DA levels. Behavioural facets include sociability, cooperation, pair bonding and parental care. In addition, we provide insights into OXT-DA interplay in animal models of social stress, autism, and schizophrenia. Emphasis is placed on the complex relationship between the OXT and DA systems and their collective influence on social behaviour across physiological and pathological conditions. Understanding OXT and DA imbalance is fundamental for unravelling the neurobiological underpinnings of social interaction and reward processing deficits observed in psychiatric conditions.
Collapse
Affiliation(s)
- Virginie Rappeneau
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany.
| | - Fernando Castillo Díaz
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany
| |
Collapse
|
21
|
Sayegh FJP, Mouledous L, Macri C, Pi Macedo J, Lejards C, Rampon C, Verret L, Dahan L. Ventral tegmental area dopamine projections to the hippocampus trigger long-term potentiation and contextual learning. Nat Commun 2024; 15:4100. [PMID: 38773091 PMCID: PMC11109191 DOI: 10.1038/s41467-024-47481-4] [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: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
In most models of neuronal plasticity and memory, dopamine is thought to promote the long-term maintenance of Long-Term Potentiation (LTP) underlying memory processes, but not the initiation of plasticity or new information storage. Here, we used optogenetic manipulation of midbrain dopamine neurons in male DAT::Cre mice, and discovered that stimulating the Schaffer collaterals - the glutamatergic axons connecting CA3 and CA1 regions - of the dorsal hippocampus concomitantly with midbrain dopamine terminals within a 200 millisecond time-window triggers LTP at glutamatergic synapses. Moreover, we showed that the stimulation of this dopaminergic pathway facilitates contextual learning in awake behaving mice, while its inhibition hinders it. Thus, activation of midbrain dopamine can operate as a teaching signal that triggers NeoHebbian LTP and promotes supervised learning.
Collapse
Affiliation(s)
- Fares J P Sayegh
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France.
| | - Lionel Mouledous
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France
| | - Catherine Macri
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France
| | - Juliana Pi Macedo
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France
| | - Camille Lejards
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France
| | - Laure Verret
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse, France.
| |
Collapse
|
22
|
Velazquez-Delgado C, Perez-Becerra J, Calderon V, Hernandez-Ortiz E, Bermudez-Rattoni F, Carrillo-Reid L. Paradoxical Boosting of Weak and Strong Spatial Memories by Hippocampal Dopamine Uncaging. eNeuro 2024; 11:ENEURO.0469-23.2024. [PMID: 38755011 PMCID: PMC11138129 DOI: 10.1523/eneuro.0469-23.2024] [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: 11/03/2023] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
The ability to remember changes in the surroundings is fundamental for daily life. It has been proposed that novel events producing dopamine release in the hippocampal CA1 region could modulate spatial memory formation. However, the role of hippocampal dopamine increase on weak or strong spatial memories remains unclear. We show that male mice exploring two objects located in a familiar environment for 5 min created a short-term memory (weak) that cannot be retrieved 1 d later, whereas 10 min exploration created a long-term memory (strong) that can be retrieved 1 d later. Remarkably, hippocampal dopamine elevation during the encoding of weak object location memories (OLMs) allowed their retrieval 1 d later but dopamine elevation during the encoding of strong OLMs promoted the preference for a familiar object location over a novel object location after 24 h. Moreover, dopamine uncaging after the encoding of OLMs did not have effect on weak memories whereas on strong memories diminished the exploration of the novel object location. Additionally, hippocampal dopamine elevation during the retrieval of OLMs did not allow the recovery of weak memories and did not affect the retrieval of strong memory traces. Finally, dopamine elevation increased hippocampal theta oscillations, indicating that dopamine promotes the recurrent activation of specific groups of neurons. Our experiments demonstrate that hippocampal dopaminergic modulation during the encoding of OLMs depends on memory strength indicating that hyperdopaminergic levels that enhance weak experiences could compromise the normal storage of strong memories.
Collapse
Affiliation(s)
| | - Job Perez-Becerra
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, México
| | - Vladimir Calderon
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, México
| | - Eduardo Hernandez-Ortiz
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México 04510, México
| | - Federico Bermudez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México 04510, México
| | - Luis Carrillo-Reid
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla 76230, México
| |
Collapse
|
23
|
Wilmot JH, Diniz CRAF, Crestani AP, Puhger KR, Roshgadol J, Tian L, Wiltgen BJ. Phasic locus coeruleus activity enhances trace fear conditioning by increasing dopamine release in the hippocampus. eLife 2024; 12:RP91465. [PMID: 38592773 PMCID: PMC11003744 DOI: 10.7554/elife.91465] [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: 04/10/2024] Open
Abstract
Locus coeruleus (LC) projections to the hippocampus play a critical role in learning and memory. However, the precise timing of LC-hippocampus communication during learning and which LC-derived neurotransmitters are important for memory formation in the hippocampus are currently unknown. Although the LC is typically thought to modulate neural activity via the release of norepinephrine, several recent studies have suggested that it may also release dopamine into the hippocampus and other cortical regions. In some cases, it appears that dopamine release from LC into the hippocampus may be more important for memory than norepinephrine. Here, we extend these data by characterizing the phasic responses of the LC and its projections to the dorsal hippocampus during trace fear conditioning in mice. We find that the LC and its projections to the hippocampus respond to task-relevant stimuli and that amplifying these responses with optogenetic stimulation can enhance long-term memory formation. We also demonstrate that LC activity increases both norepinephrine and dopamine content in the dorsal hippocampus and that the timing of hippocampal dopamine release during trace fear conditioning is similar to the timing of LC activity. Finally, we show that hippocampal dopamine is important for trace fear memory formation, while norepinephrine is not.
Collapse
Affiliation(s)
- Jacob H Wilmot
- Department of Psychology, University of California, DavisDavisUnited States
- Center for Neuroscience, University of California, DavisDavisUnited States
| | - Cassiano RAF Diniz
- Center for Neuroscience, University of California, DavisDavisUnited States
| | - Ana P Crestani
- Center for Neuroscience, University of California, DavisDavisUnited States
| | - Kyle R Puhger
- Department of Psychology, University of California, DavisDavisUnited States
- Center for Neuroscience, University of California, DavisDavisUnited States
| | - Jacob Roshgadol
- Center for Neuroscience, University of California, DavisDavisUnited States
- Department of Biomedical Engineering, University of California, DavisDavisUnited States
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, University of California, DavisDavisUnited States
| | - Brian Joseph Wiltgen
- Department of Psychology, University of California, DavisDavisUnited States
- Center for Neuroscience, University of California, DavisDavisUnited States
| |
Collapse
|
24
|
Rocha GS, Freire MAM, Paiva KM, Oliveira RF, Morais PLAG, Santos JR, Cavalcanti JRLP. The neurobiological effects of senescence on dopaminergic system: A comprehensive review. J Chem Neuroanat 2024; 137:102415. [PMID: 38521203 DOI: 10.1016/j.jchemneu.2024.102415] [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: 12/26/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
Over time, the body undergoes a natural, multifactorial, and ongoing process named senescence, which induces changes at the molecular, cellular, and micro-anatomical levels in many body systems. The brain, being a highly complex organ, is particularly affected by this process, potentially impairing its numerous functions. The brain relies on chemical messengers known as neurotransmitters to function properly, with dopamine being one of the most crucial. This catecholamine is responsible for a broad range of critical roles in the central nervous system, including movement, learning, cognition, motivation, emotion, reward, hormonal release, memory consolidation, visual performance, sexual drive, modulation of circadian rhythms, and brain development. In the present review, we thoroughly examine the impact of senescence on the dopaminergic system, with a primary focus on the classic delimitations of the dopaminergic nuclei from A8 to A17. We provide in-depth information about their anatomy and function, particularly addressing how senescence affects each of these nuclei.
Collapse
Affiliation(s)
- Gabriel S Rocha
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | - Marco Aurelio M Freire
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | - Karina M Paiva
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - Rodrigo F Oliveira
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - Paulo Leonardo A G Morais
- Laboratory of Experimental Neurology, State University of Rio Grande do Norte (UERN), Mossoró, Brazil
| | - José Ronaldo Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Federal University of Sergipe (UFS), Itabaiana, Brazil
| | | |
Collapse
|
25
|
Zhao F, Li C, Zhuang Y, Yan Y, Gao Y, Behnisch T. Apoptosis signal-regulating kinase 1 ( Ask1) deficiency alleviates MPP +-induced impairment of evoked dopamine release in the mouse hippocampus. Front Cell Neurosci 2024; 18:1288991. [PMID: 38414754 PMCID: PMC10896914 DOI: 10.3389/fncel.2024.1288991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/19/2024] [Indexed: 02/29/2024] Open
Abstract
The dopaminergic system is susceptible to dysfunction in numerous neurological diseases, including Parkinson's disease (PD). In addition to motor symptoms, some PD patients may experience non-motor symptoms, including cognitive and memory deficits. A possible explanation for their manifestation is a disturbed pattern of dopamine release in brain regions involved in learning and memory, such as the hippocampus. Therefore, investigating neuropathological alterations in dopamine release prior to neurodegeneration is imperative. This study aimed to characterize evoked hippocampal dopamine release and assess the impact of the neurotoxin MPP+ using a genetically encoded dopamine sensor and gene expression analysis. Additionally, considering the potential neuroprotective attributes demonstrated by apoptosis signal-regulating kinase 1 (Ask1) in various animal-disease-like models, the study also aimed to determine whether Ask1 knockdown restores MPP+-altered dopamine release in acute hippocampal slices. We applied variations of low- and high-frequency stimulation to evoke dopamine release within different hippocampal regions and discovered that acute application of MPP+ reduced the amount of dopamine released and hindered the recovery of dopamine release after repeated stimulation. In addition, we observed that Ask1 deficiency attenuated the detrimental effects of MPP+ on the recovery of dopamine release after repeated stimulation. RNA sequencing analysis indicated that genes associated with the synaptic pathways are involved in response to MPP+ exposure. Notably, Ask1 deficiency was found to downregulate the expression of Slc5a7, a gene encoding a sodium-dependent high-affinity choline transporter that regulates acetylcholine levels. Respective follow-up experiments indicated that Slc5a7 plays a role in Ask1 deficiency-mediated protection against MPP+ neurotoxicity. In addition, increasing acetylcholine levels using an acetylcholinesterase inhibitor could exacerbate the toxicity of MPP+. In conclusion, our data imply that the modulation of the dopamine-acetylcholine balance may be a crucial mechanism of action underlying the neuroprotective effects of Ask1 deficiency in PD.
Collapse
Affiliation(s)
- Fang Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chuhan Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yinghan Zhuang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yan Yan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Lazarova M, Tsvetanova E, Georgieva A, Stefanova M, Uzunova D, Denev P, Vassileva V, Tasheva K. Extracts of Sideritis scardica and Clinopodium vulgare Alleviate Cognitive Impairments in Scopolamine-Induced Rat Dementia. Int J Mol Sci 2024; 25:1840. [PMID: 38339117 PMCID: PMC10855470 DOI: 10.3390/ijms25031840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Sideritis scardica Griseb. and Clinopodium vulgare L., belonging to the Lamiaceae family, are rich in terpenoids and phenolics and exhibit various pharmacological effects, including antioxidant, anti-inflammatory and anti-cancer activities. While the memory-enhancing impacts of S. scardica are well documented, the cognitive benefits of C. vulgare remain unexplored. This study assessed the potential effect of C. vulgare on learning and memory in healthy and scopolamine (Sco)-induced memory-impaired male Wistar rats, comparing it with the effects of S. scardica. Over a 21-day period, rats orally received extracts of cultivated S. scardica (200 mg/kg) and C. vulgare (100 mg/kg), either individually or in combination, with administration starting 10 days before and continuing 11 days simultaneously with Sco injection at a dose of 2 mg/kg intraperitoneally. The results showed that both extracts effectively mitigated Sco-induced memory impairment. Their combination significantly improved recognition memory and maintained monoaminergic function. S. scardica excelled in preserving spatial working memory, while C. vulgare exhibited comparable retention of recognition memory, robust antioxidant activity and acetylcholinesterase inhibitory activity. The extracts alleviated Sco-induced downregulation of p-CREB/BDNF signaling, suggesting neuroprotective mechanisms. The extract combination positively affected most of the Sco-induced impairments, underscoring the potential for further investigation of these extracts for therapeutic development.
Collapse
Affiliation(s)
- Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Petko Denev
- Laboratory of Biologically Active Substances, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 4000 Plovdiv, Bulgaria
| | - Valya Vassileva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 21, 1113 Sofia, Bulgaria;
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 21, 1113 Sofia, Bulgaria;
| |
Collapse
|
27
|
Xu S, Ren W. Distinct processing of the state prediction error signals in frontal and parietal correlates in learning the environment model. Cereb Cortex 2024; 34:bhad449. [PMID: 38037370 DOI: 10.1093/cercor/bhad449] [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: 09/18/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Goal-directed reinforcement learning constructs a model of how the states in the environment are connected and prospectively evaluates action values by simulating experience. State prediction error (SPE) is theorized as a crucial signal for learning the environment model. However, the underlying neural mechanisms remain unclear. Here, using electroencephalogram, we verified in a two-stage Markov task two neural correlates of SPEs: an early negative correlate transferring from frontal to central electrodes and a late positive correlate over parietal regions. Furthermore, by investigating the effects of explicit knowledge about the environment model and rewards in the environment, we found that, for the parietal correlate, rewards enhanced the representation efficiency (beta values of regression coefficient) of SPEs, whereas explicit knowledge elicited a larger SPE representation (event-related potential activity) for rare transitions. However, for the frontal and central correlates, rewards increased activities in a content-independent way and explicit knowledge enhanced activities only for common transitions. Our results suggest that the parietal correlate of SPEs is responsible for the explicit learning of state transition structure, whereas the frontal and central correlates may be involved in cognitive control. Our study provides novel evidence for distinct roles of the frontal and the parietal cortices in processing SPEs.
Collapse
Affiliation(s)
- Shuyuan Xu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wei Ren
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
- Faculty of Education, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
28
|
Bénac N, Ezequiel Saraceno G, Butler C, Kuga N, Nishimura Y, Yokoi T, Su P, Sasaki T, Petit-Pedrol M, Galland R, Studer V, Liu F, Ikegaya Y, Sibarita JB, Groc L. Non-canonical interplay between glutamatergic NMDA and dopamine receptors shapes synaptogenesis. Nat Commun 2024; 15:27. [PMID: 38167277 PMCID: PMC10762086 DOI: 10.1038/s41467-023-44301-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Direct interactions between receptors at the neuronal surface have long been proposed to tune signaling cascades and neuronal communication in health and disease. Yet, the lack of direct investigation methods to measure, in live neurons, the interaction between different membrane receptors at the single molecule level has raised unanswered questions on the biophysical properties and biological roles of such receptor interactome. Using a multidimensional spectral single molecule-localization microscopy (MS-SMLM) approach, we monitored the interaction between two membrane receptors, i.e. glutamatergic NMDA (NMDAR) and G protein-coupled dopamine D1 (D1R) receptors. The transient interaction was randomly observed along the dendritic tree of hippocampal neurons. It was higher early in development, promoting the formation of NMDAR-D1R complexes in an mGluR5- and CK1-dependent manner, favoring NMDAR clusters and synaptogenesis in a dopamine receptor signaling-independent manner. Preventing the interaction in the neonate, and not adult, brain alters in vivo spontaneous neuronal network activity pattern in male mice. Thus, a weak and transient interaction between NMDAR and D1R plays a structural and functional role in the developing brain.
Collapse
Affiliation(s)
- Nathan Bénac
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | | | - Corey Butler
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Nahoko Kuga
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | - Yuya Nishimura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taiki Yokoi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Takuya Sasaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | | | - Rémi Galland
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Vincent Studer
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Center for Information and Neural Networks, Suita City, Osaka, 565-0871, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Laurent Groc
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
29
|
Ciampa CJ, Morin TM, Murphy A, Joie RL, Landau SM, Berry AS. DAT1 and BDNF polymorphisms interact to predict Aβ and tau pathology. Neurobiol Aging 2024; 133:115-124. [PMID: 37948982 PMCID: PMC10872994 DOI: 10.1016/j.neurobiolaging.2023.10.009] [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: 05/05/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023]
Abstract
Previous work has associated polymorphisms in the dopamine transporter gene (rs6347 in DAT1/SLC6A3) and brain derived neurotrophic factor gene (Val66Met in BDNF) with atrophy and memory decline. However, it is unclear whether these polymorphisms relate to atrophy and cognition through associations with Alzheimer's disease pathology. We tested for effects of DAT1 and BDNF polymorphisms on cross-sectional and longitudinal β-amyloid (Aβ) and tau pathology (measured with positron emission tomography (PET)), hippocampal volume, and cognition. We analyzed a sample of cognitively normal older adults (cross-sectional n = 321) from the Alzheimer's Disease Neuroimaging Initiative (ADNI). DAT1 and BDNF interacted to predict Aβ-PET, tau-PET, and hippocampal atrophy. Carriers of both "non-boptimal" DAT1 C and BDNF Met alleles demonstrated greater pathology and atrophy. Our findings provide novel links between dopamine and neurotrophic factor genes and AD pathology, consistent with previous research implicating these variants in greater risk for developing AD.
Collapse
Affiliation(s)
- Claire J Ciampa
- Department of Biology, Brandeis University, Waltham, MA 02453, USA.
| | - Thomas M Morin
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA 02155, USA
| | - Alice Murphy
- Hellen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Susan M Landau
- Hellen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
30
|
Lima KR, Alves N, Lopes LF, Picua SS, da Silva de Vargas L, Daré LR, Ramborger B, Roehrs R, de Gomes MG, Mello-Carpes PB. Novelty facilitates the persistence of aversive memory extinction by dopamine regulation in the hippocampus and ventral tegmental area. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110832. [PMID: 37463639 DOI: 10.1016/j.pnpbp.2023.110832] [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: 05/10/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/20/2023]
Abstract
Aversive memory extinction comprises a novel learning that blocks retrieving a previously formed traumatic memory. In this sense, aversive memory extinction is an excellent tool for decreasing fear responses. However, this tool it's not effective in the long term because of original memory spontaneous recovery. Thus, searching for alternative strategies that strengthen extinction learning is essential. In the current study, we evaluated the effects of a novel context (i.e., novelty) exposure on aversive memory extinction enhancement over days and the dopaminergic system requirement. Given the purpose, experiments were conducted using 3-month-old male Wistar rats. Animals were trained in inhibitory avoidance (IA). Twenty-four hours later, rats were submitted to a weak extinction protocol. Still, 30 min before the first extinction session, animals were submitted to an exploration of a novel context for 5 min. After, memory retention and persistence were evaluated 24 h, 3, 7, 14, and 21 days later. The exposition of a novel context caused a decrease in aversive responses in all days analyzed and an increase in dopamine levels in the hippocampus. The intrahippocampal infusion of dopamine in the CA1 area or the stimulation of the ventral tegmental area (VTA) by a glutamatergic agonist (NMDA) showed similar effects of novelty. In contrast, VTA inhibition by a gabaergic agonist (muscimol) impaired the persistence of extinction learning induced by novelty exposition and caused a decrease in hippocampal dopamine levels. In summary, we show that novel context exposure promotes persistent aversive memory extinction, revealing the significant role of the dopaminergic system.
Collapse
Affiliation(s)
- Karine Ramires Lima
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Niege Alves
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Luiza Freitas Lopes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Steffanie Severo Picua
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Liane da Silva de Vargas
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | | | - Bruna Ramborger
- Interdisciplinary Group of Research in Teaching Practice, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Rafael Roehrs
- Interdisciplinary Group of Research in Teaching Practice, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Marcelo Gomes de Gomes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil.
| |
Collapse
|
31
|
Nachtigall EG, de Freitas JDR, Marcondes LA, Furini CRG. Memory persistence induced by environmental enrichment is dependent on different brain structures. Physiol Behav 2023; 272:114375. [PMID: 37806510 DOI: 10.1016/j.physbeh.2023.114375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/24/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Environmental enrichment (EE) has been demonstrated to have a beneficial effect on different functions of the central nervous system in several mammal species, being used to improve behavior and cell damage in various neurological and psychiatric diseases. However, little has been investigated on the effect of EE in healthy animals, particularly regarding its impact on memory persistence and the brain structures involved. Therefore, here we verified in male Wistar rats that contextual fear conditioning (CFC) memory persistence, tested 28 days after the CFC training session, was facilitated by 5 weeks of exposure to EE, with no effect in groups tested 7 or 14 days after CFC training. However, a two-week exposure to EE did not affect memory persistence. Moreover, we investigated the role of specific brain regions in mediating the effect of EE on memory persistence. We conducted inactivation experiments using the GABAergic agonist Muscimol to target the basolateral amygdala (BLA), medial prefrontal cortex (mPFC), and CA1 region of the hippocampus (CA1). Inactivation of the BLA immediately and 12 h after CFC training impaired the effect of EE on memory persistence. Similarly, inactivation of the CA1 region and mPFC 12 h after training, but not immediately, also impaired the effect of EE on memory persistence. These results have important scientific implications as they shed new light on the effect of an enriched environment on memory persistence and the brain structures involved, thereby helping elucidate how an environment rich in experiences can modify the persistence of learned information.
Collapse
Affiliation(s)
- Eduarda G Nachtigall
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, 90610-000, Porto Alegre, RS, Brazil
| | - Júlia D R de Freitas
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, 90610-000, Porto Alegre, RS, Brazil
| | - Lucas Aschidamini Marcondes
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, 90610-000, Porto Alegre, RS, Brazil
| | - Cristiane R G Furini
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, 90610-000, Porto Alegre, RS, Brazil.
| |
Collapse
|
32
|
Lima KR, Neves BHSD, Carrazoni GS, Rosa ACDSD, Carriço MRS, Roehrs R, Mello-Carpes PB. Acute physical exercise improves recognition memory via locus coeruleus activation but not via ventral tegmental area activation. Physiol Behav 2023; 272:114370. [PMID: 37797663 DOI: 10.1016/j.physbeh.2023.114370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
Both animals and humans have been studied to explore the impact of acute physical exercise (PE) on memory. In rats, a single session of PE enhances the persistence of novel object recognition (NOR) memory, which depends on dopamine and noradrenaline activity in the hippocampus. However, limited research has examined the involvement of other brain regions in this phenomenon. In this study, we investigated the role of the ventral tegmental area (VTA) and locus coeruleus (LC) in modulating the persistence of NOR memory induced by acute PE. After NOR training, some animals underwent a 30 min treadmill PE session, followed by infusion of either vehicle (VEH) or muscimol (MUS) in either the VTA or LC. Other animals did not undergo PE and only received VEH, MUS, or NMDA within the same time window. We evaluated memory recall 1, 7, and 14 days later. Acute PE promoted memory persistence for up to 14 days afterward, similar to NMDA glutamatergic stimulation of the VTA or LC. Moreover, only the LC region was required for the memory improvement induced by acute PE since blocking this region with MUS impaired NOR encoding. Our findings suggest that acute PE can improve learning within a closed time window, and this effect depends on LC, but not VTA, activity.
Collapse
Affiliation(s)
- Karine Ramires Lima
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Ben-Hur Souto das Neves
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Guilherme Salgado Carrazoni
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil
| | | | - Murilo Ricardo Sigal Carriço
- Laboratory of Environmental Chemical and Toxicological Analysis, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Rafael Roehrs
- Laboratory of Environmental Chemical and Toxicological Analysis, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Federal University of Pampa, Uruguaiana, RS, Brazil.
| |
Collapse
|
33
|
Traina G, Tuszynski JA. The Neurotransmission Basis of Post-Traumatic Stress Disorders by the Fear Conditioning Paradigm. Int J Mol Sci 2023; 24:16327. [PMID: 38003517 PMCID: PMC10671801 DOI: 10.3390/ijms242216327] [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: 10/13/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Fear conditioning constitutes the best and most reproducible paradigm to study the neurobiological mechanisms underlying emotions. On the other hand, studies on the synaptic plasticity phenomena underlying fear conditioning present neural circuits enforcing this learning pattern related to post-traumatic stress disorder (PTSD). Notably, in both humans and the rodent model, fear conditioning and context rely on dependent neurocircuitry in the amygdala and prefrontal cortex, cingulate gyrus, and hippocampus. In this review, an overview of the role that classical neurotransmitters play in the contextual conditioning model of fear, and therefore in PTSD, was reported.
Collapse
Affiliation(s)
- Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, Via Romana, 06126 Perugia, Italy
| | - Jack A. Tuszynski
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy;
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
- Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| |
Collapse
|
34
|
Dalto JF, Medina JH. Time-dependent inhibition of Rac1 in the VTA enhances long-term aversive memory: implications in active forgetting mechanisms. Sci Rep 2023; 13:13507. [PMID: 37598223 PMCID: PMC10439914 DOI: 10.1038/s41598-023-40434-9] [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: 05/19/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
The fate of memories depends mainly on two opposing forces: the mechanisms required for the storage and maintenance of memory and the mechanisms underlying forgetting, being the latter much less understood. Here, we show the effect of inhibiting the small Rho GTPase Rac1 on the fate of inhibitory avoidance memory in male rats. The immediate post-training micro-infusion of the specific Rac1 inhibitor NSC23766 (150 ng/0.5 µl/ side) into the ventral tegmental area (VTA) enhanced long-term memory at 1, 7, and 14 days after a single training. Additionally, an opposed effect occurred when the inhibitor was infused at 12 h after training while no effect was observed immediately after testing animals at 1 day. Control experiments ruled out the possibility that post-training memory enhancement was due to facilitation of memory formation since no effect was found when animals were tested at 1 h after acquisition and no memory enhancement was observed after the formation of a weak memory. Immediate post-training micro-infusion of Rac1 inhibitor into the dorsal hippocampus, or the amygdala did not affect memory. Our findings support the idea of a Rac1-dependent time-specific active forgetting mechanism in the VTA controlling the strength of a long-term aversive memory.
Collapse
Affiliation(s)
- Juliana F Dalto
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo de Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155, 3rd Floor, C1121ABG, Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo de Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155, 3rd Floor, C1121ABG, Buenos Aires, Argentina.
- Instituto Tecnológico de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
35
|
Wang W, Wang Z, Cao J, Dong Y, Chen Y. Roles of Rac1-Dependent Intrinsic Forgetting in Memory-Related Brain Disorders: Demon or Angel. Int J Mol Sci 2023; 24:10736. [PMID: 37445914 DOI: 10.3390/ijms241310736] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Animals are required to handle daily massive amounts of information in an ever-changing environment, and the resulting memories and experiences determine their survival and development, which is critical for adaptive evolution. However, intrinsic forgetting, which actively deletes irrelevant information, is equally important for memory acquisition and consolidation. Recently, it has been shown that Rac1 activity plays a key role in intrinsic forgetting, maintaining the balance of the brain's memory management system in a controlled manner. In addition, dysfunctions of Rac1-dependent intrinsic forgetting may contribute to memory deficits in neurological and neurodegenerative diseases. Here, these new findings will provide insights into the neurobiology of memory and forgetting, pathological mechanisms and potential therapies for brain disorders that alter intrinsic forgetting mechanisms.
Collapse
Affiliation(s)
- Wei Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zixu Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jing Cao
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yulan Dong
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yaoxing Chen
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, Beijing Laboratory of Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| |
Collapse
|
36
|
Pastor V, Dalto JF, Medina JH. α7 nicotinic acetylcholine receptors in the medial prefrontal cortex control rewarding but not aversive memory expression in a dopamine-sensitive manner. Pharmacol Biochem Behav 2023; 227-228:173594. [PMID: 37385456 DOI: 10.1016/j.pbb.2023.173594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Emotional learning involves the association between sensory cues and rewarding or aversive stimuli, and this stored information can be recalled during memory retrieval. In this process, the medial prefrontal cortex (mPFC) plays an essential role. We have previously shown that the antagonism of α7 nicotinic acetylcholine receptors (nAChRs) by methyllycaconitine (MLA) in the mPFC blocked cue-induced cocaine memory retrieval. However, little is known about the involvement of prefrontal α7 nAChRs in the retrieval of aversive memories. Here, by using pharmacology and different behavioral tasks, we found that MLA did not affect aversive memory retrieval, indicating a differential effect of cholinergic prefrontal control of appetitive and aversive memories. Despite being shown that acetylcholine modulates dopamine release in the mPFC, it remains unknown if those modulatory systems act together to control reward-based behavior. We examined that question and found that dopamine type 1 receptor (D1R) activation prevented MLA-induced blockade of cocaine CPP retrieval. Our results suggest that α7 nAChRs and D1R signaling interact in the mPFC to modulate cocaine-associated memory retrieval.
Collapse
Affiliation(s)
- Verónica Pastor
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Buenos Aires, Argentina.
| | - Juliana F Dalto
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), Buenos Aires, Argentina
| | - Jorge H Medina
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), Buenos Aires, Argentina; Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| |
Collapse
|
37
|
Nazari M, Rashidy-Pour A, Ali Vafaei A, Raise-Abdullahi P. Systemic corticosterone administration impairs the late fear memory reconsolidation via basolateral amygdala glucocorticoid receptors: dependence on the time window and memory age. Neurobiol Learn Mem 2023:107797. [PMID: 37385522 DOI: 10.1016/j.nlm.2023.107797] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Glucocorticoid receptors (GRs) of the basolateral amygdala (BLA) play an important role in memory reconsolidation. The present study investigated the role of the BLA GRs in the late reconsolidation of fear memory using an inhibitory avoidance (IA) task in male Wistar rats. Stainless steel cannulae were implanted bilaterally into the BLA of the rats. After 7 days of recovery, the animals were trained in a one-trial IA task (1mA, 3s). In Experiment one, 48h after the training session, the animals received 3 systemic doses of corticosterone (CORT; 1, 3, or 10 mg/kg, i.p.) followed by an intra-BLA microinjection of the vehicle (0.3µl/side) at different time points (immediately, 12, or 24h) after memory reactivation. Memory reactivation was performed by returning the animals to the light compartment while the sliding door was open. No shock was delivered during memory reactivation. CORT (10 mg/kg) injection 12h after memory reactivation most effectively impaired the late memory reconsolidation (LMR). In the second part of Experiment one, immediately, 12, or 24h after memory reactivation, GR antagonist RU38486 (RU; 1ng/0.3µl/side) was injected into BLA following a systemic injection of CORT (10 mg/kg) to examine whether it would block the CORT effect. RU inhibited the impairing effects of CORT on LMR. In Experiment two, the animals received CORT (10 mg/kg) with time windows immediately, 3, 6, 12, and 24h after memory reactivation. Again, CORT (10 mg/kg) injection 12h after MR impaired LMR. Memory reactivation was performed in the third Experiment, 7, 14, 28, or 56 days after the training session. Injection of CORT (10 mg/kg) 12h later had no significant effect on the LMR. The impairing effect of CORT was seen only in 2-day-old but not 7, 14, 28, and 56-day-old memories. GRs located in BLA seem to play an important role in the LMR of young memory, as with increasing the age of memories, they become less sensitive to manipulation.
Collapse
Affiliation(s)
- Maryam Nazari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Ali Vafaei
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | |
Collapse
|
38
|
Luckey AM, McLeod LS, Huang Y, Mohan A, Vanneste S. Making memories last using the peripheral effect of direct current stimulation. eLife 2023; 12:e75586. [PMID: 37204308 PMCID: PMC10241520 DOI: 10.7554/elife.75586] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/18/2023] [Indexed: 05/20/2023] Open
Abstract
Most memories that are formed are forgotten, while others are retained longer and are subject to memory stabilization. We show that non-invasive transcutaneous electrical stimulation of the greater occipital nerve (NITESGON) using direct current during learning elicited a long-term memory effect. However, it did not trigger an immediate effect on learning. A neurobiological model of long-term memory proposes a mechanism by which memories that are initially unstable can be strengthened through subsequent novel experiences. In a series of studies, we demonstrate NITESGON's capability to boost the retention of memories when applied shortly before, during, or shortly after the time of learning by enhancing memory consolidation via activation and communication in and between the locus coeruleus pathway and hippocampus by plausibly modulating dopaminergic input. These findings may have a significant impact for neurocognitive disorders that inhibit memory consolidation such as Alzheimer's disease.
Collapse
Affiliation(s)
- Alison M Luckey
- Global Brain Health Institute and Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Lauren S McLeod
- School of Medicine, Texas Tech School of MedicineLubbockUnited States
| | - Yuefeng Huang
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Anusha Mohan
- Global Brain Health Institute and Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Sven Vanneste
- Global Brain Health Institute and Institute of Neuroscience, Trinity College DublinDublinIreland
| |
Collapse
|
39
|
Isotalus HK, Carr WJ, Blackman J, Averill GG, Radtke O, Selwood J, Williams R, Ford E, McCullagh L, McErlane J, O’Donnell C, Durant C, Bartsch U, Jones MW, Muñoz-Neira C, Wearn AR, Grogan JP, Coulthard EJ. L-DOPA increases slow-wave sleep duration and selectively modulates memory persistence in older adults. Front Behav Neurosci 2023; 17:1096720. [PMID: 37091594 PMCID: PMC10113484 DOI: 10.3389/fnbeh.2023.1096720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction Millions of people worldwide take medications such as L-DOPA that increase dopamine to treat Parkinson's disease. Yet, we do not fully understand how L-DOPA affects sleep and memory. Our earlier research in Parkinson's disease revealed that the timing of L-DOPA relative to sleep affects dopamine's impact on long-term memory. Dopamine projections between the midbrain and hippocampus potentially support memory processes during slow wave sleep. In this study, we aimed to test the hypothesis that L-DOPA enhances memory consolidation by modulating NREM sleep. Methods We conducted a double-blind, randomised, placebo-controlled crossover trial with healthy older adults (65-79 years, n = 35). Participants first learned a word list and were then administered long-acting L-DOPA (or placebo) before a full night of sleep. Before sleeping, a proportion of the words were re-exposed using a recognition test to strengthen memory. L-DOPA was active during sleep and the practice-recognition test, but not during initial learning. Results The single dose of L-DOPA increased total slow-wave sleep duration by approximately 11% compared to placebo, while also increasing spindle amplitudes around slow oscillation peaks and around 1-4 Hz NREM spectral power. However, behaviourally, L-DOPA worsened memory of words presented only once compared to re-exposed words. The coupling of spindles to slow oscillation peaks correlated with these differential effects on weaker and stronger memories. To gauge whether L-DOPA affects encoding or retrieval of information in addition to consolidation, we conducted a second experiment targeting L-DOPA only to initial encoding or retrieval and found no behavioural effects. Discussion Our results demonstrate that L-DOPA augments slow wave sleep in elderly, perhaps tuning coordinated network activity and impacting the selection of information for long-term storage. The pharmaceutical modification of slow-wave sleep and long-term memory may have clinical implications. Clinical trial registration Eudract number: 2015-002027-26; https://doi.org/10.1186/ISRCTN90897064, ISRCTN90897064.
Collapse
Affiliation(s)
- Hanna K. Isotalus
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Digital Health, Faculty of Engineering, University of Bristol, Bristol, United Kingdom
| | - Will J. Carr
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jonathan Blackman
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
| | - George G. Averill
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Oliver Radtke
- Department of Neurosurgery, Heinrich-Heine-University Clinic, Düsseldorf, Germany
| | - James Selwood
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
| | - Rachel Williams
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elizabeth Ford
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Liz McCullagh
- Production Pharmacy, Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Trust, Bristol, United Kingdom
| | - James McErlane
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Cian O’Donnell
- School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| | - Claire Durant
- Experimental Psychology, University of Bristol, Bristol, United Kingdom
| | - Ullrich Bartsch
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Matt W. Jones
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Carlos Muñoz-Neira
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Alfie R. Wearn
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - John P. Grogan
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Elizabeth J. Coulthard
- Clinical Neurosciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
| |
Collapse
|
40
|
Possemato E, La Barbera L, Nobili A, Krashia P, D'Amelio M. The role of dopamine in NLRP3 inflammasome inhibition: Implications for neurodegenerative diseases. Ageing Res Rev 2023; 87:101907. [PMID: 36893920 DOI: 10.1016/j.arr.2023.101907] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/10/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
In the Central Nervous System (CNS), neuroinflammation orchestrated by microglia and astrocytes is an innate immune response to counteract stressful and dangerous insults. One of the most important and best characterized players in the neuroinflammatory response is the NLRP3 inflammasome, a multiproteic complex composed by NOD-like receptor family Pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC) and pro-caspase-1. Different stimuli mediate NLRP3 activation, resulting in the NLRP3 inflammasome assembly and the pro-inflammatory cytokine (IL-1β and IL-18) maturation and secretion. The persistent and uncontrolled NLRP3 inflammasome activation has a leading role during the pathophysiology of neuroinflammation in age-related neurodegenerative diseases such as Parkinson's (PD) and Alzheimer's (AD). The neurotransmitter dopamine (DA) is one of the players that negatively modulate NLRP3 inflammasome activation through DA receptors expressed in both microglia and astrocytes. This review summarizes recent findings linking the role of DA in the modulation of NLRP3-mediated neuroinflammation in PD and AD, where early deficits of the dopaminergic system are well characterized. Highlighting the relationship between DA, its glial receptors and the NLRP3-mediated neuroinflammation can provide insights to novel diagnostic strategies in early disease phases and new pharmacological tools to delay the progression of these diseases.
Collapse
Affiliation(s)
- Elena Possemato
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Livia La Barbera
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Annalisa Nobili
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Paraskevi Krashia
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy; Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy.
| |
Collapse
|
41
|
Yang J, Zhai X, Dong X, Zhao L, Zhang Y, Xiao H, Ju P, Duan J, Tang X, Hou B. Peroxidase-like phosphate hydrate nanosheets bio-synthesized by a marine Shewanella algae strain for highly sensitive dopamine detection. Colloids Surf B Biointerfaces 2023; 225:113248. [PMID: 36905834 DOI: 10.1016/j.colsurfb.2023.113248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023]
Abstract
The sensitive and rapid detection of dopamine (DA) is of great significance for early diagnosis of related diseases. Current detection strategies of DA are time-consuming, expensive and inaccurate, while biosynthetic nanomaterials were considered highly stable and environment friendly, which were promising on colorimetric sensing. Thus, in this study, novel zinc phosphate hydrate nanosheets (SA@ZnPNS) biosynthesized by Shewanella algae were designed for the detection of DA. SA@ZnPNS showed high peroxidase-like activity which catalyzed the oxidation reaction of 3,3',5,5'-tetramethylbenzidine in the presence of H2O2. Results showed that the catalytic reaction of SA@ZnPNS followed Michaelis-Menton kinetics, and catalytic process conformed to ping-pong mechanism with chief active species of hydroxyl radicals. The colorimetric detection of DA in human serum samples was performed based on SA@ZnPNS peroxidase-like activity. The linear range of DA detection was 0.1-40 μM, and the detection limit was 0.083 μM. This study provided a simple and practical method for the detection of DA and expanded the application of biosynthesized nanoparticles to biosensing fields.
Collapse
Affiliation(s)
- Jing Yang
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, PR China; CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China
| | - Xiaofan Zhai
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China; Sanya Institute of Ocean Eco-Environmental Engineering, Zhenzhou Road, Sanya 572000, PR China; Institute of Marine Corrosion Protection, Guangxi Academy of Sciences, No. 98 Daling Road, Nanning 530007, PR China.
| | - Xucheng Dong
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China
| | - Liuhui Zhao
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China; School of Bioengineering, Qilu University of Technology, No. 3501 Daxue Road, Jinan 250353, PR China
| | - Yu Zhang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China
| | - Hui Xiao
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, PR China.
| | - Peng Ju
- Key Laboratory of Marine Eco-Environmental Science and Technology, Marine Bioresource and Environment Research Center, First Institute of Oceanography, Ministry of Natural Resources, No. 6 Xianxialing Road, Qingdao 266061, PR China
| | - Jizhou Duan
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China; Sanya Institute of Ocean Eco-Environmental Engineering, Zhenzhou Road, Sanya 572000, PR China; Institute of Marine Corrosion Protection, Guangxi Academy of Sciences, No. 98 Daling Road, Nanning 530007, PR China
| | - Xuexi Tang
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, PR China
| | - Baorong Hou
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, PR China; Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266235, PR China; Sanya Institute of Ocean Eco-Environmental Engineering, Zhenzhou Road, Sanya 572000, PR China; Institute of Marine Corrosion Protection, Guangxi Academy of Sciences, No. 98 Daling Road, Nanning 530007, PR China
| |
Collapse
|
42
|
Wang Y, Liu J, Hui Y, Wu Z, Wang L, Wu X, Bai Y, Zhang Q, Li L. Dose and time-dependence of acute intermittent theta-burst stimulation on hippocampus-dependent memory in parkinsonian rats. Front Neurosci 2023; 17:1124819. [PMID: 36866328 PMCID: PMC9972116 DOI: 10.3389/fnins.2023.1124819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
Background The treatment options for cognitive impairments in Parkinson's disease (PD) are limited. Repetitive transcranial magnetic stimulation has been applied in various neurological diseases. However, the effect of intermittent theta-burst stimulation (iTBS) as a more developed repetitive transcranial magnetic stimulation paradigm on cognitive dysfunction in PD remains largely unclear. Objective Our aim was to explore the effect of acute iTBS on hippocampus-dependent memory in PD and the mechanism underlying it. Methods Different blocks of iTBS protocols were applied to unilateral 6-hydroxidopamine-induced parkinsonian rats followed by the behavioral, electrophysiological and immunohistochemical analyses. The object-place recognition and hole-board test were used to assess hippocampus-dependent memory. Results Sham-iTBS and 1 block-iTBS (300 stimuli) didn't alter hippocampus-dependent memory, hippocampal theta rhythm and the density of c-Fos- and parvalbumin-positive neurons in the hippocampus and medial septum. 3 block-iTBS (900 stimuli) alleviated 6-hydroxidopamine-induced memory impairments, and increased the density of hippocampal c-Fos-positive neurons at 80 min post-stimulation but not 30 min compared to sham-iTBS. Interestingly, 3 block-iTBS first decreased and then increased normalized theta power during a period of 2 h following stimulation. Moreover, 3 block-iTBS decreased the density of parvalbumin-positive neurons in the medial septum at 30 min post-stimulation compared to sham-iTBS. Conclusion The results indicate that multiple blocks of iTBS elicit dose and time-dependent effects on hippocampus-dependent memory in PD, which may be attributed to changes in c-Fos expression and the power of theta rhythm in the hippocampus.
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yanping Hui
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhongheng Wu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ling Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiang Wu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yihua Bai
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiaojun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Libo Li
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
43
|
Tsetsenis T, Broussard JI, Dani JA. Dopaminergic regulation of hippocampal plasticity, learning, and memory. Front Behav Neurosci 2023; 16:1092420. [PMID: 36778837 PMCID: PMC9911454 DOI: 10.3389/fnbeh.2022.1092420] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023] Open
Abstract
The hippocampus is responsible for encoding behavioral episodes into short-term and long-term memory. The circuits that mediate these processes are subject to neuromodulation, which involves regulation of synaptic plasticity and local neuronal excitability. In this review, we present evidence to demonstrate the influence of dopaminergic neuromodulation on hippocampus-dependent memory, and we address the controversy surrounding the source of dopamine innervation. First, we summarize historical and recent retrograde and anterograde anatomical tracing studies of direct dopaminergic projections from the ventral tegmental area and discuss dopamine release from the adrenergic locus coeruleus. Then, we present evidence of dopaminergic modulation of synaptic plasticity in the hippocampus. Plasticity mechanisms are examined in brain slices and in recordings from in vivo neuronal populations in freely moving rodents. Finally, we review pharmacological, genetic, and circuitry research that demonstrates the importance of dopamine release for learning and memory tasks while dissociating anatomically distinct populations of direct dopaminergic inputs.
Collapse
Affiliation(s)
- Theodoros Tsetsenis
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Theodoros Tsetsenis John I. Broussard John A. Dani
| | - John I. Broussard
- Department of Neurobiology and Anatomy, UT Health Houston McGovern Medical School, Houston, TX, United States,*Correspondence: Theodoros Tsetsenis John I. Broussard John A. Dani
| | - John A. Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Theodoros Tsetsenis John I. Broussard John A. Dani
| |
Collapse
|
44
|
Hagena H, Stacho M, Laja A, Manahan-Vaughan D. Strain-dependent regulation of hippocampal long-term potentiation by dopamine D1/D5 receptors in mice. Front Behav Neurosci 2022; 16:1023361. [PMID: 36545120 PMCID: PMC9760685 DOI: 10.3389/fnbeh.2022.1023361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/26/2022] [Indexed: 12/12/2022] Open
Abstract
The magnitude and persistency of long-term potentiation (LTP) in the rodent hippocampus is species-dependent: rats express more robust and more prolonged LTP in response to a broader afferent frequency range than mice. The C57Bl/6 mouse is an extremely popular murine strain used in studies of hippocampal synaptic plasticity and spatial learning. Recently it was reported that it expresses impoverished LTP compared to other murine strains. Given the important role of the dopamine D1/D5 receptor (D1/D5R) in the maintenance of LTP and in memory consolidation, we explored to what extent strain-dependent differences in LTP in mice are determined by differences in D1/D5R-control. In CaOlaHsd mice, robust LTP was induced that lasted for over 24 h and which was significantly greater in magnitude than LTP induced in C57Bl/6 mice. Intracerebral treatment with a D1/D5R-antagonist (SCH23390) prevented both the early and late phase of LTP in CaOlaHsd mice, whereas only late-LTP was impaired in C57Bl/6 mice. Treatment with a D1/D5R-agonist (Chloro-PB) facilitated short-term potentiation (STP) into LTP (> 24 h) in both strains, whereby effects became evident earlier in CaOlaHsd compared to C57Bl/6 mice. Immunohistochemical analysis revealed a significantly higher expression of D1-receptors in the stratum lacunosum moleculare of CaOlaHsd compared to C57Bl/6 mice. These findings highlight differences in D1/D5R- dependent regulation of strain-dependent variations in hippocampal LTP in C57Bl/6 and CaOlaHsd mice, that may be mediated, in part, by differences in the expression of D1R in the hippocampus.
Collapse
|
45
|
Chowdhury A, Luchetti A, Fernandes G, Filho DA, Kastellakis G, Tzilivaki A, Ramirez EM, Tran MY, Poirazi P, Silva AJ. A locus coeruleus-dorsal CA1 dopaminergic circuit modulates memory linking. Neuron 2022; 110:3374-3388.e8. [PMID: 36041433 PMCID: PMC10508214 DOI: 10.1016/j.neuron.2022.08.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 07/31/2022] [Indexed: 11/20/2022]
Abstract
Individual memories are often linked so that the recall of one triggers the recall of another. For example, contextual memories acquired close in time can be linked, and this is known to depend on a temporary increase in excitability that drives the overlap between dorsal CA1 (dCA1) hippocampal ensembles that encode the linked memories. Here, we show that locus coeruleus (LC) cells projecting to dCA1 have a key permissive role in contextual memory linking, without affecting contextual memory formation, and that this effect is mediated by dopamine. Additionally, we found that LC-to-dCA1-projecting neurons modulate the excitability of dCA1 neurons and the extent of overlap between dCA1 memory ensembles as well as the stability of coactivity patterns within these ensembles. This discovery of a neuromodulatory system that specifically affects memory linking without affecting memory formation reveals a fundamental separation between the brain mechanisms modulating these two distinct processes.
Collapse
Affiliation(s)
- Ananya Chowdhury
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
| | - Alessandro Luchetti
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
| | - Giselle Fernandes
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
| | - Daniel Almeida Filho
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
| | - George Kastellakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas (FORTH), Vassilica Vouton, PO Box 1527, GR 711 10 Heraklion, Crete, Greece
| | - Alexandra Tzilivaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas (FORTH), Vassilica Vouton, PO Box 1527, GR 711 10 Heraklion, Crete, Greece
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Charitéplatz 1, 10117 Berlin Germany
- Einstein Center for Neurosciences Berlin Charitéplatz 1, 10117 Berlin Germany
- Neurocure Cluster of Excellence Charitéplatz 1, 10117 Berlin, Germany
| | - Erica M Ramirez
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
| | - Mary Y Tran
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas (FORTH), Vassilica Vouton, PO Box 1527, GR 711 10 Heraklion, Crete, Greece
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA 90095
- Lead contact
| |
Collapse
|
46
|
Castillo Díaz F, Dalto JF, Pereyra M, Medina JH. Dopamine neurotransmission in the VTA regulates aversive memory formation and persistence. Physiol Behav 2022; 253:113854. [PMID: 35609721 DOI: 10.1016/j.physbeh.2022.113854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022]
Abstract
Dopamine (DA) neurons in the ventral tegmental area (VTA) innervating several limbic and neocortical regions of the mammalian brain have long been implicated in motivation, rewarding and aversive behaviors, and memory processing. Recently, we demonstrated that somatodendritic release of DA in the VTA regulates the formation and maintenance of appetitive long-term memories (LTM). However, less is known about the impact of DA neurotransmission in the VTA on aversive LTM. Here, we describe the modulation of negative-valence memories by D1/D5-type DA (D1R)-receptor-mediated neurotransmission in the VTA. As aversive stimuli elicit both active and passive behavioral responses, we used two single-trial aversive training protocols: inhibitory avoidance task and conditioned place aversion. We bilaterally microinfused SCH23390, an antagonist of D1R, into the VTA immediately after training and found that DA neurotransmission in the VTA modulates LTM consolidation and persistence of aversive experiences. Together with previous findings demonstrating that D1R-mediated DA neurotransmission in the medial prefrontal cortex and hippocampus is involved in the formation and persistence of LTM for aversive events, our present results indicate that memory processing of environmental stimuli with negative-valence depends on the integration of information mediated by D1R activation in both the VTA region and in selected downstream target areas.
Collapse
Affiliation(s)
- Fernando Castillo Díaz
- Facultad de Medicina, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina; Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Via Balzaretti 9, Milano 20133, Italy.
| | - Juliana F Dalto
- Facultad de Medicina, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina
| | - Magdalena Pereyra
- Facultad de Medicina, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina
| | - Jorge H Medina
- Facultad de Medicina, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina; Instituto Tecnológico Buenos Aires, Iguazú 341, CABA C1437, Argentina
| |
Collapse
|
47
|
A Novel and Selective Dopamine Transporter Inhibitor, (S)-MK-26, Promotes Hippocampal Synaptic Plasticity and Restores Effort-Related Motivational Dysfunctions. Biomolecules 2022; 12:biom12070881. [PMID: 35883437 PMCID: PMC9312958 DOI: 10.3390/biom12070881] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
Dopamine (DA), the most abundant human brain catecholaminergic neurotransmitter, modulates key behavioral and neurological processes in young and senescent brains, including motricity, sleep, attention, emotion, learning and memory, and social and reward-seeking behaviors. The DA transporter (DAT) regulates transsynaptic DA levels, influencing all these processes. Compounds targeting DAT (e.g., cocaine and amphetamines) were historically used to shape mood and cognition, but these substances typically lead to severe negative side effects (tolerance, abuse, addiction, and dependence). DA/DAT signaling dysfunctions are associated with neuropsychiatric and progressive brain disorders, including Parkinson’s and Alzheimer diseases, drug addiction and dementia, resulting in devastating personal and familial concerns and high socioeconomic costs worldwide. The development of low-side-effect, new/selective medicaments with reduced abuse-liability and which ameliorate DA/DAT-related dysfunctions is therefore crucial in the fields of medicine and healthcare. Using the rat as experimental animal model, the present work describes the synthesis and pharmacological profile of (S)-MK-26, a new modafinil analogue with markedly improved potency and selectivity for DAT over parent drug. Ex vivo electrophysiology revealed significantly augmented hippocampal long-term synaptic potentiation upon acute, intraperitoneally delivered (S)-MK-26 treatment, whereas in vivo experiments in the hole-board test showed only lesser effects on reference memory performance in aged rats. However, in effort-related FR5/chow and PROG/chow feeding choice experiments, (S)-MK-26 treatment reversed the depression-like behavior induced by the dopamine-depleting drug tetrabenazine (TBZ) and increased the selection of high-effort alternatives. Moreover, in in vivo microdialysis experiments, (S)-MK-26 significantly increased extracellular DA levels in the prefrontal cortex and in nucleus accumbens core and shell. These studies highlight (S)-MK-26 as a potent enhancer of transsynaptic DA and promoter of synaptic plasticity, with predominant beneficial effects on effort-related behaviors, thus proposing therapeutic potentials for (S)-MK-26 in the treatment of low-effort exertion and motivational dysfunctions characteristic of depression and aging-related disorders.
Collapse
|
48
|
Tancheva L, Lazarova M, Velkova L, Dolashki A, Uzunova D, Minchev B, Petkova-Kirova P, Hassanova Y, Gavrilova P, Tasheva K, Taseva T, Hodzhev Y, Atanasov AG, Stefanova M, Alexandrova A, Tzvetanova E, Atanasov V, Kalfin R, Dolashka P. Beneficial Effects of Snail Helix aspersa Extract in an Experimental Model of Alzheimer’s Type Dementia. J Alzheimers Dis 2022; 88:155-175. [DOI: 10.3233/jad-215693] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: Alzheimer’s disease (AD) is a complex neurodegenerative disease with multifactorial etiology, unsatisfactory treatment, and a necessity for broad-spectrum active substances for cure. The mucus from Helix aspersa snail is a mixture of bioactive molecules with antimicrobial, anti-inflammatory, antioxidant, and anti-apoptotic effects. So far there are no data concerning the capacity of snail extract (SE) to affect neurodegenerative disorders. Objective: The effects of SE from Helix aspersa on learning and memory deficits in Alzheimer’s type dementia (ATD) induced by scopolamine (Sco) in male Wistar rats were examined and some mechanisms of action underlying these effects were evaluated. Methods: SE (0.5 mL/100 g) was applied orally through a food tube for 16 consecutive days: 5 days before and 11 days simultaneously with Sco (2 mg/kg, intraperitoneally). At the end of Sco treatment, using behavioral methods, we evaluated memory performance. Additionally, in cortex and hippocampus the acetylcholinesterase (AChE) activity, acetylcholine and monoamines (dopamine, noradrenaline, and serotonin) content, levels of main oxidative stress markers, and expression of brain-derived neurotrophic factor (BDNF) and cAMP response element-binding protein (CREB) were determined. Results: We demonstrated that, according to all behavioral tests used, SE significantly improved the cognitive deficits induced by Sco. Furthermore, SE possessed AChE inhibitory activity, moderate antioxidant properties and the ability to modulate monoamines content in two brain structures. Moreover, multiple SE applications not only restored the depressed by Sco expression of CREB and BDNF, but significantly upregulated it. Conclusion: Summarizing results, we conclude that complex mechanisms underlie the beneficial effects of SE on impaired memory in Alzheimer’s type dementia.
Collapse
Affiliation(s)
- Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
- Weston Professor of Weizmann Institute of Science, Israel
| | - Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Lyudmila Velkova
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Alexander Dolashki
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | | | - Yozljam Hassanova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Petja Gavrilova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Teodora Taseva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yordan Hodzhev
- National Center for Infectious and Parasitic Diseases, Sofia, Bulgaria
| | - Atanas G. Atanasov
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Magdalenka, Poland
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | | | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Elina Tzvetanova
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Ventseslav Atanasov
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Science, Sofia, Bulgaria
- Department of Healthcare, South-West University “Neofit Rilski”, Blagoevgrad, Bulgaria
| | - Pavlina Dolashka
- Institute of Organic Chemistry with Center for Phytochemistry, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
49
|
Guzmán-Ramos K, Osorio-Gómez D, Bermúdez-Rattoni F. Cognitive impairment in alzheimer’s and metabolic diseases: A catecholaminergic hypothesis. Neuroscience 2022; 497:308-323. [DOI: 10.1016/j.neuroscience.2022.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022]
|
50
|
Zhu H, Chu Z, Wang Y, Chen J, Zhang Z, Wu X. Strong Out-of-Plane Vibrations and Ultrasensitive Detection of Dopamine-like Neurotransmitters. J Phys Chem Lett 2022; 13:3325-3331. [PMID: 35394786 DOI: 10.1021/acs.jpclett.2c00737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The detection of monoamine neurotransmitters has become a vital research subject due to their high correlations with nervous system diseases, but insufficient detection precisions have obstructed diagnosis of some related diseases. Here, we focus on four monoamine neurotransmitters, dopamine, norepinephrine, epinephrine, and serotonin, to conduct their rapid and ultrasensitive detection. We find that the low-frequency (<200 cm-1) Raman vibrations of these molecules show some sharp peaks, and their intensities are significantly stronger than those of the high-frequency side. Theoretical calculations identify these peaks to be from strong out-of-plane vibrations of the C-C single bonds at the joint point of the ring-like molecule and its side chain. Using our surface enhanced low-frequency Raman scattering substrates, we show that the detection limit of dopamine as an example can reach 10 nM in artificial cerebrospinal fluid. This work provides a useful way for ultrasensitive and rapid detection of some neurotransmitters.
Collapse
Affiliation(s)
- Haogang Zhu
- National Laboratory of Solid States Microstructures and Key Laboratory of Modern Acoustics, MOE, Institute of Acoustics, Nanjing University, Nanjing 210093, China
| | - Zhiqiang Chu
- National Laboratory of Solid States Microstructures and Key Laboratory of Modern Acoustics, MOE, Institute of Acoustics, Nanjing University, Nanjing 210093, China
| | - Yixian Wang
- National Laboratory of Solid States Microstructures and Key Laboratory of Modern Acoustics, MOE, Institute of Acoustics, Nanjing University, Nanjing 210093, China
| | - Jian Chen
- National Laboratory of Solid States Microstructures and Research Institute of Superconductor Electronics, Nanjing University, Nanjing 210093, China
| | - Zhiyong Zhang
- National Laboratory of Solid States Microstructures and Key Laboratory of Modern Acoustics, MOE, Institute of Acoustics, Nanjing University, Nanjing 210093, China
| | - Xinglong Wu
- National Laboratory of Solid States Microstructures and Key Laboratory of Modern Acoustics, MOE, Institute of Acoustics, Nanjing University, Nanjing 210093, China
| |
Collapse
|