1
|
Radahmadi M, Halabian A, Halabian A. An overview of extracellular field potentials: Different potentiation and measurable components, interpretations, and hippocampal synaptic activity models. Methods 2025; 239:50-63. [PMID: 40147603 DOI: 10.1016/j.ymeth.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
The hippocampus and some other brain regions are critically involved in synaptic plasticity. Electrophysiological recordings using extracellular field potentials (EFPs) reveal diverse synaptic activity within the hippocampus, including input/output functions (reflecting neural excitability), paired-pulse responses (reflecting short-term plasticity), and long-term potentiation (reflecting long-term plasticity). EFP techniques offer various measurable components for assessing multiple neural functions. These include fEPSP slope, amplitude, and area under curve (AUC), as well as latency (fEPSP onset or peak after stimulation), width at half amplitude, fiber volley, decay time, time-course (fEPSP rise and decay time constants; tau), initial slope/initial area and -/late area derived from a fEPSP waveform sample. Each of these parameters is separately evaluated and provides distinct electrophysiological interpretations. Despite the rich data offered by EFP techniques, many studies adopt a limited approach, focusing solely on fEPSP slope, amplitude, and occasionally AUC, thereby neglecting the potential insights provided by other parameters. Given the inherent variability of fEPSP components within a single recording and timeframe, a comprehensive analysis of synaptic activity within a specific hippocampal region is necessary for obtaining the full spectrum of fEPSP-related data. Researchers should consider the potential influence of additional factors contributing to the variability of synaptic activity magnitude. A detailed analysis considering different parts of extracellular fEPSP recordings and their properties is crucial for a deeper understanding of synaptic activity changes within the brain. Therefore, this review aims to provide a comprehensive overview of diverse forms of hippocampal synaptic activity, measurable components of EFP recordings, and their corresponding interpretations.
Collapse
Affiliation(s)
- Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Alireza Halabian
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Arshia Halabian
- Department of Electrical Engineering, University of Isfahan, Isfahan, Iran
| |
Collapse
|
2
|
Hernández-Frausto M, Galván EJ, López-Rubalcava C. Dopamine D1 receptors activation rescues hippocampal synaptic plasticity and cognitive impairments in the MK-801 neonatal schizophrenia model. Behav Brain Res 2025; 476:115250. [PMID: 39277140 DOI: 10.1016/j.bbr.2024.115250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/27/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
Schizophrenia is a disorder with a higher cognitive decline in early adulthood, causing impaired retention of episodic memories. However, the physiological and behavioral functions that underlie cognitive deficits with a potential mechanism to ameliorate and improve cognitive performance are unknown. In this study, we used the MK-801 neurodevelopmental schizophrenia-like model. Rats were divided into two groups: one received MK-801, and the other received saline for five consecutive days (7-11 postnatal days, PND). We evaluated synaptic plasticity late-LTP and spatial memory consolidation in early adolescence and young adulthood using extracellular field recordings in acute hippocampal slices and the Barnes maze task. Next, we examined D1 receptor (D1R) activation as a mechanism to ameliorate cognitive impairments. Our results suggest that MK-801 neonatal treatment induces impairment in late-LTP expression and deficits in spatial memory retrieval in early adolescence that is maintained until young adulthood. Furthermore, we found that activation of dopamine D1R ameliorates the impairments and promotes a robust expression of late-LTP and an improved performance in the Barnes maze task, suggesting a novel and potential therapeutic role in treating cognitive impairments in schizophrenia.
Collapse
Affiliation(s)
- Melissa Hernández-Frausto
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - Emilio J Galván
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - Carolina López-Rubalcava
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico.
| |
Collapse
|
3
|
Tadinada SM, Walsh EN, Mukherjee U, Abel T. Differential effects of Phosphodiesterase 4A5 on cAMP-dependent forms of long-term potentiation. J Physiol 2024. [PMID: 39693518 DOI: 10.1113/jp286801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
cAMP signalling is critical for memory consolidation and certain forms of long-term potentiation (LTP). Phosphodiesterases (PDEs), enzymes that degrade the second messengers cAMP and cGMP, are highly conserved during evolution and represent a unique set of drug targets, given the involvement of these enzymes in several pathophysiological states including brain disorders. The PDE4 family of cAMP-selective PDEs exert regulatory roles in memory and synaptic plasticity, but the specific roles of distinct PDE4 isoforms in these processes are poorly understood. Building on our previous work demonstrating that spatial and contextual memory deficits were caused by expressing selectively the long isoform of the PDE4A subfamily, PDE4A5, in hippocampal excitatory neurons, we now investigate the effects of PDE4A isoforms on different cAMP-dependent forms of LTP. We found that PDE4A5 impairs long-lasting LTP induced by theta burst stimulation (TBS) while sparing long-lasting LTP induced by spaced four-train stimulation (4 × 100 Hz). This effect requires the unique N-terminus of PDE4A5 and is specific to this long isoform. Targeted overexpression of PDE4A5 in area CA1 is sufficient to impair TBS-LTP, suggesting that cAMP levels in the postsynaptic neuron are critical for TBS-LTP. Our results shed light on the inherent differences among the PDE4A subfamily isoforms, emphasizing the importance of the long isoforms, which have a unique N-terminal region. Advancing our understanding of the function of specific PDE isoforms will pave the way for developing isoform-selective approaches to treat the cognitive deficits that are debilitating aspects of psychiatric, neurodevelopmental and neurodegenerative disorders. KEY POINTS: Hippocampal overexpression of PDE4A5, but not PDE4A1 or the N-terminus-truncated PDE4A5 (PDE4A5Δ4), selectively impairs long-term potentiation (LTP) induced by theta burst stimulation (TBS-LTP). Expression of PDE4A5 in area CA1 is sufficient to cause deficits in TBS-LTP. Hippocampal overexpression of the PDE4A isoforms PDE4A1 and PDE4A5 does not impair LTP induced by repeated tetanic stimulation at the CA3-CA1 synapses. These results suggest that PDE4A5, through its N-terminus, regulates cAMP pools that are critical for memory consolidation and expression of specific forms of long-lasting synaptic plasticity at CA3-CA1 synapses.
Collapse
Affiliation(s)
- Satya Murthy Tadinada
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
| | - Emily N Walsh
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
| | - Utsav Mukherjee
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, The University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
4
|
Daume J, Kamiński J, Salimpour Y, Gómez Palacio Schjetnan A, Anderson WS, Valiante TA, Mamelak AN, Rutishauser U. Persistent activity during working memory maintenance predicts long-term memory formation in the human hippocampus. Neuron 2024; 112:3957-3968.e3. [PMID: 39406238 PMCID: PMC11624075 DOI: 10.1016/j.neuron.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/22/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024]
Abstract
Working memory (WM) and long-term memory (LTM) are often viewed as separate cognitive systems. Little is known about how these systems interact when forming memories. We recorded single neurons in the human medial temporal lobe while patients maintained novel items in WM and completed a subsequent recognition memory test for the same items. In the hippocampus, but not in the amygdala, the level of WM content-selective persistent activity during WM maintenance was predictive of whether the item was later recognized with high confidence or forgotten. By contrast, visually evoked activity in the same cells was not predictive of LTM formation. During LTM retrieval, memory-selective neurons responded more strongly to familiar stimuli for which persistent activity was high while they were maintained in WM. Our study suggests that hippocampal persistent activity of the same cells supports both WM maintenance and LTM encoding, thereby revealing a common single-neuron component of these two memory systems.
Collapse
Affiliation(s)
- Jonathan Daume
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jan Kamiński
- Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Yousef Salimpour
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | - William S Anderson
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Taufik A Valiante
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada; Department of Electrical and Computer Engineering, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Adam N Mamelak
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ueli Rutishauser
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
5
|
Cui H, Ding H, Hu L, Zhao Y, Shu Y, Voon V. A novel dual-site OFC-dlPFC accelerated repetitive transcranial magnetic stimulation for depression: a pilot randomized controlled study. Psychol Med 2024; 54:1-14. [PMID: 39440449 PMCID: PMC11578911 DOI: 10.1017/s0033291724002289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND This study aimed to evaluate a novel rTMS protocol for treatment-resistant depression (TRD), using an EEG 10-20 system guided dual-target accelerated approach of right lateral orbitofrontal cortex (lOFC) inhibition followed by left dorsolateral prefrontal cortex (dlPFC) excitation, along with comparing 20 Hz dlPFC accelerated TMS v. sham. METHODS Seventy five patients participated in this trial consisting of 20 sessions over 5 consecutive days comparing dual-site (cTBS of right lOFC followed sequentially by 20 Hz rTMS of left dlPFC), active control (sham right lOFC followed by 20 Hz rTMS of left dlPFC) and sham control (sham for both targets). Resting-state fMRI was acquired prior to and following treatment. RESULTS Hamilton Rating Scale for Depression (HRSD-24) scores were similarly significantly improved at 4 weeks in both the Dual and Single group relative to Sham. Planned comparisons immediately after treatment highlighted greater HRSD-24 clinical responders (Dual: 47.8% v. Single:18.2% v. Sham:4.3%, χ2 = 13.0, p = 0.002) and in PHQ-9 scores by day 5 in the Dual relative to Sham group. We further showed that accelerated 20 Hz stimulation targeting the left dlPFC (active control) is significantly better than sham at 4 weeks. Dual stimulation decreased lOFC-subcallosal cingulate functional connectivity. Greater baseline lOFC-thalamic connectivity predicted better therapeutic response, while decreased lOFC-thalamic connectivity correlated with better response. CONCLUSIONS Our novel accelerated dual TMS protocol shows rapid clinically relevant antidepressant efficacy which may be related to state-modulation. This study has implications for community-based accessible TMS without neuronavigation and rapid onset targeting suicidal ideation and accelerated discharge from hospital.
Collapse
Affiliation(s)
- Hailun Cui
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Ding
- Department of Radiology, The Second People's Hospital of Guizhou Province, Guiyang, China
| | - Lingyan Hu
- Department of Psychiatric Rehabilitation, The Second People's Hospital of Guizhou Province, Guiyang, China
| | - Yijie Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
- Zhangjiang Fudan International Innovation Centre, Shanghai, China
| | - Yanping Shu
- Department of Psychiatry of Women and Children, The Second People's Hospital of Guizhou Province, Guiyang, China
| | - Valerie Voon
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
- Zhangjiang Fudan International Innovation Centre, Shanghai, China
| |
Collapse
|
6
|
Azarfarin M, Ghadiri T, Dadkhah M, Sahab-Negah S. The interaction between cannabinoids and long-term synaptic plasticity: A survey on memory formation and underlying mechanisms. Cell Biochem Funct 2024; 42:e4100. [PMID: 39090824 DOI: 10.1002/cbf.4100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
Synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD), is an essential phenomenon in memory formation as well as maintenance along with many other cognitive functions, such as those needed for coping with external stimuli. Synaptic plasticity consists of gradual changes in the biochemistry and morphology of pre- and postsynaptic neurons, particularly in the hippocampus. Consuming marijuana as a primary source of exocannabinoids immediately impairs attention and working memory-related tasks. Evidence regarding the effects of cannabinoids on LTP and memory is contradictory. While cannabinoids can affect a variety of specific cannabinoid receptors (CBRs) and nonspecific receptors throughout the body and brain, they exert miscellaneous systemic and local cerebral effects. Given the increasing use of cannabis, mainly among the young population, plus its potential adverse long-term effects on learning and memory processes, it could be a future global health challenge. Indeed, the impact of cannabinoids on memory is multifactorial and depends on the dosage, timing, formula, and route of consumption, plus the background complex interaction of the endocannabinoids system with other cerebral networks. Herein, we review how exogenously administrated organic cannabinoids, CBRs agonists or antagonists, and endocannabinoids can affect LTP and synaptic plasticity through various receptors in interaction with other cerebral pathways and primary neurotransmitters.
Collapse
Affiliation(s)
- Maryam Azarfarin
- Department of Neuroscience,Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tahereh Ghadiri
- Department of Neuroscience,Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoomeh Dadkhah
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Sahab-Negah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Daume J, Kamiński J, Salimpour Y, Anderson WS, Valiante TA, Mamelak AN, Rutishauser U. Persistent activity during working memory maintenance predicts long-term memory formation in the human hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.15.603630. [PMID: 39071325 PMCID: PMC11275810 DOI: 10.1101/2024.07.15.603630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Working Memory (WM) and Long-Term Memory (LTM) are often viewed as separate cognitive systems. Little is known about how these systems interact when forming memories. We recorded single neurons in the human medial temporal lobe while patients maintained novel items in WM and a subsequent recognition memory test for the same items. In the hippocampus but not the amygdala, the level of WM content-selective persist activity during WM maintenance was predictive of whether the item was later recognized with high confidence or forgotten. In contrast, visually evoked activity in the same cells was not predictive of LTM formation. During LTM retrieval, memory-selective neurons responded more strongly to familiar stimuli for which persistent activity was high while they were maintained in WM. Our study suggests that hippocampal persistent activity of the same cell supports both WM maintenance and LTM encoding, thereby revealing a common single-neuron component of these two memory systems.
Collapse
Affiliation(s)
- Jonathan Daume
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jan Kamiński
- Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Yousef Salimpour
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - William S Anderson
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Taufik A Valiante
- Krembil Research Institute, Toronto Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering | Department of Electrical and Computer Engineering, University of Toronto, Toronto, Canada
| | - Adam N Mamelak
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Ueli Rutishauser
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
8
|
Latif‐Hernandez A, Yang T, Butler RR, Losada PM, Minhas PS, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson KI, Wyss‐Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. Alzheimers Dement 2024; 20:4434-4460. [PMID: 38779814 PMCID: PMC11247716 DOI: 10.1002/alz.13857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Tropomyosin related kinase B (TrkB) and C (TrkC) receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid beta (Aβ) toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. METHODS PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APPL/S) and wild-type controls. Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA sequencing. RESULTS In APPL/S mice, BD10-2 treatment improved memory and LTP deficits. This was accompanied by normalized phosphorylation of protein kinase B (Akt), calcium-calmodulin-dependent kinase II (CaMKII), and AMPA-type glutamate receptors containing the subunit GluA1; enhanced activity-dependent recruitment of synaptic proteins; and increased excitatory synapse number. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. DISCUSSION BD10-2 prevented APPL/S/Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response. HIGHLIGHTS Small molecule modulation of tropomyosin related kinase B (TrkB) and C (TrkC) restores long-term potentiation (LTP) and behavior in an Alzheimer's disease (AD) model. Modulation of TrkB and TrkC regulates synaptic activity-dependent transcription. TrkB and TrkC receptors are candidate targets for translational therapeutics. Electrophysiology combined with transcriptomics elucidates synaptic restoration. LTP identifies neuron and microglia AD-relevant human-mouse co-expression modules.
Collapse
Affiliation(s)
- Amira Latif‐Hernandez
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Tao Yang
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Robert R. Butler
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Patricia Moran Losada
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| | - Paras S. Minhas
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Halle White
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Kevin C. Tran
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Harry Liu
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Danielle A. Simmons
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Vanessa Langness
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Katrin I. Andreasson
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Frank M. Longo
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
9
|
Chowdhury KU, Holden ME, Wiley MT, Suppiramaniam V, Reed MN. Effects of Cannabis on Glutamatergic Neurotransmission: The Interplay between Cannabinoids and Glutamate. Cells 2024; 13:1130. [PMID: 38994982 PMCID: PMC11240741 DOI: 10.3390/cells13131130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
There has been a significant increase in the consumption of cannabis for both recreational and medicinal purposes in recent years, and its use can have long-term consequences on cognitive functions, including memory. Here, we review the immediate and long-term effects of cannabis and its derivatives on glutamatergic neurotransmission, with a focus on both the presynaptic and postsynaptic alterations. Several factors can influence cannabinoid-mediated changes in glutamatergic neurotransmission, including dosage, sex, age, and frequency of use. Acute exposure to cannabis typically inhibits glutamate release, whereas chronic use tends to increase glutamate release. Conversely, the postsynaptic alterations are more complicated than the presynaptic effects, as cannabis can affect the glutamate receptor expression and the downstream signaling of glutamate. All these effects ultimately influence cognitive functions, particularly memory. This review will cover the current research on glutamate-cannabis interactions, as well as the future directions of research needed to understand cannabis-related health effects and neurological and psychological aspects of cannabis use.
Collapse
Affiliation(s)
- Kawsar U. Chowdhury
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 30144, USA
| | | | - Miles T. Wiley
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, GA 30144, USA
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA; (K.U.C.); (M.T.W.)
- Center for Neuroscience Initiative, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
10
|
Bonilla-Quintana M, Rangamani P. Biophysical Modeling of Actin-Mediated Structural Plasticity Reveals Mechanical Adaptation in Dendritic Spines. eNeuro 2024; 11:ENEURO.0497-23.2024. [PMID: 38383589 DOI: 10.1523/eneuro.0497-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/13/2024] [Indexed: 02/23/2024] Open
Abstract
Synaptic plasticity is important for learning and memory formation; it describes the strengthening or weakening of connections between synapses. The postsynaptic part of excitatory synapses resides in dendritic spines, which are small protrusions on the dendrites. One of the key features of synaptic plasticity is its correlation with the size of these spines. A long-lasting synaptic strength increase [long-term potentiation (LTP)] is only possible through the reconfiguration of the actin spine cytoskeleton. Here, we develop an experimentally informed three-dimensional computational model in a moving boundary framework to investigate this reconfiguration. Our model describes the reactions between actin and actin-binding proteins leading to the cytoskeleton remodeling and their effect on the spine membrane shape to examine the spine enlargement upon LTP. Moreover, we find that the incorporation of perisynaptic elements enhances spine enlargement upon LTP, exhibiting the importance of accounting for these elements when studying structural LTP. Our model shows adaptation to repeated stimuli resulting from the interactions between spine proteins and mechanical forces.
Collapse
Affiliation(s)
- Mayte Bonilla-Quintana
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California 92093
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
11
|
Bartsch CJ, Jacobs JT, Mojahed N, Qasem E, Smith M, Caldwell O, Aaflaq S, Nordman JC. Visualizing traumatic stress-induced structural plasticity in a medial amygdala pathway using mGRASP. Front Mol Neurosci 2023; 16:1313635. [PMID: 38098941 PMCID: PMC10720331 DOI: 10.3389/fnmol.2023.1313635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023] Open
Abstract
Traumatic stress has been shown to contribute to persistent behavioral changes, yet the underlying neural pathways are not fully explored. Structural plasticity, a form of long-lasting neural adaptability, offers a plausible mechanism. To scrutinize this, we used the mGRASP imaging technique to visualize synaptic modifications in a pathway formed between neurons of the posterior ventral segment of the medial amygdala and ventrolateral segment of the ventromedial hypothalamus (MeApv-VmHvl), areas we previously showed to be involved in stress-induced excessive aggression. We subjected mice (7-8 weeks of age) to acute stress through foot shocks, a reliable and reproducible form of traumatic stress, and compared synaptic changes to control animals. Our data revealed an increase in synapse formation within the MeApv-VmHvl pathway post-stress as evidenced by an increase in mGRASP puncta and area. Chemogenetic inhibition of CaMKIIα-expressing neurons in the MeApv during the stressor led to reduced synapse formation, suggesting that the structural changes were driven by excitatory activity. To elucidate the molecular mechanisms, we administered the NMDAR antagonist MK-801, which effectively blocked the stress-induced synaptic changes. These findings suggest a strong link between traumatic stress and enduring structural changes in an MeApv-VmHvl neural pathway. Furthermore, our data point to NMDAR-dependent mechanisms as key contributors to these synaptic changes. This structural plasticity could offer insights into persistent behavioral consequences of traumatic stress, such as symptoms of PTSD and social deficits.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jacob C. Nordman
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, United States
| |
Collapse
|
12
|
Latif-Hernandez A, Yang T, Raymond-Butler R, Losada PM, Minhas P, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson K, Wyss-Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558138. [PMID: 37781573 PMCID: PMC10541128 DOI: 10.1101/2023.09.18.558138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Introduction TrkB and TrkC receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid-β (Aβ)-toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. Methods PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APP L/S ) and wild-type controls (WT). Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA-sequencing. Results Memory and LTP deficits in APP L/S mice were attenuated by treatment with BD10-2. BD10-2 prevented aberrant AKT, CaMKII, and GLUA1 phosphorylation, and enhanced activity-dependent recruitment of synaptic proteins. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. Conclusions BD10-2 prevented APP L/S /Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response.
Collapse
|
13
|
Grover FM, Chen B, Perez MA. Increased paired stimuli enhance corticospinal-motoneuronal plasticity in humans with spinal cord injury. J Neurophysiol 2023; 129:1414-1422. [PMID: 36752493 PMCID: PMC10259851 DOI: 10.1152/jn.00499.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
Paired corticospinal-motoneuronal stimulation (PCMS) has been used to enhance corticospinal excitability and functional outcomes in humans with spinal cord injury (SCI). Here, we examined the effect of increasing the number of paired pulses on PCMS-induced plasticity. During PCMS, corticospinal volleys evoked by transcranial magnetic stimulation (TMS) over the hand motor cortex were timed to arrive at corticospinal-motoneuronal synapses of the first dorsal interosseous (FDI) muscle 1-2 ms before the arrival of antidromic potentials elicited in motoneurons by electrical stimulation of the ulnar nerve. We tested motor-evoked potentials (MEPs) elicited by TMS over the hand motor cortex and electrical stimulation at the cervicomedullary junction (CMEPs) in the FDI muscle before and after 180 paired pulses (PCMS-180) followed up by another 180 paired pulses (PCMS-360) in humans with and without chronic incomplete cervical SCI. The nine-hole-peg-test (9HPT) was measured before and after PCMS paired pulses in individuals with SCI. We found that the size of MEPs and CMEPs increased after PCMS-180 in both groups compared with baseline and further increased after PCMS-360 in participants with SCI, suggesting a spinal origin for these effects. Notably, in people with SCI, the time to complete the 9HPT decreased after PCMS-180 and further decreased after PCMS-360 compared with baseline but not when the 9HPT was repeated overtime. Our findings demonstrate that increasing the number of PCMS paired pulses potentiates corticospinal excitability and voluntary motor output after SCI, likely through spinal plasticity. This proof-of-principle study suggests that increasing the PCMS dose represents a strategy to boost voluntary motor output after SCI.NEW & NOTEWORTHY Paired corticospinal-motoneuronal stimulation (PCMS) has been used to enhance corticospinal excitability and functional outcomes in humans with spinal cord injury (SCI). Here, we demonstrate that 360 paired pulses resulted in larger increases in motor-evoked potential size in a hand muscle and in a better ability to complete the nine-hold-peg-test compared with 180 paired pulses in people with SCI. This proof-of-principle study suggests that increasing the PCMS dose represents a strategy to boost motor output after SCI.
Collapse
Affiliation(s)
- Francis M Grover
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago, Illinois, United States
| | - Bing Chen
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Edward Hines Jr. VA Hospital, Chicago, Illinois, United States
| | - Monica A Perez
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago, Illinois, United States
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, United States
- Edward Hines Jr. VA Hospital, Chicago, Illinois, United States
| |
Collapse
|
14
|
Champagne PL, Blanchette AK, Schneider C. Continuous, and not intermittent, theta-burst stimulation of the unlesioned hemisphere improved brain and hand function in chronic stroke: A case study. BRAIN DISORDERS 2023. [DOI: 10.1016/j.dscb.2022.100062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
15
|
Jain A, Woolley CS. Mechanisms That Underlie Expression of Estradiol-Induced Excitatory Synaptic Potentiation in the Hippocampus Differ between Males and Females. J Neurosci 2023; 43:1298-1309. [PMID: 36650060 PMCID: PMC9987570 DOI: 10.1523/jneurosci.2080-19.2023] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
17β-estradiol (E2) is synthesized in the hippocampus of both sexes and acutely potentiates excitatory synapses in each sex. Previously, we found that the mechanisms for initiation of E2-induced synaptic potentiation differ between males and females, including in the molecular signaling involved. Here, we used electrical stimulation and two-photon glutamate uncaging in hippocampal slices from adult male and female rats to investigate whether the downstream consequences of distinct molecular signaling remain different between the sexes or converge to the same mechanism(s) of expression of potentiation. This showed that synaptic activity is necessary for expression of E2-induced potentiation in females but not males, which paralleled a sex-specific requirement in females for calcium-permeable AMPARs (cpAMPARs) to stabilize potentiation. Nonstationary fluctuation analysis of two-photon evoked unitary synaptic currents showed that the postsynaptic component of E2-induced potentiation occurs either through an increase in AMPAR conductance or in nonconductive properties of AMPARs (number of channels × open probability) and never both at the same synapse. In females, most synapses (76%) were potentiated via increased AMPAR conductance, whereas in males, more synapses (60%) were potentiated via an increase in nonconductive AMPAR properties. Inhibition of cpAMPARs eliminated E2-induced synaptic potentiation in females, whereas some synapses in males were unaffected by cpAMPAR inhibition; these synapses in males potentiated exclusively via increased AMPAR nonconductive properties. This sex bias in expression mechanisms of E2-induced synaptic potentiation underscores the concept of latent sex differences in mechanisms of synaptic plasticity in which the same outcome in each sex is achieved through distinct underlying mechanisms.SIGNIFICANCE STATEMENT Estrogens are synthesized in the brains of both sexes and potentiate excitatory synapses to the same degree in each sex. Despite this apparent similarity, the molecular signaling that initiates estrogen-induced synaptic potentiation differs between the sexes. Here we show that these differences extend to the mechanisms of expression of synaptic potentiation and result in distinct patterns of postsynaptic neurotransmitter receptor modulation in each sex. Such latent sex differences, in which the same outcome is achieved through distinct underlying mechanisms in males versus females, indicate that molecular mechanisms targeted for drug development may differ between the sexes even in the absence of an overt sex difference in behavior or disease.
Collapse
Affiliation(s)
- Anant Jain
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
| | - Catherine S Woolley
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
16
|
Zulkifli NA, Hassan Z, Mustafa MZ, Azman WNW, Hadie SNH, Ghani N, Mat Zin AA. The potential neuroprotective effects of stingless bee honey. Front Aging Neurosci 2023; 14:1048028. [PMID: 36846103 PMCID: PMC9945235 DOI: 10.3389/fnagi.2022.1048028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/29/2022] [Indexed: 02/11/2023] Open
Abstract
Tropical Meliponini bees produce stingless bee honey (SBH). Studies have shown beneficial properties, including antibacterial, bacteriostatic, anti-inflammatory, neurotherapeutic, neuroprotective, wound, and sunburn healing capabilities. High phenolic acid and flavonoid concentrations offer SBH its benefits. SBH can include flavonoids, phenolic acids, ascorbic acid, tocopherol, organic acids, amino acids, and protein, depending on its botanical and geographic origins. Ursolic acid, p-coumaric acid, and gallic acid may diminish apoptotic signals in neuronal cells, such as nuclear morphological alterations and DNA fragmentation. Antioxidant activity minimizes reactive oxygen species (ROS) formation and lowers oxidative stress, inhibiting inflammation by decreasing enzymes generated during inflammation. Flavonoids in honey reduce neuroinflammation by decreasing proinflammatory cytokine and free radical production. Phytochemical components in honey, such as luteolin and phenylalanine, may aid neurological problems. A dietary amino acid, phenylalanine, may improve memory by functioning on brain-derived neurotrophic factor (BDNF) pathways. Neurotrophin BDNF binds to its major receptor, TrkB, and stimulates downstream signaling cascades, which are crucial for neurogenesis and synaptic plasticity. Through BDNF, SBH can stimulate synaptic plasticity and synaptogenesis, promoting learning and memory. Moreover, BDNF contributes to the adult brain's lasting structural and functional changes during limbic epileptogenesis by acting through the cognate receptor tyrosine receptor kinase B (TrkB). Given the higher antioxidants activity of SBH than the Apis sp. honey, it may be more therapeutically helpful. There is minimal research on SBH's neuroprotective effects, and the related pathways contribute to it is unclear. More research is needed to elucidate the underlying molecular process of SBH on BDNF/TrkB pathways in producing neuroprotective effects.
Collapse
Affiliation(s)
- Nurdarina Ausi Zulkifli
- Department of Pathology, School of Medical Sciences Universiti Sains Malaysia and Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia
| | - Mohd Zulkifli Mustafa
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Wan Norlina Wan Azman
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia and Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Siti Nurma Hanim Hadie
- Department of Anatomy, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Nurhafizah Ghani
- Basic and Medical Sciences Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Anani Aila Mat Zin
- Department of Pathology, School of Medical Sciences Universiti Sains Malaysia and Hospital Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
17
|
Chemogenetic Enhancement of cAMP Signaling Renders Hippocampal Synaptic Plasticity Resilient to the Impact of Acute Sleep Deprivation. eNeuro 2023; 10:ENEURO.0380-22.2022. [PMID: 36635248 PMCID: PMC9829098 DOI: 10.1523/eneuro.0380-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/24/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022] Open
Abstract
Sleep facilitates memory storage and even brief periods of sleep loss lead to impairments in memory, particularly memories that are hippocampus dependent. In previous studies, we have shown that the deficit in memory seen after sleep loss is accompanied by deficits in synaptic plasticity. Our previous work has also found that sleep deprivation (SD) is associated with reduced levels of cyclic adenosine monophosphate (cAMP) in the hippocampus and that the reduction of cAMP mediates the diminished memory observed in sleep-deprived animals. Based on these findings, we hypothesized that cAMP acts as a mediator for not only the cognitive deficits caused by sleep deprivation, but also the observed deficits in synaptic plasticity. In this study, we expressed the heterologous Drosophila melanogaster Gαs-protein-coupled octopamine receptor (DmOctβ1R) in mouse hippocampal neurons. This receptor is selectively activated by the systemically injected ligand (octopamine), thus allowing us to increase cAMP levels in hippocampal neurons during a 5-h sleep deprivation period. Our results show that chemogenetic enhancement of cAMP during the period of sleep deprivation prevents deficits in a persistent form of long-term potentiation (LTP) that is induced at the Schaffer collateral synapses in the hippocampal CA1 region. We also found that elevating cAMP levels in either the first or second half of sleep deprivation successfully prevented LTP deficits. These findings reveal that cAMP-dependent signaling pathways are key mediators of sleep deprivation at the synaptic level. Targeting these pathways could be useful in designing strategies to prevent the impact of sleep loss.
Collapse
|
18
|
Yesiltepe M, Yin T, Tambini MD, Breuillaud L, Zehntner SP, D’Adamio L. Late-long-term potentiation magnitude, but not Aβ levels and amyloid pathology, is associated with behavioral performance in a rat knock-in model of Alzheimer disease. Front Aging Neurosci 2022; 14:1040576. [PMID: 36438008 PMCID: PMC9691854 DOI: 10.3389/fnagi.2022.1040576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/31/2022] [Indexed: 09/23/2023] Open
Abstract
Cleavage of Amyloid precursor protein by β- and γ-secretases lead to Aβ formation. The widely accepted pathogenic model states that these mutations cause AD via an increase in Aβ formation and accumulation of Aβ in Amyloid plaques. APP mutations cause early onset familial forms of Alzheimer's disease (FAD) in humans. We generated App-Swedish (Apps ) knock-in rats, which carry a pathogenic APP mutation in the endogenous rat App gene. This mutation increases β-secretase processing of APP leading to both augmented Aβ production and facilitation of glutamate release in Apps/s rats, via a β-secretase and APP-dependent glutamate release mechanism. Here, we studied 11 to 14-month-old male and female Apps/s rats. To determine whether the Swedish App mutation leads to behavioral deficits, Apps/s knock-in rats were subjected to behavioral analysis using the IntelliCage platform, an automated behavioral testing system. This system allows behavioral assessment in socially housed animals reflecting a more natural, less stress-inducing environment and eliminates experimenter error and bias while increasing precision of measurements. Surprisingly, a spatial discrimination and flexibility task that can reveal deficits in higher order brain function showed that Apps/s females, but not Apps/s male rats, performed significantly worse than same sex controls. Moreover, female control rats performed significantly better than control and Apps/s male rats. The Swedish mutation causes a significant increase in Aβ production in 14-month-old animals of both sexes. Yet, male and female Apps/s rats showed no evidence of AD-related amyloid pathology. Finally, Apps/s rats did not show signs of significant neuroinflammation. Given that the APP Swedish mutation causes alterations in glutamate release, we analyzed Long-term potentiation (LTP), a long-lasting form of synaptic plasticity that is a cellular basis for learning and memory. Strikingly, LTP was significantly increased in Apps/s control females compared to both Apps/s sexes and control males. In conclusion, this study shows that behavioral performances are sex and App-genotype dependent. In addition, they are associated with LTP values and not Aβ or AD-related pathology. These data, and the failures of anti-Aβ therapies in humans, suggest that alternative pathways, such as those leading to LTP dysfunction, should be targeted for disease-modifying AD therapy.
Collapse
Affiliation(s)
- Metin Yesiltepe
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Tao Yin
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Marc D. Tambini
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | | | | | - Luciano D’Adamio
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
19
|
Shetty MS, Ris L, Schindler RFR, Mizuno K, Fedele L, Giese KP, Brand T, Abel T. Mice Lacking the cAMP Effector Protein POPDC1 Show Enhanced Hippocampal Synaptic Plasticity. Cereb Cortex 2022; 32:3457-3471. [PMID: 34937090 PMCID: PMC9376866 DOI: 10.1093/cercor/bhab426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Extensive research has uncovered diverse forms of synaptic plasticity and an array of molecular signaling mechanisms that act as positive or negative regulators. Specifically, cyclic 3',5'-cyclic adenosine monophosphate (cAMP)-dependent signaling pathways are crucially implicated in long-lasting synaptic plasticity. In this study, we examine the role of Popeye domain-containing protein 1 (POPDC1) (or blood vessel epicardial substance (BVES)), a cAMP effector protein, in modulating hippocampal synaptic plasticity. Unlike other cAMP effectors, such as protein kinase A (PKA) and exchange factor directly activated by cAMP, POPDC1 is membrane-bound and the sequence of the cAMP-binding cassette differs from canonical cAMP-binding domains, suggesting that POPDC1 may have an unique role in cAMP-mediated signaling. Our results show that Popdc1 is widely expressed in various brain regions including the hippocampus. Acute hippocampal slices from Popdc1 knockout (KO) mice exhibit PKA-dependent enhancement in CA1 long-term potentiation (LTP) in response to weaker stimulation paradigms, which in slices from wild-type mice induce only transient LTP. Loss of POPDC1, while not affecting basal transmission or input-specificity of LTP, results in altered response during high-frequency stimulation. Popdc1 KO mice also show enhanced forskolin-induced potentiation. Overall, these findings reveal POPDC1 as a novel negative regulator of hippocampal synaptic plasticity and, together with recent evidence for its interaction with phosphodiesterases (PDEs), suggest that POPDC1 is involved in modulating activity-dependent local cAMP-PKA-PDE signaling.
Collapse
Affiliation(s)
- Mahesh Shivarama Shetty
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Laurence Ris
- Department of Neuroscience, University of Mons, Research Institute for Health Sciences and Technology, 7000 Mons, Belgium
| | | | - Keiko Mizuno
- Department of Neuroscience, King’s College, London SE5 9NU, UK
| | - Laura Fedele
- National Heart and Lung Institute, Imperial College London, London W12 ONN, UK
| | | | - Thomas Brand
- National Heart and Lung Institute, Imperial College London, London W12 ONN, UK
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
20
|
Belén Sanz-Martos A, Fernández-Felipe J, Merino B, Cano V, Ruiz-Gayo M, Del Olmo N. Butyric Acid Precursor Tributyrin Modulates Hippocampal Synaptic Plasticity and Prevents Spatial Memory Deficits: Role of PPARγ and AMPK. Int J Neuropsychopharmacol 2022; 25:498-511. [PMID: 35152284 PMCID: PMC9211015 DOI: 10.1093/ijnp/pyac015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Short chain fatty acids (SCFA), such as butyric acid (BA), derived from the intestinal fermentation of dietary fiber and contained in dairy products, are gaining interest in relation to their possible beneficial effects on neuropsychological disorders. METHODS C57BL/6J male mice were used to investigate the effect of tributyrin (TB), a prodrug of BA, on hippocampus (HIP)-dependent spatial memory, HIP synaptic transmission and plasticity mechanisms, and the expression of genes and proteins relevant to HIP glutamatergic transmission. RESULTS Ex vivo studies, carried out in HIP slices, revealed that TB can transform early-LTP into late-LTP (l-LTP) and to rescue LTP-inhibition induced by scopolamine. The facilitation of l-LTP induced by TB was blocked both by GW9662 (a PPARγ antagonist) and C-Compound (an AMPK inhibitor), suggesting the involvement of both PPARγ and AMPK on TB effects. Moreover, 48-hour intake of a diet containing 1% TB prevented, in adolescent but not in adult mice, scopolamine-induced impairment of HIP-dependent spatial memory. In the adolescent HIP, TB upregulated gene expression levels of Pparg, leptin, and adiponectin receptors, and that of the glutamate receptor subunits AMPA-2, NMDA-1, NMDA-2A, and NMDA-2B. CONCLUSIONS Our study shows that TB has a positive influence on LTP and HIP-dependent spatial memory, which suggests that BA may have beneficial effects on memory.
Collapse
Affiliation(s)
- Ana Belén Sanz-Martos
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Jesús Fernández-Felipe
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Beatriz Merino
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Victoria Cano
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | | | - Nuria Del Olmo
- Correspondence: Nuria Del Olmo, PhD, Department of Psychobiology, School of Psychology, National University for Distance Education (UNED), C/ Juan del Rosal 10, 28040 Madrid, Spain ()
| |
Collapse
|
21
|
Rodrigues NC, Silva-Cruz A, Caulino-Rocha A, Bento-Oliveira A, Alexandre Ribeiro J, Cunha-Reis D. Hippocampal CA1 theta burst-induced LTP from weaning to adulthood: Cellular and molecular mechanisms in young male rats revisited. Eur J Neurosci 2021; 54:5272-5292. [PMID: 34251729 DOI: 10.1111/ejn.15390] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/08/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023]
Abstract
Long-term potentiation (LTP) is a highly studied cellular process, yet determining the transduction and gamma aminobutyric acid (GABAergic) pathways that are the essential versus modulatory for LTP elicited by theta burst stimulation (TBS) in the hippocampal Cornu Ammonis 1 (CA1) area is still elusive, due to the use of different TBS intensities, patterns or different rodent/cellular models. We now characterised the developmental maturation and the transduction and GABAergic pathways required for mild TBS-induced LTP in hippocampal CA1 area in male rats. LTP induced by TBS (5x4) (five bursts of four pulses delivered at 100 Hz) lasted for up to 3 h and was increasingly larger from weaning to adulthood. Stronger TBS patterns - TBS (15x4) or three TBS (15x4) separated by 6 min induced nearly maximal LTP not being the best choice to study the value of LTP-enhancing drugs. LTP induced by TBS (5x4) in young adults was fully dependent on N-methyl D-aspartate (NMDA) receptor and calmodulin-dependent protein kinase II (CaMKII) activity but independent of protein kinase A (PKA) or protein kinase C (PKC) activity. Furthermore, it was partially dependent on GABAB receptor activation and was potentiated by GABAA receptor blockade and less by GAT-1 transporter blockade. AMPA GluA1 phosphorylation on Ser831 (CaMKII target) but not GluA1 Ser845 (PKA target) was essential for LTP expression. The phosphorylation of the Kv4.2 channel was observed at Ser438 (CaMKII target) but not at Thr602 or Thr607 (ERK/MAPK pathway target). This suggests that cellular kinases like PKA, PKC, or kinases of the ERK/MAPK family although important modulators of TBS (5x4)-induced LTP may not be essential for its expression in the CA1 area of the hippocampus.
Collapse
Affiliation(s)
| | - Armando Silva-Cruz
- Instituto de Medicina Molecular, Unidade de Neurociências, Lisbon, Portugal
| | - Ana Caulino-Rocha
- Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal.,BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Bento-Oliveira
- Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal.,BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim Alexandre Ribeiro
- Instituto de Medicina Molecular, Unidade de Neurociências, Lisbon, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Diana Cunha-Reis
- Instituto de Medicina Molecular, Unidade de Neurociências, Lisbon, Portugal.,Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal.,BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
Historical perspective and progress on protein ubiquitination at glutamatergic synapses. Neuropharmacology 2021; 196:108690. [PMID: 34197891 DOI: 10.1016/j.neuropharm.2021.108690] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Transcription-translation coupling leads to the production of proteins that are key for controlling essential neuronal processes that include neuronal development and changes in synaptic strength. Although these events have been a prevailing theme in neuroscience, the regulation of proteins via posttranslational signaling pathways are equally relevant for these neuronal processes. Ubiquitin is one type of posttranslational modification that covalently attaches to its targets/substrates. Ubiquitination of proteins play a key role in multiple signaling pathways, the predominant being removal of its substrates by a large molecular machine called the proteasome. Here, I review 40 years of progress on ubiquitination in the nervous system at glutamatergic synapses focusing on axon pathfinding, synapse formation, presynaptic release, dendritic spine formation, and regulation of postsynaptic glutamate receptors. Finally, I elucidate emerging themes in ubiquitin biology that may challenge our current understanding of ubiquitin signaling in the nervous system.
Collapse
|
23
|
Bin Ibrahim MZ, Benoy A, Sajikumar S. Long-term plasticity in the hippocampus: maintaining within and 'tagging' between synapses. FEBS J 2021; 289:2176-2201. [PMID: 34109726 DOI: 10.1111/febs.16065] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/15/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Synapses between neurons are malleable biochemical structures, strengthening and diminishing over time dependent on the type of information they receive. This phenomenon known as synaptic plasticity underlies learning and memory, and its different forms, long-term potentiation (LTP) and long-term depression (LTD), perform varied cognitive roles in reinforcement, relearning and associating memories. Moreover, both LTP and LTD can exist in an early transient form (early-LTP/LTD) or a late persistent form (late-LTP/LTD), which are triggered by different induction protocols, and also differ in their dependence on protein synthesis and the involvement of key molecular players. Beyond homosynaptic modifications, synapses can also interact with one another. This is encapsulated in the synaptic tagging and capture hypothesis (STC), where synapses expressing early-LTP/LTD present a 'tag' that can capture the protein synthesis products generated during a temporally proximal late-LTP/LTD induction. This 'tagging' phenomenon forms the framework of synaptic interactions in various conditions and accounts for the cellular basis of the time-dependent associativity of short-lasting and long-lasting memories. All these synaptic modifications take place under controlled neuronal conditions, regulated by subcellular elements such as epigenetic regulation, proteasomal degradation and neuromodulatory signals. Here, we review current understanding of the different forms of synaptic plasticity and its regulatory mechanisms in the hippocampus, a brain region critical for memory formation. We also discuss expression of plasticity in hippocampal CA2 area, a long-overlooked narrow hippocampal subfield and the behavioural correlate of STC. Lastly, we put forth perspectives for an integrated view of memory representation in synapses.
Collapse
Affiliation(s)
- Mohammad Zaki Bin Ibrahim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore
| | - Amrita Benoy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
24
|
Ahnaou A, Broadbelt T, Biermans R, Huysmans H, Manyakov NV, Drinkenburg WHIM. The phosphodiesterase-4 and glycine transporter-1 inhibitors enhance in vivo hippocampal theta network connectivity and synaptic plasticity, whereas D-serine does not. Transl Psychiatry 2020; 10:197. [PMID: 32555167 PMCID: PMC7303193 DOI: 10.1038/s41398-020-00875-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Dysfunctional N-methyl-D-aspartate receptors (NMDARs) and cyclic adenosine monophosphate (cAMP) have been associated with deficits in synaptic plasticity and cognition found in neurodegenerative and neuropsychiatric disorders such as Alzheimer's disease (AD) and schizophrenia. Therapeutic approaches that indirectly enhance NMDAR function through increases in glycine and/or D-serine levels as well as inhibition of phosphodiesterases that reduces degradation of cAMP, are expected to enhance synaptic strength, connectivity and to potentially impact cognition processes. The present in vivo study investigated effects of subcutaneous administration of D-serine, the glycine transporter 1 (GlyT1) inhibitor SSR504734 and the PDE4 inhibitor rolipram, on network oscillations, connectivity and long-term potentiation (LTP) at the hippocampi circuits in Sprague-Dawley rats. In conscious animals, multichannel EEG recordings assessed network oscillations and connectivity at frontal and hippocampal CA1-CA3 circuits. Under urethane anaesthesia, field excitatory postsynaptic potentials (fEPSPs) were measured in the CA1 subfield of the hippocampus after high-frequency stimulation (HFS) of the Schaffer collateral-CA1 (SC) pathway. SSR504734 and rolipram significantly increased slow theta oscillations (4-6.5 Hz) at the CA1-CA3, slow gamma oscillations (30-50 Hz) in the frontal areas and enhanced coherence in the CA1-CA3 network, which were dissociated from motor behaviour. SSR504734 enhanced short-term potentiation (STP) and fEPSP responses were extended into LTP response, whereas the potentiation of EPSP slope was short-lived to STP with rolipram. Unlike glycine, increased levels of D-serine had no effect on network oscillations and limits the LTP induction and expression. The present data support a facilitating role of glycine and cAMP on network oscillations and synaptic efficacy at the CA3-CA1 circuit in rats, whereas raising endogenous D-serine levels had no such beneficial effects.
Collapse
Affiliation(s)
- A. Ahnaou
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - T. Broadbelt
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - R. Biermans
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - H. Huysmans
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - N. V. Manyakov
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - W. H. I. M. Drinkenburg
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
25
|
Zheng Y, Ma XX, Dong L, Ma W, Cheng JH. Effects of uninterrupted sinusoidal LF-EMF stimulation on LTP induced by different combinations of TBS/HFS at the Schaffer collateral-CA1 of synapses. Brain Res 2019; 1725:146487. [PMID: 31580873 DOI: 10.1016/j.brainres.2019.146487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/31/2019] [Accepted: 09/29/2019] [Indexed: 10/25/2022]
Abstract
Long-term potentiation (LTP) is an important aspect of synaptic plasticity and is one of the main mechanisms involved in memory. Low-frequency electromagnetic fields (LF-EMFs) such as transcranial magnetic stimulation are emerging neuromodulation tools for the regulation of LTP. However, whether LF-EMFs have different effects on different types of LTP has not yet been verified. Herein, we studied the regulatory effects of 15 Hz/2 mT sinusoidal magnetic field as pre-magnetic stimulation on several types of LTP, which were induced by theta-burst(TBS) or high-frequency stimulation (HFS) or some combination of them, and applied N-methyl-D-aspartate receptor(NMDAR) antagonists to observe the relationship between the regulation of LTP by LF-EMFs and NMDAR in the Schaffer collateral pathway of rat brain slices in vitro. The results presented in this paper are the performance of TBS and HFS was not exactly the same and the recovery speed of TBS-LTP was faster than HFS-LTP after receiving the regulation of LF-EMFs; moreover, the LTP level was affected by the order of combination and the effect of pre-magnetic stimulation could maintain the entire process of the combined induction experiment, while NMDAR antagonists could not completely offset the influence of LF-EMFs. The memory patterns are diverse, and this study has shown LF-EMFs can regulate LTP such as TBS-LTP and HFS-LTP and can continuously affect multiple LTP induction processes. However, different memory processes may have different performance in the face of LF-EMFs regulation. In terms of the mechanism of LF-EMFs-induced LTP regulation, NMDARs may be involved in the process of LF-EMF regulation of LTP, but are not the only factor.
Collapse
Affiliation(s)
- Yu Zheng
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China.
| | - Xiao-Xu Ma
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China
| | - Lei Dong
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Wei Ma
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China
| | - Jian-Hao Cheng
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China
| |
Collapse
|
26
|
Brandwein NJ, Nguyen PV. A requirement for epigenetic modifications during noradrenergic stabilization of heterosynaptic LTP in the hippocampus. Neurobiol Learn Mem 2019; 161:72-82. [PMID: 30930287 DOI: 10.1016/j.nlm.2019.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/31/2022]
Abstract
Beta-adrenergic receptor (b-AR) activation by noradrenaline (NA) enhances memory formation and long-term potentiation (LTP), a form of synaptic plasticity characterized by an activity-dependent increase in synaptic strength. LTP is believed to be a cellular mechanism for contextual learning and memory. In the mammalian hippocampus, LTP can be observed at multiple synaptic pathways after strong stimulation of a single synaptic pathway. This heterosynaptic LTP is believed to involve synaptic tagging of active synapses and capture of plasticity-related proteins that enable heterosynaptic transfer of persistent potentiation. These processes may permit distinct neural pathways to associate information transmitted by separate, but convergent, synaptic inputs. We had previously shown that transcription and epigenetic modifications were necessary for stabilization of homosynaptic LTP. However, it is unclear whether transfer of LTP to a second, heterosynaptic pathway involves b-ARs signalling to the nucleus. Using electrophysiologic recordings in area CA1 of murine hippocampal slices, we show here that pharmacologically inhibiting b-AR activation, transcription, DNA methyltransferase or histone acetyltransferase activation, prevents stabilization of heterosynaptic LTP. Our data suggest that noradrenergic stabilization of heterosynaptic ("tagged") LTP requires not only transcription, but specifically, DNA methylation and histone acetylation. NA promotes stable heterosynaptic plasticity through engagement of nuclear processes that may contribute to prompt consolidation of short-term memories into resilient long-term memories under conditions when the brain's noradrenergic system is recruited.
Collapse
Affiliation(s)
- N J Brandwein
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada
| | - P V Nguyen
- Department of Physiology, University of Alberta School of Medicine, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
27
|
Park P, Kang H, Sanderson TM, Bortolotto ZA, Georgiou J, Zhuo M, Kaang BK, Collingridge GL. On the Role of Calcium-Permeable AMPARs in Long-Term Potentiation and Synaptic Tagging in the Rodent Hippocampus. Front Synaptic Neurosci 2019; 11:4. [PMID: 30923499 PMCID: PMC6426746 DOI: 10.3389/fnsyn.2019.00004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/01/2019] [Indexed: 12/21/2022] Open
Abstract
Classically, long-term potentiation (LTP) at hippocampal CA1 synapses is triggered by the synaptic activation of NMDA receptors (NMDARs). More recently, it has been shown that calcium-permeable (CP)-AMPARs can also trigger synaptic plasticity at these synapses. Specifically, their activation is required for the PKA and protein synthesis dependent component of LTP that is typically induced by delivery of spaced trains of high frequency stimulation. Here we present new data that build upon these ideas, including the requirement for low frequency synaptic activation and NMDAR dependence. We also show that a spaced theta burst stimulation (sTBS) protocol induces a heterosynaptic potentiation of baseline responses via activation of CP-AMPARs. Finally, we present data that implicate CP-AMPARs in synaptic tagging and capture, a fundamental process that is associated with the protein synthesis-dependent component of LTP. We have studied how a sTBS can augment the level of LTP generated by a weak TBS (wTBS), delivered 30 min later to an independent input. We show that inhibition of CP-AMPARs during the sTBS eliminates, and that inhibition of CP-AMPARs during the wTBS reduces, this facilitation of LTP. These data suggest that CP-AMPARs are crucial for the protein synthesis-dependent component of LTP and its heterosynaptic nature.
Collapse
Affiliation(s)
- Pojeong Park
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Heather Kang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Thomas M Sanderson
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Zuner A Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Min Zhuo
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Graham L Collingridge
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
28
|
Long-term population spike-timing-dependent plasticity promotes synaptic tagging but not cross-tagging in rat hippocampal area CA1. Proc Natl Acad Sci U S A 2019; 116:5737-5746. [PMID: 30819889 DOI: 10.1073/pnas.1817643116] [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] [Indexed: 12/18/2022] Open
Abstract
In spike-timing-dependent plasticity (STDP), the direction and degree of synaptic modification are determined by the coherence of pre- and postsynaptic activities within a neuron. However, in the adult rat hippocampus, it remains unclear whether STDP-like mechanisms in a neuronal population induce synaptic potentiation of a long duration. Thus, we asked whether the magnitude and maintenance of synaptic plasticity in a population of CA1 neurons differ as a function of the temporal order and interval between pre- and postsynaptic activities. Modulation of the relative timing of Schaffer collateral fibers (presynaptic component) and CA1 axons (postsynaptic component) stimulations resulted in an asymmetric population STDP (pSTDP). The resulting potentiation in response to 20 pairings at 1 Hz was largest in magnitude and most persistent (4 h) when presynaptic activity coincided with or preceded postsynaptic activity. Interestingly, when postsynaptic activation preceded presynaptic stimulation by 20 ms, an immediate increase in field excitatory postsynaptic potentials was observed, but it eventually transformed into a synaptic depression. Furthermore, pSTDP engaged in selective forms of late-associative activity: It facilitated the maintenance of tetanization-induced early long-term potentiation (LTP) in neighboring synapses but not early long-term depression, reflecting possible mechanistic differences with classical tetanization-induced LTP. The data demonstrate that a pairing of pre- and postsynaptic activities in a neuronal population can greatly reduce the required number of synaptic plasticity-evoking events and induce a potentiation of a degree and duration similar to that with repeated tetanization. Thus, pSTDP determines synaptic efficacy in the hippocampal CA3-CA1 circuit and could bias the CA1 neuronal population toward potentiation in future events.
Collapse
|
29
|
Baltaci SB, Mogulkoc R, Baltaci AK. Molecular Mechanisms of Early and Late LTP. Neurochem Res 2019; 44:281-296. [PMID: 30523578 DOI: 10.1007/s11064-018-2695-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/31/2018] [Accepted: 12/04/2018] [Indexed: 12/01/2022]
Abstract
LTP is the most intensively studied cellular model of the memory and generally divided at least two distinct phases as early and late. E-LTP requires activation of CaMKII that initiates biochemical events and trafficking of proteins, which eventually potentiate synaptic transmission, and is independent of de novo protein synthesis. In contrast, L-LTP requires gene expression and local protein synthesis regulated via TrkB receptor- and functional prions CPEB2-3-mediated translation. Maintenance of LTP for longer periods depends on constitutively active PKMζ. Throughout this review, current knowledge about early and late phases of LTP will be reviewed.
Collapse
Affiliation(s)
- Saltuk Bugra Baltaci
- Faculty of Medicine, Department of Physiology, Selcuk University, 42031, Konya, Turkey
| | - Rasim Mogulkoc
- Faculty of Medicine, Department of Physiology, Selcuk University, 42031, Konya, Turkey
| | | |
Collapse
|
30
|
Park P, Kang H, Sanderson TM, Bortolotto ZA, Georgiou J, Zhuo M, Kaang BK, Collingridge GL. The Role of Calcium-Permeable AMPARs in Long-Term Potentiation at Principal Neurons in the Rodent Hippocampus. Front Synaptic Neurosci 2018; 10:42. [PMID: 30524263 PMCID: PMC6262052 DOI: 10.3389/fnsyn.2018.00042] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/05/2018] [Indexed: 11/25/2022] Open
Abstract
Long-term potentiation (LTP) at hippocampal CA1 synapses is classically triggered by the synaptic activation of NMDA receptors (NMDARs). More recently, it has been shown that calcium-permeable (CP) AMPA receptors (AMPARs) can also trigger synaptic plasticity at these synapses. Here, we review this literature with a focus on recent evidence that CP-AMPARs are critical for the induction of the protein kinase A (PKA)- and protein synthesis-dependent component of LTP.
Collapse
Affiliation(s)
- Pojeong Park
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Heather Kang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Thomas M Sanderson
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Zuner A Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Min Zhuo
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bong-Kiun Kaang
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Graham L Collingridge
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
31
|
Long noncoding RNA GM12371 acts as a transcriptional regulator of synapse function. Proc Natl Acad Sci U S A 2018; 115:E10197-E10205. [PMID: 30297415 PMCID: PMC6205475 DOI: 10.1073/pnas.1722587115] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuronal functions of long noncoding RNAs (lncRNAs) are poorly understood. Here we describe identification and function of lncRNA GM12371 in regulating synaptic transmission, synapse density, and dendritic arborization in primary hippocampal neurons. GM12371 expression is regulated by cAMP signaling and is critical for the activity regulated synaptic transmission. Importantly, GM12371 is associated with transcriptionally active chromatin and regulates expression of several genes involved in neuronal growth and development. Taken together, these results suggest that GM12371 acts as a transcriptional regulator of synapse function. Despite the growing evidence suggesting that long noncoding RNAs (lncRNAs) are critical regulators of several biological processes, their functions in the nervous system remain elusive. We have identified an lncRNA, GM12371, in hippocampal neurons that is enriched in the nucleus and necessary for synaptic communication, synapse density, synapse morphology, and dendritic tree complexity. Mechanistically, GM12371 regulates the expression of several genes involved in neuronal development and differentiation, as well as expression of specific lncRNAs and their cognate mRNA targets. Furthermore, we find that cAMP-PKA signaling up-regulates the expression of GM12371 and that its expression is essential for the activity-dependent changes in synaptic transmission in hippocampal neurons. Taken together, our data establish a key role for GM12371 in regulating synapse function.
Collapse
|
32
|
Yang H, Zhu H, Ibrahim JG. MILFM: Multiple index latent factor model based on high-dimensional features. Biometrics 2018; 74:834-844. [PMID: 29665616 PMCID: PMC6158073 DOI: 10.1111/biom.12866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/01/2018] [Accepted: 11/01/2017] [Indexed: 11/30/2022]
Abstract
The aim of this article is to develop a multiple-index latent factor modeling (MILFM) framework to build an accurate prediction model for clinical outcomes based on a massive number of features. We develop a three-stage estimation procedure to build the prediction model. MILFM uses an independent screening method to select a set of informative features, which may have a complex nonlinear relationship with the outcome variables. Moreover, we develop a latent factor model to project all informative predictors onto a small number of local subspaces, which lead to a few key features that capture reliable and informative covariate information. Finally, we fit the regularized empirical estimate to those key features in order to accurately predict clinical outcomes. We systematically investigate the theoretical properties of MILFM, such as risk bounds and selection consistency. Our simulation results and real data analysis show that MILFM outperforms many state-of-the-art methods in terms of prediction accuracy.
Collapse
Affiliation(s)
- Hojin Yang
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina 27599-7420, U.S.A
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina 27599-7420, U.S.A
| | - Joseph G Ibrahim
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina 27599-7420, U.S.A
| |
Collapse
|
33
|
Calmodulin shuttling mediates cytonuclear signaling to trigger experience-dependent transcription and memory. Nat Commun 2018; 9:2451. [PMID: 29934532 PMCID: PMC6015085 DOI: 10.1038/s41467-018-04705-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 05/14/2018] [Indexed: 12/18/2022] Open
Abstract
Learning and memory depend on neuronal plasticity originating at the synapse and requiring nuclear gene expression to persist. However, how synapse-to-nucleus communication supports long-term plasticity and behavior has remained elusive. Among cytonuclear signaling proteins, γCaMKII stands out in its ability to rapidly shuttle Ca2+/CaM to the nucleus and thus activate CREB-dependent transcription. Here we show that elimination of γCaMKII prevents activity-dependent expression of key genes (BDNF, c-Fos, Arc), inhibits persistent synaptic strengthening, and impairs spatial memory in vivo. Deletion of γCaMKII in adult excitatory neurons exerts similar effects. A point mutation in γCaMKII, previously uncovered in a case of intellectual disability, selectively disrupts CaM sequestration and CaM shuttling. Remarkably, this mutation is sufficient to disrupt gene expression and spatial learning in vivo. Thus, this specific form of cytonuclear signaling plays a key role in learning and memory and contributes to neuropsychiatric disease. Activity-dependent gene expression is thought to involve translocation of Ca2+/calmodulin (CaM) to the nucleus. Here, the authors examine a translocation-deficient mutant of γCaMKII, a Ca2+/CaM shuttle protein, to show that translocation of Ca2+/CaM is required for memory and synaptic plasticity.
Collapse
|
34
|
Saavedra A, Ballesteros JJ, Tyebji S, Martínez-Torres S, Blázquez G, López-Hidalgo R, Azkona G, Alberch J, Martín ED, Pérez-Navarro E. Proteolytic Degradation of Hippocampal STEP 61 in LTP and Learning. Mol Neurobiol 2018; 56:1475-1487. [PMID: 29948948 DOI: 10.1007/s12035-018-1170-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/31/2018] [Indexed: 10/14/2022]
Abstract
Striatal-enriched protein tyrosine phosphatase (STEP) modulates key signaling molecules involved in synaptic plasticity and neuronal function. It is postulated that STEP opposes the development of long-term potentiation (LTP) and that it exerts a restraint on long-term memory (LTM). Here, we examined whether STEP61 levels are regulated during hippocampal LTP and after training in hippocampal-dependent tasks. We found that after inducing LTP by high frequency stimulation or theta-burst stimulation STEP61 levels were significantly reduced, with a concomitant increase of STEP33 levels, a product of calpain cleavage. Importantly, inhibition of STEP with TC-2153 improved LTP in hippocampal slices. Moreover, we observed that after training in the passive avoidance and the T-maze spontaneous alternation task, hippocampal STEP61 levels were significantly reduced, but STEP33 levels were unchanged. Yet, hippocampal BDNF content and TrkB levels were increased in trained mice, and it is known that BDNF promotes STEP degradation through the proteasome. Accordingly, hippocampal pTrkBTyr816, pPLCγTyr783, and protein ubiquitination levels were increased in T-SAT trained mice. Remarkably, injection of the TrkB antagonist ANA-12 (2 mg/Kg, but not 0.5 mg/Kg) elicited LTM deficits and promoted STEP61 accumulation in the hippocampus. Also, STEP knockout mice outperformed wild-type animals in an age- and test-dependent manner. Summarizing, STEP61 undergoes proteolytic degradation in conditions leading to synaptic strengthening and memory formation, thus highlighting its role as a molecular constrain, which is removed to enable the activation of pathways important for plasticity processes.
Collapse
Affiliation(s)
- Ana Saavedra
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús J Ballesteros
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shiraz Tyebji
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - Sara Martínez-Torres
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Gloria Blázquez
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rosa López-Hidalgo
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Grupo de Patología Celular y Molecular del Alcohol, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Garikoitz Azkona
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eduardo D Martín
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Instituto Cajal, CSIC, Madrid, Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
35
|
Sung JY, Bae JH, Lee JH, Kim YN, Kim DK. The Melatonin Signaling Pathway in a Long-Term Memory In Vitro Study. Molecules 2018; 23:molecules23040737. [PMID: 29570621 PMCID: PMC6017053 DOI: 10.3390/molecules23040737] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/15/2018] [Accepted: 03/20/2018] [Indexed: 12/20/2022] Open
Abstract
The activation of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) via phosphorylation in the hippocampus is an important signaling mechanism for enhancing memory processing. Although melatonin is known to increase CREB expression in various animal models, the signaling mechanism between melatonin and CREB has been unknown in vitro. Thus, we confirmed the signaling pathway between the melatonin receptor 1 (MT1) and CREB using melatonin in HT-22 cells. Melatonin increased MT1 and gradually induced signals associated with long-term memory processing through phosphorylation of Raf, ERK, p90RSK, CREB, and BDNF expression. We also confirmed that the calcium, JNK, and AKT pathways were not involved in this signaling pathway by melatonin in HT-22 cells. Furthermore, we investigated whether melatonin regulated the expressions of CREB-BDNF associated with long-term memory processing in aged HT-22 cells. In conclusion, melatonin mediated the MT1-ERK-p90RSK-CREB-BDNF signaling pathway in the in vitro long-term memory processing model and increased the levels of p-CREB and BDNF expression in melatonin-treated cells compared to untreated HT-22 cells in the cellular aged state. Therefore, this paper suggests that melatonin induces CREB signaling pathways associated with long-term memory processing in vitro.
Collapse
Affiliation(s)
- Jin-Young Sung
- Department of Medical Genetics, Hanvit Institutute for Medical Genetics, School of Medicine, Keimyung University, Daegu 42601, Korea.
| | - Ji-Hyun Bae
- Department of Medical Genetics, Hanvit Institutute for Medical Genetics, School of Medicine, Keimyung University, Daegu 42601, Korea.
| | - Jong-Ha Lee
- Department of Biomedical Engineering, School of Medicine, Keimyung University, Daegu 42601, Korea.
| | - Yoon-Nyun Kim
- Dongsan Medical Center, Department of Internal Medicine, Keimyung University, Daegu 42931, Korea.
| | - Dae-Kwang Kim
- Department of Medical Genetics, Hanvit Institutute for Medical Genetics, School of Medicine, Keimyung University, Daegu 42601, Korea.
| |
Collapse
|
36
|
Pharmacological activation of protein kinase A improves memory loss and neuropathological changes in a mouse model of dementia of Alzheimer's type. Behav Pharmacol 2018; 28:187-198. [PMID: 28177982 DOI: 10.1097/fbp.0000000000000294] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The study investigates the therapeutic potential of the protein kinase A (PKA) activator forskolin in cognitive deficits of mice. Streptozotocin (STZ) [3 mg/kg, intracerebroventricularly (i.c.v.)] was used to induce memory deficits in mice, whereas aged mice served as natural model of dementia. Forskolin (2.5, 5, and 10 mg/kg/day, oral) treatment was administered to i.c.v. STZ-treated and aged mice for 14 days. The Morris Water Maze test was used to evaluate learning and memory. Estimation of brain acetylcholinesterase (AChE) activity, brain glutathione, thiobarbituric acid-reactive species, brain myeloperoxidase levels, and histopathological studies were also performed. Both STZ i.c.v. and aging resulted in a marked decline in Morris Water Maze performance, reflecting impairment of learning and memory. STZ i.c.v.-treated mice and aged mice showed a marked accentuation of AChE activity, thiobarbituric acid-reactive species and myeloperoxidase levels along with a decrease in the glutathione level. Further, the stained micrographs of STZ-treated mice and aged mice indicated pathological changes, severe neutrophilic infiltration, and amyloid deposition. Forskolin treatment significantly attenuated STZ-induced and age-related memory deficits, and biochemical and histopathological alterations. The findings indicate that the PKA activator forskolin probably alleviated memory deficits by virtue of its anticholinesterase, antiamyloid, antioxidative, and anti-inflammatory effects. It is concluded that PKA could be explored as a potential therapeutic target in dementia.
Collapse
|
37
|
Campbell SL, van Groen T, Kadish I, Smoot LHM, Bolger GB. Altered phosphorylation, electrophysiology, and behavior on attenuation of PDE4B action in hippocampus. BMC Neurosci 2017; 18:77. [PMID: 29197324 PMCID: PMC5712142 DOI: 10.1186/s12868-017-0396-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 11/28/2017] [Indexed: 01/19/2023] Open
Abstract
Background PDE4 cyclic nucleotide phosphodiesterases regulate 3′, 5′ cAMP abundance in the CNS and thereby regulate PKA activity and phosphorylation of CREB, which has been implicated in learning and memory, depression and other functions. The PDE4 isoform PDE4B1 also interacts with the DISC1 protein, implicated in neural development and behavioral disorders. The cellular functions of PDE4B1 have been investigated extensively, but its function(s) in the intact organism remained unexplored. Results To specifically disrupt PDE4B1, we developed mice that express a PDE4B1-D564A transgene in the hippocampus and forebrain. The transgenic mice showed enhanced phosphorylation of CREB and ERK1/2 in hippocampus. Hippocampal neurogenesis was increased in the transgenic mice. Hippocampal electrophysiological studies showed increased baseline synaptic transmission and enhanced LTP in male transgenic mice. Behaviorally, male transgenic mice showed increased activity in prolonged open field testing, but neither male nor female transgenic mice showed detectable anxiety-like behavior or antidepressant effects in the elevated plus-maze, tail-suspension or forced-swim tests. Neither sex showed any significant differences in associative fear conditioning or showed any demonstrable abnormalities in pre-pulse inhibition. Conclusions These data support the use of an isoform-selective approach to the study of PDE4B1 function in the CNS and suggest a probable role of PDE4B1 in synaptic plasticity and behavior. They also provide additional rationale and a refined approach to the development of small-molecule PDE4B1-selective inhibitors, which have potential functions in disorders of cognition, memory, mood and affect.
Collapse
Affiliation(s)
- Susan L Campbell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA
| | - Thomas van Groen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Inga Kadish
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Lisa High Mitchell Smoot
- Department of Medicine, University of Alabama at Birmingham, NP 2501, 1720 2nd Ave S, Birmingham, AL, 35294-3300, USA
| | - Graeme B Bolger
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA. .,Department of Medicine, University of Alabama at Birmingham, NP 2501, 1720 2nd Ave S, Birmingham, AL, 35294-3300, USA. .,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA.
| |
Collapse
|
38
|
Schulze L, Feffer K, Lozano C, Giacobbe P, Daskalakis ZJ, Blumberger DM, Downar J. Number of pulses or number of sessions? An open-label study of trajectories of improvement for once-vs. twice-daily dorsomedial prefrontal rTMS in major depression. Brain Stimul 2017; 11:327-336. [PMID: 29153439 DOI: 10.1016/j.brs.2017.11.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/03/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) shows efficacy in the treatment of major depressive episodes (MDEs), but can require ≥4-6 weeks for maximal effect. Recent studies suggest that multiple daily sessions of rTMS can accelerate response without reducing therapeutic efficacy. However, it is unresolved whether therapeutic effects track cumulative number of pulses, or cumulative number of sessions. OBJECTIVE This open-label study reviewed clinical outcomes over a 20-30 session course of high-frequency bilateral dorsomedial prefrontal cortex (DMPFC)-rTMS among patients receiving 6000 pulses/day delivered either in twice-daily sessions 80 min apart (at 20 Hz) or single, longer, once-daily sessions (at 10 Hz). METHODS A retrospective chart review identified 130 MDD patients who underwent 20-30 daily sessions of bilateral DMPFC-rTMS (Once-daily, n = 65; Twice-daily, n = 65) at a single Canadian clinic. RESULTS Mixed-effects modeling revealed significantly faster improvement (group-by-time interaction) for twice-daily versus once-daily DMPFC-rTMS. Across both groups, the pace of improvement showed a consistent relationship with number of cumulative sessions, but not with cumulative number of pulses. Although the twice-daily group completed treatment in half as many days, final clinical outcomes did not differ significantly between groups on dichotomous measures (response/remission rates: once-daily, 35.4%/33.8%; twice-daily, 41.5%/35.4%), or continuous measures, or on overall response distribution. CONCLUSIONS Twice-daily rTMS appears feasible, tolerable, and capable of achieving comparable results to once-daily rTMS, while also reducing course length approximately twofold. Therapeutic gains tracked the cumulative number of sessions, not pulses. Future randomized studies comparing once-daily to multiple-daily rTMS sessions, while controlling for number of pulses, may be warranted.
Collapse
Affiliation(s)
- Laura Schulze
- MRI-Guided rTMS Clinic, Department of Psychiatry, University Health Network, Canada; Institute of Medical Science, University of Toronto, Canada
| | - Kfir Feffer
- MRI-Guided rTMS Clinic, Department of Psychiatry, University Health Network, Canada; Shalvata Mental Health Center, Hod-Hasharon, Israel; Department of Psychiatry, University of Toronto, Canada
| | | | - Peter Giacobbe
- MRI-Guided rTMS Clinic, Department of Psychiatry, University Health Network, Canada; Institute of Medical Science, University of Toronto, Canada; Department of Psychiatry, University of Toronto, Canada
| | - Zafiris J Daskalakis
- Institute of Medical Science, University of Toronto, Canada; Department of Psychiatry, University of Toronto, Canada; Campbell Family Mental Health Research Institute and Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Canada
| | - Daniel M Blumberger
- Institute of Medical Science, University of Toronto, Canada; Department of Psychiatry, University of Toronto, Canada; Campbell Family Mental Health Research Institute and Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Canada
| | - Jonathan Downar
- MRI-Guided rTMS Clinic, Department of Psychiatry, University Health Network, Canada; Institute of Medical Science, University of Toronto, Canada; Department of Psychiatry, University of Toronto, Canada; Krembil Research Institute, University Health Network, Canada.
| |
Collapse
|
39
|
Rossetti T, Banerjee S, Kim C, Leubner M, Lamar C, Gupta P, Lee B, Neve R, Lisman J. Memory Erasure Experiments Indicate a Critical Role of CaMKII in Memory Storage. Neuron 2017; 96:207-216.e2. [PMID: 28957669 DOI: 10.1016/j.neuron.2017.09.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/24/2017] [Accepted: 09/11/2017] [Indexed: 12/29/2022]
Abstract
The abundant synaptic protein CaMKII is necessary for long-term potentiation (LTP) and memory. However, whether CaMKII is required only during initial processes or whether it also mediates memory storage remains unclear. The most direct test of a storage role is the erasure test. In this test, a putative memory molecule is inhibited after learning. The key prediction is that this should produce persistent memory erasure even after the inhibitory agent is removed. We conducted this test using transient viral (HSV) expression of dominant-negative CaMKII-alpha (K42M) in the hippocampus. This produced persistent erasure of conditioned place avoidance. As an additional test, we found that expression of activated CaMKII (T286D/T305A/T306A) impaired place avoidance, a result not expected if a process other than CaMKII stores memory. Our behavioral results, taken together with prior experiments on LTP, strongly support a critical role of CaMKII in LTP maintenance and memory storage.
Collapse
Affiliation(s)
- Tom Rossetti
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Somdeb Banerjee
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Chris Kim
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Megan Leubner
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Casey Lamar
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Pooja Gupta
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Bomsol Lee
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Rachael Neve
- Gene Delivery Technology Core, Department of Neurology, MGH, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - John Lisman
- Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02453, USA.
| |
Collapse
|
40
|
Akiba C, Nakajima M, Miyajima M, Ogino I, Miura M, Inoue R, Nakamura E, Kanai F, Tada N, Kunichika M, Yoshida M, Nishimura K, Kondo A, Sugano H, Arai H. Leucine-rich α2-glycoprotein overexpression in the brain contributes to memory impairment. Neurobiol Aging 2017; 60:11-19. [PMID: 28917663 DOI: 10.1016/j.neurobiolaging.2017.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/28/2017] [Accepted: 08/15/2017] [Indexed: 12/19/2022]
Abstract
We previously reported increase in leucine-rich α2-glycoprotein (LRG) concentration in cerebrospinal fluid is associated with cognitive decline in humans. To investigate relationship between LRG expression in the brain and memory impairment, we analyzed transgenic mice overexpressing LRG in the brain (LRG-Tg) focusing on hippocampus. Immunostaining and Western blotting revealed age-related increase in LRG expression in hippocampal neurons in 8-, 24-, and 48-week-old controls and LRG-Tg. Y-maze and Morris water maze tests indicated retained spatial memory in 8- and 24-week-old LRG-Tg, while deteriorated in 48-week-old LRG-Tg compared with age-matched controls. Field excitatory postsynaptic potentials declined with age in LRG-Tg compared with controls at 8, 24, and 48 weeks. Paired-pulse ratio decreased with age in LRG-Tg, while increased in controls. As a result, long-term potentiation was retained in 8- and 24-week-old LRG-Tg, whereas diminished in 48-week-old LRG-Tg compared with age-matched controls. Electron microscopy observations revealed fewer synaptic vesicles and junctions in LRG-Tg compared with age-matched controls, which became significant with age. Hippocampal LRG overexpression contributes to synaptic dysfunction, which leads to memory impairment with advance of age.
Collapse
Affiliation(s)
- Chihiro Akiba
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Madoka Nakajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ikuko Ogino
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masami Miura
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Ritsuko Inoue
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Eri Nakamura
- Laboratory of Disease Model Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumio Kanai
- Laboratory of Disease Model Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norihiro Tada
- Laboratory of Disease Model Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miyuki Kunichika
- Laboratory of Morpheme Analysis Imaging Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mitsutaka Yoshida
- Laboratory of Morpheme Analysis Imaging Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kinya Nishimura
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan; Department of Anesthesiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hidenori Sugano
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
41
|
Jȩdrzejewska-Szmek J, Luczak V, Abel T, Blackwell KT. β-adrenergic signaling broadly contributes to LTP induction. PLoS Comput Biol 2017; 13:e1005657. [PMID: 28742159 PMCID: PMC5546712 DOI: 10.1371/journal.pcbi.1005657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 08/07/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022] Open
Abstract
Long-lasting forms of long-term potentiation (LTP) represent one of the major cellular mechanisms underlying learning and memory. One of the fundamental questions in the field of LTP is why different molecules are critical for long-lasting forms of LTP induced by diverse experimental protocols. Further complexity stems from spatial aspects of signaling networks, such that some molecules function in the dendrite and some are critical in the spine. We investigated whether the diverse experimental evidence can be unified by creating a spatial, mechanistic model of multiple signaling pathways in hippocampal CA1 neurons. Our results show that the combination of activity of several key kinases can predict the occurrence of long-lasting forms of LTP for multiple experimental protocols. Specifically Ca2+/calmodulin activated kinase II, protein kinase A and exchange protein activated by cAMP (Epac) together predict the occurrence of LTP in response to strong stimulation (multiple trains of 100 Hz) or weak stimulation augmented by isoproterenol. Furthermore, our analysis suggests that activation of the β-adrenergic receptor either via canonical (Gs-coupled) or non-canonical (Gi-coupled) pathways underpins most forms of long-lasting LTP. Simulations make the experimentally testable prediction that a complete antagonist of the β-adrenergic receptor will likely block long-lasting LTP in response to strong stimulation. Collectively these results suggest that converging molecular mechanisms allow CA1 neurons to flexibly utilize signaling mechanisms best tuned to temporal pattern of synaptic input to achieve long-lasting LTP and memory storage. Long-term potentiation of the strength of synaptic connections is a mechanism of learning and memory storage. One of the most confusing aspects of hippocampal synaptic potentiation is that numerous experiments have revealed the requirement for a plethora of signaling molecules. Furthermore the degree to which molecules activated by the stress response modify hippocampal synaptic potentiation and memory is still unclear. We used a computational model to demonstrate that this molecular diversity can be explained by considering a combination of several key molecules. We also show that activation of β-adrenergic receptors by the stress response appears to be involved in most forms of synaptic potentiation, though in some cases unconventional mechanisms are utilized. This suggests that novel treatments for stress-related disorders may have more success if they target unconventional mechanisms activated by β-adrenergic receptors.
Collapse
Affiliation(s)
- Joanna Jȩdrzejewska-Szmek
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
| | - Vincent Luczak
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kim T Blackwell
- The Krasnow Institute for Advanced Studies, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
42
|
Cash RF, Dar A, Hui J, De Ruiter L, Baarbé J, Fettes P, Peters S, Fitzgerald PB, Downar J, Chen R. Influence of inter-train interval on the plastic effects of rTMS. Brain Stimul 2017; 10:630-636. [DOI: 10.1016/j.brs.2017.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/14/2017] [Accepted: 02/28/2017] [Indexed: 01/16/2023] Open
|
43
|
Memory-enhancing effects of GEBR-32a, a new PDE4D inhibitor holding promise for the treatment of Alzheimer's disease. Sci Rep 2017; 7:46320. [PMID: 28402318 PMCID: PMC5389348 DOI: 10.1038/srep46320] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/13/2017] [Indexed: 11/20/2022] Open
Abstract
Memory loss characterizes several neurodegenerative disorders, including Alzheimer’s disease (AD). Inhibition of type 4 phosphodiesterase (PDE4) and elevation of cyclic adenosine monophosphate (cAMP) has emerged as a promising therapeutic approach to treat cognitive deficits. However, PDE4 exists in several isoforms and pan inhibitors cannot be used in humans due to severe emesis. Here, we present GEBR-32a, a new PDE4D full inhibitor that has been characterized both in vitro and in vivo using biochemical, electrophysiological and behavioural analyses. GEBR-32a efficiently enhances cAMP in neuronal cultures and hippocampal slices. In vivo pharmacokinetic analysis shows that GEBR-32a is rapidly distributed within the central nervous system with a very favourable brain/blood ratio. Specific behavioural tests (object location and Y-maze continuous alternation tasks) demonstrate that this PDE4D inhibitor is able to enhance memory in AD transgenic mice and concomitantly rescues their hippocampal long-term potentiation deficit. Of great relevance, our preliminary toxicological analysis indicates that GEBR-32a is not cytotoxic and genotoxic, and does not seem to possess emetic-like side effects. In conclusion, GEBR-32a could represent a very promising cognitive-enhancing drug with a great potential for the treatment of Alzheimer’s disease.
Collapse
|
44
|
Altered Cortical Dynamics and Cognitive Function upon Haploinsufficiency of the Autism-Linked Excitatory Synaptic Suppressor MDGA2. Neuron 2016; 91:1052-1068. [DOI: 10.1016/j.neuron.2016.08.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/13/2016] [Accepted: 07/29/2016] [Indexed: 11/17/2022]
|
45
|
Li Y, Kulvicius T, Tetzlaff C. Induction and Consolidation of Calcium-Based Homo- and Heterosynaptic Potentiation and Depression. PLoS One 2016; 11:e0161679. [PMID: 27560350 PMCID: PMC4999190 DOI: 10.1371/journal.pone.0161679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 08/10/2016] [Indexed: 11/19/2022] Open
Abstract
The adaptive mechanisms of homo- and heterosynaptic plasticity play an important role in learning and memory. In order to maintain plasticity-induced changes for longer time scales (up to several days), they have to be consolidated by transferring them from a short-lasting early-phase to a long-lasting late-phase state. The underlying processes of this synaptic consolidation are already well-known for homosynaptic plasticity, however, it is not clear whether the same processes also enable the induction and consolidation of heterosynaptic plasticity. In this study, by extending a generic calcium-based plasticity model with the processes of synaptic consolidation, we show in simulations that indeed heterosynaptic plasticity can be induced and, furthermore, consolidated by the same underlying processes as for homosynaptic plasticity. Furthermore, we show that by local diffusion processes the heterosynaptic effect can be restricted to a few synapses neighboring the homosynaptically changed ones. Taken together, this generic model reproduces many experimental results of synaptic tagging and consolidation, provides several predictions for heterosynaptic induction and consolidation, and yields insights into the complex interactions between homo- and heterosynaptic plasticity over a broad variety of time (minutes to days) and spatial scales (several micrometers).
Collapse
Affiliation(s)
- Yinyun Li
- III. Institute of Physics – Biophysics, Georg-August-University, 37077 Göttingen, Germany
- Bernstein Center for Computational Neuroscience, Georg-August-University, 37077 Göttingen, Germany
- School of System Science, Beijing Normal University, 100875 Beijing, China
- * E-mail:
| | - Tomas Kulvicius
- III. Institute of Physics – Biophysics, Georg-August-University, 37077 Göttingen, Germany
- Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark
| | - Christian Tetzlaff
- Bernstein Center for Computational Neuroscience, Georg-August-University, 37077 Göttingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
- Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
46
|
Cortese GP, Burger C. Neuroinflammatory challenges compromise neuronal function in the aging brain: Postoperative cognitive delirium and Alzheimer's disease. Behav Brain Res 2016; 322:269-279. [PMID: 27544872 DOI: 10.1016/j.bbr.2016.08.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that targets memory and cognition, and is the most common form of dementia among the elderly. Although AD itself has been extensively studied, very little is known about early-stage preclinical events and/or mechanisms that may underlie AD pathogenesis. Since the majority of AD cases are sporadic in nature, advancing age remains the greatest known risk factor for AD. However, additional environmental and epigenetic factors are thought to accompany increasing age to play a significant role in the pathogenesis of AD. Postoperative cognitive delirium (POD) is a behavioral syndrome that primarily occurs in elderly patients following a surgical procedure or injury and is characterized by disruptions in cognition. Individuals that experience POD are at an increased risk for developing dementia and AD compared to normal aging individuals. One way in which cognitive function is affected in cases of POD is through activation of the inflammatory cascade following surgery or injury. There is compelling evidence that immune challenges (surgery and/or injury) associated with POD trigger the release of pro-inflammatory cytokines into both the periphery and central nervous system. Thus, it is possible that cognitive impairments following an inflammatory episode may lead to more severe forms of dementia and AD pathogenesis. Here we will discuss the inflammation associated with POD, and highlight the advantages of using POD as a model to study inflammation-evoked cognitive impairment. We will explore the possibility that advancing age and immune challenges may provide mechanistic evidence correlating early life POD with AD. We will review and propose neural mechanisms by which cognitive impairments occur in cases of POD, and discuss how POD may augment the onset of AD.
Collapse
Affiliation(s)
- Giuseppe P Cortese
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, 1300 University Ave, Room 73 Bardeen Madison, WI 53706, USA.
| | - Corinna Burger
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, 1300 University Ave, Room 73 Bardeen Madison, WI 53706, USA
| |
Collapse
|
47
|
Piracetam prevents memory deficit induced by postnatal propofol exposure in mice. Eur J Pharmacol 2016; 779:59-65. [DOI: 10.1016/j.ejphar.2016.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 02/22/2016] [Accepted: 03/04/2016] [Indexed: 11/18/2022]
|
48
|
Balakrishnan S, Niebert M, Richter DW. Rescue of Cyclic AMP Mediated Long Term Potentiation Impairment in the Hippocampus of Mecp2 Knockout (Mecp2(-/y) ) Mice by Rolipram. Front Cell Neurosci 2016; 10:15. [PMID: 26869885 PMCID: PMC4737891 DOI: 10.3389/fncel.2016.00015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/15/2016] [Indexed: 11/13/2022] Open
Abstract
Rett syndrome (RTT) patients experience learning difficulties and memory loss. Analogous deficits of hippocampal plasticity are reported in mouse models of RTT. To elucidate the underlying pathophysiology, we studied long term potentiation (LTP) at the CA3 to CA1 synapses in the hippocampus in acute brain slices from WT and Mecp2(-/y) mice, by either activating cAMP dependent pathway or using high frequency stimulation, by means of patch clamp. We have observed that, the NMDA channel current characteristics remain unchanged in the Mecp2(-/y) mice. The adenylyl cyclase (AC) agonist forskolin evoked a long lasting potentiation of evoked EPSCs in WT CA1 neurons, but only minimally enhanced the EPSCs in the Mecp2(-/y) mice. This weaker potentiation in Mecp2 (-/) (y) mice was ameliorated by application of phosphodiesterase 4 inhibitor rolipram. The hyperpolarization activated cyclic nucleotide gated channel current (I h) was potentiated to similar extent by forskolin in both phenotypes. Multiple tetanus induced cAMP-dependent plasticity was also impaired in the Mecp2 (-/) (y) mice, and was also partially rescued by rolipram. Western blot analysis of CA region of Mecp2 (-/) (y) mice hippocampus revealed more than twofold up-regulation of protein kinase A (PKA) regulatory subunits, while the expression of the catalytic subunit remained unchanged. We hypothesize that the overexpressed PKA regulatory subunits buffer cAMP and restrict the PKA mediated phosphorylation of target proteins necessary for LTP. Blocking the degradation of cAMP, thereby saturating the regulatory subunits alleviated this defect.
Collapse
Affiliation(s)
- Saju Balakrishnan
- Institute for Neuro and Sensory Physiology, University of Göttingen Göttingen, Germany
| | - Marcus Niebert
- Institute for Neuro and Sensory Physiology, University of Göttingen Göttingen, Germany
| | - Diethelm W Richter
- Institute for Neuro and Sensory Physiology, University of Göttingen Göttingen, Germany
| |
Collapse
|
49
|
Serotonin dependent masking of hippocampal sharp wave ripples. Neuropharmacology 2016; 101:188-203. [DOI: 10.1016/j.neuropharm.2015.09.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/04/2015] [Accepted: 09/21/2015] [Indexed: 11/21/2022]
|
50
|
Fitzpatrick SC, Luu BL, Butler JE, Taylor JL. More conditioning stimuli enhance synaptic plasticity in the human spinal cord. Clin Neurophysiol 2016; 127:724-731. [DOI: 10.1016/j.clinph.2015.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/06/2015] [Accepted: 03/23/2015] [Indexed: 10/23/2022]
|