Pathological reorganization of NMDA receptors subunits and postsynaptic protein PSD-95 distribution in Alzheimer's disease.

In Alzheimer's disease (AD), synaptic alterations play a major role and are often correlated with cognitive changes. In order to better understand synaptic modifications, we compared alterations in NMDA receptors and postsynaptic protein PSD-95 expression in the entorhinal cortex (EC) and frontal cortex (FC; area 9) of AD and control brains. We combined immunohistochemical and image analysis methods to quantify on consecutive sections the distribution of PSD-95 and NMDA receptors GluN1, GluN2A and GluN2B in EC and FC from 25 AD and control cases. The density of stained receptors was analyzed using multivariate statistical methods to assess the effect of neurodegeneration. In both regions, the number of neuronal profiles immunostained for GluN1 receptors subunit and PSD-95 protein was significantly increased in AD compared to controls (3-6 fold), while the number of neuronal profiles stained for GluN2A and GluN2B receptors subunits was on the contrary decreased (3-4 fold). The increase in marked neuronal profiles was more prominent in a cortical band corresponding to layers 3 to 5 with large pyramidal cells. Neurons positive for GluN1 or PSD-95 staining were often found in the same localization on consecutive sections and they were also reactive for the anti-tau antibody AD2, indicating a neurodegenerative process. Differences in the density of immunoreactive puncta representing neuropile were not statistically significant. Altogether these data indicate that GluN1 and PSD-95 accumulate in the neuronal perikarya, but this is not the case for GluN2A and GluN2B, while the neuropile compartment is less subject to modifications. Thus, important variations in the pattern of distribution of the NMDA receptors subunits and PSD-95 represent a marker in AD and by impairing the neuronal network, contribute to functional deterioration.

[1]  C. Cotman,et al.  Density and distribution of NMDA receptors in the human hippocampus in Alzheimer's disease , 1986, Brain Research.

[2]  G. Knott,et al.  PSD-95 promotes synaptogenesis and multiinnervated spine formation through nitric oxide signaling , 2008, The Journal of cell biology.

[3]  W. Klein,et al.  Aβ Oligomer-Induced Aberrations in Synapse Composition, Shape, and Density Provide a Molecular Basis for Loss of Connectivity in Alzheimer's Disease , 2007, The Journal of Neuroscience.

[4]  T. Sacktor,et al.  Postsynaptic degeneration as revealed by PSD-95 reduction occurs after advanced Aβ and tau pathology in transgenic mouse models of Alzheimer’s disease , 2011, Acta Neuropathologica.

[5]  M. Bear,et al.  Ubiquitination Regulates PSD-95 Degradation and AMPA Receptor Surface Expression , 2003, Neuron.

[6]  Suneil K. Kalia,et al.  NMDA receptors in clinical neurology: excitatory times ahead , 2008, The Lancet Neurology.

[7]  D. Bennett,et al.  Decreases in soluble α-synuclein in frontal cortex correlate with cognitive decline in the elderly , 2004, Neuroscience Letters.

[8]  P. Greengard,et al.  Regulation of NMDA receptor trafficking by amyloid-β , 2005, Nature Neuroscience.

[9]  E. Masliah,et al.  Alterations in glutamate receptor 2/3 subunits and amyloid precursor protein expression during the course of Alzheimer’s disease and Lewy body variant , 1997, Acta Neuropathologica.

[10]  A. El-Husseini,et al.  Excitation Control: Balancing PSD-95 Function at the Synapse , 2008, Frontiers in molecular neuroscience.

[11]  X. Zhao,et al.  The effects of aging on N-methyl-d-aspartate receptor subunits in the synaptic membrane and relationships to long-term spatial memory , 2009, Neuroscience.

[12]  D. Pei,et al.  NR2B-Containing NMDA Receptors Expression and Their Relationship to Apoptosis in Hippocampus of Alzheimer’s Disease-Like Rats , 2012, Neurochemical Research.

[13]  Vishnu Suppiramaniam,et al.  Amyloid beta peptides and glutamatergic synaptic dysregulation , 2008, Experimental Neurology.

[14]  M. Sheng,et al.  Synaptic Accumulation of PSD-95 and Synaptic Function Regulated by Phosphorylation of Serine-295 of PSD-95 , 2007, Neuron.

[15]  Roger A. Nicoll,et al.  Rapid Bidirectional Switching of Synaptic NMDA Receptors , 2007, Neuron.

[16]  Masahiko Watanabe,et al.  PSD-95 Uncouples Dopamine–Glutamate Interaction in the D1/PSD-95/NMDA Receptor Complex , 2009, The Journal of Neuroscience.

[17]  R. Wenthold,et al.  Distribution of Glutamate Receptor Subunit NMDAR1 in the Hippocampus of Normal Elderly and Patients with Alzheimer's Disease , 1999, Experimental Neurology.

[18]  R. Malenka,et al.  Destabilization of the Postsynaptic Density by PSD-95 Serine 73 Phosphorylation Inhibits Spine Growth and Synaptic Plasticity , 2009, Neuron.

[19]  W. Klein,et al.  N‐Methyl‐d‐aspartate receptors are required for synaptic targeting of Alzheimer’s toxic amyloid‐β peptide oligomers , 2010, Journal of neurochemistry.

[20]  S. Love,et al.  Premorbid effects of APOE on synaptic proteins in human temporal neocortex , 2006, Neurobiology of Aging.

[21]  Shaomin Li,et al.  Amyloid-β protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory , 2008, Nature Medicine.

[22]  Kristina D. Micheva,et al.  Oligomeric amyloid β associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques , 2009, Proceedings of the National Academy of Sciences.

[23]  P. Riederer,et al.  Functional Neurochemistry of Alzheimers Disease , 2004 .

[24]  S. Love,et al.  Measurement of pre- and post-synaptic proteins in cerebral cortex: effects of post-mortem delay , 2004, Journal of Neuroscience Methods.

[25]  N. Inestrosa,et al.  β-Amyloid Causes Depletion of Synaptic Vesicles Leading to Neurotransmission Failure* , 2009, The Journal of Biological Chemistry.

[26]  Ning Zhang,et al.  Different expression of NR2B and PSD‐95 in rat hippocampal subregions during postnatal development , 2009, Microscopy research and technique.

[27]  H. Bading,et al.  Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways , 2002, Nature Neuroscience.

[28]  A. Buisson,et al.  Synapses, NMDA receptor activity and neuronal Aβ production in Alzheimer’s disease , 2011, Reviews in the neurosciences.

[29]  M. Constantine‐Paton,et al.  BDNF induces transport of PSD-95 to dendrites through PI3K-AKT signaling after NMDA receptor activation , 2007, Nature Neuroscience.

[30]  R. Tanzi The synaptic Aβ hypothesis of Alzheimer disease , 2005, Nature Neuroscience.

[31]  Jürgen Götz,et al.  Amyloid-β and tau — a toxic pas de deux in Alzheimer's disease , 2011, Nature Reviews Neuroscience.

[32]  M. Martín-Satué,et al.  Amyloid β peptide oligomers directly activate NMDA receptors. , 2011, Cell calcium.

[33]  E B Mukaetova-Ladinska,et al.  Staging of cytoskeletal and beta-amyloid changes in human isocortex reveals biphasic synaptic protein response during progression of Alzheimer's disease. , 2000, The American journal of pathology.

[34]  C. Bouras,et al.  Ubiquitination and cysteine nitrosylation during aging and Alzheimer's disease , 2009, Brain Research Bulletin.

[35]  C. Lippa,et al.  Review: Disruption of the Postsynaptic Density in Alzheimer’s Disease and Other Neurodegenerative Dementias , 2010, American journal of Alzheimer's disease and other dementias.

[36]  Yen-Chung Chang,et al.  Heavy chain of cytoplasmic dynein is a major component of the postsynaptic density fraction , 2006, Journal of neuroscience research.

[37]  R. Roberts,et al.  Dual use of immunohistochemistry for film densitometry and light microscopy , 2012, Journal of Neuroscience Methods.

[38]  Eric Tardif,et al.  Differential changes in synaptic proteins in the Alzheimer frontal cortex with marked increase in PSD-95 postsynaptic protein. , 2008, Journal of Alzheimer's disease : JAD.

[39]  P. Dodd,et al.  Reduction in post-synaptic scaffolding PSD-95 and SAP-102 protein levels in the Alzheimer inferior temporal cortex is correlated with disease pathology. , 2010, Journal of Alzheimer's disease : JAD.

[40]  E. Mandelkow,et al.  Linking Amyloid-β and Tau: Amyloid-β Induced Synaptic Dysfunction via Local Wreckage of the Neuronal Cytoskeleton , 2011, Neurodegenerative Diseases.

[41]  Jürgen Götz,et al.  Dendritic Function of Tau Mediates Amyloid-β Toxicity in Alzheimer's Disease Mouse Models , 2010, Cell.

[42]  D. Purpura,et al.  NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders , 2007, Nature Reviews Neuroscience.

[43]  V. Nimmrich,et al.  Is Alzheimer's Disease a Result of Presynaptic Failure? - Synaptic Dysfunctions Induced by Oligomeric β-Amyloid , 2009, Reviews in the neurosciences.

[44]  André Schrattenholz,et al.  NMDA receptors are not alone: dynamic regulation of NMDA receptor structure and function by neuregulins and transient cholesterol-rich membrane domains leads to disease-specific nuances of glutamate-signalling. , 2006, Current topics in medicinal chemistry.

[45]  M. Constantine‐Paton,et al.  Receptor compartmentalization and trafficking at glutamate synapses: a developmental proposal , 2004, Trends in Neurosciences.

[46]  Shigeo Okabe,et al.  Differential Control of Postsynaptic Density Scaffolds via Actin-Dependent and -Independent Mechanisms , 2006, The Journal of Neuroscience.

[47]  G. Leuba,et al.  The role of the ubiquitin proteasome system in Alzheimer's disease , 2011, Experimental biology and medicine.

[48]  C. Parsons,et al.  Alzheimer's disease, β‐amyloid, glutamate, NMDA receptors and memantine – searching for the connections , 2012, British journal of pharmacology.

[49]  J. Kaye,et al.  Differential loss of synaptic proteins in Alzheimer's disease: implications for synaptic dysfunction. , 2005, Journal of Alzheimer's disease : JAD.

[50]  G. Leuba,et al.  Postsynaptic density protein PSD-95 expression in Alzheimer's disease and okadaic acid induced neuritic retraction , 2008, Neurobiology of Disease.

[51]  M. Sheng,et al.  PDZ domain proteins of synapses , 2004, Nature Reviews Neuroscience.

[52]  R. Rissman,et al.  Biochemical and molecular studies of NMDA receptor subunits NR1/2A/2B in hippocampal subregions throughout progression of Alzheimer's disease pathology , 2004, Neurobiology of Disease.

[53]  Ghiam Yamin NMDA receptor–dependent signaling pathways that underlie amyloid β‐protein disruption of LTP in the hippocampus , 2009, Journal of neuroscience research.

[54]  K. Magnusson,et al.  Frontiers in Aging Neuroscience Aging Neuroscience Review Article , 2022 .

[55]  P. Dodd,et al.  Post-synaptic scaffolding protein interactions with glutamate receptors in synaptic dysfunction and Alzheimer's disease , 2011, Progress in Neurobiology.

[56]  Tsutomu Hashikawa,et al.  Retrograde modulation of presynaptic release probability through signaling mediated by PSD-95–neuroligin , 2007, Nature Neuroscience.

[57]  D. Winder,et al.  Plasticity and behavior New genetic techniques to address multiple forms and functions , 2001, Physiology & Behavior.

[58]  R. Petralia Distribution of Extrasynaptic NMDA Receptors on Neurons , 2012, TheScientificWorldJournal.