101
|
Mäki-Marttunen V. Pupil-based States of Brain Integration across Cognitive States. Neuroscience 2021; 471:61-71. [PMID: 34303781 DOI: 10.1016/j.neuroscience.2021.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 06/14/2021] [Accepted: 07/15/2021] [Indexed: 01/21/2023]
Abstract
Arousal is a potent mechanism that provides the brain with functional flexibility and adaptability to external conditions. Within the wake state, arousal levels driven by activity in the neuromodulatory systems are related to specific signatures of neural activation and brain synchrony. However, direct evidence is still lacking on the varying effects of arousal on macroscopic brain characteristics and across a variety of cognitive states in humans. Using a concurrent fMRI-pupillometry approach, we used pupil size as a proxy for arousal and obtained patterns of brain integration associated with increasing arousal levels. We carried out this analysis on resting-state data and data from two attentional tasks implicating different cognitive processes. We found that an increasing level of arousal was related to a state of increased brain integration. This effect was prominent in the salience, visual and default-mode networks in all conditions, while other regions showed task-specificity. Increased integration in the salience network was also related to faster pupil dilation in the two attentional tasks. Furthermore, task performance was related to arousal level, with lower accuracy at higher level of arousal. Taken together, our study provides evidence in humans for pupil size as an index of brain network state, and supports the role of arousal as a switch that drives brain coordination in specific brain regions according to the cognitive state.
Collapse
Affiliation(s)
- Verónica Mäki-Marttunen
- Department of Psychology, University of Oslo, Postboks 1094, Blindern, 0317 Oslo, Norway; Cognitive Psychology Unit, Faculty of Social Sciences, Leiden University, Pieter de la Court, Wassenaarseweg 52, 2333 AK Leiden, Netherlands.
| |
Collapse
|
102
|
The association of WTELS as a master motivator with higher executive functioning and better mental health. CURRENT PSYCHOLOGY 2021; 42:7309-7320. [PMID: 34276169 PMCID: PMC8272615 DOI: 10.1007/s12144-021-02078-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2021] [Indexed: 12/12/2022]
Abstract
The goal is to test the validity of the “Will to exist-live and survive (WTELS) as a master motivator that activates executive functions. A sample of 262 adults administered different measures that included WTELS and executive functions. We conducted hierarchical regressions with working memory deficits (WMD) and inhibition deficits (ID) as dependent variables. We entered in the last steps resilience and WTELS as independent variables. We conducted path analysis with WTELS as independent variables and WMD and ID as outcome variables and resilience and social support as mediating variables. WTELS accounted for the high effect size for lower working memory deficits and medium effect size for lower inhibition deficits. In path analysis, the effects of WTELS on decreased WMD were direct, while its effects on the ID were indirect. PROCESS analysis indicated that WTELS was directly associated with lower depression, anxiety, PTSD, and COVID-19 traumatic stress, and its indirect effects were mediated by lower executive function deficits (Kira et al., Psych 12:992-1024 2021c, Kira et al., in press). The path model discussed was generally superior to the alternative models and was strictly invariant across genders (male/ female).
Collapse
|
103
|
Samkaria A, Punjabi K, Sharma S, Joon S, Sandal K, Dasgupta T, Sharma P, Mandal PK. Brain Stress Mapping in COVID-19 Survivors Using MR Spectroscopy: New Avenue of Mental Health Status Monitoring$. J Alzheimers Dis 2021; 83:523-530. [PMID: 34250939 DOI: 10.3233/jad-210287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronavirus (COVID-19) has emerged as a human catastrophe worldwide, and it has impacted human life more detrimentally than the combined effect of World Wars I and II. Various research studies reported that the disease is not confined to the respiratory system but also leads to neurological and neuropsychiatric disorders suggesting that the virus is potent to affect the central nervous system (CNS). Moreover, the damage to CNS may continue to rise even after the COVID-19 infection subsides which may further induce a long-term impact on the brain, resulting in cognitive impairment. Neuroimaging techniques is the ideal platform to detect and quantify pathological manifestations in the brain of COVID-19 survivors. In this context, a scheme based on structural, spectroscopic, and behavioral studies could be executed to monitor the gradual changes in the brain non-invasively due to COVID-19 which may further help in quantifying the impact of COVID-19 on the mental health of the survivors. Extensive research is required in this direction for identifying the mechanism and implications of COVID-19 in the brain. Cohort studies are urgently required for monitoring the effects of this pandemic on individuals of various subtypes longitudinally.
Collapse
Affiliation(s)
- Avantika Samkaria
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon, India
| | - Khushboo Punjabi
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon, India
| | - Shallu Sharma
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon, India
| | - Shallu Joon
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon, India
| | - Kanika Sandal
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon, India
| | | | - Pooja Sharma
- Medanta Institute of Education and Research, Medicity, Gurgaon, India
| | - Pravat K Mandal
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon, India.,Florey Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Melbourne, Australia
| |
Collapse
|
104
|
Hou Y, Chen M, Wang C, Liu L, Mao H, Qu X, Shen X, Yu B, Liu S. Electroacupuncture Attenuates Anxiety-Like Behaviors in a Rat Model of Post-traumatic Stress Disorder: The Role of the Ventromedial Prefrontal Cortex. Front Neurosci 2021; 15:690159. [PMID: 34248490 PMCID: PMC8264195 DOI: 10.3389/fnins.2021.690159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Electroacupuncture (EA) is a promising clinical approach to treating posttraumatic stress disorder (PTSD), yet the mechanisms whereby EA can alleviate anxiety and other PTSD symptoms have yet to be clarified. In the present report, rats underwent EA for 14 consecutive days following modified single prolonged stress (MSPS) exposure. These animals were then evaluated in open field and elevated plus maze tests (OFT and EPM), while Fos immunohistochemical staining was performed to assess ventromedial prefrontal cortex (vmPFC) functional activation. In addition, an extracellular recording and stimulation system was used to analyze vmPFC inputs into the ventral tegmental area (VTA) in these rats. Temporary vmPFC inactivation was further performed to assess whether this was sufficient to reverse the anxiolytic effects of EA. Overall, rats that underwent EA treatment spent more time in the central region (OFT) and the open arm (EPM) relative to MSPS model animals (P < 0.05). These MSPS model animals also exhibited significantly fewer activated Fos-positive nuclei in the vmPFC following behavioral testing, while EA was associated with a significant relative increase in c-Fos expression in this region. The transient inactivation of the vmPFC was sufficient to reverse the effects of EA treatment on anxiety-like behaviors in MSPS model rats. MSPS and SEA rats exhibiting no differences in bursting activity between baseline and vmPFC stimulation, whereas bursting activity rose relative to baseline upon ventral mPFC stimulation in EA treated and control rats. Together, these findings indicate that the vmPFC and its inputs into the VTA are functionally linked to the anxiolytic activity of EA, implicating this pathway in the EA-mediated treatment of PTSD.
Collapse
Affiliation(s)
- Yuchao Hou
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meiyu Chen
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Can Wang
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lumin Liu
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huijuan Mao
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyi Qu
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xueyong Shen
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bo Yu
- Department of Human Anatomy, School of Basic Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Department of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
105
|
Delayed motor learning in a 16p11.2 deletion mouse model of autism is rescued by locus coeruleus activation. Nat Neurosci 2021; 24:646-657. [PMID: 33753944 DOI: 10.1038/s41593-021-00815-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/05/2021] [Indexed: 01/30/2023]
Abstract
Children with autism spectrum disorder often exhibit delays in achieving motor developmental milestones such as crawling, walking and speech articulation. However, little is known about the neural mechanisms underlying motor-related deficits. Here, we reveal that mice with a syntenic deletion of the chromosome 16p11.2, a common copy number variation associated with autism spectrum disorder, also exhibit delayed motor learning without showing gross motor deficits. Using in vivo two-photon imaging in awake mice, we find that layer 2/3 excitatory neurons in the motor cortex of adult male 16p11.2-deletion mice show abnormally high activity during the initial phase of learning, and the process of learning-induced spine reorganization is prolonged. Pharmacogenetic activation of locus coeruleus noradrenergic neurons was sufficient to rescue the circuit deficits and the delayed motor learning in these mice. Our results unveil an unanticipated role of noradrenergic neuromodulation in improving the delayed motor learning in 16p11.2-deletion male mice.
Collapse
|
106
|
Louth EL, Jørgensen RL, Korshoej AR, Sørensen JCH, Capogna M. Dopaminergic Neuromodulation of Spike Timing Dependent Plasticity in Mature Adult Rodent and Human Cortical Neurons. Front Cell Neurosci 2021; 15:668980. [PMID: 33967700 PMCID: PMC8102156 DOI: 10.3389/fncel.2021.668980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/29/2021] [Indexed: 11/29/2022] Open
Abstract
Synapses in the cerebral cortex constantly change and this dynamic property regulated by the action of neuromodulators such as dopamine (DA), is essential for reward learning and memory. DA modulates spike-timing-dependent plasticity (STDP), a cellular model of learning and memory, in juvenile rodent cortical neurons. However, it is unknown whether this neuromodulation also occurs at excitatory synapses of cortical neurons in mature adult mice or in humans. Cortical layer V pyramidal neurons were recorded with whole cell patch clamp electrophysiology and an extracellular stimulating electrode was used to induce STDP. DA was either bath-applied or optogenetically released in slices from mice. Classical STDP induction protocols triggered non-hebbian excitatory synaptic depression in the mouse or no plasticity at human cortical synapses. DA reverted long term synaptic depression to baseline in mouse via dopamine 2 type receptors or elicited long term synaptic potentiation in human cortical synapses. Furthermore, when DA was applied during an STDP protocol it depressed presynaptic inhibition in the mouse but not in the human cortex. Thus, DA modulates excitatory synaptic plasticity differently in human vs. mouse cortex. The data strengthens the importance of DA in gating cognition in humans, and may inform on therapeutic interventions to recover brain function from diseases.
Collapse
Affiliation(s)
- Emma Louise Louth
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | | | | | | | - Marco Capogna
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark.,Center for Proteins in Memory-PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| |
Collapse
|
107
|
Roles of the Functional Interaction between Brain Cholinergic and Dopaminergic Systems in the Pathogenesis and Treatment of Schizophrenia and Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094299. [PMID: 33919025 PMCID: PMC8122651 DOI: 10.3390/ijms22094299] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Most physiologic processes in the brain and related diseases involve more than one neurotransmitter system. Thus, elucidation of the interaction between different neurotransmitter systems could allow for better therapeutic approaches to the treatments of related diseases. Dopaminergic (DAergic) and cholinergic neurotransmitter system regulate various brain functions that include cognition, movement, emotion, etc. This review focuses on the interaction between the brain DAergic and cholinergic systems with respect to the pathogenesis and treatment of schizophrenia and Parkinson’s disease (PD). We first discussed the selection of motor plans at the level of basal ganglia, the major DAergic and cholinergic pathways in the brain, and the receptor subtypes involved in the interaction between the two signaling systems. Next, the roles of each signaling system were discussed in the context of the negative symptoms of schizophrenia, with a focus on the α7 nicotinic cholinergic receptor and the dopamine D1 receptor in the prefrontal cortex. In addition, the roles of the nicotinic and dopamine receptors were discussed in the context of regulation of striatal cholinergic interneurons, which play crucial roles in the degeneration of nigrostriatal DAergic neurons and the development of L-DOPA-induced dyskinesia in PD patients. Finally, we discussed the general mechanisms of nicotine-induced protection of DAergic neurons.
Collapse
|
108
|
Yamada Y, Frith EM, Wong V, Spitz RW, Bell ZW, Chatakondi RN, Abe T, Loenneke JP. Acute exercise and cognition: A review with testable questions for future research into cognitive enhancement with blood flow restriction. Med Hypotheses 2021; 151:110586. [PMID: 33848917 DOI: 10.1016/j.mehy.2021.110586] [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: 02/04/2021] [Revised: 03/12/2021] [Accepted: 03/24/2021] [Indexed: 12/01/2022]
Abstract
Blood flow restriction, in combination with low load/intensity exercise, has consistently been shown to increase both muscle size and strength. In contrast, the effects of blood flow restricted exercise on cognition have not been well studied. Therefore, the purpose of this paper is 1) to review the currently available literature investigating the impact of blood flow restricted exercise on cognition and 2) to provide some hypotheses for how blood flow restriction might provide an additive stimulus for augmenting specific cognitive domains above exercise alone. Given the lack of research in this area, the effects of blood flow restricted exercise on cognition are still unclear. We hypothesize that blood flow restricted exercise could potentially enhance several cognitive domains (such as attention, executive functioning, and memory) through increases in lactate production, catecholamine concentration, and PGC-1α expression. We review work that suggests that blood flow restriction is not only a beneficial strategy to improve musculoskeletal function but could also be a favorable method for enhancing multiple domains of cognition. Nonetheless, it must be emphasized this is a hypothesis that currently has only minimal experimental support, and further investigations in the future are necessary to test the hypothesis.
Collapse
Affiliation(s)
- Yujiro Yamada
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA
| | - Emily M Frith
- Department of Psychology, Cognitive Neuroscience of Creativity Laboratory, Pennsylvania State University, PA 16801, USA
| | - Vickie Wong
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA
| | - Robert W Spitz
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA
| | - Zachary W Bell
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA
| | - Raksha N Chatakondi
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA
| | - Takashi Abe
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA
| | - Jeremy P Loenneke
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
109
|
Speranza L, di Porzio U, Viggiano D, de Donato A, Volpicelli F. Dopamine: The Neuromodulator of Long-Term Synaptic Plasticity, Reward and Movement Control. Cells 2021; 10:735. [PMID: 33810328 PMCID: PMC8066851 DOI: 10.3390/cells10040735] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA) is a key neurotransmitter involved in multiple physiological functions including motor control, modulation of affective and emotional states, reward mechanisms, reinforcement of behavior, and selected higher cognitive functions. Dysfunction in dopaminergic transmission is recognized as a core alteration in several devastating neurological and psychiatric disorders, including Parkinson's disease (PD), schizophrenia, bipolar disorder, attention deficit hyperactivity disorder (ADHD) and addiction. Here we will discuss the current insights on the role of DA in motor control and reward learning mechanisms and its involvement in the modulation of synaptic dynamics through different pathways. In particular, we will consider the role of DA as neuromodulator of two forms of synaptic plasticity, known as long-term potentiation (LTP) and long-term depression (LTD) in several cortical and subcortical areas. Finally, we will delineate how the effect of DA on dendritic spines places this molecule at the interface between the motor and the cognitive systems. Specifically, we will be focusing on PD, vascular dementia, and schizophrenia.
Collapse
Affiliation(s)
- Luisa Speranza
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Umberto di Porzio
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, CNR, 80131 Naples, Italy
| | - Davide Viggiano
- Department of Translational Medical Sciences, Genetic Research Institute “Gaetano Salvatore”, University of Campania “L. Vanvitelli”, IT and Biogem S.c.a.r.l., 83031 Ariano Irpino, Italy; (D.V.); (A.d.D.)
| | - Antonio de Donato
- Department of Translational Medical Sciences, Genetic Research Institute “Gaetano Salvatore”, University of Campania “L. Vanvitelli”, IT and Biogem S.c.a.r.l., 83031 Ariano Irpino, Italy; (D.V.); (A.d.D.)
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| |
Collapse
|
110
|
Tan T, Wang W, Liu T, Zhong P, Conrow-Graham M, Tian X, Yan Z. Neural circuits and activity dynamics underlying sex-specific effects of chronic social isolation stress. Cell Rep 2021; 34:108874. [PMID: 33761364 DOI: 10.1016/j.celrep.2021.108874] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/09/2020] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
Exposure to prolonged stress in critical developmental periods induces heightened vulnerability to psychiatric disorders, which may have sex-specific consequences. Here we investigate the neuronal circuits mediating behavioral changes in mice after chronic adolescent social isolation stress. Escalated aggression is exhibited in stressed males, while social withdrawal is shown in stressed females. In vivo multichannel recordings of free-moving animals indicate that pyramidal neurons in prefrontal cortex (PFC) from stressed males exhibit the significantly decreased spike activity during aggressive attacks, while PFC pyramidal neurons from stressed females show a blunted increase of discharge rates during sociability tests. Chemogenetic and electrophysiological evidence shows that PFC hypofunctioning and BLA principal neuron hyperactivity contribute to the elevated aggression in stressed males, while PFC hypofunctioning and VTA dopamine neuron hypoactivity contribute to the diminished sociability in stressed females. These results establish a framework for understanding the circuit and physiological mechanisms underlying sex-specific divergent effects of stress.
Collapse
Affiliation(s)
- Tao Tan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Wei Wang
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Tiaotiao Liu
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Ping Zhong
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Megan Conrow-Graham
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Xin Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
111
|
Iwai Y, Ozawa K, Yahagi K, Mishima T, Akther S, Vo CT, Lee AB, Tanaka M, Itohara S, Hirase H. Transient Astrocytic Gq Signaling Underlies Remote Memory Enhancement. Front Neural Circuits 2021; 15:658343. [PMID: 33828463 PMCID: PMC8019746 DOI: 10.3389/fncir.2021.658343] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 01/31/2023] Open
Abstract
Astrocytes elicit transient Ca2+ elevations induced by G protein-coupled receptors (GPCRs), yet their role in vivo remains unknown. To address this, transgenic mice with astrocytic expression of the optogenetic Gq-type GPCR, Optoα1AR, were established, in which transient Ca2+ elevations similar to those in wild type mice were induced by brief blue light illumination. Activation of cortical astrocytes resulted in an adenosine A1 receptor-dependent inhibition of neuronal activity. Moreover, sensory stimulation with astrocytic activation induced long-term depression of sensory evoked response. At the behavioral level, repeated astrocytic activation in the anterior cortex gradually affected novel open field exploratory behavior, and remote memory was enhanced in a novel object recognition task. These effects were blocked by A1 receptor antagonism. Together, we demonstrate that GPCR-triggered Ca2+ elevation in cortical astrocytes has causal impacts on neuronal activity and behavior.
Collapse
Affiliation(s)
- Youichi Iwai
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
| | - Katsuya Ozawa
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
| | - Kazuko Yahagi
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
| | - Tsuneko Mishima
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sonam Akther
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Trang Vo
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ashley Bomin Lee
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mika Tanaka
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Wako, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Center for Brain Science, Wako, Japan
| | - Hajime Hirase
- Laboratory for Neuron-Glia Circuitry, RIKEN Center for Brain Science, Wako, Japan
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
112
|
Noradrenergic Suppression of Persistent Firing in Hippocampal CA1 Pyramidal Cells through cAMP-PKA Pathway. eNeuro 2021; 8:ENEURO.0440-20.2020. [PMID: 33637539 PMCID: PMC8009666 DOI: 10.1523/eneuro.0440-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/03/2020] [Accepted: 12/20/2020] [Indexed: 12/15/2022] Open
Abstract
Persistent firing is believed to be a cellular correlate of working memory. While the effects of noradrenaline (NA) on working memory have widely been described, its effect on the cellular mechanisms of persistent firing remains largely unknown. Using in vitro intracellular recordings, we demonstrate that persistent firing is supported by individual neurons in hippocampal CA1 pyramidal cells through cholinergic receptor activation, but is dramatically attenuated by NA. In contrast to the classical theory that recurrent synaptic excitation supports persistent firing, suppression of persistent firing by NA was independent of synaptic transmission, indicating that the mechanism is intrinsic to individual cells. In agreement with detrimental effects of cAMP on working memory, we demonstrate that the suppressive effect of NA was through cAMP-PKA pathway. In addition, activation of β1 and/or β3 adrenergic receptors, which increases cAMP levels, suppressed persistent firing. These results are in line with working memory decline observed during high levels of NA and cAMP, which are implicated in high stress, aging, and schizophrenia.
Collapse
|
113
|
Vijayraghavan S, Everling S. Neuromodulation of Persistent Activity and Working Memory Circuitry in Primate Prefrontal Cortex by Muscarinic Receptors. Front Neural Circuits 2021; 15:648624. [PMID: 33790746 PMCID: PMC8005543 DOI: 10.3389/fncir.2021.648624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/11/2021] [Indexed: 12/31/2022] Open
Abstract
Neuromodulation by acetylcholine plays a vital role in shaping the physiology and functions of cerebral cortex. Cholinergic neuromodulation influences brain-state transitions, controls the gating of cortical sensory stimulus responses, and has been shown to influence the generation and maintenance of persistent activity in prefrontal cortex. Here we review our current understanding of the role of muscarinic cholinergic receptors in primate prefrontal cortex during its engagement in the performance of working memory tasks. We summarize the localization of muscarinic receptors in prefrontal cortex, review the effects of muscarinic neuromodulation on arousal, working memory and cognitive control tasks, and describe the effects of muscarinic M1 receptor stimulation and blockade on the generation and maintenance of persistent activity of prefrontal neurons encoding working memory representations. Recent studies describing the pharmacological effects of M1 receptors on prefrontal persistent activity demonstrate the heterogeneity of muscarinic actions and delineate unexpected modulatory effects discovered in primate prefrontal cortex when compared with studies in rodents. Understanding the underlying mechanisms by which muscarinic receptors regulate prefrontal cognitive control circuitry will inform the search of muscarinic-based therapeutic targets in the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Susheel Vijayraghavan
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Stefan Everling
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Robarts Research Institute, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
114
|
Zeng F, Zhang Q, Liu Y, Sun G, Li A, Talay RS, Wang J. AMPAkines potentiate the corticostriatal pathway to reduce acute and chronic pain. Mol Brain 2021; 14:45. [PMID: 33653395 PMCID: PMC7923831 DOI: 10.1186/s13041-021-00757-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022] Open
Abstract
The corticostriatal circuit plays an important role in the regulation of reward- and aversion-types of behaviors. Specifically, the projection from the prelimbic cortex (PL) to the nucleus accumbens (NAc) has been shown to regulate sensory and affective aspects of pain in a number of rodent models. Previous studies have shown that enhancement of glutamate signaling through the NAc by AMPAkines, a class of agents that specifically potentiate the function of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, reduces acute and persistent pain. However, it is not known whether postsynaptic potentiation of the NAc with these agents can achieve the full anti-nociceptive effects of PL activation. Here we compared the impact of AMPAkine treatment in the NAc with optogenetic activation of the PL on pain behaviors in rats. We found that not only does AMPAkine treatment partially reconstitute the PL inhibition of sensory withdrawals, it fully occludes the effect of the PL on reducing the aversive component of pain. These results indicate that the NAc is likely one of the key targets for the PL, especially in the regulation of pain aversion. Furthermore, our results lend support for neuromodulation or pharmacological activation of the corticostriatal circuit as an important analgesic approach.
Collapse
Affiliation(s)
- Fei Zeng
- Department of Pain, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, People's Republic of China
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA
| | - Qiaosheng Zhang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA
| | - Yaling Liu
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA
| | - Guanghao Sun
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA
| | - Anna Li
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA
| | - Robert S Talay
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
115
|
Di Benedetto G, Iannucci LF, Surdo NC, Zanin S, Conca F, Grisan F, Gerbino A, Lefkimmiatis K. Compartmentalized Signaling in Aging and Neurodegeneration. Cells 2021; 10:464. [PMID: 33671541 PMCID: PMC7926881 DOI: 10.3390/cells10020464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic AMP (cAMP) signalling cascade is necessary for cell homeostasis and plays important roles in many processes. This is particularly relevant during ageing and age-related diseases, where drastic changes, generally decreases, in cAMP levels have been associated with the progressive decline in overall cell function and, eventually, the loss of cellular integrity. The functional relevance of reduced cAMP is clearly supported by the finding that increases in cAMP levels can reverse some of the effects of ageing. Nevertheless, despite these observations, the molecular mechanisms underlying the dysregulation of cAMP signalling in ageing are not well understood. Compartmentalization is widely accepted as the modality through which cAMP achieves its functional specificity; therefore, it is important to understand whether and how this mechanism is affected during ageing and to define which is its contribution to this process. Several animal models demonstrate the importance of specific cAMP signalling components in ageing, however, how age-related changes in each of these elements affect the compartmentalization of the cAMP pathway is largely unknown. In this review, we explore the connection of single components of the cAMP signalling cascade to ageing and age-related diseases whilst elaborating the literature in the context of cAMP signalling compartmentalization.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Liliana F. Iannucci
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta C. Surdo
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Sofia Zanin
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Filippo Conca
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Francesca Grisan
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy;
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
116
|
Wang J, John Y, Barbas H. Pathways for Contextual Memory: The Primate Hippocampal Pathway to Anterior Cingulate Cortex. Cereb Cortex 2021; 31:1807-1826. [PMID: 33207365 PMCID: PMC7869091 DOI: 10.1093/cercor/bhaa333] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/27/2022] Open
Abstract
The anterior cingulate cortex (ACC) is one of the few prefrontal areas that receives robust direct hippocampal terminations. This pathway may enable current context and past experience to influence goal-directed actions and emotional regulation by prefrontal cortices. We investigated the still ill-understood organization of the pathway from anterior hippocampus to ACC (A24a, A25, A32) to identify laminar termination patterns and their postsynaptic excitatory and inhibitory targets from system to synapse in rhesus monkeys. The densest hippocampal terminations targeted posterior A25, a region that is involved in affective and autonomic regulation. Hippocampal terminations innervated mostly excitatory neurons (~90%), suggesting strong excitatory effects. Among the smaller fraction of inhibitory targets, hippocampal terminations in A25 preferentially innervated calretinin neurons, a pattern that differs markedly from rodents. Further, hippocampal terminations innervated spines with D1 receptors, particularly in the deep layers of A25, where D1 receptors are enriched in comparison with the upper layers. The proximity of hippocampal terminations to D1 receptors may enable dopamine to enhance information transfer from the hippocampus to A25 and contribute to dopaminergic influence downstream on goal-directed action and emotional control by prefrontal cortices, in processes that may be disrupted by excessive dopamine release during uncontrollable stress.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Health Sciences, Neural Systems Laboratory, Boston University, Boston, MA 02215, USA
| | - Yohan John
- Department of Health Sciences, Neural Systems Laboratory, Boston University, Boston, MA 02215, USA
| | - Helen Barbas
- Department of Health Sciences, Neural Systems Laboratory, Boston University, Boston, MA 02215, USA
- Graduate Program in Neuroscience, Boston University and School of Medicine, Boston, MA 02215, USA
| |
Collapse
|
117
|
Nuno-Perez A, Trusel M, Lalive AL, Congiu M, Gastaldo D, Tchenio A, Lecca S, Soiza-Reilly M, Bagni C, Mameli M. Stress undermines reward-guided cognitive performance through synaptic depression in the lateral habenula. Neuron 2021; 109:947-956.e5. [PMID: 33535028 PMCID: PMC7980092 DOI: 10.1016/j.neuron.2021.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/05/2020] [Accepted: 01/08/2021] [Indexed: 01/11/2023]
Abstract
Weighing alternatives during reward pursuit is a vital cognitive computation that, when disrupted by stress, yields aspects of neuropsychiatric disorders. To examine the neural mechanisms underlying these phenomena, we employed a behavioral task in which mice were confronted by a reward and its omission (i.e., error). The experience of error outcomes engaged neuronal dynamics within the lateral habenula (LHb), a subcortical structure that supports appetitive behaviors and is susceptible to stress. A high incidence of errors predicted low strength of habenular excitatory synapses. Accordingly, stressful experiences increased error choices while decreasing glutamatergic neurotransmission onto LHb neurons. This synaptic adaptation required a reduction in postsynaptic AMPA receptors (AMPARs), irrespective of the anatomical source of glutamate. Bidirectional control of habenular AMPAR transmission recapitulated and averted stress-driven cognitive deficits. Thus, a subcortical synaptic mechanism vulnerable to stress underlies behavioral efficiency during cognitive performance.
Collapse
Affiliation(s)
- Alvaro Nuno-Perez
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Massimo Trusel
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Arnaud L Lalive
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Mauro Congiu
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Denise Gastaldo
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Anna Tchenio
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Salvatore Lecca
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | | | - Claudia Bagni
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland
| | - Manuel Mameli
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005 Lausanne, Switzerland; Inserm, UMR-S 839, 75005 Paris, France.
| |
Collapse
|
118
|
Talay RS, Liu Y, Michael M, Li A, Friesner ID, Zeng F, Sun G, Chen ZS, Zhang Q, Wang J. Pharmacological restoration of anti-nociceptive functions in the prefrontal cortex relieves chronic pain. Prog Neurobiol 2021; 201:102001. [PMID: 33545233 DOI: 10.1016/j.pneurobio.2021.102001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/14/2021] [Accepted: 01/24/2021] [Indexed: 12/30/2022]
Abstract
Chronic pain affects one in four adults, and effective non-sedating and non-addictive treatments are urgently needed. Chronic pain causes maladaptive changes in the cerebral cortex, which can lead to impaired endogenous nociceptive processing. However, it is not yet clear if drugs that restore endogenous cortical regulation could provide an effective therapeutic strategy for chronic pain. Here, we studied the nociceptive response of neurons in the prelimbic region of the prefrontal cortex (PL-PFC) in freely behaving rats using a spared nerve injury (SNI) model of chronic pain, and the impact of AMPAkines, a class of drugs that increase central glutamate signaling, on such response. We found that neurons in the PL-PFC increase their firing rates in response to noxious stimulations; chronic neuropathic pain, however, suppressed this important cortical pain response. Meanwhile, CX546, a well-known AMPAkine, restored the anti-nociceptive response of PL-PFC neurons in the chronic pain condition. In addition, both systemic administration and direct delivery of CX546 into the PL-PFC inhibited symptoms of chronic pain, whereas optogenetic inactivation of the PFC neurons or administration of AMPA receptor antagonists in the PL-PFC blocked the anti-nociceptive effects of CX546. These results indicate that restoration of the endogenous anti-nociceptive functions in the PL-PFC by pharmacological agents such as AMPAkines constitutes a successful strategy to treat chronic neuropathic pain.
Collapse
Affiliation(s)
- Robert S Talay
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States
| | - Yaling Liu
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States; Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Matthew Michael
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States
| | - Anna Li
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States
| | - Isabel D Friesner
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States
| | - Fei Zeng
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States
| | - Guanghao Sun
- Department of Psychiatry, New York University Langone Health, New York, NY 10016, United States
| | - Zhe Sage Chen
- Department of Psychiatry, New York University Langone Health, New York, NY 10016, United States; Neuroscience Institute, New York University Langone Health, New York, NY 10016, United States
| | - Qiaosheng Zhang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States.
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Langone Health, New York, NY 10016, United States; Neuroscience Institute, New York University Langone Health, New York, NY 10016, United States; Department of Neuroscience and Physiology, New York University Langone Health, New York, NY 10016, United States.
| |
Collapse
|
119
|
Ross JA, Van Bockstaele EJ. The Locus Coeruleus- Norepinephrine System in Stress and Arousal: Unraveling Historical, Current, and Future Perspectives. Front Psychiatry 2021; 11:601519. [PMID: 33584368 PMCID: PMC7873441 DOI: 10.3389/fpsyt.2020.601519] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2023] Open
Abstract
Arousal may be understood on a spectrum, with excessive sleepiness, cognitive dysfunction, and inattention on one side, a wakeful state in the middle, and hypervigilance, panic, and psychosis on the other side. However, historically, the concepts of arousal and stress have been challenging to define as measurable experimental variables. Divergent efforts to study these subjects have given rise to several disciplines, including neurobiology, neuroendocrinology, and cognitive neuroscience. We discuss technological advancements that chronologically led to our current understanding of the arousal system, focusing on the multifaceted nucleus locus coeruleus. We share our contemporary perspective and the hypotheses of others in the context of our current technological capabilities and future developments that will be required to move forward in this area of research.
Collapse
Affiliation(s)
- Jennifer A. Ross
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, United States
| | | |
Collapse
|
120
|
Hayes J, Laursen B, Eneberg E, Kehler J, Rasmussen LK, Langgard M, Bastlund JF, Gerdjikov TV. Phosphodiesterase type 1 inhibition alters medial prefrontal cortical activity during goal-driven behaviour and partially reverses neurophysiological deficits in the rat phencyclidine model of schizophrenia. Neuropharmacology 2021; 186:108454. [PMID: 33444639 DOI: 10.1016/j.neuropharm.2021.108454] [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: 07/14/2020] [Revised: 11/27/2020] [Accepted: 01/04/2021] [Indexed: 10/22/2022]
Abstract
Positive modulation of cAMP signalling by phosphodiesterase (PDE) inhibitors has recently been explored as a potential target for the reversal of cognitive and behavioural deficits implicating the corticoaccumbal circuit. Previous studies show that PDE type 1 isoform B (PDE1B) inhibition may improve memory function in rodent models; however, the contribution of PDE1B inhibition to impulsivity, attentional and motivational functions as well as its neurophysiological effects have not been investigated. To address this, we recorded single unit activity in medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) in Lister Hooded rats treated with the PDE1B inhibitor Lu AF64386 and tested in the 5-choice serial reaction time task (5-CSRTT). We also asked whether PDE1B inhibition modulates neurophysiological deficits produced by subchronic phencyclidine (PCP) treatment, a rat pharmacological model of schizophrenia. Lu AF64386 significantly affected behavioural parameters consistent with a reduction in goal-directed behaviour, however without affecting accuracy. Additionally, it reduced mPFC neuronal activity. Pre-treatment with PCP did not affect behavioural parameters, however it significantly disrupted overall neuronal firing while increasing phasic responses to reward-predicting cues and disrupting mPFC-NAc cross-talk. The latter two effects were reversed by Lu AF64386. These findings suggest PDE1B inhibition may be beneficial in disorders implicating a dysfunction of the mPFC-NAc network.
Collapse
Affiliation(s)
- Jessica Hayes
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, United Kingdom
| | | | | | - Jan Kehler
- Molecular Discovery and Innovation, Lundbeck A/S, Denmark
| | | | | | | | - Todor V Gerdjikov
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, United Kingdom.
| |
Collapse
|
121
|
Sinha N, Berg CN, Yassa MA, Gluck MA. Increased dynamic flexibility in the medial temporal lobe network following an exercise intervention mediates generalization of prior learning. Neurobiol Learn Mem 2021; 177:107340. [PMID: 33186745 PMCID: PMC7861122 DOI: 10.1016/j.nlm.2020.107340] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/29/2020] [Accepted: 11/01/2020] [Indexed: 11/28/2022]
Abstract
Recent work has conceptualized the brain as a network comprised of groups of sub-networks or modules. "Flexibility" of brain network(s) indexes the dynamic reconfiguration of comprising modules. Using novel techniques from dynamic network neuroscience applied to high-resolution resting-state functional magnetic resonance imaging (fMRI), the present study investigated the effects of an aerobic exercise intervention on the dynamic rearrangement of modular community structure-a measure of neural flexibility-within the medial temporal lobe (MTL) network. The MTL is one of the earliest brain regions impacted by Alzheimer's disease. It is also a major site of neuroplasticity that is sensitive to the effects of exercise. In a two-group non-randomized, repeated measures and matched control design with 34 healthy older adults, we observed an exercise-related increase in flexibility within the MTL network. Furthermore, MTL network flexibility mediated the beneficial effect aerobic exercise had on mnemonic flexibility, as measured by the ability to generalize past learning to novel task demands. Our results suggest that exercise exerts a rehabilitative and protective effect on MTL function, resulting in dynamically evolving networks of regions that interact in complex communication patterns. These reconfigurations may underlie exercise-induced improvements on cognitive measures of generalization, which are sensitive to subtle changes in the MTL.
Collapse
Affiliation(s)
- Neha Sinha
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, NJ, USA.
| | - Chelsie N Berg
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, NJ, USA.
| | - Michael A Yassa
- Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
| | - Mark A Gluck
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, NJ, USA.
| |
Collapse
|
122
|
van Dyck CH, Arnsten AFT, Padala PR, Brawman-Mintzer O, Lerner AJ, Porsteinsson AP, Scherer RW, Levey AI, Herrmann N, Jamil N, Mintzer JE, Lanctôt KL, Rosenberg PB. Neurobiologic Rationale for Treatment of Apathy in Alzheimer's Disease With Methylphenidate. Am J Geriatr Psychiatry 2021; 29:51-62. [PMID: 32461027 PMCID: PMC7641967 DOI: 10.1016/j.jagp.2020.04.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 11/23/2022]
Abstract
The public health burden of Alzheimer's disease (AD) is related not only to cognitive symptoms, but also to neuropsychiatric symptoms, including apathy. Apathy is defined as a quantitative reduction of goal-directed activity in comparison to a previous level of functioning and affects 30%-70% of persons with AD. Previous attempts to treat apathy in AD-both nonpharmacologically and pharmacologically-have been wanting. Catecholaminergic treatment with methylphenidate has shown encouraging results in initial trials of apathy in AD. Understanding the neuronal circuits underlying motivated behavior and their reliance on catecholamine actions helps provide a rationale for methylphenidate actions in the treatment of apathy in patients with AD. Anatomical, physiological, and behavioral studies have identified parallel, cortical-basal ganglia circuits that govern action, cognition, and emotion and play key roles in motivated behavior. Understanding the distinct contributions to motivated behavior of subregions of the prefrontal cortex-dorsolateral, orbital-ventromedial, and dorsomedial-helps to explain why degeneration of these areas in AD results in apathetic behaviors. We propose that the degeneration of the prefrontal cortex in AD produces symptoms of apathy. We further propose that methylphenidate treatment may ameliorate those symptoms by boosting norepinephrine and dopamine actions in prefrontal-striatal-thalamocortical circuits.
Collapse
Affiliation(s)
| | | | - Prasad R Padala
- University of Arkansas for Medical Sciences, Central Arkansas Veterans Healthcare System (PRP), Little Rock, AR
| | - Olga Brawman-Mintzer
- Medical University of South Carolina and Ralph H. Johnson Veterans Administration Medical Center (OB-M, JEM), Charleston, SC
| | - Alan J Lerner
- University Hospitals - Case Western Reserve University (AJL), Cleveland, OH
| | | | - Roberta W Scherer
- Johns Hopkins University Bloomberg School of Public Health (RWS), Baltimore, MD
| | | | - Nathan Herrmann
- Sunnybrook Research Institute (NH, KLL), Toronto, ON, Canada
| | - Nimra Jamil
- Johns Hopkins University School of Medicine (NJ, PBR), Baltimore, MD
| | - Jacobo E Mintzer
- Medical University of South Carolina and Ralph H. Johnson Veterans Administration Medical Center (OB-M, JEM), Charleston, SC
| | | | - Paul B Rosenberg
- Johns Hopkins University School of Medicine (NJ, PBR), Baltimore, MD
| |
Collapse
|
123
|
Effects of blocking mGluR5 on primate dorsolateral prefrontal cortical neuronal firing and working memory performance. Psychopharmacology (Berl) 2021; 238:97-106. [PMID: 32939596 PMCID: PMC7794104 DOI: 10.1007/s00213-020-05661-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022]
Abstract
RATIONALE Metabotropic glutamate type 5 receptor (mGluR5) antagonists are under development for treating cognitive disorders such as Fragile X syndrome and Alzheimer's disease, largely based on success in mouse models, where post-synaptic mGluR5 stimulation weakens synaptic functions in hippocampus. However, human trials of mGluR5 antagonists have yet to be successful. This may be due in part to the differing effects of mGluR5 in hippocampus vs. prefrontal cortex, as mGluR5 are primarily post-synaptic in rodent hippocampus, but are both pre- and post-synaptic in the dorsolateral prefrontal cortical (dlPFC) circuits known to subserve working memory. OBJECTIVES AND METHODS The current study examined the effects of the selective mGluR5 negative allosteric modulator, MTEP (3-((2-Methyl-1,3-thiazol-4-yl)ethynyl)pyridine hydrochloride), on neuronal firing and working memory performance in aging rhesus monkeys with naturally occurring impairments in neuronal firing and cognitive performance. RESULTS We found that iontophoresis of MTEP directly onto dlPFC "Delay cells" had an inverted U dose-response, where low doses tended to enhance task-related firing, but higher doses suppressed neuronal firing. Similar effects were seen on cognitive performance following systemic MTEP administration (0.0001-0.1 mg/kg), with MTEP producing erratic dose-response curves. In the subset of monkeys (50%) that showed replicable improvement with MTEP, co-administration with the mGluR5 PAM, CDPPB (3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide), blocked MTEP beneficial effects, consistent with mGluR5 actions. CONCLUSIONS The mixed effects of MTEP on cognitive performance may arise from opposing actions at pre- vs. post-synaptic mGluR5 in dlPFC. These data from monkeys suggest that future clinical trials should include low doses, and identification of potential subgroup responders.
Collapse
|
124
|
Glausier JR, Datta D, Fish KN, Chung DW, Melchitzky DS, Lewis DA. Laminar Differences in the Targeting of Dendritic Spines by Cortical Pyramidal Neurons and Interneurons in Human Dorsolateral Prefrontal Cortex. Neuroscience 2021; 452:181-191. [PMID: 33212224 PMCID: PMC7770119 DOI: 10.1016/j.neuroscience.2020.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 01/05/2023]
Abstract
Activation of specific neural circuits in different layers of the primate dorsolateral prefrontal cortex (DLPFC) is essential for working memory, a core cognitive function. Recurrent excitation between pyramidal neurons in middle and deep layers of the DLPFC contributes to the laminar-specific activity associated with different working memory subprocesses. Excitation between cortical pyramidal neurons is mediated by glutamatergic synapses on dendritic spines, but whether the relative abundance of spines receiving cortical inputs differs between middle and deep cortical layers in human DLPFC is unknown. Additionally, GABAergic inputs to spines sculpt pyramidal neuron activity. Whether dendritic spines that receive a glutamatergic input from a cortical pyramidal neuron are targeted by GABAergic interneurons in the human DLPFC is unknown. Using triple-label fluorescence confocal microscopy, we found that 1) the density of spines receiving an input from a cortical pyramidal neuron is greater in the middle than in the deep laminar zone, 2) dendritic spines dually innervated by a cortical pyramidal neuron and an interneuron are present in the human DLPFC, and 3) the density of spines dually innervated by a cortical pyramidal neuron and an interneuron is also greater in the middle than in the deep laminar zone. Ultrastructural analyses support the presence of spines that receive a cortical pyramidal neuron synapse and an interneuron synapse in human and monkey DLPFC. These data support the notion that the DLPFC middle laminar zone is particularly endowed with a microcircuit structure that supports the gating, integrating and fine-tuning of synaptic information in recurrent excitatory microcircuits.
Collapse
Affiliation(s)
- Jill R Glausier
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| | - Dibyadeep Datta
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA; Department of Neuroscience, Yale University, Sterling Hall of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Kenneth N Fish
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Daniel W Chung
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Darlene S Melchitzky
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA.
| |
Collapse
|
125
|
Stavroulaki V, Giakoumaki SG, Sidiropoulou K. Working memory training effects across the lifespan: Evidence from human and experimental animal studies. Mech Ageing Dev 2020; 194:111415. [PMID: 33338498 DOI: 10.1016/j.mad.2020.111415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
Working memory refers to a cognitive function that provides temporary storage and manipulation of the information necessary for complex cognitive tasks. Due to its central role in general cognition, several studies have investigated the possibility that training on working memory tasks could improve not only working memory function but also increase other cognitive abilities or modulate other behaviors. This possibility is still highly controversial, with prior studies providing contradictory findings. The lack of systematic approaches and methodological shortcomings complicates this debate even more. This review highlights the impact of working memory training at different ages on humans. Finally, it demonstrates several findings about the neural substrate of training in both humans and experimental animals, including non-human primates and rodents.
Collapse
Affiliation(s)
| | - Stella G Giakoumaki
- Laboratory of Neuropsychology, Department of Psychology, Gallos University Campus, University of Crete, Rethymno, 74100, Crete, Greece; University of Crete Research Center for the Humanities, The Social and Educational Sciences, University of Crete, Rethymno, 74100, Crete, Greece
| | - Kyriaki Sidiropoulou
- Dept of Biology, University of Crete, Greece; Institute of Molecular Biology and Biotechnology - Foundation for Research and Technology Hellas, Greece.
| |
Collapse
|
126
|
Zhang Q, Weber MA, Narayanan NS. Medial prefrontal cortex and the temporal control of action. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 158:421-441. [PMID: 33785154 DOI: 10.1016/bs.irn.2020.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Across species, the medial prefrontal cortex guides actions in time. This process can be studied using behavioral paradigms such as simple reaction-time and interval-timing tasks. Temporal control of action can be influenced by prefrontal neurotransmitters such as dopamine and acetylcholine and is highly relevant to human diseases such as Parkinson's disease, schizophrenia, and attention-deficit hyperactivity disorder (ADHD). We review evidence that across species, medial prefrontal lesions impair the temporal control of action. We then consider neurophysiological correlates in humans, primates, and rodents that might encode temporal processing and relate to cognitive-control mechanisms. These data have informed brain-stimulation studies in rodents and humans that can compensate for timing deficits. This line of work illuminates basic mechanisms of temporal control of action in the medial prefrontal cortex, which underlies a range of high-level cognitive processing and could contribute to new biomarkers and therapies for human brain diseases.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | - Matthew A Weber
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | | |
Collapse
|
127
|
Choi JY, Jang HJ, Ornelas S, Fleming WT, Fürth D, Au J, Bandi A, Engel EA, Witten IB. A Comparison of Dopaminergic and Cholinergic Populations Reveals Unique Contributions of VTA Dopamine Neurons to Short-Term Memory. Cell Rep 2020; 33:108492. [PMID: 33326775 PMCID: PMC8038523 DOI: 10.1016/j.celrep.2020.108492] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 09/18/2020] [Accepted: 11/17/2020] [Indexed: 10/22/2022] Open
Abstract
We systematically compare the contributions of two dopaminergic and two cholinergic ascending populations to a spatial short-term memory task in rats. In ventral tegmental area dopamine (VTA-DA) and nucleus basalis cholinergic (NB-ChAT) populations, trial-by-trial fluctuations in activity during the delay period relate to performance with an inverted-U, despite the fact that both populations have low activity during that time. Transient manipulations reveal that only VTA-DA neurons, and not the other three populations we examine, contribute causally and selectively to short-term memory. This contribution is most significant during the delay period, when both increases and decreases in VTA-DA activity impair short-term memory. Our results reveal a surprising dissociation between when VTA-DA neurons are most active and when they have the biggest causal contribution to short-term memory, and they also provide support for classic ideas about an inverted-U relationship between neuromodulation and cognition.
Collapse
Affiliation(s)
- Jung Yoon Choi
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Psychology, Princeton University, Princeton, NJ 08544, USA
| | - Hee Jae Jang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Sharon Ornelas
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Weston T Fleming
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Daniel Fürth
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Jennifer Au
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Akhil Bandi
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Esteban A Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Psychology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
128
|
Pharmacological inhibition of phosphodiesterase 7 enhances consolidation processes of spatial memory. Neurobiol Learn Mem 2020; 177:107357. [PMID: 33278592 DOI: 10.1016/j.nlm.2020.107357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022]
Abstract
Augmentation of cAMP signaling through inhibition of phosphodiesterases (PDE) is known to enhance plasticity and memory. Inhibition of PDE4 enhances consolidation into memory, but less is known about the role of other cAMP specific PDEs. Here, we tested the effects of oral treatment with a selective inhibitor of PDE7 of nanomolar potency on spatial and contextual memory. In an object location task, doses of 0.3-3 mg/kg administered 3 h after training dose-dependently attenuated time-dependent forgetting in rats. Significant enhancement of memory occurred at a dose of 3 mg/kg with corresponding brain levels consistent with PDE7 inhibition. The same dose given prior to training augmented contextual fear conditioning. In mice, daily dosing before training enhanced spatial memory in two different incremental learning paradigms in the Barnes Maze. Drug treated mice made significantly less errors locating the escape in a probe-test 24 h after the end of training, and they exhibited hippocampal-dependent spatial search strategies more frequently than controls, which tended to show serial sampling of escape locations. Acquisition and short-term memory, in contrast, were unaffected. Our data provide evidence for a role of PDE7 in the consolidation of hippocampal-dependent memory. We suggest that targeting PDE7 for memory enhancement may provide an alternative to PDE4 inhibitors, which tend to have undesirable gastrointestinal side-effects.
Collapse
|
129
|
Arnsten AFT. Guanfacine's mechanism of action in treating prefrontal cortical disorders: Successful translation across species. Neurobiol Learn Mem 2020; 176:107327. [PMID: 33075480 PMCID: PMC7567669 DOI: 10.1016/j.nlm.2020.107327] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/13/2020] [Indexed: 01/18/2023]
Abstract
The selective norepinephrine (NE) α2A-adrenoceptor (α2A-AR) agonist, guanfacine (Intuniv™), is FDA-approved for treating Attention Deficit Hyperactivity Disorder (ADHD) based on research in animals, a translational success story. Guanfacine is also widely used off-label in additional mental disorders that involve impaired functioning of the prefrontal cortex (PFC), including stress-related disorders such as substance abuse, schizotypic cognitive deficits, and traumatic brain injury. The PFC subserves high order cognitive and executive functions including working memory, abstract reasoning, insight and judgment, and top-down control of attention, action and emotion. These abilities arise from PFC microcircuits with extensive recurrent excitation through NMDAR synapses. There is powerful modulation of these synapses, where cAMP-PKA opening of nearby potassium (K+) channels can rapidly and dynamically alter synaptic strength to coordinate arousal state with cognitive state, e.g. to take PFC "offline" during uncontrollable stress. A variety of evidence shows that guanfacine acts within the PFC via post-synaptic α2A-AR on dendritic spines to inhibit cAMP-PKA-K+ channel signaling, thus strengthening network connectivity, enhancing PFC neuronal firing, and improving PFC cognitive functions. Although guanfacine's beneficial effects are present in rodent, they are especially evident in primates, where the PFC greatly expands and differentiates. In addition to therapeutic actions in PFC, stress-related disorders may also benefit from additional α2-AR actions, such as weakening plasticity in the amygdala, reducing NE release, and anti-inflammatory actions by deactivating microglia. Altogether, these NE α2-AR actions optimize top-down control by PFC networks, which may explain guanfacine's benefits in a variety of mental disorders.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Dept. Neuroscience, Yale Medical School, 333 Cedar St., New Haven, CT 06510, USA.
| |
Collapse
|
130
|
Datta D, Enwright JF, Arion D, Paspalas CD, Morozov YM, Lewis DA, Arnsten AFT. Mapping Phosphodiesterase 4D (PDE4D) in Macaque Dorsolateral Prefrontal Cortex: Postsynaptic Compartmentalization in Layer III Pyramidal Cell Circuits. Front Neuroanat 2020; 14:578483. [PMID: 33328902 PMCID: PMC7714912 DOI: 10.3389/fnana.2020.578483] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
cAMP signaling has powerful, negative effects on cognitive functions of the primate dorsolateral prefrontal cortex (dlPFC), opening potassium channels to reduce firing and impair working memory, and increasing tau phosphorylation in aging neurons. This contrasts with cAMP actions in classic circuits, where it enhances plasticity and transmitter release. PDE4 isozymes regulate cAMP actions, and thus have been a focus of research and drug discovery. Previous work has focused on the localization of PDE4A and PDE4B in dlPFC, but PDE4D is also of great interest, as it is the predominant PDE4 isoform in primate association cortex, and PDE4D expression decreases with aging in human dlPFC. Here we used laser-capture microdissection transcriptomics and found that PDE4D message is enriched in pyramidal cells compared to GABAergic PV-interneurons in layer III of the human dlPFC. A parallel study in rhesus macaques using high-spatial resolution immunoelectron microscopy revealed the ultrastructural locations of PDE4D in primate dlPFC with clarity not possible in human post-mortem tissue. PDE4D was especially prominent in dendrites associated with microtubules, mitochondria, and likely smooth endoplasmic reticulum (SER). There was substantial postsynaptic labeling in dendritic spines, associated with the SER spine-apparatus near glutamatergic-like axospinous synapses, but sparse labeling in axon terminals. We also observed dense PDE4D labeling perisynaptically in astroglial leaflets ensheathing glutamatergic connections. These data suggest that PDE4D is strategically positioned to regulate cAMP signaling in dlPFC glutamatergic synapses and circuits, especially in postsynaptic compartments where it is localized to influence cAMP actions on intracellular trafficking, mitochondrial physiology, and internal calcium release.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - John F. Enwright
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dominique Arion
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Constantinos D. Paspalas
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Yury M. Morozov
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - David A. Lewis
- Department of Psychiatry, Translational Neuroscience Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amy F. T. Arnsten
- Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
131
|
Reynaud AJ, Blini E, Koun E, Macaluso E, Meunier M, Hadj-Bouziane F. Atomoxetine modulates the contribution of low-level signals during free viewing of natural images in rhesus monkeys. Neuropharmacology 2020; 182:108377. [PMID: 33137343 DOI: 10.1016/j.neuropharm.2020.108377] [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: 07/09/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
Visuo-spatial attentional orienting is fundamental to selectively process behaviorally relevant information, depending on both low-level visual attributes of stimuli in the environment and higher-level factors, such as goals, expectations and prior knowledge. Growing evidence suggests an impact of the locus-cœruleus-norepinephrine (LC-NE) system in attentional orienting that depends on taskcontext. Nonetheless, most of previous studies used visual displays encompassing a target and various distractors, often preceded by cues to orient the attentional focus. This emphasizes the contribution of goal-driven processes, at the expense of other factors related to the stimulus content. Here, we aimed to determine the impact of NE on attentional orienting in more naturalistic conditions, using complex images and without any explicit task manipulation. We tested the effects of atomoxetine (ATX) injections, a NE reuptake inhibitor, on four monkeys during free viewing of images belonging to three categories: landscapes, monkey faces and scrambled images. Analyses of the gaze exploration patterns revealed, first, that the monkeys spent more time on each fixation under ATX compared to the control condition, regard less of the image content. Second, we found that, depending on the image content, ATX modulated the impact of low-level visual salience on attentional orienting. This effect correlated with the effect of ATX on the number and duration of fixations. Taken together, our results demonstrate that ATX adjusts the contribution of salience on attentional orienting depending on the image content, indicative of its role in balancing the role of stimulus-driven and top-down control during free viewing of complex stimuli.
Collapse
Affiliation(s)
- Amélie J Reynaud
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, ImpAct Team, Lyon, F-69000, France; University UCBL, Lyon 1, F-69000, France.
| | - Elvio Blini
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, ImpAct Team, Lyon, F-69000, France; University UCBL, Lyon 1, F-69000, France
| | - Eric Koun
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, ImpAct Team, Lyon, F-69000, France; University UCBL, Lyon 1, F-69000, France
| | - Emiliano Macaluso
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, ImpAct Team, Lyon, F-69000, France; University UCBL, Lyon 1, F-69000, France
| | - Martine Meunier
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, ImpAct Team, Lyon, F-69000, France; University UCBL, Lyon 1, F-69000, France
| | - Fadila Hadj-Bouziane
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, ImpAct Team, Lyon, F-69000, France; University UCBL, Lyon 1, F-69000, France.
| |
Collapse
|
132
|
Leslie SN, Datta D, Christensen KR, van Dyck CH, Arnsten AFT, Nairn AC. Phosphodiesterase PDE4D Is Decreased in Frontal Cortex of Aged Rats and Positively Correlated With Working Memory Performance and Inversely Correlated With PKA Phosphorylation of Tau. Front Aging Neurosci 2020; 12:576723. [PMID: 33192469 PMCID: PMC7655962 DOI: 10.3389/fnagi.2020.576723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/24/2020] [Indexed: 01/16/2023] Open
Abstract
Age is the largest risk factor for Alzheimer’s disease (AD) and contributes to cognitive impairment in otherwise healthy individuals. Thus, it is critical that we better understand the risk aging presents to vulnerable regions of the brain and carefully design therapeutics to address those effects. In this study we examined age-related changes in cAMP-regulatory protein, phosphodiesterase 4D (PDE4D). Inhibition of PDE4D is currently under investigation as a therapeutic target for AD based on memory-enhancing effects in rodent hippocampus. Therefore, it is important to understand the role of PDE4D in brain regions particularly vulnerable to disease such as the frontal association cortex (FC), where cAMP signaling can impair working memory via opening of potassium channels. We found that PDE4D protein level was decreased in the FC of both moderately and extremely aged rats, and that PDE4D level was correlated with performance on a FC-dependent working memory task. In extremely aged rats, PDE4D was also inversely correlated with levels of phosphorylated tau at serine 214 (S214), a site phosphorylated by protein kinase A. In vitro studies of the PDE4D inhibitor, GEBR-7b, further illustrated that inhibition of PDE4D activity enhanced phosphorylation of tau. pS214-tau phosphorylation is associated with early AD tau pathology, promotes tau dissociation from microtubules and primes subsequent tau hyperphosphorylation at other critical AD-related sites. Age-related loss of PDE4D may thus contribute to the specific vulnerability of the FC to degeneration in AD, and play a critical role in normal cAMP regulation, cautioning against the use of pan-PDE4D inhibitors as therapeutics.
Collapse
Affiliation(s)
- Shannon N Leslie
- Interdepartmental Neuroscience Program, Yale University, School of Medicine, New Haven, CT, United States.,Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University, School of Medicine, New Haven, CT, United States
| | - Kyle R Christensen
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Christopher H van Dyck
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University, School of Medicine, New Haven, CT, United States.,Department of Neurology, Yale University, School of Medicine, New Haven, CT, United States
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University, School of Medicine, New Haven, CT, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| |
Collapse
|
133
|
Shang P, Lindberg D, Starski P, Peyton L, Hong SI, Choi S, Choi DS. Chronic Alcohol Exposure Induces Aberrant Mitochondrial Morphology and Inhibits Respiratory Capacity in the Medial Prefrontal Cortex of Mice. Front Neurosci 2020; 14:561173. [PMID: 33192248 PMCID: PMC7646256 DOI: 10.3389/fnins.2020.561173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
Alcohol use disorder (AUD) is characterized as a chronic, relapsing disease with a pattern of excessive drinking despite negative consequences to an individual's life. Severe chronic alcohol use impairs the function of the medial prefrontal cortex (mPFC), which contributes to alcohol-induced cognitive and executive dysfunction. The mPFC contains more mitochondria compared to other cortical areas, which suggests mitochondrial damage may occur in AUD and trigger subsequent behavior change. Here, we identified morphological and functional changes in mitochondria in the mPFC in C57BL6/J mice after 8 h of withdrawal from chronic intermittent alcohol (CIA) exposure. Three-dimensional serial block-face scanning electron microscopy (SBFSEM) reconstruction revealed that CIA exposure elongated mPFC mitochondria and formed mitochondria-on-a-string (MOAS). Furthermore, alcohol significantly affected mitochondrial bioenergetics, including oxidative phosphorylation and electron transport, with inhibited aerobic respiration in mPFC mitochondria after CIA exposure. We also found decreased expression of fusion (mitofusin 2, Mfn2) and increased fission (mitochondrial fission 1 protein, Fis1) proteins in the mPFC of alcohol-treated mice. In sum, our study suggests that CIA exposure impairs mitochondrial dynamics and function in the mPFC.
Collapse
Affiliation(s)
- Pei Shang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Neurology, First Hospital of Jilin University, Changchun, China
| | - Daniel Lindberg
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Phillip Starski
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Lee Peyton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sa-Ik Hong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Sun Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Neuroscience Program, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
134
|
Upright NA, Baxter MG. Effect of chemogenetic actuator drugs on prefrontal cortex-dependent working memory in nonhuman primates. Neuropsychopharmacology 2020; 45:1793-1798. [PMID: 32193513 PMCID: PMC7608232 DOI: 10.1038/s41386-020-0660-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The most common chemogenetic neuromodulatory system, designer receptors exclusively activated by designer drugs (DREADDs), uses a non-endogenous actuator ligand to activate a modified muscarinic acetylcholine receptor that is insensitive to acetylcholine. It is crucial in studies using these systems to test the potential effects of DREADD actuators prior to any DREADD transduction, so that effects of DREADDs can be attributed to the chemogenetic system rather than the actuator drug, particularly in experiments using nonhuman primates. We investigated working memory performance after injections of three DREADD actuators, clozapine, olanzapine, and deschloroclozapine, in four male rhesus monkeys tested in a spatial delayed response task before any DREADD transduction took place. Performance at 0.1 mg/kg clozapine and 0.1 mg/kg deschloroclozapine did not differ from vehicle in any of the four subjects. 0.2 mg/kg clozapine impaired working memory function in three of the four monkeys. Two monkeys were impaired after 0.1 mg/kg olanzapine and two were impaired after 0.3 mg/kg deschloroclozapine. We speculate that the unique neuropharmacology of prefrontal cortex function makes the primate prefrontal cortex especially vulnerable to off-target effects of DREADD actuator drugs with affinity for endogenous monoaminergic receptor systems. These findings underscore the importance of within-subject controls for DREADD actuator drugs in the specific tasks under study to confirm that effects following DREADD receptor transduction are not owing to the actuator drug itself. They also suggest that off-target effects of DREADD actuators may limit translational applications of chemogenetic neuromodulation.
Collapse
Affiliation(s)
- Nicholas A Upright
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mark G Baxter
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
135
|
Gagnon SA, Waskom ML, Brown TI, Wagner AD. Stress Impairs Episodic Retrieval by Disrupting Hippocampal and Cortical Mechanisms of Remembering. Cereb Cortex 2020; 29:2947-2964. [PMID: 30060134 DOI: 10.1093/cercor/bhy162] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/14/2018] [Accepted: 06/18/2018] [Indexed: 01/12/2023] Open
Abstract
Despite decades of science investigating the neural underpinnings of episodic memory retrieval, a critical question remains: how does stress influence remembering and the neural mechanisms of recollection in humans? Here, we used functional magnetic resonance imaging and multivariate pattern analyses to examine the effects of acute stress during retrieval. We report that stress reduced the probability of recollecting the details of past experience, and that this impairment was driven, in part, by a disruption of the relationship between hippocampal activation, cortical reinstatement, and memory performance. Moreover, even memories expressed with high confidence were less accurate under stress, and this stress-induced decline in accuracy was explained by reduced posterior hippocampal engagement despite similar levels of category-level cortical reinstatement. Finally, stress degraded the relationship between the engagement of frontoparietal control networks and retrieval decision uncertainty. Collectively, these findings demonstrate the widespread consequences of acute stress on the neural systems of remembering.
Collapse
Affiliation(s)
| | - Michael L Waskom
- Department of Psychology, Stanford University, Stanford, CA, USA.,Center for Neural Science, New York University, New York, NY, USA
| | - Thackery I Brown
- Department of Psychology, Stanford University, Stanford, CA, USA.,School of Psychology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Anthony D Wagner
- Department of Psychology, Stanford University, Stanford, CA, USA.,Stanford Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
136
|
Aru J, Siclari F, Phillips WA, Storm JF. Apical drive-A cellular mechanism of dreaming? Neurosci Biobehav Rev 2020; 119:440-455. [PMID: 33002561 DOI: 10.1016/j.neubiorev.2020.09.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 11/17/2022]
Abstract
Dreams are internally generated experiences that occur independently of current sensory input. Here we argue, based on cortical anatomy and function, that dream experiences are tightly related to the workings of a specific part of cortical pyramidal neurons, the apical integration zone (AIZ). The AIZ receives and processes contextual information from diverse sources and could constitute a major switch point for transitioning from externally to internally generated experiences such as dreams. We propose that during dreams the output of certain pyramidal neurons is mainly driven by input into the AIZ. We call this mode of functioning "apical drive". Our hypothesis is based on the evidence that the cholinergic and adrenergic arousal systems, which show different dynamics between waking, slow wave sleep, and rapid eye movement sleep, have specific effects on the AIZ. We suggest that apical drive may also contribute to waking experiences, such as mental imagery. Future studies, investigating the different modes of apical function and their regulation during sleep and wakefulness are likely to be richly rewarded.
Collapse
Affiliation(s)
- Jaan Aru
- Institute of Computer Science, University of Tartu, Estonia; Institute of Biology, Humboldt University Berlin, Germany.
| | - Francesca Siclari
- Center for Investigation and Research on Sleep, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Faculty of Natural Sciences, Psychology, University of Stirling, Stirling, United Kingdom.
| | - William A Phillips
- Faculty of Natural Sciences, Psychology, University of Stirling, Stirling, United Kingdom.
| | - Johan F Storm
- Brain Signalling Group, Section for Physiology, Faculty of Medicine, Domus Medica, University of Oslo, PB 1104 Blindern, 0317 Oslo, Norway.
| |
Collapse
|
137
|
Slow electroencephalographic oscillations and behavioral measures as predictors of high executive processing in early postmenopausal females: A discriminant analysis approach. Brain Cogn 2020; 145:105613. [PMID: 32911233 DOI: 10.1016/j.bandc.2020.105613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 08/10/2020] [Accepted: 08/16/2020] [Indexed: 11/22/2022]
Abstract
Decline in cognitive function is frequent in early postmenopause. There are postmenopausal females who show high performance while others display low performance in executive function, modulated by the prefrontal cortex. These differences have led to confusing and inconclusive results, which have not been explained entirely by the decline in estrogens, which affect the prefrontal cortex functions. An analysis of brain function and the application of a discriminant analysis can help to clarify the deficits in executive function shown by some postmenopausal females. The objective was to examine electroencephalographic recording during the performance of an executive function test in early postmenopausal females, ten with a high level of performance and ten with a low level of performance. Absolute power of delta, theta, alpha1, alpha2, beta1 and beta2 and the numbers of completed categories, trials, perseverative errors and overall errors were submitted to stepwise discriminant analysis to identify predictor variables. Four predictors emerged as significant of group membership based on cognitive performance, with the high-performance group characterized by more completed categories, more delta power, less theta power and more alpha1 power. These findings suggest that postmenopausal females classified in the high-performance group displayed appropriate temporary activation in slow oscillations during executive processing.
Collapse
|
138
|
Shanmugan S, Cao W, Satterthwaite TD, Sammel MD, Ashourvan A, Bassett DS, Ruparel K, Gur RC, Epperson CN, Loughead J. Impact of childhood adversity on network reconfiguration dynamics during working memory in hypogonadal women. Psychoneuroendocrinology 2020; 119:104710. [PMID: 32563173 PMCID: PMC7745207 DOI: 10.1016/j.psyneuen.2020.104710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/25/2022]
Abstract
Many women with no history of cognitive difficulties experience executive dysfunction during menopause. Significant adversity during childhood negatively impacts executive function into adulthood and may be an indicator of women at risk of a mid-life cognitive decline. Previous studies have indicated that alterations in functional network connectivity underlie these negative effects of childhood adversity. There is growing evidence that functional brain networks are not static during executive tasks; instead, such networks reconfigure over time. Optimal dynamics are necessary for efficient executive function; while too little reconfiguration is insufficient for peak performance, too much reconfiguration (supra-optimal reconfiguration) is also maladaptive and associated with poorer performance. Here we examined the impact of adverse childhood experiences (ACEs) on network flexibility, a measure of dynamic reconfiguration, during a letter n-back task within three networks that support executive function: frontoparietal, salience, and default mode networks. Several animal and human subject studies have suggested that childhood adversity exerts lasting effects on executive function via serotonergic mechanisms. Tryptophan depletion (TD) was used to examine whether serotonin function drives ACE effects on network flexibility. We hypothesized that ACE would be associated with higher flexibility (supra-optimal flexibility) and that TD would further increase this measure. Forty women underwent functional imaging at two time points in this double-blind, placebo controlled, crossover study. Participants also completed the Penn Conditional Exclusion Test, a task assessing abstraction and mental flexibility. The effects of ACE and TD were evaluated using generalized estimating equations. ACE was associated with higher flexibility across networks (frontoparietal β = 0.00748, D = 2.79, p = 0.005; salience β = 0.00679, D = 3.02, p = 0.003; and default mode β = 0.00910, D = 3.53, p = 0.0004). While there was no interaction between ACE and TD, active TD increased network flexibility in both ACE groups in comparison to sham depletion (frontoparietal β = 0.00489, D = 2.15, p = 0.03; salience β = 0.00393, D = 1.91, p = 0.06; default mode β = 0.00334, D = 1.73, p = 0.08). These results suggest that childhood adversity has lasting impacts on dynamic reconfiguration of functional brain networks supporting executive function and that decreasing serotonin levels may exacerbate these effects.
Collapse
Affiliation(s)
- Sheila Shanmugan
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Penn PROMOTES Research on Sex and Gender in Health, University of Pennsylvania, Philadelphia, PA, USA.
| | - Wen Cao
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Theodore D Satterthwaite
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mary D Sammel
- Penn PROMOTES Research on Sex and Gender in Health, University of Pennsylvania, Philadelphia, PA, USA; Department of Biostatistics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Arian Ashourvan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Danielle S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA; Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Kosha Ruparel
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben C Gur
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - C Neill Epperson
- Department of Psychiatry, Anschutz Medical Campus, University of Colorado, Aurora, CO USA
| | - James Loughead
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
139
|
Kim JI, Kim JW, Park S, Hong SB, Lee DS, Paek SH, Han DH, Cheong JH, Kim BN. The GRIN2B and GRIN2A Gene Variants Are Associated With Continuous Performance Test Variables in ADHD. J Atten Disord 2020; 24:1538-1546. [PMID: 27199241 DOI: 10.1177/1087054716649665] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Objective: To examine the association between variants of N-methyl-D-aspartate (NMDA) receptor subunit-encoding genes (GRIN2A and GRIN2B) and continuous performance test (CPT) variables in ADHD and healthy controls. Method: In all, 253 ADHD patients and 98 controls were recruited. The diagnosis, genotype, and diagnosis-genotype interaction effects for the CPT variables were examined. Results: Significant diagnosis effects were detected for all CPT variables. There were significant genotype and interaction effects on response time variability (RTV) by the GRIN2B variant. The C/C subgroup had higher RTV than the C/T + T/T subgroup in ADHD, but not in controls. There were significant genotype effects on omission errors by the GRIN2A variant. The G/G subgroup had more omission errors than the G/A + A/A subgroup in ADHD patients, but not in controls. Conclusion: These results suggest that the genetic variants of GRIN2B and GRIN2A confer an increased susceptibility to attentional impairment in ADHD patients.
Collapse
Affiliation(s)
- Johanna Inhyang Kim
- Seoul National University College of Medicine, Chongno-gu, Republic of Korea
| | - Jae-Won Kim
- Seoul National University College of Medicine, Chongno-gu, Republic of Korea
| | - Subin Park
- National Center for Mental Health, Kwangjin-ju, Republic of Korea
| | - Soon-Beom Hong
- Seoul National University College of Medicine, Chongno-gu, Republic of Korea
| | - Dong Soo Lee
- Seoul National University College of Medicine, Chongno-gu, Republic of Korea
| | - Sun Ha Paek
- Seoul National University College of Medicine, Chongno-gu, Republic of Korea
| | - Doug Hyun Han
- Chung Ang University, Dongjak-gu, Seoul, Republic of Korea
| | | | - Bung-Nyun Kim
- Seoul National University College of Medicine, Chongno-gu, Republic of Korea
| |
Collapse
|
140
|
Keshmiri S. Stress Changes the Resting-State Cortical Flow of Information from Distributed to Frontally Directed Patterns. BIOLOGY 2020; 9:E236. [PMID: 32824879 PMCID: PMC7464349 DOI: 10.3390/biology9080236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 11/16/2022]
Abstract
Despite converging evidence on the involvement of large-scale distributed brain networks in response to stress, the effect of stress on the components of these networks is less clear. Although some studies identify higher regional activities in response to stress, others observe an opposite effect in the similar regions. Studies based on synchronized activities and coactivation of these components also yield similar differing results. However, these differences are not necessarily contradictory once we observe the effect of stress on these functional networks in terms of the change in information processing capacity of their components. In the present study, we investigate the utility of such a shift in the analysis of the effect of stress on distributed cortical regions through quantification of the flow of information among them. For this purpose, we use the self-assessed responses of 216 individuals to stress-related questionnaires and systematically select 20 of them whose responses showed significantly higher and lower susceptibility to stress. We then use these 20 individuals' resting-state multi-channel electroencephalography (EEG) recordings (both Eyes-Closed (EC) and Eyes-Open (EO) settings) and compute the distributed flow of information among their cortical regions using transfer entropy (TE). The contribution of the present study is three-fold. First, it identifies that the stress-susceptibility is characterized by the change in flow of information in fronto-parietal brain network. Second, it shows that these regions are distributed bi-hemispherically and are sufficient to significantly differentiate between the individuals with high versus low stress-susceptibility. Third, it verifies that the high stress-susceptibility is markedly associated with a higher parietal-to-frontal flow of information. These results provide further evidence for the viewpoint in which the brain's modulation of information is not necessarily accompanied by the change in its regional activity. They further construe the effect of stress in terms of a disturbance that disrupts the flow of information among the brain's distributed cortical regions. These observations, in turn, suggest that some of the differences in the previous findings perhaps reflect different aspects of impaired distributed brain information processing in response to stress. From a broader perspective, these results posit the use of TE as a potential diagnostic/prognostic tool in identification of the effect of stress on distributed brain networks that are involved in stress-response.
Collapse
Affiliation(s)
- Soheil Keshmiri
- The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International (ATR), Kyoto 619-0237, Japan
| |
Collapse
|
141
|
Mäki-Marttunen V, Andreassen OA, Espeseth T. The role of norepinephrine in the pathophysiology of schizophrenia. Neurosci Biobehav Rev 2020; 118:298-314. [PMID: 32768486 DOI: 10.1016/j.neubiorev.2020.07.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 07/01/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Several lines of evidence have suggested for decades a role for norepinephrine (NE) in the pathophysiology and treatment of schizophrenia. Recent experimental findings reveal anatomical and physiological properties of the locus coeruleus-norepinephrine (LC-NE) system and its involvement in brain function and cognition. Here, we integrate these two lines of evidence. First, we review the functional and structural properties of the LC-NE system and its impact on functional brain networks, cognition, and stress, with special emphasis on recent experimental and theoretical advances. Subsequently, we present an update about the role of LC-associated functions for the pathophysiology of schizophrenia, focusing on the cognitive and motivational deficits. We propose that schizophrenia phenomenology, in particular cognitive symptoms, may be explained by an abnormal interaction between genetic susceptibility and stress-initiated LC-NE dysfunction. This in turn, leads to imbalance between LC activity modes, dysfunctional regulation of brain network integration and neural gain, and deficits in cognitive functions. Finally, we suggest how recent development of experimental approaches can be used to characterize LC function in schizophrenia.
Collapse
Affiliation(s)
| | - Ole A Andreassen
- CoE NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Building 49, P.O. Box 4956 Nydalen, N-0424 Oslo, Norway
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Postboks 1094, Blindern, 0317 Oslo, Norway; Bjørknes College, Lovisenberggata 13, 0456 Oslo, Norway
| |
Collapse
|
142
|
Moraes NC, Muela HCS, MemÓria CM, Costa-Hong VAD, Machado MF, Cechinhi MA, Nitrini R, Bortolotto LA, Yassuda MS. Systemic arterial hypertension and cognition in adults: effects on executive functioning. ARQUIVOS DE NEURO-PSIQUIATRIA 2020; 78:412-418. [PMID: 32627812 DOI: 10.1590/0004-282x20200039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Central nervous system changes associated to systemic arterial hypertension (SAH) are progressive and may cause negative effects on cognitive performance. The objective of this study was to investigate the relation between SAH and the components of executive functions (EF), inhibitory control (IC), updating and shifting, comparing a control group (without SAH) to patients with SAH, in two levels of severity. METHODS The protocol included the following tests to evaluate EF components: T.O.V.A. Test (IC), Backward Digit Span from Wechsler Adults Intelligence Scale (WAIS-III), Phonemic and Semantic Verbal Fluency (updating), and Trail Making Test Part B (shifting). RESULTS A total of 204 participants was included: 56 from the Control Group (CG), 87 SAH stage 1, and 61 SAH stage 2. The groups were not different for age (52.37±12.29) and education (10.98±4.06). As to controlled blood pressure (BP), duration of hypertension treatment and number of drugs, the SAH 2 group had a worse BP control, longer duration of hypertension treatment and use of more drugs when compared to the SAH 1. The findings revealed that patients with more severe hypertension presented worse performance in updating (Backward Digit Span, phonemic and semantics VF) and shifting (Trail Making Test Part B). CONCLUSION The results suggest that patients with SAH have a significant impairment in EF, more specifically in updating and shifting. Besides that, such damage may be directly proportional to the severity of SAH. It is suggested that future studies include neuroimaging exams to exclude possible cerebrovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ricardo Nitrini
- Department of Neurology, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | |
Collapse
|
143
|
Yahaya R, Zahary MN, Othman Z, Ismail R, Nik Him NAS, Abd Aziz A, Dahlan R, Jusoh AF. Tualang honey supplementation as cognitive enhancer in patients with schizophrenia. Heliyon 2020; 6:e03948. [PMID: 32426546 PMCID: PMC7226648 DOI: 10.1016/j.heliyon.2020.e03948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/12/2020] [Accepted: 05/05/2020] [Indexed: 01/31/2023] Open
Abstract
Introduction Schizophrenia is a chronic mental illness with clusters of symptoms, including cognitive impairment. This study aimed to explore the effect of Tualang Honey (TH) on cognitive domains, especially as it pertained to the verbal memory of schizophrenia patients. Method This was a cross-sectional study involved 80 individuals, diagnosed with schizophrenia. The Malay Version Auditory Verbal Learning Test (MVAVLT) was used. Data were analysed using SPSS 20.0 software. Intention to treat analysis was applied. Result A comparison of the total learning score at eight weeks between the two groups based on time effect and time-treatment interaction favoured TH group. Conclusion This study concludes that by supplementing schizophrenia patients with 8-week of TH did improve total learning performance across domains in the immediate memory among patients with schizophrenia.
Collapse
Affiliation(s)
- Rosliza Yahaya
- Faculty of Medicine, Medical Campus, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
| | - Mohd Nizam Zahary
- Faculty of Health Sciences, Gong Badak Campus, Universiti Sultan Zainal Abidin, 21300 Kuala Nerus, Terengganu, Malaysia
| | - Zahiruddin Othman
- School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Rusli Ismail
- Faculty of Medicine, Medical Campus, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
| | - Nik Ahmad Shaiffudin Nik Him
- Faculty of Medicine, Medical Campus, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
| | - Aniza Abd Aziz
- Faculty of Medicine, Medical Campus, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
| | - Rahima Dahlan
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Azizul Fadzli Jusoh
- Faculty of Medicine, Medical Campus, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia
- Corresponding author.
| |
Collapse
|
144
|
Ma Q, Tang Y, Wang F, Liao X, Jiang X, Wei S, Mechelli A, He Y, Xia M. Transdiagnostic Dysfunctions in Brain Modules Across Patients with Schizophrenia, Bipolar Disorder, and Major Depressive Disorder: A Connectome-Based Study. Schizophr Bull 2020; 46:699-712. [PMID: 31755957 PMCID: PMC7147584 DOI: 10.1093/schbul/sbz111] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Psychiatric disorders, including schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD), share clinical and neurobiological features. Because previous investigations of functional dysconnectivity have mainly focused on single disorders, the transdiagnostic alterations in the functional connectome architecture of the brain remain poorly understood. We collected resting-state functional magnetic resonance imaging data from 512 participants, including 121 with SCZ, 100 with BD, 108 with MDD, and 183 healthy controls. Individual functional brain connectomes were constructed in a voxelwise manner, and the modular architectures were examined at different scales, including (1) global modularity, (2) module-specific segregation and intra- and intermodular connections, and (3) nodal participation coefficients. The correlation of these modular measures with clinical scores was also examined. We reliably identify common alterations in modular organization in patients compared to controls, including (1) lower global modularity; (2) lower modular segregation in the frontoparietal, subcortical, visual, and sensorimotor modules driven by more intermodular connections; and (3) higher participation coefficients in several network connectors (the dorsolateral prefrontal cortex and angular gyrus) and the thalamus. Furthermore, the alterations in the SCZ group are more widespread than those of the BD and MDD groups and involve more intermodular connections, lower modular segregation and higher connector integrity. These alterations in modular organization significantly correlate with clinical scores in patients. This study demonstrates common hyper-integrated modular architectures of functional brain networks among patients with SCZ, BD, and MDD. These findings reveal a transdiagnostic mechanism of network dysfunction across psychiatric disorders from a connectomic perspective.
Collapse
Affiliation(s)
- Qing Ma
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuhong Liao
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Xiaowei Jiang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shengnan Wei
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Andrea Mechelli
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Yong He
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Mingrui Xia
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
145
|
Dehais F, Lafont A, Roy R, Fairclough S. A Neuroergonomics Approach to Mental Workload, Engagement and Human Performance. Front Neurosci 2020; 14:268. [PMID: 32317914 PMCID: PMC7154497 DOI: 10.3389/fnins.2020.00268] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
The assessment and prediction of cognitive performance is a key issue for any discipline concerned with human operators in the context of safety-critical behavior. Most of the research has focused on the measurement of mental workload but this construct remains difficult to operationalize despite decades of research on the topic. Recent advances in Neuroergonomics have expanded our understanding of neurocognitive processes across different operational domains. We provide a framework to disentangle those neural mechanisms that underpin the relationship between task demand, arousal, mental workload and human performance. This approach advocates targeting those specific mental states that precede a reduction of performance efficacy. A number of undesirable neurocognitive states (mind wandering, effort withdrawal, perseveration, inattentional phenomena) are identified and mapped within a two-dimensional conceptual space encompassing task engagement and arousal. We argue that monitoring the prefrontal cortex and its deactivation can index a generic shift from a nominal operational state to an impaired one where performance is likely to degrade. Neurophysiological, physiological and behavioral markers that specifically account for these states are identified. We then propose a typology of neuroadaptive countermeasures to mitigate these undesirable mental states.
Collapse
Affiliation(s)
- Frédéric Dehais
- ISAE-SUPAERO, Université de Toulouse, Toulouse, France
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - Alex Lafont
- ISAE-SUPAERO, Université de Toulouse, Toulouse, France
| | - Raphaëlle Roy
- ISAE-SUPAERO, Université de Toulouse, Toulouse, France
| | - Stephen Fairclough
- School of Psychology, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
146
|
Abstract
Neural activity and behavior are both notoriously variable, with responses differing widely between repeated presentation of identical stimuli or trials. Recent results in humans and animals reveal that these variations are not random in their nature, but may in fact be due in large part to rapid shifts in neural, cognitive, and behavioral states. Here we review recent advances in the understanding of rapid variations in the waking state, how variations are generated, and how they modulate neural and behavioral responses in both mice and humans. We propose that the brain has an identifiable set of states through which it wanders continuously in a nonrandom fashion, owing to the activity of both ascending modulatory and fast-acting corticocortical and subcortical-cortical neural pathways. These state variations provide the backdrop upon which the brain operates, and understanding them is critical to making progress in revealing the neural mechanisms underlying cognition and behavior.
Collapse
Affiliation(s)
- David A McCormick
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403, USA;
| | - Dennis B Nestvogel
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403, USA;
| | - Biyu J He
- Departments of Neurology, Neuroscience and Physiology, and Radiology, Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
147
|
Agrawal P, Kao D, Chung P, Looger LL. The neuropeptide Drosulfakinin regulates social isolation-induced aggression in Drosophila. J Exp Biol 2020; 223:jeb207407. [PMID: 31900346 PMCID: PMC7033730 DOI: 10.1242/jeb.207407] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/19/2019] [Indexed: 01/09/2023]
Abstract
Social isolation strongly modulates behavior across the animal kingdom. We utilized the fruit fly Drosophila melanogaster to study social isolation-driven changes in animal behavior and gene expression in the brain. RNA-seq identified several head-expressed genes strongly responding to social isolation or enrichment. Of particular interest, social isolation downregulated expression of the gene encoding the neuropeptide Drosulfakinin (Dsk), the homologue of vertebrate cholecystokinin (CCK), which is critical for many mammalian social behaviors. Dsk knockdown significantly increased social isolation-induced aggression. Genetic activation or silencing of Dsk neurons each similarly increased isolation-driven aggression. Our results suggest a U-shaped dependence of social isolation-induced aggressive behavior on Dsk signaling, similar to the actions of many neuromodulators in other contexts.
Collapse
Affiliation(s)
- Pavan Agrawal
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Damian Kao
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Phuong Chung
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| |
Collapse
|
148
|
Ranjbar-Slamloo Y, Fazlali Z. Dopamine and Noradrenaline in the Brain; Overlapping or Dissociate Functions? Front Mol Neurosci 2020; 12:334. [PMID: 32038164 PMCID: PMC6986277 DOI: 10.3389/fnmol.2019.00334] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/27/2019] [Indexed: 12/21/2022] Open
Abstract
Dopamine and noradrenaline are crucial neuromodulators controlling brain states, vigilance, action, reward, learning, and memory processes. Ventral tegmental area (VTA) and Locus Coeruleus (LC) are canonically described as the main sources of dopamine (DA) and noradrenaline (NA) with dissociate functions. A comparison of diverse studies shows that these neuromodulators largely overlap in multiple domains such as shared biosynthetic pathway and co-release from the LC terminals, convergent innervations, non-specificity of receptors and transporters, and shared intracellular signaling pathways. DA–NA interactions are mainly studied in prefrontal cortex and hippocampus, yet it can be extended to the whole brain given the diversity of catecholamine innervations. LC can simultaneously broadcast both dopamine and noradrenaline across the brain. Here, we briefly review the molecular, cellular, and physiological overlaps between DA and NA systems and point to their functional implications. We suggest that DA and NA may function in parallel to facilitate learning and maintain the states required for normal cognitive processes. Various signaling modules of NA and DA have been targeted for developing of therapeutics. Understanding overlaps of the two systems is crucial for more effective interventions in a range of neuropsychiatric conditions.
Collapse
Affiliation(s)
- Yadollah Ranjbar-Slamloo
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Zeinab Fazlali
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| |
Collapse
|
149
|
Ibañez S, Luebke JI, Chang W, Draguljić D, Weaver CM. Network Models Predict That Pyramidal Neuron Hyperexcitability and Synapse Loss in the dlPFC Lead to Age-Related Spatial Working Memory Impairment in Rhesus Monkeys. Front Comput Neurosci 2020; 13:89. [PMID: 32009920 PMCID: PMC6979278 DOI: 10.3389/fncom.2019.00089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/18/2019] [Indexed: 01/04/2023] Open
Abstract
Behavioral studies have shown spatial working memory impairment with aging in several animal species, including humans. Persistent activity of layer 3 pyramidal dorsolateral prefrontal cortex (dlPFC) neurons during delay periods of working memory tasks is important for encoding memory of the stimulus. In vitro studies have shown that these neurons undergo significant age-related structural and functional changes, but the extent to which these changes affect neural mechanisms underlying spatial working memory is not understood fully. Here, we confirm previous studies showing impairment on the Delayed Recognition Span Task in the spatial condition (DRSTsp), and increased in vitro action potential firing rates (hyperexcitability), across the adult life span of the rhesus monkey. We use a bump attractor model to predict how empirically observed changes in the aging dlPFC affect performance on the Delayed Response Task (DRT), and introduce a model of memory retention in the DRSTsp. Persistent activity-and, in turn, cognitive performance-in both models was affected much more by hyperexcitability of pyramidal neurons than by a loss of synapses. Our DRT simulations predict that additional changes to the network, such as increased firing of inhibitory interneurons, are needed to account for lower firing rates during the DRT with aging reported in vivo. Synaptic facilitation was an essential feature of the DRSTsp model, but it did not compensate fully for the effects of the other age-related changes on DRT performance. Modeling pyramidal neuron hyperexcitability and synapse loss simultaneously led to a partial recovery of function in both tasks, with the simulated level of DRSTsp impairment similar to that observed in aging monkeys. This modeling work integrates empirical data across multiple scales, from synapse counts to cognitive testing, to further our understanding of aging in non-human primates.
Collapse
Affiliation(s)
- Sara Ibañez
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Jennifer I. Luebke
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Wayne Chang
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Danel Draguljić
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
| | - Christina M. Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, United States
| |
Collapse
|
150
|
Blockage of NMDA- and GABA(A) Receptors Improves Working Memory Selectivity of Primate Prefrontal Neurons. J Neurosci 2020; 40:1527-1537. [PMID: 31911457 DOI: 10.1523/jneurosci.2009-19.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022] Open
Abstract
The ongoing activity of prefrontal neurons after a stimulus has disappeared is considered a neuronal correlate of working memory. It depends on the delicate but poorly understood interplay between excitatory glutamatergic and inhibitory GABAergic receptor effects. We administered the NMDA receptor antagonist MK-801 and the GABA(A) receptor antagonist bicuculline methiodide while recording cellular activity in PFC of male rhesus monkeys performing a delayed decision task requiring working memory. The blockade of GABA(A) receptors strongly improved the selectivity of the neurons' delay activity, causing an increase in signal-to-noise ratio during working memory periods as well as an enhancement of the neurons' coding selectivity. The blockade of NMDA receptors resulted in a slight enhancement of selectivity and encoding capacity of the neurons. Our findings emphasize the delicate and more complex than expected interplay of excitatory and inhibitory transmitter systems in modulating working memory coding in prefrontal circuits.SIGNIFICANCE STATEMENT Ongoing delay activity of prefrontal neurons constitutes a neuronal correlate of working memory. However, how this delay activity is generated by the delicate interplay of synaptic excitation and inhibition is unknown. We probed the effects of excitatory neurotransmitter glutamate and inhibitory neurotransmitter GABA in regulating delay activity in rhesus monkeys performing a delayed decision task requiring working memory. Surprisingly, the blockade of both glutamatergic NMDA and GABA(A) receptors improved neuronal selectivity of delay activity, causing an increase in neuronal signal-to-noise ratio. Moreover, individual neurons were similarly affected by blockade of both receptors. This emphasizes the delicate and more complex than expected interplay of excitatory and inhibitory transmitter systems in modulating working memory coding in prefrontal circuits.
Collapse
|