1
|
Zhu Y, Mei Y, Baby N, Teo HY, Binte Hanafi Z, Mohd Salleh SN, Sajikumar S, Liu H. Tumor-mediated microbiota alteration impairs synaptic tagging/capture in the hippocampal CA1 area via IL-1β production. Commun Biol 2023; 6:685. [PMID: 37400621 DOI: 10.1038/s42003-023-05036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 06/12/2023] [Indexed: 07/05/2023] Open
Abstract
Cancer patients often experience impairments in cognitive function. However, the evidence for tumor-mediated neurological impairment and detailed mechanisms are still lacking. Gut microbiota has been demonstrated to be involved in the immune system homeostasis and brain functions. Here we find that hepatocellular carcinoma (HCC) growth alters the gut microbiota and impedes the cognitive functions. The synaptic tagging and capture (STC), an associative cellular mechanism for the formation of associative memory, is impaired in the tumor-bearing mice. STC expression is rescued after microbiota sterilization. Transplantation of microbiota from HCC tumor-bearing mice induces similar STC impairment in wide type mice. Mechanistic study reveals that HCC growth significantly elevates the serum and hippocampus IL-1β levels. IL-1β depletion in the HCC tumor-bearing mice restores the STC. Taken together, these results demonstrate that gut microbiota plays a crucial role in mediating the tumor-induced impairment of the cognitive function via upregulating IL-1β production.
Collapse
Affiliation(s)
- Ying Zhu
- Immunology Translational Research Programme, Department of Microbiology of Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Yu Mei
- Immunology Translational Research Programme, Department of Microbiology of Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Nimmi Baby
- Department of Physiology, National University of Singapore, Singapore, 117597, Singapore
| | - Huey Yee Teo
- Immunology Translational Research Programme, Department of Microbiology of Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Zuhairah Binte Hanafi
- Immunology Translational Research Programme, Department of Microbiology of Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Siti Nazihah Mohd Salleh
- Human Monoclonal Antibody Platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore, 117597, Singapore.
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore, 117456, Singapore.
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
| | - Haiyan Liu
- Immunology Translational Research Programme, Department of Microbiology of Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
2
|
Raghuraman R, Navakkode S, Sajikumar S. Alteration of hippocampal CA2 plasticity and social memory in adult rats impacted by juvenile stress. Hippocampus 2023; 33:745-758. [PMID: 36965045 PMCID: PMC10946601 DOI: 10.1002/hipo.23531] [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: 06/15/2022] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/27/2023]
Abstract
The hippocampal CA2 region has received greater attention in recent years due to its fundamental role in social memory and hippocampus-dependent memory processing. Unlike entorhinal cortical inputs, the Schaffer collateral inputs to CA2 do not support activity-dependent long-term potentiation (LTP), which serves as the basis for long-term memories. This LTP-resistant zone also expresses genes that restrict plasticity. With the aim of exploring social interaction and sociability in rats that were subjected to juvenile stress, we addressed questions about how the neural circuitry is altered and its effects on social behavior. Although there was induction of LTP in both Schaffer collateral and entorhinal cortical pathways in juvenile-stressed rats, LTP declined in both pathways after 2-3 h. Moreover, exogenous bath application of substance P, a neuropeptide that resulted in slow onset long-lasting potentiation in control animals while it failed to induce LTP in juvenile-stressed rats. Our study reveals that juvenile-stressed rats show behavioral and cellular abnormalities with a long-lasting impact in adulthood.
Collapse
Affiliation(s)
- Radha Raghuraman
- Department of PhysiologyNational University of SingaporeSingapore117593Singapore
- Life Sciences Institute Neurobiology ProgrammeCentre for Life Sciences, National University of SingaporeSingapore117456Singapore
- Present address:
Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew York10032USA
| | - Sheeja Navakkode
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore308232Singapore
| | - Sreedharan Sajikumar
- Department of PhysiologyNational University of SingaporeSingapore117593Singapore
- Life Sciences Institute Neurobiology ProgrammeCentre for Life Sciences, National University of SingaporeSingapore117456Singapore
- Healthy Longevity Translational Research ProgrammeYong Loo Lin School of Medicine, National University of SingaporeSingapore117456Singapore
| |
Collapse
|
3
|
Hydroxychloroquine lowers Alzheimer's disease and related dementias risk and rescues molecular phenotypes related to Alzheimer's disease. Mol Psychiatry 2023; 28:1312-1326. [PMID: 36577843 PMCID: PMC10005941 DOI: 10.1038/s41380-022-01912-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022]
Abstract
We recently nominated cytokine signaling through the Janus-kinase-signal transducer and activator of transcription (JAK/STAT) pathway as a potential AD drug target. As hydroxychloroquine (HCQ) has recently been shown to inactivate STAT3, we hypothesized that it may impact AD pathogenesis and risk. Among 109,124 rheumatoid arthritis patients from routine clinical care, HCQ initiation was associated with a lower risk of incident AD compared to methotrexate initiation across 4 alternative analyses schemes addressing specific types of biases including informative censoring, reverse causality, and outcome misclassification (hazard ratio [95% confidence interval] of 0.92 [0.83-1.00], 0.87 [0.81-0.93], 0.84 [0.76-0.93], and 0.87 [0.75-1.01]). We additionally show that HCQ exerts dose-dependent effects on late long-term potentiation (LTP) and rescues impaired hippocampal synaptic plasticity prior to significant accumulation of amyloid plaques and neurodegeneration in APP/PS1 mice. Additionally, HCQ treatment enhances microglial clearance of Aβ1-42, lowers neuroinflammation, and reduces tau phosphorylation in cell culture-based phenotypic assays. Finally, we show that HCQ inactivates STAT3 in microglia, neurons, and astrocytes suggesting a plausible mechanism associated with its observed effects on AD pathogenesis. HCQ, a relatively safe and inexpensive drug in current use may be a promising disease-modifying AD treatment. This hypothesis merits testing through adequately powered clinical trials in at-risk individuals during preclinical stages of disease progression.
Collapse
|
4
|
Chong YS, Wong LW, Gaunt J, Lee YJ, Goh CS, Morris RGM, Ch'ng TH, Sajikumar S. Distinct contributions of ventral CA1/amygdala co-activation to the induction and maintenance of synaptic plasticity. Cereb Cortex 2023; 33:676-690. [PMID: 35253866 DOI: 10.1093/cercor/bhac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 02/03/2023] Open
Abstract
The amygdala is known to modulate hippocampal synaptic plasticity. One role could be an immediate effect of basolateral amygdala (BLA) in priming synaptic plasticity in the hippocampus. Another role could be through associative synaptic co-operation and competition that triggers events involved in the maintenance of synaptic potentiation. We present evidence that the timing and activity level of BLA stimulation are important factors for the induction and maintenance of long-term potentiation (LTP) in ventral hippocampal area CA1. A 100 Hz BLA co-stimulation facilitated the induction of LTP, whereas 200 Hz co-stimulation attenuated induction. A 100 Hz BLA co-stimulation also caused enhanced persistence, sufficient to prevent synaptic competition. This maintenance effect is likely through translational mechanisms, as mRNA expression of primary response genes was unaffected, whereas protein level of plasticity-related products was increased. Further understanding of the neural mechanisms of amygdala modulation on hippocampus could provide insights into the mechanisms of emotional disorders.
Collapse
Affiliation(s)
- Yee Song Chong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Jessica Gaunt
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.,Interdisciplinary Graduate School, Nanyang Technological University, Singapore 637335, Singapore
| | - Cai Shan Goh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Richard G M Morris
- Laboratory for Cognitive Neuroscience, Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh EH8 9JZ, Scotland
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, SIngapore 117597, Singapore.,Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
5
|
Chatterjee S, Bahl E, Mukherjee U, Walsh EN, Shetty MS, Yan AL, Vanrobaeys Y, Lederman JD, Giese KP, Michaelson J, Abel T. Endoplasmic reticulum chaperone genes encode effectors of long-term memory. SCIENCE ADVANCES 2022; 8:eabm6063. [PMID: 35319980 PMCID: PMC8942353 DOI: 10.1126/sciadv.abm6063] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/28/2022] [Indexed: 06/10/2023]
Abstract
The mechanisms underlying memory loss associated with Alzheimer's disease and related dementias (ADRD) remain unclear, and no effective treatments exist. Fundamental studies have shown that a set of transcriptional regulatory proteins of the nuclear receptor 4a (Nr4a) family serve as molecular switches for long-term memory. Here, we show that Nr4a proteins regulate the transcription of genes encoding chaperones that localize to the endoplasmic reticulum (ER). These chaperones fold and traffic plasticity-related proteins to the cell surface during long-lasting forms of synaptic plasticity and memory. Dysregulation of Nr4a transcription factors and ER chaperones is linked to ADRD, and overexpressing Nr4a1 or the chaperone Hspa5 ameliorates long-term memory deficits in a tau-based mouse model of ADRD, pointing toward innovative therapeutic approaches for treating memory loss. Our findings establish a unique molecular concept underlying long-term memory and provide insights into the mechanistic basis of cognitive deficits in dementia.
Collapse
Affiliation(s)
- Snehajyoti Chatterjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Ethan Bahl
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Utsav Mukherjee
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Emily N. Walsh
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Mahesh Shivarama Shetty
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Amy L. Yan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Joseph D. Lederman
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - K. Peter Giese
- Department of Basic and Clinical Neuroscience, King’s College London, London, UK
| | - Jacob Michaelson
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA 52242, USA
- Department of Communication Sciences and Disorders, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
6
|
Navakkode S, Gaunt JR, Pavon MV, Bansal VA, Abraham RP, Chong YS, Ch'ng TH, Sajikumar S. Sex-specific accelerated decay in time/activity-dependent plasticity and associative memory in an animal model of Alzheimer's disease. Aging Cell 2021; 20:e13502. [PMID: 34796608 PMCID: PMC8672784 DOI: 10.1111/acel.13502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/02/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical studies have shown that female brains are more predisposed to neurodegenerative diseases such as Alzheimer's disease (AD), but the cellular and molecular mechanisms behind this disparity remain unknown. In several mouse models of AD, synaptic plasticity dysfunction is an early event and appears before significant accumulation of amyloid plaques and neuronal degeneration. However, it is unclear whether sexual dimorphism at the synaptic level contributes to the higher risk and prevalence of AD in females. Our studies on APP/PS1 (APPSwe/PS1dE9) mouse model show that AD impacts hippocampal long‐term plasticity in a sex‐specific manner. Long‐term potentiation (LTP) induced by strong tetanic stimulation (STET), theta burst stimulation (TBS) and population spike timing‐dependent plasticity (pSTDP) show a faster decay in AD females compared with age‐matched AD males. In addition, behavioural tagging (BT), a model of associative memory, is specifically impaired in AD females with a faster decay in memory compared with males. Together with the plasticity and behavioural data, we also observed an upregulation of neuroinflammatory markers, along with downregulation of transcripts that regulate cellular processes associated with synaptic plasticity and memory in females. Immunohistochemistry of AD brains confirms that female APP/PS1 mice carry a higher amyloid plaque burden and have enhanced microglial activation compared with male APP/PS1 mice. Their presence in the diseased mice also suggests a link between the impairment of LTP and the upregulation of the inflammatory response. Overall, our data show that synaptic plasticity and associative memory impairments are more prominent in females and this might account for the faster progression of AD in females.
Collapse
Affiliation(s)
- Sheeja Navakkode
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Department of Physiology National University of Singapore Singapore Singapore
| | - Jessica Ruth Gaunt
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Maria Vazquez Pavon
- Department of Physiology National University of Singapore Singapore Singapore
| | | | - Riya Prasad Abraham
- Department of Physiology National University of Singapore Singapore Singapore
| | - Yee Song Chong
- Department of Physiology National University of Singapore Singapore Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Science Nanyang Technological University Singapore Singapore
| | - Sreedharan Sajikumar
- Department of Physiology National University of Singapore Singapore Singapore
- Healthy Longevity Translational Research Programme Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Life Sciences Institute Neurobiology Programme National University of Singapore Singapore Singapore
| |
Collapse
|
7
|
Metaplastic Reinforcement of Long-Term Potentiation in Hippocampal Area CA2 by Cholinergic Receptor Activation. J Neurosci 2021; 41:9082-9098. [PMID: 34561235 DOI: 10.1523/jneurosci.2885-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 09/13/2021] [Accepted: 09/18/2021] [Indexed: 11/21/2022] Open
Abstract
Hippocampal CA2, an inconspicuously positioned area between the well-studied CA1 and CA3 subfields, has captured research interest in recent years because of its role in social memory formation. However, the role of cholinergic inputs to the CA2 area for the regulation of synaptic plasticity remains to be fully understood. We show that cholinergic receptor activation with the nonselective cholinergic agonist, carbachol (CCh), triggers a protein synthesis-dependent and NMDAR-independent long-term synaptic depression (CCh-LTD) at entorhinal cortical (EC)-CA2 and Schaffer collateral (SC)-CA2 synapses in the hippocampus of adult male Wistar rats. The activation of muscarinic acetylcholine receptors (mAChRs) is critical for the induction of CCh-LTD with the results suggesting an involvement of M3 and M1 mAChRs in the early facilitation of CCh-LTD, while nicotinic AChR activation plays a role in the late maintenance of CCh-LTD at CA2 synapses. Remarkably, we find that CCh priming lowers the threshold for the subsequent induction of persistent long-term potentiation (LTP) of synaptic transmission at EC-CA2 and the plasticity-resistant SC-CA2 pathways. The effects of such a cholinergic-dependent synaptic depression on subsequent LTP at EC-CA2 and SC-CA2 synapses have not been previously explored. Collectively, the results demonstrate that CA2 synaptic learning rules are regulated in a metaplastic manner, whereby modifications triggered by prior cholinergic stimulation can dictate the outcome of future plasticity events. Moreover, the reinforcement of LTP at EC inputs to CA2 following the priming stimulus coexists with concurrent sustained CCh-LTD at the SC-CA2 pathway and is dynamically scaled by modulation of SC-CA2 synaptic transmission.SIGNIFICANCE STATEMENT The release of the neuromodulator acetylcholine is critically involved in processes of hippocampus-dependent memory formation. Cholinergic afferents originating in the medial septum and diagonal bands of Broca terminating in the hippocampal area CA2 might play an important role in the modulation of area-specific synaptic plasticity. Our findings demonstrate that cholinergic receptor activation induces an LTD of synaptic transmission at entorhinal cortical- and Schaffer collateral-CA2 synapses. This cholinergic activation-mediated LTD displays a bidirectional metaplastic switch to LTP on a future timescale. This suggests that such bidirectional synaptic modifications triggered by the dynamic modulation of tonic cholinergic receptor activation may support the formation of CA2-dependent memories given the increased hippocampal cholinergic tone during active wakefulness observed in exploratory behavior.
Collapse
|
8
|
Wong L, Chong YS, Lin W, Kisiswa L, Sim E, Ibáñez CF, Sajikumar S. Age-related changes in hippocampal-dependent synaptic plasticity and memory mediated by p75 neurotrophin receptor. Aging Cell 2021; 20:e13305. [PMID: 33448137 PMCID: PMC7884039 DOI: 10.1111/acel.13305] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 11/25/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022] Open
Abstract
The plasticity mechanisms in the nervous system that are important for learning and memory are greatly impacted during aging. Notably, hippocampal-dependent long-term plasticity and its associative plasticity, such as synaptic tagging and capture (STC), show considerable age-related decline. The p75 neurotrophin receptor (p75NTR ) is a negative regulator of structural and functional plasticity in the brain and thus represents a potential candidate to mediate age-related alterations. However, the mechanisms by which p75NTR affects synaptic plasticity of aged neuronal networks and ultimately contribute to deficits in cognitive function have not been well characterized. Here, we report that mutant mice lacking the p75NTR were resistant to age-associated changes in long-term plasticity, associative plasticity, and associative memory. Our study shows that p75NTR is responsible for age-dependent disruption of hippocampal homeostatic plasticity by modulating several signaling pathways, including BDNF, MAPK, Arc, and RhoA-ROCK2-LIMK1-cofilin. p75NTR may thus represent an important therapeutic target for limiting the age-related memory and cognitive function deficits.
Collapse
Affiliation(s)
- Lik‐Wei Wong
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
- Healthy Longevity Translational Research ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
| | - Yee Song Chong
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
| | - Wei Lin
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
| | - Lilian Kisiswa
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
| | - Eunice Sim
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
| | - Carlos F. Ibáñez
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
- Department of NeuroscienceKarolinska InstituteStockholmSweden
| | - Sreedharan Sajikumar
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences Institute Neurobiology ProgrammeNational University of SingaporeSingapore CitySingapore
- Healthy Longevity Translational Research ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
| |
Collapse
|
9
|
Yi C, Goh KY, Wong L, Ramanujan A, Tanaka K, Sajikumar S, Ibáñez CF. Inactive variants of death receptor p75 NTR reduce Alzheimer's neuropathology by interfering with APP internalization. EMBO J 2021; 40:e104450. [PMID: 33258176 PMCID: PMC7809794 DOI: 10.15252/embj.2020104450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 01/04/2023] Open
Abstract
A prevalent model of Alzheimer's disease (AD) pathogenesis postulates the generation of neurotoxic fragments derived from the amyloid precursor protein (APP) after its internalization to endocytic compartments. The molecular pathways that regulate APP internalization and intracellular trafficking in neurons are incompletely understood. Here, we report that 5xFAD mice, an animal model of AD, expressing signaling-deficient variants of the p75 neurotrophin receptor (p75NTR ) show greater neuroprotection from AD neuropathology than animals lacking this receptor. p75NTR knock-in mice lacking the death domain or transmembrane Cys259 showed lower levels of Aβ species, amyloid plaque burden, gliosis, mitochondrial stress, and neurite dystrophy than global knock-outs. Strikingly, long-term synaptic plasticity and memory, which are completely disrupted in 5xFAD mice, were fully recovered in the knock-in mice. Mechanistically, we found that p75NTR interacts with APP at the plasma membrane and regulates its internalization and intracellular trafficking in hippocampal neurons. Inactive p75NTR variants internalized considerably slower than wild-type p75NTR and showed increased association with the recycling pathway, thereby reducing APP internalization and co-localization with BACE1, the critical protease for generation of neurotoxic APP fragments, favoring non-amyloidogenic APP cleavage. These results reveal a novel pathway that directly and specifically regulates APP internalization, amyloidogenic processing, and disease progression, and suggest that inhibitors targeting the p75NTR transmembrane domain may be an effective therapeutic strategy in AD.
Collapse
Affiliation(s)
- Chenju Yi
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
- Present address:
The Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
| | - Ket Yin Goh
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
| | - Lik‐Wei Wong
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
| | - Ajeena Ramanujan
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
| | - Kazuhiro Tanaka
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
| | - Sreedharan Sajikumar
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
| | - Carlos F. Ibáñez
- Department of PhysiologyNational University of SingaporeSingapore CitySingapore
- Life Sciences InstituteNational University of SingaporeSingapore CitySingapore
- Department of NeuroscienceKarolinska InstituteStockholmSweden
- Stellenbosch Institute for Advanced StudyWallenberg Research Centre at Stellenbosch UniversityStellenboschSouth Africa
| |
Collapse
|
10
|
Sharma M, Sajikumar S. G9a/GLP Complex Acts as a Bidirectional Switch to Regulate Metabotropic Glutamate Receptor-Dependent Plasticity in Hippocampal CA1 Pyramidal Neurons. Cereb Cortex 2020; 29:2932-2946. [PMID: 29982412 DOI: 10.1093/cercor/bhy161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/14/2018] [Accepted: 06/17/2018] [Indexed: 02/01/2023] Open
Abstract
Metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD) is conventionally considered to be solely dependent on local protein synthesis. Given the impact of epigenetics on memory, the intriguing question is whether epigenetic regulation influences mGluR-LTD as well. G9a/GLP histone lysine methyltransferase complex is crucial for brain development and goal-directed learning as well as for drug-addiction. In this study, we analyzed whether the epigenetic regulation by G9a/GLP complex affects mGluR-LTD in CA1 hippocampal pyramidal neurons of 5-7 weeks old male Wistar rats. In hippocampal slices with intact CA1 dendritic regions, inhibition of G9a/GLP activity abolished mGluR-LTD. The inhibition of this complex upregulated the expression of plasticity proteins like PKMζ, which mediated the prevention of mGluR-LTD expression by regulating the NSF-GluA2-mediated trafficking of AMPA receptors towards the postsynaptic site. G9a/GLP inhibition during the induction of mGluR-LTD also downregulated the protein levels of phosphorylated-GluA2 and Arc. Interestingly, G9a/GLP inhibition could not impede the mGluR-LTD when the cell-body was severed. Our study highlights the role of G9a/GLP complex in intact neuronal network as a bidirectional switch; when turned on, it facilitates the expression of mGluR-LTD, and when turned off, it promotes the expression of long-term potentiation.
Collapse
Affiliation(s)
- Mahima Sharma
- Department of Physiology, National University of Singapore, 2 Medical Drive, MD9, Singapore, Singapore.,Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, 2 Medical Drive, MD9, Singapore, Singapore.,Neurobiology/Aging Programme, Life Sciences Institute, Centre for Life Sciences, 28 Medical Drive, Singapore, Singapore
| |
Collapse
|
11
|
Dasgupta A, Lim YJ, Kumar K, Baby N, Pang KLK, Benoy A, Behnisch T, Sajikumar S. Group III metabotropic glutamate receptors gate long-term potentiation and synaptic tagging/capture in rat hippocampal area CA2. eLife 2020; 9:e55344. [PMID: 32310084 PMCID: PMC7170650 DOI: 10.7554/elife.55344] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) play an important role in synaptic plasticity and memory and are largely classified based on amino acid sequence homology and pharmacological properties. Among group III metabotropic glutamate receptors, mGluR7 and mGluR4 show high relative expression in the rat hippocampal area CA2. Group III metabotropic glutamate receptors are known to down-regulate cAMP-dependent signaling pathways via the activation of Gi/o proteins. Here, we provide evidence that inhibition of group III mGluRs by specific antagonists permits an NMDA receptor- and protein synthesis-dependent long-lasting synaptic potentiation in the apparently long-term potentiation (LTP)-resistant Schaffer collateral (SC)-CA2 synapses. Moreover, long-lasting potentiation of these synapses transforms a transient synaptic potentiation of the entorhinal cortical (EC)-CA2 synapses into a stable long-lasting LTP, in accordance with the synaptic tagging/capture hypothesis (STC). Furthermore, this study also sheds light on the role of ERK/MAPK protein signaling and the downregulation of STEP protein in the group III mGluR inhibition-mediated plasticity in the hippocampal CA2 region, identifying them as critical molecular players. Thus, the regulation of group III mGluRs provides a conducive environment for the SC-CA2 synapses to respond to events that could lead to activity-dependent synaptic plasticity.
Collapse
Affiliation(s)
- Ananya Dasgupta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Yu Jia Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Krishna Kumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Nimmi Baby
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Ka Lam Karen Pang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Amrita Benoy
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Life Sciences Institute Neurobiology Programme, National University of SingaporeSingaporeSingapore
| |
Collapse
|
12
|
Baby N, Alagappan N, Dheen ST, Sajikumar S. MicroRNA-134-5p inhibition rescues long-term plasticity and synaptic tagging/capture in an Aβ(1-42)-induced model of Alzheimer's disease. Aging Cell 2020; 19:e13046. [PMID: 31625272 PMCID: PMC6974725 DOI: 10.1111/acel.13046] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/31/2019] [Accepted: 08/28/2019] [Indexed: 12/21/2022] Open
Abstract
Progressive memory loss is one of the most common characteristics of Alzheimer's disease (AD), which has been shown to be caused by several factors including accumulation of amyloid β peptide (Aβ) plaques and neurofibrillary tangles. Synaptic plasticity and associative plasticity, the cellular basis of memory, are impaired in AD. Recent studies suggest a functional relevance of microRNAs (miRNAs) in regulating plasticity changes in AD, as their differential expressions were reported in many AD brain regions. However, the specific role of these miRNAs in AD has not been elucidated. We have reported earlier that late long-term potentiation (late LTP) and its associative mechanisms such as synaptic tagging and capture (STC) were impaired in Aβ (1-42)-induced AD condition. This study demonstrates that expression of miR-134-5p, a brain-specific miRNA is upregulated in Aβ (1-42)-treated AD hippocampus. Interestingly, the loss of function of miR-134-5p restored late LTP and STC in AD. In AD brains, inhibition of miR-134-5p elevated the expression of plasticity-related proteins (PRPs), cAMP-response-element binding protein (CREB-1) and brain-derived neurotrophic factor (BDNF), which are otherwise downregulated in AD condition. The results provide the first evidence that the miR-134-mediated post-transcriptional regulation of CREB-1 and BDNF is an important molecular mechanism underlying the plasticity deficit in AD; thus demonstrating the critical role of miR-134-5p as a potential therapeutic target for restoring plasticity in AD condition.
Collapse
Affiliation(s)
- Nimmi Baby
- Department of PhysiologyYong Loo Lin School of MedicineNational University Health SystemNational University of SingaporeSingapore
- Centre for Life SciencesLife Sciences Institute, Neurobiology ProgrammeNational University of SingaporeSingapore
| | - Nithyakalyani Alagappan
- Department of PhysiologyYong Loo Lin School of MedicineNational University Health SystemNational University of SingaporeSingapore
- Centre for Life SciencesLife Sciences Institute, Neurobiology ProgrammeNational University of SingaporeSingapore
| | - Shaikali Thameem Dheen
- Department of AnatomyYong Loo Lin School of MedicineNational University Health SystemNational University of SingaporeSingapore
| | - Sreedharan Sajikumar
- Department of PhysiologyYong Loo Lin School of MedicineNational University Health SystemNational University of SingaporeSingapore
- Centre for Life SciencesLife Sciences Institute, Neurobiology ProgrammeNational University of SingaporeSingapore
| |
Collapse
|
13
|
Tsokas P, Rivard B, Hsieh C, Cottrell JE, Fenton AA, Sacktor TC. Antisense Oligodeoxynucleotide Perfusion Blocks Gene Expression of Synaptic Plasticity-related Proteins without Inducing Compensation in Hippocampal Slices. Bio Protoc 2019; 9:e3387. [PMID: 31803793 PMCID: PMC6892586 DOI: 10.21769/bioprotoc.3387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/29/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
The elucidation of the molecular mechanisms of long-term synaptic plasticity has been hindered by both the compensation that can occur after chronic loss of the core plasticity molecules and by ex vivo conditions that may not reproduce in vivo plasticity. Here we describe a novel method to rapidly suppress gene expression by antisense oligodeoxynucleotides (ODNs) applied to rodent brain slices in an "Oslo-type" interface chamber. The method has three advantageous features: 1) rapid blockade of new synthesis of the targeted proteins that avoids genetic compensation, 2) efficient oxygenation of the brain slice, which is critical for reproducing in vivo conditions of long-term synaptic plasticity, and 3) a recirculation system that uses only small volumes of bath solution (< 5 ml), reducing the amount of reagents required for long-term experiments lasting many hours. The method employs a custom-made recirculation system involving piezoelectric micropumps and was first used for the acute translational blockade of protein kinase Mζ (PKMζ) synthesis during long-term potentiation (LTP) by Tsokas et al., 2016. In that study, applying antisense-ODN rapidly prevents the synthesis of PKMζ and blocks late-LTP without inducing the compensation by other protein kinase C (PKC) isoforms that occurs in PKCζ/PKMζ knockout mice. In addition, we show that in a low-oxygenation submersion-type chamber, applications of the atypical PKC inhibitor, zeta inhibitory peptide (ZIP), can result in unstable baseline synaptic transmission, but in the high-oxygenation, "Oslo-type" interface electrophysiology chamber, the drug reverses late-LTP without affecting baseline synaptic transmission. This comparison reveals that the interface chamber, but not the submersion chamber, reproduces the effects of ZIP in vivo. Therefore, the protocol combines the ability to acutely block new synthesis of specific proteins for the study of long-term synaptic plasticity, while maintaining properties of synaptic transmission that reproduce in vivo conditions relevant for long-term memory.
Collapse
Affiliation(s)
- Panayiotis Tsokas
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, United States
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, United States
| | - Bruno Rivard
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, United States
| | - Changchi Hsieh
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, United States
| | - James E. Cottrell
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, United States
| | - André Antonio Fenton
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, United States
- Center for Neural Science, New York University, New York, United States
| | - Todd Charlton Sacktor
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, United States
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, United States
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, United States
| |
Collapse
|
14
|
Activation of microglia in acute hippocampal slices affects activity-dependent long-term potentiation and synaptic tagging and capture in area CA1. Neurobiol Learn Mem 2019; 163:107039. [DOI: 10.1016/j.nlm.2019.107039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
|
15
|
Wong LW, Tann JY, Ibanez CF, Sajikumar S. The p75 Neurotrophin Receptor Is an Essential Mediator of Impairments in Hippocampal-Dependent Associative Plasticity and Memory Induced by Sleep Deprivation. J Neurosci 2019; 39:5452-5465. [PMID: 31085607 PMCID: PMC6616296 DOI: 10.1523/jneurosci.2876-18.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/22/2023] Open
Abstract
Sleep deprivation (SD) interferes with hippocampal structural and functional plasticity, formation of long-term memory and cognitive function. The molecular mechanisms underlying these effects are incompletely understood. Here, we show that SD impaired synaptic tagging and capture and behavioral tagging, two major mechanisms of associative learning and memory. Strikingly, mutant male mice lacking the p75 neurotrophin receptor (p75NTR) were resistant to the detrimental effects of SD on hippocampal plasticity at both cellular and behavioral levels. Mechanistically, SD increased p75NTR expression and its interaction with phosphodiesterase. p75NTR deletion preserved hippocampal structural and functional plasticity by preventing SD-mediated effects on hippocampal cAMP-CREB-BDNF, cAMP-PKA-LIMK1-cofilin, and RhoA-ROCK2 pathways. Our study identifies p75NTR as an important mediator of hippocampal structural and functional changes associated with SD, and suggests that targeting p75NTR could be a promising strategy to limit the memory and cognitive deficits that accompany sleep loss.SIGNIFICANCE STATEMENT The lack of sufficient sleep is a major health concern in today's world. Sleep deprivation (SD) affects cognitive functions such as memory. We have investigated how associative memory mechanisms, synaptic tagging and capture (STC), was impaired in SD mice at cellular and behavioral level. Interestingly, mutant male mice that lacked the p75 neurotrophin receptor (p75NTR) were seen to be resistant to the SD-induced impairments in hippocampal synaptic plasticity and STC. Additionally, we elucidated the molecular pathways responsible for this rescue of plasticity in the mutant mice. Our study has thus identified p75NTR as a promising target to limit the cognitive deficits associated with SD.
Collapse
Affiliation(s)
- Lik-Wei Wong
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| | - Jason Y Tann
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| | - Carlos F Ibanez
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm S-17177, Sweden
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore,
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| |
Collapse
|
16
|
Mak A, Dharmadhikari B, Kow NY, Thamboo TP, Tang Q, Wong LW, Sajikumar S, Wong HY, Schwarz H. Deletion of CD137 Ligand Exacerbates Renal and Cutaneous but Alleviates Cerebral Manifestations in Lupus. Front Immunol 2019; 10:1411. [PMID: 31297111 PMCID: PMC6607944 DOI: 10.3389/fimmu.2019.01411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/04/2019] [Indexed: 01/01/2023] Open
Abstract
The CD137—CD137 ligand (CD137L) costimulatory system is a critical immune checkpoint with pathophysiological implications in autoimmunity. In this study, we investigated the role of CD137L-mediated costimulation on renal, cutaneous and cerebral manifestations in lupus and the underlying immunological mechanism. Lupus-prone C57BL/6lpr−/− (B6.lpr) mice were crossed to C57BL/6.CD137L−/− mice to obtain CD137L-deficient B6.lpr [double knock out (DKO)] mice. We investigated the extent of survival, glomerulonephritis, skin lesions, cerebral demyelination, immune deviation and long-term synaptic plasticity among the two mouse groups. Cytokine levels, frequency of splenic leukocyte subsets and phenotypes were compared between DKO, B6.lpr and B6.WT mice. A 22 month observation of 226 DKO and 137 B6.lpr mice demonstrated significantly more frequent proliferative glomerulonephritis, larger skin lesions and shorter survival in DKO than in B6.lpr mice. Conversely, microglial activation and cerebral demyelination were less pronounced while long-term synaptic plasticity, was superior in DKO mice. Splenic Th17 cells were significantly higher in DKO than in B6.lpr and B6.WT mice while Th1 and Th2 cell frequencies were comparable between DKO and B6.lpr mice. IL-10 and IL-17 expression by T cells was not affected but there were fewer IL-10-producing myeloid (CD11b+) cells, and also lower serum IL-10 levels in DKO than in B6.lpr mice. The absence of CD137L causes an immune deviation toward Th17, fewer IL-10-producing CD11b+ cells and reduced serum IL-10 levels which potentially explain the more severe lupus in DKO mice while leading to reduced microglia activation, lesser cerebral damage and less severe neurological deficits.
Collapse
Affiliation(s)
- Anselm Mak
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Department of Medicine, National University of Singapore, Singapore, Singapore.,Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Bhushan Dharmadhikari
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| | - Nien Yee Kow
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Qianqiao Tang
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| | - Lik Wei Wong
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Hiu Yi Wong
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, National University of Singapore, Singapore, Singapore.,Immunlogy Programme, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Li D, Jing D, Liu Z, Chen Y, Huang F, Behnisch T. Enhanced Expression of Secreted α-Klotho in the Hippocampus Alters Nesting Behavior and Memory Formation in Mice. Front Cell Neurosci 2019; 13:133. [PMID: 31001090 PMCID: PMC6454015 DOI: 10.3389/fncel.2019.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/18/2019] [Indexed: 12/30/2022] Open
Abstract
The klotho gene family consists of α-, β-, and γ-Klotho, which encode type I single-pass transmembrane proteins with large extracellular domains. α-Klotho exists as a full-length membrane-bound and as a soluble form after cleavage of the extracellular domain. Due to gene splicing, a short extracellular Klotho form can be expressed and secreted. Inactivation of α-Klotho leads to a phenotype that resembles accelerated aging, as the expression level of the α-Klotho protein in the hippocampal formation of mice decreases with age. Here, we show that intrahippocampal viral expression of secreted human α-Klotho alters social behavior and memory formation. Interestingly, overexpression of secreted human α-Klotho in the CA1 changed the nest-building behavior and improved object recognition, object location and passive avoidance memory. Moreover, α-Klotho overexpression increased hippocampal synaptic transmission in response to standardized stimulation strengths, altered paired-pulse facilitation of synaptic transmission, and enhanced activity-dependent synaptic plasticity. These results indicate that memory formation benefits from an augmented level of secreted α-Klotho.
Collapse
Affiliation(s)
- Dongxue Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Dongqing Jing
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ziyang Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Fang Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Tann JY, Wong LW, Sajikumar S, Ibáñez CF. Abnormal TDP-43 function impairs activity-dependent BDNF secretion, synaptic plasticity, and cognitive behavior through altered Sortilin splicing. EMBO J 2019; 38:embj.2018100989. [PMID: 30692134 DOI: 10.15252/embj.2018100989] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
Aberrant function of the RNA-binding protein TDP-43 has been causally linked to multiple neurodegenerative diseases. Due to its large number of targets, the mechanisms through which TDP-43 malfunction cause disease are unclear. Here, we report that knockdown, aggregation, or disease-associated mutation of TDP-43 all impair intracellular sorting and activity-dependent secretion of the neurotrophin brain-derived neurotrophic factor (BDNF) through altered splicing of the trafficking receptor Sortilin. Adult mice lacking TDP-43 specifically in hippocampal CA1 show memory impairment and synaptic plasticity defects that can be rescued by restoring Sortilin splicing or extracellular BDNF. Human neurons derived from patient iPSCs carrying mutated TDP-43 also show altered Sortilin splicing and reduced levels of activity-dependent BDNF secretion, which can be restored by correcting the mutation. We propose that major disease phenotypes caused by aberrant TDP-43 activity may be explained by the abnormal function of a handful of critical proteins, such as BDNF.
Collapse
Affiliation(s)
- Jason Y Tann
- Department of Physiology, National University of Singapore, Singapore City, Singapore.,Life Sciences Institute, National University of Singapore, Singapore City, Singapore
| | - Lik-Wei Wong
- Department of Physiology, National University of Singapore, Singapore City, Singapore.,Life Sciences Institute, National University of Singapore, Singapore City, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore City, Singapore.,Life Sciences Institute, National University of Singapore, Singapore City, Singapore
| | - Carlos F Ibáñez
- Department of Physiology, National University of Singapore, Singapore City, Singapore .,Life Sciences Institute, National University of Singapore, Singapore City, Singapore.,Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
19
|
Weng W, Li D, Peng C, Behnisch T. Recording Synaptic Plasticity in Acute Hippocampal Slices Maintained in a Small-volume Recycling-, Perfusion-, and Submersion-type Chamber System. J Vis Exp 2018. [PMID: 29364264 DOI: 10.3791/55936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Even though experiments on brain slices have been in use since 1951, problems remain that reduce the probability of achieving a stable and successful analysis of synaptic transmission modulation when performing field potential or intracellular recordings. This manuscript describes methodological aspects that might be helpful in improving experimental conditions for the maintenance of acute brain slices and for recording field excitatory postsynaptic potentials in a commercially available submersion chamber with an outflow-carbogenation unit. The outflow-carbogenation helps to stabilize the oxygen level in experiments that rely on the recycling of a small buffer reservoir to enhance the cost-efficiency of drug experiments. In addition, the manuscript presents representative experiments that examine the effects of different carbogenation modes and stimulation paradigms on the activity-dependent synaptic plasticity of synaptic transmission.
Collapse
Affiliation(s)
- Weiguang Weng
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, the Collaborative Innovation Center for Brain Science, Fudan University
| | - Dongxue Li
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, the Collaborative Innovation Center for Brain Science, Fudan University
| | - Cheng Peng
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, the Collaborative Innovation Center for Brain Science, Fudan University
| | - Thomas Behnisch
- The Institutes of Brain Science, the State Key Laboratory of Medical Neurobiology, the Collaborative Innovation Center for Brain Science, Fudan University;
| |
Collapse
|
20
|
Dasgupta A, Kim J, Manakkadan A, Arumugam TV, Sajikumar S. Intermittent fasting promotes prolonged associative interactions during synaptic tagging/capture by altering the metaplastic properties of the CA1 hippocampal neurons. Neurobiol Learn Mem 2017; 154:70-77. [PMID: 29277679 DOI: 10.1016/j.nlm.2017.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 01/18/2023]
Abstract
Metaplasticity is the inherent property of a neuron or neuronal population to undergo activity-dependent changes in neural function that modulate subsequent synaptic plasticity. Here we studied the effect of intermittent fasting (IF) in governing the interactions of associative plasticity mechanisms in the pyramidal neurons of rat hippocampal area CA1. Late long-term potentiation and its associative mechanisms such as synaptic tagging and capture at an interval of 120 min were evaluated in four groups of animals, AL (Ad libitum), IF12 (daily IF for 12 h), IF16 (daily IF for 16 h) and EOD (every other day IF for 24 h). IF had no visible effect on the early or late plasticity but it manifested a critical role in prolonging the associative interactions between weak and strong synapses at an interval of 120 min in IF16 and EOD animals. However, both IF12 and AL did not show associativity at 120 min. Plasticity genes such as Bdnf and Prkcz, which are well known for their expressions in late plasticity and synaptic tagging and capture, were significantly upregulated in IF16 and EOD in comparison to AL. Specific inhibition of brain derived neurotropic factor (BDNF) prevented the prolonged associativity expressed in EOD. Thus, daily IF for 16 h or more can be considered to enhance the metaplastic properties of synapses by improving their associative interactions that might translate into animprovedmemoryformation.
Collapse
Affiliation(s)
- Ananya Dasgupta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, #04-44, 28 Medical Drive, Singapore 117 456, Singapore
| | - Joonki Kim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Republic of Korea
| | - Anoop Manakkadan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, #04-44, 28 Medical Drive, Singapore 117 456, Singapore
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, #04-44, 28 Medical Drive, Singapore 117 456, Singapore.
| |
Collapse
|
21
|
Sharma M, Dierkes T, Sajikumar S. Epigenetic regulation by G9a/GLP complex ameliorates amyloid-beta 1-42 induced deficits in long-term plasticity and synaptic tagging/capture in hippocampal pyramidal neurons. Aging Cell 2017; 16:1062-1072. [PMID: 28665013 PMCID: PMC5595698 DOI: 10.1111/acel.12634] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2017] [Indexed: 12/11/2022] Open
Abstract
Altered epigenetic mechanisms are implicated in the cognitive decline associated with neurodegenerative diseases such as in Alzheimer's disease (AD). AD is the most prevalent form of dementia worldwide; amyloid plaques and neurofibrillary tangles are the histopathological hallmarks of AD. We have recently reported that the inhibition of G9a/GLP complex promotes long-term potentiation (LTP) and its associative mechanisms such as synaptic tagging and capture (STC). However, the role of this complex in plasticity impairments remains elusive. Here, we investigated the involvement of G9a/GLP complex in alleviating the effects of soluble Amyloid-β 1-42 oligomers (oAβ) on neuronal plasticity and associativity in the CA1 region of acute hippocampal slices from 5- to 7-week-old male Wistar rats. Our findings demonstrate that the regulation of G9a/GLP complex by inhibiting its catalytic activity reverses the amyloid-β oligomer-induced deficits in late-LTP and STC. This is achieved by releasing the transcription repression of the brain-derived neurotrophic factor (Bdnf) gene. The catalytic inhibition of G9a/GLP complex leads to the upregulation of Bdnf expression in the slices treated with oAβ. This further ensures the availability of BDNF that subsequently binds its receptor tyrosine kinase B (TrkB) and maintains the late-LTP. Furthermore, the capture of BDNF by weakly activated synapses re-establishes STC. Our findings regarding the reinstatement of functional plasticity and associativity in AD-like conditions provide the first evidence for the role of G9a/GLP complex in AD. We propose G9a/GLP complex as the possible target for preventing oAβ-induced plasticity deficits in hippocampal neurons.
Collapse
Affiliation(s)
- Mahima Sharma
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| | - Tobias Dierkes
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Institute of Innate Immunity; Biomedical Centre; University hospital Bonn; Sigmund-Freud-Str. 25 Bonn 53127 Germany
- Division of Cellular Neurobiology; Zoological Institute; Technical University Braunschweig; Braunschweig Germany
| | - Sreedharan Sajikumar
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| |
Collapse
|
22
|
Substance P induces plasticity and synaptic tagging/capture in rat hippocampal area CA2. Proc Natl Acad Sci U S A 2017; 114:E8741-E8749. [PMID: 28973908 DOI: 10.1073/pnas.1711267114] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The hippocampal area Cornu Ammonis (CA) CA2 is important for social interaction and is innervated by Substance P (SP)-expressing supramammillary (SuM) nucleus neurons. SP exerts neuromodulatory effects on pain processing and central synaptic transmission. Here we provide evidence that SP can induce a slowly developing NMDA receptor- and protein synthesis-dependent potentiation of synaptic transmission that can be induced not only at entorhinal cortical (EC)-CA2 synapses but also at long-term potentiation (LTP)-resistant Schaffer collateral (SC)-CA2 synapses. In addition, SP-induced potentiation of SC-CA2 synapses transforms a short-term potentiation of EC-CA2 synaptic transmission into LTP, consistent with the synaptic tagging and capture hypothesis. Interestingly, this SP-induced potentiation and associative interaction between the EC and SC inputs of CA2 neurons is independent of the GABAergic system. In addition, CaMKIV and PKMζ play a critical role in the SP-induced effects on SC-CA2 and EC-CA2 synapses. Thus, afferents from SuM neurons are ideally situated to prime CA2 synapses for the formation of long-lasting plasticity and associativity.
Collapse
|
23
|
Shetty MS, Sharma M, Sajikumar S. Chelation of hippocampal zinc enhances long-term potentiation and synaptic tagging/capture in CA1 pyramidal neurons of aged rats: implications to aging and memory. Aging Cell 2017; 16:136-148. [PMID: 27633878 PMCID: PMC5242293 DOI: 10.1111/acel.12537] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2016] [Indexed: 01/08/2023] Open
Abstract
Aging is associated with decline in cognitive functions, prominently in the memory consolidation and association capabilities. Hippocampus plays a crucial role in the formation and maintenance of long‐term associative memories, and a significant body of evidence shows that impairments in hippocampal function correlate with aging‐related memory loss. A number of studies have implicated alterations in hippocampal synaptic plasticity, such as long‐term potentiation (LTP), in age‐related cognitive decline although exact mechanisms underlying are not completely clear. Zinc deficiency and the resultant adverse effects on cognition have been well studied. However, the role of excess of zinc in synaptic plasticity, especially in aging, is not addressed well. Here, we have investigated the hippocampal zinc levels and the impairments in synaptic plasticity, such as LTP and synaptic tagging and capture (STC), in the CA1 region of acute hippocampal slices from 82‐ to 84‐week‐old male Wistar rats. We report increased zinc levels in the hippocampus of aged rats and also deficits in the tetani‐induced and dopaminergic agonist‐induced late‐LTP and STC. The observed deficits in synaptic plasticity were restored upon chelation of zinc using a cell‐permeable chelator. These data suggest that functional plasticity and associativity can be successfully established in aged neural networks by chelating zinc with cell‐permeable chelating agents.
Collapse
Affiliation(s)
- Mahesh Shivarama Shetty
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| | - Mahima Sharma
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| | - Sreedharan Sajikumar
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| |
Collapse
|
24
|
Krishna K, Behnisch T, Sajikumar S. Inhibition of Histone Deacetylase 3 Restores Amyloid-β Oligomer-Induced Plasticity Deficit in Hippocampal CA1 Pyramidal Neurons. J Alzheimers Dis 2016; 51:783-91. [PMID: 26890755 DOI: 10.3233/jad-150838] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD) are associated with alterations in epigenetic factors leading to cognitive decline. Histone deacetylase 3 (HDAC3) is a known critical epigenetic negative regulator of learning and memory. In this study, attenuation of long-term potentiation by amyloid-β oligomer, and its reversal by specific HDAC3 inhibitor RGFP966, was performed in rat CA1 pyramidal neurons using whole cell voltage-clamp and field recording techniques. Our findings provide the first evidence that amyloid-β oligomer-induced synaptic plasticity impairment can be prevented by inhibition of HDAC3 enzyme both at the single neuron as well as in a population of neurons, thus identifying HDAC3 as a potential target for ameliorating AD related plasticity impairments.
Collapse
Affiliation(s)
- Kumar Krishna
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thomas Behnisch
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, Singapore
| |
Collapse
|
25
|
Shetty MS, Sajikumar S. Differential involvement of Ca 2+/calmodulin-dependent protein kinases and mitogen-activated protein kinases in the dopamine D1/D5 receptor-mediated potentiation in hippocampal CA1 pyramidal neurons. Neurobiol Learn Mem 2016; 138:111-120. [PMID: 27470093 DOI: 10.1016/j.nlm.2016.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/21/2016] [Accepted: 07/23/2016] [Indexed: 10/21/2022]
Abstract
Dopaminergic neurotransmission modulates and influences hippocampal CA1 synaptic plasticity, learning and long-term memory mechanisms. Investigating the mechanisms involved in the slow-onset potentiation induced by the dopamine D1/D5 receptor agonists in hippocampal CA1 region, we have reported recently that it could play a role in regulating synaptic cooperation and competition. We have also shown that a sustained activation of MEK/MAP kinase pathway was involved in the maintenance of this long-lasting potentiation (Shivarama Shetty, Gopinadhan, & Sajikumar, 2016). However, the molecular aspects of the induction of dopaminergic slow-onset potentiation are not known. Here, we investigated the involvement of MEK/MAPK pathway and Ca2+-calmodulin-dependent protein kinases (CaMKII and CaMKIV) in the induction and maintenance phases of the D1/D5 receptor-mediated slow-onset potentiation. We report differential involvement of these kinases in a dose-dependent manner wherein at weaker levels of dopaminergic activation, both CaMKII and MEK1/2 activation is necessary for the establishment of potentiation and with sufficiently stronger dopaminergic activation, the role of CaMKII becomes dispensable whereas MEK activation remains crucial for the long-lasting potentiation. The results are interesting in view of the involvement of the hippocampal dopaminergic system in a variety of cognitive abilities including memory formation and also in neurological diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Mahesh Shivarama Shetty
- Department of Physiology, Block MD9, 2 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Neurobiology/Aging Program, 28 Medical Drive, Life Sciences Institute, National University of Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Block MD9, 2 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Neurobiology/Aging Program, 28 Medical Drive, Life Sciences Institute, National University of Singapore, Singapore.
| |
Collapse
|