Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(5): e0304555, 2024.
Article in English | MEDLINE | ID: mdl-38820269

ABSTRACT

Inflammation is a key driver in the pathogenesis of cystic fibrosis (CF). We assessed the effectiveness of elexacaftor/tezacaftor/ivacaftor (ETI) therapy on downregulating systemic and immune cell-derived inflammatory cytokines. We also monitored the impact of ETI therapy on clinical outcome. Adults with CF, heterozygous for F508del (n = 19), were assessed at baseline, one month and three months following ETI therapy, and clinical outcomes were measured, including sweat chloride, lung function, weight, neutrophil count and C-reactive protein (CRP). Cytokine quantifications were measured in serum and following stimulation of peripheral blood mononuclear cells (PBMCs) with lipopolysaccharide (LPS) and adenosine triphosphate and analysed using LEGEND plex™ Human Inflammation Panel 1 by flow cytometry (n = 19). ASC specks were measured in serum and caspase-1 activity and mRNA levels determined from stimulated PBMCs were determined. Patients remained stable over the study period. ETI therapy resulted in decreased sweat chloride concentrations (p < 0.0001), CRP (p = 0.0112) and neutrophil count (p = 0.0216) and increased percent predicted forced expiratory volume (ppFEV1) (p = 0.0399) from baseline to three months, alongside a trend increase in weight. Three months of ETI significantly decreased IL-18 (p< 0.0011, p < 0.0001), IL-1ß (p<0.0013, p = 0.0476), IL-6 (p = 0.0109, p = 0.0216) and TNF (p = 0.0028, p = 0.0033) levels in CF serum and following PBMCs stimulation respectively. The corresponding mRNA levels were also found to be reduced in stimulated PBMCs, as well as reduced ASC specks and caspase-1 levels, indicative of NLRP3-mediated production of pro-inflammatory cytokines, IL-1ß and IL-18. While ETI therapy is highly effective at reducing sweat chloride and improving lung function, it also displays potent anti-inflammatory properties, which are likely to contribute to improved long-term clinical outcomes.


Subject(s)
Aminophenols , Anti-Inflammatory Agents , Benzodioxoles , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Cytokines , Indoles , Quinolones , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Benzodioxoles/therapeutic use , Benzodioxoles/pharmacology , Adult , Aminophenols/therapeutic use , Female , Indoles/therapeutic use , Indoles/pharmacology , Male , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Quinolones/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Cytokines/blood , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Young Adult , Pyridines/therapeutic use , Pyridines/pharmacology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , C-Reactive Protein/metabolism , Pyrroles/therapeutic use , Pyrroles/pharmacology , Sweat/chemistry , Sweat/metabolism , Pyrrolidines
2.
Sci Rep ; 11(1): 8194, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33854181

ABSTRACT

Hydrogen sulfide (H2S) is gaining interest as a mammalian signalling molecule with wide ranging effects. S-sulfhydration is one mechanism that is emerging as a key post translational modification through which H2S acts. Ion channels and neuronal receptors are key target proteins for S-sulfhydration and this can influence a range of neuronal functions. Voltage-gated K+ channels, including Kv2.1, are fundamental components of neuronal excitability. Here, we show that both recombinant and native rat Kv2.1 channels are inhibited by the H2S donors, NaHS and GYY4137. Biochemical investigations revealed that NaHS treatment leads to S-sulfhydration of the full length wild type Kv2.1 protein which was absent (as was functional regulation by H2S) in the C73A mutant form of the channel. Functional experiments utilising primary rat hippocampal neurons indicated that NaHS augments action potential firing and thereby increases neuronal excitability. These studies highlight an important role for H2S in shaping cellular excitability through S-sulfhydration of Kv2.1 at C73 within the central nervous system.


Subject(s)
Hippocampus/cytology , Hydrogen Sulfide/pharmacology , Shab Potassium Channels/genetics , Shab Potassium Channels/metabolism , Action Potentials , Animals , Cells, Cultured , Down-Regulation , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Morpholines/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organothiophosphorus Compounds/pharmacology , Phosphorylation , Primary Cell Culture , Rats
3.
Front Pharmacol ; 12: 643254, 2021.
Article in English | MEDLINE | ID: mdl-33776778

ABSTRACT

The prevalence of neurodegenerative disease has increased significantly in recent years, and with a rapidly aging global population, this trend is expected to continue. These diseases are characterised by a progressive neuronal loss in the brain or peripheral nervous system, and generally involve protein aggregation, as well as metabolic abnormalities and immune dysregulation. Although the vast majority of neurodegeneration is idiopathic, there are many known genetic and environmental triggers. In the past decade, research exploring low-grade systemic inflammation and its impact on the development and progression of neurodegenerative disease has increased. A particular research focus has been whether systemic inflammation arises only as a secondary effect of disease or is also a cause of pathology. The inflammasomes, and more specifically the NLRP3 inflammasome, a crucial component of the innate immune system, is usually activated in response to infection or tissue damage. Dysregulation of the NLRP3 inflammasome has been implicated in the progression of several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and prion diseases. This review aims to summarise current literature on the role of the NLRP3 inflammasome in the pathogenesis of neurodegenerative diseases, and recent work investigating NLRP3 inflammasome inhibition as a potential future therapy.

4.
Cell Mol Life Sci ; 77(22): 4485-4503, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32367193

ABSTRACT

Cystic fibrosis (CF) is one of the most common life-limiting recessive genetic disorders in Caucasians, caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). CF is a multi-organ disease that involves the lungs, pancreas, sweat glands, digestive and reproductive systems and several other tissues. This debilitating condition is associated with recurrent lower respiratory tract bacterial and viral infections, as well as inflammatory complications that may eventually lead to pulmonary failure. Immune cells play a crucial role in protecting the organs against opportunistic infections and also in the regulation of tissue homeostasis. Innate immune cells are generally affected by CFTR mutations in patients with CF, leading to dysregulation of several cellular signalling pathways that are in continuous use by these cells to elicit a proper immune response. There is substantial evidence to show that airway epithelial cells, neutrophils, monocytes and macrophages all contribute to the pathogenesis of CF, underlying the importance of the CFTR in innate immune responses. The goal of this review is to put into context the important role of the CFTR in different innate immune cells and how CFTR dysfunction contributes to the pathogenesis of CF, highlighting several signalling pathways that may be dysregulated in cells with CFTR mutations.


Subject(s)
Cystic Fibrosis/genetics , Cystic Fibrosis/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Mutation/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Humans , Mutation/immunology
5.
Elife ; 92020 03 02.
Article in English | MEDLINE | ID: mdl-32118580

ABSTRACT

Previously, we showed that serum and monocytes from patients with CF exhibit an enhanced NLRP3-inflammasome signature with increased IL-18, IL-1ß, caspase-1 activity and ASC speck release (Scambler et al. eLife 2019). Here we show that CFTR modulators down regulate this exaggerated proinflammatory response following LPS/ATP stimulation. In vitro application of ivacaftor/lumacaftor or ivacaftor/tezacaftor to CF monocytes showed a significant reduction in IL-18, whereas IL-1ß was only reduced with ivacaftor/tezacaftor. Thirteen adults starting ivacaftor/lumacaftor and eight starting ivacaftor/tezacaftor were assessed over three months. Serum IL-18 and TNF decreased significantly with treatments, but IL-1ß only declined following ivacaftor/tezacaftor. In (LPS/ATP-stimulated) PBMCs, IL-18/TNF/caspase-1 were all significantly decreased and IL-10 was increased with both combinations. Ivacaftor/tezacaftor alone showed a significant reduction in IL-1ß and pro-IL-1ß mRNA. This study demonstrates that these CFTR modulator combinations have potent anti-inflammatory properties, in addition to their ability to stimulate CFTR function, which could contribute to improved clinical outcomes.


Subject(s)
Aminophenols/therapeutic use , Aminopyridines/therapeutic use , Benzodioxoles/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/metabolism , Indoles/therapeutic use , Inflammation/metabolism , Quinolones/therapeutic use , Adult , Aminophenols/administration & dosage , Aminopyridines/administration & dosage , Benzodioxoles/administration & dosage , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytokines/metabolism , Down-Regulation , Drug Therapy, Combination , Female , Humans , Indoles/administration & dosage , Inflammation/diet therapy , Interleukin-18/blood , Interleukin-1beta/blood , Male , Monocytes/drug effects , Monocytes/metabolism , Quinolones/administration & dosage , Tumor Necrosis Factor-alpha/blood , Young Adult
6.
Elife ; 82019 09 18.
Article in English | MEDLINE | ID: mdl-31532390

ABSTRACT

Cystic Fibrosis (CF) is a monogenic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR-mediated chloride and bicarbonate transport, with dysregulation of epithelial sodium channels (ENaC). These changes alter fluid and electrolyte homeostasis and result in an exaggerated proinflammatory response driven, in part, by infection. We tested the hypothesis that NLRP3 inflammasome activation and ENaC upregulation drives exaggerated innate-immune responses in this multisystem disease. We identify an enhanced proinflammatory signature, as evidenced by increased levels of IL-18, IL-1ß, caspase-1 activity and ASC-speck release in monocytes, epithelia and serum with CF-associated mutations; these differences were reversed by pretreatment with NLRP3 inflammasome inhibitors and notably, inhibition of amiloride-sensitive sodium (Na+) channels. Overexpression of ß-ENaC, in the absence of CFTR dysfunction, increased NLRP3-mediated inflammation, indicating that dysregulated, ENaC-dependent signalling may drive exaggerated inflammatory responses in CF. These data support a role for sodium in modulating NLRP3 inflammasome activation.


Subject(s)
Cystic Fibrosis/pathology , Epithelial Sodium Channels/metabolism , Inflammation/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sodium/metabolism , Cell Line , Humans , Immunity, Innate
7.
Front Immunol ; 10: 1789, 2019.
Article in English | MEDLINE | ID: mdl-31428093

ABSTRACT

Cystic Fibrosis (CF) is a recessive genetic disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). CFTR mutations cause dysregulation of channel function with intracellular accumulation of misfolded proteins and endoplasmic reticulum (ER) stress, with activation of the IRE1α-XBP1 pathway that regulates a subset of unfolded protein response (UPR) genes. This pathway regulates a group of genes that control proinflammatory and metabolic responses in different immune cells; however, the metabolic state of immune cells and the role of this pathway in CF remain elusive. Our results indicate that only innate immune cells from CF patients present increased levels of ER stress, mainly affecting neutrophils, monocytes, and macrophages. An overactive IRE1α-XBP1 pathway reprograms CF M1 macrophages toward an increased metabolic state, with increased glycolytic rates and mitochondrial function, associated with exaggerated production of TNF and IL-6. This hyper-metabolic state, seen in CF macrophages, is reversed by inhibiting the RNase domain of IRE1α, thereby decreasing the increased glycolic rates, mitochondrial function and inflammation. Altogether, our results indicate that innate immune cells from CF patients are primarily affected by ER stress. Moreover, the IRE1α-XBP1 pathway of the UPR is responsible for the hyper-metabolic state seen in CF macrophages, which is associated with the exaggerated inflammatory response. Modulating ER stress, metabolism and inflammation, by targeting IRE1α, may improve the metabolic fitness of macrophages, and other immune cells in CF and other immune-related disorders.


Subject(s)
Cystic Fibrosis/metabolism , Endoribonucleases/metabolism , Inflammation/metabolism , Macrophages/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/physiology , Adult , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Female , Humans , Male , Middle Aged , Mitochondria/metabolism , Signal Transduction/physiology , X-Box Binding Protein 1/metabolism , Young Adult
8.
Int Immunol ; 31(10): 639-648, 2019 09 18.
Article in English | MEDLINE | ID: mdl-30838383

ABSTRACT

Autoinflammatory syndromes are a group of disorders characterized by recurring episodes of inflammation as a result of specific defects in the innate immune system. Patients with autoinflammatory disease present with recurrent outbreaks of chronic systemic inflammation that are mediated by innate immune cells, for the most part. A number of these diseases arise from defects in the tumour necrosis factor receptor (TNFR) signalling pathway leading to elevated levels of inflammatory cytokines. Elucidation of the molecular mechanisms of these recently defined autoinflammatory diseases has led to a greater understanding of the mechanisms of action of key molecules involved in TNFR signalling, particularly those involved in ubiquitination, as found in haploinsufficiency of A20 (HA20), otulipenia/OTULIN-related autoinflammatory syndrome (ORAS) and linear ubiquitin chain assembly complex (LUBAC) deficiency. In this review, we also address other TNFR signalling disorders such as TNFR-associated periodic syndrome (TRAPS), RELA haploinsufficiency, RIPK1-associated immunodeficiency and autoinflammation, X-linked ectodermal dysplasia and immunodeficiency (X-EDA-ID) and we review the most recent advances surrounding these diseases and therapeutic approaches currently used to target these diseases. Finally, we explore therapeutic advances in TNF-related immune-based therapies and explore new approaches to target disease-specific modulation of autoinflammatory diseases.


Subject(s)
Autoimmune Diseases/immunology , Inflammation/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction , Animals , Autoimmune Diseases/genetics , Humans , Inflammation/genetics , Signal Transduction/immunology
9.
J Biol Chem ; 294(17): 7085-7097, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30872401

ABSTRACT

The cellular prion protein (PrPC) is a key neuronal receptor for ß-amyloid oligomers (AßO), mediating their neurotoxicity, which contributes to the neurodegeneration in Alzheimer's disease (AD). Similarly to the amyloid precursor protein (APP), PrPC is proteolytically cleaved from the cell surface by a disintegrin and metalloprotease, ADAM10. We hypothesized that ADAM10-modulated PrPC shedding would alter the cellular binding and cytotoxicity of AßO. Here, we found that in human neuroblastoma cells, activation of ADAM10 with the muscarinic agonist carbachol promotes PrPC shedding and reduces the binding of AßO to the cell surface, which could be blocked with an ADAM10 inhibitor. Conversely, siRNA-mediated ADAM10 knockdown reduced PrPC shedding and increased AßO binding, which was blocked by the PrPC-specific antibody 6D11. The retinoic acid receptor analog acitretin, which up-regulates ADAM10, also promoted PrPC shedding and decreased AßO binding in the neuroblastoma cells and in human induced pluripotent stem cell (iPSC)-derived cortical neurons. Pretreatment with acitretin abolished activation of Fyn kinase and prevented an increase in reactive oxygen species caused by AßO binding to PrPC Besides blocking AßO binding and toxicity, acitretin also increased the nonamyloidogenic processing of APP. However, in the iPSC-derived neurons, Aß and other amyloidogenic processing products did not exhibit a reciprocal decrease upon acitretin treatment. These results indicate that by promoting the shedding of PrPC in human neurons, ADAM10 activation prevents the binding and cytotoxicity of AßO, revealing a potential therapeutic benefit of ADAM10 activation in AD.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Biopolymers/metabolism , Membrane Proteins/metabolism , ADAM10 Protein/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Cell Line, Tumor , Enzyme Activation , Gene Knockdown Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/genetics , Prion Proteins/metabolism , Protein Binding , Proteolysis , Reactive Oxygen Species/metabolism
10.
Alzheimers Dement ; 14(3): 306-317, 2018 03.
Article in English | MEDLINE | ID: mdl-29055813

ABSTRACT

INTRODUCTION: Synapse loss is the structural correlate of the cognitive decline indicative of dementia. In the brains of Alzheimer's disease sufferers, amyloid ß (Aß) peptides aggregate to form senile plaques but as soluble peptides are toxic to synapses. We previously demonstrated that Aß induces Dickkopf-1 (Dkk1), which in turn activates the Wnt-planar cell polarity (Wnt-PCP) pathway to drive tau pathology and neuronal death. METHODS: We compared the effects of Aß and of Dkk1 on synapse morphology and memory impairment while inhibiting or silencing key elements of the Wnt-PCP pathway. RESULTS: We demonstrate that Aß synaptotoxicity is also Dkk1 and Wnt-PCP dependent, mediated by the arm of Wnt-PCP regulating actin cytoskeletal dynamics via Daam1, RhoA and ROCK, and can be blocked by the drug fasudil. DISCUSSION: Our data add to the importance of aberrant Wnt signaling in Alzheimer's disease neuropathology and indicate that fasudil could be repurposed as a treatment for the disease.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amyloid beta-Peptides/metabolism , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Synapses/metabolism , Wnt Signaling Pathway , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacokinetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Female , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Neuroprotective Agents/pharmacokinetics , Nootropic Agents/pharmacokinetics , Primary Cell Culture , RNA, Messenger/metabolism , Rats , Synapses/drug effects , Synapses/pathology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology
11.
J Biol Chem ; 291(7): 3174-83, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26719327

ABSTRACT

Several different receptor proteins have been identified that bind monomeric, oligomeric, or fibrillar forms of amyloid-ß (Aß). "Good" receptors internalize Aß or promote its transcytosis out of the brain, whereas "bad" receptors bind oligomeric forms of Aß that are largely responsible for the synapticloss, memory impairments, and neurotoxicity that underlie Alzheimer disease. The prion protein both removes Aß from the brain and transduces the toxic actions of Aß. The clustering of distinct receptors in cell surface signaling platforms likely underlies the actions of distinct oligomeric species of Aß. These Aß receptor-signaling platforms provide opportunities for therapeutic intervention in Alzheimer disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Cell Surface/agonists , Signal Transduction , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Animals , Apoptosis/drug effects , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/agonists , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Molecular Targeted Therapy , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Neurons/drug effects , Neurons/pathology , Nootropic Agents/pharmacology , Nootropic Agents/therapeutic use , PrPC Proteins/agonists , PrPC Proteins/antagonists & inhibitors , PrPC Proteins/metabolism , Protein Aggregates/drug effects , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/prevention & control , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptor Aggregation/drug effects , Receptor for Advanced Glycation End Products/agonists , Receptor for Advanced Glycation End Products/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Transcytosis/drug effects
12.
Front Cell Dev Biol ; 2: 41, 2014.
Article in English | MEDLINE | ID: mdl-25364748

ABSTRACT

Dysregulation of neuronal zinc homeostasis plays a major role in many processes related to brain aging and neurodegenerative diseases, including Alzheimer's disease (AD). Yet, despite the critical role of zinc in neuronal function, the cellular mechanisms underpinning its homeostatic control are far from clear. We reported that the cellular prion protein (PrP(C)) is involved in the uptake of zinc into neurons. This PrP(C)-mediated zinc influx required the metal-binding octapeptide repeats in PrP(C) and the presence of the zinc permeable AMPA channel with which PrP(C) directly interacted. Together with the observation that PrP(C) is evolutionarily related to the ZIP family of zinc transporters, these studies indicate that PrP(C) plays a key role in neuronal zinc homeostasis. Therefore, PrP(C) could contribute to cognitive health and protect against age-related zinc dyshomeostasis but PrP(C) has also been identified as a receptor for amyloid-ß oligomers which accumulate in the brains of those with AD. We propose that the different roles that PrP(C) has are due to its interaction with different ligands and/or co-receptors in lipid raft-based signaling/transport complexes.

13.
Biosens Bioelectron ; 56: 83-90, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24480125

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia, with over 37 million sufferers worldwide and a global cost of over $600 billion. There is currently no cure for AD and no reliable method of diagnosis other than post-mortem brain examination. The development of a point-of-care test for AD is an urgent requirement in order to provide earlier diagnosis and, thus, useful therapeutic intervention. Here, we present a novel, label-free impedimetric biosensor for the specific detection of amyloid-beta oligomers (AßO), which are the primary neurotoxic species in AD. AßO have been proposed as the best biomarker for AD and levels of AßO in the blood have been found to correlate with cerebrospinal fluid load. The biorecognition element of our biosensor is a fragment of the cellular prion protein (PrP(C), residues 95-110), a highly expressed synaptic protein which mediates the neuronal binding and toxicity of AßO. During the layer-by-layer sensor construction, biotinylated PrP(C) (95-110) was attached via a biotin/NeutrAvidin bridge to polymer-functionalised gold screen-printed electrodes. Electrochemical impedance spectroscopy (EIS), cyclic voltammetry and scanning electron microscopy were used to validate biosensor assembly and functionality. EIS was employed for biosensor interrogation in the presence of Aß oligomers or monomers. The biosensor was specific for the detection of synthetic AßO and gave a linear response, without significant detection of monomeric Aß, down to an equivalent AßO concentration of ~0.5 pM. The biosensor was also able to detect natural, cell-derived AßO present in conditioned medium. The eventual commercialisation of this biosensor system could allow for the early diagnosis and disease monitoring of AD.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/analysis , Biosensing Techniques/methods , Dielectric Spectroscopy/methods , Amino Acid Sequence , Animals , CHO Cells , Cricetulus , Equipment Design , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , PrPC Proteins/chemistry
14.
Prion ; 7(3): 203-8, 2013.
Article in English | MEDLINE | ID: mdl-23764834

ABSTRACT

Zinc, the most abundant trace metal in the brain, has numerous functions in health and disease. It is released into the synaptic cleft alongside glutamate and this connection between zinc and glutamatergic neurotransmission allows the ion to modulate overall excitability of the brain and influence synaptic plasticity. To maintain healthy synapses, extracellular zinc levels need to be tightly regulated. We recently reported that the cellular prion protein (PrP (C) ) can directly influence neuronal zinc concentrations by promoting zinc uptake via AMPA receptors. The octapeptide repeat region of PrP (C) is involved in zinc sensing or scavenging and the AMPA receptor provides the channel for transport of the metal across the membrane, facilitated by a direct interaction between the N-terminal polybasic region of PrP (C) and AMPA receptors. PrP (C) has been evolutionarily linked to the Zrt/Irt-like protein (ZIP) metal ion transport family with the C-terminus of PrP (C) sharing sequence similarities with the N-terminal extracellular domains of ZIP 5, 6 and 10. By incorporating the properties of ZIP transporters (both zinc sensing and zinc transport) into two existing neuronal proteins, (PrP (C) as zinc sensor, AMPA receptor as zinc transporter), neuronal cells are enhancing their biological efficiency and functionality.


Subject(s)
Brain/metabolism , Neurons/metabolism , PrPC Proteins/metabolism , Prion Diseases/metabolism , Receptors, AMPA/metabolism , Zinc/metabolism , Animals , Carrier Proteins/metabolism , Humans , Models, Molecular
15.
J Biol Chem ; 288(13): 8935-51, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23386614

ABSTRACT

Soluble oligomers of the amyloid-ß (Aß) peptide cause neurotoxicity, synaptic dysfunction, and memory impairments that underlie Alzheimer disease (AD). The cellular prion protein (PrP(C)) was recently identified as a high affinity neuronal receptor for Aß oligomers. We report that fibrillar Aß oligomers recognized by the OC antibody, which have been shown to correlate with the onset and severity of AD, bind preferentially to cells and neurons expressing PrP(C). The binding of Aß oligomers to cell surface PrP(C), as well as their downstream activation of Fyn kinase, was dependent on the integrity of cholesterol-rich lipid rafts. In SH-SY5Y cells, fluorescence microscopy and co-localization with subcellular markers revealed that the Aß oligomers co-internalized with PrP(C), accumulated in endosomes, and subsequently trafficked to lysosomes. The cell surface binding, internalization, and downstream toxicity of Aß oligomers was dependent on the transmembrane low density lipoprotein receptor-related protein-1 (LRP1). The binding of Aß oligomers to cell surface PrP(C) impaired its ability to inhibit the activity of the ß-secretase BACE1, which cleaves the amyloid precursor protein to produce Aß. The green tea polyphenol (-)-epigallocatechin gallate and the red wine extract resveratrol both remodeled the fibrillar conformation of Aß oligomers. The resulting nonfibrillar oligomers displayed significantly reduced binding to PrP(C)-expressing cells and were no longer cytotoxic. These data indicate that soluble, fibrillar Aß oligomers bind to PrP(C) in a conformation-dependent manner and require the integrity of lipid rafts and the transmembrane LRP1 for their cytotoxicity, thus revealing potential targets to alleviate the neurotoxic properties of Aß oligomers in AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Prions/chemistry , Animals , Caspase 3/metabolism , Catechin/analogs & derivatives , Catechin/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Hippocampus/metabolism , Humans , Microscopy, Fluorescence/methods , Neurons/metabolism , Protein Binding , Proto-Oncogene Proteins c-fyn/metabolism , RNA Interference , Rats , Rats, Wistar , Resveratrol , Stilbenes/pharmacology , Tea/metabolism
16.
Nat Commun ; 3: 1134, 2012.
Article in English | MEDLINE | ID: mdl-23072804

ABSTRACT

Zinc is released into the synaptic cleft upon exocytotic stimuli, although the mechanism for its reuptake into neurons is unresolved. Here we show that the cellular prion protein enhances the uptake of zinc into neuronal cells. This prion-protein-mediated zinc influx requires the octapeptide repeats and amino-terminal polybasic region in the prion protein, but not its endocytosis. Selective antagonists of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptors block the prion protein-mediated zinc uptake, and the prion protein co-immunoprecipitates with both GluA1 and GluA2 AMPA receptor subunits. Zinc-sensitive intracellular tyrosine phosphatase activity is decreased in cells expressing prion protein and increased in the brains of prion-protein-null mice, providing evidence of a physiological consequence of this process. Prion protein-mediated zinc uptake is ablated in cells expressing familial associated mutants of the protein and in prion-infected cells. These data suggest that alterations in the cellular prion protein-mediated zinc uptake may contribute to neurodegeneration in prion and other neurodegenerative diseases.


Subject(s)
Endocytosis , Neurons/metabolism , Prions/metabolism , Zinc/metabolism , Animals , Cell Line, Tumor , GPI-Linked Proteins/metabolism , Humans , Mice , Nerve Tissue Proteins/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Prion Proteins , Prions/chemistry , Protein Subunits/metabolism , Protein Tyrosine Phosphatases/metabolism , Rats , Receptors, AMPA/metabolism , Transfection
17.
Prion ; 6(3): 217-22, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22449984

ABSTRACT

Alzheimer disease (AD) is characterized by the amyloidogenic processing of the amyloid precursor protein (APP), culminating in the accumulation of amyloid-ß peptides in the brain. The enzymatic action of the ß-secretase, BACE1 is the rate-limiting step in this amyloidogenic processing of APP. BACE1 cleavage of wild-type APP (APPWT) is inhibited by the cellular prion protein (PrP (C) ). Our recent study has revealed the molecular and cellular mechanisms behind this observation by showing that PrP (C) directly interacts with the pro-domain of BACE1 in the trans-Golgi network (TGN), decreasing the amount of BACE1 at the cell surface and in endosomes where it cleaves APPWT, while increasing BACE1 in the TGN where it preferentially cleaves APP with the Swedish mutation (APPSwe). PrP (C) deletion in transgenic mice expressing the Swedish and Indiana familial mutations (APPSwe,Ind) failed to affect amyloid-ß accumulation, which is explained by the differential subcellular sites of action of BACE1 toward APPWT and APPSwe. This, together with our observation that PrP (C) is reduced in sporadic but not familial AD brain, suggests that PrP (C) plays a key protective role against sporadic AD. It also highlights the need for an APPWT transgenic mouse model to understand the molecular and cellular mechanisms underlying sporadic AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , PrPC Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Brain/metabolism , Humans , Mice
18.
J Biol Chem ; 286(38): 33489-500, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21795680

ABSTRACT

In Alzheimer disease amyloid-ß (Aß) peptides derived from the amyloid precursor protein (APP) accumulate in the brain. Cleavage of APP by the ß-secretase BACE1 is the rate-limiting step in the production of Aß. We have reported previously that the cellular prion protein (PrP(C)) inhibited the action of BACE1 toward human wild type APP (APP(WT)) in cellular models and that the levels of endogenous murine Aß were significantly increased in PrP(C)-null mouse brain. Here we investigated the molecular and cellular mechanisms underlying this observation. PrP(C) interacted directly with the prodomain of the immature Golgi-localized form of BACE1. This interaction decreased BACE1 at the cell surface and in endosomes where it preferentially cleaves APP(WT) but increased it in the Golgi where it preferentially cleaves APP with the Swedish mutation (APP(Swe)). In transgenic mice expressing human APP with the Swedish and Indiana familial mutations (APP(Swe,Ind)), PrP(C) deletion had no influence on APP proteolytic processing, Aß plaque deposition, or levels of soluble Aß or Aß oligomers. In cells, although PrP(C) inhibited the action of BACE1 on APP(WT), it did not inhibit BACE1 activity toward APP(Swe). The differential subcellular location of the BACE1 cleavage of APP(Swe) relative to APP(WT) provides an explanation for the failure of PrP(C) deletion to affect Aß accumulation in APP(Swe,Ind) mice. Thus, although PrP(C) exerts no control on cleavage of APP(Swe) by BACE1, it has a profound influence on the cleavage of APP(WT), suggesting that PrP(C) may be a key protective player against sporadic Alzheimer disease.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Prions/metabolism , Amino Acid Sequence , Amyloid Precursor Protein Secretases/chemistry , Animals , Aspartic Acid Endopeptidases/chemistry , Binding Sites , Cell Line , Gene Deletion , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/metabolism , Subcellular Fractions/metabolism
19.
Expert Opin Ther Targets ; 12(6): 693-704, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18479216

ABSTRACT

BACKGROUND: The amyloid beta (Abeta) peptide is critical to the development of Alzheimer's disease (AD), the major neurodegenerative disease of the elderly for which there is currently no cure. OBJECTIVE: To review the literature on emerging treatments and potential therapeutic strategies for AD. METHODS: Available published literature and information from pharmaceutical companies was utilised. RESULTS/CONCLUSION: Several of the current treatments to combat AD are aimed at inhibiting the production, blocking the oligomerisation/aggregation or enhancing the degradation of Abeta. In our opinion, albeit based on limited available data, a future potential therapeutic strategy is to mimic the mechanism by which the normal cellular form of the prion protein inhibits the beta-secretase beta-site amyloid precursor protein cleaving enzyme-1 (BACE1), and hence the production of Abeta.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Humans , Prions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...