Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 75(4): 1361-1376, 2020.
Article in English | MEDLINE | ID: mdl-32390638

ABSTRACT

BACKGROUND: Porphyromonas gingivalis (P. gingivalis) and its gingipain virulence factors have been identified as pathogenic effectors in Alzheimer's disease (AD). In a recent study we demonstrated the presence of gingipains in over 90% of postmortem AD brains, with gingipains localizing to the cytoplasm of neurons. However, infection of neurons by P. gingivalis has not been previously reported. OBJECTIVE: To demonstrate intraneuronal P. gingivalis and gingipain expression in vitro after infecting neurons derived from human inducible pluripotent stem cells (iPSC) with P. gingivalis for 24, 48, and 72 h. METHODS: Infection was characterized by transmission electron microscopy, confocal microscopy, and bacterial colony forming unit assays. Gingipain expression was monitored by immunofluorescence and RT-qPCR, and protease activity monitored with activity-based probes. Neurodegenerative endpoints were assessed by immunofluorescence, western blot, and ELISA. RESULTS: Neurons survived the initial infection and showed time dependent, infection induced cell death. P. gingivalis was found free in the cytoplasm or in lysosomes. Infected neurons displayed an accumulation of autophagic vacuoles and multivesicular bodies. Tau protein was strongly degraded, and phosphorylation increased at T231. Over time, the density of presynaptic boutons was decreased. CONCLUSION: P. gingivalis can invade and persist in mature neurons. Infected neurons display signs of AD-like neuropathology including the accumulation of autophagic vacuoles and multivesicular bodies, cytoskeleton disruption, an increase in phospho-tau/tau ratio, and synapse loss. Infection of iPSC-derived mature neurons by P. gingivalis provides a novel model system to study the cellular mechanisms leading to AD and to investigate the potential of new therapeutic approaches.


Subject(s)
Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Bacteroidaceae Infections/complications , Gingipain Cysteine Endopeptidases/metabolism , Neurons/microbiology , Neurons/pathology , Alzheimer Disease/enzymology , Animals , Cells, Cultured , Mice , Neural Stem Cells/enzymology , Neural Stem Cells/microbiology , Neural Stem Cells/pathology , Neurons/enzymology , Porphyromonas gingivalis
2.
Pharmacol Res Perspect ; 8(1): e00562, 2020 02.
Article in English | MEDLINE | ID: mdl-31999052

ABSTRACT

COR388, a small-molecule lysine-gingipain inhibitor, is currently being investigated in a Phase 2/3 clinical trial for Alzheimer's disease (AD) with exploratory endpoints in periodontal disease. Gingipains are produced by two species of bacteria, Porphyromonas gingivalis and Porphyromonas gulae, typically associated with periodontal disease and systemic infections in humans and dogs, respectively. P. gulae infection in dogs is associated with periodontal disease, which provides a physiologically relevant model to investigate the pharmacology of COR388. In the current study, aged dogs with a natural oral infection of P. gulae and periodontal disease were treated with COR388 by oral administration for up to 90 days to assess lysine-gingipain target engagement and reduction of bacterial load and downstream pathology. In a 28-day dose-response study, COR388 inhibited the lysine-gingipain target and reduced P. gulae load in saliva, buccal cells, and gingival crevicular fluid. The lowest effective dose was continued for 90 days and was efficacious in continuous reduction of bacterial load and downstream periodontal disease pathology. In a separate histology study, dog brain tissue showed evidence of P. gulae DNA and neuronal lysine-gingipain, demonstrating that P. gulae infection is systemic and spreads beyond its oral reservoir, similar to recent observations of P. gingivalis in humans. Together, the pharmacokinetics and pharmacodynamics of COR388 lysine-gingipain inhibition, along with reduction of bacterial load and periodontal disease in naturally occurring P. gulae infection in the dog, support the use of COR388 in targeting lysine-gingipain and eliminating P. gingivalis infection in humans.


Subject(s)
Bacteroidaceae Infections/drug therapy , Dog Diseases/microbiology , Gingipain Cysteine Endopeptidases/antagonists & inhibitors , Organic Chemicals/administration & dosage , Periodontal Diseases/drug therapy , Porphyromonas/enzymology , Small Molecule Libraries/administration & dosage , Administration, Oral , Aging/blood , Animals , Bacterial Load , Bacterial Proteins/antagonists & inhibitors , Bacteroidaceae Infections/veterinary , Brain/drug effects , Brain/microbiology , Dog Diseases/drug therapy , Dogs , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gingival Crevicular Fluid/drug effects , Gingival Crevicular Fluid/microbiology , Organic Chemicals/chemistry , Organic Chemicals/pharmacology , Periodontal Diseases/veterinary , Porphyromonas/drug effects , Porphyromonas/pathogenicity , Saliva/drug effects , Saliva/microbiology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
3.
eNeuro ; 6(6)2019.
Article in English | MEDLINE | ID: mdl-31611335

ABSTRACT

Current perceptions of genetic and environmental vulnerabilities in the developing fetus are biased toward male outcomes. An argument is made that males are more vulnerable to gestational complications and neurodevelopmental disorders, the implication being that an understanding of disrupted development in males is sufficient to understand causal mechanisms that are assumed to be similar but attenuated in females. Here we examine this assumption in the context of immune-driven alterations in fetal brain development and related outcomes in female and male mice. Pregnant C57BL/6 mice were treated with low-dose lipopolysaccharide at embryonic day 12.5. Placental pathology, acute fetal brain inflammation and hypoxia, long-term changes in adult cortex cytoarchitecture, altered densities and ratio of excitatory (Satb2+) to inhibitory (parvalbumin+) neuronal subtypes, postnatal growth, and behavior outcomes were compared between male and female offspring. We find that while males experience more pronounced placental pathology, fetal brain hypoxia, depleted PV and Satb2+ densities, and social and learning-related behavioral abnormalities, females exhibit unique acute inflammatory signaling in fetal brain, postnatal growth delay, opposite alterations in cortical PV densities, changes in juvenile behavior, delayed postnatal body growth, and elevated anxiety-related behavior as adults. While males are more severely impacted by prenatal immune disruption by several measures, females exposed to the same insult exhibit a unique set of vulnerabilities and developmental consequences that is not present in males. Our results clearly outline disparate sex-specific features of prenatal vulnerability to inflammatory insults and warn against the casual extrapolation of male disease mechanisms to females.


Subject(s)
Brain/drug effects , Inflammation/immunology , Lipopolysaccharides/pharmacology , Placenta/drug effects , Prenatal Exposure Delayed Effects/immunology , Animals , Brain/immunology , Brain/metabolism , Cytokines/metabolism , Female , Male , Mice , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Placenta/immunology , Placenta/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Sex Factors
4.
Sci Adv ; 5(1): eaau3333, 2019 01.
Article in English | MEDLINE | ID: mdl-30746447

ABSTRACT

Porphyromonas gingivalis, the keystone pathogen in chronic periodontitis, was identified in the brain of Alzheimer's disease patients. Toxic proteases from the bacterium called gingipains were also identified in the brain of Alzheimer's patients, and levels correlated with tau and ubiquitin pathology. Oral P. gingivalis infection in mice resulted in brain colonization and increased production of Aß1-42, a component of amyloid plaques. Further, gingipains were neurotoxic in vivo and in vitro, exerting detrimental effects on tau, a protein needed for normal neuronal function. To block this neurotoxicity, we designed and synthesized small-molecule inhibitors targeting gingipains. Gingipain inhibition reduced the bacterial load of an established P. gingivalis brain infection, blocked Aß1-42 production, reduced neuroinflammation, and rescued neurons in the hippocampus. These data suggest that gingipain inhibitors could be valuable for treating P. gingivalis brain colonization and neurodegeneration in Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/microbiology , Bacteroidaceae Infections/drug therapy , Brain/microbiology , Brain/pathology , Neuroprotective Agents/therapeutic use , Porphyromonas gingivalis/enzymology , Small Molecule Libraries/therapeutic use , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Bacteroidaceae Infections/microbiology , Cell Line, Tumor , Disease Models, Animal , Female , Gingipain Cysteine Endopeptidases/antagonists & inhibitors , Gingipain Cysteine Endopeptidases/metabolism , Gingipain Cysteine Endopeptidases/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Pilot Projects , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/genetics , Prospective Studies , Saliva/microbiology , Small Molecule Libraries/pharmacology , tau Proteins/metabolism
5.
Mol Psychiatry ; 24(8): 1178-1188, 2019 08.
Article in English | MEDLINE | ID: mdl-29311652

ABSTRACT

Chronic stress is a recognized risk factor for psychiatric and psychological disorders and a potent modulator of adult neurogenesis. Numerous studies have shown that during stress, neurogenesis decreases; however, during the recovery from the stress, neurogenesis increases. Despite the increased number of neurons born after stress, it is unknown if the function and morphology of those neurons are altered. Here we asked whether neurons in adult mice, born during the final 5 days of chronic social stress and matured during recovery from chronic social stress, are similar to neurons born with no stress conditions from a quantitative, functional and morphological perspective, and whether those neurons are uniquely adapted to respond to a subsequent stressful challenge. We observed an increased number of newborn neurons incorporated in the dentate gyrus of the hippocampus during the 10-week post-stress recovery phase. Interestingly, those new neurons were more responsive to subsequent chronic stress, as they showed more of a stress-induced decrease in spine density and branching nodes than in neurons born during a non-stress period. Our results replicate findings that the neuronal survival and incorporation of neurons in the adult dentate gyrus increases after chronic stress and suggest that such neurons are uniquely adapted in the response to future social stressors. This finding provides a potential mechanism for some of the long-term hippocampal effects of stress.


Subject(s)
Neurogenesis/physiology , Neurons/physiology , Stress, Psychological/physiopathology , Age Factors , Animals , Brain/metabolism , Dentate Gyrus/metabolism , Hippocampus/metabolism , Male , Mice
6.
J Neurosci ; 33(43): 16874-88, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24155294

ABSTRACT

We have previously shown in mice that cytokine-mediated damage to the placenta can temporarily limit the flow of nutrients and oxygen to the fetus. The placental vulnerability is pronounced before embryonic day 11, when even mild immune challenge results in fetal loss. As gestation progresses, the placenta becomes increasingly resilient to maternal inflammation, but there is a narrow window in gestation when the placenta is still vulnerable to immune challenge yet resistant enough to allow for fetal survival. This gestational window correlates with early cortical neurogenesis in the fetal brain. Here, we show that maternal illness during this period selectively alters the abundance and laminar positioning of neuronal subtypes influenced by the Tbr1, Satb2, and Ctip2/Fezf2 patterning axis. The disturbances also lead to a laminar imbalance in the proportions of projection neurons and interneurons in the adult and are sufficient to cause changes in social behavior and cognition. These data illustrate how the timing of an illness-related placental vulnerability causes developmental alterations in neuroanatomical systems and behaviors that are relevant to autism spectrum disorders.


Subject(s)
Cerebral Cortex/embryology , Neurogenesis , Placenta Diseases/pathology , Placenta/pathology , Pregnancy Complications, Infectious/pathology , Animals , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Cognition , Cognition Disorders/etiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Interneurons/metabolism , Interneurons/pathology , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mental Disorders/etiology , Mice , Mice, Inbred C57BL , Placenta/physiopathology , Pregnancy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Social Behavior , T-Box Domain Proteins , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
J Neurosci ; 33(30): 12229-41, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884931

ABSTRACT

Hippocampus-dependent learning and memory relies on synaptic plasticity as well as network adaptations provided by the addition of adult-born neurons. We have previously shown that activity-induced intracellular signaling through the Rho family small GTPase Rac1 is necessary in forebrain projection neurons for normal synaptic plasticity in vivo, and here we show that selective loss of neuronal Rac1 also impairs the learning-evoked increase in neurogenesis in the adult mouse hippocampus. Earlier work has indicated that experience elevates the abundance of adult-born neurons in the hippocampus primarily by enhancing the survival of neurons produced just before the learning event. Loss of Rac1 in mature projection neurons did reduce learning-evoked neurogenesis but, contrary to our expectations, these effects were not mediated by altering the survival of young neurons in the hippocampus. Instead, loss of neuronal Rac1 activation selectively impaired a learning-evoked increase in the proliferation and accumulation of neural precursors generated during the learning event itself. This indicates that experience-induced alterations in neurogenesis can be mechanistically resolved into two effects: (1) the well documented but Rac1-independent signaling cascade that enhances the survival of young postmitotic neurons; and (2) a previously unrecognized Rac1-dependent signaling cascade that stimulates the proliferative production and retention of new neurons generated during learning itself.


Subject(s)
Adult Stem Cells/physiology , Maze Learning/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Neuropeptides/physiology , rac GTP-Binding Proteins/physiology , Adult Stem Cells/cytology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation , Cell Survival/physiology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Hippocampus/physiology , Male , Memory, Long-Term/physiology , Mice , Mice, Knockout , Mitosis/physiology , Neural Stem Cells/cytology , Neuronal Plasticity/physiology , Neurons/cytology , Neuropeptides/genetics , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, AMPA/physiology , Space Perception/physiology , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
8.
Brain Behav Immun ; 30: 33-44, 2013 May.
Article in English | MEDLINE | ID: mdl-23041279

ABSTRACT

Cranial irradiation for the treatment of brain tumors causes a delayed and progressive cognitive decline that is pronounced in young patients. Dysregulation of neural stem and progenitor cells is thought to contribute to these effects by altering early childhood brain development. Earlier work has shown that irradiation creates a chronic neuroinflammatory state that severely and selectively impairs postnatal and adult neurogenesis. Here we show that irradiation induces a transient non-classical cytokine response with selective upregulation of CCL2/monocyte chemoattractant protein-1 (MCP-1). Absence of CCL2 signaling in the hours after irradiation is alone sufficient to attenuate chronic microglia activation and allow the recovery of neurogenesis in the weeks following irradiation. This identifies CCL2 signaling as a potential clinical target for moderating the long-term defects in neural stem cell function following cranial radiation in children.


Subject(s)
Chemokine CCL2/metabolism , Cranial Irradiation , Hippocampus/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Cells, Cultured , Chemokine CCL2/genetics , Hippocampus/metabolism , Hippocampus/radiation effects , Male , Mice , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Microglia/radiation effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Neurogenesis/radiation effects , Neurons/metabolism , Neurons/radiation effects
9.
Brain Behav Immun ; 29: 28-38, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23108061

ABSTRACT

The detrimental effects of illness on cognition are familiar to virtually everyone. Some effects resolve quickly while others may linger after the illness resolves. We found that a transient immune response stimulated by lipopolysaccharide (LPS) compromised hippocampal neurogenesis and impaired hippocampus-dependent spatial memory. The immune event caused an ∼50% reduction in the number of neurons generated during the illness and the onset of the memory impairment was delayed and coincided with the time when neurons generated during the illness would have become functional within the hippocampus. Broad spectrum non-steroidal anti-inflammatory drugs attenuated these effects but selective Cox-2 inhibition was ineffective while PPARγ activation was surprisingly effective at protecting both neurogenesis and memory from the effects of LPS-produced transient illness. These data may highlight novel mechanisms behind chronic inflammatory and neuroinflammatory episodes that are known to compromise hippocampus-dependent forms of learning and memory.


Subject(s)
Illness Behavior/physiology , Lipopolysaccharides/pharmacology , Memory/physiology , Neurogenesis/physiology , PPAR gamma/metabolism , Space Perception/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antimetabolites , Bromodeoxyuridine , Cell Count , Data Interpretation, Statistical , Female , Hippocampus/physiology , Illness Behavior/drug effects , Immunohistochemistry , Learning/physiology , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects
10.
Mol Cell Neurosci ; 41(4): 409-19, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19394428

ABSTRACT

Rac1 is a member of the Rho family of small GTPases that are important for structural aspects of the mature neuronal synapse including basal spine density and shape, activity-dependent spine enlargement, and AMPA receptor clustering in vitro. Here we demonstrate that selective elimination of Rac1 in excitatory neurons in the forebrain in vivo not only affects spine structure, but also impairs synaptic plasticity in the hippocampus with consequent defects in hippocampus-dependent spatial learning. Furthermore, Rac1 mutants display deficits in working/episodic-like memory in the delayed matching-to-place (DMP) task suggesting that Rac1 is a central regulator of rapid encoding of novel spatial information in vivo.


Subject(s)
Hippocampus/cytology , Learning/physiology , Memory/physiology , Neuronal Plasticity/physiology , Spatial Behavior/physiology , rac1 GTP-Binding Protein/physiology , Analysis of Variance , Animals , Biophysics/methods , Disks Large Homolog 4 Protein , Electric Stimulation/methods , Green Fluorescent Proteins/genetics , Guanylate Kinases , Hippocampus/physiology , Hippocampus/ultrastructure , Intracellular Signaling Peptides and Proteins/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Reaction Time/genetics , beta-Galactosidase/metabolism , rac1 GTP-Binding Protein/deficiency
11.
Eur J Neurosci ; 18(5): 1279-85, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12956726

ABSTRACT

Two types of synaptic depression have been described in the hippocampus, long-term depression and depotentiation of long-term potentiation known to recruit the serine/threonine protein phosphatases PP1, PP2A and PP2B (calcineurin). The contribution of each of these protein phosphatases is controversial. To examine the role of the Ca2+/calmodulin-dependent protein phosphatase calcineurin in long-term depression and depotentiation, we analysed the effect of genetically inhibiting calcineurin reversibly in the hippocampus, using the doxycycline-dependent rtTA system in transgenic mice. We show that reducing calcineurin activity has no effect on long-term depression but reversibly affects depotentiation. Consistently, the calcineurin inhibitor FK-506 reproduces the depotentiation impairment observed in the mutant mice but does not affect long-term depression in control animals. In contrast, the PP1/PP2A inhibitor okadaic acid fully blocks both long-term depression and depotentiation. These data demonstrate that the nature of signalling cascades induced by synaptic activity depends on the initial synaptic state. While depression of potentiated synaptic responses requires activation of PP1/PP2A and/or calcineurin, depression of basal synaptic responses depends only on PP1/PP2A activation.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Phosphoric Monoester Hydrolases/physiology , Animals , Electric Stimulation , Electrophysiology/methods , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/anatomy & histology , In Vitro Techniques , Long-Term Potentiation/genetics , Long-Term Synaptic Depression/genetics , Mice , Mice, Transgenic , Mutation/physiology , N-Methylaspartate/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Okadaic Acid/pharmacology , Phosphoric Monoester Hydrolases/classification , Presynaptic Terminals/physiology , Tacrolimus/pharmacology , Time Factors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
12.
J Neurosci ; 23(3): 826-36, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12574411

ABSTRACT

Long-term depression (LTD) is an activity-dependent weakening of synaptic efficacy at individual inhibitory synapses, a possible cellular model of learning and memory. Here, we show that the induction of LTD of inhibitory transmission recruits activated calcineurin (CaN) to dephosphorylate type-A GABA receptor (GABA(A)Rs) via the direct binding of CaN catalytic domain to the second intracellular domain of the GABA(A)R-gamma(2) subunits. Prevention of the CaN-GABA(A) receptor complex formation by expression of an autoinhibitory domain of CaN in the hippocampus of transgenic mice blocks the induction of LTD. Conversely, genetic expression of the CaN catalytic domain in the hippocampus depresses inhibitory synaptic responses, occluding LTD. Thus, an activity-dependent physical and functional interaction between CaN and GABA(A) receptors is both necessary and sufficient for inducing LTD at CA1 individual inhibitory synapses.


Subject(s)
Calcineurin/metabolism , Hippocampus/metabolism , Long-Term Synaptic Depression/physiology , Neural Inhibition/physiology , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Electric Stimulation , Hippocampus/cytology , In Vitro Techniques , Macromolecular Substances , Mice , Mice, Mutant Strains , Mice, Transgenic , Patch-Clamp Techniques , Phosphorylation , Protein Binding/physiology , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
13.
Nature ; 418(6901): 970-5, 2002 Aug 29.
Article in English | MEDLINE | ID: mdl-12198546

ABSTRACT

Repetition in learning is a prerequisite for the formation of accurate and long-lasting memory. Practice is most effective when widely distributed over time, rather than when closely spaced or massed. But even after efficient learning, most memories dissipate with time unless frequently used. The molecular mechanisms of these time-dependent constraints on learning and memory are unknown. Here we show that protein phosphatase 1 (PP1) determines the efficacy of learning and memory by limiting acquisition and favouring memory decline. When PP1 is genetically inhibited during learning, short intervals between training episodes are sufficient for optimal performance. The enhanced learning correlates with increased phosphorylation of cyclic AMP-dependent response element binding (CREB) protein, of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and of the GluR1 subunit of the AMPA receptor; it also correlates with CREB-dependent gene expression that, in control mice, occurs only with widely distributed training. Inhibition of PP1 prolongs memory when induced after learning, suggesting that PP1 also promotes forgetting. This property may account for ageing-related cognitive decay, as old mutant animals had preserved memory. Our findings emphasize the physiological importance of PP1 as a suppressor of learning and memory, and as a potential mediator of cognitive decline during ageing.


Subject(s)
Hippocampus/enzymology , Hippocampus/physiology , Learning/physiology , Memory/physiology , Phosphoprotein Phosphatases/metabolism , Aging/genetics , Aging/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cognition/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Discrimination, Psychological/physiology , Gene Expression Regulation , Mice , Mice, Transgenic , Mutation , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Phosphatase 1 , Protein Subunits , Receptors, AMPA/metabolism , Space Perception/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...