1
|
Trent S, Abdullah MH, Parwana K, Valdivieso MA, Hassan Z, Müller CP. Fear conditioning: Insights into learning, memory and extinction and its relevance to clinical disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111310. [PMID: 40056965 DOI: 10.1016/j.pnpbp.2025.111310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
Fear, whether innate or learned, is an essential emotion required for survival. The learning, and subsequent memory, of fearful events enhances our ability to recognise and respond to threats, aiding adaptation to new, ever-changing environments. Considerable research has leveraged associative learning protocols such as contextual or auditory forms of fear conditioning in rodents, to understand fear learning, memory consolidation and extinction phases of memory. Such assays have led to detailed characterisation of the underlying neurocircuitry and neurobiology supporting fear learning processes. Given fear processing is conserved across rodents and humans, fear conditioning experiments provide translational insights into fundamental memory processes and fear-related pathologies. This review examines associative learning protocols used to measure fear learning, memory and extinction, before providing an overview on the underlying complex neurocircuitry including the amygdala, hippocampus and medial prefrontal cortex. This is followed by an in-depth commentary on the neurobiology, particularly synaptic plasticity mechanisms, which regulate fear learning, memory and extinction. Next, we consider how fear conditioning assays in rodents can inform our understanding of disrupted fear memory in human disorders such as post-traumatic stress disorder (PTSD), anxiety and psychiatric disorders including schizophrenia. Lastly, we critically evaluate fear conditioning protocols, highlighting some of the experimental and theoretical limitations and the considerations required when conducting such assays, alongside recent methodological advancements in the field. Overall, rodent-based fear conditioning assays remain central to making progress in uncovering fundamental memory phenomena and understanding the aetiological mechanisms that underpin fear associated disorders, alongside the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Simon Trent
- School of Life Sciences, Faculty of Natural Sciences, Huxley Building, Keele University, Keele ST5 5BG, UK.
| | | | - Krishma Parwana
- School of Life Sciences, Faculty of Natural Sciences, Huxley Building, Keele University, Keele ST5 5BG, UK
| | - Maria Alcocer Valdivieso
- School of Life Sciences, Faculty of Natural Sciences, Huxley Building, Keele University, Keele ST5 5BG, UK
| | - Zurina Hassan
- Centre for Drug Research, Universiti Malaysia (USM), 11800 Penang, Malaysia
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University of Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany; Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
2
|
McClarty BM, Rodriguez G, Dong H. Class 1 histone deacetylases differentially modulate memory and synaptic genes in a spatial and temporal manner in aged and APP/PS1 mice. Brain Res 2024; 1837:148951. [PMID: 38642789 PMCID: PMC11182336 DOI: 10.1016/j.brainres.2024.148951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Epigenetics plays a vital role in aging and Alzheimer's disease (AD); however, whether epigenetic alterations during aging can initiate AD and exacerbate AD progression remains unclear. In this study, using 3-, 12- and 18- month-old APP/PS1 mice and age matched WT littermates, we conducted a series of memory tests, measured synapse-related gene expression, class 1 histone deacetylases (HDACs) abundance, and H3K9ac levels at target gene promoters in the hippocampus and prefrontal cortex (PFC). Our results showed impaired recognition and long-term spatial memory in 18-month-old WT mice and impaired recognition, short-term working, and long-term spatial reference memory in 12-and 18- month-old APP/PS1 mice. These memory impairments are associated with changes of synapse-related gene (nr2a, glur1, glur2, psd95) expression, HDAC abundance, and H3K9ac levels; more specifically, increased HDAC2 was associated with synapse-related gene expression changes through modulation of H3K9ac at the gene promoters during aging and AD progression in the hippocampus. Conversely, increased HDAC3 was associated with synapse-related gene expression changes through modulation of H3K9ac at the gene promoters during AD progression in the PFC. These findings suggest memory impairments in aging and AD may associated with a differential HDAC modulation of synapse-related gene expression in the brain.
Collapse
Affiliation(s)
- Bryan M McClarty
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Ward 7-103, Chicago, IL 60611, USA.
| |
Collapse
|
3
|
Richard SA. Elucidating the pivotal molecular mechanisms, therapeutic and neuroprotective effects of lithium in traumatic brain injury. Brain Behav 2024; 14:e3595. [PMID: 38874089 PMCID: PMC11177180 DOI: 10.1002/brb3.3595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Traumatic brain injury (TBI) refers to damage to brain tissue by mechanical or blunt force via trauma. TBI is often associated with impaired cognitive abilities, like difficulties in memory, learning, attention, and other higher brain functions, that typically remain for years after the injury. Lithium is an elementary light metal that is only utilized in salt form due to its high intrinsic reactivity. This current review discusses the molecular mechanisms and therapeutic and neuroprotective effects of lithium in TBI. METHOD The "Boolean logic" was used to search for articles on the subject matter in PubMed and PubMed Central, as well as Google Scholar. RESULTS Lithium's therapeutic action is extremely complex, involving multiple effects on gene secretion, neurotransmitter or receptor-mediated signaling, signal transduction processes, circadian modulation, as well as ion transport. Lithium is able to normalize multiple short- as well as long-term modifications in neuronal circuits that ultimately result in disparity in cortical excitation and inhibition activated by TBI. Also, lithium levels are more distinct in the hippocampus, thalamus, neo-cortex, olfactory bulb, amygdala as well as the gray matter of the cerebellum following treatment of TBI. CONCLUSION Lithium attenuates neuroinflammation and neuronal toxicity as well as protects the brain from edema, hippocampal neurodegeneration, loss of hemispheric tissues, and enhanced memory as well as spatial learning after TBI.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Medicine, Princefield University, Ho, Ghana
- Institute of Neuroscience, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Yunusa S, Hassan Z, Müller CP. Mitragynine inhibits hippocampus neuroplasticity and its molecular mechanism. Pharmacol Rep 2023; 75:1488-1501. [PMID: 37924443 PMCID: PMC10661785 DOI: 10.1007/s43440-023-00541-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Mitragynine (MIT), the primary indole alkaloid of kratom (Mitragyna speciosa), has been associated with addictive and cognitive decline potentials. In acute studies, MIT decreases spatial memory and inhibits hippocampal synaptic transmission in long-term potentiation (LTP). This study investigated the impacts of 14-day MIT treatment on hippocampus synaptic transmission and its possible underlying mechanisms. METHODS Under urethane anesthesia, field excitatory post-synaptic potentials (fEPSP) of the hippocampal CA1 region were recorded in the Sprague Dawley (SD) rats that received MIT (1, 5, and 10 mg/kg), morphine (MOR) 5 mg/kg, or vehicle (ip). The effects of the treatments on basal synaptic transmission, paired-pulse facilitation (PPF), and LTP were assessed in the CA1 region. Analysis of the brain's protein expression linked to neuroplasticity was then performed using a western blot. RESULTS The baseline synaptic transmission's amplitude was drastically decreased by MIT at 5 and 10 mg/kg doses, although the PPF ratio before TBS remained unchanged, the PPF ratio after TBS was significantly reduced by MIT (10 mg/kg). Strong and persistent inhibition of LTP was generated in the CA1 region by MIT (5 and 10 mg/kg) doses; this effect was not seen in MIT (1 mg/kg) treated rats. In contrast to MIT (1 mg/kg), MIT (5 and 10 mg/kg) significantly raised the extracellular glutamate levels. After exposure to MIT, GluR-1 receptor expression remained unaltered. However, NMDAε2 receptor expression was markedly downregulated. The expression of pCaMKII, pERK, pCREB, BDNF, synaptophysin, PSD-95, Delta fosB, and CDK-5 was significantly downregulated in response to MIT (5 and 10 mg/kg) exposure, while MOR (5 mg/kg) significantly raised synaptophysin and Delta fosB expression. CONCLUSION Findings from this work reveal that a smaller dose of MIT (1 mg/kg) poses no risk to hippocampal synaptic transmission. Alteration in neuroplasticity-associated proteins may be a molecular mechanism for MIT (5 and 10 mg/kg)-induced LTP disruption and cognitive impairments. Data from this work posit that MIT acted differently from MOR on neuroplasticity and its underlying mechanisms.
Collapse
Affiliation(s)
- Suleiman Yunusa
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Penang, Malaysia
- Department of Pharmacology, Bauchi State University Gadau, PMB 65 Itas/Gadau, Bauchi, Bauchi State, Nigeria
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Christian P Müller
- Centre for Drug Research, Universiti Sains Malaysia, 11800, Penang, Malaysia.
- Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany.
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine Mannheim, University of Heidelberg, Heidelberg, Germany.
- Psychiatric and Psychotherapeutic University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| |
Collapse
|
5
|
Sepahvand T, Power KD, Qin T, Yuan Q. The Basolateral Amygdala: The Core of a Network for Threat Conditioning, Extinction, and Second-Order Threat Conditioning. BIOLOGY 2023; 12:1274. [PMID: 37886984 PMCID: PMC10604397 DOI: 10.3390/biology12101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
Threat conditioning, extinction, and second-order threat conditioning studied in animal models provide insight into the brain-based mechanisms of fear- and anxiety-related disorders and their treatment. Much attention has been paid to the role of the basolateral amygdala (BLA) in such processes, an overview of which is presented in this review. More recent evidence suggests that the BLA serves as the core of a greater network of structures in these forms of learning, including associative and sensory cortices. The BLA is importantly regulated by hippocampal and prefrontal inputs, as well as by the catecholaminergic neuromodulators, norepinephrine and dopamine, that may provide important prediction-error or learning signals for these forms of learning. The sensory cortices may be required for the long-term storage of threat memories. As such, future research may further investigate the potential of the sensory cortices for the long-term storage of extinction and second-order conditioning memories.
Collapse
Affiliation(s)
| | | | | | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University, St John’s, NL A1B 3V6, Canada; (T.S.); (K.D.P.); (T.Q.)
| |
Collapse
|
6
|
Choopani S, Kiani B, Aliakbari S, Babaie J, Golkar M, Pourbadie HG, Sayyah M. Latent toxoplasmosis impairs learning and memory yet strengthens short-term and long-term hippocampal synaptic plasticity at perforant pathway-dentate gyrus, and Schaffer collatterals-CA1 synapses. Sci Rep 2023; 13:8959. [PMID: 37268701 DOI: 10.1038/s41598-023-35971-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023] Open
Abstract
Investigating long-term potentiation (LTP) in disease models provides essential mechanistic insight into synaptic dysfunction and relevant behavioral changes in many neuropsychiatric and neurological diseases. Toxoplasma (T) gondii is an intracellular parasite causing bizarre changes in host's mind including losing inherent fear of life-threatening situations. We examined hippocampal-dependent behavior as well as in vivo short- and long-term synaptic plasticity (STP and LTP) in rats with latent toxoplasmosis. Rats were infected by T. gondii cysts. Existence of REP-529 genomic sequence of the parasite in the brain was detected by RT-qPCR. Four and eight weeks after infection, spatial, and inhibitory memories of rats were assessed by Morris water maze and shuttle box tests, respectively. Eight weeks after infection, STP was assessed in dentate gyrus (DG) and CA1 by double pulse stimulation of perforant pathway and Shaffer collaterals, respectively. High frequency stimulation (HFS) was applied to induce LTP in entorhinal cortex-DG (400 Hz), and CA3-CA1 (200 Hz) synapses. T. gondii infection retarded spatial learning and memory performance at eight weeks post-infection period, whereas inhibitory memory was not changed. Unlike uninfected rats that normally showed paired-pulse depression, the infected rats developed paired-pulse facilitation, indicating an inhibitory synaptic network disruption. T. gondii-infected rats displayed strengthened LTP of both CA1-pyramidal and DG-granule cell population spikes. These data indicate that T. gondii disrupts inhibition/excitation balance and causes bizarre changes to the post-synaptic neuronal excitability, which may ultimately contribute to the abnormal behavior of the infected host.
Collapse
Affiliation(s)
- Samira Choopani
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Bahereh Kiani
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
- Department of Biology, Damghan University, Damghan, Iran
| | - Shayan Aliakbari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Jalal Babaie
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Majid Golkar
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
Abstract
Neuroplasticity and evolutionary biology have been prominent fields of study for well over a century. However, they have advanced largely independently, without consideration of the benefits of integration. We propose a new framework by which researchers can begin to examine the evolutionary causes and consequences of neuroplasticity. Neuroplasticity can be defined as changes to the structure, function or connections of the nervous system in response to individual experience. Evolution can alter levels of neuroplasticity if there is variation in neuroplasticity traits within and between populations. Neuroplasticity may be favored or disfavored by natural selection depending on the variability of the environment and the costs of neuroplasticity. Additionally, neuroplasticity may affect rates of genetic evolution in many ways: for example, decreasing rates of evolution by buffering against selection or increasing them via the Baldwin effect, by increasing genetic variation or by incorporating evolved peripheral changes to the nervous system. These mechanisms can be tested using comparative and experimental approaches and by examining patterns and consequences of variation in neuroplasticity among species, populations and individuals.
Collapse
Affiliation(s)
- Caleb J Axelrod
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853, USA
| | - Swanne P Gordon
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY 14853, USA
| | - Bruce A Carlson
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
8
|
Dong TN, Kramár EA, Beardwood JH, Al-Shammari A, Wood MA, Keiser AA. Temporal endurance of exercise-induced benefits on hippocampus-dependent memory and synaptic plasticity in female mice. Neurobiol Learn Mem 2022; 194:107658. [PMID: 35811066 PMCID: PMC9901197 DOI: 10.1016/j.nlm.2022.107658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023]
Abstract
Exercise facilitates hippocampal neurogenesis and neuroplasticity that in turn, promotes cognitive function. Our previous studies have demonstrated that in male mice, voluntary exercise enables hippocampus-dependent learning in conditions that are normally subthreshold for long-term memory formation in sedentary animals. Such cognitive enhancement can be maintained long after exercise has ceased and can be re-engaged by a subsequent subthreshold exercise session, suggesting exercise-induced benefits are temporally dynamic. In females, the extent to which the benefits of exercise can be maintained and the mechanisms underlying this maintenance have yet to be defined. Here, we examined the exercise parameters required to initiate and maintain the benefits of exercise in female C57BL/6J mice. Using a subthreshold version of the hippocampus-dependent task called object-location memory (OLM) task, we show that 14d of voluntary exercise enables learning under subthreshold acquisition conditions in female mice. Following the initial exercise, a 7d sedentary delay results in diminished performance, which can be re-facilitated when animals receive 2d of reactivating exercise following the sedentary delay. Assessment of estrous cycle reveals enhanced wheel running activity during the estrus phase relative to the diestrus phase, whereas estrous phase on training or test had no effect on OLM performance. Utilizing the same exercise parameters, we demonstrate that 14d of exercise enhances long-term potentiation (LTP) in the CA1 region of the hippocampus, an effect that persists throughout the sedentary delay and following the reactivating exercise session. Previous studies have proposed exercise-induced BDNF upregulation as the mechanism underlying exercise-mediated benefits on synaptic plasticity and cognition. However, our assessment of hippocampal Bdnf mRNA expression following memory retrieval reveals no difference between exercise conditions and control, suggesting that persistent Bdnf upregulation may not be required for maintenance of exercise-induced benefits. Together, our data indicate that 14d of voluntary exercise can initiate long-lasting benefits on neuroplasticity and cognitive function in female mice, establishing the first evidence on the temporal endurance of exercise-induced benefits in females.
Collapse
Affiliation(s)
- T N Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - E A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - J H Beardwood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A Al-Shammari
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - M A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - A A Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States.
| |
Collapse
|
9
|
Saleki K, Banazadeh M, Saghazadeh A, Rezaei N. Aging, testosterone, and neuroplasticity: friend or foe? Rev Neurosci 2022; 34:247-273. [PMID: 36017670 DOI: 10.1515/revneuro-2022-0033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
Neuroplasticity or neural plasticity implicates the adaptive potential of the brain in response to extrinsic and intrinsic stimuli. The concept has been utilized in different contexts such as injury and neurological disease. Neuroplasticity mechanisms have been classified into neuroregenerative and function-restoring processes. In the context of injury, neuroplasticity has been defined in three post-injury epochs. Testosterone plays a key yet double-edged role in the regulation of several neuroplasticity alterations. Research has shown that testosterone levels are affected by numerous factors such as age, stress, surgical procedures on gonads, and pharmacological treatments. There is an ongoing debate for testosterone replacement therapy (TRT) in aging men; however, TRT is more useful in young individuals with testosterone deficit and more specific subgroups with cognitive dysfunction. Therefore, it is important to pay early attention to testosterone profile and precisely uncover its harms and benefits. In the present review, we discuss the influence of environmental factors, aging, and gender on testosterone-associated alterations in neuroplasticity, as well as the two-sided actions of testosterone in the nervous system. Finally, we provide practical insights for further study of pharmacological treatments for hormonal disorders focusing on restoring neuroplasticity.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,USERN Office, Babol University of Medical Sciences, 47176 47745 Babol, Iran.,Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| | - Mohammad Banazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, 76169 13555 Kerman, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, 14197 33151 Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 14176 13151 Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 14197 33151 Tehran, Iran
| |
Collapse
|
10
|
Reis AL, Hammond JH, Stevanovski I, Arnold JC, McGregor IS, Deveson IW, Gururajan A. Sex-specific transcriptomic and epitranscriptomic signatures of PTSD-like fear acquisition. iScience 2022; 25:104861. [PMID: 36039298 PMCID: PMC9418440 DOI: 10.1016/j.isci.2022.104861] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/03/2022] [Accepted: 07/26/2022] [Indexed: 11/22/2022] Open
Abstract
Our understanding of the molecular pathology of posttraumatic stress disorder (PTSD) is evolving due to advances in sequencing technologies. With the recent emergence of Oxford Nanopore direct RNA-seq (dRNA-seq), it is now also possible to interrogate diverse RNA modifications, collectively known as the “epitranscriptome.”. Here, we present our analyses of the male and female mouse amygdala transcriptome and epitranscriptome, obtained using parallel Illumina RNA-seq and Oxford Nanopore dRNA-seq, associated with the acquisition of PTSD-like fear induced by Pavlovian cued-fear conditioning. We report significant sex-specific differences in the amygdala transcriptional response during fear acquisition and a range of shared and dimorphic epitranscriptomic signatures. Differential RNA modifications are enriched among mRNA transcripts associated with neurotransmitter regulation and mitochondrial function, many of which have been previously implicated in PTSD. Very few differentially modified transcripts are also differentially expressed, suggesting an influential, expression-independent role for epitranscriptional regulation in PTSD-like fear acquisition. PTSD-like trauma has sexually dimorphic effects on the amygdala transcriptome Most RNA modifications identified adhere to the known patterns associated with m6A There was enrichment of RNA modifications in neurological/PTSD-related genes There was little overlap between transcriptomic and epitranscriptomic signatures
Collapse
|
11
|
Zappelli E, Daniele S, Ceccarelli L, Vergassola M, Ragni L, Mangano G, Martini C. α-glyceryl-phosphoryl-ethanolamine protects human hippocampal neurons from aging-induced cellular alterations. Eur J Neurosci 2022; 56:4514-4528. [PMID: 35902984 PMCID: PMC9545488 DOI: 10.1111/ejn.15783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022]
Abstract
Brain ageing has been related to a decrease in cellular metabolism, to an accumulation of misfolded proteins and to an alteration of the lipid membrane composition. These alterations act as contributive aspects of age‐related memory decline by reducing membrane excitability and neurotransmitter release. In this sense, precursors of phospholipids (PLs) can restore the physiological composition of cellular membranes and ameliorate the cellular defects associated with brain ageing. In particular, phosphatidylcholine (PC) and phosphatidylethanolamine (PE) have been shown to restore mitochondrial function, reduce the accumulation of amyloid beta (Aβ) and, at the same time, provide the amount of acetylcholine needed to reduce memory deficit. Among PL precursors, alpha‐glycerylphosphorylethanolamine (GPE) has shown to protect astrocytes from Aβ injuries and to slow‐down ageing of human neural stem cells. GPE has been evaluated in aged human hippocampal neurons, which are implicated in learning and memory, and constitute a good in vitro model to investigate the beneficial properties of GPE. In order to mimic cellular ageing, the cells have been maintained 21 days in vitro and challenged with GPE. Results of the present paper showed GPE ability to increase PE and PC content, glucose uptake and the activity of the chain respiratory complex I and of the GSK‐3β pathway. Moreover, the nootropic compound showed an increase in the transcriptional/protein levels of neurotrophic and well‐being related genes. Finally, GPE counteracted the accumulation of ageing‐related misfolded proteins (a‐synuclein and tau). Overall, our data underline promising effects of GPE in counteracting cellular alterations related to brain ageing and cognitive decline.
Collapse
Affiliation(s)
| | | | | | | | - Lorella Ragni
- Global R&D PLCM -Angelini Pharma S.p.A, Ancona, Italy
| | | | | |
Collapse
|
12
|
Alexandrov YI, Pletnikov MV. Neuronal metabolism in learning and memory: The anticipatory activity perspective. Neurosci Biobehav Rev 2022; 137:104664. [PMID: 35439520 DOI: 10.1016/j.neubiorev.2022.104664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/30/2022] [Accepted: 04/10/2022] [Indexed: 12/20/2022]
Abstract
Current research on the molecular mechanisms of learning and memory is based on the "stimulus-response" paradigm, in which the neural circuits connecting environmental events with behavioral responses are strengthened. By contrast, cognitive and systems neuroscience emphasize the intrinsic activity of the brain that integrates information, establishes anticipatory actions, executes adaptive actions, and assesses the outcome via regulatory feedback mechanisms. We believe that the difference in the perspectives of systems and molecular studies is a major roadblock to further progress toward understanding the mechanisms of learning and memory. Here, we briefly overview the current studies in molecular mechanisms of learning and memory and propose that studying the predictive properties of neuronal metabolism will significantly advance our knowledge of how intrinsic, predictive activity of neurons shapes a new learning event. We further suggest that predictive metabolic changes in the brain may also take place in non-neuronal cells, including those of peripheral tissues. Finally, we present a path forward toward more in-depth studies of the role of cell metabolism in learning and memory.
Collapse
Affiliation(s)
- Yuri I Alexandrov
- V. B. Shvyrkov Laboratory for the Neural Bases of the Mind, Institute of Psychology, the Russian Academy of Sciences, Moscow, Russia; Department of Psychology, Institute for Cognitive Neuroscience, HSE University, Moscow, Russia.
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY 14203, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
13
|
Farahani M, Rezaei-Tavirani M, Zali A, Zamanian-Azodi M. Systematic Analysis of Protein-Protein and Gene-Environment Interactions to Decipher the Cognitive Mechanisms of Autism Spectrum Disorder. Cell Mol Neurobiol 2022; 42:1091-1103. [PMID: 33165687 PMCID: PMC11441303 DOI: 10.1007/s10571-020-00998-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD), a heterogeneous neurodevelopmental disorder resulting from both genetic and environmental risk factors, is manifested by deficits in cognitive function. Elucidating the cognitive disorder-relevant biological mechanisms may open up promising therapeutic approaches. In this work, we mined ASD cognitive phenotype proteins to construct and analyze protein-protein and gene-environment interaction networks. Incorporating the protein-protein interaction (PPI), human cognition proteins, and connections of autism-cognition proteins enabled us to generate an autism-cognition network (ACN). With the topological analysis of ACN, important proteins, highly clustered modules, and 3-node motifs were identified. Moreover, the impact of environmental exposures in cognitive impairment was investigated through chemicals that target the cognition-related proteins. Functional enrichment analysis of the ACN-associated modules and chemical targets revealed biological processes involved in the cognitive deficits of ASD. Among the 17 identified hub-bottlenecks in the ACN, PSD-95 was recognized as an important protein through analyzing the module and motif interactions. PSD-95 and its interacting partners constructed a cognitive-specific module. This hub-bottleneck interacted with the 89 cognition-related 3-node motifs. The identification of gene-environment interactions indicated that most of the cognitive-related proteins interact with bisphenol A (BPA) and valproic acid (VPA). Moreover, we detected significant expression changes of 56 cognitive-specific genes using four ASD microarray datasets in the GEO database, including GSE28521, GSE26415, GSE18123 and GSE29691. Our outcomes suggest future endeavors for dissecting the PSD-95 function in ASD and evaluating the various environmental conditions to discover possible mechanisms of the different levels of cognitive impairment.
Collapse
Affiliation(s)
- Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, 19716-53313, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, 19716-53313, Tehran, Iran.
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Zamanian-Azodi
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, 19716-53313, Tehran, Iran
| |
Collapse
|
14
|
Jin T, Wang R, Peng S, Liu X, Zhang H, He X, Teng W, Teng X. Developmental Hypothyroidism Influences the Development of the Entorhinal-Dentate Gyrus Pathway of Rat Offspring. Endocrinol Metab (Seoul) 2022; 37:290-302. [PMID: 35390249 PMCID: PMC9081305 DOI: 10.3803/enm.2021.1343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/21/2022] [Accepted: 02/10/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Developmental hypothyroidism impairs learning and memory in offspring, which depend on extensive neuronal circuits in the entorhinal cortex, together with the hippocampus and neocortex. The entorhinal-dentate gyrus pathway is the main entrance of memory circuits. We investigated whether developmental hypothyroidism impaired the morphological development of the entorhinal-dentate gyrus pathway. METHODS We examined the structure and function of the entorhinal-dentate gyrus pathway in response to developmental hypothyroidism induced using 2-mercapto-1-methylimidazole. RESULTS 1,1´-Dioctadecyl-3,3,3´,3´-tetramethylindocarbocyanine perchlorate tract tracing indicated that entorhinal axons showed delayed growth in reaching the outer molecular layer of the dentate gyrus at postnatal days 2 and 4 in hypothyroid conditions. The proportion of fibers in the outer molecular layer was significantly smaller in the hypothyroid group than in the euthyroid group at postnatal day 4. At postnatal day 10, the pathway showed a layer-specific distribution in the outer molecular layer, similar to the euthyroid group. However, the projected area of entorhinal axons was smaller in the hypothyroid group than in the euthyroid group. An electrophysiological examination showed that hypothyroidism impaired the long-term potentiation of the perforant and the cornu ammonis 3-cornu ammonis 1 pathways. Many repulsive axon guidance molecules were involved in the formation of the entorhinaldentate gyrus pathway. The hypothyroid group had higher levels of erythropoietin-producing hepatocyte ligand A3 and semaphorin 3A than the euthyroid group. CONCLUSION We demonstrated that developmental hypothyroidism might influence the development of the entorhinal-dentate gyrus pathway, contributing to impaired long-term potentiation. These findings improve our understanding of neural mechanisms for memory function.
Collapse
Affiliation(s)
- Ting Jin
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ranran Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Endocrinology, Chifeng College Affiliated Hospital, Chifeng, China
| | - Shiqiao Peng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Liu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hanyi Zhang
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xue He
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, National Health Commission Key Laboratory of Thyroid Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Liu C, Zhao H, Ji ZH, Yu XY. Hyperhomocysteinemia Induces Rat Memory Impairment Via Injuring Hippocampal CA3 Neurons and Downregulating cAMP Response Element-Binding Protein (CREB) Phosphorylation. Neurochem Res 2021; 47:762-767. [PMID: 34787820 DOI: 10.1007/s11064-021-03485-w] [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] [Received: 08/27/2021] [Revised: 10/22/2021] [Accepted: 11/08/2021] [Indexed: 11/30/2022]
Abstract
Accumulated evidence demonstrated that an elevated plasma homocysteine level, hyperhomocysteinemia, induced cognitive impairment in animals, elderly and the patients with neurodegenerative diseases. To date, the underlying cellular and molecular mechanisms by which hyperhomocysteinemia induces cognitive impairment has not been clearly defined. The purpose of this study was to investigate the possible cellular and molecular mechanisms behind hyperhomocysteinemia signaling in rat memory impairment. The results from this study demonstrated that hyperhomocysteinemia induced neuronal damage and loss in hippocampal CA3 region and downregulated the cAMP response element-binding protein (CREB) phosphorylation. The findings of this study provide evidence that hyperhomocysteinemia induces rat memory impairment via injuring hippocampal CA3 neurons and downregulating CREB phosphorylation.
Collapse
Affiliation(s)
- Chao Liu
- Laboratory of Neuroscience, College of Medicine, Dalian University, Dalian, 116622, People's Republic of China
| | - Hong Zhao
- Laboratory of Neuroscience, College of Medicine, Dalian University, Dalian, 116622, People's Republic of China
| | - Zhi-Hong Ji
- Laboratory of Neuroscience, College of Medicine, Dalian University, Dalian, 116622, People's Republic of China
| | - Xin-Yu Yu
- Laboratory of Neuroscience, College of Medicine, Dalian University, Dalian, 116622, People's Republic of China.
| |
Collapse
|
16
|
Peng X, Wang J, Peng J, Jiang H, Le K. Resveratrol Improves Synaptic Plasticity in Hypoxic-Ischemic Brain Injury in Neonatal Mice via Alleviating SIRT1/NF-κB Signaling-Mediated Neuroinflammation. J Mol Neurosci 2021; 72:113-125. [PMID: 34549339 DOI: 10.1007/s12031-021-01908-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is an obstinate disease that troubles neonatologists. At present, cognitive impairment after HIE has received increasing attention. Synaptic plasticity determines the development of cognitive function, so it is urgent to develop new drugs that can improve HIE-induced cognitive impairment. Hypoxia-ischemia (HI)-induced neuroinflammation affects synaptic plasticity. As a SIRT1 agonist, resveratrol has a powerful anti-inflammatory effect, but whether it has an effect on impaired synaptic plasticity in HIE and the potential mechanism remain unclear. In the present study, resveratrol was used to intervene in hypoxic-ischemic brain injury (HIBI) mice, and the effects on hippocampal synaptic plasticity and further mechanisms were explored through performing neurobehavioral, morphological observations, Golgi sliver staining, western blotting, and quantitative real-time polymerase chain reaction experiments. We first found that resveratrol improves HI-induced long-term cognitive and memory deficits, and then we found that resveratrol reduces hippocampal neuronal damage and increases dendritic spine density and the expression of synaptic proteins. Finally, we found that this effect may be exerted by regulating the neuroinflammatory response mediated by the SIRT1/NF-κB axis. This study provides a new theoretical basis for resveratrol to prevent long-term neurological dysfunction following HIBI.
Collapse
Affiliation(s)
- Xin Peng
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, No.17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.,Department of Otolaryngology, Jiangxi Province Children's Hospital, No.122 Yangming Road, Nanchang, Jiangxi Province, 330006, China
| | - Jun Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, No.17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Juan Peng
- Department of Rehabilitation Medicine, PingXiang No.2 People's Hospital, No. 89 Pingan South Avenue, Danjiang Street, PingXiang, Jiangxi Province, 337000, China
| | - Hongqun Jiang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, No.17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, No.17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
17
|
CPEB3-dowregulated Nr3c1 mRNA translation confers resilience to developing posttraumatic stress disorder-like behavior in fear-conditioned mice. Neuropsychopharmacology 2021; 46:1669-1679. [PMID: 33941859 PMCID: PMC8280193 DOI: 10.1038/s41386-021-01017-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/12/2021] [Accepted: 04/08/2021] [Indexed: 02/03/2023]
Abstract
Susceptibility or resilience to posttraumatic stress disorder (PTSD) depends on one's ability to appropriately adjust synaptic plasticity for coping with the traumatic experience. Activity-regulated mRNA translation synthesizes plasticity-related proteins to support long-term synaptic changes and memory. Hence, cytoplasmic polyadenylation element-binding protein 3-knockout (CPEB3-KO) mice, showing dysregulated translation-associated synaptic rigidity, may be susceptible to PTSD-like behavior. Here, using a context-dependent auditory fear conditioning and extinction paradigm, we found that CPEB3-KO mice exhibited traumatic intensity-dependent PTSD-like fear memory. A genome-wide screen of CPEB3-bound transcripts revealed that Nr3c1, encoding glucocorticoid receptor (GR), was translationally suppressed by CPEB3. Thus, CPEB3-KO neurons with elevated GR expression exhibited increased corticosterone-induced calcium influx and decreased mRNA and protein levels of brain-derived neurotrophic factor (Bdnf). Moreover, the reduced expression of BDNF was associated with increased GR level during fear extinction in CPEB3-KO hippocampi. Intracerebroventricular delivery of BDNF before extinction training mitigated spontaneous fear intrusion in CPEB3-KO mice during extinction recall. Analysis of two GEO datasets revealed decreased transcriptomic expression of CPEB3 but not NR3C1 in peripheral blood mononuclear cells of humans with PTSD. Collectively, this study reveals that CPEB3, as a potential PTSD-risk gene, downregulates Nr3c1 translation to maintain proper GR-BDNF signaling for fear extinction.
Collapse
|
18
|
Chia CH, Tang XW, Cao Y, Cao HT, Zhang W, Wu JF, Zhu YL, Chen Y, Lin Y, Wu Y, Zhang Z, Yuan TF, Hu RP. Cortical excitability signatures for the degree of sleepiness in human. eLife 2021; 10:65099. [PMID: 34313218 PMCID: PMC8373378 DOI: 10.7554/elife.65099] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Sleep is essential in maintaining physiological homeostasis in the brain. While the underlying mechanism is not fully understood, a 'synaptic homeostasis' theory has been proposed that synapses continue to strengthen during awake and undergo downscaling during sleep. This theory predicts that brain excitability increases with sleepiness. Here, we collected transcranial magnetic stimulation measurements in 38 subjects in a 34 hr program and decoded the relationship between cortical excitability and self-report sleepiness using advanced statistical methods. By utilizing a combination of partial least squares regression and mixed-effect models, we identified a robust pattern of excitability changes, which can quantitatively predict the degree of sleepiness. Moreover, we found that synaptic strengthen occurred in both excitatory and inhibitory connections after sleep deprivation. In sum, our study provides supportive evidence for the synaptic homeostasis theory in human sleep and clarifies the process of synaptic strength modulation during sleepiness.
Collapse
Affiliation(s)
- Chin-Hsuan Chia
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin-Wei Tang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yue Cao
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hua-Teng Cao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Wei Zhang
- Institute of Brain Science, Fudan University, Shanghai, China
| | - Jun-Fa Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Lian Zhu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Chen
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Lin
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhe Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental HealthCenter, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Rui-Ping Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Micro and Nano Plastics Distribution in Fish as Model Organisms: Histopathology, Blood Response and Bioaccumulation in Different Organs. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11135768] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Micro- and nano-plastic (MP/NP) pollution represents a threat not only to marine organisms and ecosystems, but also a danger for humans. The effects of these small particles resulting from the fragmentation of waste of various types have been well documented in mammals, although the consequences of acute and chronic exposure are not fully known yet. In this review, we summarize the recent results related to effects of MPs/NPs in different species of fish, both saltwater and freshwater, including zebrafish, used as model organisms for the evaluation of human health risk posed by MNPs. The expectation is that discoveries made in the model will provide insight regarding the risks of plastic particle toxicity to human health, with a focus on the effect of long-term exposure at different levels of biological complexity in various tissues and organs, including the brain. The current scientific evidence shows that plastic particle toxicity depends not only on factors such as particle size, concentration, exposure time, shape, and polymer type, but also on co-factors, which make the issue extremely complex. We describe and discuss the possible entry pathways of these particles into the fish body, as well as their uptake mechanisms and bioaccumulation in different organs and the role of blood response (hematochemical and hematological parameters) as biomarkers of micro- and nano-plastic water pollution.
Collapse
|
20
|
Keiser AA, Kramár EA, Dong T, Shanur S, Pirodan M, Ru N, Acharya MM, Baulch JE, Limoli CL, Wood MA. Systemic HDAC3 inhibition ameliorates impairments in synaptic plasticity caused by simulated galactic cosmic radiation exposure in male mice. Neurobiol Learn Mem 2021; 178:107367. [PMID: 33359392 PMCID: PMC8456980 DOI: 10.1016/j.nlm.2020.107367] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022]
Abstract
Deep space travel presents a number of measurable risks including exposure to a spectrum of radiations of varying qualities, termed galactic cosmic radiation (GCR) that are capable of penetrating the spacecraft, traversing through the body and impacting brain function. Using rodents, studies have reported that exposure to simulated GCR leads to cognitive impairments associated with changes in hippocampus function that can persist as long as one-year post exposure with no sign of recovery. Whether memory can be updated to incorporate new information in mice exposed to GCR is unknown. Further, mechanisms underlying long lasting impairments in cognitive function as a result of GCR exposure have yet to be defined. Here, we examined whether whole body exposure to simulated GCR using 6 ions and doses of 5 or 30 cGy interfered with the ability to update an existing memory or impact hippocampal synaptic plasticity, a cellular mechanism believed to underlie memory processes, by examining long term potentiation (LTP) in acute hippocampal slices from middle aged male mice 3.5-5 months after radiation exposure. Using a modified version of the hippocampus-dependent object location memory task developed by our lab termed "Objects in Updated Locations" (OUL) task we find that GCR exposure impaired hippocampus-dependent memory updating and hippocampal LTP 3.5-5 months after exposure. Further, we find that impairments in LTP are reversed through one-time systemic subcutaneous injection of the histone deacetylase 3 inhibitor RGFP 966 (10 mg/kg), suggesting that long lasting impairments in cognitive function may be mediated at least in part, through epigenetic mechanisms.
Collapse
Affiliation(s)
- A A Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - E A Kramár
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - T Dong
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - S Shanur
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - M Pirodan
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States
| | - N Ru
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States
| | - M M Acharya
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States
| | - J E Baulch
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States
| | - C L Limoli
- Department of Radiation Oncology, University of California, Irvine 92697-2695, United States.
| | - M A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences University of California, Irvine 92697-2695, United States; Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine 92697-2695, United States; Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine 92697-2695, United States.
| |
Collapse
|
21
|
So LY, Miller JE. Social context-dependent singing alters molecular markers of synaptic plasticity signaling in finch basal ganglia Area X. Behav Brain Res 2020; 398:112955. [PMID: 33031871 DOI: 10.1016/j.bbr.2020.112955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/14/2020] [Accepted: 09/30/2020] [Indexed: 12/23/2022]
Abstract
Vocal communication is a crucial skill required throughout life. However, there is a critical gap in our understanding of the underlying molecular brain mechanisms, thereby motivating our use of the zebra finch songbird model. Adult male zebra finches show differences in neural activity patterns in song-dedicated brain nuclei when they sing in two distinct social contexts: a male singing by himself (undirected, UD) and a male singing to a female (female-directed, FD). In our prior work, we showed that in song-dedicated basal ganglia Area X, protein levels of a N-methyl-D-aspartate receptor subtype 2B (NMDAR2B) increased with more UD song and decreased with more FD song. We hypothesized that molecules downstream of this receptor would show differential protein expression levels in Area X between UD and FD song. Specifically, we investigated calcium/calmodulin dependent protein kinase II beta (CaMKIIB), homer scaffold protein 1 (HOMER1), serine/threonine protein kinase (Akt), and mechanistic target of rapamycin kinase (mTOR) following singing and non-singing states in Area X. We show relationships between social context and protein levels. HOMER1 protein levels decreased with time spent singing FD song, and mTOR protein levels decreased with the amount of and time spent singing FD song. For both HOMER1 and mTOR, there were no differences with the amount of UD song. With time spent singing UD, CaMKIIB protein levels trended in a U-shaped curve whereas Akt protein levels trended down. Both molecules showed no change with FD song. Our results support differential involvement of molecules in synaptic plasticity pathways between UD and FD song behaviors.
Collapse
Affiliation(s)
- Lisa Y So
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Gould-Simpson Building, Tucson, AZ, 85721, United States; Department of Neuroscience, University of Arizona, Gould-Simpson Building, Tucson, AZ, 85721, United States
| | - Julie E Miller
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Gould-Simpson Building, Tucson, AZ, 85721, United States; Department of Neuroscience, University of Arizona, Gould-Simpson Building, Tucson, AZ, 85721, United States; Department of Speech, Language, and Hearing Sciences, University of Arizona, Speech, Language, and Hearing Sciences Building, Tucson, AZ, 85721, United States.
| |
Collapse
|
22
|
Wu J, Dou Y, Ladiges WC. Adverse Neurological Effects of Short-Term Sleep Deprivation in Aging Mice Are Prevented by SS31 Peptide. Clocks Sleep 2020; 2:325-333. [PMID: 33089207 PMCID: PMC7573804 DOI: 10.3390/clockssleep2030024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 08/04/2020] [Indexed: 12/01/2022] Open
Abstract
Sleep deprivation is a potent stress factor that disrupts regulatory pathways in the brain resulting in cognitive dysfunction and increased risk of neurodegenerative disease with increasing age. Prevention of the adverse effects of sleep deprivation could be beneficial in older individuals by restoring healthy brain function. We report here on the ability of SS31, a mitochondrial specific peptide, to attenuate the negative neurological effects of short-term sleep deprivation in aging mice. C57BL/6 female mice, 20 months old, were subcutaneously injected with SS31 (3 mg/kg) or saline daily for four days. Sleep deprivation was 4 h daily for the last two days of SS31 treatment. Mice were immediately tested for learning ability followed by collection of brain and other tissues. In sleep deprived mice treated with SS31, learning impairment was prevented, brain mitochondrial ATP levels and synaptic plasticity regulatory proteins were restored, and reactive oxygen species (ROS) and inflammatory cytokines levels were decreased in the hippocampus. This observation suggests possible therapeutic benefits of SS31 for alleviating adverse neurological effects of short-term sleep loss.
Collapse
Affiliation(s)
- Jinzi Wu
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA; (J.W.); (Y.D.)
| | - Yan Dou
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA; (J.W.); (Y.D.)
| | - Warren C Ladiges
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195, USA; (J.W.); (Y.D.)
| |
Collapse
|
23
|
Tomášková A, Šlamberová R, Černá M. Influence of Prenatal Methamphetamine Abuse on the Brain. EPIGENOMES 2020; 4:14. [PMID: 34968287 PMCID: PMC8594709 DOI: 10.3390/epigenomes4030014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 11/17/2022] Open
Abstract
Methamphetamine (MA), a psychostimulant, has become a serious problem in recent years. It is one of the most widely abused psychostimulants in the world. In the Czech Republic, ecstasy is the most commonly used non-cannabis drug, followed by hallucinogenic fungi, LSD, MA, cocaine, and finally heroin. The prevalence of the usage of all addictive substances is highest in the age category of 15-34. Approximately 17.2% of registered drug addicts, both male and female, in the Czech Republic use MA as their first-choice drug. This group consists mostly of women who are unemployed and addicted to MA (85%). Almost half of the addicted women switched to MA from other drugs in the course of pregnancy. Psychostimulants such as amphetamine and its synthetic derivate MA induce feelings of calm and happiness by suppressing anxiety and depression. When MA is abused for longer periods, it mimics symptoms of mania and can lead to the development of psychosis. MA is often abused for its anorectic effect, its simple preparation, and compared to heroin and cocaine, its low price. There are significant differences in the susceptibility of users to the stimulant, with reactions to MA fluctuating from person to person. Molecular mechanisms related to the variable response among users might represent an explanation for increased addiction-associated bipolar disorder and psychosis. Currently, there is limited information regarding genetic mechanisms linked to these disorders and the transmission of drug addiction. As such, animal models of drug addiction represent significant sources of information and assets in the research of these issues. The aim of this review is to summarize the mechanism of action of methamphetamine and its effect on pregnant addicted women and their children, including a detailed description of the anatomical structures involved.
Collapse
Affiliation(s)
- Anežka Tomášková
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Marie Černá
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| |
Collapse
|
24
|
TRPM4 inhibition improves spatial memory impairment and hippocampal long-term potentiation deficit in chronic cerebral hypoperfused rats. Behav Brain Res 2020; 393:112781. [PMID: 32619565 DOI: 10.1016/j.bbr.2020.112781] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) been well characterized as a common pathological status contributing to neurodegenerative diseases such as Alzheimer's disease and vascular dementia. CCH is an important factor that leads to cognitive impairment, but the underlying neurobiological mechanism is poorly understood and no effective treatment is available. Recently, transient receptor potential melastatin 4 (TRPM4) cation channel has been identified as an important molecular element in focal cerebral ischemia. Over activation of the channel is a major molecular mechanism of oncotic cell death. However, the role of TRPM4 in CCH that propagates global brain hypoxia have not been explored. Therefore, the present study is designed to investigate the effect of TRPM4 inhibition on the cognitive functions of the rats following CCH via permanent bilateral occlusion of common carotid arteries (PBOCCA) model. In this model, treatment with siRNA suppressed TRPM4 expression at both the mRNA and protein levels and improved cognitive deficits of the CCH rats without affecting their motor function. Furthermore, treatment with siRNA rescued the LTP impairment in CCH-induced rats. Consistent with the restored of LTP, western blot analysis revealed that siRNA treatment prevented the reduction of synaptic proteins, including calcium/calmodulin-dependent kinase II alpha (CaMKIIα) and brain-derived neurotrophic factor (BDNF) in brain regions of CCH rats. The present findings provide a novel role of TRPM4 in restricting cognitive functions in CCH and suggest inhibiting TRPM4 may represent a promising therapeutic strategy in targeting ion channels to prevent the progression of cognitive deficits induced by ischemia.
Collapse
|
25
|
Abstract
The hippocampus is central to spatial learning and stress responsiveness, both of which differ in form and function in males versus females, yet precisely how the hippocampus contributes to these sex differences is largely unknown. In reproductively mature individuals, sex differences in the steroid hormone milieu undergirds many sex differences in hippocampal-related endpoints. However, there is also evidence for developmental programming of adult hippocampal function, with a central role for androgens as well as their aromatized byproduct, estrogens. These include sex differences in cell genesis, synapse formation, dendritic arborization, and excitatory/inhibitory balance. Enduring effects of steroid hormone modulation occur during two developmental epochs, the first being the classic perinatal critical period of sexual differentiation of the brain and the other being adolescence and the associated hormonal changes of puberty. The cellular mechanisms by which steroid hormones enduringly modify hippocampal form and function are poorly understood, but we here review what is known and highlight where attention should be focused.
Collapse
|
26
|
Baajour SJ, Chowdury A, Thomas P, Rajan U, Khatib D, Zajac-Benitez C, Falco D, Haddad L, Amirsadri A, Bressler S, Stanley JA, Diwadkar VA. Disordered directional brain network interactions during learning dynamics in schizophrenia revealed by multivariate autoregressive models. Hum Brain Mapp 2020; 41:3594-3607. [PMID: 32436639 PMCID: PMC7416040 DOI: 10.1002/hbm.25032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/09/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Directional network interactions underpin normative brain function in key domains including associative learning. Schizophrenia (SCZ) is characterized by altered learning dynamics, yet dysfunctional directional functional connectivity (dFC) evoked during learning is rarely assessed. Here, nonlinear learning dynamics were induced using a paradigm alternating between conditions (Encoding and Retrieval). Evoked fMRI time series data were modeled using multivariate autoregressive (MVAR) models, to discover dysfunctional direction interactions between brain network constituents during learning stages (Early vs. Late), and conditions. A functionally derived subnetwork of coactivated (healthy controls [HC] ∩ SCZ] nodes was identified. MVAR models quantified directional interactions between pairs of nodes, and coefficients were evaluated for intergroup differences (HC ≠ SCZ). In exploratory analyses, we quantified statistical effects of neuroleptic dosage on performance and MVAR measures. During Early Encoding, SCZ showed reduced dFC within a frontal–hippocampal–fusiform network, though during Late Encoding reduced dFC was associated with pathways toward the dorsolateral prefrontal cortex (dlPFC). During Early Retrieval, SCZ showed increased dFC in pathways to and from the dorsal anterior cingulate cortex, though during Late Retrieval, patients showed increased dFC in pathways toward the dlPFC, but decreased dFC in pathways from the dlPFC. These discoveries constitute novel extensions of our understanding of task‐evoked dysconnection in schizophrenia and motivate understanding of the directional aspect of the dysconnection in schizophrenia. Disordered directionality should be investigated using computational psychiatric approaches that complement the MVAR method used in our work.
Collapse
Affiliation(s)
- Shahira J Baajour
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Asadur Chowdury
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Patricia Thomas
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Usha Rajan
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dalal Khatib
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Caroline Zajac-Benitez
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dimitri Falco
- Center for Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, Florida, USA
| | - Luay Haddad
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Alireza Amirsadri
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Steven Bressler
- Center for Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, Florida, USA.,Department of Psychology, Florida Atlantic University, Boca Raton, Florida, USA
| | - Jeffery A Stanley
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Vaibhav A Diwadkar
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
27
|
Khatib T, Chisholm DR, Whiting A, Platt B, McCaffery P. Decay in Retinoic Acid Signaling in Varied Models of Alzheimer's Disease and In-Vitro Test of Novel Retinoic Acid Receptor Ligands (RAR-Ms) to Regulate Protective Genes. J Alzheimers Dis 2020; 73:935-954. [PMID: 31884477 PMCID: PMC7081102 DOI: 10.3233/jad-190931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2019] [Indexed: 12/22/2022]
Abstract
Retinoic acid has been previously proposed in the treatment of Alzheimer's disease (AD). Here, five transgenic mouse models expressing AD and frontotemporal dementia risk genes (i.e., PLB2APP, PLB2TAU, PLB1Double, PLB1Triple, and PLB4) were used to investigate if consistent alterations exist in multiple elements of the retinoic acid signaling pathway in these models. Many steps of the retinoic acid signaling pathway including binding proteins and metabolic enzymes decline, while the previously reported increase in RBP4 was only consistent at late (6 months) but not early (3 month) ages. The retinoic acid receptors were exceptional in their consistent decline in mRNA and protein with transcript decline of retinoic acid receptors β and γ by 3 months, before significant pathology, suggesting involvement in early stages of disease. Decline in RBP1 transcript may also be an early but not late marker of disease. The decline in the retinoic acid signaling system may therefore be a therapeutic target for AD and frontotemporal dementia. Thus, novel stable retinoic acid receptor modulators (RAR-Ms) activating multiple genomic and non-genomic pathways were probed for therapeutic control of gene expression in rat primary hippocampal and cortical cultures. RAR-Ms promoted the non-amyloidogenic pathway, repressed lipopolysaccharide induced inflammatory genes and induced genes with neurotrophic action. RAR-Ms had diverse effects on gene expression allowing particular RAR-Ms to be selected for maximal therapeutic effect. Overall the results demonstrated the early decline of retinoic acid signaling in AD and frontotemporal dementia models and the activity of stable and potent alternatives to retinoic acid as potential therapeutics.
Collapse
Affiliation(s)
- Thabat Khatib
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - David R. Chisholm
- Department of Chemistry, Durham University, Science Laboratories, South Road, Durham, UK
| | - Andrew Whiting
- Department of Chemistry, Durham University, Science Laboratories, South Road, Durham, UK
| | - Bettina Platt
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| |
Collapse
|
28
|
Bothe K, Hirschauer F, Wiesinger HP, Edfelder JM, Gruber G, Hoedlmoser K, Birklbauer J. Gross motor adaptation benefits from sleep after training. J Sleep Res 2019; 29:e12961. [PMID: 31868978 PMCID: PMC7540033 DOI: 10.1111/jsr.12961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 01/26/2023]
Abstract
Sleep has been shown to facilitate the consolidation of newly acquired motor memories. However, the role of sleep in gross motor learning, especially in motor adaptation, is less clear. Thus, we investigated the effects of nocturnal sleep on the performance of a gross motor adaptation task, i.e. riding an inverse steering bicycle. Twenty‐six male participants (M = 24.19, SD = 3.70 years) were randomly assigned to a PM‐AM‐PM (n = 13) or an AM‐PM‐AM (n = 13) group, i.e. they trained in the evening/morning and were re‐tested the next morning/evening and the following evening/morning (PM‐AM‐PM/AM‐PM‐AM group) so that every participant spent one sleep as well as one wake interval between the three test sessions. Inverse cycling performance was assessed by speed (riding time) and accuracy (standard deviation of steering angle) measures. Behavioural results showed that in the PM‐AM‐PM group a night of sleep right after training stabilized performance (accuracy and speed) and was further improved over the subsequent wake interval. In the AM‐PM‐AM group, a significant performance deterioration after the initial wake interval was followed by the restoration of subjects' performance levels from right after training when a full night of sleep was granted. Regarding sleep, right hemispheric fast N2 sleep spindle activity was related to better stabilization of inverse cycling skills, thus possibly reflecting the ongoing process of updating the participants' mental model from “how to ride a bicycle” to “how to ride an inverse steering bicycle”. Our results demonstrate that sleep facilitates the consolidation of gross motor adaptation, thus adding further insights to the role of sleep for tasks with real‐life relevance.
Collapse
Affiliation(s)
- Kathrin Bothe
- Laboratory for Sleep, Cognition and Consciousness Research, Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| | - Franziska Hirschauer
- Laboratory for Sleep, Cognition and Consciousness Research, Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| | - Hans-Peter Wiesinger
- Department of Sport and Exercise Science, University of Salzburg, Salzburg, Austria
| | - Janina M Edfelder
- Department of Sport and Exercise Science, University of Salzburg, Salzburg, Austria
| | - Georg Gruber
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Kerstin Hoedlmoser
- Laboratory for Sleep, Cognition and Consciousness Research, Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| | - Juergen Birklbauer
- Department of Sport and Exercise Science, University of Salzburg, Salzburg, Austria
| |
Collapse
|
29
|
Rivi V, Benatti C, Colliva C, Radighieri G, Brunello N, Tascedda F, Blom JMC. Lymnaea stagnalis as model for translational neuroscience research: From pond to bench. Neurosci Biobehav Rev 2019; 108:602-616. [PMID: 31786320 DOI: 10.1016/j.neubiorev.2019.11.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/24/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Abstract
The purpose of this review is to illustrate how a reductionistic, but sophisticated, approach based on the use of a simple model system such as the pond snail Lymnaea stagnalis (L. stagnalis), might be useful to address fundamental questions in learning and memory. L. stagnalis, as a model, provides an interesting platform to investigate the dialog between the synapse and the nucleus and vice versa during memory and learning. More importantly, the "molecular actors" of the memory dialogue are well-conserved both across phylogenetic groups and learning paradigms, involving single- or multi-trials, aversion or reward, operant or classical conditioning. At the same time, this model could help to study how, where and when the memory dialog is impaired in stressful conditions and during aging and neurodegeneration in humans and thus offers new insights and targets in order to develop innovative therapies and technology for the treatment of a range of neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- V Rivi
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - C Benatti
- Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - C Colliva
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - G Radighieri
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - N Brunello
- Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F Tascedda
- Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - J M C Blom
- Dept. of Education and Human Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
30
|
Mak CW, Ching-Fong Yeung K, Chan KM. Acute toxic effects of polyethylene microplastic on adult zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 182:109442. [PMID: 31352214 DOI: 10.1016/j.ecoenv.2019.109442] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 06/10/2023]
Abstract
To identify the physical effects, behavioral changes, and gene expression profiles of the phase 1 detoxification-related gene (cyp 1a) and oogenesis-related gene (vtg 1) induced by microplastics, high-density polyethylene microplastics of various sizes were used because of their dominance in coastal areas and effluent samples in Hong Kong. Adult zebrafish were used as the model organism to identify the upper and lower boundaries of microplastics ingestion and were exposed to individual polyethylene microplastics in five size ranges (10-22 μm, 45-53 μm, 90-106 μm, 212-250 μm, and 500-600 μm) at a concentration of 2 mg/L for 96 h. To study behavioral changes and targeted gene expression profiles via real-time PCR (qPCR), a mixture of microplastics in three size ranges at effluent-related (11 particles/L), moderate (110 particles/L), and high concentrations (1,100 particles/L) were applied for 96 h. The zebrafish behavior was recorded by a video camera and by two observers (interrater reliability, >85%). The results implied that the upper and lower size boundaries for microplastic ingestion were 558.4 ± 26.2 μm (yellow) and 19.7 ± 3.1 μm (red), respectively. In addition, 61 ± 10% of fish in medium concentration treatments and 61 ± 10% of fish in high concentration treatments were found with the microplastic ingestion and remaining in their intestine. In addition, 28 ± 10% of fish in high concentration treatments were found with microplastic retaining in their gills (No. of fishes = 18 in each treatment). The presence of microplastics, which occupied 89 ± 6% of intestine area, reduced the voids inside the intestine for feed. The expression of cyp1a in the intestine (medium concentration) and vtg1 in the liver (medium and high concentration) showed significant up-regulation, and abnormal behavior (i.e., seizures and tail bent downward) was observed (medium and high concentration). In summary, the effects on the aryl hydrocarbon receptor (AHR) pathway, disruption of the oogenesis process, and neurotoxicity could be caused by acute exposure of adult zebrafish to microplastics.
Collapse
Affiliation(s)
- Chu Wa Mak
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, N.T., Hong Kong
| | | | - King Ming Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, N.T., Hong Kong.
| |
Collapse
|
31
|
Microglia Adopt Longitudinal Transcriptional Changes After Traumatic Brain Injury. J Surg Res 2019; 246:113-122. [PMID: 31563831 DOI: 10.1016/j.jss.2019.08.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/18/2019] [Accepted: 08/29/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) is an under-recognized public health threat. Even mild brain injuries can lead to long-term neurologic impairment. Microglia play a fundamental role in the development and progression of this ensuing neurologic impairment. Despite this, a microglia-specific injury signature has yet to be identified. We hypothesized that TBI would lead to long-term changes in the transcriptional profile of microglial pathways associated with the development of subsequent neurologic impairment. MATERIALS AND METHODS Male C57BL/6 mice underwent TBI via a controlled cortical impact and were followed longitudinally. FACSorted microglia from TBI mice were subjected to Quantiseq 3'-biased RNA sequencing at 7, 30, and 90 d after TBI. K-means clustering on 396 differentially expressed genes was performed, and gene ontology enrichment analysis was used to determine corresponding enriched processes. RESULTS Differentially expressed genes in microglia exhibited four main patterns of expression over the course of TBI. In particular, we identified four gene clusters which corresponded to the host defense response, synaptic plasticity, lipid remodeling, and membrane polarization. CONCLUSIONS Transcriptional profiling within individual populations of microglia after TBI remains a critical unmet research need within the field of TBI. This focused study identified several physiologic processes within microglia that may be associated with development of long-term neurologic impairment after TBI. These data demonstrate the capability of longitudinal transcriptional profiling to uncover potential cell-specific targets for the treatment of TBI.
Collapse
|
32
|
Heydari A, Esmaeilpour K, Sheibani V. Maternal separation impairs long term-potentiation in CA3-CA1 synapses in adolescent female rats. Behav Brain Res 2019; 376:112239. [PMID: 31526768 DOI: 10.1016/j.bbr.2019.112239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/11/2019] [Accepted: 09/13/2019] [Indexed: 12/28/2022]
Abstract
Mother-infant interactions influence the development of physiology and behavior during the first weeks after birth. As an adverse early life experience, maternal separation (MS) produces behavioral and neuroendocrine functions disorders associated with the hippocampus. Considering the critical role of long-term potentiation (LTP) in learning and memory, we investigated whether MS affects LTP in adolescent female rats. In this study, female rat pups were exposed to daily 3-h (MS180) or 15-min (MS15) periods of maternal separation on postnatal days (PND) 1-14 and control offspring remained with the dams all the time before weaning. Extracellular evoked field excitatory postsynaptic potentials (fEPSPs) were recorded in the stratum radiatum of the CA1 area of the slice at 28-35 days of age. Our results indicate that a significant difference existed in the magnitude of LTP between the control group and MS180 group, but the MS15 group was not different from control. In conclusion, these findings suggest that MS may impair LTP induction in the CA1 area of the hippocampus in adolescent female rats.
Collapse
Affiliation(s)
- Arefe Heydari
- Department of Physiology, Faculty of Medicine, Kerman university of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology, Faculty of Medicine, Kerman university of Medical Sciences, Kerman, Iran.
| |
Collapse
|
33
|
Zeng K, Li Y, Yang W, Ge Y, Xu L, Ren T, Zhang H, Zhuo R, Peng L, Chen C, Zhou Y, Zhao Y, Li WJ, Jin X, Yang L. Moringa oleifera seed extract protects against brain damage in both the acute and delayed stages of ischemic stroke. Exp Gerontol 2019; 122:99-108. [DOI: 10.1016/j.exger.2019.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/11/2019] [Accepted: 04/21/2019] [Indexed: 10/26/2022]
|
34
|
Feng J, Chen S, Wang Y, Liu Q, Yang M, Li X, Nie C, Qin J, Chen H, Yuan X, Huang Y, Zhang Q. Maternal exposure to cadmium impairs cognitive development of male offspring by targeting the Coronin-1a signaling pathway. CHEMOSPHERE 2019; 225:765-774. [PMID: 30903850 DOI: 10.1016/j.chemosphere.2019.03.094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 06/09/2023]
Abstract
Direct exposure to cadmium (Cd) may induce persistent impairment in learning and memory. However, the outcomes of maternal exposure on the neurological development of offspring are much less clear, and the underlying mechanism leading to toxicity remains undisclosed. Following chronic exposure of female rats during gestation and lactation, low level of Cd was detectable in the cerebral cortex but not in the hippocampus of F1 male offspring. The synapses and neurites in hippocampus were destroyed by high Cd exposure level as evidenced by abnormal morphology and cognitive behavior deficit lasting from childhood to adulthood. The membrane glycoprotein M6a (GPM6A) regulates the filopodium formation, neurite outgrowth and synaptogenesis, and is a possible target which Cd acts upon. The signaling pathway Coronin-1a (CORO1A), Ras-related C3 botulinum toxin substrate 1 (RAC1) and p21-activated kinase 1 (PAK1) promotes GPM6A-induced filopodium formation. Our results showed that maternal exposure dramatically down-regulated the level of CORO1A as well as the expression of downstream effectors RAC1, PAK1 and GPM6A. CORO1A-knockdown by siRNA caused decreases in the expression of RAC1, PAK1 and GPM6A; and siRNA targeting combined with Cd insult further decreased the expression of these proteins. Following CORO1A overexpression, the neurites were lengthened with increased expression of all the effector proteins in SH-SY5Y cells exposed to Cd, confirming the significance of CORO1A in mediating the Cd neurotoxicity. These findings may help to disclose how Cd impairs the learning and cognitive development in children, and facilitate finding of potential therapeutic targets for the treatment of Cd poisoning.
Collapse
Affiliation(s)
- Jianfeng Feng
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Shaomin Chen
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Youjin Wang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Qunxing Liu
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Mengqi Yang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xin Li
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Chuan Nie
- Guangdong Women and Children Hospital, Guangzhou, 510000, China
| | - Jianxiang Qin
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Hongxia Chen
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiaohui Yuan
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, 510632, China
| | - Yadong Huang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, 510632, China
| | - Qihao Zhang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, 510632, China; National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
35
|
Majdi A, Kamari F, Sadigh-Eteghad S, Gjedde A. Molecular Insights Into Memory-Enhancing Metabolites of Nicotine in Brain: A Systematic Review. Front Neurosci 2019; 12:1002. [PMID: 30697142 PMCID: PMC6341027 DOI: 10.3389/fnins.2018.01002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/12/2018] [Indexed: 01/25/2023] Open
Abstract
Background: The alleged procognitive effects of nicotine and its metabolites in brain are controversial. Objective: Here, we review the pharmacologically active metabolites of nicotine in brain and their effects on neuronal mechanisms involving two main cognitive domains, i.e., learning and memory. Methods: We searched Embase, Medline via PubMed, Scopus, and Web of Science databases for entries no later than May 2018, and restricted the search to articles about nicotine metabolites and cognitive behavior or cognitive mechanisms. Results: The initial search yielded 425 articles, of which 17 were eligible for inclusion after application of exclusion criteria. Of these, 13 were experimental, two were clinical, and two were conference papers. Conclusions: The results revealed three pharmacologically active biotransformations of nicotine in the brain, including cotinine, norcotinine, and nornicotine, among which cotinine and nornicotine both had a procognitive impact without adverse effects. The observed effect was significant only for cotinine.
Collapse
Affiliation(s)
- Alireza Majdi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzin Kamari
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Albert Gjedde
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
36
|
Zhang T, Shi Z, Wang Y, Wang L, Zhang B, Chen G, Wan Q, Chen L. Akt3 deletion in mice impairs spatial cognition and hippocampal CA1 long long-term potentiation through downregulation of mTOR. Acta Physiol (Oxf) 2019; 225:e13167. [PMID: 30053339 DOI: 10.1111/apha.13167] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 07/20/2018] [Accepted: 07/24/2018] [Indexed: 11/30/2022]
Abstract
AIM Loss-of-function mutation of Akt3 in humans has been associated with microcephaly and cognitive defects. Two Akt isoforms, Akt1 and Akt3, are highly expressed in hippocampal pyramidal cells. We explored the roles of Akt1 and Akt3, respectively, in spatial cognition and underlying mechanisms. METHODS We used Akt1 knockout (Akt1-KO) and Akt3 knockout (Akt3-KO) mice to examine the influence of Akt1 and Akt3 deficiency on spatial memory, as well as induction and maintenance of hippocampal CA1 NMDA receptor-dependent and protein synthesis-dependent long-term potentiation (LTP). RESULTS Long-term spatial memory was impaired in Akt3-KO mice, but not in Akt1-KO mice, as assessed by the Morris water maze task. Akt3-KO and Akt1-KO mice displayed reductions in brain size without concurrent changes in the number of pyramidal cells or basal properties of synaptic transmission. One-train high-frequency stimulation (HFS × 1) induced NMDA receptor-dependent LTP in Akt3-KO mice and Akt1-KO mice. Four-train HFS (HFS × 4) induced rapamycin-sensitive long-LTP in Akt1-KO mice, but not Akt3-KO mice. Basal level of mTOR phosphorylation was reduced in Akt3-KO mice rather than Akt1-KO mice. HFS × 4 induced an elevation of mTOR and p70S6K phosphorylation in Akt1-KO mice, which led to enhanced 4EBP2 and eIF4E phosphorylation along with an increase in AMPA receptor protein. However, the same protocol of HFS × 4 failed to trigger the mTOR-p70S6K signalling cascade or increase 4EBP2 and eIF4E phosphorylation in Akt3-KO mice. CONCLUSION The Akt3 deficiency via inactivation of mTOR suppresses HFS × 4-induced mTOR-p70S6K signalling to reduce phosphorylation of 4EBP and eIF4E, which impairs protein synthesis-dependent long-LTP and long-term spatial cognitive function.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Lab of Reproductive Medicine; Nanjing Medical University; Nanjing China
- Department of Physiology; Nanjing Medical University; Nanjing China
| | - Zhaochun Shi
- Department of Neurology; First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Ya Wang
- Department of Physiology; Nanjing Medical University; Nanjing China
| | - Ling Wang
- Department of Physiology; Nanjing Medical University; Nanjing China
| | - Baofeng Zhang
- Department of Physiology; Nanjing Medical University; Nanjing China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology; MOE Key Laboratory of Model Animal for Disease Study; Model Animal Research Center; Nanjing University; Nanjing China
| | - Qi Wan
- Department of Neurology; First Affiliated Hospital of Nanjing Medical University; Nanjing China
| | - Ling Chen
- State Key Lab of Reproductive Medicine; Nanjing Medical University; Nanjing China
- Department of Physiology; Nanjing Medical University; Nanjing China
| |
Collapse
|
37
|
Ghanbari A, Moradi Kor N, Rashidy-Pour A. Bombesin-induced enhancement of memory consolidation in male and female rat pups: Role of glutamatergic and dopaminergic systems. Neuropeptides 2018; 70:101-106. [PMID: 29880391 DOI: 10.1016/j.npep.2018.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/18/2018] [Accepted: 05/30/2018] [Indexed: 02/04/2023]
Abstract
Previous studies have shown that the neuropeptide bombesin (BBS) enhances consolidation of specifically for inhibitory avoidance memory in adult rats. However, its effect on memory consolidation during premature period is not clear as well. Thus, this study evaluated the effect of BBS and its interaction with glutamatergic and dopaminergic systems on memory consolidation in rat pups. Male and female rat pups (30 days old) were trained in an inhibitory avoidance (IA) task (0.5 mA, 3 s footshock). Memory retention was tested 24 h later during which the latency to re-enter to the shock compartment was recorded. First, the effects of different doses (0.001, 0.0025, 0.005, 0.01 and 0.02 mg/kg) of BBS injected immediately following training were tested. Then, the effect of the most effective dose of BBS obtained in the previous experiment was examined in the presence of the glutamate NMDA receptor antagonist MK-801 (0.05 mg/kg), the dopamine D1 receptor antagonist SCH-23390 (0.05 mg/kg) and the dopamine D2 receptor antagonist sulpiride (20 mg/kg). Findings indicate that BBS significantly enhances memory consolidation at all tested doses in male pups and at a dose of 0.01 mg/kg in female pups. MK-801, SCH-23390 and sulpiride administration before BBS injection in individual groups significantly blocked BBS-induced memory enhancement. Our findings indicate that similar to adult rats, BBS enhances memory consolidation in developing rat. This enhancing effect is mediated, at least in part, via an interaction with glutamatergic and dopaminergic systems.
Collapse
Affiliation(s)
- Ali Ghanbari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Nasroallah Moradi Kor
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Student Research Committee and Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Research Center of Physiology, Department of Physiology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
38
|
Feng J, Lu W, Wang GY, Zhu ZM, Sun Y, Du K, Wang JH. Cell-specific plasticity associated with integrative memory of triple sensory signals in the barrel cortex. Oncotarget 2018; 9:30962-30978. [PMID: 30123420 PMCID: PMC6089555 DOI: 10.18632/oncotarget.25740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/06/2018] [Indexed: 01/08/2023] Open
Abstract
Neuronal plasticity occurs in associative memory. Associative memory cells are recruited for the integration and storage of associated signals. The coordinated refinements and interactions of associative memory cells including glutamatergic and GABAergic neurons remain elusive, which we have examined in a mouse model of associative learning. Paired olfaction, tail and whisker stimulations lead to odorant-induced and tail-induced whisker motions alongside whisker-induced whisker motion. In mice that show this cross-modal associative memory, barrel cortical glutamatergic and GABAergic neurons are recruited to encode the newly learned odor and tail signals alongside the innate whisker signal. These glutamatergic neurons are functionally upregulated, and GABAergic neurons are refined in a homeostatic manner. The mutual innervations between these glutamatergic and GABAergic neurons are upregulated. Therefore, the co-activations of sensory cortices by pairing the input signals recruit their glutamatergic and GABAergic neurons to be associative memory cells, which undergo coordinated refinement among glutamatergic and GABAergic neurons as well as homeostatic plasticity among subcellular compartments in order to drive these cells toward the optimal state for the integrative storage of associated signals.
Collapse
Affiliation(s)
- Jing Feng
- Department of Biology, University of Science and Technology China, Hefei, China
- Institute of Biophysics and University of Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wei Lu
- Department of Biology, University of Science and Technology China, Hefei, China
- Institute of Biophysics and University of Chinese Academy of Sciences, Beijing, China
- Qingdao University, School of Pharmacy, Shandong, China
| | | | - Zhao-Ming Zhu
- Qingdao University, School of Pharmacy, Shandong, China
| | - Yan Sun
- Qingdao University, School of Pharmacy, Shandong, China
| | - Kaixin Du
- Qingdao University, School of Pharmacy, Shandong, China
| | - Jin-Hui Wang
- Department of Biology, University of Science and Technology China, Hefei, China
- Institute of Biophysics and University of Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
39
|
Usuda K, Kawase T, Shigeno Y, Fukuzawa S, Fujii K, Zhang H, Tsukahara T, Tomonaga S, Watanabe G, Jin W, Nagaoka K. Hippocampal metabolism of amino acids by L-amino acid oxidase is involved in fear learning and memory. Sci Rep 2018; 8:11073. [PMID: 30038322 PMCID: PMC6056520 DOI: 10.1038/s41598-018-28885-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 07/02/2018] [Indexed: 12/18/2022] Open
Abstract
Amino acids participate directly and indirectly in many important biochemical functions in the brain. We focused on one amino acid metabolic enzyme, L-amino acid oxidase (LAO), and investigated the importance of LAO in brain function using LAO1 knockout (KO) mice. Compared to wild-type mice, LAO1 KO mice exhibited impaired fear learning and memory function in a passive avoidance test. This impairment in LAO1 KO mice coincided with significantly reduced hippocampal acetylcholine levels compared to wild-type mice, while treatment with donepezil, a reversible acetylcholine esterase inhibitor, inhibited this reduction. Metabolomic analysis revealed that knocking out LAO1 affected amino acid metabolism (mainly of phenylalanine [Phe]) in the hippocampus. Specifically, Phe levels were elevated in LAO1 KO mice, while phenylpyruvic acid (metabolite of Phe produced largely by LAO) levels were reduced. Moreover, knocking out LAO1 decreased hippocampal mRNA levels of pyruvate kinase, the enzymatic activity of which is known to be inhibited by Phe. Based on our findings, we propose that LAO1 KO mice exhibited impaired fear learning and memory owing to low hippocampal acetylcholine levels. Furthermore, we speculate that hippocampal Phe metabolism is an important physiological mechanism related to glycolysis and may underlie cognitive impairments, including those observed in Alzheimer's disease.
Collapse
Affiliation(s)
- Kento Usuda
- United Graduate School of Veterinarian Science, Gifu University, Gifu, Gifu, Japan.,Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Takahiro Kawase
- Kyoto Institute of Nutrition and Pathology, Tsuzuki, Kyoto, Japan
| | - Yuko Shigeno
- Laboratory of Benno, RIKEN Innovation Center, Wako, Saitama, Japan
| | - Susumu Fukuzawa
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Kazuki Fujii
- Life Science Research Center, Toyama University, Toyama, Toyama, Japan
| | - Haolin Zhang
- College of Biological Science and Technology, Beijing Forestry University, Haidian, Beijing, China
| | | | - Shozo Tomonaga
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Kyoto, Japan
| | - Gen Watanabe
- United Graduate School of Veterinarian Science, Gifu University, Gifu, Gifu, Japan.,Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan
| | - Wanzhu Jin
- Institute of Zoology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Kentaro Nagaoka
- United Graduate School of Veterinarian Science, Gifu University, Gifu, Gifu, Japan. .,Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Fuchu, Tokyo, Japan.
| |
Collapse
|
40
|
Ge Y, Chen L, Yin Z, Song X, Ruan T, Hua L, Liu J, Wang J, Ning H. Fluoride-induced alterations of synapse-related proteins in the cerebral cortex of ICR offspring mouse brain. CHEMOSPHERE 2018; 201:874-883. [PMID: 29567471 DOI: 10.1016/j.chemosphere.2018.02.167] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 06/08/2023]
Abstract
Fluoride (F) exposure causes cognitive dysfunction in humans and animals. However, the precise molecular mechanisms by which fluoride exerts its neurotoxic effects are poorly understood. In this study, an animal model of fluoride exposure was created by providing ICR mice were treated with vehicle F at a dose of 0 (control group), 50 (low-fluoride group) or 100 mg/L (high-fluoride group) in water for one month. After the mice mated, parents and offspring were treated and maintained under these conditions. The cognitive abilities of the mice were examined using a Morris water maze test. Results indicated that fluoride exposure significantly prolonged the escape latency period and decreased the number of crossings in a particular zone. Histopathologic analysis revealed the shrinkage and fragmentation of glial cells in the fluoride-treated groups. Pyramidal cells in the cerebral cortices of fluoride-treated groups were fewer than those of the control group. The expression of microtubule-associated protein 2 (MAP2) and synaptic proteins of the cerebral cortex in mouse offspring was assayed using RT-PCR and Western blot. Fluoride exposure possibly induced a significantly decreased expression of MAP2, synaptophysin (SYP) and developmentally regulated brain protein (Dbn) at protein and mRNA levels. Glutamate receptor (N-methyl-d-aspartate receptor, NMDAR) was also expressed, and this finding was consistent with the reduced MAP2, SYP and Dbn expression. Therefore, fluoride-mediated reduction in cognitive dysfunction is likely caused by the disruption of the expression of these synapse-associated proteins, resulting in attenuated neuronal functioning.
Collapse
Affiliation(s)
- Yaming Ge
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China
| | - Lingli Chen
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China; Shanxi Key Laboratory of Ecological Animal Science and Environmental Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, PR China
| | - Zhihong Yin
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China
| | - Xiaochao Song
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China
| | - Tao Ruan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, PR China
| | - Liushuai Hua
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China
| | - Junwei Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China
| | - Jundong Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, PR China.
| | - Hongmei Ning
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, 453003, PR China; Shanxi Key Laboratory of Ecological Animal Science and Environmental Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, PR China.
| |
Collapse
|
41
|
Li D, Huang Y, Cheng B, Su J, Zhou WX, Zhang YX. Streptozotocin Induces Mild Cognitive Impairment at Appropriate Doses in Mice as Determined by Long-Term Potentiation and the Morris Water Maze. J Alzheimers Dis 2018; 54:89-98. [PMID: 27472873 DOI: 10.3233/jad-150979] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and effective therapeutic drugs in the clinic are still lacking. Ideally, AD progression could be stopped at an early stage, such as at the mild cognitive impairment (MCI) stage. MCI refers to the clinical condition between normal aging and dementia. Patients with MCI experience memory loss but do not meet the criteria for the diagnosis of clinically probable AD. However, few MCI animal models have been established. Here, we used in vivo long-term potentiation (LTP) recording and the Morris water maze (MWM) to evaluate the effects of intracerebroventricular injection of streptozotocin (ICV-STZ) in mice. We found a relationship between cognitive behavior and LTP in vivo and determined the appropriate doses of STZ for a putative MCI animal model. Animals that received≥150μg of STZ exhibited cognitive impairment in the MWM test, and few changes in behavior tests were observed in animals receiving less than 150μg of STZ. In vivo LTP recordings revealed that the induction of LTP decreased significantly in STZ-treated animals, even at the lowest dose (25μg/mouse), in a dose-dependent manner. Pathology analysis revealed STZ-induced neuron loss in a dose-dependent manner, both in the cortex and in the hippocampus, as evidenced by a significantly decreased neuronal number in the cohort treated with 75μg of STZ/mouse. Our study indicated that a low dose (25μg/mouse) of STZ impaired neural plasticity; at a higher dose of 75μg/mouse STZ, further LTP deficits were noted along with induced neuronal loss in both the cortex and the hippocampus, which could be considered a possible MCI or pre-MCI animal model; and finally, at 150μg/mouse STZ, dementia was induced, feasibly indicating a state of AD.
Collapse
|
42
|
Papaleonidopoulos V, Trompoukis G, Koutsoumpa A, Papatheodoropoulos C. A gradient of frequency-dependent synaptic properties along the longitudinal hippocampal axis. BMC Neurosci 2017; 18:79. [PMID: 29233091 PMCID: PMC5727934 DOI: 10.1186/s12868-017-0398-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/05/2017] [Indexed: 12/29/2022] Open
Abstract
Background The hippocampus is a functionally heterogeneous brain structure and specializations of the intrinsic neuronal network may crucially support the functional segregation along the longitudinal axis of the hippocampus. Short-term synaptic plasticity plays fundamental roles in information processing and may be importantly involved in diversifying the properties of local neuronal network along the hippocampus long axis. Therefore, we aimed to examine the properties of the cornu ammonis 1 (CA1) synapses along the entire dorsoventral axis of the rat hippocampus using field excitatory postsynaptic potentials from transverse rat hippocampal slices and a frequency stimulation paradigm. Results Applying a ten-pulse stimulus train at frequencies from 0.1 to 100 Hz to the Schaffer collaterals we found a gradually diversified pattern of frequency-dependent synaptic effects along the dorsoventral hippocampus axis. The first conditioned response was facilitated along the whole hippocampus for stimulus frequencies 10–40 Hz. However, steady-state responses or averaged responses generally ranged from maximum synaptic facilitation in the most dorsal segment of the hippocampus to maximum synaptic depression in the most ventral segment of the hippocampus. In particular, dorsal synapses facilitated for stimulus frequency up to 50 Hz while they depressed at higher frequencies (75–100 Hz). Facilitation at dorsal synapses was maximal at stimulus frequency of 20 Hz. On the contrary, the most ventral synapses showed depression regardless of the stimulus frequency, only displaying a transient facilitation at the beginning of 10–50 Hz stimulation. Importantly, the synapses in the medial hippocampus displayed a transitory behavior. Finally, as a whole the hippocampal synapses maximally facilitated at 20 Hz and increasingly depressed at 50–100 Hz. Conclusion The short-term synaptic dynamics change gradually along the hippocampal long axis in a frequency-dependent fashion conveying distinct properties of information processing to successive segments of the structure, thereby crucially supporting functional segregation along the dorsoventral axis of the hippocampus.
Collapse
Affiliation(s)
| | - George Trompoukis
- Department of Medicine, Laboratory of Physiology, University of Patras, 26504, Rion, Greece
| | - Andriana Koutsoumpa
- Department of Medicine, Laboratory of Physiology, University of Patras, 26504, Rion, Greece
| | | |
Collapse
|
43
|
Feng J, Lu W, Wang D, Ma K, Song Z, Chen N, Sun Y, Du K, Shen M, Cui S, Wang JH. Barrel Cortical Neuron Integrates Triple Associated Signals for Their Memory Through Receiving Epigenetic-Mediated New Synapse Innervations. Cereb Cortex 2017; 27:5858-5871. [PMID: 29121184 DOI: 10.1093/cercor/bhx292] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022] Open
Abstract
Associative learning is common way for information acquisition. Associative memory is essential to logical reasoning and associative thinking. The storages of multiple associated signals in individual neurons facilitate their integration, expand memory volume, and strengthen cognition ability. Associative memory cells that encode multiple signals have been reported, however, the mechanisms underlying their recruitment and working principle remain to be addressed. We have examined the recruitment of associative memory cells that integrate and store triple sensory signals as well as the potential mechanism of their recruitment. Paired mouse whisker, olfaction, and tail stimulations lead to odorant-induced motion and tail-induced whisker motion. In mice of expressing this cross-modal response, their barrel cortical neurons become to encode odor and tail signals alongside whisker signal. These barrel cortical neurons receive new synapse innervations from piriform and S1-tail cortical neurons. The emergence of cross-modal responses as well as the recruitments of new synapse innervations and associative memory cells in the barrel cortex need miRNA-324 and miRNA-133a, which downregulate Ttbk1 and Tet3. The co-activations of sensory cortices recruit their mutual synapse innervations and associative memory cells that integrate and store multiple associated signals through epigenetic-mediated process.
Collapse
Affiliation(s)
- Jing Feng
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Science, University of Science and Technology of China, Hefei Anhui 230026, China
| | - Wei Lu
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dangui Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ke Ma
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
| | - Zhenhua Song
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
| | - Na Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Sun
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
| | - Kaixin Du
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
| | - Mengmeng Shen
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
| | - Shan Cui
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jin-Hui Wang
- Qingdao University, School of Pharmacy, Qingdao, Shandong 266021, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Science, University of Science and Technology of China, Hefei Anhui 230026, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
44
|
Gerashchenko D, Pasumarthi RK, Kilduff TS. Plasticity-Related Gene Expression During Eszopiclone-Induced Sleep. Sleep 2017; 40:3866746. [PMID: 28605546 DOI: 10.1093/sleep/zsx098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Study Objectives Experimental evidence suggests that restorative processes depend on synaptic plasticity changes in the brain during sleep. We used the expression of plasticity-related genes to assess synaptic plasticity changes during drug-induced sleep. Methods We first characterized sleep induced by eszopiclone in mice during baseline conditions and during the recovery from sleep deprivation. We then compared the expression of 18 genes and two miRNAs critically involved in synaptic plasticity in these mice. Gene expression was assessed in the cerebral cortex and hippocampus by the TaqMan reverse transcription polymerase chain reaction and correlated with sleep parameters. Results Eszopiclone reduced the latency to nonrapid eye movement (NREM) sleep and increased NREM sleep amounts. Eszopiclone had no effect on slow wave activity (SWA) during baseline conditions but reduced the SWA increase during recovery sleep (RS) after sleep deprivation. Gene expression analyses revealed three distinct patterns: (1) four genes had higher expression either in the cortex or hippocampus in the group of mice with increased amounts of wakefulness; (2) a large proportion of plasticity-related genes (7 out of 18 genes) had higher expression during RS in the cortex but not in the hippocampus; and (3) six genes and the two miRNAs showed no significant changes across conditions. Even at a relatively high dose (20 mg/kg), eszopiclone did not reduce the expression of plasticity-related genes during RS period in the cortex. Conclusions These results indicate that gene expression associated with synaptic plasticity occurs in the cortex in the presence of a hypnotic medication.
Collapse
Affiliation(s)
| | - Ravi K Pasumarthi
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| |
Collapse
|
45
|
Ge Q, Wang Z, Wu Y, Huo Q, Qian Z, Tian Z, Ren W, Zhang X, Han J. High salt diet impairs memory-related synaptic plasticity via increased oxidative stress and suppressed synaptic protein expression. Mol Nutr Food Res 2017; 61. [PMID: 28654221 PMCID: PMC5656827 DOI: 10.1002/mnfr.201700134] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/03/2017] [Accepted: 06/02/2017] [Indexed: 12/27/2022]
Abstract
Scope A high salt (HS) diet is detrimental to cognitive function, in addition to having a role in cardiovascular disorders. However, the method by which an HS diet impairs cognitive functions such as learning and memory remains open. Methods and results In this study, we found that mice on a 7 week HS diet demonstrated disturbed short‐term memory in an object‐place recognition task, and both 4 week and 7 week HS treatments impaired long‐term memory, as evidenced in a fear conditioning test. Mechanistically, the HS diet inhibited memory‐related long‐term potentiation (LTP) in the hippocampus, while also increasing the levels of reactive oxygen species (ROS) in hippocampal cells and downregulating the expression of synapsin I, synaptophysin, and brain‐derived neurotrophic factor in specific encephalic region. Conclusion This suggests that oxidative stress or synaptic protein/neurotrophin deregulation was involved in the HS diet‐induced memory impairment. Thus, the present study provides novel insights into the mechanisms of memory impairment caused by excessive dietary salt, and underlined the importance of controlling to salt absorb quantity.
Collapse
Affiliation(s)
- Qian Ge
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Zhengjun Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yuwei Wu
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Qing Huo
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Zhaoqiang Qian
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Zhongmin Tian
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Wei Ren
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xia Zhang
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Jing Han
- Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
46
|
Kundap UP, Kumari Y, Othman I, Shaikh MF. Zebrafish as a Model for Epilepsy-Induced Cognitive Dysfunction: A Pharmacological, Biochemical and Behavioral Approach. Front Pharmacol 2017; 8:515. [PMID: 28824436 PMCID: PMC5541063 DOI: 10.3389/fphar.2017.00515] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/21/2017] [Indexed: 12/31/2022] Open
Abstract
Epilepsy is a neuronal disorder allied with distinct neurological and behavioral alterations characterized by recurrent spontaneous epileptic seizures. Impairment of the cognitive performances such as learning and memory is frequently observed in epileptic patients. Anti-epileptic drugs (AEDs) are efficient to the majority of patients. However, 30% of this population seems to be refractory to the drug treatment. These patients are not seizure-free and frequently they show impaired cognitive functions. Unfortunately, as a side effect, some AEDs could contribute to such impairment. The major problem associated with conducting studies on epilepsy-related cognitive function is the lack of easy, rapid, specific and sensitive in vivo testing models. However, by using a number of different techniques and parameters in the zebrafish, we can incorporate the unique feature of specific disorder to study the molecular and behavior basis of this disease. In the view of current literature, the goal of the study was to develop a zebrafish model of epilepsy induced cognitive dysfunction. In this study, the effect of AEDs on locomotor activity and seizure-like behavior was tested against the pentylenetetrazole (PTZ) induced seizures in zebrafish and epilepsy associated cognitive dysfunction was determined using T-maze test followed by neurotransmitter estimation and gene expression analysis. It was observed that all the AEDs significantly reversed PTZ induced seizure in zebrafish, but had a negative impact on cognitive functions of zebrafish. AEDs were found to modulate neurotransmitter levels, especially GABA, glutamate, and acetylcholine and gene expression in the drug treated zebrafish brains. Therefore, combination of behavioral, neurochemical and genenetic information, makes this model a useful tool for future research and discovery of newer and safer AEDs.
Collapse
Affiliation(s)
- Uday P Kundap
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University MalaysiaSelangor, Malaysia
| | - Yatinesh Kumari
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University MalaysiaSelangor, Malaysia
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University MalaysiaSelangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University MalaysiaSelangor, Malaysia
| |
Collapse
|
47
|
Wang L, Wang J, Wang F, Liu C, Yang X, Yang J, Ming D. VEGF-Mediated Cognitive and Synaptic Improvement in Chronic Cerebral Hypoperfusion Rats Involves Autophagy Process. Neuromolecular Med 2017; 19:423-435. [PMID: 28766254 DOI: 10.1007/s12017-017-8458-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/27/2017] [Indexed: 12/23/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) is associated with various neurodegenerative diseases characterized by cognitive impairment. Dozens of studies including ours have indicated that exogenous administration of vascular endothelial growth factor (VEGF) could exert effective cognitive protection during ischemia. Nevertheless, the underlying mechanism has not been well clarified. To address this issue, we explored the synaptic mechanisms in vivo since hippocampal synaptic function is essential to the learning and memory process. Besides, the role of autophagy in cognitive dysfunction under conditions of CCH is still controversial. And abnormal autophagy could threaten normal neurotransmission at synapse where a large amount of protein synthesis and degradation take place. Hence, we further examined whether the altered synaptic function was associated with autophagy. The results showed that CCH impaired spatial cognition as evidenced in Morris water maze. We further found that VEGF mitigated impaired hippocampal synaptic function including basal synaptic transmission, paired-pulse facilitation, short-term, long-term plasticity, depotentiation, and the level of synaptic proteins as assessed by electrophysiological examination and western blot assay. Furthermore, our results demonstrated that CCH could induce excessive autophagy which could be inhibited by VEGF. Thus, we speculated that VEGF could ameliorate impaired synaptic function induced by CCH because of its ability to inhibit excessive autophagy, and eventually improve spatial learning and memory function. Importantly, our findings shed light on potential therapeutic strategies to be exploited in the usage of VEGF.
Collapse
Affiliation(s)
- Ling Wang
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jingyu Wang
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, 300072, People's Republic of China.,State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Faqi Wang
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Chunhua Liu
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Xuening Yang
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jiajia Yang
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, 300072, People's Republic of China.
| | - Dong Ming
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin, 300072, People's Republic of China.
| |
Collapse
|
48
|
Zhao X, Huang L, Guo R, Liu Y, Zhao S, Guan S, Ge R, Cui S, Wang S, Wang JH. Coordinated Plasticity among Glutamatergic and GABAergic Neurons and Synapses in the Barrel Cortex Is Correlated to Learning Efficiency. Front Cell Neurosci 2017; 11:221. [PMID: 28798668 PMCID: PMC5526921 DOI: 10.3389/fncel.2017.00221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/12/2017] [Indexed: 01/29/2023] Open
Abstract
Functional plasticity at cortical synapses and neurons is presumably associated with learning and memory. Additionally, coordinated refinement between glutamatergic and GABAergic neurons occurs in associative memory. If these assumptions are present, neuronal plasticity strength and learning efficiency should be correlated. We have examined whether neuronal plasticity strength and learning efficiency are quantitatively correlated in a mouse model of associative learning. Paired whisker and odor stimulations in mice induce odorant-induced whisker motions. The fully establishment of this associative memory appears fast and slow, which are termed as high learning efficiency and low learning efficiency, respectively. In the study of cellular mechanisms underlying this differential learning efficiency, we have compared the strength of neuronal plasticity in the barrel cortices that store associative signals from the mice with high vs. low learning efficiencies. Our results indicate that the levels of learning efficiency are linearly correlated with the upregulated strengths of excitatory synaptic transmission on glutamatergic neurons and their excitability, as well as the downregulated strengths of GABAergic neurons' excitability, their excitatory synaptic inputs and inhibitory synaptic outputs in layers II~III of barrel cortices. The correlations between learning efficiency in associative memory formation and coordinated plasticity at cortical glutamatergic and GABAergic neurons support the notion that the plasticity of associative memory cells is a basis for memory strength.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Li Huang
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Rui Guo
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Yulong Liu
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Shidi Zhao
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Sudong Guan
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Rongjing Ge
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
| | - Shan Cui
- Laboratory of Brain and Cognitive Science, Institute of Biophysics and University of Chinese Academy of SciencesBeijing, China
- University of Chinese Academy of SciencesBeijing, China
| | - Shirlene Wang
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of MedicineChicago, IL, United States
| | - Jin-Hui Wang
- Department of Pathophysiology, Bengbu Medical CollegeBengbu, China
- Laboratory of Brain and Cognitive Science, Institute of Biophysics and University of Chinese Academy of SciencesBeijing, China
- University of Chinese Academy of SciencesBeijing, China
- School of Pharmacy, Qingdao UniversityQingdao, China
| |
Collapse
|
49
|
García-Rojo G, Gámiz F, Ampuero E, Rojas-Espina D, Sandoval R, Rozas C, Morales B, Wyneken U, Pancetti F. In Vivo Sub-chronic Treatment with Dichlorvos in Young Rats Promotes Synaptic Plasticity and Learning by a Mechanism that Involves Acylpeptide Hydrolase Instead of Acetylcholinesterase Inhibition. Correlation with Endogenous β-Amyloid Levels. Front Pharmacol 2017; 8:483. [PMID: 28790916 PMCID: PMC5524899 DOI: 10.3389/fphar.2017.00483] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/05/2017] [Indexed: 11/21/2022] Open
Abstract
Acylpeptide hydrolase (APEH) is a serine hydrolase that displays two catalytic activities, acting both as an exopeptidase toward short N-acylated peptides and as an endopeptidase toward oxidized peptides or proteins. It has been demonstrated that this enzyme can degrade monomers, dimers, and trimers of the Aβ1-40 peptide in the conditioned media of neuroblastoma cells. In a previous report, we showed that the specific inhibition of this enzyme by the organophosphate molecule dichlorvos (DDVP) triggers an enhancement of long-term potentiation in rat hippocampal slices. In this study, we demonstrate that the same effect can be accomplished in vivo by sub-chronic treatment of young rats with a low dose of DDVP (0.1 mg/kg). Besides exhibiting a significant enhancement of LTP, the treated animals also showed improvements in parameters of spatial learning and memory. Interestingly, higher doses of DDVP such as 2 mg/kg did not prove to be beneficial for synaptic plasticity or behavior. Due to the fact that at 2 mg/kg we observed inhibition of both APEH and acetylcholinesterase, we interpret that in order to achieve positive effects on the measured parameters only APEH inhibition should be obtained. The treatment with both DDVP doses produced an increase in the endogenous concentration of Aβ1-40, although this was statistically significant only at the dose of 0.1 mg/kg. We propose that APEH represents an interesting pharmacological target for cognitive enhancement, acting through the modulation of the endogenous concentration of Aβ1-40.
Collapse
Affiliation(s)
- Gonzalo García-Rojo
- Laboratory of Environmental Neurotoxicology, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del NorteCoquimbo, Chile
| | - Fernando Gámiz
- Laboratory of Environmental Neurotoxicology, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del NorteCoquimbo, Chile
| | - Estíbaliz Ampuero
- Laboratory of Neuroscience, Faculty of Medicine, Universidad de Los AndesSantiago, Chile
| | - Daniel Rojas-Espina
- Laboratory of Environmental Neurotoxicology, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del NorteCoquimbo, Chile
| | - Rodrigo Sandoval
- Laboratory of Environmental Neurotoxicology, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del NorteCoquimbo, Chile
| | - Carlos Rozas
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de ChileSantiago, Chile
| | - Bernardo Morales
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de ChileSantiago, Chile
| | - Ursula Wyneken
- Laboratory of Neuroscience, Faculty of Medicine, Universidad de Los AndesSantiago, Chile
| | - Floria Pancetti
- Laboratory of Environmental Neurotoxicology, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del NorteCoquimbo, Chile
| |
Collapse
|
50
|
Guo R, Ge R, Zhao S, Liu Y, Zhao X, Huang L, Guan S, Lu W, Cui S, Wang S, Wang JH. Associative Memory Extinction Is Accompanied by Decayed Plasticity at Motor Cortical Neurons and Persistent Plasticity at Sensory Cortical Neurons. Front Cell Neurosci 2017; 11:168. [PMID: 28659764 PMCID: PMC5469894 DOI: 10.3389/fncel.2017.00168] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/31/2017] [Indexed: 01/04/2023] Open
Abstract
Associative memory is essential for cognition, in which associative memory cells and their plasticity presumably play important roles. The mechanism underlying associative memory extinction vs. maintenance remains unclear, which we have studied in a mouse model of cross-modal associative learning. Paired whisker and olfaction stimulations lead to a full establishment of odorant-induced whisker motion in training day 10, which almost disappears if paired stimulations are not given in a week, and then recovers after paired stimulation for an additional day. In mice that show associative memory, extinction and recovery, we have analyzed the dynamical plasticity of glutamatergic neurons in layers II-III of the barrel cortex and layers IV-V of the motor cortex. Compared with control mice, the rate of evoked spikes as well as the amplitude and frequency of excitatory postsynaptic currents increase, whereas the amplitude and frequency of inhibitory postsynaptic currents (IPSC) decrease at training day 10 in associative memory mice. Without paired training for a week, these plastic changes are persistent in the barrel cortex and decayed in the motor cortex. If paired training is given for an additional day to revoke associative memory, neuronal plasticity recovers in the motor cortex. Our study indicates persistent neuronal plasticity in the barrel cortex for cross-modal memory maintenance as well as the dynamical change of neuronal plasticity in the motor cortex for memory retrieval and extinction. In other words, the sensory cortices are essential for long-term memory while the behavior-related cortices with the inability of memory retrieval are correlated to memory extinction.
Collapse
Affiliation(s)
- Rui Guo
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Rongjing Ge
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Shidi Zhao
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Yulong Liu
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Xin Zhao
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Li Huang
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Sodong Guan
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
| | - Wei Lu
- School of Pharmacy, Qingdao UniversityQingdao, China
| | - Shan Cui
- Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
| | - Shirlene Wang
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern UniversityChicago, IL, United States
| | - Jin-Hui Wang
- Department of Pathophysiology, Bengbu Medical CollegeAnhui, China
- School of Pharmacy, Qingdao UniversityQingdao, China
- Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of SciencesBeijing, China
- Department of Biology, University of Chinese Academy of SciencesBeijing, China
| |
Collapse
|