1
|
Wang X, Jiang Y, Feng B, Ma X, Zhang K, Yang F, Liu Z, Yang L, Yue J, Lu L, Song D, Guo Q, Qi J, Li X, Wang M, Zhang H, Huang J, Zhao M, Liu S. PJA1 mediates the effects of astrocytic GPR30 on learning and memory in female mice. J Clin Invest 2023; 133:e165812. [PMID: 37712419 PMCID: PMC10503807 DOI: 10.1172/jci165812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 07/19/2023] [Indexed: 09/16/2023] Open
Abstract
Hormone replacement therapy (HRT) is not recommended for treating learning and memory decline in menopausal women because it exerts adverse effects by activating classic estrogen receptors ERα and ERβ. The membrane estrogen receptor G protein-coupled receptor 30 (GPR30) has been reported to be involved in memory modulation; however, the underlying mechanisms are poorly understood. Here, we found that GPR30 deletion in astrocytes, but not in neurons, impaired learning and memory in female mice. Astrocytic GPR30 depletion induced A1 phenotype transition, impairing neuronal function. Further exploration revealed that Praja1 (PJA1), a RING ubiquitin ligase, mediated the effects of astrocytic GPR30 on learning and memory by binding to Serpina3n, which is a molecular marker of neuroinflammation in astrocytes. GPR30 positively modulated PJA1 expression through the CREB signaling pathway in cultured murine and human astrocytes. Additionally, the mRNA levels of GPR30 and PJA1 were reduced in exosomes isolated from postmenopausal women while Serpina3n levels were increased in the plasma. Together, our findings suggest a key role for astrocytic GPR30 in the learning and memory abilities of female mice and identify GPR30/PJA1/Serpina3n as potential therapeutic targets for learning and memory loss in peri- and postmenopausal women.
Collapse
Affiliation(s)
| | - Yongli Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Ban Feng
- Department of Pharmacology, School of Pharmacy and
| | - Xue Ma
- Department of Pharmacology, School of Pharmacy and
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy and
| | - Fan Yang
- Department of Pharmacology, School of Pharmacy and
| | - Zhenguo Liu
- Department of Pharmacy, Northwest Women’s and Children’s Hospital, Xi’an, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Jiao Yue
- Department of Pharmacology, School of Pharmacy and
| | - Liang Lu
- Department of Pharmacology, School of Pharmacy and
| | - Dake Song
- Department of Pharmacology, School of Pharmacy and
| | - Qingjuan Guo
- Department of Pharmacology, School of Pharmacy and
| | - Jingyu Qi
- Department of Pharmacology, School of Pharmacy and
| | - Xubo Li
- Department of Pharmacology, School of Pharmacy and
| | - Min Wang
- Department of Pharmacology, School of Pharmacy and
| | - Huinan Zhang
- Department of Health Management, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Jing Huang
- Department of Health Management, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Minggao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Shuibing Liu
- Department of Pharmacology, School of Pharmacy and
| |
Collapse
|
2
|
Ghosh B, Karmakar S, Prasad M, Mandal AK. Praja1 ubiquitin ligase facilitates degradation of polyglutamine proteins and suppresses polyglutamine-mediated toxicity. Mol Biol Cell 2021; 32:1579-1593. [PMID: 34161122 PMCID: PMC8351749 DOI: 10.1091/mbc.e20-11-0747] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A network of chaperones and ubiquitin ligases sustain intracellular proteostasis and is integral in preventing aggregation of misfolded proteins associated with various neurodegenerative diseases. Using cell-based studies of polyglutamine (polyQ) diseases, spinocerebellar ataxia type 3 (SCA3) and Huntington's disease (HD), we aimed to identify crucial ubiquitin ligases that protect against polyQ aggregation. We report here that Praja1 (PJA1), a Ring-H2 ubiquitin ligase abundantly expressed in the brain, is diminished when polyQ repeat proteins (ataxin-3/huntingtin) are expressed in cells. PJA1 interacts with polyQ proteins and enhances their degradation, resulting in reduced aggregate formation. Down-regulation of PJA1 in neuronal cells increases polyQ protein levels vis-a-vis their aggregates, rendering the cells vulnerable to cytotoxic stress. Finally, PJA1 suppresses polyQ toxicity in yeast and rescues eye degeneration in a transgenic Drosophila model of SCA3. Thus, our findings establish PJA1 as a robust ubiquitin ligase of polyQ proteins and induction of which might serve as an alternative therapeutic strategy in handling cytotoxic polyQ aggregates.
Collapse
Affiliation(s)
- Baijayanti Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - Susnata Karmakar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Mohit Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Atin K Mandal
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| |
Collapse
|
3
|
Watabe K, Kato Y, Sakuma M, Murata M, Niida-Kawaguchi M, Takemura T, Hanagata N, Tada M, Kakita A, Shibata N. Praja1 RING-finger E3 ubiquitin ligase suppresses neuronal cytoplasmic TDP-43 aggregate formation. Neuropathology 2020; 40:570-586. [PMID: 32686212 PMCID: PMC7818255 DOI: 10.1111/neup.12694] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
Abstract
Transactivation response DNA-binding protein of 43 kDa (TDP-43) is a major constituent of cytoplasmic aggregates in neuronal and glial cells in cases of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). We have previously shown neuronal cytoplasmic aggregate formation induced by recombinant adenoviruses expressing human wild-type and C-terminal fragment (CTF) TDP-43 under the condition of proteasome inhibition in vitro and in vivo. In the present study, we demonstrated that the formation of the adenoviral TDP-43 aggregates was markedly suppressed in rat neural stem cell-derived neuronal cells by co-infection of an adenovirus expressing heat shock transcription factor 1 (HSF1), a master regulator of heat shock response. We performed DNA microarray analysis and searched several candidate molecules, located downstream of HSF1, which counteract TDP-43 aggregate formation. Among these, we identified Praja 1 RING-finger E3 ubiquitin ligase (PJA1) as a suppressor of phosphorylation and aggregate formation of TDP-43. Co-immunoprecipitation assay revealed that PJA1 binds to CTF TDP-43 and the E2-conjugating enzyme UBE2E3. PJA1 also suppressed formation of cytoplasmic phosphorylated TDP-43 aggregates in mouse facial motor neurons in vivo. Furthermore, phosphorylated TDP-43 aggregates were detected in PJA1-immunoreactive human ALS motor neurons. These results indicate that PJA1 is one of the principal E3 ubiquitin ligases for TDP-43 to counteract its aggregation propensity and could be a potential therapeutic target for ALS and FTLD.
Collapse
Affiliation(s)
- Kazuhiko Watabe
- Department of Medical Technology (Neuropathology), Kyorin University, Tokyo, Japan.,Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yoichiro Kato
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Miho Sakuma
- School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Makiko Murata
- Department of Medical Technology (Neuropathology), Kyorin University, Tokyo, Japan
| | | | - Taro Takemura
- Research Network and Facility Services Division, National Institute for Materials Science, Tsukuba, Japan
| | - Nobutaka Hanagata
- Research Network and Facility Services Division, National Institute for Materials Science, Tsukuba, Japan
| | - Mari Tada
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
4
|
Mulligan MK, Lu L, Cavigelli SA, Mormède P, Terenina E, Zhao W, Williams RW, Jones BC. Impact of Genetic Variation on Stress-Related Ethanol Consumption. Alcohol Clin Exp Res 2019; 43:1391-1402. [PMID: 31034606 DOI: 10.1111/acer.14073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/23/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND The effect of stress on alcohol consumption in humans is highly variable, and the underlying processes are not yet understood. Attempts to model a positive relationship between stress and increased ethanol (EtOH) consumption in animals have been only modestly successful. Our hypothesis is that individual differences in stress effects on EtOH consumption are mediated by genetics. METHODS We measured alcohol consumption, using the drinking-in-the-dark (DID) paradigm in females from 2 inbred mouse strains, C57BL/6J (B6) and DBA/2J (D2), and 35 of their inbred progeny (the BXD family). A control group was maintained in normal housing and a stress group was exposed to chronic mild stress (CMS), consisting of unpredictable stressors over 7 weeks. These included predator, social, and environmental perturbations. Alcohol intake was measured over 16 weeks in both groups during baseline (preceding 5-week period), CMS (intervening 7-week period), and post-CMS (final 4-week period). RESULTS We detected a strong effect of CMS on alcohol intake. A few strains demonstrated CMS-related increased alcohol consumption; however, most showed decreased intake. We identified 1 nearly significant quantitative trait locus on chromosome 5 that contains the neuronal nitric oxide synthase gene (Nos1). The expression of Nos1 is frequently changed following alcohol exposure, and variants in this gene segregating among the BXD population may modulate alcohol intake in response to stress. CONCLUSIONS The results we present here represent the first study to combine chronic stress and alcohol consumption in a genetic reference population of mice. Differences in susceptibility to the effects of stressful environments vis-à-vis alcohol use disorders would suggest that the differences have at least some basis in genetic constitution. We have also nominated a likely candidate gene underlying the large individual differences in effects of stress on alcohol consumption.
Collapse
Affiliation(s)
- Megan K Mulligan
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Lu Lu
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Pierre Mormède
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet-Tolosan, France
| | - Elena Terenina
- GenPhySE, Université de Toulouse, INRA, ENVT, Castanet-Tolosan, France
| | - Wenyuan Zhao
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Robert W Williams
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Byron C Jones
- The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
5
|
PJA1 Coordinates with the SMC5/6 Complex To Restrict DNA Viruses and Episomal Genes in an Interferon-Independent Manner. J Virol 2018; 92:JVI.00825-18. [PMID: 30185588 PMCID: PMC6206484 DOI: 10.1128/jvi.00825-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
DNA viruses, including hepatitis B virus and herpes simplex virus, induce a series of immune responses in the host and lead to human public health concerns worldwide. In addition to cytokines in the cytoplasm, restriction of viral DNA in the nucleus is an important approach of host immunity. However, the mechanism of foreign DNA recognition and restriction in the cell nucleus is largely unknown. This work demonstrates that an important cellular factor (PJA1) suppresses DNA viruses and transfected plasmids independent of type I and II interferon (IFN) pathways. Instead, PJA1 interacts with the chromosome maintenance complex (SMC5/6), facilitates the complex to recognize and bind viral and episomal DNAs, and recruits DNA topoisomerases to restrict the foreign molecules. These results reveal a distinct mechanism underlying the silencing of viral and episomal invaders in the cell nuclei and suggest that PJA1 acts as a potential agent to prevent infectious and inflammatory diseases. Viral and episomal DNAs, as signs of infections and dangers, induce a series of immune responses in the host, and cells must sense foreign DNAs to eliminate the invaders. The cell nucleus is not “immune privileged” and exerts intrinsic mechanisms to control nuclear-replicating DNA viruses. Thus, it is important to understand the action of viral DNA sensing in the cell nucleus. Here, we reveal a mechanism of restriction of DNA viruses and episomal plasmids mediated by PJA1, a RING-H2 E3 ubiquitin ligase. PJA1 restricts the DNA viruses hepatitis B virus (HBV) and herpes simplex virus 1 (HSV-1) but not the RNA viruses enterovirus 71 (EV71) and vesicular stomatitis virus (VSV). Similarly, PJA1 inhibits episomal plasmids but not chromosome-integrated reporters or endogenous genes. In addition, PJA1 has no effect on endogenous type I and II interferons (IFNs) and interferon-stimulated genes (ISGs), suggesting that PJA1 silences DNA viruses independent of the IFN pathways. Interestingly, PJA1 interacts with the SMC5/6 complex (a complex essential for chromosome maintenance and HBV restriction) to facilitate the binding of the complex to viral and episomal DNAs in the cell nucleus. Moreover, treatment with inhibitors of DNA topoisomerases (Tops) and knockdown of Tops release PJA1-mediated silencing of viral and extrachromosomal DNAs. Taken together, results of this work demonstrate that PJA1 interacts with SMC5/6 and facilitates the complex to bind and eliminate viral and episomal DNAs through DNA Tops and thus reveal a distinct mechanism underlying restriction of DNA viruses and foreign genes in the cell nucleus. IMPORTANCE DNA viruses, including hepatitis B virus and herpes simplex virus, induce a series of immune responses in the host and lead to human public health concerns worldwide. In addition to cytokines in the cytoplasm, restriction of viral DNA in the nucleus is an important approach of host immunity. However, the mechanism of foreign DNA recognition and restriction in the cell nucleus is largely unknown. This work demonstrates that an important cellular factor (PJA1) suppresses DNA viruses and transfected plasmids independent of type I and II interferon (IFN) pathways. Instead, PJA1 interacts with the chromosome maintenance complex (SMC5/6), facilitates the complex to recognize and bind viral and episomal DNAs, and recruits DNA topoisomerases to restrict the foreign molecules. These results reveal a distinct mechanism underlying the silencing of viral and episomal invaders in the cell nuclei and suggest that PJA1 acts as a potential agent to prevent infectious and inflammatory diseases.
Collapse
|
6
|
Olsen RHJ, Weber SJ, Akinyeke T, Raber J. Enhanced cued fear memory following post-training whole body irradiation of 3-month-old mice. Behav Brain Res 2017; 319:181-187. [PMID: 27865918 PMCID: PMC5924676 DOI: 10.1016/j.bbr.2016.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 11/12/2016] [Accepted: 11/15/2016] [Indexed: 12/25/2022]
Abstract
Typically, in studies designed to assess effects of irradiation on cognitive performance the animals are trained and tested for cognitive function following irradiation. Little is known about post-training effects of irradiation on cognitive performance. In the current study, 3-month-old male mice were irradiated with X-rays 24h following training in a fear conditioning paradigm and cognitively tested starting two weeks later. Average motion during the extinction trials, measures of anxiety in the elevated zero maze, and body weight changes over the course of the study were assessed as well. Exposure to whole body irradiation 24h following training in a fear conditioning resulted in greater freezing levels 2 weeks after training. In addition, motion during both contextual and cued extinction trials was lower in irradiated than sham-irradiated mice. In mice trained for cued fear conditioning, activity levels in the elevated zero maze 12days after sham-irradiation or irradiation were also lower in irradiated than sham-irradiated mice. Finally, the trajectory of body weight changes was affected by irradiation, with lower body weights in irradiated than sham-irradiated mice, with the most profound effect 7days after training. These effects were associated with reduced c-Myc protein levels in the amygdala of the irradiated mice. These data indicate that whole body X ray irradiation of mice at 3 months of age causes persistent alterations in the fear response and activity levels in a novel environment, while the effects on body weight seem more transient.
Collapse
Affiliation(s)
- Reid H J Olsen
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sydney J Weber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tunde Akinyeke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
7
|
Taguchi YH. Principal component analysis based unsupervised feature extraction applied to publicly available gene expression profiles provides new insights into the mechanisms of action of histone deacetylase inhibitors. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.nepig.2016.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
Identification and Characterization of the V(D)J Recombination Activating Gene 1 in Long-Term Memory of Context Fear Conditioning. Neural Plast 2015; 2016:1752176. [PMID: 26843989 PMCID: PMC4710954 DOI: 10.1155/2016/1752176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/12/2015] [Indexed: 12/17/2022] Open
Abstract
An increasing body of evidence suggests that mechanisms related to the introduction and repair of DNA double strand breaks (DSBs) may be associated with long-term memory (LTM) processes. Previous studies from our group suggested that factors known to function in DNA recombination/repair machineries, such as DNA ligases, polymerases, and DNA endonucleases, play a role in LTM. Here we report data using C57BL/6 mice showing that the V(D)J recombination-activating gene 1 (RAG1), which encodes a factor that introduces DSBs in immunoglobulin and T-cell receptor genes, is induced in the amygdala, but not in the hippocampus, after context fear conditioning. Amygdalar induction of RAG1 mRNA, measured by real-time PCR, was not observed in context-only or shock-only controls, suggesting that the context fear conditioning response is related to associative learning processes. Furthermore, double immunofluorescence studies demonstrated the neuronal localization of RAG1 protein in amygdalar sections prepared after perfusion and fixation. In functional studies, intra-amygdalar injections of RAG1 gapmer antisense oligonucleotides, given 1 h prior to conditioning, resulted in amygdalar knockdown of RAG1 mRNA and a significant impairment in LTM, tested 24 h after training. Overall, these findings suggest that the V(D)J recombination-activating gene 1, RAG1, may play a role in LTM consolidation.
Collapse
|
9
|
Stockton SD, Gomes I, Liu T, Moraje C, Hipólito L, Jones MR, Ma'ayan A, Morón JA, Li H, Devi LA. Morphine Regulated Synaptic Networks Revealed by Integrated Proteomics and Network Analysis. Mol Cell Proteomics 2015; 14:2564-76. [PMID: 26149443 DOI: 10.1074/mcp.m115.047977] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Indexed: 01/12/2023] Open
Abstract
Despite its efficacy, the use of morphine for the treatment of chronic pain remains limited because of the rapid development of tolerance, dependence and ultimately addiction. These undesired effects are thought to be because of alterations in synaptic transmission and neuroplasticity within the reward circuitry including the striatum. In this study we used subcellular fractionation and quantitative proteomics combined with computational approaches to investigate the morphine-induced protein profile changes at the striatal postsynaptic density. Over 2,600 proteins were identified by mass spectrometry analysis of subcellular fractions enriched in postsynaptic density associated proteins from saline or morphine-treated striata. Among these, the levels of 34 proteins were differentially altered in response to morphine. These include proteins involved in G-protein coupled receptor signaling, regulation of transcription and translation, chaperones, and protein degradation pathways. The altered expression levels of several of these proteins was validated by Western blotting analysis. Using Genes2Fans software suite we connected the differentially expressed proteins with proteins identified within the known background protein-protein interaction network. This led to the generation of a network consisting of 116 proteins with 40 significant intermediates. To validate this, we confirmed the presence of three proteins predicted to be significant intermediates: caspase-3, receptor-interacting serine/threonine protein kinase 3 and NEDD4 (an E3-ubiquitin ligase identified as a neural precursor cell expressed developmentally down-regulated protein 4). Because this morphine-regulated network predicted alterations in proteasomal degradation, we examined the global ubiquitination state of postsynaptic density proteins and found it to be substantially altered. Together, these findings suggest a role for protein degradation and for the ubiquitin/proteasomal system in the etiology of opiate dependence and addiction.
Collapse
Affiliation(s)
- Steven D Stockton
- From the ‡Department of Pharmacology and Systems Therapeutics, §Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029
| | - Ivone Gomes
- From the ‡Department of Pharmacology and Systems Therapeutics
| | - Tong Liu
- ¶Center for Advanced Proteomic Research and Department of Biochemistry and Molecular Biology, New Jersey Medical School Cancer Center, Rutgers University, Newark, New Jersey, 07103
| | | | - Lucia Hipólito
- ‖Department of Anesthesiology, Columbia University Medical Center, New York, New York, 10027
| | - Matthew R Jones
- From the ‡Department of Pharmacology and Systems Therapeutics
| | - Avi Ma'ayan
- From the ‡Department of Pharmacology and Systems Therapeutics
| | - Jose A Morón
- ‖Department of Anesthesiology, Columbia University Medical Center, New York, New York, 10027
| | - Hong Li
- ¶Center for Advanced Proteomic Research and Department of Biochemistry and Molecular Biology, New Jersey Medical School Cancer Center, Rutgers University, Newark, New Jersey, 07103
| | - Lakshmi A Devi
- From the ‡Department of Pharmacology and Systems Therapeutics, §Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029;
| |
Collapse
|
10
|
Savalli G, Diao W, Berger S, Ronovsky M, Partonen T, Pollak DD. Anhedonic behavior in cryptochrome 2-deficient mice is paralleled by altered diurnal patterns of amygdala gene expression. Amino Acids 2015; 47:1367-77. [PMID: 25820768 PMCID: PMC4458264 DOI: 10.1007/s00726-015-1968-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/17/2015] [Indexed: 11/30/2022]
Abstract
Mood disorders are frequently paralleled by disturbances in circadian rhythm-related physiological and behavioral states and genetic variants of clock genes have been associated with depression. Cryptochrome 2 (Cry2) is one of the core components of the molecular circadian machinery which has been linked to depression, both, in patients suffering from the disease and animal models of the disorder. Despite this circumstantial evidence, a direct causal relationship between Cry2 expression and depression has not been established. Here, a genetic mouse model of Cry2 deficiency (Cry2 (-/-) mice) was employed to test the direct relevance of Cry2 for depression-like behavior. Augmented anhedonic behavior in the sucrose preference test, without alterations in behavioral despair, was observed in Cry2 (-/-) mice. The novelty suppressed feeding paradigm revealed reduced hyponeophagia in Cry2 (-/-) mice compared to wild-type littermates. Given the importance of the amygdala in the regulation of emotion and their relevance for the pathophysiology of depression, potential alterations in diurnal patterns of basolateral amygdala gene expression in Cry2 (-/-) mice were investigated focusing on core clock genes and neurotrophic factor systems implicated in the pathophysiology of depression. Differential expression of the clock gene Bhlhe40 and the neurotrophic factor Vegfb were found in the beginning of the active (dark) phase in Cry2 (-/-) compared to wild-type animals. Furthermore, amygdala tissue of Cry2 (-/-) mice contained lower levels of Bdnf-III. Collectively, these results indicate that Cry2 exerts a critical role in the control of depression-related emotional states and modulates the chronobiological gene expression profile in the mouse amygdala.
Collapse
Affiliation(s)
- Giorgia Savalli
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
11
|
Teuber J, Mueller B, Fukabori R, Lang D, Albrecht A, Stork O. The ubiquitin ligase Praja1 reduces NRAGE expression and inhibits neuronal differentiation of PC12 cells. PLoS One 2013; 8:e63067. [PMID: 23717400 PMCID: PMC3661586 DOI: 10.1371/journal.pone.0063067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/27/2013] [Indexed: 02/05/2023] Open
Abstract
Evidence suggests that regulated ubiquitination of proteins plays a critical role in the development and plasticity of the central nervous system. We have previously identified the ubiquitin ligase Praja1 as a gene product induced during fear memory consolidation. However, the neuronal function of this enzyme still needs to be clarified. Here, we investigate its involvement in the nerve growth factor (NGF)-induced differentiation of rat pheochromocytoma (PC12) cells. Praja1 co-localizes with cytoskeleton components and the neurotrophin receptor interacting MAGE homologue (NRAGE). We observed an enhanced expression of Praja1 after 3 days of NGF treatment and a suppression of neurite formation upon Praja1 overexpression in stably transfected PC12 cell lines, which was associated with a proteasome-dependent reduction of NRAGE levels. Our data suggest that Praja1, through ubiquitination and degradation of NRAGE, inhibits neuronal differentiation. The two murine isoforms, Praja1.1 and Praja1.2, appear to be functionally homologous in this respect.
Collapse
Affiliation(s)
- Jan Teuber
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Bettina Mueller
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ryoji Fukabori
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Daniel Lang
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Anne Albrecht
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Center for Behavioural Brain Sciences, Magdeburg, Germany
- * E-mail:
| |
Collapse
|
12
|
Effect of acute stressor and serotonin transporter genotype on amygdala first wave transcriptome in mice. PLoS One 2013; 8:e58880. [PMID: 23536833 PMCID: PMC3594195 DOI: 10.1371/journal.pone.0058880] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/07/2013] [Indexed: 12/17/2022] Open
Abstract
The most prominent brain region evaluating the significance of external stimuli immediately after their onset is the amygdala. Stimuli evaluated as being stressful actuate a number of physiological processes as an immediate stress response. Variation in the serotonin transporter gene has been associated with increased anxiety- and depression-like behavior, altered stress reactivity and adaptation, and pathophysiology of stress-related disorders. In this study the instant reactions to an acute stressor were measured in a serotonin transporter knockout mouse model. Mice lacking the serotonin transporter were verified to be more anxious than their wild-type conspecifics. Genome-wide gene expression changes in the amygdala were measured after the mice were subjected to control condition or to an acute stressor of one minute exposure to water. The dissection of amygdalae and stabilization of RNA was conducted within nine minutes after the onset of the stressor. This extremely short protocol allowed for analysis of first wave primary response genes, typically induced within five to ten minutes of stimulation, and was performed using Affymetrix GeneChip Mouse Gene 1.0 ST Arrays. RNA profiling revealed a largely new set of differentially expressed primary response genes between the conditions acute stress and control that differed distinctly between wild-type and knockout mice. Consequently, functional categorization and pathway analysis indicated genes related to neuroplasticity and adaptation in wild-types whereas knockouts were characterized by impaired plasticity and genes more related to chronic stress and pathophysiology. Our study therefore disclosed different coping styles dependent on serotonin transporter genotype even directly after the onset of stress and accentuates the role of the serotonergic system in processing stressors and threat in the amygdala. Moreover, several of the first wave primary response genes that we found might provide promising targets for future therapeutic interventions of stress-related disorders also in humans.
Collapse
|
13
|
Oliveira DR, Sanada PF, Filho ACS, Conceição GMS, Cerutti JM, Cerutti SM. Long-term treatment with standardized extract of Ginkgo biloba L. enhances the conditioned suppression of licking in rats by the modulation of neuronal and glial cell function in the dorsal hippocampus and central amygdala. Neuroscience 2013; 235:70-86. [PMID: 23321541 DOI: 10.1016/j.neuroscience.2013.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/21/2012] [Accepted: 01/08/2013] [Indexed: 01/02/2023]
Abstract
Our group previously demonstrated that short-term treatment with a standardized extract of Ginkgo biloba (EGb) changed fear-conditioned memory by modulating gene expression in the hippocampus, amygdaloid complex and prefrontal cortex. Although there are few controlled studies that support the long-term use of EGb for the prevention and/or treatment of memory impairment, the chronic use of Ginkgo is common. This study evaluated the effects of chronic treatment with EGb on the conditioned emotional response, assessed by the suppression of ongoing behavior and in the modulation of gene and protein expression. Male adult Wistar rats were treated over 28days and assigned to five groups (n=10) as follows: positive control (4mgkg(-1) Diazepam), negative control (12% Tween 80), EGb groups (0.5 and 1.0gkg(-1)) and the naïve group. The suppression of the licking response was calculated for each rat in six trials. Our results provide further evidence for the efficacy of EGb on memory. For the first time, we show that long-term treatment with the highest dose of EGb improves the fear memory and suggests that increased cAMP-responsive element-binding protein (CREB)-1 and glial fibrillary acidic protein (GFAP) mRNA and protein (P<0.001) in the dorsal hippocampus and amygdaloid complex and reduced growth and plasticity-associated protein 43 (GAP-43) (P<0.01) in the hippocampus are involved in this process. The fear memory/treatment-dependent changes observed in our study suggest that EGb might be effective for memory enhancement through its effect on the dorsal hippocampus and amygdaloid complex.
Collapse
Affiliation(s)
- D R Oliveira
- Behavior Pharmacology and Etnopharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
14
|
Ke Y, Dramiga J, Schütz U, Kril JJ, Ittner LM, Schröder H, Götz J. Tau-mediated nuclear depletion and cytoplasmic accumulation of SFPQ in Alzheimer's and Pick's disease. PLoS One 2012; 7:e35678. [PMID: 22558197 PMCID: PMC3338448 DOI: 10.1371/journal.pone.0035678] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 03/20/2012] [Indexed: 11/19/2022] Open
Abstract
Tau dysfunction characterizes neurodegenerative diseases such as Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD). Here, we performed an unbiased SAGE (serial analysis of gene expression) of differentially expressed mRNAs in the amygdala of transgenic pR5 mice that express human tau carrying the P301L mutation previously identified in familial cases of FTLD. SAGE identified 29 deregulated transcripts including Sfpq that encodes a nuclear factor implicated in the splicing and regulation of gene expression. To assess the relevance for human disease we analyzed brains from AD, Pick's disease (PiD, a form of FTLD), and control cases. Strikingly, in AD and PiD, both dementias with a tau pathology, affected brain areas showed a virtually complete nuclear depletion of SFPQ in both neurons and astrocytes, along with cytoplasmic accumulation. Accordingly, neurons harboring either AD tangles or Pick bodies were also depleted of SFPQ. Immunoblot analysis of human entorhinal cortex samples revealed reduced SFPQ levels with advanced Braak stages suggesting that the SFPQ pathology may progress together with the tau pathology in AD. To determine a causal role for tau, we stably expressed both wild-type and P301L human tau in human SH-SY5Y neuroblastoma cells, an established cell culture model of tau pathology. The cells were differentiated by two independent methods, mitomycin C-mediated cell cycle arrest or neuronal differentiation with retinoic acid. Confocal microscopy revealed that SFPQ was confined to nuclei in non-transfected wild-type cells, whereas in wild-type and P301L tau over-expressing cells, irrespective of the differentiation method, it formed aggregates in the cytoplasm, suggesting that pathogenic tau drives SFPQ pathology in post-mitotic cells. Our findings add SFPQ to a growing list of transcription factors with an altered nucleo-cytoplasmic distribution under neurodegenerative conditions.
Collapse
Affiliation(s)
- Yazi Ke
- Alzheimer's and Parkinson's Disease Laboratory, Brain & Mind Research Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Joe Dramiga
- Department II of Anatomy and Neuroanatomy, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Ulrich Schütz
- Department II of Anatomy and Neuroanatomy, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
| | - Jillian J. Kril
- Disciplines of Medicine and Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Lars M. Ittner
- Alzheimer's and Parkinson's Disease Laboratory, Brain & Mind Research Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Hannsjörg Schröder
- Department II of Anatomy and Neuroanatomy, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany
- * E-mail: (HS); (JG)
| | - Jürgen Götz
- Alzheimer's and Parkinson's Disease Laboratory, Brain & Mind Research Institute, University of Sydney, Camperdown, New South Wales, Australia
- * E-mail: (HS); (JG)
| |
Collapse
|
15
|
Fonslow BR, Niessen SM, Singh M, Wong CCL, Xu T, Carvalho PC, Choi J, Park SK, Yates JR. Single-step inline hydroxyapatite enrichment facilitates identification and quantitation of phosphopeptides from mass-limited proteomes with MudPIT. J Proteome Res 2012; 11:2697-709. [PMID: 22509746 DOI: 10.1021/pr300200x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Herein we report the characterization and optimization of single-step inline enrichment of phosphopeptides directly from small amounts of whole cell and tissue lysates (100-500 μg) using a hydroxyapatite (HAP) microcolumn and Multidimensional Protein Identification Technology (MudPIT). In comparison to a triplicate HILIC-IMAC phosphopeptide enrichment study, ∼80% of the phosphopeptides identified using HAP-MudPIT were unique. Similarly, analysis of the consensus phosphorylation motifs between the two enrichment methods illustrates the complementarity of calcium- and iron-based enrichment methods and the higher sensitivity and selectivity of HAP-MudPIT for acidic motifs. We demonstrate how the identification of more multiply phosphorylated peptides from HAP-MudPIT can be used to quantify phosphorylation cooperativity. Through optimization of HAP-MudPIT on a whole cell lysate we routinely achieved identification and quantification of ca. 1000 phosphopeptides from a ∼1 h enrichment and 12 h MudPIT analysis on small quantities of material. Finally, we applied this optimized method to identify phosphorylation sites from a mass-limited mouse brain region, the amygdala (200-500 μg), identifying up to 4000 phosphopeptides per run.
Collapse
Affiliation(s)
- Bryan R Fonslow
- Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Johansen JP, Cain CK, Ostroff LE, LeDoux JE. Molecular mechanisms of fear learning and memory. Cell 2011; 147:509-24. [PMID: 22036561 DOI: 10.1016/j.cell.2011.10.009] [Citation(s) in RCA: 727] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Indexed: 01/08/2023]
Abstract
Pavlovian fear conditioning is a particularly useful behavioral paradigm for exploring the molecular mechanisms of learning and memory because a well-defined response to a specific environmental stimulus is produced through associative learning processes. Synaptic plasticity in the lateral nucleus of the amygdala (LA) underlies this form of associative learning. Here, we summarize the molecular mechanisms that contribute to this synaptic plasticity in the context of auditory fear conditioning, the form of fear conditioning best understood at the molecular level. We discuss the neurotransmitter systems and signaling cascades that contribute to three phases of auditory fear conditioning: acquisition, consolidation, and reconsolidation. These studies suggest that multiple intracellular signaling pathways, including those triggered by activation of Hebbian processes and neuromodulatory receptors, interact to produce neural plasticity in the LA and behavioral fear conditioning. Collectively, this body of research illustrates the power of fear conditioning as a model system for characterizing the mechanisms of learning and memory in mammals and potentially for understanding fear-related disorders, such as PTSD and phobias.
Collapse
Affiliation(s)
- Joshua P Johansen
- Center for Neural Science, New York University, New York, NY 10003, USA
| | | | | | | |
Collapse
|
17
|
Abstract
In addition to its role in cellular development and proliferation, there are emerging in vitro data implicating the Wnt/β-catenin pathway in synaptic plasticity. Yet in vivo studies have not examined whether Wnt activity is required for learning and memory. In the amygdala during fear memory formation, we found that many Wnt-signaling genes were dynamically regulated, with an immediate decrease, followed by an eventual normalization during memory consolidation. This rapid decrease in Wnt mRNA was confirmed with individual quantitative PCR and in situ hybridization. We then manipulated Wnt signaling with a specific peptide antagonist (Dkk-1) or agonist (Wnt1) injected stereotaxically into the adult amygdala during fear learning. We found that neither manipulation had an effect on locomotion, anxiety, fear acquisition, or fear expression. However, both Wnt modulators prevented long-term fear memory consolidation without affecting short-term memory. Dkk-1 and Wnt infusions had destabilizing, but opposite, effects on the requisite β-catenin/cadherin dynamic interactions that occur during consolidation. These data suggest that dynamic modulation of Wnt/β-catenin signaling during consolidation is critical for the structural basis of long-term memory formation.
Collapse
|
18
|
Protein microarrays for the identification of praja1 e3 ubiquitin ligase substrates. Cell Biochem Biophys 2011; 60:127-35. [PMID: 21461837 DOI: 10.1007/s12013-011-9180-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although they are the primary determinants of substrate specificity, few E3-substrate pairs have been positively identified, and few E3's profiled in a proteomic fashion. Praja1 is an E3 implicated in bone development and highly expressed in brain. Although it has been well studied relative to the majority of E3's, little is known concerning the repertoire of proteins it ubiquitylates. We sought to identify high confidence substrates for Praja1 from an unbiased proteomic profile of thousands of human proteins using protein microarrays. We first profiled Praja1 activity against a panel of E2's to identify its optimal partner in vitro. We then ubiquitylated multiple, identical protein arrays and detected putative substrates with reagents that vary in ubiquitin recognition according to the extent of chain formation. Gene ontology clustering identified putative substrates consistent with information previously known about Praja1 function, and provides clues into novel aspects of this enzyme's function.
Collapse
|
19
|
Fioravante D, Byrne JH. Protein degradation and memory formation. Brain Res Bull 2011; 85:14-20. [PMID: 21078374 PMCID: PMC3079012 DOI: 10.1016/j.brainresbull.2010.11.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 10/21/2010] [Accepted: 11/03/2010] [Indexed: 11/16/2022]
Abstract
Long-term memories are created when labile short-term memory traces are converted to more enduring forms. This process, called consolidation, is associated with changes in the synthesis of proteins that alter the biophysical properties of neurons and the strength of their synaptic connections. Recently, it has become clear that the consolidation process requires not only protein synthesis but also degradation. Here, we discuss recent findings on the roles of ubiquitination and protein degradation in synaptic plasticity and learning and memory.
Collapse
Affiliation(s)
| | - John H. Byrne
- Dept. Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, The University of Texas Medical School at Houston, Houston TX 77030
| |
Collapse
|
20
|
Yao I, Takao K, Miyakawa T, Ito S, Setou M. Synaptic E3 ligase SCRAPPER in contextual fear conditioning: extensive behavioral phenotyping of Scrapper heterozygote and overexpressing mutant mice. PLoS One 2011; 6:e17317. [PMID: 21390313 PMCID: PMC3044740 DOI: 10.1371/journal.pone.0017317] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 01/31/2011] [Indexed: 11/22/2022] Open
Abstract
SCRAPPER, an F-box protein coded by FBXL20, is a subunit of SCF type E3 ubiquitin ligase. SCRAPPER localizes synapses and directly binds to Rab3-interacting molecule 1 (RIM1), an essential factor for synaptic vesicle release, thus it regulates neural transmission via RIM1 degradation. A defect in SCRAPPER leads to neurotransmission abnormalities, which could subsequently result in neurodegenerative phenotypes. Because it is likely that the alteration of neural transmission in Scrapper mutant mice affect their systemic condition, we have analyzed the behavioral phenotypes of mice with decreased or increased the amount of SCRAPPER. We carried out a series of behavioral test batteries for Scrapper mutant mice. Scrapper transgenic mice overexpressing SCRAPPER in the hippocampus did not show any significant difference in every test argued in this manuscript by comparison with wild-type mice. On the other hand, heterozygotes of Scrapper knockout [SCR (+/−)] mice showed significant difference in the contextual but not cued fear conditioning test. In addition, SCR (+/−) mice altered in some tests reflecting anxiety, which implies the loss of functions of SCRAPPER in the hippocampus. The behavioral phenotypes of Scrapper mutant mice suggest that molecular degradation conferred by SCRAPPER play important roles in hippocampal-dependent fear memory formation.
Collapse
Affiliation(s)
- Ikuko Yao
- Department of Medical Chemistry, Kansai Medical University, Moriguchi, Osaka, Japan
- Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo, Japan
- * E-mail: (IY); (MS)
| | - Keizo Takao
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine Kyoto University, Kyoto, Japan
- Section of Behavior Analysis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Tsuyoshi Miyakawa
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine Kyoto University, Kyoto, Japan
- Section of Behavior Analysis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Seiji Ito
- Department of Medical Chemistry, Kansai Medical University, Moriguchi, Osaka, Japan
| | - Mitsutoshi Setou
- Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo, Japan
- Department of Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
- * E-mail: (IY); (MS)
| |
Collapse
|
21
|
Role of the neural cell adhesion molecule (NCAM) in amygdalo-hippocampal interactions and salience determination of contextual fear memory. Int J Neuropsychopharmacol 2010; 13:661-74. [PMID: 20003620 DOI: 10.1017/s1461145709991106] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Evidence suggests that the neural cell adhesion molecule (NCAM) is an important molecular constituent of adaptive and maladaptive circuit (re-)organization in the central nervous system. Here, we further investigate its putative involvement in amygdala and hippocampus functions during context fear memory formation. Using laser capture microdissection and quantitative RT-PCR, we show high NCAM mRNA expression levels in the lateral and basolateral subnuclei of the amygdala, as well as their training intensity- and context-dependent regulation during fear memory consolidation. Moreover, we demonstrate that deficits of NCAM-/- mice in context fear memory can be overcome through contextual pre-exposure, i.e. by reducing the modulatory influence of the amygdala on this hippocampus-dependent memory. On the contrary, NCAM-/- mice failed to increase contextual fear memory after salient overtraining, although they adequately increased their response to auditory-cued fear stimuli. Finally, we demonstrate a reduction of amygdalo-hippocampal theta synchronization in NCAM-/- mice during fear memory retrieval. Together, these results suggest an involvement of NCAM-mediated cell recognition processes in information processing of the amygdalo-hippocampal system and in the amygdala-mediated modulation of context fear memory according to stimulus salience.
Collapse
|
22
|
Pape HC, Pare D. Plastic synaptic networks of the amygdala for the acquisition, expression, and extinction of conditioned fear. Physiol Rev 2010; 90:419-63. [PMID: 20393190 DOI: 10.1152/physrev.00037.2009] [Citation(s) in RCA: 766] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The last 10 years have witnessed a surge of interest for the mechanisms underlying the acquisition and extinction of classically conditioned fear responses. In part, this results from the realization that abnormalities in fear learning mechanisms likely participate in the development and/or maintenance of human anxiety disorders. The simplicity and robustness of this learning paradigm, coupled with the fact that the underlying circuitry is evolutionarily well conserved, make it an ideal model to study the basic biology of memory and identify genetic factors and neuronal systems that regulate the normal and pathological expressions of learned fear. Critical advances have been made in determining how modified neuronal functions upon fear acquisition become stabilized during fear memory consolidation and how these processes are controlled in the course of fear memory extinction. With these advances came the realization that activity in remote neuronal networks must be coordinated for these events to take place. In this paper, we review these mechanisms of coordinated network activity and the molecular cascades leading to enduring fear memory, and allowing for their extinction. We will focus on Pavlovian fear conditioning as a model and the amygdala as a key component for the acquisition and extinction of fear responses.
Collapse
Affiliation(s)
- Hans-Christian Pape
- Institute of Physiology, Westfaelische Wilhelms-University, Muenster, Germany; and Rutgers State University, Newark, New Jersey, USA.
| | | |
Collapse
|
23
|
Rehberg K, Bergado-Acosta JR, Koch JC, Stork O. Disruption of fear memory consolidation and reconsolidation by actin filament arrest in the basolateral amygdala. Neurobiol Learn Mem 2010; 94:117-26. [PMID: 20416387 DOI: 10.1016/j.nlm.2010.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 03/19/2010] [Accepted: 04/16/2010] [Indexed: 10/19/2022]
Abstract
The dynamic re-arrangement of actin filaments is an essential process in the plasticity of synaptic connections during memory formation. In this study, we determined in mice effects of actin filament arrest in the basolateral complex of the amygdala (BLA) at different time points after memory acquisition and re-activation, using the fungal cytotoxin phalloidin. Our data show a selective disruption of auditory cued but not contextual fear memory, when phalloidin was injected 6h after conditioning. In contrast, no effect was observed when phalloidin was applied after 24h, ruling out an interference with the retrieval or expression of conditioned fear. A comparable result was obtained after memory re-activation, hence suggesting similar actin-dependent mechanisms to be active during consolidation and reconsolidation of auditory fear memory. Biochemical analysis showed that phalloidin-mediated filament arrest leads to a transient increase of highly cross-linked actin filaments in the BLA, evident 2h after injection. Together, these observations indicate that dynamic re-arrangements of actin filaments in the BLA during a late phase of fear memory consolidation and reconsolidation are critical for fear memory storage.
Collapse
Affiliation(s)
- Kati Rehberg
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | | | | | | |
Collapse
|
24
|
Ploski JE, Park KW, Ping J, Monsey MS, Schafe GE. Identification of plasticity-associated genes regulated by Pavlovian fear conditioning in the lateral amygdala. J Neurochem 2009; 112:636-50. [PMID: 19912470 DOI: 10.1111/j.1471-4159.2009.06491.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Most recent studies aimed at defining the cellular and molecular mechanisms of Pavlovian fear conditioning have focused on protein kinase signaling pathways and the transcription factor cAMP-response element binding protein (CREB) that promote fear memory consolidation in the lateral nucleus of the amygdala (LA). Despite this progress, there still remains a paucity of information regarding the genes downstream of CREB that are required for long-term fear memory formation in the LA. We have adopted a strategy of using microarray technology to initially identify genes induced within the dentate gyrus following in vivo long-term potentiation (LTP) followed by analysis of whether these same genes are also regulated by fear conditioning within the LA. In the present study, we first identified 34 plasticity-associated genes that are induced within 30 min following LTP induction utilizing a combination of DNA microarray, qRT-PCR, and in situ hybridization. To determine whether these genes are also induced in the LA following Pavlovian fear conditioning, we next exposed rats to an auditory fear conditioning protocol or to control conditions that do not support fear learning followed by qRT-PCR on mRNA from microdissected LA samples. Finally, we asked whether identified genes induced by fear learning in the LA are downstream of the extracellular-regulated kinase/mitogen-activated protein kinase signaling cascade. Collectively, our findings reveal a comprehensive list of genes that represent the first wave of transcription following both LTP induction and fear conditioning that largely belong to a class of genes referred to as 'neuronal activity dependent genes' that are likely calcium, extracellular-regulated kinase/mitogen-activated protein kinase, and CREB-dependent.
Collapse
Affiliation(s)
- Jonathan E Ploski
- Department of Psychology, Yale University, New Haven, Connecticut 06520, USA
| | | | | | | | | |
Collapse
|
25
|
Kwapis JL, Jarome TJ, Lonergan ME, Helmstetter FJ. Protein kinase Mzeta maintains fear memory in the amygdala but not in the hippocampus. Behav Neurosci 2009; 123:844-50. [PMID: 19634944 PMCID: PMC2782955 DOI: 10.1037/a0016343] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recent work on the long-term stability of memory and synaptic plasticity has identified a potentially critical role for protein kinase Mzeta (PKMzeta). PKMzeta is a constitutively active, atypical isoform of protein kinase C that is believed to maintain long term potentiation at hippocampal synapses in vitro. In behaving animals, local inhibition of PKMzeta disrupts spatial memory in the hippocampus and conditioned taste aversion memory in the insular cortex. The role of PKMzeta in context fear memory is less clear. This study examined the role of PKMzeta in amygdala and hippocampal neurons following a standard fear conditioning protocol. The results indicate that PKMzeta inhibition in the amygdala, but not in the hippocampus, can disrupt fear memory. This suggests that PKMzeta may only maintain select forms of memory in specific brain structures and does not participate in a universal memory storage mechanism.
Collapse
|
26
|
Lamprecht R, Dracheva S, Assoun S, LeDoux JE. Fear conditioning induces distinct patterns of gene expression in lateral amygdala. GENES BRAIN AND BEHAVIOR 2009; 8:735-43. [PMID: 19689454 DOI: 10.1111/j.1601-183x.2009.00515.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The lateral nucleus of the amygdala (LA) has been implicated in the formation of long-term associative memory (LTM) of stimuli associated with danger through fear conditioning. The current study aims to detect genes that are expressed in LA following associative fear conditioning. Using oligonucleotide microarrays, we monitored gene expression in rats subjected to paired training where a tone co-terminates with a footshock, or unpaired training where the tone and footshock are presented in a non-overlapping manner. The paired protocol consistently leads to auditory fear conditioning memory formation, whereas the unpaired protocol does not. When the paired group was compared with the unpaired group 5 h after training, the expression of genes coding for the limbic system-associated membrane protein (Lsamp), kinesin heavy chain member 2 (Kif2), N-ethylmaleimide-sensitive fusion protein (NSF) and Hippocalcin-like 4 protein (Hpcal4) was higher in the paired group. These genes encode proteins that regulate neuronal axonal morphology (Lsamp, Kif2), presynaptic vesicle cycling and release (Hpcal4 and NSF), and AMPA receptor maintenance in synapses (NSF). Quantitative real-time PCR (qPCR) showed that Kif2 and Lsamp are expressed hours following fear conditioning but minutes after unpaired training. Hpcal4 is induced by paired stimulation only 5 h after the training. These results show that fear conditioning induces a unique temporal activation of molecular pathways involved in regulating synaptic transmission and axonal morphology in LA, which is different from non-associative stimulation.
Collapse
Affiliation(s)
- R Lamprecht
- Department of Neurobiology and Ethology, University of Haifa, Haifa, Israel.
| | | | | | | |
Collapse
|
27
|
Joo Y, Choi KM, Lee YH, Kim G, Lee DH, Roh GS, Kang SS, Cho GJ, Choi WS, Kim HJ. Chronic immobilization stress induces anxiety- and depression-like behaviors and decreases transthyretin in the mouse cortex. Neurosci Lett 2009; 461:121-5. [PMID: 19539719 DOI: 10.1016/j.neulet.2009.06.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 06/02/2009] [Accepted: 06/10/2009] [Indexed: 11/28/2022]
Abstract
In this study, we examined the changes in gene expression in the mouse cortex following chronic stress and behavioral tests. Mice were subjected to immobilization stress for 2h per day for 15 consecutive days and the behavior of the mice was examined. The mice in the experimental group were more anxious and depressive than the control mice. The expression of mRNA in the cortex was analyzed by microarray analysis and 63 genes were found to show a greater than twofold change in expression between the control and experimental groups. Transthyretin was further investigated because its expression showed the greatest fold change. Transthyretin mRNA expression decreased in a chronic stress-specific manner, and protein levels were reduced in the cortex but not in the choroid plexus.
Collapse
Affiliation(s)
- Yeon Joo
- Department of Anatomy and Neurobiology, Institute of Health Sciences, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju 660-751, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Oliveira DR, Sanada PF, Saragossa Filho AC, Innocenti LR, Oler G, Cerutti JM, Cerutti SM. Neuromodulatory property of standardized extract Ginkgo biloba L. (EGb 761) on memory: behavioral and molecular evidence. Brain Res 2009; 1269:68-89. [PMID: 19146837 DOI: 10.1016/j.brainres.2008.11.105] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2008] [Revised: 10/03/2008] [Accepted: 11/14/2008] [Indexed: 11/19/2022]
Abstract
Although it has been suggested that the standardized Ginkgo biloba leaf extract (Egb 761) may have a beneficial effect on memory, the cellular and molecular changes that underlie this process are not yet well defined. The present study evaluated the effects of acute (one dose) or subacute treatments (one daily dose/seven days) with EGb 761 (0.5 g kg(-1) and 1.0 g kg(-1)) on rats submitted to a conditioned emotional response (CER) in comparison with positive (4 mg kg(-1) Diazepam) and negative (12%Tween 80) control groups. To this end, eighty (n=10/group) adult, male, Wistar rats (+/-250-300 g) were used in an off-baseline CER procedure. We here observed that the rats submitted to an acute and subacute EGb 761 treatments had acquisition of fear conditioning. Additionally, we investigate if the expression of genes previously associated with classical conditioning (CREB-1 and GAP-43) and new candidate genes (GFAP) are modulated following EGb 761 acute treatment. CREB-1, GAP-43 and GFAP mRNA and protein expressions were evaluated using both quantitative PCR (qPCR) and immunohistochemical analysis, respectively. We here show, for the first time, that EGb 761 modulated GAP-43, CREB-1 and GFAP expression in the prefrontal cortex, amygdala and hippocampus. We observed an underexpression of GAP-43 in all structures evaluated and over-expression of GFAP in the amygdala and hippocampus following acute G. biloba treatment when compared to control group (Tween; p<0.01). GAP-43 expression was decreased in prefrontal cortex and hippocampus in the subacute treatment with EGb 761. Subacute treatment with EGb 761 lead to a decreased CREB-1 in mPFC (p<0.001) and increased in the hippocampus to 1.0 g kg(-1)G. biloba group (p<0.001). The results obtained from immunohistochemical analysis support our aforementioned findings and revealed that the changes in expression occurred within specific regions in the areas evaluated. All together, our findings not only provide new evidence for a role of EGb 761 on memory but also identify molecular changes that underlie the fear memory consolidation.
Collapse
Affiliation(s)
- Daniela R Oliveira
- Department of Biological Science, Federal University of Sao Paulo, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
29
|
Pistell PJ, Falls WA. Extended fear conditioning reveals a role for both N-methyl-D-aspartic acid and non-N-methyl-D-aspartic acid receptors in the amygdala in the acquisition of conditioned fear. Neuroscience 2008; 155:1011-20. [PMID: 18675886 DOI: 10.1016/j.neuroscience.2008.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/03/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
Pavlovian conditioning is a useful tool for elucidating the neural mechanisms involved with learning and memory, especially in regard to the stimuli associated with aversive events. The amygdala has been repeatedly implicated as playing a significant role in the acquisition and expression of fear. If the amygdala is critical for the acquisition of fear, then it should contribute to this processes regardless of the parameters used to induce or evaluate conditioned fear. A series of experiments using reversible inactivation techniques evaluated the role of the amygdala in the acquisition of conditioned fear when training was conducted over several days in rats. Fear-potentiated startle was used to evaluate the acquisition of conditioned fear. Pretraining infusions of N-methyl-d-aspartic acid (NMDA) or non-NMDA receptor antagonists alone into the amygdala interfered with the acquisition of fear early in training, but not later. Pretraining infusions of a cocktail consisting of both an NMDA and non-NMDA antagonist interfered with the acquisition of conditioned fear across all days of training. Taken together these results suggest the amygdala may potentially be critical for the acquisition of conditioned fear regardless of the parameters utilized.
Collapse
Affiliation(s)
- P J Pistell
- Department of Psychology, The University of Vermont, Burlington, VT 05405, USA.
| | | |
Collapse
|
30
|
Bergado-Acosta JR, Sangha S, Narayanan RT, Obata K, Pape HC, Stork O. Critical role of the 65-kDa isoform of glutamic acid decarboxylase in consolidation and generalization of Pavlovian fear memory. Learn Mem 2008; 15:163-71. [PMID: 18323571 DOI: 10.1101/lm.705408] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Evidence suggests that plasticity of the amygdalar and hippocampal GABAergic system is critical for fear memory formation. In this study we investigated in wild-type and genetically manipulated mice the role of the activity-dependent 65-kDa isozyme of glutamic acid decarboxylase (GAD65) in the consolidation and generalization of conditioned fear. First, we demonstrate a transient reduction of GAD65 gene expression in the dorsal hippocampus (6 h post training) and in the basolateral complex of the amygdala (24 h post training) during distinct phases of fear memory consolidation. Second, we show that targeted ablation of the GAD65 gene in Gad65(-/-) mice results in a pronounced context-independent, intramodal generalization of auditory fear memory during long-term (24 h or 14 d) but not short-term (30 min) memory retrieval. The temporal specificity of both gene regulation and memory deficits in Gad65 mutant mice suggests that GAD65-mediated GABA synthesis is critical for the consolidation of stimulus-specific fear memory. This function appears to involve a modulation of neural activity patterns in the amygdalo-hippocampal pathway as indicated by a reduction in theta frequency synchronization between the amygdala and hippocampus of Gad65(-/-) mice during the expression of generalized fear memory.
Collapse
Affiliation(s)
- Jorge R Bergado-Acosta
- Institute for Physiology, Otto-von-Guericke University Magdeburg, D-39120 Magdeburg, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Helmstetter FJ, Parsons RG, Gafford GM. Macromolecular synthesis, distributed synaptic plasticity, and fear conditioning. Neurobiol Learn Mem 2008; 89:324-37. [PMID: 17977027 PMCID: PMC2297466 DOI: 10.1016/j.nlm.2007.09.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2007] [Revised: 08/23/2007] [Accepted: 09/06/2007] [Indexed: 01/21/2023]
Abstract
Recent work from a number of laboratories has provided new and important insights about how gene expression is altered by experience and how these molecular changes may provide a substrate for the long-term storage of new memories. Here, we review a series of recent studies using aversive Pavlovian conditioning in rats as a well characterized model system in which experience-dependent alterations in gene expression can be manipulated and quantified within a specific neural circuit. We highlight some of the issues involved in using broad-spectrum inhibitors of mRNA and protein synthesis to study cellular changes underlying the formation and long-term stability of memory and discuss the idea that these changes occur over widespread, behaviorally-defined, networks of cells. We also discuss the idea that the maintenance of memory and its susceptibly to disruption after retrieval may relate to local protein synthesis in dendrites. Finally, a series of recent experiments from our laboratory studying the role of a specific signaling pathway (mTOR) which regulates translational processes and memory formation in the amygdala and hippocampus during fear conditioning are reviewed.
Collapse
Affiliation(s)
- Fred J Helmstetter
- Department of Psychology, University of Wisconsin, P.O. Box 413, Milwaukee, WI 53201, USA.
| | | | | |
Collapse
|
32
|
Matsuoka T, Tsunoda M, Sumiyoshi T, Takasaki I, Tabuchi Y, Seo T, Tanaka K, Uehara T, Itoh H, Suzuki M, Kurachi M. Effect of MK-801 on gene expressions in the amygdala of rats. Synapse 2008; 62:1-7. [PMID: 17948890 DOI: 10.1002/syn.20455] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rodents treated with N-methyl-D-aspartate (NMDA) antagonists have been thought to be an animal model of schizophrenia. In this study, we examined gene expression in the amygdala of rats chronically treated with MK-801, as well as behavioral changes, such as social behavior, in these animals. The social interaction test, a measure of social behavior, and locomotor activity was performed in male Wistar rats injected with MK-801 (0.13 mg/kg i.p.) or saline for 14 days. Changes in mRNA levels were analyzed using a GeneChip microarray system. Real-time quantitative PCR (RT-qPCR) assay was subsequently conducted to confirm the results of the microarray analysis. MK-801 decreased social interaction and increased locomotor activity in rats, consistent with previous reports. We found 23 downregulated genes and 16 upregulated genes, with the gene encoding arginine-vasopressin (AVP) being most downregulated, and that for transthyretin (Ttr) most upregulated. mRNA levels, quantified by RT-qPCR assay, were altered for genes related to neuropeptides (AVP, Sstr2), the arachidonic cascade (Ptgds), myelination (Mobp, Enpp2), neurotrophic factors (Igfbp2), and hormonal milieu (Ttr). Downregulation of the AVP gene in the amygdala of MK-801-treated rats may provide a basis for the ability of AVP-analogues to ameliorate the behavioral disturbances caused by blockade of the NMDA receptor. The results of this study provide an insight into the neural substrates responsible for the generation of psychotic symptoms.
Collapse
Affiliation(s)
- Tadasu Matsuoka
- Department of Neuropsychiatry, University of Toyama, Graduate School of Medicine and Pharmaceutical Sciences, Toyama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Swofford JA, DeBello WM. Transcriptome changes associated with instructed learning in the barn owl auditory localization pathway. Dev Neurobiol 2007; 67:1457-77. [PMID: 17526003 DOI: 10.1002/dneu.20458] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Owls reared wearing prismatic spectacles learn to make adaptive orienting movements. This instructed learning depends on re-calibration of the midbrain auditory space map, which in turn involves the formation of new synapses. Here we investigated whether these processes are associated with differential gene expression, using longSAGE. Newly fledged owls were reared for 8-36 days with prism or control lenses at which time the extent of learning was quantified by electrophysiological mapping. Transciptome profiles were obtained from the inferior colliculus (IC), the major site of synaptic plasticity, and the optic tectum (OT), which provides an instructive signal that controls the direction and extent of plasticity. Twenty-two differentially expressed sequence tags were identified in IC and 36 in OT, out of more than 35,000 unique tags. Of these, only four were regulated in both structures. These results indicate that regulation of two largely independent gene clusters is associated with synaptic remodeling (in IC) and generation of the instructive signal (in OT). Real-time PCR data confirmed the changes for two transcripts, ubiquitin/polyubiquitin and tyrosine 3-monooxgenase/tryotophan 5-monooxygenase activation protein, theta subunit (YWHAQ; also referred to as 14-3-3 protein). Ubiquitin was downregulated in IC, consistent with a model in which protein degradation pathways act as an inhibitory constraint on synaptogenesis. YWHAQ was up-regulated in OT, indicating a role in the synthesis or delivery of instructive information. In total, our results provide a path towards unraveling molecular cascades that link naturalistic experience with synaptic remodeling and, ultimately, with the expression of learned behavior.
Collapse
Affiliation(s)
- Janet A Swofford
- Department of Neurobiology, Physiology, and Behavior, Center for Neuroscience, University of California-Davis, Davis, CA 95616, USA
| | | |
Collapse
|
34
|
Wieland I, Weidner C, Ciccone R, Lapi E, McDonald-McGinn D, Kress W, Jakubiczka S, Collmann H, Zuffardi O, Zackai E, Wieacker P. Contiguous gene deletions involving EFNB1, OPHN1, PJA1 and EDA in patients with craniofrontonasal syndrome. Clin Genet 2007; 72:506-16. [PMID: 17941886 DOI: 10.1111/j.1399-0004.2007.00905.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Craniofrontonasal syndrome (CFNS [MIM 304110]) is an X-linked malformation syndrome characterized by craniofrontonasal dysplasia and extracranial manifestations in heterozygous females. In the majority of patients CFNS is caused by mutations in the EFNB1 gene (MIM 300035). We identified three girls with classical CFNS and mild developmental delay harboring de novo deletions of the EFNB1 gene. Applying haplotype analysis, Southern blot hybridization and array-comparative genomic hybridization, deletion of EFNB1 was found to be part of contiguous gene deletions in the patients. In one patient the deletion interval includes the genes for oligophrenin-1 (OPHN1 [MIM 300127]) and praja 1 (PJA1 [MIM 300420]). In the second patient the deletion includes OPHN1, PJA1 and the gene for ectodysplasin A (EDA [MIM 300451]). In the third patient EFNB1 gene deletion may include deletion of regulatory regions 5' of OPHN1. Previously, the OPHN1 gene has been shown to be responsible for recessive X-linked mental retardation. Although it is too early to predict the future cognitive performance of the two infant patients with contiguous gene deletions of OPHN1-EFNB1-PJA1, mild learning disabilities have been recognized in the older, third patient. It is important for genetic counseling to be aware that their male offspring may not only be carriers of CFNS but may also be affected by mental retardation and anhidrotic ectodermal dysplasia.
Collapse
Affiliation(s)
- I Wieland
- Institut für Humangenetik, Otto-von-Guericke-Universität, Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Narayanan RT, Seidenbecher T, Kluge C, Bergado J, Stork O, Pape HC. Dissociated theta phase synchronization in amygdalo- hippocampal circuits during various stages of fear memory. Eur J Neurosci 2007; 25:1823-31. [PMID: 17408428 DOI: 10.1111/j.1460-9568.2007.05437.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The amygdala and the hippocampus are critically involved in the formation and retention of fear memories. However, their precise contribution to, and their interplay during, fear memory formation are not fully understood. In the present study we investigated network activities in the amygdalo-hippocampal system of freely behaving mice at different stages of fear memory consolidation and retention. Our data show enhanced theta phase synchronization in this pathway during the retrieval of fear memory at long-term (24 h post-training), but not short-term (2 min, 30 min and 2 h post-training) stages, following both contextual and auditory cued conditioning. However, retrieval of remotely conditioned fear (30 days post-training) failed to induce an increase in synchronization despite there still being memory retention. Thus, our data indicate that the amygdalo-hippocampal interaction reflects a dynamic interaction of ensemble activities related to various stages of fear memory consolidation and/or retention, and support the notion that recent and remote memories are organized through different network principles.
Collapse
Affiliation(s)
- Rajeevan T Narayanan
- Institut für Physiologie I, Westfälische Wilhelms-Universität Münster, Robert-Koch-Str 27a, Münster, Germany
| | | | | | | | | | | |
Collapse
|
36
|
Stoppel C, Albrecht A, Pape HC, Stork O. Genes and neurons: molecular insights to fear and anxiety. GENES BRAIN AND BEHAVIOR 2006; 5 Suppl 2:34-47. [PMID: 16681799 DOI: 10.1111/j.1601-183x.2006.00229.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Experimental animal models provide an important tool for the identification of inheritable components of fear and anxiety. 'Pavlovian' fear conditioning has been tremendously successful to characterize the neuronal circuitry and cellular mechanisms of the formation, consolidation and extinction of fear memories. Here we summarize recent progress that has led to the identification of gene products contributing to such experience-dependent changes in fear and anxiety and may guide the search for genetic factors involved in the development and treatment of human anxiety disorders.
Collapse
Affiliation(s)
- C Stoppel
- Institute of Physiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | | | | |
Collapse
|
37
|
Sousa JC, Cardoso I, Marques F, Saraiva MJ, Palha JA. Transthyretin and Alzheimer's disease: where in the brain? Neurobiol Aging 2006; 28:713-8. [PMID: 16698124 DOI: 10.1016/j.neurobiolaging.2006.03.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Revised: 03/24/2006] [Accepted: 03/31/2006] [Indexed: 12/17/2022]
Abstract
Transthyretin (TTR), a carrier protein for thyroxine and retinol in plasma and cerebrospinal fluid (CSF), has been shown to bind the amyloid beta peptide. Accordingly, TTR has been suggested to protect against amyloid beta deposition, a key pathological feature in Alzheimer's disease (AD). Supporting this view are the reduced TTR levels found in CSF of patients with AD, as well as reports of altered TTR expression in the cortex and hippocampus of AD rodent models. Importantly, early characterization of TTR distribution revealed the choroid plexus as the site of TTR synthesis within the brain. To resolve this controversy we used precise laser microdissection technology to assay for TTR mRNA expression. Our results clearly demonstrate that TTR is not produced in the brain parenchyma of wild-type mice nor in two different transgenic mouse models of AD, suggesting that contamination by choroid plexus contributed to the recent results indicating TTR production in various brain regions. The relevance of TTR to AD should now take into consideration TTR production by the choroid plexus and its ability, in the CSF, to sequester the amyloid beta peptide.
Collapse
Affiliation(s)
- João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal
| | | | | | | | | |
Collapse
|
38
|
Feldker DEM, Morsink MC, Veenema AH, Datson NA, Proutski V, Lathouwers D, de Kloet ER, Vreugdenhil E. The effect of chronic exposure to highly aggressive mice on hippocampal gene expression of non-aggressive subordinates. Brain Res 2006; 1089:10-20. [PMID: 16678802 DOI: 10.1016/j.brainres.2006.02.110] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 01/31/2006] [Accepted: 02/26/2006] [Indexed: 01/12/2023]
Abstract
Exposure to a chronic psychosocial stressor changes the behavioral and neuroendocrine response pattern and causes structural changes in the rodent hippocampus. However, the underlying molecular mechanism of these changes induced by chronic stress is largely unknown. Recently, it was shown that exposure to a dominant highly aggressive mouse in the sensory contact model induced long-lasting stress symptoms in subordinate mice genetically selected for long attack latency (LAL mice). The aim of the present study was to study the effect of chronic stress on hippocampal gene expression in these subordinate LAL mice. GeneChips (Affymetrix) were used to compare gene expression profiles of LAL mice exposed to a sensory contact stressor for 25 days and their controls (one array per mouse, n=5 per line). After this stress paradigm, 131 genes were found differentially expressed (P<0.01). Strikingly, all of these genes showed a subtle downregulation in response to a chronic stressor. Interestingly, a significant overrepresentation of genes encoding structural components of ribosomes were found, suggesting diminished protein biosynthesis in the hippocampus of chronically stressed LAL mice. In addition, several genes of the NFkappaB signaling cascade, a pathway crucially involved in neuronal viability and neurite growth, were found to be downregulated. Together, we hypothesize that reduced NFkappaB signaling and diminished protein biosynthesis form part of the molecular mechanisms by which a chronic psychosocial stressor induces structural alterations in hippocampus of LAL mice.
Collapse
Affiliation(s)
- Dorine E M Feldker
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University Medical Center, PO Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Berger-Sweeney J, Zearfoss NR, Richter JD. Reduced extinction of hippocampal-dependent memories in CPEB knockout mice. Learn Mem 2006; 13:4-7. [PMID: 16452649 DOI: 10.1101/lm.73706] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
CPEB is a sequence-specific RNA binding protein that regulates translation at synapses. In neurons of CPEB knockout mice, synaptic efficacy is reduced. Here, we have performed a battery of behavioral tests and find that relative to wild-type animals, CPEB knockout mice, although similar on many baseline behaviors, have reduced extinction of memories on two hippocampal-dependent tasks. A corresponding microarray analysis reveals that about 0.14% of hippocampal genes have an altered expression in the CPEB knockout mouse. These data suggest that CPEB-dependent local protein synthesis may be an important cellular mechanism underlying extinction of hippocampal-dependent memories.
Collapse
|
40
|
Keeley MB, Wood MA, Isiegas C, Stein J, Hellman K, Hannenhalli S, Abel T. Differential transcriptional response to nonassociative and associative components of classical fear conditioning in the amygdala and hippocampus. Learn Mem 2006; 13:135-42. [PMID: 16547164 PMCID: PMC1409829 DOI: 10.1101/lm.86906] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Classical fear conditioning requires the recognition of conditioned stimuli (CS) and the association of the CS with an aversive stimulus. We used Affymetrix oligonucleotide microarrays to characterize changes in gene expression compared to naive mice in both the amygdala and the hippocampus 30 min after classical fear conditioning and 30 min after exposure to the CS in the absence of an aversive stimulus. We found that in the hippocampus, levels of gene regulation induced by classical fear conditioning were not significantly greater than those induced by CS alone, whereas in the amygdala, classical fear conditioning did induce significantly greater levels of gene regulation compared to the CS. Computational studies suggest that transcriptional changes in the hippocampus and amygdala are mediated by large and overlapping but distinct combinations of molecular events. Our results demonstrate that an increase in gene regulation in the amygdala was partially correlated to associative learning and partially correlated to nonassociative components of the task, while gene regulation in the hippocampus was correlated to nonassociative components of classical fear conditioning, including configural learning.
Collapse
Affiliation(s)
- Michael B Keeley
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Pape HC, Driesang RB, Heinbockel T, Laxmi TR, Meis S, Seidenbecher T, Szinyei C, Frey U, Stork O. Cellular processes in the amygdala: gates to emotional memory? ZOOLOGY 2006; 104:232-40. [PMID: 16351838 DOI: 10.1078/0944-2006-00029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The amygdala is considered a core structure of the so-called limbic system and has been implicated in a variety of functions, including emotional interpretation of sensory information, emotional arousal, emotional memory, fear and anxiety, and related clinical disorders. Despite the clinical and functional importance of the amygdala, it is only recently that some general principles of intra-amygdaloid mechanisms of signal processing that are relevant for fear behavior and memory have emerged from behavioral, anatomical, electrophysiological, and neurochemical studies performed in the amygdala of various mammalian species in vivo, in situ and in vitro.
Collapse
Affiliation(s)
- H C Pape
- Institute of Physiology, Medical School, Otto-von-Guericke-University, Magdeburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kai N, Iwase K, Imai K, Nakahira E, Soma M, Ohtsuka S, Yagi T, Kobayashi K, Koga H, Takiguchi M, Yuasa S. Altered gene expression in the subdivisions of the amygdala of Fyn-deficient mice as revealed by laser capture microdissection and mKIAA cDNA array analysis. Brain Res 2006; 1073-1074:60-70. [PMID: 16427614 DOI: 10.1016/j.brainres.2005.12.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Revised: 11/18/2005] [Accepted: 12/11/2005] [Indexed: 11/20/2022]
Abstract
Fyn-tyrosine-kinase-deficient mice exhibit increased fearfulness and display enhanced excitability in the amygdala. To gain insight into the molecular changes associated with the increased excitability of the amygdala, we used a newly developed cDNA array system comprising mouse KIAA cDNA clones to identify novel genes differentially expressed in the amygdala of fyn(-/-) and fyn(+/-) mice following administration of N-methyl-D-aspartate (NMDA). Laser capture microdissection in combination with PCR-based cDNA amplification allowed us to analyze gene expression in each amygdalar subdivision. The statistical significance of the differential expressions was tested by one-way analysis of variance (ANOVA) by the false discovery rate controlling approach. Among the 805 mKIAA cDNA clones tested, only the expression level of mKIAA1577 (Zinc finger SWIM domain containing protein 6; gene name, Zswim6) showed statistically significant change in regard to the genotype and amygdalar subdivision. Namely, only the lowered expression of mKIAA1577 in the central nucleus of fyn(-/-) mice 1 h after NMDA administration (2.1-fold lower relative to fyn(+/-) mice) was statistically significant. In situ hybridization analysis confirmed the downregulation of the mRNA in the central nucleus of the fyn(-/-) mice 1 h after NMDA administration (3.2-fold lower relative to fyn(+/-) mice). The NMDA-induced change in gene expression was partially blocked by the NMDA antagonist D-AP-5. These results suggest that Fyn deficiency was responsible for the NMDA-induced downregulation of a specific gene in the amygdalar central nucleus.
Collapse
Affiliation(s)
- Nobuyuki Kai
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Mei B, Li C, Dong S, Jiang CH, Wang H, Hu Y. Distinct gene expression profiles in hippocampus and amygdala after fear conditioning. Brain Res Bull 2005; 67:1-12. [PMID: 16140156 DOI: 10.1016/j.brainresbull.2005.03.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 03/17/2005] [Accepted: 03/28/2005] [Indexed: 11/23/2022]
Abstract
It is well known that the hippocampus and amygdala are involved in the formations of fear conditioning memories, and both contextual and cued fear memory requires activation of the NMDA receptors. However, the global molecular responses in the hippocampus and amygdala have not been investigated. By applying high-density microarrays containing 11,000 genes and expressed sequence tags, we examined fear conditioning-induced gene expression profiles in these two brain regions at 0.5, 6, and 24 h. We found that 222 genes in the amygdala and 145 genes in the hippocampus showed dynamic changes in their expression levels. Surprisingly, the overall patterns of gene expression as well as the individual genes for the amygdala and hippocampus were drastically different and only small number of genes exhibited the similar regulation in both brain regions. Based on expression kinetics, the genes from the amygdala can be further grouped into eight unique clusters, whereas the genes from the hippocampus were placed into six clusters. Therefore, our study suggests that different genomic responses are initiated in the hippocampus and amygdala which are known to play distinct roles in fear memory formation.
Collapse
Affiliation(s)
- Bing Mei
- Key Lab of Brain Functional Genomics, MOE & STCSM, Shanghai Institute of Brain Functional Genomics, East China Normal University, 3663 Zhongshan Road N., Shanghai 200062, China
| | | | | | | | | | | |
Collapse
|
45
|
Salzmann J, Canestrelli C, Noble F, Marie-Claire C. Analysis of transcriptional responses in the mouse dorsal striatum following acute 3,4-methylenedioxymethamphetamine (ecstasy): identification of extracellular signal-regulated kinase-controlled genes. Neuroscience 2005; 137:473-82. [PMID: 16289835 PMCID: PMC1993848 DOI: 10.1016/j.neuroscience.2005.09.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Revised: 08/05/2005] [Accepted: 09/20/2005] [Indexed: 11/21/2022]
Abstract
3,4-Methylenedioxymethamphetamine (ecstasy), a widely used recreational drug with psychoactive properties, induces both serotonin and dopamine release in the brain. However, little is known about its intracellular effects. We previously showed that 3,4-methylenedioxymethamphetamine rewarding effects in mice were dependent upon extracellular signal-regulated kinase activation and that dorsal striatum was a critical region for mediating extracellular signal-regulated kinase-dependent Egr1 3,4-methylenedioxymethamphetamine-induced transcription. Here, we extend these findings by showing that 3,4-methylenedioxymethamphetamine is indeed able to activate extracellular signal-regulated kinase within this structure. To identify genes regulated by acute 3,4-methylenedioxymethamphetamine in the mice dorsal striatum, and selectively controlled by this kinase, we performed microarray experiments by using a selective inhibitor of extracellular signal-regulated kinase activation, SL327. Of the approximately 24,000 genes from the microarray, 27 showed altered expression after exposure to 3,4-methylenedioxymethamphetamine, and among these, 59% were partially or totally inhibited by SL327 pretreatment. Our results showed that the genes regulated by 3,4-methylenedioxymethamphetamine encode proteins that belong to transcription factors family, signaling pathways (phosphatases, cytoskeleton regulation), and synaptic functions. These early changes, and especially those controlled by extracellular signal-regulated kinase activation might play significant roles in the expression of many of the behaviors that occur following 3,4-methylenedioxymethamphetamine taking.
Collapse
|
46
|
Stoss O, Novoyatleva T, Gencheva M, Olbrich M, Benderska N, Stamm S. p59(fyn)-mediated phosphorylation regulates the activity of the tissue-specific splicing factor rSLM-1. Mol Cell Neurosci 2005; 27:8-21. [PMID: 15345239 DOI: 10.1016/j.mcn.2004.04.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2003] [Revised: 04/22/2004] [Accepted: 04/29/2004] [Indexed: 02/08/2023] Open
Abstract
The Sam68-like mammalian protein SLM-1 is a member of the STAR protein family and is related to SAM68 and SLM-2. Here, we demonstrate that rSLM-1 interacts with itself, scaffold-attachment factor B, YT521-B, SAM68, rSLM-2, SRp30c, and hnRNP G. rSLM-1 regulates splice site selection in vivo via a purine-rich enhancer. In contrast to the widely expressed SAM68 and rSLM-2 proteins, rSLM-1 is found primarily in brain and, to a much smaller degree, in testis. In the brain, rSLM-1 and rSLM-2 are predominantly expressed in different neurons. In the hippocampal formation, rSLM-1 is present only in the dentate gyrus, whereas rSLM-2 is found in the pyramidal cells of the CA1, CA3, and CA4 regions. rSLM-1, but not rSLM-2, is phosphorylated by p59(fyn). p59(fyn)-mediated phosphorylation abolishes the ability of rSLM-1 to regulate splice site selection, but has no effect on rSLM-2 activity. This suggests that rSLM-1-positive cells could respond with a change of their splicing pattern to p59(fyn) activation, whereas rSLM-2-positive cells would not be affected. Together, our data indicate that rSLM-1 is a tissue-specific splicing factor whose activity is regulated by tyrosine phosphorylation signals emanating from p59(fyn).
Collapse
Affiliation(s)
- Oliver Stoss
- Klinikum Kassel, Pathology, Mönchebergstr. 41-43, D-34125 Kassel, Germany
| | | | | | | | | | | |
Collapse
|
47
|
Pintér M, Lent DD, Strausfeld NJ. Memory consolidation and gene expression in Periplaneta americana. Learn Mem 2005; 12:30-8. [PMID: 15647592 PMCID: PMC548493 DOI: 10.1101/lm.87905] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A unique behavioral paradigm has been developed for Periplaneta americana that assesses the timing and success of memory consolidation leading to long-term memory of visual-olfactory associations. The brains of trained and control animals, removed at the critical consolidation period, were screened by two-directional suppression subtractive hybridization. Screens identified neurobiologically relevant as well as novel genes that are differentially expressed at the consolidation phase of memory. The differential expression of six transcripts was confirmed with real-time RT-PCR experiments. There are mitochondrial DNA encoded transcripts among the up-regulated ones (COX, ATPase6). One of the confirmed down-regulated transcripts is RNA polymerase II largest subunit. The mitochondrial genes are of particular interest because mitochondria represent autonomous DNA at synapses. These transcripts will be used as one of several tools in the identification of neuronal circuits, such as in the mushroom bodies, that are implicated in memory consolidation.
Collapse
Affiliation(s)
- Marianna Pintér
- Arizona Research Laboratories, Division of Neurobiology, The University of Arizona, Tucson, Arizona 85721, USA.
| | | | | |
Collapse
|
48
|
Rho S, Kang M, Choi B, Sim D, Lee J, Lee E, Cho C, Oh JW, Park S, Ko S, Shin M, Hong M, Bae H. Effects of Yukmijihwang-tang Derivatives (YMJd), a Memory Enhancing Herbal Extract, on the Gene-Expression Profile in the Rat Hippocampus. Biol Pharm Bull 2005; 28:87-93. [PMID: 15635169 DOI: 10.1248/bpb.28.87] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The herbal extract Yukmijihwang-tang (YMJ) has been widely used for centuries as an anti-aging herbal medicine in Asian countries. Among the various modified prescriptions of YMJ, YMJ derivatives (YMJd) were formulated to enhance memory retention. This study has three goals: 1) to quantitatively evaluate the memory-enhancing effect of YMJd using behavior tasks; 2) to use cDNA micro-array tools to identify candidate genes responsible for enhancing memory; and 3) to statistically evaluate the specific gene expression patterns using Real-time PCR. Memory retention abilities are addressed by the passive avoidance task with SD male rat. The retention time of the YMJd group was significantly delayed (ca. 100%), whereas with Ginkgo biloba and Soya lecithin treatment, this was only delayed 20% and 10%, respectively. The cDNA from the hippocampi of YMJd and rat control groups were applied to an Incyte rat GEM2 cDNA microarray. The microarray results showed that transthyretin and PEP-19 were abundantly expressed in the YMJd treated group. Importantly, PEP-19 is a neuron-specific protein that inhibits apoptotic processes. On the other hand, neuronal genes involved in neuronal death or neurodegeneration, such as pentraxin and spectrin, were abundantly expressed in the control group. The list of differentially expressed genes may provide further insight into the action and mechanism behind the memory-enhancing effect of herbal extracts of YMJd.
Collapse
Affiliation(s)
- Samwoong Rho
- College of Oriental Medicine, Kyung-Hee University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Learning and memory processes are thought to underlie a variety of human psychiatric disorders, including generalised anxiety disorder and post-traumatic stress disorder. Basic research performed in laboratory animals may help to elucidate the aetiology of the respective diseases. This chapter gives a short introduction into theoretical and practical aspects of animal experiments aimed at investigating acquisition, consolidation and extinction of aversive memories. It describes the behavioural paradigms most commonly used as well as neuroanatomical, cellular and molecular correlates of aversive memories. Finally, it discusses clinical implications of the results obtained in animal experiments in respect to the development of novel pharmacotherapeutic strategies for the treatment of human patients.
Collapse
Affiliation(s)
- C T Wotjak
- Research Group Neuronal Plasticity/Mouse Behaviour, Max-Planck-Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany.
| |
Collapse
|
50
|
Rodrigues SM, Schafe GE, LeDoux JE. Molecular Mechanisms Underlying Emotional Learning and Memory in the Lateral Amygdala. Neuron 2004; 44:75-91. [PMID: 15450161 DOI: 10.1016/j.neuron.2004.09.014] [Citation(s) in RCA: 359] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fear conditioning is a valuable behavioral paradigm for studying the neural basis of emotional learning and memory. The lateral nucleus of the amygdala (LA) is a crucial site of neural changes that occur during fear conditioning. Pharmacological manipulations of the LA, strategically timed with respect to training and testing, have shed light on the molecular events that mediate the acquisition of fear associations and the formation and maintenance of long-term memories of those associations. Similar mechanisms have been found to underlie long-term potentiation (LTP) in LA, an artificial means of inducing synaptic plasticity and a physiological model of learning and memory. Thus, LTP-like changes in synaptic plasticity may underlie fear conditioning. Given that the neural circuit underlying fear conditioning has been implicated in emotional disorders in humans, the molecular mechanisms of fear conditioning are potential targets for psychotherapeutic drug development.
Collapse
|