1
|
Sordo L, Ubele MF, Boaz KA, Mefford JL, Jones ED, McCarty KL, van Rooyen HY, Smiley J, Bembenek Bailey SA, Perpich JA, Meacham B, Powell DK, Bresch F, Crump JW, Phelan MJ, Noche JA, Stark CE, Puskás LG, Norris CM, Head E. Calcineurin/NFAT inhibitors maintain cognition in a preclinical prevention study in an aging canine model of Alzheimer disease. Neurobiol Aging 2024; 146:1-14. [PMID: 39547056 DOI: 10.1016/j.neurobiolaging.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Brain signaling of calcineurin (CN) and nuclear factor of activated T-cells (NFAT) transcription factor increases in Alzheimer disease (AD) and is associated with synaptic loss, neurodegeneration, neuroinflammation, amyloid-β (Aβ) production, and cognitive decline. CN/NFAT inhibitors ameliorate these neuropathologies in mouse models of AD. Further, chronic use of tacrolimus in transplant patients reduces risk of AD. Beagles naturally develop Aβ plaques and cognitive dysfunction. We evaluated the impact of FDA-approved CN inhibitor, tacrolimus, and experimental NFAT inhibitor, Q134R, on cognitive outcomes during a three-year prevention study in 37 middle-aged beagles. While beagles treated with CN/NFAT inhibitors showed differences in the pattern of cognitive maintenance and duration of their effect, there was improvement in spatial learning, as well as maintenance of memory, attention, and working memory relative to placebo dogs. CN/NFAT inhibition is a promising target for prevention of cognitive decline that may be rapidly implemented in human clinical trials.
Collapse
Affiliation(s)
- Lorena Sordo
- Department of Pathology and Laboratory Medicine, University of California, 837 Health Sciences Rd., Irvine, CA 92697, USA
| | - Margo F Ubele
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Kathy A Boaz
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Jennifer L Mefford
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Erin Dehnart Jones
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Katie L McCarty
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Hollie Y van Rooyen
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Jeffrey Smiley
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Stasia A Bembenek Bailey
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Jessica A Perpich
- Division of Laboratory Animal Resources, University of Kentucky, 204 Health Sciences Research Building, Lexington, KY 19104, USA
| | - Beverly Meacham
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, 740 Rose Street, Lexington, KY 19104, USA
| | - David K Powell
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, 740 Rose Street, Lexington, KY 19104, USA
| | - Frederick Bresch
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Jacob W Crump
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Michael J Phelan
- Department of Pathology and Laboratory Medicine, University of California, 837 Health Sciences Rd., Irvine, CA 92697, USA
| | - Jessica A Noche
- Department of Neurobiology and Behavior, Department of Cognitive Sciences, University of California, 2205 McGaugh Hall, Irvine, CA 92697, USA
| | - Craig E Stark
- Department of Neurobiology and Behavior, Department of Cognitive Sciences, University of California, 2205 McGaugh Hall, Irvine, CA 92697, USA
| | | | - Christopher M Norris
- Sanders Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, 800 S. Limestone Street, Lexington, KY 19104, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, 837 Health Sciences Rd., Irvine, CA 92697, USA.
| |
Collapse
|
2
|
Dong L, Yuan S, Wei G, Zhu P, Ma S, Xu B, Yang Y. Artificial Optoelectronic Synapse Based on Violet Phosphorus Microfiber Arrays. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2306998. [PMID: 37963849 DOI: 10.1002/smll.202306998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/31/2023] [Indexed: 11/16/2023]
Abstract
Memristor-based artificial synapses are regarded as the most promising candidate to develop brain-like neuromorphic network computers and overcome the bottleneck of Von-Neumann architecture. Violet phosphorus (VP) as a new allotrope of available phosphorus with outstanding electro-optical properties and stability has attracted more and more attention in the past several years. In this study, large-scale, high-yield VP microfiber vertical arrays have been successfully developed on a Sn-coated graphite paper and are used as the memristor functional layers to build reliable, low-power artificial synaptic devices. The VP devices can well mimic the major synaptic functions such as short-term memory (STM), long-term memory (LTM), paired-pulse facilitation (PPF), spike timing-dependent plasticity (STDP), and spike rate-dependent plasticity (SRDP) under both electrical and light stimulation conditions, even the dendritic synapse functions and simple logical operations. By virtue of the excellent performance, the VP artificial synapse devices can be conductive to building high-performance optic-neural synaptic devices simulating the human-like optic nerve system. On this basis, Pavlov's associative memory can be successfully implemented optically. This study provides a promising approach for the design and manufacture of VP-based artificial synaptic devices and outlines a direction with multifunctional neural devices.
Collapse
Affiliation(s)
- Liyan Dong
- Materials Institute of Atomic and Molecular Science, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Shuai Yuan
- Materials Institute of Atomic and Molecular Science, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Guodong Wei
- Materials Institute of Atomic and Molecular Science, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Peifen Zhu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65211, USA
| | - Shufang Ma
- Materials Institute of Atomic and Molecular Science, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
| | - Bingshe Xu
- Materials Institute of Atomic and Molecular Science, Shaanxi University of Science and Technology, Xi'an, 710021, P. R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030024, P. R. China
| | - Ya Yang
- CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Micro-nano Energy and Sensor, Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, P. R. China
| |
Collapse
|
3
|
Malter JS. Pin1 and Alzheimer's disease. Transl Res 2023; 254:24-33. [PMID: 36162703 PMCID: PMC10111655 DOI: 10.1016/j.trsl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is an immense and growing public health crisis. Despite over 100 years of investigation, the etiology remains elusive and therapy ineffective. Despite current gaps in knowledge, recent studies have identified dysfunction or loss-of-function of Pin1, a unique cis-trans peptidyl prolyl isomerase, as an important step in AD pathogenesis. Here I review the functionality of Pin1 and its role in neurodegeneration.
Collapse
Affiliation(s)
- James S Malter
- Department of Pathology, UT Southwestern Medical Center, 5333 Harry Hines Blvd, Dallas, TX 75390.
| |
Collapse
|
4
|
Norris CM. Is It Time to Repurpose Calcineurin Inhibitors for the Treatment of Cognitive Impairment and Dementia? J Alzheimers Dis 2023; 95:599-602. [PMID: 37661889 PMCID: PMC10947791 DOI: 10.3233/jad-230780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Numerous preclinical and human tissue studies implicate the protein phosphatase calcineurin (CN) as a pathophysiologic mechanism in Alzheimer's disease (AD) and other neurodegenerative conditions. Using public electronic records of tens of thousands of individuals across the United States, Silva et al. (2023) show that use of the FDA-approved CN inhibitor, tacrolimus (for purposes of immunosuppression) is also associated with reduced prevalence of dementia-related symptoms. Notably, the study controls for age, sex, and race as well as multiple risk factors for AD. The results suggest that tacrolimus, and possibly other immunosuppressants could be repurposed for the treatment of AD-related dementia.
Collapse
Affiliation(s)
- Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
5
|
Liu C, Liu T, Lv Z, Qin M, Qu Z, Zhang Z, Li F, Chen D, Zhang X, Chen XL, Shen M. A Calcineurin Regulator MoRCN1 Is Important for Asexual Development, Stress Response, and Plant Infection of Magnaporthe oryzae. FRONTIERS IN PLANT SCIENCE 2022; 13:925645. [PMID: 35783935 PMCID: PMC9244802 DOI: 10.3389/fpls.2022.925645] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 06/12/2023]
Abstract
The calcium/calcineurin signaling pathway plays a key role in the development and virulence of plant pathogenic fungi, but the regulation of this signaling pathway is still not clear. In this study, we identified a calcineurin regulator MoRCN1 in the plant pathogenic fungus Magnaporthe oryzae and found it is important for virulence by regulating the calcineurin pathway. MoRCN1 deletion mutants were severely decreased in colony growth and conidia formation. More importantly, the deletion of MoRCN1 led to a significant reduction in virulence due to defects in appressorium formation and invasive growth. The ΔMorcn1 mutants were more sensitive to different stresses and induced host ROS accumulation, suggesting a role of MoRCN1 in stress adaptation. We found that MoRCN1 directly interacted with the calcineurin catalytic subunit MoCNA and affected its protein stability, which was therefore important for regulating the calcineurin pathway. Transcriptome analysis showed that MoRCN1 significantly activated 491 genes and suppressed 337 genes in response to calcium ion, partially overlapped with the MoCRZ1-bound genes. Gene Ontology and KEGG pathway analyses indicated that MoRCN1-regulated genes were enriched in stress adaptation, lipid metabolism, and secondary metabolite biosynthesis, reflecting a function of MoRCN1 in host cell adaptation. Altogether, these results suggest MoRCN1 functions as a regulator of the calcium/calcineurin signaling pathway for fungal development and infection of host cells.
Collapse
Affiliation(s)
- Caiyun Liu
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, China
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Tiangu Liu
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ziwei Lv
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mengyuan Qin
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhiguang Qu
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ziwei Zhang
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Fuyan Li
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Deng Chen
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xinrong Zhang
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiao-Lin Chen
- State Key Laboratory of Agricultural Microbiology and Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mi Shen
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, College of Biology and Agricultural Resources, Huanggang Normal University, Huanggang, China
| |
Collapse
|
6
|
Fujii H, Bito H. Deciphering Ca2+-controlled biochemical computation governing neural circuit dynamics via multiplex imaging. Neurosci Res 2022; 179:79-90. [DOI: 10.1016/j.neures.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/25/2022]
|
7
|
Miura E, Watanabe M. Coexpression of calcineurin A and B subunits in various subcellular and synaptic compartments of cerebellar neurons and glia with particular abundance at parallel fiber-Purkinje cell synapses. Neurosci Res 2022; 180:13-22. [PMID: 35247520 DOI: 10.1016/j.neures.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 10/19/2022]
Abstract
Calcineurin (CN) is a Ca2+/calmodulin-dependent serine/threonine protein phosphatase consisting of catalytic CNA and regulatory CNB subunits, and links activity-dependent Ca2+ signals to various neural functions. Here we studied CN expression in the mouse brain by producing subunit-specific probes and antibodies. Of five CN subunits. CNAα, CNAβ, and CNB1 mRNAs were predominantly expressed over the brain from early embryonic to adult stage, and all were high in the telencephalon and cerebellum. Protein localization was examined in the cerebellum by immunofluorescence with cellular and terminal markers and by preembedding silver-enhanced immunogold microscopy. CNB1 and CNAβ were co-distributed in subcellular and synaptic elements of various cerebellar neurons and glia, whereas CNAα was exclusive in granule cell elements, including parallel fiber terminals. The present study thus discloses that CNB1 subunit well coexists with one or two CNA subunits in various cerebellar compartments. Moreover, high CN contents are provided to parallel fiber-Purkinje cell synapses, i.e., CNAα, CNAβ, and CNB1 in their presynaptic side and CNAβ and CNB1 in their postsynaptic side. These findings will be the anatomical basis, at least partly, for the known regulatory roles of postsynaptic CNs in long-term depression and presynaptic CNs in transmitter release function.
Collapse
Affiliation(s)
- Eriko Miura
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| |
Collapse
|
8
|
Raha S, Ghosh A, Dutta D, Patel DR, Pahan K. Activation of PPARα enhances astroglial uptake and degradation of β-amyloid. Sci Signal 2021; 14:eabg4747. [PMID: 34699252 DOI: 10.1126/scisignal.abg4747] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Arunava Ghosh
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Dhruv R Patel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
9
|
Hill EM, Howard CD, Bale TL, Jašarević E. Perinatal exposure to tetracycline contributes to lasting developmental effects on offspring. Anim Microbiome 2021; 3:37. [PMID: 33975649 PMCID: PMC8111738 DOI: 10.1186/s42523-021-00099-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND For more than 30 years, the tetracycline on/off system of inducible gene expression has been leveraged to study disease mechanisms across many research areas, especially that of metabolism and neuroscience. This system requires acute or chronic exposure to tetracycline derivatives, such as doxycycline, to manipulate gene expression in a temporal and tissue-specific manner, with exposure often being restricted to gestational and early developmental windows. Despite evidence showing that early life antibiotic exposure has adverse effects on gut microbiota, metabolism, physiology, immunity and behavior, little is known regarding the lasting impact of doxycycline treatment on relevant outcomes in experimental offspring. RESULTS To examine the hypothesis that early life doxycycline exposure produces effects on offspring growth, behavior, and gut microbiota, we employed the most commonly used method for tetracycline on/off system by administering a low dose of doxycycline (0.5 mg/ml) in the drinking water to C57Bl/6J and C57BL/6J:129S1/SvImJ dams from embryonic day 15.5 to postnatal day 28. Developmental exposure to low dose doxycycline resulted in significant alterations to growth trajectories and body weight in both strains, which persisted beyond cessation of doxycycline exposure. Developmental doxycycline exposure influenced offspring bacterial community assembly in a temporal and sex-specific manner. Further, gut microbiota composition failed to recover by adulthood, suggesting a lasting imprint of developmental antibiotic exposure. CONCLUSIONS Our results demonstrated that early life doxycycline exposure shifts the homeostatic baseline of prior exposed animals that may subsequently impact responses to experimental manipulations. These results highlight the gut microbiota as an important factor to consider in systems requiring methods of chronic antibiotic administration during pregnancy and critical periods of postnatal development.
Collapse
Affiliation(s)
- Elizabeth M Hill
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher D Howard
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tracy L Bale
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eldin Jašarević
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Serine/Threonine Phosphatases in LTP: Two B or Not to Be the Protein Synthesis Blocker-Induced Impairment of Early Phase. Int J Mol Sci 2021; 22:ijms22094857. [PMID: 34064311 PMCID: PMC8125358 DOI: 10.3390/ijms22094857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/25/2022] Open
Abstract
Dephosphorylation of target proteins at serine/threonine residues is one of the most crucial mechanisms regulating their activity and, consequently, the cellular functions. The role of phosphatases in synaptic plasticity, especially in long-term depression or depotentiation, has been reported. We studied serine/threonine phosphatase activity during the protein synthesis blocker (PSB)-induced impairment of long-term potentiation (LTP). Established protein phosphatase 2B (PP2B, calcineurin) inhibitor cyclosporin A prevented the LTP early phase (E-LTP) decline produced by pretreatment of hippocampal slices with cycloheximide or anisomycin. For the first time, we directly measured serine/threonine phosphatase activity during E-LTP, and its significant increase in PSB-treated slices was demonstrated. Nitric oxide (NO) donor SNAP also heightened phosphatase activity in the same manner as PSB, and simultaneous application of anisomycin + SNAP had no synergistic effect. Direct measurement of the NO production in hippocampal slices by the NO-specific fluorescent probe DAF-FM revealed that PSBs strongly stimulate the NO concentration in all studied brain areas: CA1, CA3, and dentate gyrus (DG). Cyclosporin A fully abolished the PSB-induced NO production in the hippocampus, suggesting a close relationship between nNOS and PP2B activity. Surprisingly, cyclosporin A alone impaired short-term plasticity in CA1 by decreasing paired-pulse facilitation, which suggests bi-directionality of the influences of PP2B in the hippocampus. In conclusion, we proposed a minimal model of signaling events that occur during LTP induction in normal conditions and the PSB-treated slices.
Collapse
|
11
|
Abstract
Neuromorphic devices and systems have attracted attention as next-generation computing due to their high efficiency in processing complex data. So far, they have been demonstrated using both machine-learning software and complementary metal-oxide-semiconductor-based hardware. However, these approaches have drawbacks in power consumption and learning speed. An energy-efficient neuromorphic computing system requires hardware that can mimic the functions of a brain. Therefore, various materials have been introduced for the development of neuromorphic devices. Here, recent advances in neuromorphic devices are reviewed. First, the functions of biological synapses and neurons are discussed. Also, deep neural networks and spiking neural networks are described. Then, the operation mechanism and the neuromorphic functions of emerging devices are reviewed. Finally, the challenges and prospects for developing neuromorphic devices that use emerging materials are discussed.
Collapse
Affiliation(s)
- Min-Kyu Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Youngjun Park
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Ik-Jyae Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jang-Sik Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| |
Collapse
|
12
|
Moreno A. Molecular mechanisms of forgetting. Eur J Neurosci 2020; 54:6912-6932. [DOI: 10.1111/ejn.14839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Andrea Moreno
- Danish Institute of Translational Neuroscience (DANDRITE) Aarhus University Aarhus C Denmark
| |
Collapse
|
13
|
Poon CH, Tse LSR, Lim LW. DNA methylation in the pathology of Alzheimer's disease: from gene to cognition. Ann N Y Acad Sci 2020; 1475:15-33. [PMID: 32491215 DOI: 10.1111/nyas.14373] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a debilitating disorder that manifests with amyloid beta plaque deposition, neurofibrillary tangles, neuronal loss, and severe cognitive impairment. Although much effort has been made to decipher the pathogenesis of this disease, the mechanisms causing these detrimental outcomes remain obscure. Over the past few decades, neuroepigenetics has emerged as an important field that, among other things, explores how reversible modifications can change gene expression to control behavior and cognitive abilities. Among epigenetic modifications, DNA methylation requires further elucidation for the conflicting observations from AD research and its pivotal role in learning and memory. In this review, we focus on the essential components of DNA methylation, the effects of aberrant methylation on gene expressions in the amyloidogenic pathway and neurochemical processes, as well as memory epigenetics in Alzheimer's disease.
Collapse
Affiliation(s)
- Chi Him Poon
- Neuromodulation Laboratory, Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, the University of Hong Kong, Hong Kong, P. R. China
| | - Long Sum Rachel Tse
- Neuromodulation Laboratory, Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, the University of Hong Kong, Hong Kong, P. R. China
| | - Lee Wei Lim
- Neuromodulation Laboratory, Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, the University of Hong Kong, Hong Kong, P. R. China
| |
Collapse
|
14
|
Yu H, He L, Li ZQ, Li N, Ou-Yang YY, Huang GH. Altering of host larval (Spodoptera exigua) calcineurin activity in response to ascovirus infection. PEST MANAGEMENT SCIENCE 2020; 76:1048-1059. [PMID: 31515935 DOI: 10.1002/ps.5615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Calcineurin (CaN) is involved in numerous cellular processes and Ca2+ -dependent signal transduction pathways. According to our previous transcriptome studies, thousands of host larval (Spodoptera exigua) transcripts were downregulated after the infection of Heliothis virescent ascovirus 3h (HvAV-3h), while the Spodoptera exigua calcineurin genes (SeCaNs) were significantly upregulated. To understand the regulation of SeCaNs in S. exigua larvae during the infection of HvAV-3h, the functions of CaN subunit A (SeCaN-SubA) and CaN binding protein (SeCaN-BP) were analysed. RESULTS The in vitro assays indicated that the bacterial expressed SeCaN-SubA is an acid phosphatase, but no phosphatase activity was detected with the purified SeCaN-BP. The transcription level of SeCaN-SubA was upregulated after HvAV-3h infection and the CaN activity was significantly increased after HvAV-3h infection in S. exigua larvae. Interestingly, the SeCaN-BP transcripts were only detectable in the HvAV-3h infected larvae. Further immunoblotting results consistently agree with those obtained by qPCR, indicating that the infection of HvAV-3h causes the upregulated expression of SeCaN-SubA and the appearance of SeCaN-BP. An interaction between the cleaved SeCaN-SubA and SeCaN-BP was detected by co-immunoprecipitation assays, and the expression of SeCaN-BP in Spodoptera frugiperda-9 (Sf9) cells can help to increase the CaN activity of SeCaN-SubA. Further investigations with CaN inhibitors suggested that HvAV-3h. Further investigations with CaN inhibitors suggested that the inhibition on host larval CaN activity can also inhibit the viral replication of HvAV-3h. CONCLUSION The increase in CaN activity caused by HvAV-3h infection might be due to the upregulation of SeCaN-SubA and the induced expression of SeCaN-BP, and increased CaN activity is essential for ascoviral replication. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Huan Yu
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, P.R. China
- College of Plant Protection, Hunan Agricultural University, Changsha, P.R. China
| | - Lei He
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, P.R. China
- College of Plant Protection, Hunan Agricultural University, Changsha, P.R. China
| | - Zi-Qi Li
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, P.R. China
- College of Plant Protection, Hunan Agricultural University, Changsha, P.R. China
| | - Ni Li
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, P.R. China
- College of Plant Protection, Hunan Agricultural University, Changsha, P.R. China
| | - Yi-Yi Ou-Yang
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, P.R. China
- College of Plant Protection, Hunan Agricultural University, Changsha, P.R. China
| | - Guo-Hua Huang
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, Hunan Agricultural University, Changsha, P.R. China
- College of Plant Protection, Hunan Agricultural University, Changsha, P.R. China
| |
Collapse
|
15
|
Navabpour S, Kwapis JL, Jarome TJ. A neuroscientist's guide to transgenic mice and other genetic tools. Neurosci Biobehav Rev 2020; 108:732-748. [PMID: 31843544 PMCID: PMC8049509 DOI: 10.1016/j.neubiorev.2019.12.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
The past decade has produced an explosion in the number and variety of genetic tools available to neuroscientists, resulting in an unprecedented ability to precisely manipulate the genome and epigenome in behaving animals. However, no single resource exists that describes all of the tools available to neuroscientists. Here, we review the genetic, transgenic, and viral techniques that are currently available to probe the complex relationship between genes and cognition. Topics covered include types of traditional transgenic mouse models (knockout, knock-in, reporter lines), inducible systems (Cre-loxP, Tet-On, Tet-Off) and cell- and circuit-specific systems (TetTag, TRAP, DIO-DREADD). Additionally, we provide details on virus-mediated and siRNA/shRNA approaches, as well as a comprehensive discussion of the myriad manipulations that can be made using the CRISPR-Cas9 system, including single base pair editing and spatially- and temporally-regulated gene-specific transcriptional control. Collectively, this review will serve as a guide to assist neuroscientists in identifying and choosing the appropriate genetic tools available to study the complex relationship between the brain and behavior.
Collapse
Affiliation(s)
- Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, College Park, PA, USA; Center for the Molecular Investigation of Neurological Disorders (CMIND), Pennsylvania State University, College Park, PA, USA.
| | - Timothy J Jarome
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA; Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
16
|
Guo LB, Yu C, Ling QL, Fu Y, Wang YJ, Liu JG. Proteomic analysis of male rat nucleus accumbens, dorsal hippocampus and amygdala on conditioned place aversion induced by morphine withdrawal. Behav Brain Res 2019; 372:112008. [PMID: 31173798 DOI: 10.1016/j.bbr.2019.112008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/20/2019] [Accepted: 06/03/2019] [Indexed: 01/17/2023]
Abstract
Addiction is characterized by compulsive drug seeking and taking behavior, which is thought to result from persistent neuroadaptations, encoded by changes of gene expression. We previously demonstrated that the changes in synaptic plasticity were required for the formation of aversive memories associated with morphine withdrawal. However, the proteins involved in synaptic plasticity and aversive memory formation have not been well explored. In the present study, we employed a two-dimensional gel electrophoresis (2-DE)-based proteomic technique to detect the changes of protein expression in the nucleus accumbens, amygdala and dorsal hippocampus of the rats that had developed conditioned morphine withdrawal. We found that twenty-three proteins were significantly altered in the amygdala and dorsal hippocampus after conditioned morphine withdrawal. These proteins can be classified into multiple categories, such as energy metabolism, signal transduction, synaptic transmission, cytoskeletal proteins, chaperones, and protein metabolism according to their biological functions. Eight proteins related to synaptic plasticity were further confirmed by western blot analysis. It is very likely that these identified proteins may contribute to conditioned morphine withdrawal-induced neural plasticity and aversive memory formation. Thus, our work will help understand the potential mechanism associated with generation of drug withdrawal memories.
Collapse
Affiliation(s)
- Liu-Bin Guo
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Chuan Yu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica and Collaborative Innovation Center for Brain Science, Chinese Academy of Sciences, Shanghai, 201203, People's Republic of China
| | - Qing-Lan Ling
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica and Collaborative Innovation Center for Brain Science, Chinese Academy of Sciences, Shanghai, 201203, People's Republic of China
| | - Yu Fu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica and Collaborative Innovation Center for Brain Science, Chinese Academy of Sciences, Shanghai, 201203, People's Republic of China
| | - Yu-Jun Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica and Collaborative Innovation Center for Brain Science, Chinese Academy of Sciences, Shanghai, 201203, People's Republic of China
| | - Jing-Gen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica and Collaborative Innovation Center for Brain Science, Chinese Academy of Sciences, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
17
|
Pagani MR, Merlo E. Kinase and Phosphatase Engagement Is Dissociated Between Memory Formation and Extinction. Front Mol Neurosci 2019; 12:38. [PMID: 30842725 PMCID: PMC6391346 DOI: 10.3389/fnmol.2019.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/31/2019] [Indexed: 01/18/2023] Open
Abstract
Associative long-term memories (LTMs) support long-lasting behavioral changes resulting from sensory experiences. Retrieval of a stable LTM by means of a large number of conditioned stimulus (CS) alone presentations produces inhibition of the original memory through extinction. Currently, there are two opposing hypotheses to account for the neural mechanisms supporting extinction. The unlearning hypothesis posits that extinction affects the original memory trace by reverting the synaptic changes supporting LTM. On the contrary, the new learning hypothesis proposes that extinction is simply the formation of a new associative memory that inhibits the expression of the original one. We propose that detailed analysis of extinction-associated molecular mechanisms could help distinguish between these hypotheses. Here we will review experimental evidence regarding the role of protein kinases and phosphatases (K&P) on LTM formation and extinction. Even though K&P regulate both memory processes, their participation appears to be dissociated. LTM formation recruits kinases, but is constrained by phosphatases. Memory extinction presents a more diverse molecular landscape, requiring phosphatases and some kinases, but also being constrained by kinase activity. Based on the available evidence, we propose a new theoretical model for memory extinction: a neuronal segregation of K&P supports a combination of time-dependent reversible inhibition of the original memory [CS-unconditioned stimulus (US)], with establishment of a new associative memory trace (CS-noUS).
Collapse
Affiliation(s)
- Mario Rafael Pagani
- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)-Houssay, Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Emiliano Merlo
- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)-Houssay, Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Rangasamy SB, Jana M, Roy A, Corbett GT, Kundu M, Chandra S, Mondal S, Dasarathi S, Mufson EJ, Mishra RK, Luan CH, Bennett DA, Pahan K. Selective disruption of TLR2-MyD88 interaction inhibits inflammation and attenuates Alzheimer's pathology. J Clin Invest 2018; 128:4297-4312. [PMID: 29990310 PMCID: PMC6159992 DOI: 10.1172/jci96209] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
Induction of TLR2 activation depends on its association with the adapter protein MyD88. We have found that TLR2 and MyD88 levels are elevated in the hippocampus and cortex of patients with Alzheimer's disease (AD) and in a 5XFAD mouse model of AD. Since there is no specific inhibitor of TLR2, to target induced TLR2 from a therapeutic angle, we engineered a peptide corresponding to the TLR2-interacting domain of MyD88 (TIDM) that binds to the BB loop of only TLR2, and not other TLRs. Interestingly, WT TIDM peptide inhibited microglial activation induced by fibrillar Aβ1-42 and lipoteichoic acid, but not 1-methyl-4-phenylpyridinium, dsRNA, bacterial lipopolysaccharide, flagellin, or CpG DNA. After intranasal administration, WT TIDM peptide reached the hippocampus, reduced hippocampal glial activation, lowered Aβ burden, attenuated neuronal apoptosis, and improved memory and learning in 5XFAD mice. However, WT TIDM peptide was not effective in 5XFAD mice lacking TLR2. In addition to its effects in 5XFAD mice, WT TIDM peptide also suppressed the disease process in mice with experimental allergic encephalomyelitis and collagen-induced arthritis. Therefore, selective targeting of the activated status of 1 component of the innate immune system by WT TIDM peptide may be beneficial in AD as well as other disorders in which TLR2/MyD88 signaling plays a role in disease pathogenesis.
Collapse
Affiliation(s)
- Suresh B. Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Grant T. Corbett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sujyoti Chandra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sridevi Dasarathi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Rama K. Mishra
- Medicinal and Synthetic Chemistry Core, Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, USA
| | - Chi-Hao Luan
- High Throughput Analysis Laboratory and Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
19
|
Hopp SC, Bihlmeyer NA, Corradi JP, Vanderburg C, Cacace AM, Das S, Clark TW, Betensky RA, Hyman BT, Hudry E. Neuronal calcineurin transcriptional targets parallel changes observed in Alzheimer disease brain. J Neurochem 2018; 147:24-39. [PMID: 29806693 DOI: 10.1111/jnc.14469] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/11/2018] [Accepted: 05/09/2018] [Indexed: 01/23/2023]
Abstract
Synaptic dysfunction and loss are core pathological features in Alzheimer disease (AD). In the vicinity of amyloid-β plaques in animal models, synaptic toxicity occurs and is associated with chronic activation of the phosphatase calcineurin (CN). Indeed, pharmacological inhibition of CN blocks amyloid-β synaptotoxicity. We therefore hypothesized that CN-mediated transcriptional changes may contribute to AD neuropathology and tested this by examining the impact of CN over-expression on neuronal gene expression in vivo. We found dramatic transcriptional down-regulation, especially of synaptic mRNAs, in neurons chronically exposed to CN activation. Importantly, the transcriptional profile parallels the changes in human AD tissue. Bioinformatics analyses suggest that both nuclear factor of activated T cells and numerous microRNAs may all be impacted by CN, and parallel findings are observed in AD. These data and analyses support the hypothesis that at least part of the synaptic failure characterizing AD may result from aberrant CN activation leading to down-regulation of synaptic genes, potentially via activation of specific transcription factors and expression of repressive microRNAs. OPEN PRACTICES Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ Read the Editorial Highlight for this article on page 8.
Collapse
Affiliation(s)
- Sarah C Hopp
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Nathan A Bihlmeyer
- MIND Informatics, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| | - John P Corradi
- Exploratory Biology and Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, USA
| | - Charles Vanderburg
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Angela M Cacace
- Exploratory Biology and Genomics, Bristol-Myers Squibb, Wallingford, Connecticut, USA
| | - Sudeshna Das
- MIND Informatics, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Timothy W Clark
- MIND Informatics, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| | - Rebecca A Betensky
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Bradley T Hyman
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Eloise Hudry
- Alzheimer's disease Research Laboratory, Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
20
|
Shahidi S, Komaki A, Sadeghian R, Soleimani Asl S. Effect of a 5-HT 1D receptor agonist on the reinstatement phase of the conditioned place preference test and hippocampal long-term potentiation in methamphetamine-treated rats. Brain Res 2018; 1698:151-160. [PMID: 30076792 DOI: 10.1016/j.brainres.2018.07.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH)-seeking relapse is associated with memory and synaptic plasticity changes. Serotonin is a key neuromodulator in this process. While there is a known distribution of 5-HT1D receptors in reward and memory areas, such as the hippocampus, its physiological function is currently unknown. Here, we evaluated effect of a 5-HT1D receptor agonist, PNU142633, on the reinstatement of METH-seeking behavior and long-term potentiation. Rats were implanted with a cannula into lateral ventricle, then treated with saline or METH (5 mg/kg) during the acquisition phase of the conditioned place preference (CPP) test. On day 13 of the extinction phase, METH groups were divided into four groups: METH (0: saline, 1, or 2.5 (priming METH) mg/kg; i.p.) + vehicle (5 µl/rat) or a priming dose of METH (2.5 mg/kg; i.p.) + PNU (2 µg/5 µl; i.c.v.) and their preference scores were calculated on reinstatement day (day 14). Immediately following this, electrophysiology was performed to assay the field excitatory postsynaptic potential (fEPSP) slope and population spike (PS) amplitude between groups. The results showed that CPP induction by METH gradually declined to extinction on days 12 and 13. A priming METH treatment significantly increased preference for the METH-paired chamber when compared with other groups, but pre-treatment with PNU significantly attenuated this effect. PS amplitude and fEPSP slopes in vehicle + priming METH rats were greater when compared with other groups. Furthermore, PNU attenuated the priming METH-induced increase in PS amplitude. These findings suggest that PNU can decrease synaptic transmission and prevent METH reinstatement in rats.
Collapse
Affiliation(s)
- Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Reihaneh Sadeghian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sara Soleimani Asl
- Anatomy Departments, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
21
|
Dark C, Homman-Ludiye J, Bryson-Richardson RJ. The role of ADHD associated genes in neurodevelopment. Dev Biol 2018; 438:69-83. [DOI: 10.1016/j.ydbio.2018.03.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/04/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
|
22
|
Circadian Regulation of Hippocampal-Dependent Memory: Circuits, Synapses, and Molecular Mechanisms. Neural Plast 2018; 2018:7292540. [PMID: 29593785 PMCID: PMC5822921 DOI: 10.1155/2018/7292540] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
Circadian modulation of learning and memory efficiency is an evolutionarily conserved phenomenon, occurring in organisms ranging from invertebrates to higher mammalian species, including humans. While the suprachiasmatic nucleus (SCN) of the hypothalamus functions as the master mammalian pacemaker, recent evidence suggests that forebrain regions, including the hippocampus, exhibit oscillatory capacity. This finding, as well as work on the cellular signaling events that underlie learning and memory, has opened promising new avenues of investigation into the precise cellular, molecular, and circuit-based mechanisms by which clock timing impacts plasticity and cognition. In this review, we examine the complex molecular relationship between clock timing and memory, with a focus on hippocampal-dependent tasks. We evaluate how the dysregulation of circadian timing, both at the level of the SCN and at the level of ancillary forebrain clocks, affects learning and memory. Further, we discuss experimentally validated intracellular signaling pathways (e.g., ERK/MAPK and GSK3β) and potential cellular signaling mechanisms by which the clock affects learning and memory formation. Finally, we examine how long-term potentiation (LTP), a synaptic process critical to the establishment of several forms of memory, is regulated by clock-gated processes.
Collapse
|
23
|
Medina JH. Neural, Cellular and Molecular Mechanisms of Active Forgetting. Front Syst Neurosci 2018; 12:3. [PMID: 29467630 PMCID: PMC5808127 DOI: 10.3389/fnsys.2018.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/18/2018] [Indexed: 11/16/2022] Open
Abstract
The neurobiology of memory formation attracts much attention in the last five decades. Conversely, the rules that govern and the mechanisms underlying forgetting are less understood. In addition to retroactive interference, retrieval-induced forgetting and passive decay of time, it has been recently demonstrated that the nervous system has a diversity of active and inherent processes involved in forgetting. In Drosophila, some operate mainly at an early stage of memory formation and involves dopamine (DA) neurons, specific postsynaptic DA receptor subtypes, Rac1 activation and induces rapid active forgetting. In mammals, others regulate forgetting and persistence of seemingly consolidated memories and implicate the activity of DA receptor subtypes and AMPA receptors in the hippocampus (HP) and related structures to activate parallel signaling pathways controlling active time-dependent forgetting. Most of them may involve plastic changes in synaptic and extrasynaptic receptors including specific removal of GluA2 AMPA receptors. Forgetting at longer timescales might also include changes in adult neurogenesis in the dentate gyrus (DG) of the HP. Therefore, based on relevance or value considerations neuronal circuits may regulate in a time-dependent manner what is formed, stored, and maintained and what is forgotten.
Collapse
Affiliation(s)
- Jorge H Medina
- Laboratorio de Memoria, IBCN Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (UBA-CONICET), Buenos Aires, Argentina.,Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
Local Inhibition of PERK Enhances Memory and Reverses Age-Related Deterioration of Cognitive and Neuronal Properties. J Neurosci 2017; 38:648-658. [PMID: 29196323 DOI: 10.1523/jneurosci.0628-17.2017] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 10/25/2017] [Accepted: 11/10/2017] [Indexed: 01/08/2023] Open
Abstract
Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of four known kinases that respond to cellular stress by deactivating the eukaryotic initiation factor 2 α (eIF2α) or other signal transduction cascades. Recently, both eIF2α and its kinases were found to play a role in normal and pathological brain function. Here, we show that reduction of either the amount or the activity of PERK, specifically in the CA1 region of the hippocampus in young adult male mice, enhances neuronal excitability and improves cognitive function. In addition, this manipulation rescues the age-dependent cellular phenotype of reduced excitability and memory decline. Specifically, the reduction of PERK expression in the CA1 region of the hippocampus of middle-aged male mice using a viral vector rejuvenates hippocampal function and improves hippocampal-dependent learning. These results delineate a mechanism for behavior and neuronal aging and position PERK as a promising therapeutic target for age-dependent brain malfunction.SIGNIFICANCE STATEMENT We found that local reduced protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) expression or activity in the hippocampus enhances neuronal excitability and cognitive function in young normal mice, that old CA1 pyramidal cells have reduced excitability and increased PERK expression that can be rescued by reducing PERK expression in the hippocampus, and that reducing PERK expression in the hippocampus of middle-aged mice enhances hippocampal-dependent learning and memory and restores it to normal performance levels of young mice. These findings uncover an entirely new biological link among PERK, neuronal intrinsic properties, aging, and cognitive function. Moreover, our findings propose a new way to fight mild cognitive impairment and aging-related cognitive deterioration.
Collapse
|
25
|
Opiate exposure state controls dopamine D3 receptor and cdk5/calcineurin signaling in the basolateral amygdala during reward and withdrawal aversion memory formation. Prog Neuropsychopharmacol Biol Psychiatry 2017. [PMID: 28627448 DOI: 10.1016/j.pnpbp.2017.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The dopamine (DA) D3 receptor (D3R) is highly expressed in the basolateral nucleus of the amygdala (BLA), a neural region critical for processing opiate-related reward and withdrawal aversion-related memories. Functionally, D3R transmission is linked to downstream Cdk5 and calcineurin signaling, both of which regulate D3R activity states and play critical roles in memory-related synaptic plasticity. Previous evidence links D3R transmission to opiate-related memory processing, however little is known regarding how chronic opiate exposure may alter D3R-dependent memory mechanisms. Using conditioned place preference (CPP) and withdrawal aversion (conditioned place aversion; CPA) procedures in rats, combined with molecular analyses of BLA protein expression, we examined the effects of chronic opiate exposure on the functional role of intra-BLA D3R transmission during the acquisition of opiate reward or withdrawal aversion memories. Remarkably, we report that the state of opiate exposure during behavioural conditioning (opiate-naïve/non-dependent vs. chronically exposed and in withdrawal) controlled the functional role of intra-BLA D3R transmission during the acquisition of both opiate reward memories and withdrawal-aversion associative memories. Thus, whereas intra-BLA D3R blockade had no effect on opiate reward memory formation in the non-dependent state, blockade of intra-BLA D3R transmission prevented the formation of opiate reward and withdrawal aversion memory in the chronically exposed state. This switch in the functional role of D3R transmission corresponded to significant increases in Cdk5 phosphorylation and total expression levels of calcineurin, and a corresponding decrease in intra-BLA D3R expression. Inhibition of either intra-BLA Cdk5 or calcineurin reversed these effects, switching intra-BLA associative memory formation back to a D3R-independent mechanism.
Collapse
|
26
|
Schroeder M, Jakovcevski M, Polacheck T, Lebow M, Drori Y, Engel M, Ben-Dor S, Chen A. A Methyl-Balanced Diet Prevents CRF-Induced Prenatal Stress-Triggered Predisposition to Binge Eating-like Phenotype. Cell Metab 2017; 25:1269-1281.e6. [PMID: 28576418 DOI: 10.1016/j.cmet.2017.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/30/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
Binge eating (BE) is a common aberrant form of eating behavior, characterized by overconsumption of food in a brief period of time. Recurrent episodes of BE constitute the BE disorder, which mostly affects females and is associated with early-life adversities. Here, we show that corticotropin releasing factor (CRF)-induced prenatal stress (PNS) in late gestation predisposes female offspring to BE-like behavior that coincides with hypomethylation of hypothalamic miR-1a and downstream dysregulation of the melanocortin system through Pax7/Pax3. Moreover, exposing the offspring to a methyl-balanced diet during adolescence prevents the dysregulation and predisposition from being triggered. We demonstrate that gestational programming, per se, will not lead to BE-like behavior, but pre-existing alterations due to prenatal programming are revealed only when challenged during adolescence. We provide experimental evidence for long-term epigenetic abnormalities stemming from PNS in predisposing female offspring to BE disorder as well as a potential non-invasive prevention strategy.
Collapse
Affiliation(s)
- Mariana Schroeder
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Tamar Polacheck
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Maya Lebow
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Yonat Drori
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Mareen Engel
- Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max-Planck Institute of Psychiatry, Munich 80804, Germany.
| |
Collapse
|
27
|
Shah SZA, Hussain T, Zhao D, Yang L. A central role for calcineurin in protein misfolding neurodegenerative diseases. Cell Mol Life Sci 2017; 74:1061-1074. [PMID: 27682820 PMCID: PMC11107525 DOI: 10.1007/s00018-016-2379-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/06/2016] [Accepted: 09/23/2016] [Indexed: 12/25/2022]
Abstract
Accumulation of misfolded/unfolded aggregated proteins in the brain is a hallmark of many neurodegenerative diseases affecting humans and animals. Dysregulation of calcium (Ca2+) and disruption of fast axonal transport (FAT) are early pathological events that lead to loss of synaptic integrity and axonal degeneration in early stages of neurodegenerative diseases. Dysregulated Ca2+ in the brain is triggered by accumulation of misfolded/unfolded aggregated proteins in the endoplasmic reticulum (ER), a major Ca2+ storing organelle, ultimately leading to neuronal dysfunction and apoptosis. Calcineurin (CaN), a Ca2+/calmodulin-dependent serine/threonine phosphatase, has been implicated in T cells activation through the induction of nuclear factor of activated T cells (NFAT). In addition to the involvement of several other signaling cascades, CaN has been shown to play a role in early synaptic dysfunction and neuronal death. Therefore, inhibiting hyperactivated CaN in early stages of disease might be a promising therapeutic strategy for treating patients with protein misfolding diseases. In this review, we briefly summarize the structure of CaN, inhibition mechanisms by which immunosuppressants inhibit CaN, role of CaN in maintaining neuronal and synaptic integrity and homeostasis and the role played by CaN in protein unfolding/misfolding neurodegenerative diseases.
Collapse
Affiliation(s)
- Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China
| | - Tariq Hussain
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
28
|
Pearce K, Cai D, Roberts AC, Glanzman DL. Role of protein synthesis and DNA methylation in the consolidation and maintenance of long-term memory in Aplysia. eLife 2017; 6. [PMID: 28067617 PMCID: PMC5310836 DOI: 10.7554/elife.18299] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/07/2017] [Indexed: 12/13/2022] Open
Abstract
Previously, we reported that long-term memory (LTM) in Aplysia can be reinstated by truncated (partial) training following its disruption by reconsolidation blockade and inhibition of PKM (Chen et al., 2014). Here, we report that LTM can be induced by partial training after disruption of original consolidation by protein synthesis inhibition (PSI) begun shortly after training. But when PSI occurs during training, partial training cannot subsequently establish LTM. Furthermore, we find that inhibition of DNA methyltransferase (DNMT), whether during training or shortly afterwards, blocks consolidation of LTM and prevents its subsequent induction by truncated training; moreover, later inhibition of DNMT eliminates consolidated LTM. Thus, the consolidation of LTM depends on two functionally distinct phases of protein synthesis: an early phase that appears to prime LTM; and a later phase whose successful completion is necessary for the normal expression of LTM. Both the consolidation and maintenance of LTM depend on DNA methylation.
Collapse
Affiliation(s)
- Kaycey Pearce
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, United States
| | - Diancai Cai
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, United States
| | - Adam C Roberts
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, United States
| | - David L Glanzman
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, United States.,Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Integrative Center for Learning and Memory, Brain Research Institute, UCLA, Los Angeles, United States
| |
Collapse
|
29
|
Lee AY, Choi JM, Lee J, Lee MH, Lee S, Cho EJ. Effects of Vegetable Oils with Different Fatty Acid Compositions on Cognition and Memory Ability in Aβ 25-35-Induced Alzheimer's Disease Mouse Model. J Med Food 2016; 19:912-921. [PMID: 27696934 DOI: 10.1089/jmf.2016.3737] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this study, we aimed to investigate the protective effect of three kinds of vegetable oils with different fatty acid compositions against cognitive impairment in an Alzheimer's disease (AD) mouse model. After intracerebroventricular injection of amyloid beta25-35 (Aβ25-35) into the brain of institute of cancer research mice, olive oil (rich in oleic acid, C18:1), corn oil (rich in linoleic acid, C18:2), and perilla oil (rich in α-linolenic acid [ALA], C18:3) were administered at the oral dose of 500 mg/kg/day for 14 days. The results revealed that Aβ25-35 induced learning and memory dysfunction according to the T-maze, novel object recognition, and Morris water maze tests. Among the three vegetable oils, however, the perilla oil group of mice showed marked attenuation of cognitive impairment, that is, a greater number of explorations on a new route/object than on an old route/object in the T-maze and novel object recognition tests. In the Morris water maze test, perilla oil decreased the time to reach the platform and increased the number of crossings over the target quadrant in which the platform was located previously. Furthermore, the beneficial effect of perilla oil supplementation on oxidative stress was reflected in the inhibition of malondialdehyde and nitric oxide (NO) production in Aβ25-35-injected mice. We also found that perilla oil downregulated protein expression levels of inducible NO synthase and cyclooxygenase-2 and upregulated brain-derived neurotrophic factor. These findings showed that ALA-rich perilla oil has a potential for prevention or treatment of neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Ah Young Lee
- 1 Department of Food Science and Nutrition, Research Institute of Ecology for the Elderly, Pusan National University , Busan, Korea
| | - Ji Myung Choi
- 2 Department of Southern Area Crop Science, National Institute of Crop Science , Rural Development Administration, Miryang, Korea
| | - Jaemin Lee
- 3 Department of Integrative Plant Science, Chung-Ang University , Anseong, Korea
| | - Myoung Hee Lee
- 2 Department of Southern Area Crop Science, National Institute of Crop Science , Rural Development Administration, Miryang, Korea
| | - Sanghyun Lee
- 3 Department of Integrative Plant Science, Chung-Ang University , Anseong, Korea
| | - Eun Ju Cho
- 1 Department of Food Science and Nutrition, Research Institute of Ecology for the Elderly, Pusan National University , Busan, Korea
| |
Collapse
|
30
|
Datta S, Samanta D, Sinha P, Chakrabarti N. Gender features and estrous cycle variations of nocturnal behavior of mice after a single exposure to light at night. Physiol Behav 2016; 164:113-22. [DOI: 10.1016/j.physbeh.2016.05.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 01/10/2023]
|
31
|
Popugaeva E, Pchitskaya E, Bezprozvanny I. Dysregulation of neuronal calcium homeostasis in Alzheimer's disease - A therapeutic opportunity? Biochem Biophys Res Commun 2016; 483:998-1004. [PMID: 27641664 DOI: 10.1016/j.bbrc.2016.09.053] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is the disease of lost memories. Synaptic loss is a major reason for memory defects in AD. Signaling pathways involved in memory loss in AD are under intense investigation. The role of deranged neuronal calcium (Ca2+) signaling in synaptic loss in AD is described in this review. Familial AD (FAD) mutations in presenilins are linked directly with synaptic Ca2+ signaling abnormalities, most likely by affecting endoplasmic reticulum (ER) Ca2+ leak function of presenilins. Excessive ER Ca2+ release via type 2 ryanodine receptors (RyanR2) is observed in AD spines due to increase in expression and function of RyanR2. Store-operated Ca2+ entry (nSOC) pathway is disrupted in AD spines due to downregulation of STIM2 protein. Because of these Ca2+ signaling abnormalities, a balance in activities of Ca2+-calmodulin-dependent kinase II (CaMKII) and Ca2+-dependent phosphatase calcineurin (CaN) is shifted at the synapse, tilting a balance between long-term potentiation (LTP) and long-term depression (LTD) synaptic mechanisms. As a result, synapses are weakened and eliminated in AD brains by LTD mechanism, causing memory loss. Targeting synaptic calcium signaling pathways offers opportunity for development of AD therapeutic agents.
Collapse
Affiliation(s)
- Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russian Federation.
| | - Ekaterina Pchitskaya
- Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russian Federation.
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russian Federation; Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
32
|
Taglialatela G, Rastellini C, Cicalese L. Reduced Incidence of Dementia in Solid Organ Transplant Patients Treated with Calcineurin Inhibitors. J Alzheimers Dis 2016; 47:329-33. [PMID: 26401556 PMCID: PMC4923720 DOI: 10.3233/jad-150065] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Experimental evidence suggests that the protein phosphatase calcineurin mediates the action of amyloid-β (Aβ) oligomers, the most toxic amyloid species thought to drive initial cognitive decline in Alzheimer's disease (AD). However, there is currently no evidence that inhibition of calcineurin could prevent the onset of AD in humans. Here, we report for the first time that individuals chronically treated with calcineurin inhibitors to prevent solid organ transplant rejection have a significantly lower incidence of AD/dementia as compared to the general population. This result prompts further clinical development of calcineurin inhibition as a viable treatment for AD.
Collapse
Affiliation(s)
- Giulio Taglialatela
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Cristiana Rastellini
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Luca Cicalese
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
33
|
Impaired acquisition of conditioned taste aversion memory induced by isoflurane is accompanied with calcineurin activation and Egr-1 down-regulation in amygdala in rats. Neurosci Lett 2015; 607:114-119. [PMID: 26393333 DOI: 10.1016/j.neulet.2015.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 08/10/2015] [Accepted: 09/16/2015] [Indexed: 11/22/2022]
Abstract
Compared to neutral memory, emotional memory is extremely strong and persistent immediately after acquisition, therefore it may recruit specific mechanisms during acquisition. The calcineurin-dependent mechanisms engaging early growth response 1 (Egr-1) have been proved to determine the strength of emotional memory during establishment. Isoflurane, a widely used inhalation anesthetic, can interfere with the acquisition of emotional memory. We hypothesized that isoflurane impairs the acquisition of conditioned taste aversion (CTA) memory in rats and the Egr-1 expression regulation via calcineurin (CaN) and ERK signaling pathway is involved in isoflurane-induced repression of CTA memory. To examine this, we investigated the influence of isoflurane on CTA memory and the expression and activity of CaN, the phosphorylation level of ERK and the expression of Egr-1 in amygdala in response to CTA training in rats. The results showed that isoflurane exposure (1.5%, 2h) before training impaired the acquisition of CTA memory in rats. Isoflurane exposure increased the CaN activity and decreased the p-ERK and Egr-1 in amygdala in rats. These findings suggest that isoflurane can disrupt the establishment of aversion memory, and CaN activation associating with p-ERK and Egr-1 down-regulation may contribute to the isoflurane induced impairment of aversion memory acquisition.
Collapse
|
34
|
Putting the past behind us: Social stress-induced urinary retention can be overcome. J Pediatr Urol 2015; 11:188-94. [PMID: 26052002 DOI: 10.1016/j.jpurol.2015.04.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/21/2015] [Indexed: 01/28/2023]
Abstract
INTRODUCTION To study the pathophysiology of dysfunctional voiding, we have previously developed a model of stress-induced voiding dysfunction. We have shown that cyclosporine A (CsA), an inhibitor of the Ca(2+)-calmodulin complex, can prevent social stress-induced urinary retention. However, treatment with cyclosporine has not had an effect on the increase in the stress peptide corticotrophin-releasing factor (CRF) in Barrington's nucleus, which is involved in the micturition pathway. OBJECTIVE We now investigate whether cyclosporine administered after stress can reverse the abnormal voiding phenotype, and whether it has effects on the bladder wall itself, or on the stress response within Barrington's nucleus. MATERIALS AND METHODS Six-week old Swiss-Webster mice were exposed to aggressor males for 1 h a day, followed by 23 h of barrier separation. In a long-term trial, 1 month of stress was followed by single-cage housing for 6 months. In a separate CsA reversal trial, mice either received CsA in drinking water or had plain drinking water during 1 month of single-cage housing during recovery. Bladder contractile function was examined on a Guth myograph. Nuclear translocation of myocyte enhancing factor (MEF)-2 and NFAT (nuclear factor of activated T cells) in the bladder was assessed using electrophoretic mobility shift assays (EMSAs). The expression of CRF was determined in Barrington's nucleus using in situ hybridization. RESULTS Voiding dysfunction persisted for up to 6 months after stress exposure while mice recovered in single-cage housing. In the CsA reversal trial, voiding patterns improved when they received CsA in water during single-cage housing following stress, whereas those that underwent single-cage housing alone had persistent abnormal voiding (Fig. A). There was no difference between CRF levels in Barrington's nucleus between reversal groups (p = 0.42) (Fig. B), possibly indicating a direct effect on the bladder rather than a persistent stress effect. There were no differences in the contractility of bladder wall muscle. CsA decreased the nuclear translocation of MEF-2 and NFAT induced by stress (Fig. C,D). CONCLUSION CsA reverses stress-induced urinary retention, but does not change the stress-induced CRF increase in Barrington's nucleus. Furthermore, bladder smooth muscle contractility is unchanged by CsA; however, there are changes in the levels of the downstream transcription factors MEF-2 and NFAT. We suspect that additional CsA responsive neural changes play a pivotal role in the abnormal voiding phenotype following social stress.
Collapse
|
35
|
Rangasamy SB, Corbett GT, Roy A, Modi KK, Bennett DA, Mufson EJ, Ghosh S, Pahan K. Intranasal Delivery of NEMO-Binding Domain Peptide Prevents Memory Loss in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2015; 47:385-402. [PMID: 26401561 PMCID: PMC4582676 DOI: 10.3233/jad-150040] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Despite intense investigations, no effective therapy is available to halt its progression. We found that NF-κB was activated within the hippocampus and cortex of AD subjects and that activated forms of NF-κB negatively correlated with cognitive function monitored by Mini-Mental State Examination and global cognitive z score. Accordingly, NF-κB activation was also observed in the hippocampus of a transgenic (5XFAD) mouse model of AD. It has been shown that peptides corresponding to the NF-κB essential modifier (NEMO)-binding domain (NBD) of IκB kinase α (IKKα) or IκB kinase β (IKKβ) specifically inhibit the induction of NF-κB activation without inhibiting the basal NF-κB activity. Interestingly, after intranasal administration, wild-type NBD peptide entered into the hippocampus, reduced hippocampal activation of NF-κB, suppressed hippocampal microglial activation, lowered the burden of Aβ in the hippocampus, attenuated apoptosis of hippocampal neurons, protected plasticity-related molecules, and improved memory and learning in 5XFAD mice. Mutated NBD peptide had no such protective effect, indicating the specificity of our finding. These results suggest that selective targeting of NF-κB activation by intranasal administration of NBD peptide may be of therapeutic benefit for AD patients.
Collapse
Affiliation(s)
- Suresh B. Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Grant T. Corbett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Khushbu K. Modi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A. Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | - Sankar Ghosh
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA,Correspondence to: Kalipada Pahan, PhD, Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 320, Chicago, IL 60612, USA. Tel.: +1 312 563 3592; Fax: +1 312 563 3571;
| |
Collapse
|
36
|
Modi KK, Roy A, Brahmachari S, Rangasamy SB, Pahan K. Cinnamon and Its Metabolite Sodium Benzoate Attenuate the Activation of p21rac and Protect Memory and Learning in an Animal Model of Alzheimer's Disease. PLoS One 2015; 10:e0130398. [PMID: 26102198 PMCID: PMC4478015 DOI: 10.1371/journal.pone.0130398] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/20/2015] [Indexed: 01/08/2023] Open
Abstract
This study underlines the importance of cinnamon, a commonly used natural spice and flavoring material, and its metabolite sodium benzoate (NaB) in attenuating oxidative stress and protecting memory and learning in an animal model of Alzheimer’s disease (AD). NaB, but not sodium formate, was found to inhibit LPS-induced production of reactive oxygen species (ROS) in mouse microglial cells. Similarly, NaB also inhibited fibrillar amyloid beta (Aβ)- and 1-methyl-4-phenylpyridinium(+)-induced microglial production of ROS. Although NaB reduced the level of cholesterol in vivo in mice, reversal of the inhibitory effect of NaB on ROS production by mevalonate, and geranylgeranyl pyrophosphate, but not cholesterol, suggests that depletion of intermediates, but not end products, of the mevalonate pathway is involved in the antioxidant effect of NaB. Furthermore, we demonstrate that an inhibitor of p21rac geranylgeranyl protein transferase suppressed the production of ROS and that NaB suppressed the activation of p21rac in microglia. As expected, marked activation of p21rac was observed in the hippocampus of subjects with AD and 5XFAD transgenic (Tg) mouse model of AD. However, oral feeding of cinnamon (Cinnamonum verum) powder and NaB suppressed the activation of p21rac and attenuated oxidative stress in the hippocampus of Tg mice as evident by decreased dihydroethidium (DHE) and nitrotyrosine staining, reduced homocysteine level and increased level of reduced glutathione. This was accompanied by suppression of neuronal apoptosis, inhibition of glial activation, and reduction of Aβ burden in the hippocampus and protection of memory and learning in transgenic mice. Therefore, cinnamon powder may be a promising natural supplement in halting or delaying the progression of AD.
Collapse
Affiliation(s)
- Khushbu K. Modi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, United States of America
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, United States of America
| | - Saurabh Brahmachari
- Department of Neurological Sciences, Rush University Medical Center, Chicago, United States of America
| | - Suresh B. Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, United States of America
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, United States of America
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue, Chicago, United States of America
- * E-mail:
| |
Collapse
|
37
|
Ni C, Li Z, Qian M, Zhou Y, Wang J, Guo X. Isoflurane induced cognitive impairment in aged rats through hippocampal calcineurin/NFAT signaling. Biochem Biophys Res Commun 2015; 460:889-95. [DOI: 10.1016/j.bbrc.2015.03.083] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/17/2015] [Indexed: 10/23/2022]
|
38
|
Subramaniyan S, Hajali V, Scherf T, Sase SJ, Sialana FJ, Gröger M, Bennett KL, Pollak A, Li L, Korz V, Lubec G. Hippocampal receptor complexes paralleling LTP reinforcement in the spatial memory holeboard test in the rat. Behav Brain Res 2015; 283:162-74. [PMID: 25639541 DOI: 10.1016/j.bbr.2015.01.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 01/18/2015] [Accepted: 01/21/2015] [Indexed: 12/28/2022]
Abstract
The current study was designed to examine learning-induced transformation of early-LTP into late-LTP. Recording electrodes were implanted into the dentate gyrus of the hippocampus in male rats and early-LTP was induced by weak tetanic stimulation of the medial perforant path. Dorsal right hippocampi were removed, membrane proteins were extracted, separated by blue-native gel electrophoresis with subsequent immunoblotting using brain receptor antibodies. Spatial training resulted into reinforcement of LTP and the reinforced LTP was persistent for 6h. Receptor complex levels containing GluN1 and GluN2A of NMDARs, GluA1 and GluA2 of AMPARs, nAchα7R and the D(1A) dopamine receptor were significantly-elevated in rat hippocampi of animals underwent spatial learning, whilst levels of GluA3 and 5-HT1A receptor containing complexes were significantly reduced. Evidence for complex formation between GluN1 and D(1A) dopamine receptor was provided by antibody shift assay, co-immunoprecipitation and mass spectrometric analysis. Thus our results propose that behavioural stimuli like spatial learning reinforce early LTP into late LTP and this reinforced LTP is accompanied by changes in certain receptor levels in the membrane fraction of the rat hippocampus.
Collapse
Affiliation(s)
| | - Vahid Hajali
- Institute of Biology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Scherf
- Institute of Biology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sunetra Jitkar Sase
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Fernando J Sialana
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Marion Gröger
- Core facilities, Core Facility Imaging, Medizinische Universität Wien, Anna Spiegel Forschungsgebäude, 1090 Vienna, Austria
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT25.3 and 1090 Vienna, Austria
| | - Arnold Pollak
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Lin Li
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Volker Korz
- Institute of Biology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany.
| | - Gert Lubec
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien.
| |
Collapse
|
39
|
Fakhfouri G, Mousavizadeh K, Mehr SE, Dehpour AR, Zirak MR, Ghia JE, Rahimian R. From Chemotherapy-Induced Emesis to Neuroprotection: Therapeutic Opportunities for 5-HT3 Receptor Antagonists. Mol Neurobiol 2014; 52:1670-1679. [DOI: 10.1007/s12035-014-8957-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/21/2014] [Indexed: 01/11/2023]
|
40
|
In search of a recognition memory engram. Neurosci Biobehav Rev 2014; 50:12-28. [PMID: 25280908 PMCID: PMC4382520 DOI: 10.1016/j.neubiorev.2014.09.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 09/18/2014] [Accepted: 09/22/2014] [Indexed: 01/06/2023]
Abstract
The role of the perirhinal cortex in familiarity discrimination is reviewed. Behavioural, pharmacological and electrophysiological evidence is considered. The cortex is found to be essential for memory acquisition, retrieval and storage. The evidence indicates that perirhinal synaptic weakening is critically involved.
A large body of data from human and animal studies using psychological, recording, imaging, and lesion techniques indicates that recognition memory involves at least two separable processes: familiarity discrimination and recollection. Familiarity discrimination for individual visual stimuli seems to be effected by a system centred on the perirhinal cortex of the temporal lobe. The fundamental change that encodes prior occurrence within the perirhinal cortex is a reduction in the responses of neurones when a stimulus is repeated. Neuronal network modelling indicates that a system based on such a change in responsiveness is potentially highly efficient in information theoretic terms. A review is given of findings indicating that perirhinal cortex acts as a storage site for recognition memory of objects and that such storage depends upon processes producing synaptic weakening.
Collapse
|
41
|
A physically-modified saline suppresses neuronal apoptosis, attenuates tau phosphorylation and protects memory in an animal model of Alzheimer's disease. PLoS One 2014; 9:e103606. [PMID: 25089827 PMCID: PMC4121132 DOI: 10.1371/journal.pone.0103606] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/02/2014] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia in the aging population, is characterized by the presence of neuritic plaques, neurofibrillary tangles and extensive neuronal apoptosis. Neuritic plaques are mainly composed of aggregates of amyloid-β (Aβ) protein while neurofibrillary tangles are composed of the hyperphosphorylated tau protein. Despite intense investigations, no effective therapy is currently available to halt the progression of this disease. Here, we have undertaken a novel approach to attenuate apoptosis and tau phosphorylation in cultured neuronal cells and in a transgenic animal model of AD. RNS60 is a 0.9% saline solution containing oxygenated nanobubbles that is generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. In our experiments, fibrillar Aβ1-42, but not the reverse peptide Aβ42-1, induced apoptosis and cell death in human SHSY5Y neuronal cells. RNS60, but not NS (normal saline), RNS10.3 (TCP-modified saline without excess oxygen) or PNS60 (saline containing excess oxygen without TCP modification), attenuated Aβ(1–42)-induced cell death. RNS60 inhibited neuronal cell death via activation of the type 1A phosphatidylinositol-3 (PI-3) kinase – Akt – BAD pathway. Furthermore, RNS60 also decreased Aβ(1–42)-induced tau phosphorylation via (PI-3 kinase – Akt)-mediated inhibition of GSK-3β. Similarly, RNS60 treatment suppressed neuronal apoptosis, attenuated Tau phosphorylation, inhibited glial activation, and reduced the burden of Aβ in the hippocampus and protected memory and learning in 5XFAD transgenic mouse model of AD. Therefore, RNS60 may be a promising pharmaceutical candidate in halting or delaying the progression of AD.
Collapse
|
42
|
Long CJ, Butler S, Fesi J, Frank C, Canning DA, Zderic SA. Genetic or pharmacologic disruption of the calcineurin-nuclear factor of activated T-cells axis prevents social stress-induced voiding dysfunction in a murine model. J Pediatr Urol 2014; 10:598-604. [PMID: 24909609 DOI: 10.1016/j.jpurol.2014.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 04/09/2014] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Social stress can suppress the voiding reflex, with resultant diminished voiding frequency and increased volumes. The calcineurin-NFAT (nuclear factor of activated T cells) pathway is important in memory development. It was hypothesized that interruption of the calcineurin-NFAT pathway might prevent social stress-induced voiding dysfunction. METHODS Mice were subjected to social stress in an established resident-intruder model for 1 h, followed by 23 h of barrier separation. NFATc3, NFATc4 knockout (KO) and wild-type (WT) mice were studied. At two weeks, voiding patterns were collected; this was followed by sacrifice. Corticotropin-releasing factor (CRF) mRNA expression in Barrington's nucleus (BN) was determined by in-situ hybridization. RESULTS Social stress decreased voiding frequency and increased voided volumes in WT strains. At baseline, NFATc3 KO mice showed decreased voids and increased volumes, while the NFATc4 KO mice resisted social stress. However, CRF mRNA increased in WT mice following social stress and was increased at baseline in NFATc3 KO mice. It was found that CRF mRNA did not increase following social stress in NFATc4 KO mice. The administration of CsA to WT mice normalized voiding patterns following social stress, albeit with no effect on CRF mRNA in BN. CONCLUSION Disrupting the calcineurin-NFAT axis by either genetic or pharmacologic approaches confers resistance to the development of social stress-induced voiding and dysfunction.
Collapse
Affiliation(s)
- C J Long
- John W. Duckett Center for Pediatric Urology at The Children's Hospital of Philadelphia, USA; The Perelman School of Medicine at The University of Pennsylvania, USA.
| | - S Butler
- John W. Duckett Center for Pediatric Urology at The Children's Hospital of Philadelphia, USA; The Perelman School of Medicine at The University of Pennsylvania, USA.
| | - J Fesi
- John W. Duckett Center for Pediatric Urology at The Children's Hospital of Philadelphia, USA; The Perelman School of Medicine at The University of Pennsylvania, USA.
| | - C Frank
- John W. Duckett Center for Pediatric Urology at The Children's Hospital of Philadelphia, USA; The Perelman School of Medicine at The University of Pennsylvania, USA.
| | - D A Canning
- John W. Duckett Center for Pediatric Urology at The Children's Hospital of Philadelphia, USA; The Perelman School of Medicine at The University of Pennsylvania, USA.
| | - S A Zderic
- John W. Duckett Center for Pediatric Urology at The Children's Hospital of Philadelphia, USA.
| |
Collapse
|
43
|
Mirante O, Brandalise F, Bohacek J, Mansuy IM. Distinct molecular components for thalamic- and cortical-dependent plasticity in the lateral amygdala. Front Mol Neurosci 2014; 7:62. [PMID: 25071439 PMCID: PMC4080466 DOI: 10.3389/fnmol.2014.00062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 01/05/2023] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR)-dependent long-term depression (LTD) in the lateral nucleus of the amygdala (LA) is a form of synaptic plasticity thought to be a cellular substrate for the extinction of fear memory. The LA receives converging inputs from the sensory thalamus and neocortex that are weakened following fear extinction. Combining field and patch-clamp electrophysiological recordings in mice, we show that paired-pulse low-frequency stimulation can induce a robust LTD at thalamic and cortical inputs to LA, and we identify different underlying molecular components at these pathways. We show that while LTD depends on NMDARs and activation of the protein phosphatases PP2B and PP1 at both pathways, it requires NR2B-containing NMDARs at the thalamic pathway, but NR2C/D-containing NMDARs at the cortical pathway. LTD appears to be induced post-synaptically at the thalamic input but presynaptically at the cortical input, since post-synaptic calcium chelation and NMDAR blockade prevent thalamic but not cortical LTD. These results highlight distinct molecular features of LTD in LA that may be relevant for traumatic memory and its erasure, and for pathologies such as post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Osvaldo Mirante
- Brain Research Institute, Medical Faculty, University Zürich Zürich, Switzerland ; Department of Health Science and Technology, Swiss Federal Institute of Technology Zürich, Switzerland
| | - Federico Brandalise
- Brain Research Institute, Medical Faculty, University Zürich Zürich, Switzerland
| | - Johannes Bohacek
- Brain Research Institute, Medical Faculty, University Zürich Zürich, Switzerland ; Department of Health Science and Technology, Swiss Federal Institute of Technology Zürich, Switzerland
| | - Isabelle M Mansuy
- Brain Research Institute, Medical Faculty, University Zürich Zürich, Switzerland ; Department of Health Science and Technology, Swiss Federal Institute of Technology Zürich, Switzerland
| |
Collapse
|
44
|
Lukowiak K, Heckler B, Bennett TE, Schriner EK, Wyrick K, Jewett C, Todd RP, Sorg BA. Enhanced memory persistence is blocked by a DNA methyltransferase inhibitor in the snail Lymnaea stagnalis. ACTA ACUST UNITED AC 2014; 217:2920-9. [PMID: 24902747 DOI: 10.1242/jeb.106765] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lymnaea stagnalis provides an excellent model system for studying memory because these snails have a well-described set of neurons, a single one of which controls expression of long-term memory of operantly conditioned respiratory behavior. We have shown that several different manipulations, including pre-training exposure to serotonin (5-HT) or methamphetamine, submersion of snails after training to prevent memory interference, and exposure to effluent from predatory crayfish (CE), enhance memory persistence. Changes in DNA methylation underlie formation of strong memories in mammals and 5-HT-enhanced long-term facilitation in Aplysia. Here we determined the impact of the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5-AZA; 87 μmol l(-1)), on enhanced memory persistence by all four manipulations. We found that 5-HT (100 μmol l(-1)) enhanced memory persistence, which was blocked by 5-AZA pretreatment. Snails pre-exposed to 3.3 μmol l(-1) Meth 4 h prior to training demonstrated memory 72 h later, which was not present in controls. This memory-enhancing effect was blocked by pre-treatment with 87 μmol l(-1) 5-AZA. Similarly, submersion to prevent interference learning as well as training in CE produced memory that was not present in controls, and these effects were blocked by pre-treatment with 87 μmol l(-1) 5-AZA. In contrast, 5-AZA injection did not alter expression of normal (non-enhanced) memory, suggesting that these four stimuli enhance memory persistence by increasing DNA methyltransferase activity, which, in turn, increases expression of memory-enhancing genes and/or inhibits memory suppressor genes. These studies lay important groundwork for delineating gene methylation changes that are common to persistent memory produced by different stimuli.
Collapse
Affiliation(s)
- Ken Lukowiak
- Cumming School of Medicine, University of Calgary, Calgary, AL T2N 4N1, Canada
| | - Benjamin Heckler
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Thomas E Bennett
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Ellen K Schriner
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Kathryn Wyrick
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Cynthia Jewett
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Ryan P Todd
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| | - Barbara A Sorg
- Alcohol and Drug Abuse Research Program and Translational Addiction Research Center, Department of Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA 98686, USA
| |
Collapse
|
45
|
Callaghan BL, Li S, Richardson R. The elusive engram: what can infantile amnesia tell us about memory? Trends Neurosci 2014; 37:47-53. [DOI: 10.1016/j.tins.2013.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/24/2013] [Accepted: 10/28/2013] [Indexed: 01/19/2023]
|
46
|
Effects of social instability stress in adolescence on long-term, not short-term, spatial memory performance. Behav Brain Res 2013; 256:165-71. [DOI: 10.1016/j.bbr.2013.08.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/01/2013] [Accepted: 08/05/2013] [Indexed: 01/01/2023]
|
47
|
Rahimian R, Fakhfouri G, Ejtemaei Mehr S, Ghia JE, Genazzani AA, Payandemehr B, Dehpour AR, Mousavizadeh K, Lim D. Tropisetron attenuates amyloid-beta-induced inflammatory and apoptotic responses in rats. Eur J Clin Invest 2013; 43:1039-51. [PMID: 23937291 DOI: 10.1111/eci.12141] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 07/09/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder featured by deposition of beta-amyloid (Aβ) plaques in the hippocampus and associated cortices and progressive cognitive decline. Tropisetron, a selective 5-HT3 receptor antagonist, is conventionally used to counteract chemotherapy-induced emesis. Recent investigations describe antiphlogistic properties for tropisetron. It has been shown that tropisetron protects against rat embolic stroke. We investigated protective properties of tropisetron in a beta-amyloid (Aβ) rat model of AD and possible involvement of 5-HT3 receptors. MATERIAL AND METHODS Aβ (1-42) was injected into the hippocampus of male rats. Animals were treated intracerebroventricularly with tropisetron, mCPBG (selective 5-HT3 receptor agonist) or mCPBG plus tropisetron on days 1, 3, 5 and 7. Seven days following Aβ administration, inflammatory markers (TNF-α, COX-2, iNOS and NF-κB), apoptotic markers (caspase 3 cytochrome c release) and calcineurin phosphatase activity were assessed in hippocampus. RESULTS Seven days following Aβ inoculation, control animals displayed dramatic increase in TNF-α, COX-2, iNOS, NF-κB, active caspase 3, cytochrome c release and calcineurin phosphatase activity in the hippocampus. Tropisetron significantly diminished the elevated levels of these markers and reversed the cognitive deficit. Interestingly, tropisetron was also found to be a potent inhibitor of calcineurin phosphatase activity. The selective 5-HT3 receptor agonist mCPBG, when co-administered with tropisetron, completely reversed the procognitive and anti-apoptotic properties of tropisetron while it could only partially counteract the anti-inflammatory effects. mCPBG alone significantly aggravated Aβ-induced injury. CONCLUSION Our findings indicate that tropisetron protects against Aβ-induced neurotoxicity in vivo through both 5-HT3 receptor-dependent and independent pathways.
Collapse
Affiliation(s)
- Reza Rahimian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Injury Repair Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lee SC, Li A, Calo S, Heitman J. Calcineurin plays key roles in the dimorphic transition and virulence of the human pathogenic zygomycete Mucor circinelloides. PLoS Pathog 2013; 9:e1003625. [PMID: 24039585 PMCID: PMC3764228 DOI: 10.1371/journal.ppat.1003625] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 06/27/2013] [Indexed: 11/19/2022] Open
Abstract
Many pathogenic fungi are dimorphic and switch between yeast and filamentous states. This switch alters host-microbe interactions and is critical for pathogenicity. However, in zygomycetes, whether dimorphism contributes to virulence is a central unanswered question. The pathogenic zygomycete Mucor circinelloides exhibits hyphal growth in aerobic conditions but switches to multi-budded yeast growth under anaerobic/high CO₂ conditions. We found that in the presence of the calcineurin inhibitor FK506, Mucor exhibits exclusively multi-budded yeast growth. We also found that M. circinelloides encodes three calcineurin catalytic A subunits (CnaA, CnaB, and CnaC) and one calcineurin regulatory B subunit (CnbR). Mutations in the latch region of CnbR and in the FKBP12-FK506 binding domain of CnaA result in hyphal growth of Mucor in the presence of FK506. Disruption of the cnbR gene encoding the sole calcineurin B subunit necessary for calcineurin activity yielded mutants locked in permanent yeast phase growth. These findings reveal that the calcineurin pathway plays key roles in the dimorphic transition from yeast to hyphae. The cnbR yeast-locked mutants are less virulent than the wild-type strain in a heterologous host system, providing evidence that hyphae or the yeast-hyphal transition are linked to virulence. Protein kinase A activity (PKA) is elevated during yeast growth under anaerobic conditions, in the presence of FK506, or in the yeast-locked cnbR mutants, suggesting a novel connection between PKA and calcineurin. cnaA mutants lacking the CnaA catalytic subunit are hypersensitive to calcineurin inhibitors, display a hyphal polarity defect, and produce a mixture of yeast and hyphae in aerobic culture. The cnaA mutants also produce spores that are larger than wild-type, and spore size is correlated with virulence potential. Our results demonstrate that the calcineurin pathway orchestrates the yeast-hyphal and spore size dimorphic transitions that contribute to virulence of this common zygomycete fungal pathogen.
Collapse
Affiliation(s)
- Soo Chan Lee
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Alicia Li
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Silvia Calo
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
49
|
Roy A, Jana M, Corbett GT, Ramaswamy S, Kordower JH, Gonzalez FJ, Pahan K. Regulation of cyclic AMP response element binding and hippocampal plasticity-related genes by peroxisome proliferator-activated receptor α. Cell Rep 2013; 4:724-37. [PMID: 23972989 DOI: 10.1016/j.celrep.2013.07.028] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/28/2013] [Accepted: 07/18/2013] [Indexed: 01/19/2023] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a transcription factor that regulates genes involved in fatty acid catabolism. Here, we provide evidence that PPARα is constitutively expressed in nuclei of hippocampal neurons and, surprisingly, controls calcium influx and the expression of various plasticity-related genes via direct transcriptional regulation of cyclic AMP response element binding (CREB). Accordingly, Pparα-null, but not Pparβ-null, mice are deficient in CREB and memory-associated proteins and have decreased spatial learning and memory. Small hairpin RNA knockdown of PPARα in the hippocampus suppressed CREB and NR2A, rendering wild-type animals markedly poor in consolidating spatial memory, whereas introduction of PPARα to the hippocampus of Pparα-null mice increased hippocampal CREB and NR2A and improved spatial learning and memory. Through detailed analyses of CREB and NR2A activity, as well as spatial learning and memory in bone marrow chimeric animals lacking PPARα in the CNS, we uncover a mechanism for transcriptional control of Creb and associated plasticity genes by PPARα.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Rashid AJ, Cole CJ, Josselyn SA. Emerging roles for MEF2 transcription factors in memory. GENES BRAIN AND BEHAVIOR 2013; 13:118-25. [PMID: 23790063 DOI: 10.1111/gbb.12058] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 06/19/2013] [Indexed: 01/08/2023]
Abstract
In the brain, transcription factors are critical for linking external stimuli to protein production, enabling neurons and neuronal networks to adapt to the ever-changing landscape. Gene transcription and protein synthesis are also vital for the formation of long-term memory. Members of the myocyte enhancer factor-2 (MEF2) family of transcription factors have a well-characterized role in the development of a variety of tissues, but their role in the adult brain is only beginning to be understood. Recent evidence indicates that MEF2 regulates the structural and synaptic plasticity underlying memory formation. However, in stark contrast to most other transcription factors implicated in memory, MEF2-mediated transcription constrains (rather than promotes) memory formation. Here, we review recent data examining the role of MEF2 in adult memory formation in rodents.
Collapse
Affiliation(s)
- A J Rashid
- Program in Neurosciences & Mental Health, Hospital for Sick Children; Department of Psychology; Department of Physiology; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|