Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 150(5): 1055-67, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939628

ABSTRACT

The type 2 ryanodine receptor/calcium release channel (RyR2), required for excitation-contraction coupling in the heart, is abundant in the brain. Chronic stress induces catecholamine biosynthesis and release, stimulating ß-adrenergic receptors and activating cAMP signaling pathways in neurons. In a murine chronic restraint stress model, neuronal RyR2 were phosphorylated by protein kinase A (PKA), oxidized, and nitrosylated, resulting in depletion of the stabilizing subunit calstabin2 (FKBP12.6) from the channel complex and intracellular calcium leak. Stress-induced cognitive dysfunction, including deficits in learning and memory, and reduced long-term potentiation (LTP) at the hippocampal CA3-CA1 connection were rescued by oral administration of S107, a compound developed in our laboratory that stabilizes RyR2-calstabin2 interaction, or by genetic ablation of the RyR2 PKA phosphorylation site at serine 2808. Thus, neuronal RyR2 remodeling contributes to stress-induced cognitive dysfunction. Leaky RyR2 could be a therapeutic target for treatment of stress-induced cognitive dysfunction.


Subject(s)
Cognition Disorders/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Stress Disorders, Traumatic/metabolism
2.
Cell ; 149(6): 1314-26, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682251

ABSTRACT

Hypothalamic neurons expressing Agouti-related peptide (AgRP) are critical for initiating food intake, but druggable biochemical pathways that control this response remain elusive. Thus, genetic ablation of insulin or leptin signaling in AgRP neurons is predicted to reduce satiety but fails to do so. FoxO1 is a shared mediator of both pathways, and its inhibition is required to induce satiety. Accordingly, FoxO1 ablation in AgRP neurons of mice results in reduced food intake, leanness, improved glucose homeostasis, and increased sensitivity to insulin and leptin. Expression profiling of flow-sorted FoxO1-deficient AgRP neurons identifies G-protein-coupled receptor Gpr17 as a FoxO1 target whose expression is regulated by nutritional status. Intracerebroventricular injection of Gpr17 agonists induces food intake, whereas Gpr17 antagonist cangrelor curtails it. These effects are absent in Agrp-Foxo1 knockouts, suggesting that pharmacological modulation of this pathway has therapeutic potential to treat obesity.


Subject(s)
Agouti-Related Protein/metabolism , Eating , Forkhead Transcription Factors/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Agouti-Related Protein/genetics , Animals , Energy Metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Glucose/metabolism , Leptin/metabolism , Mice
3.
Mol Ther ; 31(2): 409-419, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36369741

ABSTRACT

The accumulation of soluble oligomers of the amyloid-ß peptide (AßOs) in the brain has been implicated in synapse failure and memory impairment in Alzheimer's disease. Here, we initially show that treatment with NUsc1, a single-chain variable-fragment antibody (scFv) that selectively targets a subpopulation of AßOs and shows minimal reactivity to Aß monomers and fibrils, prevents the inhibition of long-term potentiation in hippocampal slices and memory impairment induced by AßOs in mice. As a therapeutic approach for intracerebral antibody delivery, we developed an adeno-associated virus vector to drive neuronal expression of NUsc1 (AAV-NUsc1) within the brain. Transduction by AAV-NUsc1 induced NUsc1 expression and secretion in adult human brain slices and inhibited AßO binding to neurons and AßO-induced loss of dendritic spines in primary rat hippocampal cultures. Treatment of mice with AAV-NUsc1 prevented memory impairment induced by AßOs and, remarkably, reversed memory deficits in aged APPswe/PS1ΔE9 Alzheimer's disease model mice. These results support the feasibility of immunotherapy using viral vector-mediated gene delivery of NUsc1 or other AßO-specific single-chain antibodies as a potential therapeutic approach in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Single-Chain Antibodies , Mice , Rats , Humans , Animals , Aged , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Synapses/metabolism , Neurons/metabolism , Memory Disorders/genetics , Memory Disorders/therapy
4.
Alzheimers Dement ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38934107

ABSTRACT

INTRODUCTION: Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS: We investigated the effects of HNK on hippocampal protein synthesis, long-term potentiation (LTP), and memory in AD mouse models. RESULTS: HNK activated extracellular signal-regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid-ß oligomers (AßO)-infused mice in an ERK1/2-dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION: Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. HIGHLIGHTS: The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways. HNK corrects hippocampal synaptic and memory defects in two mouse models of AD. Rescue of synaptic and memory impairments by HNK depends on ERK signaling. HNK corrects aberrant transcriptional signatures in APP/PS1 mice.

5.
Bioorg Med Chem Lett ; 92: 129409, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37453616

ABSTRACT

Phosphodiesterase 5 (PDE5) is a cyclic guanosine monophosphate-degrading enzyme involved in numerous biological pathways. Inhibitors of PDE5 are important therapeutics for the treatment of neurodegenerative diseases, including Alzheimer's disease (AD). We previously reported the first generation of quinoline-based PDE5 inhibitors for the treatment of AD. However, the short in vitro microsomal stability rendered them unsuitable drug candidates. Here we report a series of new quinoline-based PDE5 inhibitors. Among them, compound 4b, 8-cyclopropyl-3-(hydroxymethyl)-4-(((6-methoxypyridin-3-yl)methyl)amino)quinoline-6-carbonitrile, shows a PDE5 IC50 of 20 nM and improved in vitro microsomal stability (t1/2 = 44.6 min) as well as excellent efficacy in restoring long-term potentiation, a type of synaptic plasticity to underlie memory formation, in electrophysiology experiments with a mouse model of AD. These results provide an insight into the development of a new class of PDE5 inhibitors for the treatment of AD.


Subject(s)
Alzheimer Disease , Quinolines , Mice , Animals , Phosphodiesterase 5 Inhibitors/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Neuronal Plasticity , Alzheimer Disease/drug therapy , Quinolines/pharmacology , Quinolines/therapeutic use
6.
Brain ; 145(8): 2849-2868, 2022 08 27.
Article in English | MEDLINE | ID: mdl-35254410

ABSTRACT

Synaptic dysfunction is an early mechanism in Alzheimer's disease that involves progressively larger areas of the brain over time. However, how it starts and propagates is unknown. Here we show that amyloid-ß released by microglia in association with large extracellular vesicles (Aß-EVs) alters dendritic spine morphology in vitro, at the site of neuron interaction, and impairs synaptic plasticity both in vitro and in vivo in the entorhinal cortex-dentate gyrus circuitry. One hour after Aß-EV injection into the mouse entorhinal cortex, long-term potentiation was impaired in the entorhinal cortex but not in the dentate gyrus, its main target region, while 24 h later it was also impaired in the dentate gyrus, revealing a spreading of long-term potentiation deficit between the two regions. Similar results were obtained upon injection of extracellular vesicles carrying Aß naturally secreted by CHO7PA2 cells, while neither Aß42 alone nor inflammatory extracellular vesicles devoid of Aß were able to propagate long-term potentiation impairment. Using optical tweezers combined to time-lapse imaging to study Aß-EV-neuron interaction, we show that Aß-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aß-EV motility was inhibited, no propagation of long-term potentiation deficit occurred along the entorhinal-hippocampal circuit, implicating large extracellular vesicle motion at the neuron surface in the spreading of long-term potentiation impairment. Our data indicate the involvement of large microglial extracellular vesicles in the rise and propagation of early synaptic dysfunction in Alzheimer's disease and suggest a new mechanism controlling the diffusion of large extracellular vesicles and their pathogenic signals in the brain parenchyma, paving the way for novel therapeutic strategies to delay the disease.


Subject(s)
Alzheimer Disease , Extracellular Vesicles , Amyloid beta-Peptides , Animals , Hippocampus , Long-Term Potentiation , Mice , Microglia
7.
Int J Mol Sci ; 25(1)2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38203429

ABSTRACT

After several years of research in the field of Alzheimer's disease (AD), it is still unclear how amyloid-beta (Aß) and Tau, two key hallmarks of the disease, mediate the neuropathogenic events that lead to AD. Current data challenge the "Amyloid Cascade Hypothesis" that has prevailed in the field of AD, stating that Aß precedes and triggers Tau pathology that will eventually become the toxic entity in the progression of the disease. This perspective also led the field of therapeutic approaches towards the development of strategies that target Aß or Tau. In the present review, we discuss recent literature regarding the neurotoxic role of both Aß and Tau in AD, as well as their physiological function in the healthy brain. Consequently, we present studies suggesting that Aß and Tau act independently of each other in mediating neurotoxicity in AD, thereafter, re-evaluating the "Amyloid Cascade Hypothesis" that places Tau pathology downstream of Aß. More recent studies have confirmed that both Aß and Tau could propagate the disease and induce synaptic and memory impairments via the amyloid precursor protein (APP). This finding is not only interesting from a mechanistic point of view since it provides better insights into the AD pathogenesis but also from a therapeutic point of view since it renders APP a common downstream effector for both Aß and Tau. Subsequently, therapeutic strategies that act on APP might provide a more viable and physiologically relevant approach for targeting AD.


Subject(s)
Alzheimer Disease , Neurotoxicity Syndromes , Humans , Amyloid beta-Protein Precursor , Amyloid beta-Peptides , Amyloidogenic Proteins , Brain
8.
Neuropathol Appl Neurobiol ; 48(5): e12811, 2022 08.
Article in English | MEDLINE | ID: mdl-35274343

ABSTRACT

AIMS: Several studies reported that astrocytes support neuronal communication by the release of gliotransmitters, including ATP and glutamate. Astrocytes also play a fundamental role in buffering extracellular glutamate in the synaptic cleft, thus limiting the risk of excitotoxicity in neurons. We previously demonstrated that extracellular tau oligomers (ex-oTau), by specifically targeting astrocytes, affect glutamate-dependent synaptic transmission via a reduction in gliotransmitter release. The aim of this work was to determine if ex-oTau also impair the ability of astrocytes to uptake extracellular glutamate, thus further contributing to ex-oTau-dependent neuronal dysfunction. METHODS: Primary cultures of astrocytes and organotypic brain slices were exposed to ex-oTau (200 nM) for 1 h. Extracellular glutamate buffering by astrocytes was studied by: Na+ imaging; electrophysiological recordings; high-performance liquid chromatography; Western blot and immunofluorescence. Experimental paradigms avoiding ex-oTau internalisation (i.e. heparin pre-treatment and amyloid precursor protein knockout astrocytes) were used to dissect intracellular vs extracellular effects of oTau. RESULTS: Ex-oTau uploading in astrocytes significantly affected glutamate-transporter-1 expression and function, thus impinging on glutamate buffering activity. Ex-oTau also reduced Na-K-ATPase activity because of pump mislocalisation on the plasma membrane, with no significant changes in expression. This effect was independent of oTau internalisation and it caused Na+ overload and membrane depolarisation in ex-oTau-targeted astrocytes. CONCLUSIONS: Ex-oTau exerted a complex action on astrocytes, at both intracellular and extracellular levels. The net effect was dysregulated glutamate signalling in terms of both release and uptake that relied on reduced expression of glutamate-transporter-1, altered function and localisation of NKA1A1, and NKA1A2. Consequently, Na+ gradients and all Na+ -dependent transports were affected.


Subject(s)
Astrocytes , Glutamic Acid , Astrocytes/metabolism , Cells, Cultured , Down-Regulation , Neurons/metabolism , Synaptic Transmission/physiology
9.
J Neurosci ; 40(23): 4596-4608, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32341098

ABSTRACT

Beta-amyloid (Aß) is thought to play a critical role in Alzheimer's disease (AD), and application of soluble oligomeric forms of Aß produces AD-like impairments in cognition and synaptic plasticity in experimental systems. We found previously that transgenic overexpression of the PP2A methylesterase, PME-1, or the PP2A methyltransferase, LCMT-1, altered the sensitivity of mice to Aß-induced impairments, suggesting that PME-1 inhibition may be an effective approach for preventing or treating these impairments. To explore this possibility, we examined the behavioral and electrophysiological effects of acutely applied synthetic Aß oligomers in male and female mice heterozygous for either a PME-1 KO or an LCMT-1 gene-trap mutation. We found that heterozygous PME-1 KO mice were resistant to Aß-induced impairments in cognition and synaptic plasticity, whereas LCMT-1 gene-trap mice showed increased sensitivity to Aß-induced impairments. The heterozygous PME-1 KO mice produced normal levels of endogenous Aß and exhibited normal electrophysiological responses to picomolar concentrations of Aß, suggesting that reduced PME-1 expression in these animals protects against Aß-induced impairments without impacting normal physiological Aß functions. Together, these data provide additional support for roles for PME-1 and LCMT-1 in regulating sensitivity to Aß-induced impairments, and suggest that inhibition of PME-1 may constitute a viable therapeutic approach for selectively protecting against the pathologic actions of Aß in AD.SIGNIFICANCE STATEMENT Elevated levels of ß-amyloid (Aß) in the brain are thought to contribute to the cognitive impairments observed in Alzheimer's disease patients. Here we show that genetically reducing endogenous levels of the PP2A methylesterase, PME-1, prevents the cognitive and electrophysiological impairments caused by acute exposure to pathologic concentrations of Aß without impairing normal physiological Aß function or endogenous Aß production. Conversely, reducing endogenous levels of the PP2A methyltransferase, LCMT-1, increases sensitivity to Aß-induced impairments. These data offer additional insights into the molecular factors that control sensitivity to Aß-induced impairments, and suggest that inhibiting PME-1 may constitute a viable therapeutic avenue for preventing Aß-related impairments in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Carboxylic Ester Hydrolases/biosynthesis , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/enzymology , Protein O-Methyltransferase/biosynthesis , Animals , Carboxylic Ester Hydrolases/genetics , Cognitive Dysfunction/physiopathology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Female , Gene Expression , Male , Mice , Mice, Knockout , Protein O-Methyltransferase/genetics
10.
J Neurochem ; 159(6): 958-979, 2021 12.
Article in English | MEDLINE | ID: mdl-34657288

ABSTRACT

Adaptation to acute and chronic stress and/or persistent stressors is a subject of wide interest in central nervous system disorders. In this context, stress is an effector of change in organismal homeostasis and the response is generated when the brain perceives a potential threat. Herein, we discuss a nuanced and granular view whereby a wide variety of genotoxic and environmental stressors, including aging, genetic risk factors, environmental exposures, and age- and lifestyle-related changes, act as direct insults to cellular, as opposed to organismal, homeostasis. These two concepts of how stressors impact the central nervous system are not mutually exclusive. We discuss how maladaptive stressor-induced changes in protein connectivity through epichaperomes, disease-associated pathologic scaffolds composed of tightly bound chaperones, co-chaperones, and other factors, impact intracellular protein functionality altering phenotypes, that in turn disrupt and remodel brain networks ranging from intercellular to brain connectome levels. We provide an evidence-based view on how these maladaptive changes ranging from stressor to phenotype provide unique precision medicine opportunities for diagnostic and therapeutic development, especially in the context of neurodegenerative disorders including Alzheimer's disease where treatment options are currently limited.


Subject(s)
Aging/metabolism , Brain/metabolism , Environmental Exposure/adverse effects , Molecular Chaperones/metabolism , Neurodegenerative Diseases/metabolism , Neuronal Plasticity/physiology , Adaptation, Physiological/physiology , Aging/pathology , Animals , Brain/pathology , Chaperonin 60/metabolism , Heat-Shock Response/physiology , Homeostasis/physiology , Humans , Neurodegenerative Diseases/pathology , Oxidative Stress/physiology
11.
J Neurosci ; 39(30): 5986-6000, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31127002

ABSTRACT

Failure of anti-amyloid-ß peptide (Aß) therapies against Alzheimer's disease (AD), a neurodegenerative disorder characterized by high amounts of the peptide in the brain, raised the question of the physiological role of Aß released at low concentrations in the healthy brain. To address this question, we studied the presynaptic and postsynaptic mechanisms underlying the neuromodulatory action of picomolar amounts of oligomeric Aß42 (oAß42) on synaptic glutamatergic function in male and female mice. We found that 200 pm oAß42 induces an increase of frequency of miniature EPSCs and a decrease of paired pulse facilitation, associated with an increase in docked vesicle number, indicating that it augments neurotransmitter release at presynaptic level. oAß42 also produced postsynaptic changes as shown by an increased length of postsynaptic density, accompanied by an increased expression of plasticity-related proteins such as cAMP-responsive element binding protein phosphorylated at Ser133, calcium-calmodulin-dependent kinase II phosphorylated at Thr286, and brain-derived neurotrophic factor, suggesting a role for Aß in synaptic tagging. These changes resulted in the conversion of early into late long-term potentiation through the nitric oxide/cGMP/protein kinase G intracellular cascade consistent with a cGMP-dependent switch from short- to long-term memory observed in vivo after intrahippocampal administration of picomolar amounts of oAß42 These effects were present upon extracellular but not intracellular application of the peptide and involved α7 nicotinic acetylcholine receptors. These observations clarified the physiological role of oAß42 in synaptic function and memory formation providing solid fundamentals for investigating the pathological effects of high Aß levels in the AD brains.SIGNIFICANCE STATEMENT High levels of oligomeric amyloid-ß42 (oAß42) induce synaptic dysfunction leading to memory impairment in Alzheimer's disease (AD). However, at picomolar concentrations, the peptide is needed to ensure long-term potentiation (LTP) and memory. Here, we show that extracellular 200 pm oAß42 concentrations increase neurotransmitter release, number of docked vesicles, postsynaptic density length, and expression of plasticity-related proteins leading to the conversion of early LTP into late LTP and of short-term memory into long-term memory. These effects require α7 nicotinic acetylcholine receptors and are mediated through the nitric oxide/cGMP/protein kinase G pathway. The knowledge of Aß function in the healthy brain might be useful to understand the causes leading to its increase and detrimental effect in AD.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Extracellular Fluid/physiology , Memory/physiology , Neurotransmitter Agents/administration & dosage , Peptide Fragments/administration & dosage , Presynaptic Terminals/physiology , Synapses/physiology , Animals , Extracellular Fluid/drug effects , Female , Hippocampus/drug effects , Hippocampus/physiology , Injections, Intraventricular , Male , Memory/drug effects , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Presynaptic Terminals/drug effects , Rats , Rats, Wistar , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
12.
Hum Mol Genet ; 27(6): 1002-1014, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29329433

ABSTRACT

Receptor for Advanced Glycation End products (RAGE) has been implicated in amyloid ß-peptide (Aß)-induced perturbation relevant to the pathogenesis of Alzheimer's disease (AD). However, whether and how RAGE regulates Aß metabolism remains largely unknown. Aß formation arises from aberrant cleavage of amyloid pre-cursor protein (APP) by ß- and γ-secretase. To investigate whether RAGE modulates ß- and γ-secretase activity potentiating Aß formation, we generated mAPP mice with genetic deletion of RAGE (mAPP/RO). These mice displayed reduced cerebral amyloid pathology, inhibited aberrant APP-Aß metabolism by reducing ß- and γ-secretases activity, and attenuated impairment of learning and memory compared with mAPP mice. Similarly, RAGE signal transduction deficient mAPP mice (mAPP/DN-RAGE) exhibited the reduction in Aß40 and Aß42 production and decreased ß-and γ-secretase activity compared with mAPP mice. Furthermore, RAGE-deficient mAPP brain revealed suppression of activation of p38 MAP kinase and glycogen synthase kinase 3ß (GSK3ß). Finally, RAGE siRNA-mediated gene silencing or DN-RAGE-mediated signaling deficiency in the enriched human APP neuronal cells demonstrated suppression of activation of GSK3ß, accompanied with reduction in Aß levels and decrease in ß- and γ-secretases activity. Our findings highlight that RAGE-dependent signaling pathway regulates ß- and γ-secretase cleavage of APP to generate Aß, at least in part through activation of GSK3ß and p38 MAP kinase. RAGE is a potential therapeutic target to limit aberrant APP-Aß metabolism in halting progression of AD.


Subject(s)
Alzheimer Disease/metabolism , Receptor for Advanced Glycation End Products/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Memory/drug effects , Mice , Mice, Transgenic , Neurons/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
13.
PLoS Pathog ; 14(6): e1007061, 2018 06.
Article in English | MEDLINE | ID: mdl-29879225

ABSTRACT

Suppression of HIV replication by antiretroviral therapy (ART) or host immunity can prevent AIDS but not other HIV-associated conditions including neurocognitive impairment (HIV-NCI). Pathogenesis in HIV-suppressed individuals has been attributed to reservoirs of latent-inducible virus in resting CD4+ T cells. Macrophages are persistently infected with HIV but their role as HIV reservoirs in vivo has not been fully explored. Here we show that infection of conventional mice with chimeric HIV, EcoHIV, reproduces physiological conditions for development of disease in people on ART including immunocompetence, stable suppression of HIV replication, persistence of integrated, replication-competent HIV in T cells and macrophages, and manifestation of learning and memory deficits in behavioral tests, termed here murine HIV-NCI. EcoHIV established latent reservoirs in CD4+ T lymphocytes in chronically-infected mice but could be induced by epigenetic modulators ex vivo and in mice. In contrast, macrophages expressed EcoHIV constitutively in mice for up to 16 months; murine leukemia virus (MLV), the donor of gp80 envelope in EcoHIV, did not infect macrophages. Both EcoHIV and MLV were found in brain tissue of infected mice but only EcoHIV induced NCI. Murine HIV-NCI was prevented by antiretroviral prophylaxis but once established neither persistent EcoHIV infection in mice nor NCI could be reversed by long-acting antiretroviral therapy. EcoHIV-infected, athymic mice were more permissive to virus replication in macrophages than were wild-type mice, suffered cognitive dysfunction, as well as increased numbers of monocytes and macrophages infiltrating the brain. Our results suggest an important role of HIV expressing macrophages in HIV neuropathogenesis in hosts with suppressed HIV replication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Disease Reservoirs , HIV Infections/complications , HIV/physiology , Macrophages, Peritoneal/virology , Neurocognitive Disorders/virology , Adoptive Transfer , Aged , Animals , Anti-Retroviral Agents/therapeutic use , Brain/virology , Female , HIV/genetics , HIV/immunology , HIV/pathogenicity , HIV Infections/drug therapy , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Plasmids , Spleen/cytology , Spleen/immunology
14.
Adv Exp Med Biol ; 1266: 39-55, 2020.
Article in English | MEDLINE | ID: mdl-33105494

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disease caused by eventually aggregated amyloid ß (Aß) plaques in degenerating neurons of the aging brain. These aggregated protein plaques mainly consist of Aß fibrils and neurofibrillary tangles (NFTs) of phosphorylated tau protein. Even though some cholinesterase inhibitors, NMDA receptor antagonist, and monoclonal antibodies were developed to inhibit neurodegeneration or activate neural regeneration or clear off the Aß deposits, none of the treatment is effective in improving the cognitive and memory dysfunctions of the AD patients. Thus, stem cell therapy represents a powerful tool for the treatment of AD. In addition to discussing the advents in molecular pathogenesis and animal models of this disease and the treatment approaches using small molecules and immunoglobulins against AD, we will focus on the stem cell sources for AD using neural stem cells (NSCs); embryonic stem cells (ESCs); and mesenchymal stem cells (MSCs) from bone marrow, umbilical cord, and umbilical cord blood. In particular, patient-specific-induced pluripotent stem cells (iPS cells) are proposed as a future prospective and the challenges for the treatment of AD.


Subject(s)
Alzheimer Disease , Stem Cell Transplantation , Alzheimer Disease/therapy , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Animals , Disease Models, Animal , Embryonic Stem Cells , Humans , Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Mice , Mice, Transgenic , Neural Stem Cells , Plaque, Amyloid
15.
Int J Mol Sci ; 21(16)2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32781522

ABSTRACT

Many studies have revealed a central role of p38 MAPK in neuronal plasticity and the regulation of long-term changes in synaptic efficacy, such as long-term potentiation (LTP) and long-term depression (LTD). However, p38 MAPK is classically known as a responsive element to stress stimuli, including neuroinflammation. Specific to the pathophysiology of Alzheimer's disease (AD), several studies have shown that the p38 MAPK cascade is activated either in response to the Aß peptide or in the presence of tauopathies. Here, we describe the role of p38 MAPK in the regulation of synaptic plasticity and its implication in an animal model of neurodegeneration. In particular, recent evidence suggests the p38 MAPK α isoform as a potential neurotherapeutic target, and specific inhibitors have been developed and have proven to be effective in ameliorating synaptic and memory deficits in AD mouse models.


Subject(s)
Synapses/enzymology , Synapses/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Brain/pathology , Humans , Inflammation/pathology , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Synapses/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
16.
Proc Natl Acad Sci U S A ; 113(12): 3347-52, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26951658

ABSTRACT

Elevated levels of the ß-amyloid peptide (Aß) are thought to contribute to cognitive and behavioral impairments observed in Alzheimer's disease (AD). Protein phosphatase 2A (PP2A) participates in multiple molecular pathways implicated in AD, and its expression and activity are reduced in postmortem brains of AD patients. PP2A is regulated by protein methylation, and impaired PP2A methylation is thought to contribute to increased AD risk in hyperhomocysteinemic individuals. To examine further the link between PP2A and AD, we generated transgenic mice that overexpress the PP2A methylesterase, protein phosphatase methylesterase-1 (PME-1), or the PP2A methyltransferase, leucine carboxyl methyltransferase-1 (LCMT-1), and examined the sensitivity of these animals to behavioral and electrophysiological impairments caused by exogenous Aß exposure. We found that PME-1 overexpression enhanced these impairments, whereas LCMT-1 overexpression protected against Aß-induced impairments. Neither transgene affected Aß production or the electrophysiological response to low concentrations of Aß, suggesting that these manipulations selectively affect the pathological response to elevated Aß levels. Together these data identify a molecular mechanism linking PP2A to the development of AD-related cognitive impairments that might be therapeutically exploited to target selectively the pathological effects caused by elevated Aß levels in AD patients.


Subject(s)
Amyloid beta-Peptides/physiology , Cognition Disorders/physiopathology , Protein Phosphatase 2/metabolism , Animals , Behavior, Animal , Methylation , Mice , Mice, Transgenic
17.
J Neurosci ; 37(29): 6926-6937, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28626017

ABSTRACT

High levels of amyloid-ß peptide (Aß) have been related to Alzheimer's disease pathogenesis. However, in the healthy brain, low physiologically relevant concentrations of Aß are necessary for long-term potentiation (LTP) and memory. Because cGMP plays a key role in these processes, here we investigated whether the cyclic nucleotide cGMP influences Aß levels and function during LTP and memory. We demonstrate that the increase of cGMP levels by the phosphodiesterase-5 inhibitors sildenafil and vardenafil induces a parallel release of Aß due to a change in the approximation of amyloid precursor protein (APP) and the ß-site APP cleaving enzyme 1. Moreover, electrophysiological and behavioral studies performed on animals of both sexes showed that blocking Aß function, by using anti-murine Aß antibodies or APP knock-out mice, prevents the cGMP-dependent enhancement of LTP and memory. Our data suggest that cGMP positively regulates Aß levels in the healthy brain which, in turn, boosts synaptic plasticity and memory.SIGNIFICANCE STATEMENT Amyloid-ß (Aß) is a key pathogenetic factor in Alzheimer's disease. However, low concentrations of endogenous Aß, mimicking levels of the peptide in the healthy brain, enhance hippocampal long-term potentiation (LTP) and memory. Because the second messenger cGMP exerts a central role in LTP mechanisms, here we studied whether cGMP affects Aß levels and function during LTP. We show that cGMP enhances Aß production by increasing the APP/BACE-1 convergence in endolysosomal compartments. Moreover, the cGMP-induced enhancement of LTP and memory was disrupted by blockade of Aß, suggesting that the physiological effect of the cyclic nucleotide on LTP and memory is dependent upon Aß.


Subject(s)
Amyloid beta-Peptides/metabolism , Cyclic GMP/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Memory/physiology , Mental Recall/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , Task Performance and Analysis
18.
Neurobiol Dis ; 110: 154-165, 2018 02.
Article in English | MEDLINE | ID: mdl-29217476

ABSTRACT

Small ubiquitin-related modifiers (SUMOs) conjugated or bound to target proteins can affect protein trafficking, processing and solubility. SUMOylation has been suggested to play a role in the amyloid plaque and neurofibrillary tangle pathology of Alzheimer disease (AD) and related neurodegenerative diseases. The current study examines the impact of SUMO1 on processing of the amyloid precursor protein (APP) leading to the production and deposition of the amyloid-ß (Aß) peptide. An in vivo model of these pathways was developed by the generation of double transgenic mice over-expressing human SUMO1 and a mutant APP. The SUMO1-APP transgenics displayed normal APP processing but, at later ages, exhibited increased insoluble Aß and plaque density accompanied by increased dendritic spine loss, more pronounced synaptic and cognitive deficits. These findings suggest a potential impairment in Aß clearance as opposed to increased amyloid production. Examination of microglia indicated a reduction in the SUMO1-APP transgenics which is a possible mechanism for the SUMO1-mediated increase in amyloid load. These findings suggest an indirect activity of SUMO1 possibly in the removal of Aß plaques rather than a direct impact on amyloid generation.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , SUMO-1 Protein/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
19.
Proc Natl Acad Sci U S A ; 112(10): 3122-7, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713349

ABSTRACT

Gene knockout (KO) does not always result in phenotypic changes, possibly due to mechanisms of functional compensation. We have studied mice lacking cGMP-dependent kinase II (cGKII), which phosphorylates GluA1, a subunit of AMPA receptors (AMPARs), and promotes hippocampal long-term potentiation (LTP) through AMPAR trafficking. Acute cGKII inhibition significantly reduces LTP, whereas cGKII KO mice show no LTP impairment. Significantly, the closely related kinase, cGKI, does not compensate for cGKII KO. Here, we describe a previously unidentified pathway in the KO hippocampus that provides functional compensation for the LTP impairment observed when cGKII is acutely inhibited. We found that in cultured cGKII KO hippocampal neurons, cGKII-dependent phosphorylation of inositol 1,4,5-trisphosphate receptors was decreased, reducing cytoplasmic Ca(2+) signals. This led to a reduction of calcineurin activity, thereby stabilizing GluA1 phosphorylation and promoting synaptic expression of Ca(2+)-permeable AMPARs, which in turn induced a previously unidentified form of LTP as a compensatory response in the KO hippocampus. Calcineurin-dependent Ca(2+)-permeable AMPAR expression observed here is also used during activity-dependent homeostatic synaptic plasticity. Thus, a homeostatic mechanism used during activity reduction provides functional compensation for gene KO in the cGKII KO hippocampus.


Subject(s)
Calcium/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Hippocampus/enzymology , Receptors, AMPA/metabolism , Animals , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Hippocampus/drug effects , Hippocampus/physiology , Homeostasis/drug effects , Long-Term Potentiation , Mice , Mice, Knockout , Phosphorylation , Synapses/enzymology , Synapses/metabolism , Tetrodotoxin/pharmacology
20.
Glia ; 65(8): 1302-1316, 2017 08.
Article in English | MEDLINE | ID: mdl-28519902

ABSTRACT

Tau is a microtubule-associated protein exerting several physiological functions in neurons. In Alzheimer's disease (AD) misfolded tau accumulates intraneuronally and leads to axonal degeneration. However, tau has also been found in the extracellular medium. Recent studies indicated that extracellular tau uploaded from neurons causes synaptic dysfunction and contributes to tau pathology propagation. Here we report novel evidence that extracellular tau oligomers are abundantly and rapidly accumulated in astrocytes where they disrupt intracellular Ca2+ signaling and Ca2+ -dependent release of gliotransmitters, especially ATP. Consequently, synaptic vesicle release, the expression of pre- and postsynaptic proteins, and mEPSC frequency and amplitude were reduced in neighboring neurons. Notably, we found that tau uploading from astrocytes required the amyloid precursor protein, APP. Collectively, our findings suggests that astrocytes play a critical role in the synaptotoxic effects of tau via reduced gliotransmitter availability, and that astrocytes are major determinants of tau pathology in AD.


Subject(s)
Astrocytes/metabolism , Hippocampus/cytology , Neurons/physiology , Neurotransmitter Agents/metabolism , Synapses/drug effects , tau Proteins/toxicity , Adenosine Triphosphate/metabolism , Alzheimer Disease/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Coculture Techniques , Embryo, Mammalian , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholinos/toxicity , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Receptors, AMPA/metabolism , tau Proteins/chemistry , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL