1
|
Wu Y, Libby JB, Dumitrescu L, De Jager PL, Menon V, Schneider JA, Bennett DA, Hohman TJ. Association of ten VEGF family genes with Alzheimer's disease endophenotypes at single cell resolution. Alzheimers Dement 2025; 21:e14419. [PMID: 39641382 PMCID: PMC11848196 DOI: 10.1002/alz.14419] [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: 08/26/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Using a single-nucleus transcriptome derived from the dorsolateral prefrontal cortex of 424 Religious Orders Study and the Rush Memory and Aging Project (ROS/MAP) participants, we investigated the cell type-specific effect of ten vascular endothelial growth factor (VEGF) genes on Alzheimer's disease (AD) endophenotypes. METHODS Negative binomial mixed models were used for differential gene expression and association analysis with AD endophenotypes. VEGF-associated intercellular communication was also profiled. RESULTS Higher microglia FLT1, endothelial FLT4, and oligodendrocyte VEGFB are associated with greater amyloid beta (Aβ) load, whereas higher VEGFB expression in inhibitory neurons is associated with lower Aβ load. Higher astrocyte NRP1 is associated with lower tau density. Higher microglia and endothelial FLT1 are associated with worse cognition performance. Endothelial and microglial FLT1 expression was upregulated in clinical AD patients compared to cognitively normal controls. Finally, AD cells showed a significant reduction in VEGF signaling compared to controls. DISCUSSION Our results highlight key changes in VEGF receptor expression in endothelial and microglial cells during AD, and the potential protective role of VEGFB in neurons. HIGHLIGHTS The prefrontal cortical expression of FLT1 and FLT4 was associated with worse cross-sectional global cognitive function, longitudinal cognitive trajectories, and more Alzheimer's disease (AD) neuropathology. The associations between FLT1 or FLT4 and AD endophenotypes appear to be driven by endothelial and microglial cells. VEGFB expression seems to have opposing effects on the Aβ burden in AD depending on cell types, highlighting its potential protective role in neurons.
Collapse
Affiliation(s)
- Yiyang Wu
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Julia B. Libby
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Philip L. De Jager
- Center for Translational & Computational NeuroimmunologyDepartment of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Vilas Menon
- Center for Translational & Computational NeuroimmunologyDepartment of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Julie A. Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
2
|
Welle TM, Rajgor D, Kareemo DJ, Garcia JD, Zych SM, Wolfe SE, Gookin SE, Martinez TP, Dell'Acqua ML, Ford CP, Kennedy MJ, Smith KR. miRNA-mediated control of gephyrin synthesis drives sustained inhibitory synaptic plasticity. EMBO Rep 2024; 25:5141-5168. [PMID: 39294503 PMCID: PMC11549329 DOI: 10.1038/s44319-024-00253-z] [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: 12/15/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024] Open
Abstract
Activity-dependent protein synthesis is crucial for long-lasting forms of synaptic plasticity. However, our understanding of translational mechanisms controlling GABAergic synapses is limited. One distinct form of inhibitory long-term potentiation (iLTP) enhances postsynaptic clusters of GABAARs and the primary inhibitory scaffold, gephyrin, to promote sustained synaptic strengthening. While we previously found that persistent iLTP requires mRNA translation, the mechanisms controlling plasticity-induced gephyrin translation remain unknown. We identify miR153 as a novel regulator of Gphn mRNA translation which controls gephyrin protein levels and synaptic clustering, ultimately impacting inhibitory synaptic structure and function. iLTP induction downregulates miR153, reversing its translational suppression of Gphn mRNA and promoting de novo gephyrin protein synthesis and synaptic clustering during iLTP. Finally, we find that reduced miR153 expression during iLTP is driven by an excitation-transcription coupling pathway involving calcineurin, NFAT and HDACs, which also controls the miRNA-dependent upregulation of GABAARs. Together, we delineate a miRNA-dependent post-transcriptional mechanism that controls the expression of the key synaptic scaffold, gephyrin, and may converge with parallel miRNA pathways to coordinate gene upregulation to maintain inhibitory synaptic plasticity.
Collapse
Affiliation(s)
- Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Dean J Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Joshua D Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sarah M Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sarah E Wolfe
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sara E Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Tyler P Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
3
|
Sayehmiri F, Motamedi F, Batool Z, Naderi N, Shaerzadeh F, Zoghi A, Rezaei O, Khodagholi F, Pourbadie HG. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14897. [PMID: 39097920 PMCID: PMC11298206 DOI: 10.1111/cns.14897] [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: 01/05/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
Synaptic plasticity is believed to underlie the cellular and molecular basis of memory formation. Mitochondria are one of the main organelles involved in metabolism and energy maintenance as plastic organelles that change morphologically and functionally in response to cellular needs and regulate synaptic function and plasticity through multiple mechanisms, including ATP generation, calcium homeostasis, and biogenesis. An increased neuronal activity enhances synaptic efficiency, during which mitochondria's spatial distribution and morphology change significantly. These organelles build up in the pre-and postsynaptic zones to produce ATP, which is necessary for several synaptic processes like neurotransmitter release and recycling. Mitochondria also regulate calcium homeostasis by buffering intracellular calcium, which ensures proper synaptic activity. Furthermore, mitochondria in the presynaptic terminal have distinct morphological properties compared to dendritic or postsynaptic mitochondria. This specialization enables precise control of synaptic activity and plasticity. Mitochondrial dysfunction has been linked to synaptic failure in many neurodegenerative disorders, like Alzheimer's disease (AD). In AD, malfunctioning mitochondria cause delays in synaptic vesicle release and recycling, ionic gradient imbalances, and mostly synaptic failure. This review emphasizes mitochondrial plasticity's contribution to synaptic function. It also explores the profound effect of mitochondrial malfunction on neurodegenerative disorders, focusing on AD, and provides an overview of how they sustain cellular health under normal conditions and how their malfunction contributes to neurodegenerative diseases, highlighting their potential as a therapeutic target for such conditions.
Collapse
Affiliation(s)
- Fatemeh Sayehmiri
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Nima Naderi
- Department of Pharmacology and Toxicology, Faculty of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | | | - Anahita Zoghi
- Department of Neurology, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Omidvar Rezaei
- Skull Base Research CenterLoghman Hakim Hospital, Shahid Beheshti University of Medical SciencesTehranIran
| | - Fariba Khodagholi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
4
|
Ganesh A, Choudhury W, Coutellier L. Early spatial recognition memory deficits in 5XFAD female mice are associated with disruption of prefrontal parvalbumin neurons. Brain Res 2024; 1841:149122. [PMID: 39009061 DOI: 10.1016/j.brainres.2024.149122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Women have a two-fold increased risk of developing Alzheimer's disease (AD) than men, yet the underlying mechanisms of this sex-specific vulnerability remain unknown. Here, we aimed at determining in the 5XFAD mouse model whether deficits in prefrontal-dependent cognitive functions, which are impacted in the preclinical stages of AD, appear earlier in females, and whether these cognitive deficits are associated with alterations in the activity of prefrontal parvalbumin (PV)-neurons that regulate prefrontal circuits activity. We observed that 3.5-month-old 5XFAD females, but not males, display impairments in spatial short-term recognition memory, a function that relies on the integrity of the prefrontal cortex. Hippocampal-dependent cognitive functions were intact in both sexes. We then observed that 5XFAD females have more prefrontal PV neurons expressing the marker of chronic activity FosB; this was inversely correlated with prefrontal-dependent cognitive performances. Our findings show for the first time sex-specific, early deregulation of prefrontal PV neurons activity, which is associated with early appearance of prefrontal-dependent cognitive functions in 5XFAD females providing a potential novel mechanism to the increased risk to AD in females.
Collapse
Affiliation(s)
- Anish Ganesh
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Wajih Choudhury
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Laurence Coutellier
- Department of Psychology, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Li H, Lai L, Li X, Wang R, Fang X, Xu N, Zhao J. Electroacupuncture Ameliorates Cognitive Impairment by Regulating γ-Amino Butyric Acidergic Interneurons in the Hippocampus of 5 Familial Alzheimer's Disease Mice. Neuromodulation 2024; 27:730-741. [PMID: 36604241 DOI: 10.1016/j.neurom.2022.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVES γ-amino butyric acid (GABA)-ergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease (AD). Inhibitory interneurons play an important role in the regulation of E/I balance, synaptic transmission, and network oscillation through manipulation of GABAergic functions, showing positive outcomes in AD animal models. Mice expressing 5 familial AD mutation (5xFAD) exhibited a series of AD-like pathology and learning and memory deficits with age. Because electroacupuncture (EA) treatment has been used for a complementary alternative medicine therapy in patients with AD, we aimed to examine any usefulness of EA therapy in GABA interneuron function and its associated synaptic proteins, to determine whether EA could effectively improve inhibitory transmission and network oscillation and eventually alleviate cognitive impairments in 5xFAD mice, and to further elucidate the GABAergic system function underlying the antidementia response of EA. MATERIALS AND METHODS 5xFAD mice were used to evaluate the potential neuroprotective effect of electroacupuncture at Baihui (DU 20) and Dazhui (DU 14) through behavioral testing, immunofluorescence staining, electrophysiology recording, and molecular biology analysis. RESULTS First, we observed that EA improved memory deficits and inhibitory synaptic protein expression. Second, EA treatment alleviated the decrease of somatostatin-positive interneurons in the dorsal hippocampus. Third, EA attenuated E/I imbalance in 5xFAD mice. Last, EA treatment enhanced theta and gamma oscillation in the hippocampus of 5xFAD mice. CONCLUSIONS EA stimulation at DU20 and DU14 acupoints may be a potential alternative therapy to ameliorate cognitive deficits in AD through the regulation of the function of the GABAergic interneuron.
Collapse
Affiliation(s)
- Hongzhu Li
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rehabilitation, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanfeng Lai
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Li
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runyi Wang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoling Fang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaying Zhao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
6
|
Wu Y, Libby JB, Dumitrescu L, De Jager PL, Menon V, Schneider JA, Bennett DA, Hohman TJ. Association of 10 VEGF Family Genes with Alzheimer's Disease Endophenotypes at Single Cell Resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589221. [PMID: 38826287 PMCID: PMC11142115 DOI: 10.1101/2024.04.12.589221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The cell-type specific role of the vascular endothelial growth factors (VEGFs) in the pathogenesis of Alzheimer's disease (AD) is not well characterized. In this study, we utilized a single-nucleus RNA sequencing dataset from Dorsolateral Prefrontal Cortex (DLFPC) of 424 donors from the Religious Orders Study and Memory and Aging Project (ROS/MAP) to investigate the effect of 10 VEGF genes ( VEGFA, VEGFB, VEGFC, VEGFD, PGF, FLT1, FLT4, KDR, NRP1 , and NRP2 ) on AD endophenotypes. Mean age of death was 89 years, among which 68% were females, and 52% has AD dementia. Negative binomial mixed models were used for differential expression analysis and for association analysis with β-amyloid load, PHF tau tangle density, and both cross-sectional and longitudinal global cognitive function. Intercellular VEGF-associated signaling was profiled using CellChat. We discovered prefrontal cortical FLT1 expression was upregulated in AD brains in both endothelial and microglial cells. Higher FLT1 expression was also associated with worse cross-sectional global cognitive function, longitudinal cognitive trajectories, and β-amyloid load. Similarly, higher endothelial FLT4 expression was associated with more β-amyloid load. In contrast to the receptors, VEGFB showed opposing effects on β-amyloid load whereby higher levels in oligodendrocytes was associated with high amyloid burden, while higher levels in inhibitory neurons was associated with lower amyloid burden. Finally, AD cells showed significant reduction in overall VEGF signaling comparing to those from cognitive normal participants. Our results highlight key changes in VEGF receptor expression in endothelial and microglial cells during AD, and the potential protective role of VEGFB in neurons.
Collapse
|
7
|
Kiss E, Kins S, Gorgas K, Venczel Szakács KH, Kirsch J, Kuhse J. Another Use for a Proven Drug: Experimental Evidence for the Potential of Artemisinin and Its Derivatives to Treat Alzheimer's Disease. Int J Mol Sci 2024; 25:4165. [PMID: 38673751 PMCID: PMC11049906 DOI: 10.3390/ijms25084165] [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: 02/05/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Plant-derived multitarget compounds may represent a promising therapeutic strategy for multifactorial diseases, such as Alzheimer's disease (AD). Artemisinin and its derivatives were indicated to beneficially modulate various aspects of AD pathology in different AD animal models through the regulation of a wide range of different cellular processes, such as energy homeostasis, apoptosis, proliferation and inflammatory pathways. In this review, we aimed to provide an up-to-date overview of the experimental evidence documenting the neuroprotective activities of artemi-sinins to underscore the potential of these already-approved drugs for treating AD also in humans and propose their consideration for carefully designed clinical trials. In particular, the benefits to the main pathological hallmarks and events in the pathological cascade throughout AD development in different animal models of AD are summarized. Moreover, dose- and context-dependent effects of artemisinins are noted.
Collapse
Affiliation(s)
- Eva Kiss
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania;
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, 69120 Kaiserslautern, Germany;
| | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| | - Kinga Hajnal Venczel Szakács
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania;
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| | - Jochen Kuhse
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany; (K.G.); (J.K.)
| |
Collapse
|
8
|
Niraula S, Yan SS, Subramanian J. Amyloid Pathology Impairs Experience-Dependent Inhibitory Synaptic Plasticity. J Neurosci 2024; 44:e0702232023. [PMID: 38050105 PMCID: PMC10860629 DOI: 10.1523/jneurosci.0702-23.2023] [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: 04/19/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023] Open
Abstract
Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by subtle broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
- Department of Surgery, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York 10032
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
9
|
Welle TM, Rajgor D, Garcia JD, Kareemo D, Zych SM, Gookin SE, Martinez TP, Dell’Acqua ML, Ford CP, Kennedy MJ, Smith KR. miRNA-mediated control of gephyrin synthesis drives sustained inhibitory synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.570420. [PMID: 38168421 PMCID: PMC10760056 DOI: 10.1101/2023.12.12.570420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Activity-dependent protein synthesis is crucial for many long-lasting forms of synaptic plasticity. However, our understanding of the translational mechanisms controlling inhibitory synapses is limited. One distinct form of inhibitory long-term potentiation (iLTP) enhances postsynaptic clusters of GABAARs and the primary inhibitory scaffold, gephyrin, to promote sustained synaptic strengthening. While we previously found that persistent iLTP requires mRNA translation, the precise mechanisms controlling gephyrin translation during this process remain unknown. Here, we identify miR153 as a novel regulator of Gphn mRNA translation which controls gephyrin protein levels and synaptic clustering, ultimately impacting GABAergic synaptic structure and function. We find that iLTP induction downregulates miR153, reversing its translational suppression of Gphn mRNA and allowing for increased de novo gephyrin protein synthesis and synaptic clustering during iLTP. Finally, we find that reduced miR153 expression during iLTP is driven by an excitation-transcription coupling pathway involving calcineurin, NFAT and HDACs, which also controls the miRNA-dependent upregulation of GABAARs. Overall, this work delineates a miRNA-dependent post-transcriptional mechanism that controls the expression of the key synaptic scaffold, gephyrin, and may converge with parallel miRNA pathways to coordinate gene upregulation to maintain inhibitory synaptic plasticity.
Collapse
Affiliation(s)
- Theresa M. Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
- T.M.W and D.R. contributed equally to this work
| | - Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
- T.M.W and D.R. contributed equally to this work
| | - Joshua D. Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Dean Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Sarah M. Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Sara E. Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Tyler P. Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Matthew J. Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Katharine R. Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| |
Collapse
|
10
|
Hijazi S, Smit AB, van Kesteren RE. Fast-spiking parvalbumin-positive interneurons in brain physiology and Alzheimer's disease. Mol Psychiatry 2023; 28:4954-4967. [PMID: 37419975 PMCID: PMC11041664 DOI: 10.1038/s41380-023-02168-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
Fast-spiking parvalbumin (PV) interneurons are inhibitory interneurons with unique morphological and functional properties that allow them to precisely control local circuitry, brain networks and memory processing. Since the discovery in 1987 that PV is expressed in a subset of fast-spiking GABAergic inhibitory neurons, our knowledge of the complex molecular and physiological properties of these cells has been expanding. In this review, we highlight the specific properties of PV neurons that allow them to fire at high frequency and with high reliability, enabling them to control network oscillations and shape the encoding, consolidation and retrieval of memories. We next discuss multiple studies reporting PV neuron impairment as a critical step in neuronal network dysfunction and cognitive decline in mouse models of Alzheimer's disease (AD). Finally, we propose potential mechanisms underlying PV neuron dysfunction in AD and we argue that early changes in PV neuron activity could be a causal step in AD-associated network and memory impairment and a significant contributor to disease pathogenesis.
Collapse
Affiliation(s)
- Sara Hijazi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Niraula S, Yan SS, Subramanian J. Amyloid pathology impairs experience-dependent inhibitory synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539450. [PMID: 37205469 PMCID: PMC10187277 DOI: 10.1101/2023.05.04.539450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by the broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
- Department of Surgery, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
12
|
Mao R, Xu S, Sun G, Yu Y, Zuo Z, Wang Y, Yang K, Zhang Z, Yang W. Triptolide injection reduces Alzheimer's disease-like pathology in mice. Synapse 2023; 77:e22261. [PMID: 36633502 DOI: 10.1002/syn.22261] [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: 03/24/2022] [Revised: 12/21/2022] [Accepted: 01/08/2023] [Indexed: 01/13/2023]
Abstract
Triptolide is an epoxidized diterpene lactone isolated from Tripterygium wilfordii. Studies have shown that triptolide exerts organ-protective effects. However, it remains unknown whether triptolide improves Alzheimer's disease (AD)-like presentations. Thirty healthy 8-week-old male C57BL/6J mice were randomly divided into control (n = 10), model (n = 10), and triptolide (n = 10) groups. Amyloid-β (Aβ)42 was injected bilaterally into the ventricles of mice in the model group. Triptolide was injected intraperitoneally daily after injecting Aβ42 (a total of 30 days) in the triptolide group. Learning and memory were tested using the Morris water maze test. The deposition of Aβ42 in the hippocampus was detected using immunohistochemical staining. In the hippocampus, three synaptic-associated proteins-gephyrin, collybistin, and GABRA1 -were detected by western blotting. Furthermore, we used ELISA to detect proinflammatory cytokines, including TNF-α and IL-1β, in the blood and hippocampus. Moreover, superoxide dismutase (SOD), malondialdehyde (MDA), and GSH levels were measured using the corresponding kits. We found that triptolide improved spatial learning and memory in AD-like mice. Additionally, triptolide maintained the expression of gephyrin, collybistin, and GABRA1 and reduced Aβ in these mice. Additionally, triptolide reduced the expression of inflammatory cytokines and decreased oxidative damage in AD-like mice. Our study suggests that triptolide attenuates AD-like changes in the mouse brain.
Collapse
Affiliation(s)
- Rui Mao
- Department of Neurology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shihao Xu
- Department of Neurology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Guangwen Sun
- Department of Neurology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Neurology, Sinopharm Dongfeng General Hospital, Jinzhou Medical University Union Training Base, Jinzhou, China
| | - Yingying Yu
- Department of Neurology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Neurology, Sinopharm Dongfeng General Hospital, Jinzhou Medical University Union Training Base, Jinzhou, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Yuanyuan Wang
- Department of Neurology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Kun Yang
- Department of Anesthesiology, Sinopharm Dongfeng General Hospital, Jinzhou Medical University Union Training Base, Jinzhou, China
| | - Zhen Zhang
- Department of Orthopedics, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenqiong Yang
- Department of Neurology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
13
|
Melgosa-Ecenarro L, Doostdar N, Radulescu CI, Jackson JS, Barnes SJ. Pinpointing the locus of GABAergic vulnerability in Alzheimer's disease. Semin Cell Dev Biol 2023; 139:35-54. [PMID: 35963663 DOI: 10.1016/j.semcdb.2022.06.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/30/2022] [Accepted: 06/30/2022] [Indexed: 12/31/2022]
Abstract
The early stages of Alzheimer's disease (AD) have been linked to microcircuit dysfunction and pathophysiological neuronal firing in several brain regions. Inhibitory GABAergic microcircuitry is a critical feature of stable neural-circuit function in the healthy brain, and its dysregulation has therefore been proposed as contributing to AD-related pathophysiology. However, exactly how the critical balance between excitatory and inhibitory microcircuitry is modified by AD pathogenesis remains unclear. Here, we set the current evidence implicating dysfunctional GABAergic microcircuitry as a driver of early AD pathophysiology in a simple conceptual framework. Our framework is based on a generalised reductionist model of firing-rate control by local feedback inhibition. We use this framework to consider multiple loci that may be vulnerable to disruption by AD pathogenesis. We first start with evidence investigating how AD-related processes may impact the gross number of inhibitory neurons in the network. We then move to discuss how pathology may impact intrinsic cellular properties and firing thresholds of GABAergic neurons. Finally, we cover how AD-related pathogenesis may disrupt synaptic connectivity between excitatory and inhibitory neurons. We use the feedback inhibition framework to discuss and organise the available evidence from both preclinical rodent work and human studies in AD patients and conclude by identifying key questions and understudied areas for future investigation.
Collapse
Affiliation(s)
- Leire Melgosa-Ecenarro
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Nazanin Doostdar
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Carola I Radulescu
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Johanna S Jackson
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Samuel J Barnes
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
14
|
Tousley AR, Yeh PWL, Yeh HH. Precocious emergence of cognitive and synaptic dysfunction in 3xTg-AD mice exposed prenatally to ethanol. Alcohol 2023; 107:56-72. [PMID: 36038084 PMCID: PMC10183974 DOI: 10.1016/j.alcohol.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting approximately 50 million people worldwide. Early life risk factors for AD, including prenatal exposures, remain underexplored. Exposure of the fetus to alcohol (ethanol) is not uncommon during pregnancy, and may result in physical, behavioral, and cognitive changes that are first detected during childhood but result in lifelong challenges. Whether or not prenatal ethanol exposure may contribute to Alzheimer's disease risk is not yet known. Here we exposed a mouse model of Alzheimer's disease (3xTg-AD), bearing three dementia-associated transgenes, presenilin1 (PS1M146V), human amyloid precursor protein (APPSwe), and human tau (TauP301S), to ethanol on gestational days 13.5-16.5 using an established binge-type maternal ethanol exposure paradigm. We sought to investigate whether prenatal ethanol exposure resulted in a precocious onset or increased severity of AD progression, or both. We found that a brief binge-type gestational exposure to ethanol during a period of peak neuronal migration to the developing cortex resulted in an earlier onset of spatial memory deficits and behavioral inflexibility in the progeny, as assessed by performance on the modified Barnes maze task. The observed cognitive changes coincided with alterations to both GABAergic and glutamatergic synaptic transmission in layer V/VI neurons, diminished GABAergic interneurons, and increased β-amyloid accumulation in the medial prefrontal cortex. These findings provide the first preclinical evidence for prenatal ethanol exposure as a potential factor for modifying the onset of AD-like behavioral dysfunction and set the groundwork for more comprehensive investigations into the underpinnings of AD-like cognitive changes in individuals with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Adelaide R Tousley
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States; MD-PhD Program, Geisel School of Medicine at Dartmouth; Integrative Neuroscience at Dartmouth Graduate Program, Hanover, NH, United States
| | - Pamela W L Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Hermes H Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| |
Collapse
|
15
|
Kuhse J, Groeneweg F, Kins S, Gorgas K, Nawrotzki R, Kirsch J, Kiss E. Loss of Extrasynaptic Inhibitory Glycine Receptors in the Hippocampus of an AD Mouse Model Is Restored by Treatment with Artesunate. Int J Mol Sci 2023; 24:ijms24054623. [PMID: 36902054 PMCID: PMC10002537 DOI: 10.3390/ijms24054623] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by synaptic failure and neuronal loss. Recently, we demonstrated that artemisinins restored the levels of key proteins of inhibitory GABAergic synapses in the hippocampus of APP/PS1 mice, a model of cerebral amyloidosis. In the present study, we analyzed the protein levels and subcellular localization of α2 and α3 subunits of GlyRs, indicated as the most abundant receptor subtypes in the mature hippocampus, in early and late stages of AD pathogenesis, and upon treatment with two different doses of artesunate (ARS). Immunofluorescence microscopy and Western blot analysis demonstrated that the protein levels of both α2 and α3 GlyRs are considerably reduced in the CA1 and the dentate gyrus of 12-month-old APP/PS1 mice when compared to WT mice. Notably, treatment with low-dose ARS affected GlyR expression in a subunit-specific way; the protein levels of α3 GlyR subunits were rescued to about WT levels, whereas that of α2 GlyRs were not affected significantly. Moreover, double labeling with a presynaptic marker indicated that the changes in GlyR α3 expression levels primarily involve extracellular GlyRs. Correspondingly, low concentrations of artesunate (≤1 µM) also increased the extrasynaptic GlyR cluster density in hAPPswe-transfected primary hippocampal neurons, whereas the number of GlyR clusters overlapping presynaptic VIAAT immunoreactivities remained unchanged. Thus, here we provide evidence that the protein levels and subcellular localization of α2 and α3 subunits of GlyRs show regional and temporal alterations in the hippocampus of APP/PS1 mice that can be modulated by the application of artesunate.
Collapse
Affiliation(s)
- Jochen Kuhse
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69117 Heidelberg, Germany
| | - Femke Groeneweg
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69117 Heidelberg, Germany
- Institute of Neuroanatomy, Medical Faculty Mannheim, University Heidelberg, 68167 Mannheim, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69117 Heidelberg, Germany
| | - Ralph Nawrotzki
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69117 Heidelberg, Germany
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69117 Heidelberg, Germany
| | - Eva Kiss
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69117 Heidelberg, Germany
- Department of Cellular and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mures, Romania
- Correspondence:
| |
Collapse
|
16
|
Niraula S, Doderer JJ, Indulkar S, Berry KP, Hauser WL, L'Esperance OJ, Deng JZ, Keeter G, Rouse AG, Subramanian J. Excitation-inhibition imbalance disrupts visual familiarity in amyloid and non-pathology conditions. Cell Rep 2023; 42:111946. [PMID: 36640331 PMCID: PMC9939293 DOI: 10.1016/j.celrep.2022.111946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/14/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Neuronal hyperactivity induces memory deficits in Alzheimer's disease. However, how hyperactivity disrupts memory is unclear. Using in vivo synaptic imaging in the mouse visual cortex, we show that structural excitatory-inhibitory synapse imbalance in the apical dendrites favors hyperactivity in early amyloidosis. Consistent with this, natural images elicit neuronal hyperactivity in these mice. Compensatory changes that maintain activity homeostasis disrupt functional connectivity and increase population sparseness such that a small fraction of neurons dominates population activity. These properties reduce the selectivity of neural response to natural images and render visual recognition memory vulnerable to interference. Deprivation of non-specific visual experiences improves the neural representation and behavioral expression of visual familiarity. In contrast, in non-pathological conditions, deprivation of non-specific visual experiences induces disinhibition, increases excitability, and disrupts visual familiarity. We show that disrupted familiarity occurs when the fraction of high-responsive neurons and the persistence of neural representation of a memory-associated stimulus are not constrained.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Julia J Doderer
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Shreya Indulkar
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Kalen P Berry
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - William L Hauser
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Oliver J L'Esperance
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Jasmine Z Deng
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Griffin Keeter
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Adam G Rouse
- Department of Neurosurgery, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
17
|
Shu S, Xu SY, Ye L, Liu Y, Cao X, Jia JQ, Bian HJ, Liu Y, Zhu XL, Xu Y. Prefrontal parvalbumin interneurons deficits mediate early emotional dysfunction in Alzheimer's disease. Neuropsychopharmacology 2023; 48:391-401. [PMID: 36229597 PMCID: PMC9750960 DOI: 10.1038/s41386-022-01435-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and has an insidious onset. Exploring the characteristics and mechanism of the early symptoms of AD plays a critical role in the early diagnosis and intervention of AD. Here we found that depressive-like behavior and short-term spatial memory dysfunction appeared in APPswe/PS1dE9 mice (AD mice) as early as 9-11 weeks of age. Electrophysiological analysis revealed excitatory/inhibitory (E/I) imbalance in the prefrontal cortex (PFC). This E/I imbalance was induced by significant reduction in the number and activity of parvalbumin interneurons (PV+ INs) in this region. Furthermore, optogenetic and chemogenetic activation of residual PV+ INs effectively ameliorated depressive-like behavior and rescued short-term spatial memory in AD mice. These results suggest the PFC is selectively vulnerable in the early stage of AD and prefrontal PV+ INs deficits play a key role in the occurrence and development of early symptoms of AD.
Collapse
Affiliation(s)
- Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Si-Yi Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, 212013, Jiangsu, PR China
| | - Lei Ye
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
| | - Yi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Jun-Qiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
| | - Hui-Jie Bian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Ying Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
| | - Xiao-Lei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, Jiangsu, PR China.
- Institute of Brain Sciences, Nanjing University, Nanjing, 210093, Jiangsu, PR China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, Jiangsu, PR China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, 210008, Jiangsu, PR China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
18
|
Ruiter M, Herstel LJ, Wierenga CJ. Reduction of Dendritic Inhibition in CA1 Pyramidal Neurons in Amyloidosis Models of Early Alzheimer's Disease. J Alzheimers Dis 2021; 78:951-964. [PMID: 33074225 PMCID: PMC7739974 DOI: 10.3233/jad-200527] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: In an early stage of Alzheimer’s disease (AD), before the formation of amyloid plaques, neuronal network hyperactivity has been reported in both patients and animal models. This suggests an underlying disturbance of the balance between excitation and inhibition. Several studies have highlighted the role of somatic inhibition in early AD, while less is known about dendritic inhibition. Objective: In this study we investigated how inhibitory synaptic currents are affected by elevated Aβ levels. Methods: We performed whole-cell patch clamp recordings of CA1 pyramidal neurons in organotypic hippocampal slice cultures after treatment with Aβ-oligomers and in hippocampal brain slices from AppNL-F-G mice (APP-KI). Results: We found a reduction of spontaneous inhibitory postsynaptic currents (sIPSCs) in CA1 pyramidal neurons in organotypic slices after 24 h Aβ treatment. sIPSCs with slow rise times were reduced, suggesting a specific loss of dendritic inhibitory inputs. As miniature IPSCs and synaptic density were unaffected, these results suggest a decrease in activity-dependent transmission after Aβ treatment. We observed a similar, although weaker, reduction in sIPSCs in CA1 pyramidal neurons from APP-KI mice compared to control. When separated by sex, the strongest reduction in sIPSC frequency was found in slices from male APP-KI mice. Consistent with hyperexcitability in pyramidal cells, dendritically targeting interneurons received slightly more excitatory input. GABAergic action potentials had faster kinetics in APP-KI slices. Conclusion: Our results show that Aβ affects dendritic inhibition via impaired action potential driven release, possibly due to altered kinetics of GABAergic action potentials. Reduced dendritic inhibition may contribute to neuronal hyperactivity in early AD.
Collapse
Affiliation(s)
- Marvin Ruiter
- Department of Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Lotte J Herstel
- Department of Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Corette J Wierenga
- Department of Cell Biology, Neurobiology and Biophysics, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
19
|
Kurucu H, Colom-Cadena M, Davies C, Wilkins L, King D, Rose J, Tzioras M, Tulloch JH, Smith C, Spires-Jones TL. Inhibitory synapse loss and accumulation of amyloid beta in inhibitory presynaptic terminals in Alzheimer's disease. Eur J Neurol 2021; 29:1311-1323. [PMID: 34331352 DOI: 10.1111/ene.15043] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Synapse degeneration in Alzheimer's disease (AD) correlates strongly with cognitive decline. There is well-established excitatory synapse loss in AD with known contributions of pathological amyloid beta (Aβ) to excitatory synapse dysfunction and loss. Despite clear changes in circuit excitability in AD and model systems, relatively little is known about pathology in inhibitory synapses. METHODS Here human postmortem brain samples (n = 5 control, 10 AD cases) from temporal and occipital cortices were examined to investigate whether inhibitory synapses and neurons are lost in AD and whether Aβ may contribute to inhibitory synapse degeneration. Inhibitory neurons were counted in all six cortical layers using stereology software, and array tomography was used to examine synapse density and the accumulation of Aβ in synaptic terminals. RESULTS Differing inhibitory neuron densities were observed in the different cortical layers. The highest inhibitory neuron density was observed in layer 4 in both brain regions and the visual cortex had a higher inhibitory neuron density than the temporal cortex. There was significantly lower inhibitory neuron density in AD than in control cases in all six cortical layers. High-resolution array tomography imaging revealed plaque-associated loss of inhibitory synapses and accumulation of Aβ in a small subset of inhibitory presynaptic terminals with the most accumulation near amyloid plaques. CONCLUSIONS Inhibitory neuron and synapse loss in AD may contribute to disrupted excitatory/inhibitory balance and cognitive decline. Future work is warranted to determine whether targeting inhibitory synapse loss could be a useful therapeutic strategy.
Collapse
Affiliation(s)
- Hatice Kurucu
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Martí Colom-Cadena
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Caitlin Davies
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Lewis Wilkins
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Declan King
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Jamie Rose
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Makis Tzioras
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Jane H Tulloch
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences and Sudden Death Brain Bank, University of Edinburgh, Edinburgh, UK
| | - Tara L Spires-Jones
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| |
Collapse
|
20
|
Artesunate restores the levels of inhibitory synapse proteins and reduces amyloid-β and C-terminal fragments (CTFs) of the amyloid precursor protein in an AD-mouse model. Mol Cell Neurosci 2021; 113:103624. [PMID: 33933588 DOI: 10.1016/j.mcn.2021.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia, characterized histopathologically by the formation of amyloid plaques and neurofibrillary tangles in the brain. Amyloid β-peptide (Aβ) is a major component of amyloid plaques and is released together with carboxy-terminal fragments (CTFs) from the amyloid precursor protein (APP) through proteolytic cleavage, thought to contribute to synapse dysfunction and loss along the progression of AD. Artemisinins, primarily antimalarial drugs, reduce neuroinflammation and improve cognitive capabilities in mouse models of AD. Furthermore, artemisinins were demonstrated to target gephyrin, the main scaffold protein of inhibitory synapses and modulate GABAergic neurotransmission in vitro. Previously, we reported a robust decrease of inhibitory synapse proteins in the hippocampus of 12-month-old double transgenic APP-PS1 mice which overexpress in addition to the Swedish mutated form of the human APP a mutated presenilin 1 (PS1) gene and are characterized by a high plaque load at this age. Here, we provide in vivo evidence that treating these mice with artemisinin or its semisynthetic derivative artesunate in two different doses (10 mg/kg and 100 mg/kg), these compounds affect differently inhibitory synapse components, amyloid plaque load and APP-processing. Immunofluorescence microscopy demonstrated the rescue of gephyrin and γ2-GABAA-receptor protein levels in the brain of treated mice with both, artemisinin and artesunate, most efficiently with a low dose of artesunate. Remarkably, artemisinin reduced only in low dose the amyloid plaque load correlating with lower levels of mutated human APP (hAPPswe) whereas artesunate treatment in both doses resulted in significantly lower plaque numbers. Correspondingly, the level of APP-cleavage products, specifically the amount of CTFs in hippocampus homogenates was reduced significantly only by artesunate, in line with the findings in hAPPswe expressing cultured hippocampal neurons evidencing a concentration-dependent inhibition of CTF-release by artesunate already in the nanomolar range. Thus, our data support artemisinins as neuroprotective multi-target drugs, exhibiting a potent anti-amyloidogenic activity and reinforcing key proteins of inhibitory synapses.
Collapse
|
21
|
Kiss E, Kins S, Gorgas K, Orlik M, Fischer C, Endres K, Schlicksupp A, Kirsch J, Kuhse J. Artemisinin-treatment in pre-symptomatic APP-PS1 mice increases gephyrin phosphorylation at Ser270: a modification regulating postsynaptic GABA AR density. Biol Chem 2021; 403:73-87. [PMID: 33878252 DOI: 10.1515/hsz-2021-0153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/29/2021] [Indexed: 01/26/2023]
Abstract
Artemisinins, a group of plant-derived sesquiterpene lactones, are efficient antimalarial agents. They also share anti-inflammatory and anti-viral activities and were considered for treatment of neurodegenerative disorders like Alzheimer's disease (AD). Additionally, artemisinins bind to gephyrin, the multifunctional scaffold of GABAergic synapses, and modulate inhibitory neurotransmission in vitro. We previously reported an increased expression of gephyrin and GABAA receptors in early pre-symptomatic stages of an AD mouse model (APP-PS1) and in parallel enhanced CDK5-dependent phosphorylation of gephyrin at S270. Here, we studied the effects of artemisinin on gephyrin in the brain of young APP-PS1 mice. We detected an additional increase of gephyrin protein level, elevated gephyrin phosphorylation at Ser270, and an increased amount of GABAAR-γ2 subunits after artemisinin-treatment. Interestingly, the CDK5 activator p35 was also upregulated. Moreover, we demonstrate decreased density of postsynaptic gephyrin and GABAAR-γ2 immunoreactivities in cultured hippocampal neurons expressing gephyrin with alanine mutations at two CDK5 phosphorylation sites. In addition, the activity-dependent modulation of synaptic protein density was abolished in neurons expressing gephyrin lacking one or both of these phosphorylation sites. Thus, our results reveal that artemisinin modulates expression as well as phosphorylation of gephyrin at sites that might have important impact on GABAergic synapses in AD.
Collapse
Affiliation(s)
- Eva Kiss
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany.,Department of Cellular and Molecular Biology, University of Medicine, Pharmacy, Science and Technology "G.E. Palade" of Târgu Mures, Str. Gheorghe Marinescu nr. 38, 540 139Târgu Mureș, Romania
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, 67633 Kaiserslautern, Germany
| | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany
| | - Maret Orlik
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany
| | - Carolin Fischer
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Andrea Schlicksupp
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany
| | - Jochen Kuhse
- Institute of Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, D-69120Heidelberg, Germany
| |
Collapse
|
22
|
Reid HMO, Chen-Mack N, Snowden T, Christie BR. Understanding Changes in Hippocampal Interneurons Subtypes in the Pathogenesis of Alzheimer's Disease: A Systematic Review. Brain Connect 2021; 11:159-179. [PMID: 33559520 DOI: 10.1089/brain.2020.0879] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: It is becoming increasingly recognized that there is significant interneuron degeneration in Alzheimer's disease. As the hippocampus is integral for learning and memory, we performed a systematic review of primary literature focused on the relationship between Alzheimer's and hippocampal interneurons. In this study, we summarize the experimental work performed to date and identify opportunities for future experiments. Objectives: This PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses)-style systematic review seeks to summarize the findings of all accessible research focused on cholecystokinin (CCK), neuropeptide Y (NPY), parvalbumin (PV), and somatostatin (SOM) interneurons in the hippocampal formation. Results: One thousand five hundred ninety-three articles were pulled from PubMed, PsycInfo, and Web of Science, based on three blocks of search terms. There were 45 articles that met all the predetermined inclusion/exclusion criteria. There is strong evidence that PV interneurons are affected early in the disease by toxic amyloid beta (Aβ) fragments; SOM interneurons are affected indirectly while the SOM neuropeptide may act to slowly worsen toxic Aβ fragment accumulation, whereas NPY- and CCK-positive interneurons are affected later in the progression of the disease. Conclusions: Fewer studies have been performed on NPY and CCK interneurons, and there is room for further investigations regarding the role of PV interneurons in Alzheimer's to help resolve contradictory findings. This review found that PV interneurons are affected early in the disease, but only in Alzheimer's precursor protein but not tau models. NPY and CCK interneurons were found to be affected later in the disease, and SOM interneurons vary greatly. Future studies may consider reporting immunohistochemical studies inclusive of either cell location or morphology-as well as marker to give a more robust picture of the disease.
Collapse
Affiliation(s)
- Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Nathan Chen-Mack
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| | - Taylor Snowden
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| |
Collapse
|
23
|
Kim S, Kang M, Park D, Lee AR, Betz H, Ko J, Chang I, Um JW. Impaired formation of high-order gephyrin oligomers underlies gephyrin dysfunction-associated pathologies. iScience 2021; 24:102037. [PMID: 33532714 PMCID: PMC7822942 DOI: 10.1016/j.isci.2021.102037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Gephyrin is critical for the structure, function, and plasticity of inhibitory synapses. Gephyrin mutations have been linked to various neurological disorders; however, systematic analyses of the functional consequences of these mutations are lacking. Here, we performed molecular dynamics simulations of gephyrin to predict how six reported point mutations might change the structural stability and/or function of gephyrin. Additional in silico analyses revealed that the A91T and G375D mutations reduce the binding free energy of gephyrin oligomer formation. Gephyrin A91T and G375D displayed altered clustering patterns in COS-7 cells and nullified the inhibitory synapse-promoting effect of gephyrin in cultured neurons. However, only the G375D mutation reduced gephyrin interaction with GABAA receptors and neuroligin-2 in mouse brain; it also failed to normalize deficits in GABAergic synapse maintenance and neuronal hyperactivity observed in hippocampal dentate gyrus-specific gephyrin-deficient mice. Our results provide insights into biochemical, cell-biological, and network-activity effects of the pathogenic G375D mutation.
Collapse
Affiliation(s)
- Seungjoon Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Mooseok Kang
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.,Core Protein Resources Center, DGIST, Daegu 42988, Korea
| | - Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Ae-Ree Lee
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.,Core Protein Resources Center, DGIST, Daegu 42988, Korea
| | - Heinrich Betz
- Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Iksoo Chang
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.,Core Protein Resources Center, DGIST, Daegu 42988, Korea.,Supercomputing Bigdata Center, DGIST, Daegu 42988, Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea.,Core Protein Resources Center, DGIST, Daegu 42988, Korea
| |
Collapse
|
24
|
Pizzarelli R, Griguoli M, Zacchi P, Petrini EM, Barberis A, Cattaneo A, Cherubini E. Tuning GABAergic Inhibition: Gephyrin Molecular Organization and Functions. Neuroscience 2020; 439:125-136. [PMID: 31356900 PMCID: PMC7351109 DOI: 10.1016/j.neuroscience.2019.07.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 01/15/2023]
Abstract
To be highly reliable, synaptic transmission needs postsynaptic receptors (Rs) in precise apposition to the presynaptic release sites. At inhibitory synapses, the postsynaptic protein gephyrin self-assembles to form a scaffold that anchors glycine and GABAARs to the cytoskeleton, thus ensuring the accurate accumulation of postsynaptic receptors at the right place. This protein undergoes several post-translational modifications which control protein-protein interaction and downstream signaling pathways. In addition, through the constant exchange of scaffolding elements and receptors in and out of synapses, gephyrin dynamically regulates synaptic strength and plasticity. The aim of the present review is to highlight recent findings on the functional role of gephyrin at GABAergic inhibitory synapses. We will discuss different approaches used to interfere with gephyrin in order to unveil its function. In addition, we will focus on the impact of gephyrin structure and distribution at the nanoscale level on the functional properties of inhibitory synapses as well as the implications of this scaffold protein in synaptic plasticity processes. Finally, we will emphasize how gephyrin genetic mutations or alterations in protein expression levels are implicated in several neuropathological disorders, including autism spectrum disorders, schizophrenia, temporal lobe epilepsy and Alzheimer's disease, all associated with severe deficits of GABAergic signaling. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.
Collapse
Affiliation(s)
- Rocco Pizzarelli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Roma, Italy
| | - Marilena Griguoli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Roma, Italy
| | - Paola Zacchi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Enrica Maria Petrini
- Fondazione Istituto Italiano di Tecnologia (IIT), Department of Neuroscience and Brain Technologies, Plasticity of inhibitory networks Unit, Genoa, Italy
| | - Andrea Barberis
- Fondazione Istituto Italiano di Tecnologia (IIT), Department of Neuroscience and Brain Technologies, Plasticity of inhibitory networks Unit, Genoa, Italy
| | - Antonino Cattaneo
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Roma, Italy; Scuola Normale Superiore, Pisa, Italy
| | - Enrico Cherubini
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Roma, Italy; Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy.
| |
Collapse
|
25
|
Hyperexcitable Parvalbumin Interneurons Render Hippocampal Circuitry Vulnerable to Amyloid Beta. iScience 2020; 23:101271. [PMID: 32593000 PMCID: PMC7327841 DOI: 10.1016/j.isci.2020.101271] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/28/2020] [Accepted: 06/10/2020] [Indexed: 01/17/2023] Open
Abstract
Parvalbumin (PV) interneuron dysfunction is associated with various brain disorders, including Alzheimer disease (AD). Here, we asked whether early PV neuron hyperexcitability primes the hippocampus for amyloid beta-induced functional impairment. We show that prolonged chemogenetic activation of PV neurons induces long-term hyperexcitability of these cells, disrupts synaptic transmission, and causes spatial memory deficits on the short-term. On the long-term, pyramidal cells also become hyperexcitable, and synaptic transmission and spatial memory are restored. However, under these conditions of increased excitability of both PV and pyramidal cells, a single low-dose injection of amyloid beta directly into the hippocampus significantly impairs PV neuron function, increases pyramidal neuron excitability, and reduces synaptic transmission, resulting in significant spatial memory deficits. Taken together, our data show that an initial hyperexcitable state of PV neurons renders hippocampal function vulnerable to amyloid beta and may contribute to an increased risk for developing AD. Hyperexcitable hippocampal PV neurons impair spatial memory When excitability of pyramidal neurons also increases, spatial memory is restored This overall network hyperstate is particularly sensitive to amyloid-beta toxicity PV neuron hyperexcitability increases risk for Alzheimer disease
Collapse
|
26
|
Kiss E, Groeneweg F, Gorgas K, Schlicksupp A, Kins S, Kirsch J, Kuhse J. Amyloid-β Fosters p35/CDK5 Signaling Contributing to Changes of Inhibitory Synapses in Early Stages of Cerebral Amyloidosis. J Alzheimers Dis 2020; 74:1167-1187. [DOI: 10.3233/jad-190976] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Eva Kiss
- Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
- Department of Cellular and Molecular Biology, “Emil Palade” University of Medicine, Pharmacy, Science and Technology of Târgu Mureş, Târgu Mureş, Romania
| | - Femke Groeneweg
- Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Andrea Schlicksupp
- Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Jochen Kuhse
- Institute of Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
27
|
Early restoration of parvalbumin interneuron activity prevents memory loss and network hyperexcitability in a mouse model of Alzheimer's disease. Mol Psychiatry 2020; 25:3380-3398. [PMID: 31431685 PMCID: PMC7714697 DOI: 10.1038/s41380-019-0483-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 05/09/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
Abstract
Neuronal network dysfunction is increasingly recognized as an early symptom in Alzheimer's disease (AD) and may provide new entry points for diagnosis and intervention. Here, we show that amyloid-beta-induced hyperexcitability of hippocampal inhibitory parvalbumin (PV) interneurons importantly contributes to neuronal network dysfunction and memory impairment in APP/PS1 mice, a mouse model of increased amyloidosis. We demonstrate that hippocampal PV interneurons become hyperexcitable at ~16 weeks of age, when no changes are observed yet in the intrinsic properties of pyramidal cells. This hyperexcitable state of PV interneurons coincides with increased inhibitory transmission onto hippocampal pyramidal neurons and deficits in spatial learning and memory. We show that treatment aimed at preventing PV interneurons from becoming hyperexcitable is sufficient to restore PV interneuron properties to wild-type levels, reduce inhibitory input onto pyramidal cells, and rescue memory deficits in APP/PS1 mice. Importantly, we demonstrate that early intervention aimed at restoring PV interneuron activity has long-term beneficial effects on memory and hippocampal network activity, and reduces amyloid plaque deposition, a hallmark of AD pathology. Taken together, these findings suggest that early treatment of PV interneuron hyperactivity might be clinically relevant in preventing memory decline and delaying AD progression.
Collapse
|
28
|
Early alterations in hippocampal perisomatic GABAergic synapses and network oscillations in a mouse model of Alzheimer's disease amyloidosis. PLoS One 2019; 14:e0209228. [PMID: 30645585 PMCID: PMC6333398 DOI: 10.1371/journal.pone.0209228] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023] Open
Abstract
Several lines of evidence imply changes in inhibitory interneuron connectivity and subsequent alterations in oscillatory network activities in the pathogenesis of Alzheimer’s Disease (AD). Recently, we provided evidence for an increased immunoreactivity of both the postsynaptic scaffold protein gephyrin and the GABAA receptor γ2-subunit in the hippocampus of young (1 and 3 months of age), APPPS1 mice. These mice represent a well-established model of cerebral amyloidosis, which is a hallmark of human AD. In this study, we demonstrate a robust increase of parvalbumin immunoreactivity and accentuated projections of parvalbumin positive (PV+) interneurons, which target perisomatic regions of pyramidal cells within the hippocampal subregions CA1 and CA3 of 3-month-old APPPS1 mice. Colocalisation studies confirmed a significant increase in the density of PV+ projections labeled with antibodies against a presynaptic (vesicular GABA transporter) and a postsynaptic marker (gephyrin) of inhibitory synapses within the pyramidal cell layer of CA1 and CA3. As perisomatic inhibition by PV+-interneurons is crucial for the generation of hippocampal network oscillations involved in spatial processing, learning and memory formation we investigated the impact of the putative enhanced perisomatic inhibition on two types of fast neuronal network oscillations in acute hippocampal slices: 1. spontaneously occurring sharp wave-ripple complexes (SPW-R), and 2. cholinergic γ-oscillations. Interestingly, both network patterns were generally preserved in APPPS1 mice similar to WT mice. However, the comparison of simultaneous CA3 and CA1 recordings revealed that the incidence and amplitude of SPW-Rs were significantly lower in CA1 vs CA3 in APPPS1 slices, whereas the power of γ-oscillations was significantly higher in CA3 vs CA1 in WT-slices indicating an impaired communication between the CA3 and CA1 network activities in APPPS1 mice. Taken together, our data demonstrate an increased GABAergic synaptic output of PV+ interneurons impinging on pyramidal cells of CA1 and CA3, which might limit the coordinated cross-talk between these two hippocampal areas in young APPPS1 mice and mediate long-term changes in synaptic inhibition during progression of amyloidosis.
Collapse
|
29
|
Lenz M, Galanis C, Kleidonas D, Fellenz M, Deller T, Vlachos A. Denervated mouse dentate granule cells adjust their excitatory but not inhibitory synapses following in vitro entorhinal cortex lesion. Exp Neurol 2018; 312:1-9. [PMID: 30401642 DOI: 10.1016/j.expneurol.2018.10.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/16/2018] [Accepted: 10/23/2018] [Indexed: 01/04/2023]
Abstract
Neurons adjust their synaptic strength in a homeostatic manner following changes in network activity and connectivity. While this form of plasticity has been studied in detail for excitatory synapses, homeostatic plasticity of inhibitory synapses remains not well-understood. In the present study, we employed entorhinal cortex lesion (ECL) of organotypic entorhino-hippocampal tissue cultures to test for homeostatic changes in GABAergic neurotransmission onto partially denervated dentate granule cells. Using single and paired whole-cell patch-clamp recordings, as well as immunostainings for synaptic markers, we find that excitatory synaptic strength is robustly increased 3 days post lesion (dpl), whereas GABAergic neurotransmission is not changed after denervation. Even under conditions of pharmacological inhibition of glutamatergic neurotransmission, which prevents neurons to compensate for the loss of input via excitatory synaptic scaling, down-scaling of GABAergic synapses does not emerge 3 days after denervation. We conclude that granule cells maintain structural and functional properties of GABAergic synapses even in the face of substantial changes in network connectivity. Hence, alterations in inhibitory neurotransmission, as seen in pathological brain states, may not simply reflect a homeostatic response to disconnection.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany; Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Meike Fellenz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany.
| |
Collapse
|
30
|
Groeneweg FL, Trattnig C, Kuhse J, Nawrotzki RA, Kirsch J. Gephyrin: a key regulatory protein of inhibitory synapses and beyond. Histochem Cell Biol 2018; 150:489-508. [DOI: 10.1007/s00418-018-1725-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2018] [Indexed: 12/26/2022]
|
31
|
Amorim JA, Canas PM, Tomé AR, Rolo AP, Agostinho P, Palmeira CM, Cunha RA. Mitochondria in Excitatory and Inhibitory Synapses have Similar Susceptibility to Amyloid-β Peptides Modeling Alzheimer’s Disease. J Alzheimers Dis 2017; 60:525-536. [DOI: 10.3233/jad-170356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- João A. Amorim
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Paula M. Canas
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Angelo R. Tomé
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Anabela P. Rolo
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Paula Agostinho
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Carlos M. Palmeira
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Rodrigo A. Cunha
- CNC – Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|