1
|
Rajavand H, Zalouli V, Nematollahi Z, Fathy-Karkaragh F, Karimigharighi E, Jafarizadeh F, Rabiei Rad A. The Cooperation of Neurogranin with Calmodulin Promotes the Treatment of Aging-Related Diseases via Regular Exercise. Mol Neurobiol 2025:10.1007/s12035-025-04959-6. [PMID: 40285939 DOI: 10.1007/s12035-025-04959-6] [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: 08/02/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Research has demonstrated that engaging in regular exercise has the potential to enhance cognitive function, promote neuroplasticity, and mitigate the likelihood of experiencing cognitive decline. The underlying mechanisms responsible for these effects are intricate and encompass various pathways, including the interaction between neurogranin and calmodulin. The activation of calcium signaling pathways is a significant mechanism through which regular exercise facilitates the treatment of age-related diseases. The activation of neurogranin and calmodulin induced by exercise can provide protection against neurodegeneration by promoting neuronal survival, mitigating oxidative stress, and improving mitochondrial function through the regulation of calcium homeostasis and energy metabolism. In addition, there is evidence suggesting that engaging in regular exercise can lead to an upregulation of neurotrophic factors, specifically brain-derived neurotrophic factor (BDNF). These factors are crucial for the survival of neurons, the plasticity of synapses, and overall cognitive function. Researchers have discovered the involvement of neurogranin in the regulation of BDNF signaling, underscoring its significance in exercise-induced neuroprotection and cognitive enhancement. The current work offers valuable insights into how neurogranin/calmodulin cooperation, facilitated by regular exercise, promotes the treatment of aging-related diseases. The results suggest that regular exercise could enhance memory, learning, synaptic plasticity, and resilience to neurological damage; promote recovery after brain injury; and treat aging-related disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Hosniyeh Rajavand
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Vahideh Zalouli
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Zeinab Nematollahi
- 4UCL Department of Nanotechnology, Division of Surgery and Interventional Science, University College London, London, UK
| | - Farshid Fathy-Karkaragh
- Department of Psychology, Faculty of Educational Sciences and Psychology, University of Tehran, Tehran, Iran
| | - Elham Karimigharighi
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Farzad Jafarizadeh
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Rabiei Rad
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Tokat Gaziosmanpaşa University, Tokat, Turkey.
| |
Collapse
|
2
|
Aderibigbe O, Wood LB, Margulies SS. Cyclosporine A Accelerates Neurorecovery Transcriptional Trajectory in a Swine Model of Diffuse Traumatic Brain Injury. Int J Mol Sci 2025; 26:3531. [PMID: 40331981 PMCID: PMC12026708 DOI: 10.3390/ijms26083531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 05/08/2025] Open
Abstract
Mild traumatic brain injury (mTBI) is a leading cause of morbidity in children with both short- and long-term neurological, cognitive, cerebrovascular, and emotional deficits. These deficits have been attributed to ongoing pathophysiological cascades that occur acutely and persist post-injury. Given our limited understanding of the transcriptional changes associated with these pathophysiological cascades, we studied formalin-fixed paraffin-embedded (FFPE) tissues from the frontal cortex (FC) and the hippocampus + amygdala (H&A) regions of swine (N = 40) after a sagittal rapid non-impact head rotation (RNR). We then sequenced RNA to define transcriptional changes at 1 day and 1 week after injury and investigated the protective influence of cyclosporine A (CsA) treatment. Differentially expressed genes (DEGs) were classified into five temporal patterns (Early, Transient, Persistent, Intensified, Delayed, or Late). DEGs were more abundant at 1 week than 1 day. Shared significant gene ontology annotations in both regions included terms associated with neuronal distress at 1 day and neurorecovery at 1 week. CsA (20 mg/kg/day) infused for 1 day (beginning at 6 h after injury) accelerated 466 DEGs in the FC and 2794 DEGs in the H&A, such that the CsA-treated transcriptional profile was associated with neurorecovery. Overall, our data reveal the effects of anatomic region and elapsed time on gene expression post-mTBI and motivate future studies of CsA treatment.
Collapse
Affiliation(s)
- Oluwagbemisola Aderibigbe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| | - Levi B. Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Susan S. Margulies
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA;
| |
Collapse
|
3
|
Dhauria M, Mondal R, Deb S, Shome G, Chowdhury D, Sarkar S, Benito-León J. Blood-Based Biomarkers in Alzheimer's Disease: Advancing Non-Invasive Diagnostics and Prognostics. Int J Mol Sci 2024; 25:10911. [PMID: 39456697 PMCID: PMC11507237 DOI: 10.3390/ijms252010911] [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: 09/14/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is expected to rise dramatically in incidence due to the global population aging. Traditional diagnostic approaches, such as cerebrospinal fluid analysis and positron emission tomography, are expensive and invasive, limiting their routine clinical use. Recent advances in blood-based biomarkers, including amyloid-beta, phosphorylated tau, and neurofilament light, offer promising non-invasive alternatives for early AD detection and disease monitoring. This review synthesizes current research on these blood-based biomarkers, highlighting their potential to track AD pathology and enhance diagnostic accuracy. Furthermore, this review uniquely integrates recent findings on protein-protein interaction networks and microRNA pathways, exploring novel combinations of proteomic, genomic, and epigenomic biomarkers that provide new insights into AD's molecular mechanisms. Additionally, we discuss the integration of these biomarkers with advanced neuroimaging techniques, emphasizing their potential to revolutionize AD diagnostics. Although large-scale validation is still needed, these biomarkers represent a critical advancement toward more accessible, cost-effective, and early diagnostic tools for AD.
Collapse
Affiliation(s)
| | - Ritwick Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India;
| | - Shramana Deb
- Department of Stroke Medicine, Institute of Neuroscience, Kolkata 700017, India;
| | - Gourav Shome
- Department of Biological Sciences, Bose Institute, Kolkata 700054, India;
| | - Dipanjan Chowdhury
- Department of Internal Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India; (D.C.); (S.S.)
| | - Shramana Sarkar
- Department of Internal Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India; (D.C.); (S.S.)
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, ES-28041 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), ES-28041 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ES-28029 Madrid, Spain
- Department of Medicine, Complutense University, ES-28040 Madrid, Spain
| |
Collapse
|
4
|
Iacobucci GJ, Popescu GK. Calcium- and calmodulin-dependent inhibition of NMDA receptor currents. Biophys J 2024; 123:277-293. [PMID: 38140727 PMCID: PMC10870176 DOI: 10.1016/j.bpj.2023.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
Calcium ions (Ca2+) reduce NMDA receptor currents through several distinct mechanisms. Among these, calmodulin (CaM)-dependent inhibition (CDI) accomplishes rapid, reversible, and incomplete reduction of the NMDA receptor currents in response to elevations in intracellular Ca2+. Quantitative and mechanistic descriptions of CDI of NMDA receptor-mediated signals have been marred by variability originating, in part, from differences in the conditions and metrics used to evaluate this process across laboratories. Recent ratiometric approaches to measure the magnitude and kinetics of NMDA receptor CDI have facilitated rapid insights into this phenomenon. Notably, the kinetics and magnitude of NMDA receptor CDI depend on the degree of saturation of its CaM binding sites, which represent the bona fide calcium sensor for this type of inhibition, the kinetics and magnitude of the Ca2+ signal, which depends on the biophysical properties of the NMDA receptor or of adjacent Ca2+ sources, and on the relative distribution of Ca2+ sources and CaM molecules. Given that all these factors vary widely during development, across cell types, and with physiological and pathological states, it is important to understand how NMDA receptor CDI develops and how it contributes to signaling in the central nervous system. Here, we review briefly these recent advances and highlight remaining questions about the structural and kinetic mechanisms of NMDA receptor CDI. Given that pathologies can arise from several sources, including mutations in the NMDA receptor and in CaM, understanding how CaM responds to intracellular Ca2+ signals to initiate conformational changes in NMDA receptors, and mapping the structural domains responsible will help to envision novel therapeutic strategies to neuropsychiatric diseases, which presently have limited available treatments.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York.
| |
Collapse
|
5
|
Santillán-Morales V, Rodriguez-Espinosa N, Muñoz-Estrada J, Alarcón-Elizalde S, Acebes Á, Benítez-King G. Biomarkers in Alzheimer's Disease: Are Olfactory Neuronal Precursors Useful for Antemortem Biomarker Research? Brain Sci 2024; 14:46. [PMID: 38248261 PMCID: PMC10813897 DOI: 10.3390/brainsci14010046] [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: 11/16/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD), as the main cause of dementia, affects millions of people around the world, whose diagnosis is based mainly on clinical criteria. Unfortunately, the diagnosis is obtained very late, when the neurodegenerative damage is significant for most patients. Therefore, the exhaustive study of biomarkers is indispensable for diagnostic, prognostic, and even follow-up support. AD is a multifactorial disease, and knowing its underlying pathological mechanisms is crucial to propose new and valuable biomarkers. In this review, we summarize some of the main biomarkers described in AD, which have been evaluated mainly by imaging studies in cerebrospinal fluid and blood samples. Furthermore, we describe and propose neuronal precursors derived from the olfactory neuroepithelium as a potential resource to evaluate some of the widely known biomarkers of AD and to gear toward searching for new biomarkers. These neuronal lineage cells, which can be obtained directly from patients through a non-invasive and outpatient procedure, display several characteristics that validate them as a surrogate model to study the central nervous system, allowing the analysis of AD pathophysiological processes. Moreover, the ease of obtaining and harvesting endows them as an accessible and powerful resource to evaluate biomarkers in clinical practice.
Collapse
Affiliation(s)
- Valeria Santillán-Morales
- Laboratory of Neuropharmacology, Clinical Research, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (V.S.-M.); (S.A.-E.)
| | - Norberto Rodriguez-Espinosa
- Department of Neurology, University Hospital Nuestra Señora de Candelaria, 38010 Tenerife, Spain;
- Department of Internal Medicine, Dermatology and Psychiatry, Faculty of Health Sciences, University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Jesús Muñoz-Estrada
- Department of Computational Biomedicine, Cedars Sinai Medical Center, Los Angeles, CA 90069, USA;
| | - Salvador Alarcón-Elizalde
- Laboratory of Neuropharmacology, Clinical Research, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (V.S.-M.); (S.A.-E.)
| | - Ángel Acebes
- Department of Basic Medical Sciences, Institute of Biomedical Technologies (ITB), University of La Laguna (ULL), 38200 Tenerife, Spain
| | - Gloria Benítez-King
- Laboratory of Neuropharmacology, Clinical Research, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico; (V.S.-M.); (S.A.-E.)
| |
Collapse
|
6
|
Rodriguez-Jimenez FJ, Ureña-Peralta J, Jendelova P, Erceg S. Alzheimer's disease and synapse Loss: What can we learn from induced pluripotent stem Cells? J Adv Res 2023; 54:105-118. [PMID: 36646419 DOI: 10.1016/j.jare.2023.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/21/2022] [Accepted: 01/08/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Synaptic dysfunction is a major contributor to Alzheimeŕs disease (AD) pathogenesis in addition to the formation of neuritic β-amyloid plaques and neurofibrillary tangles of hyperphosphorylated Tau protein. However, how these features contribute to synaptic dysfunction and axonal loss remains unclear. While years of considerable effort have been devoted to gaining an improved understanding of this devastating disease, the unavailability of patient-derived tissues, considerable genetic heterogeneity, and lack of animal models that faithfully recapitulate human AD have hampered the development of effective treatment options. Ongoing progress in human induced pluripotent stem cell (hiPSC) technology has permitted the derivation of patient- and disease-specific stem cells with unlimited self-renewal capacity. These cells can differentiate into AD-affected cell types, which support studies of disease mechanisms, drug discovery, and the development of cell replacement therapies in traditional and advanced cell culture models. AIM OF REVIEW To summarize current hiPSC-based AD models, highlighting the associated achievements and challenges with a primary focus on neuron and synapse loss. KEY SCIENTIFIC CONCEPTS OF REVIEW We aim to identify how hiPSC models can contribute to understanding AD-associated synaptic dysfunction and axonal loss. hiPSC-derived neural cells, astrocytes, and microglia, as well as more sophisticated cellular organoids, may represent reliable models to investigate AD and identify early markers of AD-associated neural degeneration.
Collapse
Affiliation(s)
- Francisco Javier Rodriguez-Jimenez
- Stem Cell Therapies in Neurodegenerative Diseases Lab., Centro de Investigación Principe Felipe (CIPF), c/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - Juan Ureña-Peralta
- Stem Cell Therapies in Neurodegenerative Diseases Lab., Centro de Investigación Principe Felipe (CIPF), c/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - Pavla Jendelova
- Institute of Experimental Medicine, Department of Neuroregeneration, Czech Academy of Science, Prague, Czech Republic.
| | - Slaven Erceg
- Stem Cell Therapies in Neurodegenerative Diseases Lab., Centro de Investigación Principe Felipe (CIPF), c/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain; Institute of Experimental Medicine, Department of Neuroregeneration, Czech Academy of Science, Prague, Czech Republic; National Stem Cell Bank-Valencia Node, Centro de Investigacion Principe Felipe, c/ Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
7
|
Loeffler DA. Antibody-Mediated Clearance of Brain Amyloid-β: Mechanisms of Action, Effects of Natural and Monoclonal Anti-Aβ Antibodies, and Downstream Effects. J Alzheimers Dis Rep 2023; 7:873-899. [PMID: 37662616 PMCID: PMC10473157 DOI: 10.3233/adr-230025] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023] Open
Abstract
Immunotherapeutic efforts to slow the clinical progression of Alzheimer's disease (AD) by lowering brain amyloid-β (Aβ) have included Aβ vaccination, intravenous immunoglobulin (IVIG) products, and anti-Aβ monoclonal antibodies. Neither Aβ vaccination nor IVIG slowed disease progression. Despite conflicting phase III results, the monoclonal antibody Aducanumab received Food and Drug Administration (FDA) approval for treatment of AD in June 2021. The only treatments unequivocally demonstrated to slow AD progression to date are the monoclonal antibodies Lecanemab and Donanemab. Lecanemab received FDA approval in January 2023 based on phase II results showing lowering of PET-detectable Aβ; phase III results released at that time indicated slowing of disease progression. Topline results released in May 2023 for Donanemab's phase III trial revealed that primary and secondary end points had been met. Antibody binding to Aβ facilitates its clearance from the brain via multiple mechanisms including promoting its microglial phagocytosis, activating complement, dissolving fibrillar Aβ, and binding of antibody-Aβ complexes to blood-brain barrier receptors. Antibody binding to Aβ in peripheral blood may also promote cerebral efflux of Aβ by a peripheral sink mechanism. According to the amyloid hypothesis, for Aβ targeting to slow AD progression, it must decrease downstream neuropathological processes including tau aggregation and phosphorylation and (possibly) inflammation and oxidative stress. This review discusses antibody-mediated mechanisms of Aβ clearance, findings in AD trials involving Aβ vaccination, IVIG, and anti-Aβ monoclonal antibodies, downstream effects reported in those trials, and approaches which might improve the Aβ-clearing ability of monoclonal antibodies.
Collapse
Affiliation(s)
- David A. Loeffler
- Beaumont Research Institute, Department of Neurology, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
8
|
Zhang Y, Chen H, Li R, Sterling K, Song W. Amyloid β-based therapy for Alzheimer's disease: challenges, successes and future. Signal Transduct Target Ther 2023; 8:248. [PMID: 37386015 PMCID: PMC10310781 DOI: 10.1038/s41392-023-01484-7] [Citation(s) in RCA: 341] [Impact Index Per Article: 170.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 07/01/2023] Open
Abstract
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
Collapse
Affiliation(s)
- Yun Zhang
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Huaqiu Chen
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Larosa A, Wong TP. The hippocampus in stress susceptibility and resilience: Reviewing molecular and functional markers. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110601. [PMID: 35842073 DOI: 10.1016/j.pnpbp.2022.110601] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/22/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Understanding the individual variability that comes with the likelihood of developing stress-related psychopathologies is of paramount importance when addressing mechanisms of their neurobiology. This article focuses on the hippocampus as a region that is highly influenced by chronic stress exposure and that has strong ties to the development of related disorders, such as depression and post-traumatic stress disorder. We first outline three commonly used animal models that have been used to separate animals into susceptible and resilient cohorts. Next, we review molecular and functional hippocampal markers of susceptibility and resilience. We propose that the hippocampus plays a crucial role in the differences in the processing and storage of stress-related information in animals with different stress susceptibilities. These hippocampal markers not only help us attain a more comprehensive understanding of the various facets of stress-related pathophysiology, but also could be targeted for the development of new treatments.
Collapse
Affiliation(s)
- Amanda Larosa
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Dept. of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
10
|
Dulewicz M, Kulczyńska-Przybik A, Słowik A, Borawska R, Mroczko B. Neurogranin and Neuronal Pentraxin Receptor as Synaptic Dysfunction Biomarkers in Alzheimer's Disease. J Clin Med 2021; 10:jcm10194575. [PMID: 34640593 PMCID: PMC8509697 DOI: 10.3390/jcm10194575] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 12/24/2022] Open
Abstract
Synaptic loss and dysfunction are one of the earliest signs of neurodegeneration associated with cognitive decline in Alzheimer’s disease (AD). It seems that by assessing proteins related to synapses, one may reflect their dysfunction and improve the understanding of neurobiological processes in the early stage of the disease. To our best knowledge, this is the first study that analyzes the CSF concentrations of two synaptic proteins together, such as neurogranin (Ng) and neuronal pentraxins receptor (NPTXR) in relation to neurochemical dementia biomarkers in Alzheimer’s disease. Methods: Ng, NPTXR and classical AD biomarkers concentrations were measured in the CSF of patients with AD and non-demented controls (CTRL) using an enzyme-linked immunosorbent assay (ELISA) and Luminex xMAP technology. Results: The CSF level of Ng was significantly higher, whereas the NPTXR was significantly lower in the AD patients than in cognitively healthy controls. As a first, we calculated the NPTXR/Ng ratio as an indicator of synaptic disturbance. The patients with AD presented a significantly decreased NPTXR/Ng ratio. The correlation was observed between both proteins in the AD and the whole study group. Furthermore, the relationship between the Ng level and pTau181 was found in the AD group of patients. Conclusions: The Ng and NPTXR concentrations in CSF are promising synaptic dysfunction biomarkers reflecting pathological changes in AD.
Collapse
Affiliation(s)
- Maciej Dulewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (R.B.); (B.M.)
- Correspondence:
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (R.B.); (B.M.)
| | - Agnieszka Słowik
- Department of Neurology, Jagiellonian University, 30-688 Krakow, Poland;
| | - Renata Borawska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (R.B.); (B.M.)
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.-P.); (R.B.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
11
|
The Relevance of Amyloid β-Calmodulin Complexation in Neurons and Brain Degeneration in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22094976. [PMID: 34067061 PMCID: PMC8125740 DOI: 10.3390/ijms22094976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Intraneuronal amyloid β (Aβ) oligomer accumulation precedes the appearance of amyloid plaques or neurofibrillary tangles and is neurotoxic. In Alzheimer’s disease (AD)-affected brains, intraneuronal Aβ oligomers can derive from Aβ peptide production within the neuron and, also, from vicinal neurons or reactive glial cells. Calcium homeostasis dysregulation and neuronal excitability alterations are widely accepted to play a key role in Aβ neurotoxicity in AD. However, the identification of primary Aβ-target proteins, in which functional impairment initiating cytosolic calcium homeostasis dysregulation and the critical point of no return are still pending issues. The micromolar concentration of calmodulin (CaM) in neurons and its high affinity for neurotoxic Aβ peptides (dissociation constant ≈ 1 nM) highlight a novel function of CaM, i.e., the buffering of free Aβ concentrations in the low nanomolar range. In turn, the concentration of Aβ-CaM complexes within neurons will increase as a function of time after the induction of Aβ production, and free Aβ will rise sharply when accumulated Aβ exceeds all available CaM. Thus, Aβ-CaM complexation could also play a major role in neuronal calcium signaling mediated by calmodulin-binding proteins by Aβ; a point that has been overlooked until now. In this review, we address the implications of Aβ-CaM complexation in the formation of neurotoxic Aβ oligomers, in the alteration of intracellular calcium homeostasis induced by Aβ, and of dysregulation of the calcium-dependent neuronal activity and excitability induced by Aβ.
Collapse
|
12
|
Pereira JB, Janelidze S, Ossenkoppele R, Kvartsberg H, Brinkmalm A, Mattsson-Carlgren N, Stomrud E, Smith R, Zetterberg H, Blennow K, Hansson O. Untangling the association of amyloid-β and tau with synaptic and axonal loss in Alzheimer's disease. Brain 2021; 144:310-324. [PMID: 33279949 PMCID: PMC8210638 DOI: 10.1093/brain/awaa395] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/04/2020] [Accepted: 09/21/2020] [Indexed: 01/10/2023] Open
Abstract
It is currently unclear how amyloid-β and tau deposition are linked to changes in
synaptic function and axonal structure over the course of Alzheimer’s disease. Here, we
assessed these relationships by measuring presynaptic (synaptosomal-associated protein 25,
SNAP25; growth-associated protein 43, GAP43), postsynaptic (neurogranin, NRGN) and axonal
(neurofilament light chain) markers in the CSF of individuals with varying levels of
amyloid-β and tau pathology based on 18F-flutemetamol PET and
18F-flortaucipir PET. In addition, we explored the relationships between
synaptic and axonal markers with cognition as well as functional and anatomical brain
connectivity markers derived from resting-state functional MRI and diffusion tensor
imaging. We found that the presynaptic and postsynaptic markers SNAP25, GAP43 and NRGN are
elevated in early Alzheimer’s disease i.e. in amyloid-β-positive individuals without
evidence of tau pathology. These markers were associated with greater amyloid-β pathology,
worse memory and functional changes in the default mode network. In contrast,
neurofilament light chain was abnormal in later disease stages, i.e. in individuals with
both amyloid-β and tau pathology, and correlated with more tau and worse global cognition.
Altogether, these findings support the hypothesis that amyloid-β and tau might have
differential downstream effects on synaptic and axonal function in a stage-dependent
manner, with amyloid-related synaptic changes occurring first, followed by tau-related
axonal degeneration.
Collapse
Affiliation(s)
- Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology and Alzheimer Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Hlin Kvartsberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Wanderlind ML, Gonçalves R, Tomasi CD, Dal-Pizzol F, Ritter C. Association of neurogranin with delirium among critically ill patients. Biomark Med 2020; 14:1613-1617. [PMID: 33336596 DOI: 10.2217/bmm-2020-0328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Neurogranin (Ng) concentrates at dendritic spines. In patients with Alzheimer disease Ng levels are elevated. The role of Ng in delirium development has not been assessed, therefore we hypothesized that Ng levels are associated with delirium in critically ill patients. Materials & methods: From 94 critically ill patients, 47 developed delirium and 47 controls were included. Blood was collected during the first 24 h of intensive care unit (ICU) admission, and on the day of delirium diagnoses. Ng and IL-1β were determined. Results: Ng and IL-1β levels were higher in the delirium group at ICU admission and on the day of delirium diagnoses. IL-1β and Ng were independently associated with delirium occurrence. Conclusion: Ng levels are associated with delirium development in ICU patients.
Collapse
Affiliation(s)
- Márcia Lz Wanderlind
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Renata Gonçalves
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Cristiane D Tomasi
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil
| | - Cristiane Ritter
- Laboratório de Fisiopatologia Experimental Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil.,Intensive Care Unit, Hospital São José, Criciúma, SC, Brazil
| |
Collapse
|
14
|
Niu H, Xie R, Li L, Zhang X, Wei X, Deng J, Li Z. WITHDRAWN: Resveratrol partially prevents learning and memory deficits in rats exposed to gaseous formaldehyde. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020:S1382-6689(20)30179-4. [PMID: 32976996 DOI: 10.1016/j.etap.2020.103503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Huatao Niu
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, China
| | - Ran Xie
- Department of PET-CT, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, China
| | - Lanjiang Li
- College of Forensic Medicine, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Xiang Zhang
- Experimental Demonstration Center, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Xiaohan Wei
- Department of Human Anatomy and Tissue Embryology, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Jie Deng
- Department of Human Anatomy and Tissue Embryology, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China
| | - Zhongming Li
- Department of Human Anatomy and Tissue Embryology, College of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650031, China.
| |
Collapse
|
15
|
Li L, Lai M, Cole S, Le Novère N, Edelstein SJ. Neurogranin stimulates Ca2+/calmodulin-dependent kinase II by suppressing calcineurin activity at specific calcium spike frequencies. PLoS Comput Biol 2020; 16:e1006991. [PMID: 32049957 PMCID: PMC7041932 DOI: 10.1371/journal.pcbi.1006991] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 02/25/2020] [Accepted: 11/18/2019] [Indexed: 11/18/2022] Open
Abstract
Calmodulin sits at the center of molecular mechanisms underlying learning and memory. Its complex and sometimes opposite influences, mediated via the binding to various proteins, are yet to be fully understood. Calcium/calmodulin-dependent protein kinase II (CaMKII) and calcineurin (CaN) both bind open calmodulin, favoring Long-Term Potentiation (LTP) or Depression (LTD) respectively. Neurogranin binds to the closed conformation of calmodulin and its impact on synaptic plasticity is less clear. We set up a mechanistic computational model based on allosteric principles to simulate calmodulin state transitions and its interactions with calcium ions and the three binding partners mentioned above. We simulated calcium spikes at various frequencies and show that neurogranin regulates synaptic plasticity along three modalities. At low spike frequencies, neurogranin inhibits the onset of LTD by limiting CaN activation. At intermediate frequencies, neurogranin facilitates LTD, but limits LTP by precluding binding of CaMKII with calmodulin. Finally, at high spike frequencies, neurogranin promotes LTP by enhancing CaMKII autophosphorylation. While neurogranin might act as a calmodulin buffer, it does not significantly preclude the calmodulin opening by calcium. On the contrary, neurogranin synchronizes the opening of calmodulin's two lobes and promotes their activation at specific frequencies. Neurogranin suppresses basal CaN activity, thus increasing the chance of CaMKII trans-autophosphorylation at high-frequency calcium spikes. Taken together, our study reveals dynamic regulatory roles played by neurogranin on synaptic plasticity, which provide mechanistic explanations for opposing experimental findings.
Collapse
Affiliation(s)
- Lu Li
- Babraham Institute, Cambridge, United Kingdom
| | - Massimo Lai
- Quantitative Systems Pharmacology, CERTARA, Canterbury, United Kingdom
| | - Stephen Cole
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
16
|
Zhong L, Gerges NZ. Neurogranin Regulates Metaplasticity. Front Mol Neurosci 2020; 12:322. [PMID: 32038160 PMCID: PMC6992556 DOI: 10.3389/fnmol.2019.00322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/17/2019] [Indexed: 01/14/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are two major forms of synaptic plasticity that are widely accepted as cellular mechanisms involved in learning and memory. Metaplasticity is a process whereby modifications in synaptic processes shift the threshold for subsequent plasticity. While metaplasticity has been functionally observed, its molecular basis is not well understood. Here, we report that neurogranin (Ng) regulates metaplasticity by shifting the threshold toward potentiation, i.e., increasing Ng in hippocampal neurons lowers the threshold for LTP and augments the threshold for LTD. We also show that Ng does not change the ultrastructural localization of calmodulin (CaM)-dependent protein Kinase II (CaMKII) or calcineurin, critical enzymes for the induction of LTP and LTD, respectively. Interestingly, while CaMKII concentrates close to the plasma membrane, calcineurin concentrates away from the plasma membrane. These data, along with the previous observation showing Ng targets CaM closer to the plasma membrane, suggesting that shifting the localization of CaM within the dendritic spines and closer to the plasma membrane, where there is more CaMKII, may be favoring the activation of CaMKII vs. that of calcineurin. Thus, the regulation of CaM localization/targeting within dendritic spines by Ng may provide a mechanistic basis for the regulation of metaplasticity.
Collapse
Affiliation(s)
| | - Nashaat Z. Gerges
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
17
|
Yilmaz A, Fuchs D, Price RW, Spudich S, Blennow K, Zetterberg H, Gisslén M. Cerebrospinal Fluid Concentrations of the Synaptic Marker Neurogranin in Neuro-HIV and Other Neurological Disorders. Curr HIV/AIDS Rep 2020; 16:76-81. [PMID: 30649659 PMCID: PMC6420419 DOI: 10.1007/s11904-019-00420-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Purpose of Review The aim of this study was to examine the synaptic biomarker neurogranin in cerebrospinal fluid (CSF) in different stages of HIV infection and in relation to what is known about CSF neurogranin in other neurodegenerative diseases. Recent Findings CSF concentrations of neurogranin are increased in Alzheimer’s disease, but not in other neurodegenerative disorder such as Parkinson’s disease, frontotemporal dementia, and Lewy body dementia. Adults with HIV-associated dementia have been found to have decreased levels of neurogranin in the frontal cortex, which at least to some extent, may be mediated by the proinflammatory cytokines IL-1β and IL-8. Summary CSF neurogranin concentrations were in the same range for all groups of HIV-infected individuals and uninfected controls. This either indicates that synaptic injury is not an important part of HIV neuropathogenesis or that CSF neurogranin is not sensitive to the type of synaptic impairment present in HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Aylin Yilmaz
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 416 45, Gothenburg, Sweden
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Richard W Price
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Serena Spudich
- Department of Neurology, Yale University, New Haven, CT, USA
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Molndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 416 45, Gothenburg, Sweden.
| |
Collapse
|
18
|
Huang Y, Wang J, Zhu B, Fu P. CSF VEGF Was Positively Associated with Neurogranin Independent of β-Amyloid Pathology. Neuropsychiatr Dis Treat 2020; 16:1737-1744. [PMID: 32801708 PMCID: PMC7383019 DOI: 10.2147/ndt.s252008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/21/2020] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Increasing evidence suggests that both vascular endothelial growth factor (VEGF) and synaptic failure have been involved in the pathogenesis of Alzheimer's disease (AD). However, it is not clear whether CSF VEGF levels are associated with synaptic function in living human. PATIENTS AND METHODS In the present study, we included a total of 291 older individuals, including 83 individuals with normal cognition (NC), 143 individuals with mild cognitive impairment (MCI) and 65 patients with AD. Several linear regression models were conducted to examine the associations of CSF VEGF with CSF neurogranin levels (NG, reflecting synaptic degeneration) when controlling for other potential confounding factors, including age, gender, years of education, clinical diagnosis, APOE4 genotype and CSF β-amyloid 42 (Aβ 42) levels. RESULTS There was no significant difference in VEGF levels between the three diagnostic groups. In the pooled sample, females had significantly lower levels of VEGF than males. Aβ-positive (CSF Aβ 42 < 192 pg/mL) individuals had lower levels of VEGF than Aβ-negative individuals. However, the relationships between VEGF and NG levels were not modified by disease stage. Finally, we found that CSF VEGF levels were associated with NG levels with adjustment of age, gender, years of education, clinical diagnosis, APOE4 genotype and CSF Aβ 42 levels. CONCLUSION CSF VEGF levels were associated with NG independent of AD pathology and disease stage.
Collapse
Affiliation(s)
- Yangping Huang
- Department of Neurology, Taizhou First People's Hospital, Taizhou, Zhejiang, People's Republic of China
| | - Jun Wang
- Department of Neurology, Taizhou First People's Hospital, Taizhou, Zhejiang, People's Republic of China
| | - Bihong Zhu
- Department of Neurology, Taizhou First People's Hospital, Taizhou, Zhejiang, People's Republic of China
| | - Pan Fu
- Department of Neurology, Taizhou First People's Hospital, Taizhou, Zhejiang, People's Republic of China
| |
Collapse
|
19
|
Garrido-García A, de Andrés R, Jiménez-Pompa A, Soriano P, Sanz-Fuentes D, Martínez-Blanco E, Díez-Guerra FJ. Neurogranin Expression Is Regulated by Synaptic Activity and Promotes Synaptogenesis in Cultured Hippocampal Neurons. Mol Neurobiol 2019; 56:7321-7337. [PMID: 31020616 DOI: 10.1007/s12035-019-1593-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/02/2019] [Indexed: 01/09/2023]
Abstract
Neurogranin (Ng) is a calmodulin (CaM)-binding protein that is phosphorylated by protein kinase C (PKC) and is highly enriched in the dendrites and spines of telencephalic neurons. It is proposed to be involved in regulating CaM availability in the post-synaptic environment to modulate the efficiency of excitatory synaptic transmission. There is a close relationship between Ng and cognitive performance; its expression peaks in the forebrain coinciding with maximum synaptogenic activity, and it is reduced in several conditions of impaired cognition. We studied the expression of Ng in cultured hippocampal neurons and found that both protein and mRNA levels were about 10% of that found in the adult hippocampus. Long-term blockade of NMDA receptors substantially decreased Ng expression. On the other hand, treatments that enhanced synaptic activity such as long-term bicuculline treatment or co-culture with glial cells or cholesterol increased Ng expression. Chemical long-term potentiation (cLTP) induced an initial drop of Ng, with a minimum after 15 min followed by a slow recovery during the next 2-4 h. This effect was most evident in the synaptosome-enriched fraction, thus suggesting local synthesis in dendrites. Lentiviral expression of Ng led to increased density of both excitatory and inhibitory synapses in the second and third weeks of culture. These results indicate that Ng expression is regulated by synaptic activity and that Ng promotes the synaptogenesis process. Given its relationship with cognitive function, we propose targeting of Ng expression as a promising strategy to prevent or alleviate the cognitive deficits associated with aging and neuropathological conditions.
Collapse
Affiliation(s)
- Alberto Garrido-García
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto Cajal (CSIC), Av. Doctor Arce, 37, 28002, Madrid, Spain
| | - Raquel de Andrés
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Amanda Jiménez-Pompa
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patricia Soriano
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Diego Sanz-Fuentes
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Elena Martínez-Blanco
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - F Javier Díez-Guerra
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049, Madrid, Spain.
- Laboratory of Neuronal Plasticity, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain.
| |
Collapse
|
20
|
Association of cerebrospinal fluid Neurogranin with Alzheimer's disease. Aging Clin Exp Res 2019; 31:185-191. [PMID: 29667155 DOI: 10.1007/s40520-018-0948-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/07/2018] [Indexed: 01/07/2023]
Abstract
Cerebrospinal fluid (CSF) Neurogranin has recently been proposed as a potential biomarker for cognitive decline and brain injury in Alzheimer's disease (AD). To test whether CSF Neurogranin levels are increased in AD and its association with cognitive decline, we examined 99 cognitively normal (CN) subjects, 171 patients with mild cognitive impairment (MCI), and 81 patients with AD in the cross-sectional study from the Alzheimer's Disease Neuroimaging Initiative (ADNI). The results showed that CSF Neurogranin was increased in both AD and MCI compared with controls. CSF Neurogranin was particularly high in patients with MCI and AD dementia with Aβ pathologic features. Neurogranin levels were significantly higher in females compared to males with MCI. Levels of Neurogranin between the males and females with AD and CN did not differ. Neurogranin levels were significantly higher in APOE ε4 carriers compared to APOE ε4 non-carriers with MCI. Levels of Neurogranin between the APOE ε4 carriers and APOE ε4 non-carriers with AD and CN did not differ. Elevated CSF Neurogranin levels were positively correlated with levels of total tau and P-tau in AD. The results indicated that CSF Neurogranin was increased at the prodromal stage of AD and might reflect synaptic injury as cognitive decline in AD.
Collapse
|
21
|
Ye X, Zhou W, Zhang J. Association of CSF CD40 levels and synaptic degeneration across the Alzheimer's disease spectrum. Neurosci Lett 2018; 694:41-45. [PMID: 30447377 DOI: 10.1016/j.neulet.2018.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/01/2018] [Accepted: 11/13/2018] [Indexed: 01/09/2023]
Abstract
The CD40 pathway has been implicated in microglial activation, which is considered as a key factor in the pathogenesis of Alzheimer's disease (AD). However, the association of CSF CD40 and synaptic degeneration in living human is not clear. A total of 294 subjects with different severities of cognitive impairments were included in this study: 84 participants with normal cognition, 143 patients with mild cognitive impairment (MCI) and 67 patients with mild AD. Levels of CD40 in CSF were compared among the three groups. Further, several linear regression models were conducted to explore the associations of CSF CD40 and neurogranin levels (reflecting synaptic degeneration) when controlling for age, gender, educational attainment, APOE4 genotype, clinical diagnosis, CSF Aβ42 and tau proteins. We found that CSF CD40 levels were significantly decreased in patients with mild AD compared with healthy controls and MCI patients (control vs. AD, p = 0.0026; MCI vs. AD, p = 0.0268). However, there were no significant differences in CSF CD40 levels between controls and patients with MCI (p = 0.37). In addition, CSF CD40 levels were associated with neurogranin in the pooled sample when controlling for age, gender, educational attainment, APOE4 genotype and diagnosis. In summary, our findings support the notion that the CD40 pathway may contribute to an important mechanism underlying synaptic degeneration in AD.
Collapse
Affiliation(s)
- Xinwu Ye
- Wenzhou Seventh People's Hospital, Zhejiang, China.
| | - Wenjun Zhou
- Department of Pathology, Hangzhou Normal University, College of Medicine, Zhejiang, China
| | - Jie Zhang
- Independent Researcher, 25 Xuezheng road, Xiasha District, Zhejiang, 310018, China.
| |
Collapse
|
22
|
Casaletto KB, Elahi FM, Bettcher BM, Neuhaus J, Bendlin BB, Asthana S, Johnson SC, Yaffe K, Carlsson C, Blennow K, Zetterberg H, Kramer JH. Neurogranin, a synaptic protein, is associated with memory independent of Alzheimer biomarkers. Neurology 2017; 89:1782-1788. [PMID: 28939668 DOI: 10.1212/wnl.0000000000004569] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 08/07/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To determine the association between synaptic functioning as measured via neurogranin in CSF and cognition relative to established Alzheimer disease (AD) biomarkers in neurologically healthy older adults. METHODS We analyzed CSF concentrations of neurogranin, β-amyloid (Aβ42), phosphorylated tau (p-tau), and total tau (t-tau) among 132 neurologically normal older adults (mean 64.5, range 55-85), along with bilateral hippocampal volumes and a measure of episodic memory (Auditory Verbal Learning Test, delayed recall). Univariable analyses examined the relationship between neurogranin and the other AD-related biomarkers. Multivariable regression models examined the relationship between neurogranin and delayed recall, adjusting for age and sex, and interaction terms (neurogranin × AD biomarkers). RESULTS Higher neurogranin concentrations were associated with older age (ρ = 0.20, p = 0.02), lower levels of p-tau and t-tau, and smaller hippocampal volumes (p < 0.03), but not with CSF Aβ42 (p = 0.18). In addition, CSF neurogranin demonstrated a significant relationship with memory performance independent of the AD-related biomarkers; individuals with the lowest CSF neurogranin concentrations performed better on delayed recall than those with medium or high CSF neurogranin concentrations (p < 0.01). Notably, CSF p-tau, t-tau, and Aβ42 and hippocampal volumes were not significantly associated with delayed recall scores (p > 0.40), and did not interact with neurogranin to predict memory (p > 0.10). CONCLUSIONS Synaptic dysfunction (assessed via neurogranin) may be an early pathologic process in age-related neurodegeneration, and a sensitive marker of age-related cognitive abilities, potentially preceding or even acting independently from AD pathogenesis. Synaptic functioning may be a useful early marker of cognitive aging and possibly a target for future brain aging interventions.
Collapse
Affiliation(s)
- Kaitlin B Casaletto
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK.
| | - Fanny M Elahi
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Brianne M Bettcher
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - John Neuhaus
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Barbara B Bendlin
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Sanjay Asthana
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Sterling C Johnson
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kristine Yaffe
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Cynthia Carlsson
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Henrik Zetterberg
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| | - Joel H Kramer
- From the Memory and Aging Center (K.B.C., F.M.E., J.N., K.Y., J.H.K.), Department of Neurology, University of California, San Francisco; Denver Anschutz Medical Center (B.M.B., C.C.), University of Colorado, Aurora; Wisconsin Alzheimer's Disease Research Center (B.B.B., S.C.J.) and Department of Medicine (S.A.), University of Wisconsin School of Medicine and Public Health; School of Medicine and Public Health (B.B.B., S.C.J.), Wisconsin Alzheimer's Institute; Geriatric Research Education and Clinical Center (S.C.J., C.C.), William S. Middleton Memorial Veterans Hospital, Madison, WI; San Francisco Veterans Affairs Hospital (K.Y.), CA; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology (K.B., H.Z.), and Clinical Neurochemistry Laboratory (H.Z.), University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden; and Department of Molecular Neuroscience (H.Z.), UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
23
|
Lista S, Hampel H. Synaptic degeneration and neurogranin in the pathophysiology of Alzheimer’s disease. Expert Rev Neurother 2016; 17:47-57. [DOI: 10.1080/14737175.2016.1204234] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Simone Lista
- AXA Research Fund & UPMC Chair, Paris, France
- IHU-A-ICM – Paris Institute of Translational Neurosciences, Pitié-Salpêtrière University Hospital, Paris, France
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Paris, France
- Department of Neurology, Sorbonne Universities, Institute of Memory and Alzheimer’s Disease (IM2A) & Brain and Spine Institute (ICM) UMR S 1127, Pitié-Salpêtrière University Hospital, Pierre and Marie Curie University, Paris 06, Paris, France
| |
Collapse
|
24
|
Kaleka KS, Gerges NZ. Neurogranin restores amyloid β-mediated synaptic transmission and long-term potentiation deficits. Exp Neurol 2015; 277:115-123. [PMID: 26721336 DOI: 10.1016/j.expneurol.2015.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 12/08/2015] [Accepted: 12/18/2015] [Indexed: 11/16/2022]
Abstract
Amyloid β (Aβ) is widely considered one of the early causes of cognitive deficits observed in Alzheimer's disease. Many of the deficits caused by Aβ are attributed to its disruption of synaptic function represented by its blockade of long-term potentiation (LTP) and its induction of synaptic depression. Identifying pathways that reverse these synaptic deficits may open the door to new therapeutic targets. In this study, we explored the possibility that Neurogranin (Ng)-a postsynaptic calmodulin (CaM) targeting protein that enhances synaptic function-may rescue Aβ-mediated deficits in synaptic function. Our results show that Ng is able to reverse synaptic depression and LTP deficits induced by Aβ. Furthermore, Ng's restoration of synaptic transmission is through the insertion of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPARs). These restorative effects of Ng are dependent on the interaction of Ng and CaM and CaM-dependent activation of CaMKII. Overall, this study identifies a novel mechanism to rescue synaptic deficits induced by Aβ oligomers. It also suggests Ng and CaM signaling as potential therapeutic targets for Alzheimer's disease as well as important tools to further explore the pathophysiology underlying the disease.
Collapse
Affiliation(s)
- Kanwardeep Singh Kaleka
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States
| | - Nashaat Z Gerges
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States.
| |
Collapse
|
25
|
Abstract
Increasing plasticity in neurons of the prefrontal cortex (PFC) has been proposed as a possible therapeutic tool to enhance extinction, a process that is impaired in post-traumatic stress disorder, schizophrenia, and addiction. To test this hypothesis, we generated transgenic mice that overexpress neurogranin (a calmodulin-binding protein that facilitates long-term potentiation) in the PFC. Neurogranin overexpression in the PFC enhanced long-term potentiation and increased the rates of extinction learning of both fear conditioning and sucrose self-administration. Our results indicate that elevated neurogranin function within the PFC can enhance local plasticity and increase the rate of extinction learning across different behavioral tasks. Thus, neurogranin can provide a molecular link between enhanced plasticity and enhanced extinction.
Collapse
|
26
|
Neurogranin regulates CaM dynamics at dendritic spines. Sci Rep 2015; 5:11135. [PMID: 26084473 PMCID: PMC4471661 DOI: 10.1038/srep11135] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
Abstract
Calmodulin (CaM) plays a key role in synaptic function and plasticity due to its ability to mediate Ca2+ signaling. Therefore, it is essential to understand the dynamics of CaM at dendritic spines. In this study we have explored CaM dynamics using live-cell confocal microscopy and fluorescence recovery after photobleaching (FRAP) to study CaM diffusion. We find that only a small fraction of CaM in dendritic spines is immobile. Furthermore, the diffusion rate of CaM was regulated by neurogranin (Ng), a CaM-binding protein enriched at dendritic spines. Interestingly, Ng did not influence the immobile fraction of CaM at recovery plateau. We have previously shown that Ng enhances synaptic strength in a CaM-dependent manner. Taken together, these data indicate that Ng-mediated enhancement of synaptic strength is due to its ability to target, rather than sequester, CaM within dendritic spines.
Collapse
|
27
|
An L, Zhang T. Spatial cognition and sexually dimorphic synaptic plasticity balance impairment in rats with chronic prenatal ethanol exposure. Behav Brain Res 2013; 256:564-74. [PMID: 24050890 DOI: 10.1016/j.bbr.2013.09.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 09/03/2013] [Accepted: 09/07/2013] [Indexed: 01/12/2023]
Abstract
Prenatal ethanol exposure can lead to long-lasting impairments in the ability of rats to process spatial information, as well as produce long-lasting deficits in long-term potentiation (LTP), a biological model of learning and memory processing. The present study aimed to examine the sexually dimorphic effects of chronic prenatal ethanol exposure (CPEE) on behavior cognition and synaptic plasticity balance (SPB), and tried to understand a possible mechanism by evaluating the alternation of SPB. The animal model was produced by ethanol exposure throughout gestational period with 4 g/kg bodyweight. Offspring of both male and female were selected and studied on postnatal days 36. Subsequently, the data showed that chronic ethanol exposure resulted in birth weight reduction, losing bodyweight gain, microcephaly and hippocampus weight retardation. In Morris water maze (MWM) test, escape latencies were significantly higher in CPEE-treated rats than that in control ones. They also spent much less time in the target quadrant compared to that of control animals in the probe phase. In addition, it was found that there was a more severe impairment in females than that in males after CPEE treatment. Electrophysiological studies showed that CPEE considerably inhibited hippocampal LTP and facilitated depotentiation in males, while significantly enhanced LTP and suppressed depotentiation in females. A novel index, developed by us, showed that the action of CPEE on SPB was more sensitive in females than that in males, suggesting that it might be an effective index to distinguish the difference of SPB impairment between males and females.
Collapse
Affiliation(s)
- Lei An
- College of Life Sciences, Nankai University, 300071 Tianjin, PR China
| | | |
Collapse
|
28
|
Structural basis for the interaction of unstructured neuron specific substrates neuromodulin and neurogranin with Calmodulin. Sci Rep 2013; 3:1392. [PMID: 23462742 PMCID: PMC3589724 DOI: 10.1038/srep01392] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 02/21/2013] [Indexed: 01/07/2023] Open
Abstract
Neuromodulin (Nm) and neurogranin (Ng) are neuron-specific substrates of protein kinase C (PKC). Their interactions with Calmodulin (CaM) are crucial for learning and memory formation in neurons. Here, we report the structure of IQ peptides (24aa) of Nm/Ng complexed with CaM and their functional studies with full-length proteins. Nm/Ng and their respective IQ peptides are intrinsically unstructured; however, upon binding with CaM, IQ motifs adopt a helical conformation. Ser41 (Ser36) of Nm (Ng) is located in a negatively charged pocket in the apo CaM and, when phosphorylated, it will repel Nm/Ng from CaM. These observations explain the mechanism by which PKC-induced Ser phosphorylation blocks the association of Nm/Ng with CaM and interrupts several learning- and memory-associated functions. Moreover, the present study identified Arg as a key CaM interacting residue from Nm/Ng. This residue is crucial for CaM-mediated function, as evidenced by the inability of the Ng mutant (Arg-to-Ala) to potentiate synaptic transmission in CA1 hippocampal neurons.
Collapse
|