1
|
Martial C, Fritz P, Gosseries O, Bonhomme V, Kondziella D, Nelson K, Lejeune N. A neuroscientific model of near-death experiences. Nat Rev Neurol 2025:10.1038/s41582-025-01072-z. [PMID: 40159547 DOI: 10.1038/s41582-025-01072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 04/02/2025]
Abstract
Near-death experiences (NDEs) are episodes of disconnected consciousness that typically occur in situations that involve an actual or potential physical threat or are perceived as such, and the experiences are characterized by a rich content with prototypical mystical features. Several explanatory theories for NDEs have been proposed, ranging from psychological or neurophysiological to evolutionary models. However, these concepts were often formulated independently, and, owing to the fragmented nature of research in this domain, integration of these ideas has been limited. Lines of empirical evidence from different areas of neuroscience, including non-human studies, studies investigating psychedelic-induced mystical experiences in humans, and research on the dying brain, are now converging to provide a comprehensive explanation for NDEs. In this Review, we discuss processes that might underlie the rich conscious experience in NDEs, mostly focusing on prototypical examples and addressing both the potential psychological mechanisms and neurophysiological changes, including cellular and electrophysiological brain network modifications and alterations in neurotransmitter release. On the basis of this discussion, we propose a model for NDEs that encompasses a cascade of concomitant psychological and neurophysiological processes within an evolutionary framework. We also consider how NDE research can inform the debate on the emergence of consciousness in near-death conditions that arise before brain death.
Collapse
Affiliation(s)
- Charlotte Martial
- Coma Science Group, GIGA-Consciousness, GIGA Institute, University of Liège, Liège, Belgium.
- NeuroRehab & Consciousness Clinic, Neurology Department, University Hospital of Liège, Liège, Belgium.
| | - Pauline Fritz
- Coma Science Group, GIGA-Consciousness, GIGA Institute, University of Liège, Liège, Belgium
- NeuroRehab & Consciousness Clinic, Neurology Department, University Hospital of Liège, Liège, Belgium
| | - Olivia Gosseries
- Coma Science Group, GIGA-Consciousness, GIGA Institute, University of Liège, Liège, Belgium
- NeuroRehab & Consciousness Clinic, Neurology Department, University Hospital of Liège, Liège, Belgium
| | - Vincent Bonhomme
- Anaesthesia and Perioperative Neuroscience Laboratory, GIGA-Consciousness, GIGA Institute, University of Liège, Liège, Belgium
- Department of Anaesthesia and Intensive Care Medicine, University Hospital of Liège, Liège, Belgium
| | - Daniel Kondziella
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kevin Nelson
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Nicolas Lejeune
- Coma Science Group, GIGA-Consciousness, GIGA Institute, University of Liège, Liège, Belgium
- NeuroRehab & Consciousness Clinic, Neurology Department, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
2
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
3
|
Schwabe L. Memory Under Stress: From Adaptation to Disorder. Biol Psychiatry 2025; 97:339-348. [PMID: 38880463 DOI: 10.1016/j.biopsych.2024.06.005] [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: 02/16/2024] [Revised: 05/02/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Stressful events are ubiquitous in everyday life. Exposure to these stressors initiates the temporally orchestrated release of a multitude of hormones, peptides, and neurotransmitters that target brain areas that have been critically implicated in learning and memory. This review summarizes recent insights on the profound impact of stress on 4 fundamental processes of memory: memory formation, memory contextualization, memory retrieval, and memory flexibility. Stress mediators instigate dynamic alterations in these processes, thereby facilitating efficient responding under stress and the creation of a decontextualized memory representation that can effectively aid coping with novel future threats. While they are generally adaptive, the same stress-related changes may contribute to the rigid behaviors, uncontrollable intrusions, and generalized fear responding seen in anxiety disorders and posttraumatic stress disorder. Drawing on recent discoveries in cognitive neuroscience and psychiatry, this review discusses how stress-induced alterations in memory processes can simultaneously foster adaptation to stressors and fuel psychopathology. The transition from adaptive to maladaptive changes in the impact of stress on memory hinges on the nuanced interplay of stressor characteristics and individual predispositions. Thus, taking individual differences in the cognitive response to stressors into account is essential for any successful treatment of stress-related mental disorders.
Collapse
Affiliation(s)
- Lars Schwabe
- Department of Cognitive Psychology, Institute of Psychology, Universität Hamburg, Hamburg, Germany.
| |
Collapse
|
4
|
Trompoukis G, Miliou A, Papatheodoropoulos C. β-adrenergic receptor-induced E-S potentiation in the dorsal and ventral hippocampus. Front Synaptic Neurosci 2024; 16:1511485. [PMID: 39758826 PMCID: PMC11695307 DOI: 10.3389/fnsyn.2024.1511485] [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: 10/15/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
β-adrenergic receptors (β-ARs) play a critical role in modulating learning, memory, emotionality, and long-term synaptic plasticity. Recent studies indicate that β-ARs are necessary for long-term potentiation (LTP) induction in the ventral hippocampus under moderate synaptic activation conditions that do not typically induce LTP. To explore potential dorsoventral differences in β-AR-mediated effects, we applied the β-AR agonist isoproterenol (10 μM, 30 min) to dorsal and ventral hippocampal slices, recording field excitatory postsynaptic potentials (fEPSPs) and population spikes (PSs) from the CA1 region. Isoproterenol induced robust, long-lasting PS increases, with effects three times greater in the dorsal compared to the ventral hippocampus. Isoproterenol did not significantly affect fEPSP in either segment of the hippocampus, leading to strong excitatory-to-spike (E-S) potentiation-twice as large as that in the ventral hippocampus. E-S potentiation was not associated with significant paired-pulse inhibition changes in either hippocampal segment. These differences do not appear to result from β1-AR expression levels, as they are comparable across dorsal and ventral hippocampal regions. Overall, the findings suggest that β-AR activation enhances the dorsal hippocampus's role during stress, facilitating heightened alertness, rapid spatial information processing, and effective navigation necessary for "fight-or-flight" responses.
Collapse
|
5
|
Craig GE, Ramos L, Essig SR, Eagles NJ, Jaffe AE, Martinowich K, Hallock HL. Stimulation of Locus Ceruleus Inputs to the Prelimbic Cortex in Mice Induces Cell Type-Specific Expression of the Apoe Gene. eNeuro 2024; 11:ENEURO.0328-24.2024. [PMID: 39632090 PMCID: PMC11675532 DOI: 10.1523/eneuro.0328-24.2024] [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] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
The medial frontal cortex (mFC) and locus ceruleus (LC) are two brain areas that have been implicated in a range of cognitive phenomena, such as attention, memory, and decision-making. Regulators of these brain regions at the molecular level are not well understood but might help to elucidate underlying mechanisms of disorders that present with deficits in these cognitive domains. To probe this, we used chemogenetic stimulation of neurons in the LC with axonal projections to the prelimbic subregion (PrL) of the mFC and subsequent bulk RNA sequencing from the mouse PrL. We found that stimulation of this circuit caused an increase in transcription of a host of genes, including the Apoe gene. To investigate cell type-specific expression of Apoe in the PrL, we used a dual-virus approach to express either the excitatory DREADD receptor hM3Dq in LC neurons with projections to the PrL or a control virus and found that increases in Apoe expression in the PrL following depolarization of LC inputs is enriched in GABAergic neurons in a sex-dependent manner. The results of these experiments yield insights into how Apoe expression affects function in a cortical microcircuit that is important for attention, memory, and decision-making and point to interneuron-specific expression of Apoe as a potential biomarker for circuit function in disorders such as attention-deficit hyperactivity disorder, schizophrenia, and Alzheimer's disease.
Collapse
Affiliation(s)
| | - Lizbeth Ramos
- Neuroscience Program, Lafayette College, Easton, Pennsylvania 18042
| | - Samuel R Essig
- Neuroscience Program, Lafayette College, Easton, Pennsylvania 18042
| | - Nicholas J Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland 21205
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21205
| | - Henry L Hallock
- Neuroscience Program, Lafayette College, Easton, Pennsylvania 18042
| |
Collapse
|
6
|
Iguchi Y, Benton R, Kobayashi K. A chemogenetic technology using insect Ionotropic Receptors to stimulate target cell populations in the mammalian brain. Neurosci Res 2024:S0168-0102(24)00136-6. [PMID: 39532176 DOI: 10.1016/j.neures.2024.11.003] [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: 07/12/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Chemogenetics uses artificially-engineered proteins to modify the activity of cells, notably neurons, in response to small molecules. Although a common set of chemogenetic tools are the G protein-coupled receptor-based DREADDs, there has been great hope for ligand-gated, ion channel-type chemogenetic tools that directly impact neuronal excitability. We have devised such a technology by exploiting insect Ionotropic Receptors (IRs), a highly divergent subfamily of ionotropic glutamate receptors that evolved to detect diverse environmental chemicals. Here, we review a series of studies developing and applying this "IR-mediated neuronal activation" (IRNA) technology with the Drosophila melanogaster IR84a/IR8a complex, which detects phenyl-containing ligands. We also discuss how variants of IRNA could be produced by modifying the composition of the IR complex, using natural or engineered subunits, which would enable artificial activation of different cell populations in the brain in response to distinct chemicals.
Collapse
Affiliation(s)
- Yoshio Iguchi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne CH-1015, Switzerland
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima 960-1295, Japan.
| |
Collapse
|
7
|
Tanada S, Nakagomi T, Nakano-Doi A, Sawano T, Kubo S, Kuramoto Y, Uchida K, Yamahara K, Doe N, Yoshimura S. Human-Brain-Derived Ischemia-Induced Stem Cell Transplantation Is Associated with a Greater Neurological Functional Improvement Compared with Human-Bone Marrow-Derived Mesenchymal Stem Cell Transplantation in Mice After Stroke. Int J Mol Sci 2024; 25:12065. [PMID: 39596134 PMCID: PMC11593343 DOI: 10.3390/ijms252212065] [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: 09/19/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The transplantation of injury/ischemia-induced stem cells (iSCs) extracted from post-stroke human brains can improve the neurological functions of mice after stroke. However, the usefulness of iSCs as an alternative stem cell source remains unclear. The current study aimed to assess the efficacy of iSC and mesenchymal stem cell (MSC) transplantation. In this experiment, equal numbers of human brain-derived iSCs (h-iSCs) (5.0 × 104 cells/μL) and human bone marrow-derived MSCs (h-MSCs) (5.0 × 104 cells/μL) were intracranially transplanted into post-stroke mouse brains after middle cerebral artery occlusion. Results showed that not only h-iSC transplantation but also h-MSC transplantation activated endogenous neural stem/progenitor cells (NSPCs) around the grafted sites and promoted neurological functional improvement. However, mice that received h-iSC transplantation experienced improvement in a higher number of behavioral tasks compared with those that received h-MSC transplantation. To investigate the underlying mechanism, NSPCs extracted from the ischemic areas of post-stroke mouse brains were cocultured with h-iSCs or h-MSCs. After coincubation, NSPCs, h-iSCs, and h-MSCs were selectively collected via fluorescence-activated cell sorting. Next, their traits were analyzed via microarray analysis. The genes related to various neuronal lineages in NSPCs after coincubation with h-iSCs were enriched compared with those in NSPCs after coincubation with h-MSCs. In addition, the gene expression patterns of h-iSCs relative to those of h-MSCs showed that the expression of genes related to synapse formation and neurotransmitter-producing neurons increased more after coincubation with NSPCs. Hence, cell-cell interactions with NSPCs promoted transdifferentiation toward functional neurons predominantly in h-iSCs. In accordance with these findings, immunohistochemistry showed that the number of neuronal networks between NSPCs and h-iSCs was higher than that between NSPCs and h-MSCs. Therefore, compared with h-MSC transplantation, h-iSC transplantation is associated with a higher neurological functional improvement, presumably by more effectively modulating the fates of endogenous NSPCs and grafted h-iSCs themselves.
Collapse
Affiliation(s)
- Shuichi Tanada
- Department of Neurosurgery, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.T.); (Y.K.); (K.U.); (S.Y.)
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (A.N.-D.); (S.K.); (K.Y.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (A.N.-D.); (S.K.); (K.Y.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| | - Toshinori Sawano
- Department of Biomedical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu 525-8577, Japan;
| | - Shuji Kubo
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (A.N.-D.); (S.K.); (K.Y.)
| | - Yoji Kuramoto
- Department of Neurosurgery, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.T.); (Y.K.); (K.U.); (S.Y.)
| | - Kazutaka Uchida
- Department of Neurosurgery, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.T.); (Y.K.); (K.U.); (S.Y.)
| | - Kenichi Yamahara
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (A.N.-D.); (S.K.); (K.Y.)
| | - Nobutaka Doe
- Department of Rehabilitation, Hyogo Medical University (Kobe Campus), 1-3-6 Minatojima, Chuo-ku, Kobe 650-8530, Japan;
| | - Shinichi Yoshimura
- Department of Neurosurgery, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.T.); (Y.K.); (K.U.); (S.Y.)
| |
Collapse
|
8
|
Duarte JM, Nguyen R, Kyprou M, Li K, Milentijevic A, Cerquetella C, Forro T, Ciocchi S. Hippocampal contextualization of social rewards in mice. Nat Commun 2024; 15:9493. [PMID: 39489746 PMCID: PMC11532361 DOI: 10.1038/s41467-024-53866-2] [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: 05/08/2023] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Acquiring and exploiting memories of rewarding experiences is critical for survival. The spatial environment in which a rewarding stimulus is encountered regulates memory retrieval. The ventral hippocampus (vH) has been implicated in contextual memories involving rewarding stimuli such as food, social cues or drugs. Yet, the neuronal representations and circuits underlying contextual memories of socially rewarding stimuli are poorly understood. Here, using in vivo electrophysiological recordings, in vivo one-photon calcium imaging, and optogenetics during a social reward contextual conditioning paradigm in male mice, we show that vH neurons discriminate between contexts with neutral or acquired social reward value. The formation of context-discriminating vH neurons following learning was contingent upon the presence of unconditioned stimuli. Moreover, vH neurons showed distinct contextual representations during the retrieval of social reward compared to fear contextual memories. Finally, optogenetic inhibition of locus coeruleus (LC) projections in the vH selectively disrupted social reward contextual memory by impairing vH contextual representations. Collectively, our findings reveal that the vH integrates contextual and social reward information, with memory encoding of these representations supported by input from the LC.
Collapse
Affiliation(s)
- Joana Mendes Duarte
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Robin Nguyen
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Jerome L. Greene Science Center, Columbia University, New York, NY, USA
| | - Marios Kyprou
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Kaizhen Li
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Anastasija Milentijevic
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Carlo Cerquetella
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Thomas Forro
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Stéphane Ciocchi
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland.
| |
Collapse
|
9
|
Iannitelli AF, Hassenein L, Mulvey B, Blankenship HE, Liles LC, Sharpe AL, Pare JF, Segal A, Sloan SA, Martinowich K, McCann KE, Dougherty JD, Smith Y, Beckstead MJ, Weinshenker D. Tyrosinase-induced neuromelanin accumulation triggers rapid dysregulation and degeneration of the mouse locus coeruleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.07.530845. [PMID: 36945637 PMCID: PMC10028911 DOI: 10.1101/2023.03.07.530845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The locus coeruleus (LC), the major source of norepinephrine (NE) in the brain, is an early site of pathology in both Alzheimer's disease (AD) and Parkinson's disease (PD), and it undergoes catastrophic degeneration later in both disorders. Dysregulation of the LC is thought to contribute to prodromal symptoms of AD and PD such as anxiety and sleep disturbances, while frank LC-NE loss promotes cognitive decline. However, the mechanisms responsible for its selective vulnerability are unknown. The LC is among the only structures in the brain that produces appreciable amounts of neuromelanin (NM), a dark cytoplasmic pigment. It has been proposed that NM initially plays a protective role by sequestering toxic catecholamine metabolites and heavy metals, but may become harmful during aging as it overwhelms cellular machinery and is released during neurodegeneration. Rodents do not naturally produce NM, limiting the study of causal relationships between NM and LC pathology. Adapting a viral-mediated approach for expression of human tyrosinase, the enzyme responsible for peripheral melanin production, we successfully promoted pigmentation in mouse LC neurons that recapitulates key ultrastructural features of endogenous NM found in primates. Pigment expression results in LC neuron hyperactivity, reduced tissue NE levels, transcriptional changes, and novelty-induced anxiety phenotypes as early as 1-week post-injection. By 6-10 weeks, NM accumulation is associated with severe LC neuron neurodegeneration and microglial engulfment of the pigment granules, while the anxiety-like behavior is abated. These phenotypes are reminiscent of LC dysfunction and cell death in AD and PD, validating this model for studying the consequences of pigment accumulation in the LC as it relates to neurodegenerative disease.
Collapse
Affiliation(s)
- Alexa F. Iannitelli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Leslie Hassenein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bernard Mulvey
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harris E. Blankenship
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - L. Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, 73117
| | - Jean-Francoise Pare
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Arielle Segal
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katharine E. McCann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joseph D. Dougherty
- Department of Psychiatry, Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yoland Smith
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Michael J. Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Evans AK, Saw NL, Woods CE, Vidano LM, Blumenfeld SE, Lam RK, Chu EK, Reading C, Shamloo M. Impact of high-fat diet on cognitive behavior and central and systemic inflammation with aging and sex differences in mice. Brain Behav Immun 2024; 118:334-354. [PMID: 38408498 PMCID: PMC11019935 DOI: 10.1016/j.bbi.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
Aging and age-related diseases are associated with cellular stress, metabolic imbalance, oxidative stress, and neuroinflammation, accompanied by cognitive impairment. Lifestyle factors such as diet, sleep fragmentation, and stress can potentiate damaging cellular cascades and lead to an acceleration of brain aging and cognitive impairment. High-fat diet (HFD) has been associated with obesity, metabolic disorders like diabetes, and cardiovascular disease. HFD also induces neuroinflammation, impairs learning and memory, and may increase anxiety-like behavior. Effects of a HFD may also vary between sexes. The interaction between Age- and Sex- and Diet-related changes in neuroinflammation and cognitive function is an important and poorly understood area of research. This study was designed to examine the effects of HFD on neuroinflammation, behavior, and neurodegeneration in mice in the context of aging or sex differences. In a series of studies, young (2-3 months) or old (12-13 months) C57BL/6J male mice or young male and female C57Bl/6J mice were fed either a standard diet (SD) or a HFD for 5-6 months. Behavior was assessed in Activity Chamber, Y-maze, Novel Place Recognition, Novel Object Recognition, Elevated Plus Maze, Open Field, Morris Water Maze, and Fear Conditioning. Post-mortem analyses assessed a panel of inflammatory markers in the plasma and hippocampus. Additionally, proteomic analysis of the hypothalamus, neurodegeneration, neuroinflammation in the locus coeruleus, and neuroinflammation in the hippocampus were assessed in a subset of young and aged male mice. We show that HFD increased body weight and decreased locomotor activity across groups compared to control mice fed a SD. HFD altered anxiety-related exploratory behavior. HFD impaired spatial learning and recall in young male mice and impaired recall in cued fear conditioning in young and aged male mice, with no effects on spatial learning or fear conditioning in young female mice. Effects of Age and Sex were observed on neuroinflammatory cytokines, with only limited effects of HFD. HFD had a more significant impact on systemic inflammation in plasma across age and sex. Aged male mice had induction of microglial immunoreactivity in both the locus coeruleus (LC) and hippocampus an effect that HFD exacerbated in the hippocampal CA1 region. Proteomic analysis of the hypothalamus revealed changes in pathways related to metabolism and neurodegeneration with both aging and HFD in male mice. Our findings suggest that HFD induces widespread systemic inflammation and limited neuroinflammation. In addition, HFD alters exploratory behavior in male and female mice, and impairs learning and memory in male mice. These results provide valuable insight into the impact of diet on cognition and aging pathophysiology.
Collapse
Affiliation(s)
- Andrew K Evans
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Nay L Saw
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Claire E Woods
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Laura M Vidano
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Sarah E Blumenfeld
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Rachel K Lam
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | - Emily K Chu
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304
| | | | - Mehrdad Shamloo
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304.
| |
Collapse
|
11
|
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
|
12
|
Deshpande G, Zhao S, Waggoner P, Beyers R, Morrison E, Huynh N, Vodyanoy V, Denney TS, Katz JS. Two Separate Brain Networks for Predicting Trainability and Tracking Training-Related Plasticity in Working Dogs. Animals (Basel) 2024; 14:1082. [PMID: 38612321 PMCID: PMC11010877 DOI: 10.3390/ani14071082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Functional brain connectivity based on resting-state functional magnetic resonance imaging (fMRI) has been shown to be correlated with human personality and behavior. In this study, we sought to know whether capabilities and traits in dogs can be predicted from their resting-state connectivity, as in humans. We trained awake dogs to keep their head still inside a 3T MRI scanner while resting-state fMRI data was acquired. Canine behavior was characterized by an integrated behavioral score capturing their hunting, retrieving, and environmental soundness. Functional scans and behavioral measures were acquired at three different time points across detector dog training. The first time point (TP1) was prior to the dogs entering formal working detector dog training. The second time point (TP2) was soon after formal detector dog training. The third time point (TP3) was three months' post detector dog training while the dogs were engaged in a program of maintenance training for detection work. We hypothesized that the correlation between resting-state FC in the dog brain and behavior measures would significantly change during their detection training process (from TP1 to TP2) and would maintain for the subsequent several months of detection work (from TP2 to TP3). To further study the resting-state FC features that can predict the success of training, dogs at TP1 were divided into a successful group and a non-successful group. We observed a core brain network which showed relatively stable (with respect to time) patterns of interaction that were significantly stronger in successful detector dogs compared to failures and whose connectivity strength at the first time point predicted whether a given dog was eventually successful in becoming a detector dog. A second ontologically based flexible peripheral network was observed whose changes in connectivity strength with detection training tracked corresponding changes in behavior over the training program. Comparing dog and human brains, the functional connectivity between the brain stem and the frontal cortex in dogs corresponded to that between the locus coeruleus and left middle frontal gyrus in humans, suggestive of a shared mechanism for learning and retrieval of odors. Overall, the findings point toward the influence of phylogeny and ontogeny in dogs producing two dissociable functional neural networks.
Collapse
Affiliation(s)
- Gopikrishna Deshpande
- Auburn University Neuroimaging Center, Department of Electrical & Computer Engineering, Auburn University, Auburn, AL 36849, USA; (S.Z.); (R.B.); (N.H.); (T.S.D.J.)
- Department of Psychological Sciences, Auburn University, Auburn, AL 36849, USA
- Alabama Advanced Imaging Consortium, Birmingham, AL 36849, USA
- Center for Neuroscience, Auburn University, Auburn, AL 36849, USA
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
- Department of Heritage Science and Technology, Indian Institute of Technology, Hyderabad 502285, India
| | - Sinan Zhao
- Auburn University Neuroimaging Center, Department of Electrical & Computer Engineering, Auburn University, Auburn, AL 36849, USA; (S.Z.); (R.B.); (N.H.); (T.S.D.J.)
| | - Paul Waggoner
- Canine Performance Sciences Program, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA;
| | - Ronald Beyers
- Auburn University Neuroimaging Center, Department of Electrical & Computer Engineering, Auburn University, Auburn, AL 36849, USA; (S.Z.); (R.B.); (N.H.); (T.S.D.J.)
| | - Edward Morrison
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, AL 36849, USA; (E.M.); (V.V.)
| | - Nguyen Huynh
- Auburn University Neuroimaging Center, Department of Electrical & Computer Engineering, Auburn University, Auburn, AL 36849, USA; (S.Z.); (R.B.); (N.H.); (T.S.D.J.)
| | - Vitaly Vodyanoy
- Department of Anatomy, Physiology & Pharmacology, Auburn University, Auburn, AL 36849, USA; (E.M.); (V.V.)
| | - Thomas S. Denney
- Auburn University Neuroimaging Center, Department of Electrical & Computer Engineering, Auburn University, Auburn, AL 36849, USA; (S.Z.); (R.B.); (N.H.); (T.S.D.J.)
- Department of Psychological Sciences, Auburn University, Auburn, AL 36849, USA
- Alabama Advanced Imaging Consortium, Birmingham, AL 36849, USA
- Center for Neuroscience, Auburn University, Auburn, AL 36849, USA
| | - Jeffrey S. Katz
- Auburn University Neuroimaging Center, Department of Electrical & Computer Engineering, Auburn University, Auburn, AL 36849, USA; (S.Z.); (R.B.); (N.H.); (T.S.D.J.)
- Department of Psychological Sciences, Auburn University, Auburn, AL 36849, USA
- Alabama Advanced Imaging Consortium, Birmingham, AL 36849, USA
- Center for Neuroscience, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
13
|
Scognamiglio S, Aljohani YM, Olson TT, Forcelli PA, Dezfuli G, Kellar KJ. Restoration of norepinephrine release, cognitive performance, and dendritic spines by amphetamine in aged rat brain. Aging Cell 2024; 23:e14087. [PMID: 38332648 PMCID: PMC11019150 DOI: 10.1111/acel.14087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/10/2024] Open
Abstract
Age-related dysfunctions in specific neurotransmitter systems likely play an important role in cognitive decline even in its most subtle forms. Therefore, preservation or improvement of cognition via augmentation of neurotransmission is a potential therapeutic strategy to prevent further cognitive deficits. Here we identified a particular neuronal vulnerability in the aged Fischer 344 rat brain, an animal model of neurocognitive aging. Specifically, we demonstrated a marked impairment in glutamate-stimulated release of norepinephrine (NE) in the hippocampus and cerebral cortex of aged rats, and established that this release was mediated by N-methyl-D-aspartate (NMDA) receptors. Further, we also demonstrated that this decrease in NE release is fully rescued by the psychostimulant drug amphetamine (AMPH). Moreover, we showed that AMPH increases dendritic spine maturation, and importantly shows preclinical efficacy in restoring memory deficits in the aged rat through its actions to potentiate NE neurotransmission at β-adrenergic receptors. Taken together, our results suggest that deficits in glutamate-stimulated release of NE may contribute to and possibly be a determinant of neuronal vulnerability underlying cognitive decline during aging, and that these deficits can be corrected with currently available drugs. Overall these studies suggest that repurposing of psychostimulants for age-associated cognitive deficits is a potential avenue to delay or prevent cognitive decline and/or frank dementia later in life.
Collapse
Affiliation(s)
- Serena Scognamiglio
- Department of Pharmacology & PhysiologyGeorgetown University Medical CenterWashington, DCUSA
| | - Yousef M. Aljohani
- Department of Pharmacology & PhysiologyGeorgetown University Medical CenterWashington, DCUSA
| | - Thao T. Olson
- Department of Pharmacology & PhysiologyGeorgetown University Medical CenterWashington, DCUSA
| | - Patrick A. Forcelli
- Department of Pharmacology & PhysiologyGeorgetown University Medical CenterWashington, DCUSA
| | - Ghazaul Dezfuli
- Department of Pharmacology & PhysiologyGeorgetown University Medical CenterWashington, DCUSA
| | - Kenneth J. Kellar
- Department of Pharmacology & PhysiologyGeorgetown University Medical CenterWashington, DCUSA
| |
Collapse
|
14
|
Taherian N, Vaezi G, Neamati A, Hojjati V, Ghorbani-Taherdehi F, Sahebkar A, Gorji-Valokola M. The dose-dependent neuroprotective effect of norepinephrine in improving memory retrieval in an experimental model of multiple sclerosis, experimental autoimmune encephalomyelitis. Brain Res Bull 2024; 209:110907. [PMID: 38395110 DOI: 10.1016/j.brainresbull.2024.110907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/26/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Multiple sclerosis (MS) is considered an immune-mediated inflammatory disorder that causes cognitive impairments by damaging the hippocampal tissue. Conversely, norepinephrine (NEP) has anti-inflammatory and re-myelinating properties, which improve cognitive impairments. The aim of this study was to assess the neuroprotective effects of NEP on learning and memory disorders in an experimental animal model of MS. Two guide cannulas were bilaterally implanted in the rat hippocampal CA1 regions. After recovery, the animals received 3 μl of 0.01% ethidium bromide (EtB) in each of both hippocampal regions. After three days, the rats were randomly divided into 6 groups (8 rats/group), including control, sham 1, sham 2, and three groups of NEP 0.25, 0.5, and 1 mg/kg by intrahippocampal injection. Behavioral tests (e.g. shuttle box test and open-field test) were then performed. Finally, ROS, MDA, GSH, TNF-α, IL-6, and IL-1β concentrations in the left CA1 area, as well as using western-blot analysis, p-p38, p-JNK, p-AKT, p-ERK1/2, p-NMDA, p-AMPA, p-CREB, and BDNF proteins in the right CA1 region evaluated. The EtB injection increased ROS, MDA, TNF-α, IL-6, and IL-1β levels, as well as p-JNK and p-P38, except all other proteins, while decreasing GSH content, as well as step-through latency and locomotor activity in sham groups compared to the control group. Conversely, NEP (0.5 and 1 mg/kg, particularly at the dose of 1 mg/kg) counterbalanced all the alterations mentioned above in comparison to the sham groups. The EtB induced learning and memory impairment; however, NEP dose-dependently restored these impairments to normal levels.
Collapse
Affiliation(s)
- Narjes Taherian
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Gholamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Ali Neamati
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Vida Hojjati
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Faezeh Ghorbani-Taherdehi
- Department of Anatomy and Cell Biology, School of Medicine, Esfahan University of Medical Sciences, Esfahan, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Gorji-Valokola
- Department of Food and Drug Administration, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Pharmacology, Brain and Spinal Injury Repair Research Center, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
15
|
Xie H, Tian Y, Li Z, Wang K, Li R, Yi S, Chen A, Chen J, Liu J, Wei X, Gao X. Activation of Beta-adrenergic Receptors Upregulates the Signal-to-Noise Ratio of Auditory Input in the Medial Prefrontal Cortex and Mediates Auditory Fear Conditioning. Mol Neurobiol 2024; 61:1833-1844. [PMID: 37787950 DOI: 10.1007/s12035-023-03667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023]
Abstract
Norepinephrine (NE) is involved in auditory fear conditioning (AFC) in posttraumatic stress disorder (PTSD). However, it is still unclear how it acts on neurons. We aimed to investigate whether the activation of the β-adrenergic receptor (β-AR) improves AFC by sensitization of the prelimbic (PL) cortex at the animal, cellular, and molecular levels. In vivo single-cell electrophysiological recording was used to characterize the changes in neurons in the PL cortex after AFC. Then, PL neurons were locally administrated by the β-AR agonist isoproterenol (ISO), the GABAaR agonist muscimol, or intervened by optogenetic method, respectively. Western blotting and immunohistochemistry were finally used to assess molecular changes. Noise and low-frequency tones induced similar AFC. The expression of β-ARs in PL cortex neurons was upregulated after fear conditioning. Microinjection of muscimol into the PL cortex blocked the conformation of AFC, whereas ISO injection facilitated AFC. Moreover, PL neurons can be distinguished into two types, with type I but not type II neurons responding to conditioned sound and being regulated by β-ARs. Our results showed that β-ARs in the PL cortex regulate conditional fear learning by activating type I PL neurons.
Collapse
Affiliation(s)
- Haiting Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Yueqin Tian
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhongli Li
- Respiratory Medicine, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Kaitao Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Runtong Li
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Shang Yi
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Aimin Chen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Jian Chen
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Jun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang road, Guangzhou, Guangdong, 510280, People's Republic of China.
| | - Xuhong Wei
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510282, People's Republic of China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510282, People's Republic of China.
| | - Xiaoya Gao
- Department of Neurology, Zhujiang Hospital of Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China.
- Department of Pediatric Neurology, Zhujiang Hospital, Southern Medical University, 253 Gongye Avenue, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
16
|
Zhang T, Dolga AM, Eisel ULM, Schmidt M. Novel crosstalk mechanisms between GluA3 and Epac2 in synaptic plasticity and memory in Alzheimer's disease. Neurobiol Dis 2024; 191:106389. [PMID: 38142840 DOI: 10.1016/j.nbd.2023.106389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease which accounts for the most cases of dementia worldwide. Impaired memory, including acquisition, consolidation, and retrieval, is one of the hallmarks in AD. At the cellular level, dysregulated synaptic plasticity partly due to reduced long-term potentiation (LTP) and enhanced long-term depression (LTD) underlies the memory deficits in AD. GluA3 containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are one of key receptors involved in rapid neurotransmission and synaptic plasticity. Recent studies revealed a novel form of GluA3 involved in neuronal plasticity that is dependent on cyclic adenosine monophosphate (cAMP), rather than N-methyl-d-aspartate (NMDA). However, this cAMP-dependent GluA3 pathway is specifically and significantly impaired by amyloid beta (Aβ), a pathological marker of AD. cAMP is a key second messenger that plays an important role in modulating memory and synaptic plasticity. We previously reported that exchange protein directly activated by cAMP 2 (Epac2), acting as a main cAMP effector, plays a specific and time-limited role in memory retrieval. From electrophysiological perspective, Epac2 facilities the maintenance of LTP, a cellular event closely associated with memory retrieval. Additionally, Epac2 was found to be involved in the GluA3-mediated plasticity. In this review, we comprehensively summarize current knowledge regarding the specific roles of GluA3 and Epac2 in synaptic plasticity and memory, and their potential association with AD.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
17
|
Azevedo M, Martinho R, Oliveira A, Correia-de-Sá P, Moreira-Rodrigues M. Molecular pathways underlying sympathetic autonomic overshooting leading to fear and traumatic memories: looking for alternative therapeutic options for post-traumatic stress disorder. Front Mol Neurosci 2024; 16:1332348. [PMID: 38260808 PMCID: PMC10800988 DOI: 10.3389/fnmol.2023.1332348] [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: 11/02/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
The sympathoadrenal medullary system and the hypothalamic-pituitary-adrenal axis are both activated upon stressful events. The release of catecholamines, such as dopamine, norepinephrine (NE), and epinephrine (EPI), from sympathetic autonomic nerves participate in the adaptive responses to acute stress. Most theories suggest that activation of peripheral β-adrenoceptors (β-ARs) mediates catecholamines-induced memory enhancement. These include direct activation of β-ARs in the vagus nerve, as well as indirect responses to catecholamine-induced glucose changes in the brain. Excessive sympathetic activity is deeply associated with memories experienced during strong emotional stressful conditions, with catecholamines playing relevant roles in fear and traumatic memories consolidation. Recent findings suggest that EPI is implicated in fear and traumatic contextual memories associated with post-traumatic stress disorder (PTSD) by increasing hippocampal gene transcription (e.g., Nr4a) downstream to cAMP response-element protein activation (CREB). Herein, we reviewed the literature focusing on the molecular mechanisms underlying the pathophysiology of memories associated with fear and traumatic experiences to pave new avenues for the treatment of stress and anxiety conditions, such as PTSD.
Collapse
Affiliation(s)
- Márcia Azevedo
- Laboratory of General Physiology, Department of Immuno-Physiology and Pharmacology and Center for Drug Discovery and Innovative Medicines (MedInUP), School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Raquel Martinho
- Laboratory of General Physiology, Department of Immuno-Physiology and Pharmacology and Center for Drug Discovery and Innovative Medicines (MedInUP), School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Ana Oliveira
- Laboratory of General Physiology, Department of Immuno-Physiology and Pharmacology and Center for Drug Discovery and Innovative Medicines (MedInUP), School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratory of Pharmacology and Neurobiology, Department of Immuno-Physiology and Pharmacology and Center for Drug Discovery and Innovative Medicines (MedInUP), School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| | - Mónica Moreira-Rodrigues
- Laboratory of General Physiology, Department of Immuno-Physiology and Pharmacology and Center for Drug Discovery and Innovative Medicines (MedInUP), School of Medicine and Biomedical Sciences (ICBAS), University of Porto (UP), Porto, Portugal
| |
Collapse
|
18
|
Wilson LR, Plummer NW, Evsyukova IY, Patino D, Stewart CL, Smith KG, Konrad KS, Fry SA, Deal AL, Kilonzo VW, Panda S, Sciolino NR, Cushman JD, Jensen P. Partial or Complete Loss of Norepinephrine Differentially Alters Contextual Fear and Catecholamine Release Dynamics in Hippocampal CA1. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:51-60. [PMID: 38058990 PMCID: PMC10695841 DOI: 10.1016/j.bpsgos.2023.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 12/08/2023] Open
Abstract
Background Contextual fear learning is heavily dependent on the hippocampus. Despite evidence that catecholamines contribute to contextual encoding and memory retrieval, the precise temporal dynamics of their release in the hippocampus during behavior is unknown. In addition, new animal models are required to probe the effects of altered catecholamine synthesis on release dynamics and contextual learning. Methods We generated 2 new mouse models of altered locus coeruleus-norepinephrine (NE) synthesis and utilized them together with GRABNE and GRABDA sensors and in vivo fiber photometry to investigate NE and dopamine (DA) release dynamics in the dorsal hippocampal CA1 during contextual fear conditioning. Results Aversive foot shock increased both NE and DA release in the dorsal CA1, while freezing behavior associated with recall of fear memory was accompanied by decreased release. Moreover, we found that freezing at the recent time point was sensitive to both partial and complete loss of locus coeruleus-NE synthesis throughout prenatal and postnatal development, similar to previous observations of mice with global loss of NE synthesis beginning postnatally. In contrast, freezing at the remote time point was compromised only by complete loss of locus coeruleus-NE synthesis beginning prenatally. Conclusions Overall, these findings provide novel insights into the role of NE in contextual fear and the precise temporal dynamics of both NE and DA during freezing behavior and highlight complex relationships between genotype, sex, and NE signaling.
Collapse
Affiliation(s)
- Leslie R. Wilson
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
- Neurobehavioral Core Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Nicholas W. Plummer
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Irina Y. Evsyukova
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Daniela Patino
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Casey L. Stewart
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Kathleen G. Smith
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Kathryn S. Konrad
- Social and Scientific Systems, Inc., a DLH Holdings Corp Company, Durham, North Carolina
| | - Sydney A. Fry
- Neurobehavioral Core Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Alex L. Deal
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Victor W. Kilonzo
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Sambit Panda
- Neurobehavioral Core Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Natale R. Sciolino
- Department of Physiology and Neurobiology, Department of Biomedical Engineering, Institute for System Genomics, Connecticut Institute for the Brain & Cognitive Sciences, University of Connecticut, Storrs, Connecticut
| | - Jesse D. Cushman
- Neurobehavioral Core Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Patricia Jensen
- Neurobiology Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| |
Collapse
|
19
|
Gheidi A, Davidson CJ, Simpson SC, Yahya MA, Sadik N, Mascarin AT, Perrine SA. Norepinephrine depletion in the brain sex-dependently modulates aspects of spatial learning and memory in female and male rats. Psychopharmacology (Berl) 2023; 240:2585-2595. [PMID: 37658879 PMCID: PMC11069163 DOI: 10.1007/s00213-023-06453-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
RATIONALE The contribution of norepinephrine on the different phases of spatial memory processing remains incompletely understood. To address this gap, this study depleted norepinephrine in the brain and then conducted a spatial learning task with multiple phases. METHODS Male and female Wistar rats were administered 50 mg/kg/i.p. of DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine) to deplete norepinephrine. After 10 days, rats were trained on a 20-hole Barnes maze spatial navigation task for 5 days. On the fifth day, animals were euthanized and HPLC was used to confirm depletion of norepinephrine in select brain regions. In Experiment 2, rats underwent a similar Barnes maze procedure that continued beyond day 5 to investigate memory retrieval and updating via a single probe trial and two reversal learning periods. RESULTS Rats did not differ in Barnes maze acquisition between DSP-4 and saline-injected rats; however, initial acquisition differed between the sexes. HPLC analysis confirmed selective depletion of norepinephrine in dorsal hippocampus and cingulate cortex without impact to other monoamines. When retrieval was tested through a probe trial, DSP-4-improved memory retrieval in males but impaired it in females. Cognitive flexibility was transiently impacted by DSP-4 in males only. CONCLUSIONS Despite significantly reducing levels of norepinephrine, DSP-4 had only a modest impact on spatial learning and behavioral flexibility. Memory retrieval and early reversal learning were most affected and in a sex-specific manner. These data suggest that norepinephrine has sex-specific neuromodulatory effects on memory retrieval with a lesser effect on cognitive flexibility and no impact on acquisition of learned behavior.
Collapse
Affiliation(s)
- Ali Gheidi
- Department of Biomedical Sciences, Mercer University School of Medicine, 1550 College St., Macon, GA, 31207, USA.
| | - Cameron J Davidson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Serena C Simpson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Majd A Yahya
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nareen Sadik
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Alixandria T Mascarin
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
20
|
Stanley AT, Post MR, Lacefield C, Sulzer D, Miniaci MC. Norepinephrine release in the cerebellum contributes to aversive learning. Nat Commun 2023; 14:4852. [PMID: 37563141 PMCID: PMC10415399 DOI: 10.1038/s41467-023-40548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
The modulation of dopamine release from midbrain projections to the striatum has long been demonstrated in reward-based learning, but the synaptic basis of aversive learning is far less characterized. The cerebellum receives axonal projections from the locus coeruleus, and norepinephrine release is implicated in states of arousal and stress, but whether aversive learning relies on plastic changes in norepinephrine release in the cerebellum is unknown. Here we report that in mice, norepinephrine is released in the cerebellum following an unpredicted noxious event (a foot-shock) and that this norepinephrine release is potentiated powerfully with fear acquisition as animals learn that a previously neutral stimulus (tone) predicts the aversive event. Importantly, both chemogenetic and optogenetic inhibition of the locus coeruleus-cerebellum pathway block fear memory without impairing motor function. Thus, norepinephrine release in the cerebellum is modulated by experience and underlies aversive learning.
Collapse
Affiliation(s)
- Adrien T Stanley
- Departments of Psychiatry, Neurology, and Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Michael R Post
- Departments of Psychiatry, Neurology, and Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - Clay Lacefield
- Departments of Psychiatry, Neurology, and Pharmacology, Columbia University Medical Center, New York, NY, USA
| | - David Sulzer
- Departments of Psychiatry, Neurology, and Pharmacology, Columbia University Medical Center, New York, NY, USA.
| | | |
Collapse
|
21
|
Ireton KE, Xing X, Kim K, Weiner JC, Jacobi AA, Grover A, Foote M, Ota Y, Berman R, Hanks T, Hell JW. Regulation of the Ca 2+ Channel Ca V1.2 Supports Spatial Memory and Its Flexibility and LTD. J Neurosci 2023; 43:5559-5573. [PMID: 37419689 PMCID: PMC10376936 DOI: 10.1523/jneurosci.1521-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 04/30/2023] [Accepted: 05/15/2023] [Indexed: 07/09/2023] Open
Abstract
Widespread release of norepinephrine (NE) throughout the forebrain fosters learning and memory via adrenergic receptor (AR) signaling, but the molecular mechanisms are largely unknown. The β2 AR and its downstream effectors, the trimeric stimulatory Gs-protein, adenylyl cyclase (AC), and the cAMP-dependent protein kinase A (PKA), form a unique signaling complex with the L-type Ca2+ channel (LTCC) CaV1.2. Phosphorylation of CaV1.2 by PKA on Ser1928 is required for the upregulation of Ca2+ influx on β2 AR stimulation and long-term potentiation induced by prolonged theta-tetanus (PTT-LTP) but not LTP induced by two 1-s-long 100-Hz tetani. However, the function of Ser1928 phosphorylation in vivo is unknown. Here, we show that S1928A knock-in (KI) mice of both sexes, which lack PTT-LTP, express deficiencies during initial consolidation of spatial memory. Especially striking is the effect of this mutation on cognitive flexibility as tested by reversal learning. Mechanistically, long-term depression (LTD) has been implicated in reversal learning. It is abrogated in male and female S1928A knock-in mice and by β2 AR antagonists and peptides that displace β2 AR from CaV1.2. This work identifies CaV1.2 as a critical molecular locus that regulates synaptic plasticity, spatial memory and its reversal, and LTD.SIGNIFICANCE STATEMENT We show that phosphorylation of the Ca2+ channel CaV1.2 on Ser1928 is important for consolidation of spatial memory and especially its reversal, and long-term depression (LTD). Identification of Ser1928 as critical for LTD and reversal learning supports the model that LTD underlies flexibility of reference memory.
Collapse
Affiliation(s)
- Kyle E Ireton
- Department of Pharmacology, University of California, Davis, California 95616-8636
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Xiaoming Xing
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Karam Kim
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Justin C Weiner
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Ariel A Jacobi
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Aarushi Grover
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Molly Foote
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Yusuke Ota
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Robert Berman
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Timothy Hanks
- Center for Neuroscience, University of California, Davis, California 95616-8636
- Department of Neurology, University of California, Davis, California 95616-8636
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, California 95616-8636
- Center for Neuroscience, University of California, Davis, California 95616-8636
| |
Collapse
|
22
|
Larsen ME, Buonarati OR, Qian H, Hell JW, Bayer KU. Stimulating β-adrenergic receptors promotes synaptic potentiation by switching CaMKII movement from LTD to LTP mode. J Biol Chem 2023; 299:104706. [PMID: 37061000 PMCID: PMC10200978 DOI: 10.1016/j.jbc.2023.104706] [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: 09/21/2022] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/17/2023] Open
Abstract
Learning, memory, and cognition are thought to require synaptic plasticity, specifically including hippocampal long-term potentiation and depression (LTP and LTD). LTP versus LTD is induced by high-frequency stimulation versus low-frequency, but stimulating β-adrenergic receptors (βARs) enables LTP induction also by low-frequency stimulation (1 Hz) or theta frequencies (∼5 Hz) that do not cause plasticity by themselves. In contrast to high-frequency stimulation-LTP, such βAR-LTP requires Ca2+-flux through L-type voltage-gated Ca2+-channels, not N-methyl-D-aspartate-type glutamate receptors. Surprisingly, we found that βAR-LTP still required a nonionotropic scaffolding function of the N-methyl-D-aspartate-type glutamate receptor: the stimulus-induced binding of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) to its GluN2B subunit that mediates CaMKII movement to excitatory synapses. In hippocampal neurons, β-adrenergic stimulation with isoproterenol (Iso) transformed LTD-type CaMKII movement to LTP-type movement, resulting in CaMKII movement to excitatory instead of inhibitory synapses. Additionally, Iso enabled induction of a major cell-biological feature of LTP in response to LTD stimuli: increased surface expression of GluA1 fused with super-ecliptic pHluorein. Like for βAR-LTP in hippocampal slices, the Iso effects on CaMKII movement and surface expression of GluA1 fused with super-ecliptic pHluorein involved L-type Ca2+-channels and specifically required β2-ARs. Taken together, these results indicate that Iso transforms LTD stimuli to LTP signals by switching CaMKII movement and GluN2B binding to LTP mode.
Collapse
Affiliation(s)
- Matthew E Larsen
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Olivia R Buonarati
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Hai Qian
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Johannes W Hell
- Department of Pharmacology, University of Iowa, Iowa City, Iowa, USA; Department of Pharmacology, University of California at Davis, Davis, California, USA.
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
23
|
Jami SA, Wilkinson BJ, Guglietta R, Hartel N, Babiec WE, Graham NA, Coba MP, O'Dell TJ. Functional and phosphoproteomic analysis of β-adrenergic receptor signaling at excitatory synapses in the CA1 region of the ventral hippocampus. Sci Rep 2023; 13:7493. [PMID: 37161045 PMCID: PMC10170123 DOI: 10.1038/s41598-023-34401-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/28/2023] [Indexed: 05/11/2023] Open
Abstract
Activation of β-adrenergic receptors (β-ARs) not only enhances learning and memory but also facilitates the induction of long-term potentiation (LTP), a form of synaptic plasticity involved in memory formation. To identify the mechanisms underlying β-AR-dependent forms of LTP we examined the effects of the β-AR agonist isoproterenol on LTP induction at excitatory synapses onto CA1 pyramidal cells in the ventral hippocampus. LTP induction at these synapses is inhibited by activation of SK-type K+ channels, suggesting that β-AR activation might facilitate LTP induction by inhibiting SK channels. However, although the SK channel blocker apamin enhanced LTP induction, it did not fully mimic the effects of isoproterenol. We therefore searched for potential alternative mechanisms using liquid chromatography-tandem mass spectrometry to determine how β-AR activation regulates phosphorylation of postsynaptic density (PSD) proteins. Strikingly, β-AR activation regulated hundreds of phosphorylation sites in PSD proteins that have diverse roles in dendritic spine structure and function. Moreover, within the core scaffold machinery of the PSD, β-AR activation increased phosphorylation at several sites previously shown to be phosphorylated after LTP induction. Together, our results suggest that β-AR activation recruits a diverse set of signaling pathways that likely act in a concerted fashion to regulate LTP induction.
Collapse
Affiliation(s)
- Shekib A Jami
- Molecular, Cellular, and Integrative Physiology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Ryan Guglietta
- Interdepartmental PhD Program for Neuroscience, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicolas Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Walter E Babiec
- Undergraduate Interdepartmental Program for Neuroscience, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Marcelo P Coba
- Zilkha Neurogenetic Institute, Los Angeles, CA, USA
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Thomas J O'Dell
- Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Chodari L, Derafshpour L, Jafari A, Ghasemi M, Mehranfard N. Exercise may alleviate age-related spatial memory impairment by rescuing β-adrenergic receptor dysregulation via both G protein-dependent and β-arrestin-dependent mechanisms in rat hippocampus. Brain Res 2023; 1804:148250. [PMID: 36690167 DOI: 10.1016/j.brainres.2023.148250] [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: 11/10/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Hippocampal-dependent memory abilities including spatial memory decline with age. Exercise improves memory decline in aging brain, but, the precise mechanisms are still unknown. Learning and memory are recently hypothesized to be mediated by a β-arrestin (βArr)-dependent β-adrenergic pathway. Hence, we examined the effect of 8 weeks of treadmill exercise on hippocampal expression of β-adrenergic receptors (β-ARs; members of the G protein-coupled receptor family), and βArrs as well as spatial learning and memory in aged male rats to determine whether β-AR/βArr pathway could be involved in age-related memory decline. A total of 24 young (3-month-old) and aged (18-month-old) male Wistar rats were divided into young control, aged sedentary, and aged + exercise (n = 8 for each). Western blot for β1- and β2-ARs as well as βArr1 and βArr2 was performed. Spatial learning and memory were evaluated with the Morris water maze. The results showed significant up-regulation of β1-ARs as well as significant down-regulation of β2-AR and βArrs (βArr1 and βArr2) in the hippocampus of aged rats. Spatial memory, but not spatial learning, was impaired in aging, and treadmill exercise improved it. Notably, the improvement in spatial memory was accompanied by amelioration of β-ARs dysregulation and increase in βArr2 levels after exercise. There was a negative association between the expression of βArr2 and β1-AR, but not β2-AR, such that an increase in βArr2 by exercise was associated with reduced β1-AR expression, suggesting βArr2 may contribute to posttranslational down-regulation of β1-ARs. These data suggest that both G protein-dependent and β-arrestin-dependent β-AR pathways may regulate spatial learning and memory in aging brain.
Collapse
Affiliation(s)
- Leila Chodari
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Derafshpour
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
25
|
Isingrini E, Guinaudie C, Perret L, Guma E, Gorgievski V, Blum ID, Colby-Milley J, Bairachnaya M, Mella S, Adamantidis A, Storch KF, Giros B. Behavioral and Transcriptomic Changes Following Brain-Specific Loss of Noradrenergic Transmission. Biomolecules 2023; 13:biom13030511. [PMID: 36979445 PMCID: PMC10046559 DOI: 10.3390/biom13030511] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Noradrenaline (NE) plays an integral role in shaping behavioral outcomes including anxiety/depression, fear, learning and memory, attention and shifting behavior, sleep-wake state, pain, and addiction. However, it is unclear whether dysregulation of NE release is a cause or a consequence of maladaptive orientations of these behaviors, many of which associated with psychiatric disorders. To address this question, we used a unique genetic model in which the brain-specific vesicular monoamine transporter-2 (VMAT2) gene expression was removed in NE-positive neurons disabling NE release in the entire brain. We engineered VMAT2 gene splicing and NE depletion by crossing floxed VMAT2 mice with mice expressing the Cre-recombinase under the dopamine β-hydroxylase (DBH) gene promotor. In this study, we performed a comprehensive behavioral and transcriptomic characterization of the VMAT2DBHcre KO mice to evaluate the role of central NE in behavioral modulations. We demonstrated that NE depletion induces anxiolytic and antidepressant-like effects, improves contextual fear memory, alters shifting behavior, decreases the locomotor response to amphetamine, and induces deeper sleep during the non-rapid eye movement (NREM) phase. In contrast, NE depletion did not affect spatial learning and memory, working memory, response to cocaine, and the architecture of the sleep-wake cycle. Finally, we used this model to identify genes that could be up- or down-regulated in the absence of NE release. We found an up-regulation of the synaptic vesicle glycoprotein 2c (SV2c) gene expression in several brain regions, including the locus coeruleus (LC), and were able to validate this up-regulation as a marker of vulnerability to chronic social defeat. The NE system is a complex and challenging system involved in many behavioral orientations given it brain wide distribution. In our study, we unraveled specific role of NE neurotransmission in multiple behavior and link it to molecular underpinning, opening future direction to understand NE role in health and disease.
Collapse
Affiliation(s)
- Elsa Isingrini
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
- Faculté des Sciences Fondamentales et Biomédicales, Université Paris Cité, INCC UMR 8002, CNRS, F-75006 Paris, France
| | - Chloé Guinaudie
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Léa Perret
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Elisa Guma
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Victor Gorgievski
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Ian D. Blum
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Jessica Colby-Milley
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Maryia Bairachnaya
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Sébastien Mella
- Cytometry and Biomarkers Platform, Unit of Technology and Service, Institut Pasteur, Université de Paris, F-75015 Paris, France
- Bioinformatics and Biostatistics Hub Platform, Institut Pasteur, Université de Paris, F-75015 Paris, France
| | - Antoine Adamantidis
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| | - Kai-Florian Storch
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
| | - Bruno Giros
- Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, QC H4H 1R3, Canada
- Faculté des Sciences Fondamentales et Biomédicales, Université Paris Cité, INCC UMR 8002, CNRS, F-75006 Paris, France
- Correspondence:
| |
Collapse
|
26
|
Iqbal Z, Lei Z, Ramkrishnan AS, Liu S, Hasan M, Akter M, Lam YY, Li Y. Adrenergic signalling to astrocytes in anterior cingulate cortex contributes to pain-related aversive memory in rats. Commun Biol 2023; 6:10. [PMID: 36604595 PMCID: PMC9816175 DOI: 10.1038/s42003-022-04405-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 12/23/2022] [Indexed: 01/06/2023] Open
Abstract
Pain contains both sensory and affective dimensions. We identify the role of norepinephrine in colorectal distention (sub-threshold for acute pain) induced conditioned place avoidance and plasticity gene expression in the anterior cingulate cortex (ACC). Activating locus coeruleus (LC)-projecting ACC neurons facilitates pain-evoked aversive consolidation and memory, while inhibiting LC-projecting ACC neurons reversibly blocks it. Optogenetic activation of ACC astrocytes facilitates aversive behaviour. ACC astrocytic Gi manipulation suppressed aversive behaviour and early plasticity gene expression induced by opto-activation of LC neurons projecting to ACC. Evidences for the critical role of β2AR in ACC astrocytes were provided using AAV encoding β2AR miRNAi to knockdown β2AR in astrocytes. In contrast, opto-activation of ACC astrocytic β2ARs promotes aversion memory. Our findings suggest that projection-specific adrenergic astrocytic signalling in ACC is integral to system-wide neuromodulation in response to visceral stimuli, and plays a key role in mediating pain-related aversion consolidation and memory formation.
Collapse
Affiliation(s)
- Zafar Iqbal
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Zhuogui Lei
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Aruna S Ramkrishnan
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Shu Liu
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Mahadi Hasan
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Mastura Akter
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Yuk Yan Lam
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Ying Li
- Department of Neuroscience, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong.
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong.
| |
Collapse
|
27
|
Portela Moreira I, Henriques T, Vieira-Coelho MA, Guimarães J. Dysfunction of norepinephrine and its metabolites in Alzheimer's dementia - A review with meta-analysis. Ageing Res Rev 2023; 83:101784. [PMID: 36368648 DOI: 10.1016/j.arr.2022.101784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Some studies point locus coeruleus cell loss, the central nervous system main source of norepinephrine, to be one of the earliest neuropathological events of Alzheimer's disease (AD). However, there are conflicting reports regarding the level of norepinephrine and its metabolites (3-Methoxy-4-hydroxyphenylglycol (MHPG), 3,5-dihydroxyphenylglycine (DHPG) and 3,4 -dihydroxyphenylglycolaldehyde (DOPEGAL)) in AD patients. Uncover these alterations may be a key factor for understanding cognitive deficits and AD pathology. We review the literature that compare norepinephrine and its metabolites between AD patients and non-demented controls. A meta-analysis did not reveal significant statistical differences, but there was a trend towards a lower level of norepinephrine of AD, with almost no difference in MHPG in the cerebrospinal fluid. Regarding MHPG in plasma, DHPG and DOPEGAL we only performed a qualitative analyse due to the small or absent number of studies. These findings point to a decrease in norepinephrine, what is in line with locus coeluleus cell loss in AD. The absence of statistical difference and an equal level of MHGP could indicate a compensatory mechanism.
Collapse
Affiliation(s)
- Isabel Portela Moreira
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine of Porto University, Porto, Portugal; Neurology Department, Hospital Privado de Gaia do Grupo Trofa Saúde, Vila Nova de Gaia, Portugal.
| | - Teresa Henriques
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of Porto University, Porto, Portugal; Centre for Health Technology Services Research (CINTESIS), Faculty of Medicine of Porto University, Porto, Portugal
| | - Maria Augusta Vieira-Coelho
- Department of Biomedicine, Faculty of Medicine, Faculty of Medicine of Porto University, Porto, Portugal; Department of Psychiatry and Mental Health, University Hospital Center of São João, Porto, Portugal
| | - Joana Guimarães
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine of Porto University, Porto, Portugal; Neurology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
28
|
Maity S, Abbaspour R, Nahabedian D, Connor SA. Norepinephrine, beyond the Synapse: Coordinating Epigenetic Codes for Memory. Int J Mol Sci 2022; 23:ijms23179916. [PMID: 36077313 PMCID: PMC9456295 DOI: 10.3390/ijms23179916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The noradrenergic system is implicated in neuropathologies contributing to major disorders of the memory, including post-traumatic stress disorder and Alzheimer’s disease. Determining the impact of norepinephrine on cellular function and plasticity is thus essential for making inroads into our understanding of these brain conditions, while expanding our capacity for treating them. Norepinephrine is a neuromodulator within the mammalian central nervous system which plays important roles in cognition and associated synaptic plasticity. Specifically, norepinephrine regulates the formation of memory through the stimulation of β-ARs, increasing the dynamic range of synaptic modifiability. The mechanisms through which NE influences neural circuit function have been extended to the level of the epigenome. This review focuses on recent insights into how the noradrenergic recruitment of epigenetic modifications, including DNA methylation and post-translational modification of histones, contribute to homo- and heterosynaptic plasticity. These advances will be placed in the context of synaptic changes associated with memory formation and linked to brain disorders and neurotherapeutic applications.
Collapse
Affiliation(s)
- Sabyasachi Maity
- Department of Physiology, Neuroscience, and Behavioral Sciences, St. George’s University School of Medicine, True Blue FZ818, Grenada
| | - Raman Abbaspour
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
| | - David Nahabedian
- The Center for Biomedical Visualization, Department of Anatomical Sciences, St. George’s University School of Medicine, True Blue FZ818, Grenada
| | - Steven A. Connor
- Department of Biology, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 33803)
| |
Collapse
|
29
|
Rudzki S. Is PTSD an Evolutionary Survival Adaptation Initiated by Unrestrained Cytokine Signaling and Maintained by Epigenetic Change? Mil Med 2022; 188:usac095. [PMID: 35446412 DOI: 10.1093/milmed/usac095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/01/2022] [Accepted: 03/24/2022] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Treatment outcomes for PTSD with current psychological therapies are poor, with very few patients achieving sustained symptom remission. A number of authors have identified physiological and immune disturbances in Post Traumatic Stress Disorder (PTSD) patients, but there is no unifying hypothesis that explains the myriad features of the disorder. MATERIALS AND METHODS The medical literature was reviewed over a 6-year period primarily using the medical database PUBMED. RESULTS The literature contains numerous papers that have identified a range of physiological and immune dysfunction in association with PTSD. This paper proposes that unrestrained cytokine signaling induces epigenetic changes that promote an evolutionary survival adaptation, which maintains a defensive PTSD phenotype. The brain can associate immune signaling with past threat and initiate a defensive behavioral response. The sympathetic nervous system is pro-inflammatory, while the parasympathetic nervous system is anti-inflammatory. Prolonged cholinergic withdrawal will promote a chronic inflammatory state. The innate immune cytokine IL-1β has pleiotropic properties and can regulate autonomic, glucocorticoid, and glutamate receptor functions, sleep, memory, and epigenetic enzymes. Changes in epigenetic enzyme activity can potentially alter phenotype and induce an adaptation. Levels of IL-1β correlate with severity and duration of PTSD and PTSD can be prevented by bolus administration of hydrocortisone in acute sepsis, consistent with unrestrained inflammation being a risk factor for PTSD. The nervous and immune systems engage in crosstalk, governed by common receptors. The benefits of currently used psychiatric medication may arise from immune, as well as synaptic, modulation. The psychedelic drugs (3,4-Methylenedioxymethamphetamine (MDMA), psilocybin, and ketamine) have potent immunosuppressive and anti-inflammatory effects on the adaptive immune system, which may contribute to their reported benefit in PTSD. There may be distinct PTSD phenotypes induced by innate and adaptive cytokine signaling. CONCLUSION In order for an organism to survive, it must adapt to its environment. Cytokines signal danger to the brain and can induce epigenetic changes that result in a persistent defensive phenotype. PTSD may be the price individuals pay for the genomic flexibility that promotes adaptation and survival.
Collapse
Affiliation(s)
- Stephan Rudzki
- Canberra Sports Medicine, Deakin, Australian Capital Territory 2600, Australia
| |
Collapse
|
30
|
Tsetsenis T, Badyna JK, Li R, Dani JA. Activation of a Locus Coeruleus to Dorsal Hippocampus Noradrenergic Circuit Facilitates Associative Learning. Front Cell Neurosci 2022; 16:887679. [PMID: 35496910 PMCID: PMC9051520 DOI: 10.3389/fncel.2022.887679] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/25/2022] [Indexed: 01/22/2023] Open
Abstract
Processing of contextual information during a new episodic event is crucial for learning and memory. Neuromodulation in the hippocampus and prefrontal cortex plays an important role in the formation of associations between environmental cues and an aversive experience. Noradrenergic neurons in the locus coeruleus send dense projections to both regions, but their contribution to contextual associative learning has not been established. Here, we utilize selective optogenetic and pharmacological manipulations to control noradrenergic transmission in the hippocampus during the encoding of a contextual fear memory. We find that boosting noradrenergic terminal release in the dorsal CA1 enhances the acquisition of contextual associative learning and that this effect requires local activation of β-adrenenergic receptors. Moreover, we show that increasing norepinephrine release can ameliorate contextual fear learning impairments caused by dopaminergic dysregulation in the hippocampus. Our data suggest that increasing of hippocampal noradrenergic activity can have important implications in the treatment of cognitive disorders that involve problems in contextual processing.
Collapse
Affiliation(s)
- Theodoros Tsetsenis
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Julia K. Badyna
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rebecca Li
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - John A. Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
31
|
Wainstein G, Müller EJ, Taylor N, Munn B, Shine JM. The role of the locus coeruleus in shaping adaptive cortical melodies. Trends Cogn Sci 2022; 26:527-538. [DOI: 10.1016/j.tics.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
|
32
|
Examining the relationship between working memory consolidation and long-term consolidation. Psychon Bull Rev 2022; 29:1625-1648. [PMID: 35357669 DOI: 10.3758/s13423-022-02084-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 11/08/2022]
Abstract
An emerging area of research is focused on the relationship between working memory and long-term memory and the likely overlap between these processes. Of particular interest is how some information first maintained in working memory is retained for longer periods and eventually preserved in long-term memory. The process of stabilizing transient memory representations for lasting retention is referred to as consolidation in both the working memory and long-term memory literature, although these have historically been viewed as independent constructs. The present review aims to investigate the relationship between working memory consolidation and long-term memory consolidation, which both have rich, but distinct, histories. This review provides an overview of the proposed models and neural mechanisms of both types of consolidation, as well as clinical findings related to consolidation and potential approaches for the manipulation of consolidation. Finally, two hypotheses are proposed to explain the relationship between working memory consolidation and long-term memory consolidation.
Collapse
|
33
|
Borodovitsyna O, Tkaczynski JA, Corbett CM, Loweth JA, Chandler DJ. Age- and Sex-Dependent Changes in Locus Coeruleus Physiology and Anxiety-Like Behavior Following Acute Stressor Exposure. Front Behav Neurosci 2022; 16:808590. [PMID: 35283738 PMCID: PMC8914098 DOI: 10.3389/fnbeh.2022.808590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Adolescence is a critical period of development with increased sensitivity toward psychological stressors. Many psychiatric conditions emerge during adolescence and animal studies have shown that that acute stress has long-term effects on hypothalamic-pituitary-adrenal axis function and behavior. We recently demonstrated that acute stress produces long-term electrophysiological changes in locus coeruleus and long-lasting anxiety-like behavior in adolescent male rats. Based on prior reports of increased stress sensitivity during adolescence and increased sensitivity of female locus coeruleus toward corticotropin releasing factor, we hypothesized that the same acute stressor would cause different behavioral and physiological responses in adolescent female and adult male and female rats one week after stressor exposure. In this study, we assessed age and sex differences in how an acute psychological stressor affects corticosterone release, anxiety-like behavior, and locus coeruleus physiology at short- and long-term intervals. All groups of animals except adult female responded to stress with elevated corticosterone levels at the acute time point. One week after stressor exposure, adolescent females showed decreased firing of locus coeruleus neurons upon current injection and increased exploratory behavior compared to controls. The results were in direct contrast to changes observed in adolescent males, which showed increased anxiety-like behavior and increased spontaneous and induced firing in locus coeruleus neurons a week after stressor exposure. Adult males and females were both behaviorally and electrophysiologically resilient to the long-term effects of acute stress. Therefore, there may be a normal developmental trajectory for locus coeruleus neurons which promotes stress resilience in adults, but stressor exposure during adolescence perturbs their function. Furthermore, while locus coeruleus neurons are more sensitive to stressor exposure during adolescence, the effect varies between adolescent males and females. These findings suggest that endocrine, behavioral, and physiological responses to stress vary among animals of different age and sex, and therefore these variables should be taken into account when selecting models and designing experiments to investigate the effects of stress. These differences in animals may also allude to age and sex differences in the prevalence of various psychiatric illnesses within the human population.
Collapse
Affiliation(s)
- Olga Borodovitsyna
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - John A. Tkaczynski
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Claire M. Corbett
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Jessica A. Loweth
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
| | - Daniel J. Chandler
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, United States
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, United States
- *Correspondence: Daniel J. Chandler,
| |
Collapse
|
34
|
Dai ZH, Xu X, Chen WQ, Nie LN, Liu Y, Sui N, Liang J. The role of hippocampus in memory reactivation: an implication for a therapeutic target against opioid use disorder. CURRENT ADDICTION REPORTS 2022; 9:67-79. [PMID: 35223369 PMCID: PMC8857535 DOI: 10.1007/s40429-022-00407-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2022] [Indexed: 12/29/2022]
Abstract
Purpose of the review The abuse of opioids induces many terrible problems in human health and social stability. For opioid-dependent individuals, withdrawal memory can be reactivated by context, which is then associated with extremely unpleasant physical and emotional feelings during opioid withdrawal. The reactivation of withdrawal memory is considered one of the most important reasons for opioid relapse, and it also allows for memory modulation based on the reconsolidation phenomenon. However, studies exploring withdrawal memory modulation during the reconsolidation window are lacking. By summarizing the previous findings about the reactivation of negative emotional memories, we are going to suggest potential neural regions and systems for modulating opioid withdrawal memory. Recent findings Here, we first present the role of memory reactivation in its modification, discuss how the hippocampus participates in memory reactivation, and discuss the importance of noradrenergic signaling in the hippocampus for memory reactivation. Then, we review the engagement of other limbic regions receiving noradrenergic signaling in memory reactivation. We suggest that noradrenergic signaling targeting hippocampus neurons might play a potential role in strengthening the disruptive effect of withdrawal memory extinction by facilitating the degree of memory reactivation. Summary This review will contribute to a better understanding of the mechanisms underlying reactivation-dependent memory malleability and will provide new therapeutic avenues for treating opioid use disorders.
Collapse
Affiliation(s)
- Zhong-hua Dai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Xing Xu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Wei-qi Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Li-na Nie
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Liu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
35
|
Nicholas J, Daw ND, Shohamy D. Uncertainty alters the balance between incremental learning and episodic memory. eLife 2022; 11:81679. [PMID: 36458809 PMCID: PMC9810331 DOI: 10.7554/elife.81679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/01/2022] [Indexed: 12/04/2022] Open
Abstract
A key question in decision-making is how humans arbitrate between competing learning and memory systems to maximize reward. We address this question by probing the balance between the effects, on choice, of incremental trial-and-error learning versus episodic memories of individual events. Although a rich literature has studied incremental learning in isolation, the role of episodic memory in decision-making has only recently drawn focus, and little research disentangles their separate contributions. We hypothesized that the brain arbitrates rationally between these two systems, relying on each in circumstances to which it is most suited, as indicated by uncertainty. We tested this hypothesis by directly contrasting contributions of episodic and incremental influence to decisions, while manipulating the relative uncertainty of incremental learning using a well-established manipulation of reward volatility. Across two large, independent samples of young adults, participants traded these influences off rationally, depending more on episodic information when incremental summaries were more uncertain. These results support the proposal that the brain optimizes the balance between different forms of learning and memory according to their relative uncertainties and elucidate the circumstances under which episodic memory informs decisions.
Collapse
Affiliation(s)
- Jonathan Nicholas
- Department of Psychology, Columbia UniversityNew YorkUnited States,Mortimer B. Zuckerman Mind, Brain, Behavior Institute, Columbia UniversityNew YorkUnited States
| | - Nathaniel D Daw
- Department of Psychology, Princeton UniversityPrincetonUnited States,Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Daphna Shohamy
- Department of Psychology, Columbia UniversityNew YorkUnited States,Mortimer B. Zuckerman Mind, Brain, Behavior Institute, Columbia UniversityNew YorkUnited States,The Kavli Institute for Brain Science, Columbia UniversityNew YorkUnited States
| |
Collapse
|
36
|
Gao D, Wan J, Zou Y, Gong Y, Dong X, Xu Z, Tang J, Wei G, Zhang Q. Destructive Mechanism of Aβ 1-42 Protofibril by Norepinephrine revealed via Molecular Dynamics Simulations. Phys Chem Chem Phys 2022; 24:19827-19836. [DOI: 10.1039/d2cp01754g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amyloid-β (Aβ) fibrillary plaques represent the main hallmarks of Alzheimer’s disease (AD), in addition to tau neurofibrillary tangles. Disrupting early-formed Aβ protofibril is considered as one of the primary therapeutic...
Collapse
|
37
|
Cui Y, Zhang B, Zhang Z, Nie J, Liu H. Long-term repetitive exposure to excess iodine induces mitochondrial apoptosis, and alters monoamine neurotransmitters in hippocampus of rats of different genders. Toxicol Res (Camb) 2021; 10:975-982. [PMID: 34733482 DOI: 10.1093/toxres/tfab082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
The influence of excess iodine on human health has been paid more and more attention. Although numerous studies have reported that excess iodine may cause deleterious effects, the mental damage and its mechanism is yet to be identified. Using Sprague-Dawley rats exposed to excess iodine from pregnancy to 6 months post-delivery as in vivo model, this study explored the impacts of long-term repetitive excess iodine administration on the hippocampus of offspring rats, focusing on mitochondrial apoptosis pathway, with changes in monoamine neurotransmitters. The results showed that excess iodine could increase urinary iodine and brain organ coefficient in offspring of both genders, change the hippocampal cell structure, and damage the spatial learning and memory capacities. Poly ADP-ribose polymerase (PARP), P53, Cleaved Caspase-3, and cytochrome C proteins expression increased and Bcl2 protein expression decreased in hippocampus of excess iodine-treated offspring, indicating that excess iodine could activate the mitochondrial apoptosis pathway. Besides, excess iodine showed different effects on monoamine neurotransmitter in different gender. Collectively, our experimental data indicated that the learning and memory impairment induced by excess iodine may be mediated via mitochondrial apoptotic pathway. Long-term repetitive excess iodine exposure affected monoamine neurotransmitters in hippocampus of offspring rats.
Collapse
Affiliation(s)
- Yushan Cui
- Institute of Environment and Health, Tianjin Centers for Disease Control and Prevention, 6 Huayue Road, Hedong District, Tianjin 300011, P.R. China
| | - Bin Zhang
- Scientific Fitness and Health Promotion Research Center, China Institute of Sport Science, 11 Tiyuguan Road, Dongcheng District, Beijing 100061, P.R. China
| | - Zushan Zhang
- School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Junyan Nie
- School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Hongliang Liu
- School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| |
Collapse
|
38
|
Ali AA, Khalil MG, Abd El-Latif DM, Okda T, Abdelaziz AI, Abu-Elfotuh K, Kamal MM, Wahid A. The influence of vinpocetine alone or in combination with Epigallocatechin-3-gallate, Coenzyme COQ10, Vitamin E and Selenium as a potential neuroprotective combination against aluminium-induced Alzheimer's disease in Wistar Albino Rats. Arch Gerontol Geriatr 2021; 98:104557. [PMID: 34706318 DOI: 10.1016/j.archger.2021.104557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is one of such diseases that represent the most prominent cause of dementia in elderly people. To explore the possible neuroprotective effect as well as mechanism of action of Vinpocetine either alone or in combination with EGCG, CoQ10, or VE & Se in ameliorating aluminum chloride-induced AD in rats. Rats were received AlCl3 (70 mg/kg) intraperitoneal daily dose for 30 days along with EGCG (10 mg/kg, I.P), CoQ10 (200 mg/kg, P.O), VE (100 mg/kg, P.O) & Se (1 mg/kg, P.O) as well as Vinpocetine (20 mg/kg, P.O) either alone or in combination. Results revealed that the combination of Vinpocetine with EGCG showed the best neuroprotection. This protection in the brain was indicated by the significant decrease in Aβ and ACHE. The same pattern of results were shown in the levels of monoamines and BDNF. In addition, the combination of Vinpocetine with EGCG showed more pronounced anti-inflammatory (TNF-α, IL-1β) and antioxidant (MDA, SOD, TAC) effects in comparison to other combinations. These results were confirmed using histopathological examinations as well as DNA fragmentation assays. Vinpocetine with EGCG showed pronounced protection on neurons against AD induced by AlCl3 in rats.
Collapse
Affiliation(s)
- Azza A Ali
- Department of Pharmacology and Toxicology; Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mona G Khalil
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Doaa M Abd El-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Tarek Okda
- Department of Biochemistry, Faculty of pharmacy, Damanhour University, Egypt
| | - Aya I Abdelaziz
- Medical Research Center, Faculty of pharmacy, Heliopolis University, Egypt
| | - Karema Abu-Elfotuh
- Department of Pharmacology and Toxicology; Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mona M Kamal
- Department of Pharmacology and Toxicology; Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of pharmacy, Alexandria University, Egypt.
| |
Collapse
|
39
|
Martinho R, Correia G, Seixas R, Oliveira A, Silva S, Serrão P, Fernandes-Lopes C, Costa C, Moreira-Rodrigues M. Treatment With Nepicastat Decreases Contextual Traumatic Memories Persistence in Post-traumatic Stress Disorder. Front Mol Neurosci 2021; 14:745219. [PMID: 34630037 PMCID: PMC8498196 DOI: 10.3389/fnmol.2021.745219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a common anxiety mental disorder and can be manifested after exposure to a real or perceived life-threatening event. Increased noradrenaline and adrenaline in plasma and urine have been documented in PTSD. Dopamine-β-hydroxylase (DBH) catalyzes the conversion of dopamine to noradrenaline and consequently, DBH inhibition reduces catecholamines. Our aim was to evaluate if nepicastat treatment decreases PTSD signs in an animal model. Wild-type (129x1/SvJ) female mice were submitted to PTSD induction protocol. DBH-inhibitor nepicastat (30 mg/kg) or vehicle (0.2% HPMC) were administered once daily since day 0 until day 7 or 12. The percentage of freezing was calculated on days 0, 1, 2, and 7, and behavioral tests were performed. Quantification of nepicastat in plasma and DBH activity in the adrenal gland was evaluated. Catecholamines were quantified by HPLC with electrochemical detection. mRNA expression of Npas4 and Bdnf in hippocampus was evaluated by qPCR.Mice in the PTSD-group and treated with nepicastat showed a decrease in freezing, and an increase in the time spent and entries in open arms in elevated plus maze test. In mice treated with nepicastat, adrenal gland DBH activity was decreased, and catecholamines were also decreased in plasma and tissues. On day 7, in mice treated with nepicastat, there was an increase of Npas4 and Bdnf mRNA expression in the hippocampus.In conclusion, DBH inhibitor nepicastat has an effect consistent with a decrease in the persistence of traumatic memories and anxiety-like behavior in this PTSD mice model. The disruption of traumatic memories through interference with the formation, consolidation, retrieval, and/or expression processes may be important to decrease PTSD symptoms and signs. The increase in Npas4 and Bdnf mRNA expression in the hippocampus may be important to develop a weaker traumatic contextual memory after nepicastat treatment.
Collapse
Affiliation(s)
- Raquel Martinho
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Gabriela Correia
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Rafaela Seixas
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Ana Oliveira
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Soraia Silva
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| | - Paula Serrão
- Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal.,Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| | | | | | - Mónica Moreira-Rodrigues
- Laboratory of General Physiology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS/UP), Porto, Portugal.,Center for Drug Discovery and Innovative Medicines, University of Porto (MedInUP), Porto, Portugal
| |
Collapse
|
40
|
Webler RD, Berg H, Fhong K, Tuominen L, Holt DJ, Morey RA, Lange I, Burton PC, Fullana MA, Radua J, Lissek S. The neurobiology of human fear generalization: meta-analysis and working neural model. Neurosci Biobehav Rev 2021; 128:421-436. [PMID: 34242718 DOI: 10.1016/j.neubiorev.2021.06.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/04/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Fear generalization to stimuli resembling a conditioned danger-cue (CS+) is a fundamental dynamic of classical fear-conditioning. Despite the ubiquity of fear generalization in human experience and its known pathogenic contribution to clinical anxiety, neural investigations of human generalization have only recently begun. The present work provides the first meta-analysis of this growing literature to delineate brain substrates of conditioned fear-generalization and formulate a working neural model. Included studies (K = 6, N = 176) reported whole-brain fMRI results and applied generalization-gradient methodology to identify brain activations that gradually strengthen (positive generalization) or weaken (negative generalization) as presented stimuli increase in CS+ resemblance. Positive generalization was instantiated in cingulo-opercular, frontoparietal, striatal-thalamic, and midbrain regions (locus coeruleus, periaqueductal grey, ventral tegmental area), while negative generalization was implemented in default-mode network nodes (ventromedial prefrontal cortex, hippocampus, middle temporal gyrus, angular gyrus) and amygdala. Findings are integrated within an updated neural account of generalization centering on the hippocampus, its modulation by locus coeruleus and basolateral amygdala, and the excitation of threat- or safety-related loci by the hippocampus.
Collapse
Affiliation(s)
- Ryan D Webler
- Department of Psychology, University of Minnesota, 75 E River Rd, Minneapolis, MN, 55455, USA
| | - Hannah Berg
- Department of Psychology, University of Minnesota, 75 E River Rd, Minneapolis, MN, 55455, USA
| | - Kimberly Fhong
- Department of Psychology, University of Minnesota, 75 E River Rd, Minneapolis, MN, 55455, USA
| | - Lauri Tuominen
- The Royal's Institute of Mental Health Research, University of Ottawa, 1145 Carling Avenue, Ottawa, Ontario, K1Z 7K4, Canada
| | - Daphne J Holt
- Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Rajendra A Morey
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, Duke University Medical Center, Durham, NC, 27710, USA; VA Mid-Atlantic Mental Illness Research Education and Clinical Center, 508 Fulton Street, Durham VAMC, Durham, VA Medical Center, Durham, NC, 27705, USA; Duke-UNC Brain Imaging and Analysis Center, Duke University, 40 Duke Medicine Circle, Durham, NC, USA
| | - Iris Lange
- Department of Psychiatry and Psychology, School for Mental Health and Neuroscience, EURON, Maastricht University Medical Centre, Duboisdomein 30, 6229 GT, Maastricht, the Netherlands
| | - Philip C Burton
- Department of Psychology, University of Minnesota, 75 E River Rd, Minneapolis, MN, 55455, USA
| | - Miquel Angel Fullana
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBERSAM, Campus Casanova, Casanova, 143, 08036, Barcelona, Spain; Adult Psychiatry and Psychology Department, Institute of Neurosciences, Hospital Clínic, Casanovas 143, 08036, Barcelona, Spain
| | - Joaquim Radua
- Adult Psychiatry and Psychology Department, Institute of Neurosciences, Hospital Clínic, Casanovas 143, 08036, Barcelona, Spain; Early Psychosis: Interventions and Clinical-detection (EPIC) Laboratory, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, SE5 8AF, UK; Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Shmuel Lissek
- Department of Psychology, University of Minnesota, 75 E River Rd, Minneapolis, MN, 55455, USA.
| |
Collapse
|
41
|
Tillage RP, Foster SL, Lustberg D, Liles LC, McCann KE, Weinshenker D. Co-released norepinephrine and galanin act on different timescales to promote stress-induced anxiety-like behavior. Neuropsychopharmacology 2021; 46:1535-1543. [PMID: 33911187 PMCID: PMC8208976 DOI: 10.1038/s41386-021-01011-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/02/2023]
Abstract
Both the noradrenergic and galaninergic systems have been implicated in stress-related neuropsychiatric disorders, and these two neuromodulators are co-released from the stress-responsive locus coeruleus (LC); however, the individual contributions of LC-derived norepinephrine (NE) and galanin to behavioral stress responses are unclear. Here we aimed to disentangle the functional roles of co-released NE and galanin in stress-induced behavior. We used foot shock, optogenetics, and behavioral pharmacology in wild-type (WT) mice and mice lacking either NE (Dbh-/-) or galanin (GalcKO-Dbh) specifically in noradrenergic neurons to isolate the roles of these co-transmitters in regulating anxiety-like behavior in the elevated zero maze (EZM) either immediately or 24 h following stress. Foot shock and optogenetic LC stimulation produced immediate anxiety-like behavior in WT mice, and the effects of foot shock persisted for 24 h. NE-deficient mice were resistant to the anxiogenic effects of acute stress and optogenetic LC stimulation, while mice lacking noradrenergic-derived galanin displayed typical increases in anxiety-like behavior. However, when tested 24 h after foot shock, both Dbh-/- and GalcKO-Dbh mice lacked normal expression of anxiety-like behavior. Pharmacological rescue of NE, but not galanin, in knockout mice during EZM testing was anxiogenic. In contrast, restoring galanin, but not NE, signaling during foot shock normalized stress-induced anxiety-like behavior 24 h later. These results indicate that NE and noradrenergic-derived galanin play complementary, but distinguishable roles in behavioral responses to stress. NE is required for the expression of acute stress-induced anxiety, while noradrenergic-derived galanin mediates the development of more persistent responses following a stressor.
Collapse
Affiliation(s)
- Rachel P. Tillage
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - Stephanie L. Foster
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - Daniel Lustberg
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - L. Cameron Liles
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | - Katharine E. McCann
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University, Atlanta, GA USA
| | | |
Collapse
|
42
|
Campese VD. The lesser evil: Pavlovian-instrumental transfer & aversive motivation. Behav Brain Res 2021; 412:113431. [PMID: 34175357 DOI: 10.1016/j.bbr.2021.113431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 06/13/2021] [Accepted: 06/22/2021] [Indexed: 01/01/2023]
Abstract
While our understanding of appetitive motivation includes accounts of rich cognitive phenomena, such as choice, sensory-specificity and outcome valuation, the same is not true in aversive processes. A highly sophisticated picture has emerged of Pavlovian fear conditioning and extinction, but progress in aversive motivation has been somewhat limited to these fundamental behaviors. Many differences between appetitive and aversive stimuli permit different kinds of analyses; a widely used procedure in appetitive studies that can expand the scope of aversive motivation is Pavlovian-instrumental transfer (PIT). Recently, this motivational transfer effect has been used to examine issues pertaining to sensory-specificity and the nature of defensive control in avoidance learning. Given enduring controversies and unresolved criticisms surrounding avoidance research, PIT offers a valuable, well-controlled procedure with which to similarly probe this form of motivation. Furthermore, while avoidance itself can be criticized as artificial, PIT can be an effective model for how skills learned through avoidance can be practically applied to encounters with threatening or fearful stimuli and stress. Despite sensory-related challenges presented by the limited aversive unconditioned stimuli typically used in research, transfer testing can nevertheless provide valuable information on the psychological nature of this historically controversial phenomenon.
Collapse
|
43
|
Careaga MBL, Girardi CEN, Suchecki D. Propranolol failed to prevent severe stress-induced long-term behavioral changes in male rats. Prog Neuropsychopharmacol Biol Psychiatry 2021; 108:110079. [PMID: 32827609 DOI: 10.1016/j.pnpbp.2020.110079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/24/2020] [Accepted: 08/16/2020] [Indexed: 10/23/2022]
Abstract
Memories of adverse events can be maladaptive when they lead to exaggerated fear, as observed in post-traumatic stress disorder (PTSD). Fear conditioning and fear sensitization are learning processes thought to play a role in fear-related disorders, and only few animal studies have evaluated the relationship between the associative and non-associative fear memory components on the development and maintenance of PTSD-like behavioral changes. Here we assessed the effects of a single dose of propranolol (10 mg/kg) or saline after fear memory retrieval on the long-term behavioral responses induced by severe stress in male rats. Animals were submitted to contextual fear conditioning (delayed shock group) or not (non-shock group) and underwent fear memory retrieval followed by propranolol or saline administration two weeks later. Rats were then evaluated in different behavioral tests to assess the expression of the conditioned fear response, anxiety-like and exploratory behaviors, and fear response after the presentation of unknown acoustic stimulus. Post-retrieval propranolol did not disrupt the subsequent expression of neither conditioned fear response nor the exploratory deficit and fear sensitization response, indicating that propranolol failed to mitigate long-term behavioral changes induced by severe stress in rats.
Collapse
Affiliation(s)
| | - Carlos Eduardo Neves Girardi
- Departamento de Psicobiologia, Universidade Federal de São Paulo/Escola Paulista de Medicina - UNIFESP/EPM, São Paulo, Brazil
| | - Deborah Suchecki
- Departamento de Psicobiologia, Universidade Federal de São Paulo/Escola Paulista de Medicina - UNIFESP/EPM, São Paulo, Brazil.
| |
Collapse
|
44
|
O'Neill E, Griffin ÉW, O'Sullivan R, Murray C, Ryan L, Yssel J, Harkin A, Cunningham C. Acute neuroinflammation, sickness behavior and working memory responses to acute systemic LPS challenge following noradrenergic lesion in mice. Brain Behav Immun 2021; 94:357-368. [PMID: 33307172 DOI: 10.1016/j.bbi.2020.12.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Locus coeruleus (LC)-derived noradrenaline is important in cognition and decreases with age, but the impact of prior noradrenaline deficiency on vulnerability to inflammation-induced acute cognitive dysfunction is unclear. Here we assessed whether noradrenergic depletion, in female mice, impacted upon inflammation, locomotor activity and working memory directly after acute systemic immune challenge with bacterial lipopolysaccharide (LPS), a paradigm we have previously used to capture delirium-like acute cognitive deficits. Mice received 2 doses of the LC-selective noradrenergic toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4; 50 mg/kg i.p.) and were challenged, 2 weeks later, with LPS (100 μg/kg i.p.). DSP-4 dramatically reduced noradrenaline concentrations and tyrosine hydroxylase-positive afferents in the frontal cortex and hippocampus. This did not significantly alter numbers of Pu.1-positive microglia, Iba1-positive microglial morphology or mRNA expression of microglia-associated gene transcripts (Tyrobp, Sall1, Cd68, Sra2, Clec7a) in the hippocampus or frontal cortex and produced modest reductions in Cx3cr1 and P2ry12. LPS induced blood and brain cytokine levels, cFOS activation and locomotor responses that were highly similar in DSP-4- and vehicle-treated mice, although LPS-induced plasma TNF-α was significantly reduced in those treated with DSP-4. Importantly, prior noradrenergic depletion did not predispose to LPS-induced T-maze working memory deficits. The data demonstrate that significant depletion of noradrenaline in the hippocampus and frontal cortex does not prompt acutely exaggerated neuroinflammation or leave the brain vulnerable to acute, transient working memory deficits upon low dose LPS challenge. These findings have implications for our understanding of the impact of systemic inflammation on the aging and vulnerable brain during septic encephalopathy and delirium.
Collapse
Affiliation(s)
- Eoin O'Neill
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Éadaoin W Griffin
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Ruairi O'Sullivan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Carol Murray
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Lucy Ryan
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Justin Yssel
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew Harkin
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
45
|
Giri S, Ranjan A, Kumar A, Amar M, Mallick BN. Rapid eye movement sleep deprivation impairs neuronal plasticity and reduces hippocampal neuronal arborization in male albino rats: Noradrenaline is involved in the process. J Neurosci Res 2021; 99:1815-1834. [PMID: 33819353 DOI: 10.1002/jnr.24838] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/05/2021] [Accepted: 03/13/2021] [Indexed: 12/22/2022]
Abstract
Rapid eye movement sleep (REMS) favors brain development and memory, while it is decreased in neurodegenerative diseases. REMS deprivation (REMSD) affects several physiological processes including memory consolidation; however, its detailed mechanism(s) of action was unknown. REMS reduces, while REMSD elevates noradrenaline (NA) level in the brain; the latter induces several deficiencies and disorders, including changes in neuronal cytomorphology and apoptosis. Therefore, we proposed that REMS- and REMSD-associated modulation of NA level might affect neuronal plasticity and affect brain functions. Male albino rats were REMS deprived by flower-pot method for 6 days, and its effects were compared with home cage and large platform controls as well as post-REMSD recovered and REMS-deprived prazosin (α1-adrenoceptor antagonist)-treated rats. We observed that REMSD reduced CA1 and CA3 neuronal dendritic length, branching, arborization, and spine density, while length of active zone and expressions of pre- as well as post-synaptic proteins were increased as compared to controls; interestingly, prazosin prevented most of the effects in vivo. Studies on primary culture of neurons from chick embryo brain confirmed that NA at lower concentration(s) induced neuronal branching and arborization, while higher doses were destructive. The findings support our contention that REMSD adversely affects neuronal plasticity, branching, and synaptic scaffold, which explain the underlying cytoarchitectural basis of REMSD-associated patho-physio-behavioral changes. Consolidation of findings of this study along with that of our previous reports suggest that the neuronal disintegration could be due to either withdrawal of direct protective and proliferative role of low dose of NA or indirect effect of high dose of NA or both.
Collapse
Affiliation(s)
- Shatrunjai Giri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Amit Ranjan
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Awanish Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Megha Amar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
46
|
Dela Peña I, Shen G, Shi WX. Droxidopa alters dopamine neuron and prefrontal cortex activity and improves attention-deficit/hyperactivity disorder-like behaviors in rats. Eur J Pharmacol 2021; 892:173826. [PMID: 33347825 DOI: 10.1016/j.ejphar.2020.173826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
Finding alternative treatments for attention-deficit/hyperactivity disorder (ADHD) is crucial given the safety and efficacy problems of current ADHD medications. Droxidopa, also known as L-threo-dihydroxyphenylserine (L-DOPS), is a norepinephrine prodrug that enhances brain norepinephrine and dopamine levels. In this study, we used electrophysiological tests to examine effects of L-DOPS on the prefrontal cortex (PFC) and dopamine neurons in the ventral tegmental area. We also conducted behavioral tests to assess L-DOPS' effects on ADHD-like behaviors in rats. In chloral hydrate-anesthetized rats, PFC local field potentials oscillated between the active, depolarized UP state and the hyperpolarized DOWN state. Mimicking the effect of d-amphetamine, L-DOPS, given after the peripheral amino acid decarboxylase inhibitor, benserazide (BZ), increased the amount of time the PFC spent in the UP state, indicating an excitatory effect of L-DOPS on PFC neurons. Like d-amphetamine, L-DOPS also inhibited dopamine neurons, an effect significantly reversed by the D2-like receptor antagonist raclopride. In the behavioral tests, BZ + L-DOPS improved hyperactivity, inattention and impulsive action of the adolescent spontaneously hypertensive rat (SHR/NCrl), well-validated animal model of the combined type of ADHD. BZ + L-DOPS also reduced impulsive choice and impulsive action of Wistar rats, but did not ameliorate the inattentiveness of Wistar Kyoto rats (WKY/NCrl), proposed model of the ADHD-predominantly inattentive type. In conclusion, L-DOPS produced effects on the PFC and dopamine neurons characteristic of drugs used to treat ADHD. BZ + L-DOPS ameliorated ADHD-like behaviors in rats suggesting its potential as an alternative ADHD treatment.
Collapse
Affiliation(s)
- Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, CA, 92350, USA.
| | - Guofang Shen
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, CA, 92350, USA
| | - Wei-Xing Shi
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, CA, 92350, USA; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| |
Collapse
|
47
|
Kausche FM, Zerbes G, Kampermann L, Müller JC, Wiedemann K, Büchel C, Schwabe L. Noradrenergic stimulation increases fear memory expression. Eur Neuropsychopharmacol 2021; 43:71-81. [PMID: 33358539 DOI: 10.1016/j.euroneuro.2020.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/08/2020] [Accepted: 11/30/2020] [Indexed: 11/16/2022]
Abstract
Fear responses are typically not limited to the actual threatening stimulus but generalize to other stimuli resembling the threatening stimulus. Although this fear generalization is generally adaptive, fear overgeneralization is maladaptive and assumed to contribute to anxiety disorders. Despite the clinical relevance of fear (over)generalization, how the extent of fear generalization is modulated remains not well understood. Based on the known effects of stress on learning and memory, we tested here the impact of major stress mediators, glucocorticoids and noradrenergic arousal, on fear generalization. In a laboratory-based, placebo-controlled, double-blind, between-subject design, 125 healthy participants first underwent a fear conditioning procedure. About 24 h later, participants received orally either a placebo, hydrocortisone, the α2-adrenoceptor antagonist yohimbine, leading to increased noradrenergic stimulation, or both drugs before a test of fear generalization. Skin conductance responses as well as explicit rating data revealed that yohimbine intake led to enhanced fear memory expression, i.e. an enhanced responding to the CS+ but not to stimuli resembling the CS+. Moreover, neither enhanced safety learning nor a mere enhancement of perceptual discrimination ability could explain this result. In contrast to yohimbine, hydrocortisone had no significant effect on fear memory. These findings suggest that noradrenergic arousal strengthens fear memory expression and have important implications for mental disorders in which the overgeneralization of conditioned fear is prominent.
Collapse
Affiliation(s)
| | - Gundula Zerbes
- Department of Cognitive Psychology, Universität Hamburg, 20146 Hamburg, Germany
| | - Lea Kampermann
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jana Christina Müller
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Klaus Wiedemann
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Büchel
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lars Schwabe
- Department of Cognitive Psychology, Universität Hamburg, 20146 Hamburg, Germany.
| |
Collapse
|
48
|
Roy R, Paul S. Theoretical Investigation of the Inhibitory Mechanism of Norepinephrine on hIAPP Amyloid Aggregation and the Destabilization of Protofibrils. J Phys Chem B 2020; 124:10913-10929. [DOI: 10.1021/acs.jpcb.0c07830] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| |
Collapse
|
49
|
Bacon TJ, Pickering AE, Mellor JR. Noradrenaline Release from Locus Coeruleus Terminals in the Hippocampus Enhances Excitation-Spike Coupling in CA1 Pyramidal Neurons Via β-Adrenoceptors. Cereb Cortex 2020; 30:6135-6151. [PMID: 32607551 PMCID: PMC7609922 DOI: 10.1093/cercor/bhaa159] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
Release of the neuromodulator noradrenaline signals salience during wakefulness, flagging novel or important experiences to reconfigure information processing and memory representations in the hippocampus. Noradrenaline is therefore expected to enhance hippocampal responses to synaptic input; however, noradrenergic agonists have been found to have mixed and sometimes contradictory effects on Schaffer collateral synapses and the resulting CA1 output. Here, we examine the effects of endogenous, optogenetically driven noradrenaline release on synaptic transmission and spike output in mouse hippocampal CA1 pyramidal neurons. We show that endogenous noradrenaline release enhances the probability of CA1 pyramidal neuron spiking without altering feedforward excitatory or inhibitory synaptic inputs in the Schaffer collateral pathway. β-adrenoceptors mediate this enhancement of excitation-spike coupling by reducing the charge required to initiate action potentials, consistent with noradrenergic modulation of voltage-gated potassium channels. Furthermore, we find the likely effective concentration of endogenously released noradrenaline is sub-micromolar. Surprisingly, although comparable concentrations of exogenous noradrenaline cause robust depression of slow afterhyperpolarization currents, endogenous release of noradrenaline does not, indicating that endogenous noradrenaline release is targeted to specific cellular locations. These findings provide a mechanism by which targeted endogenous release of noradrenaline can enhance information transfer in the hippocampus in response to salient events.
Collapse
Affiliation(s)
- Travis J Bacon
- Centre for Synaptic Plasticity, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Anthony E Pickering
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
- Bristol Anaesthesia, Pain & Critical Care Sciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | - Jack R Mellor
- Centre for Synaptic Plasticity, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
50
|
Lustberg D, Tillage RP, Bai Y, Pruitt M, Liles LC, Weinshenker D. Noradrenergic circuits in the forebrain control affective responses to novelty. Psychopharmacology (Berl) 2020; 237:3337-3355. [PMID: 32821984 PMCID: PMC7572912 DOI: 10.1007/s00213-020-05615-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023]
Abstract
RATIONALE In rodents, exposure to novel environments elicits initial anxiety-like behavior (neophobia) followed by intense exploration (neophilia) that gradually subsides as the environment becomes familiar. Thus, innate novelty-induced behaviors are useful indices of anxiety and motivation in animal models of psychiatric disease. Noradrenergic neurons are activated by novelty and implicated in exploratory and anxiety-like responses, but the role of norepinephrine (NE) in neophobia has not been clearly delineated. OBJECTIVE We sought to define the role of central NE transmission in neophilic and neophobic behaviors. METHODS We assessed dopamine β-hydroxylase knockout (Dbh -/-) mice lacking NE and their NE-competent (Dbh +/-) littermate controls in neophilic (novelty-induced locomotion; NIL) and neophobic (novelty-suppressed feeding; NSF) behavioral tests with subsequent quantification of brain-wide c-fos induction. We complimented the gene knockout approach with pharmacological interventions. RESULTS Dbh -/- mice exhibited blunted locomotor responses in the NIL task and completely lacked neophobia in the NSF test. Neophobia was rescued in Dbh -/- mice by acute pharmacological restoration of central NE with the synthetic precursor L-3,4-dihydroxyphenylserine (DOPS), and attenuated in control mice by the inhibitory α2-adrenergic autoreceptor agonist guanfacine. Following either NSF or NIL, Dbh -/- mice demonstrated reduced c-fos in the anterior cingulate cortex, medial septum, ventral hippocampus, bed nucleus of the stria terminalis, and basolateral amygdala. CONCLUSION These findings indicate that central NE signaling is required for the expression of both neophilic and neophobic behaviors. Further, we describe a putative noradrenergic novelty network as a potential therapeutic target for treating anxiety and substance abuse disorders.
Collapse
Affiliation(s)
- Daniel Lustberg
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Rachel P Tillage
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Yu Bai
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Molly Pruitt
- University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - L Cameron Liles
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|