Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Clin Exp Rheumatol ; 40(2): 414-424, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35225225

ABSTRACT

Inclusion body myositis (IBM) is characterised by infiltration of CD8+ T-cells and signs of protein aggregation such as rimmed vacuoles and inclusion bodies. Aggregated proteins include those present in neurodegenerative diseases, and also those involved in protein homeostasis. The aim of this review is to discuss the pathological effects of protein aggregates and the process of aggregation following immune attack in IBM. Immune attack is likely to cause protein aggregation by impairing endoplasmic reticulum (ER) and mitochondrial function. Apoptotic and necrotic pathways are activated, possibly leading to nucleo-cytoplasmic coagulation. Overexpression of nuclear and ribosomal proteins in rimmed vacuoles suggests that the vacuoles develop from the collapse of myonuclei and the surrounding ER. Aggregated proteins can activate the NLR family pyrin domain containing 3 (NLRP3) inflammasome or provoke a humoral immune response. Heat shock proteins, ribosomal proteins and protein fragments may provoke interferon-gamma and cytotoxic T-cell responses in a similar manner to Mycobacterium tuberculosis antigens. Persistent provocation can lead to T-cell large granular lymphocytic leukaemia which is resistant to immunosuppression, and would explain the progression from polymyositis to IBM. Protein aggregates may impair the cellular machinery, and proteins may propagate along a myocyte in a prion-like manner. These pathological mechanisms may prevent myocyte regeneration following damage from eccentric muscle contraction, causing weakness and atrophy in a characteristic pattern. Further understanding of the mechanisms of protein aggregation in IBM may lead to additional therapies as well as novel muscle and blood biomarkers. Earlier diagnosis and treatment may result in improved outcomes when effective therapies are available.


Subject(s)
Myositis, Inclusion Body , Myositis , Biomarkers/metabolism , Heat-Shock Proteins , Humans , Myositis/pathology , Protein Aggregates
2.
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
3.
J Neurochem ; 149(3): 399-412, 2019 05.
Article in English | MEDLINE | ID: mdl-30664241

ABSTRACT

The generation of the amyloid-ß (Aß) peptides from the amyloid precursor protein (APP) through sequential proteolysis by ß- and γ-secretases is a key pathological event in the initiation and propagation of Alzheimer's disease. Aß and the transcriptionally active APP intracellular domain are generated preferentially from the APP695 isoform compared to the longer APP751 isoform. As the Aß and amyloid precursor protein intracellular domain produced from cleavage of APP695 and APP751 are identical we hypothesised that the two isoforms have differences within their interactomes which mediate the differential processing of the two isoforms. To investigate this, we applied a proteomics-based approach to identify differences in the interactomes of the APP695 and APP751 isoforms. Using stable isotope labelling of amino acids in cell culture and quantitative proteomics, we compared the interactomes of APP695 and APP751 expressed in human SH-SY5Y cells. Through this approach, we identified enrichment of proteins involved in mitochondrial function, the nuclear pore and nuclear transport specifically in the APP695 interactome. Further interrogation of the APP interactome and subsequent experimental validation (co-immunoprecipitation and siRNA knockdown) revealed GAP43 as a specific modulator of APP751 proteolysis, altering Aß generation. Our data indicate that interrogation of the APP interactome can be exploited to identify proteins which influence APP proteolysis and Aß production in an isoform dependent-manner. Cover Image for this issue: doi: 10.1111/jnc.14504.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Line, Tumor , Humans , Mitochondria/metabolism , Neurons/metabolism , Protein Isoforms , Proteomics
4.
Neurochem Res ; 43(12): 2179-2198, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30387070

ABSTRACT

Developing cellular models of sporadic Alzheimer's disease (sAD) is challenging due to the unknown initiator of disease onset and the slow disease progression that takes many years to develop in vivo. The use of human induced pluripotent stem cells (iPSCs) has revolutionised the opportunities to model AD pathology, investigate disease mechanisms and screen potential drugs. The majority of this work has, however, used cells derived from patients with familial AD (fAD) where specific genetic mutations drive disease onset. While these provide excellent models to investigate the downstream pathways involved in neuronal toxicity and ultimately neuronal death that leads to AD, they provide little insight into the causes and mechanisms driving the development of sAD. In this review we compare the data obtained from fAD and sAD iPSC-derived cell lines, identify the inconsistencies that exist in sAD models and highlight the potential role of Aß clearance mechanisms, a relatively under-investigated area in iPSC-derived models, in the study of AD. We discuss the development of more physiologically relevant models using co-culture and three-dimensional culture of iPSC-derived neurons with glial cells. Finally, we evaluate whether we can develop better, more consistent models for sAD research using genetic stratification of iPSCs and identification of genetic and environmental risk factors that could be used to initiate disease onset for modelling sAD. These considerations provide exciting opportunities to develop more relevant iPSC models of sAD which can help drive our understanding of disease mechanisms and identify new therapeutic targets.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/physiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Coculture Techniques/methods , Humans , Induced Pluripotent Stem Cells/pathology , tau Proteins/genetics , tau Proteins/metabolism
5.
Biometals ; 31(2): 267-276, 2018 04.
Article in English | MEDLINE | ID: mdl-29516299

ABSTRACT

Sporadic Alzheimer's disease (AD) is a neurodegenerative disorder that causes the most prevalent form of age-related dementia but its pathogenesis remains obscure. Altered regulation of metals, particularly pan-cerebral copper deficiency, and more regionally-localized perturbation of other metals, are prominent in AD brain although data on how these CNS perturbations are reflected in the peripheral bloodstream are inconsistent to date. To assess the potential use of metal dysregulation to generate biomarkers in AD, we performed a case-control study of seven essential metals and selenium, measured by inductively coupled plasma mass-spectrometry, in samples from AD and matched control cases. Metals were sodium, potassium, calcium, magnesium, iron, zinc, and copper. In the whole study-group and in female participants, plasma metal levels did not differ between cases and controls. In males by contrast, there was moderate evidence that zinc levels trended towards increase in AD [10.8 (10.2-11.5)] µmol/L, mean (± 95% CI; P = 0.021) compared with controls [10.2 (9.6-10.4)]. Thus alterations in plasma zinc levels differed between genders in AD. In correlational analysis, there was evidence for an increased number of 'strong' metal co-regulations in AD cases and differential co-modulations of metal pairs: copper-sodium (Rcontrol = - 0.03, RAD = 0.65; P = 0.009), and copper-calcium (Rcontrol = - 0.01, RAD = 0.65; P = 0.01) were significant in AD males, potentially consistent with reported evidence for dysregulation of copper in severely damaged brain regions in AD. In conclusion, our data suggest that the measurement of metals co-regulation in plasma may provide a useful representation of those metal perturbations taking place in the AD brain and therefore might be useful as plasma-based biomarkers.


Subject(s)
Alzheimer Disease/blood , Biomarkers/blood , Dementia/blood , Metals/blood , Calcium/blood , Copper/blood , Female , Humans , Iron/blood , Magnesium/blood , Male , Potassium/blood , Selenium/blood , Sex Characteristics , Sodium/blood , Zinc/blood
6.
J Biol Chem ; 291(37): 19235-44, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27474742

ABSTRACT

Proteolysis of the amyloid precursor protein (APP) liberates various fragments including the proposed initiator of Alzheimer disease-associated dysfunctions, amyloid-ß. However, recent evidence suggests that the accepted view of APP proteolysis by the canonical α-, ß-, and γ-secretases is simplistic, with the discovery of a number of novel APP secretases (including δ- and η-secretases, alternative ß-secretases) and additional metabolites, some of which may also cause synaptic dysfunction. Furthermore, various proteins have been identified that interact with APP and modulate its cleavage by the secretases. Here, we give an overview of the increasingly complex picture of APP proteolysis.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Proteolysis , Animals , Humans
7.
Subcell Biochem ; 76: 363-74, 2015.
Article in English | MEDLINE | ID: mdl-26219720

ABSTRACT

Tissue non-specific alkaline phosphatase (TNAP) is present on neuronal membranes and induces neuronal toxicity via tau dephosphorylation; a mechanism which could play a role in the neuronal loss seen in Alzheimer's disease (AD). TNAP increases in the plasma following brain injury and cerebrovascular disease. In this chapter we summarise our previous work which looked at changes in TNAP activity in the brain and plasma of AD individuals and discuss whether these changes may be reflective of neuronal loss. Our data demonstrate that TNAP activity is significantly increased in the brain in both the sporadic and familial forms of AD and that TNAP activity is significantly increased in the plasma in AD patients. In addition, we describe a significant inverse correlation between plasma TNAP activity and cognitive function in AD. Using these data we propose a model for TNAP-induced neurodegeneration in AD resulting from tau dephosphorylation following secretion of tau from neuronal cells.


Subject(s)
Alkaline Phosphatase/physiology , Alzheimer Disease/enzymology , Neurodegenerative Diseases/enzymology , Alkaline Phosphatase/blood , Alkaline Phosphatase/cerebrospinal fluid , Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Apoptosis , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Brain Injuries/blood , Brain Injuries/cerebrospinal fluid , Humans , Nerve Degeneration/blood , Nerve Degeneration/cerebrospinal fluid , Nerve Degeneration/etiology , tau Proteins/metabolism
8.
FEBS Lett ; 597(11): 1489-1502, 2023 06.
Article in English | MEDLINE | ID: mdl-37235726

ABSTRACT

Bone morphogenetic protein 1 (BMP1), a member of the astacin family of zinc-metalloproteases, proteolytically cleaves the low-density lipoprotein receptor (LDLR) within its ligand-binding domain, reducing the binding and cellular uptake of LDL-cholesterol. Here, we aimed to determine whether astacin proteases other than BMP1 may also cleave LDLR. Although human hepatocytes express all six astacin proteases, including the meprins and mammalian tolloid, we found through pharmacological inhibition and genetic knockdown that only BMP1 contributed to the cleavage of LDLR in its ligand-binding domain. We also found that the minimum amino acid change required to render mouse LDLR susceptible to cleavage by BMP1 is mutation at the P1' and P2 positions of the cleavage site. When expressed in cells, the resulting humanised-mouse LDLR internalised LDL-cholesterol. This work provides insight into the biological mechanisms regulating LDLR function.


Subject(s)
Bone Morphogenetic Protein 1 , Peptide Hydrolases , Receptors, LDL , Animals , Humans , Mice , Bone Morphogenetic Protein 1/metabolism , Cholesterol , Hepatocytes/metabolism , Ligands , Lipoproteins, LDL/metabolism , Mammals/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Receptors, LDL/genetics , Receptors, LDL/metabolism
9.
Neuronal Signal ; 7(4): NS20230016, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37808160

ABSTRACT

Alzheimer's disease (AD) is characterised by the aggregation and deposition of amyloid-ß (Aß) peptides in the human brain. In age-related late-onset AD, deficient degradation and clearance, rather than enhanced production, of Aß contributes to disease pathology. In the present study, we assessed the contribution of the two key Aß-degrading zinc metalloproteases, insulin-degrading enzyme (IDE) and neprilysin (NEP), to Aß degradation in human induced pluripotent stem cell (iPSC)-derived cortical neurons. Using an Aß fluorescence polarisation assay, inhibition of IDE but not of NEP, blocked the degradation of Aß by human neurons. When the neurons were grown in a 3D extracellular matrix to visualise Aß deposition, inhibition of IDE but not NEP, increased the number of Aß deposits. The resulting Aß deposits were stained with the conformation-dependent, anti-amyloid antibodies A11 and OC that recognise Aß aggregates in the human AD brain. Inhibition of the Aß-forming ß-secretase prevented the formation of the IDE-inhibited Aß deposits. These data indicate that inhibition of IDE in live human neurons grown in a 3D matrix increased the deposition of Aß derived from the proteolytic cleavage of the amyloid precursor protein. This work has implications for strategies aimed at enhancing IDE activity to promote Aß degradation in AD.

10.
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
11.
Neurodegener Dis ; 9(1): 31-7, 2012.
Article in English | MEDLINE | ID: mdl-22024719

ABSTRACT

BACKGROUND: Tissue non-specific alkaline phosphatase (TNAP) has been shown to promote the neurotoxicity of extracellular tau which contributes to the spread of pathology in Alzheimer's disease (AD). OBJECTIVE: To investigate changes in TNAP activity in the hippocampus in both sporadic and familial AD, and to examine whether changes in neuronal TNAP are reflected systemically by looking at changes in plasma TNAP activity in AD. METHODS: We measured the activity of TNAP in the hippocampus in sporadic AD, familial AD and appropriate age-matched controls, and in an ageing series (age: 25-88 years) of brains. In addition, we measured TNAP activity in plasma from 110 AD and 110 non-demented control participants. RESULTS: TNAP activity was significantly increased in the hippocampus in sporadic (by 56%; p = 0.038) and familial AD (by 121%; p = 0.042) compared with the age-matched controls. However, there was no correlation of TNAP activity with age. Furthermore, plasma TNAP activity was increased in AD (by 13%; p = 0.018) and inversely correlated with cognitive function (r(s) = -0.211; p = 0.027). CONCLUSION: Together, these data indicate that TNAP is increased in both sporadic and familial AD but not in the aged brain, indicating that the increase is likely a consequence of AD-associated changes in the brain. The neuronal change in TNAP is reflected in an increase in plasma TNAP in AD and is inversely correlated with cognitive function.


Subject(s)
Alkaline Phosphatase/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Hippocampus/metabolism , Adult , Aged , Aged, 80 and over , Aging/metabolism , Alkaline Phosphatase/blood , Alzheimer Disease/blood , Case-Control Studies , Cognition/physiology , Female , Humans , Male , Middle Aged
12.
J Biol Chem ; 285(53): 41443-54, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-20961856

ABSTRACT

Amyloidogenic processing of the amyloid precursor protein (APP) by ß- and γ-secretases generates several biologically active products, including amyloid-ß (Aß) and the APP intracellular domain (AICD). AICD regulates transcription of several neuronal genes, especially the Aß-degrading enzyme, neprilysin (NEP). APP exists in several alternatively spliced isoforms, APP(695), APP(751), and APP(770). We have examined whether each isoform can contribute to AICD generation and hence up-regulation of NEP expression. Using SH-SY5Y neuronal cells stably expressing each of the APP isoforms, we observed that only APP(695) up-regulated nuclear AICD levels (9-fold) and NEP expression (6-fold). Increased NEP expression was abolished by a ß- or γ-secretase inhibitor but not an α-secretase inhibitor. This correlated with a marked increase in both Aß(1-40) and Aß(1-42) in APP(695) cells as compared with APP(751) or APP(770) cells. Similar phenomena were observed in Neuro2a but not HEK293 cells. SH-SY5Y cells expressing the Swedish mutant of APP(695) also showed an increase in Aß levels and NEP expression as compared with wild-type APP(695) cells. Chromatin immunoprecipitation revealed that AICD was associated with the NEP promoter in APP(695), Neuro2a, and APP(Swe) cells but not APP(751) nor APP(770) cells where AICD was replaced by histone deacetylase 1 (HDAC1). AICD occupancy of the NEP promoter was replaced by HDAC1 after treatment of the APP(695) cells with a ß- but not an α-secretase inhibitor. The increased AICD and NEP levels were significantly reduced in cholesterol-depleted APP(695) cells. In conclusion, Aß and functional AICD appear to be preferentially synthesized through ß-secretase action on APP(695).


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Gene Expression Regulation , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Cholesterol/chemistry , Chromatin Immunoprecipitation , Histone Deacetylases/metabolism , Humans , Ligands , Mice , Neprilysin/biosynthesis , Neurodegenerative Diseases/metabolism , Protein Isoforms , Protein Structure, Tertiary
13.
Neuronal Signal ; 5(4): NS20210027, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34804595

ABSTRACT

The neurovascular unit (NVU), consisting of neurons, glial cells, vascular cells (endothelial cells, pericytes and vascular smooth muscle cells (VSMCs)) together with the surrounding extracellular matrix (ECM), is an important interface between the peripheral blood and the brain parenchyma. Disruption of the NVU impacts on blood-brain barrier (BBB) regulation and underlies the development and pathology of multiple neurological disorders, including stroke and Alzheimer's disease (AD). The ability to differentiate induced pluripotent stem cells (iPSCs) into the different cell types of the NVU and incorporate them into physical models provides a reverse engineering approach to generate human NVU models to study BBB function. To recapitulate the in vivo situation such NVU models must also incorporate the ECM to provide a 3D environment with appropriate mechanical and biochemical cues for the cells of the NVU. In this review, we provide an overview of the cells of the NVU and the surrounding ECM, before discussing the characteristics (stiffness, functionality and porosity) required of hydrogels to mimic the ECM when incorporated into in vitro NVU models. We summarise the approaches available to measure BBB functionality and present the techniques in use to develop robust and translatable models of the NVU, including transwell models, hydrogel models, 3D-bioprinting, microfluidic models and organoids. The incorporation of iPSCs either without or with disease-specific genetic mutations into these NVU models provides a platform in which to study normal and disease mechanisms, test BBB permeability to drugs, screen for new therapeutic targets and drugs or to design cell-based therapies.

14.
ACS Nano ; 15(4): 7357-7369, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33730479

ABSTRACT

Blood-circulating biomarkers have the potential to detect Alzheimer's disease (AD) pathology before clinical symptoms emerge and to improve the outcomes of clinical trials for disease-modifying therapies. Despite recent advances in understanding concomitant systemic abnormalities, there are currently no validated or clinically used blood-based biomarkers for AD. The extremely low concentration of neurodegeneration-associated proteins in blood necessitates the development of analytical platforms to address the "signal-to-noise" issue and to allow an in-depth analysis of the plasma proteome. Here, we aimed to discover and longitudinally track alterations of the blood proteome in a transgenic mouse model of AD, using a nanoparticle-based proteomics enrichment approach. We employed blood-circulating, lipid-based nanoparticles to extract, analyze and monitor AD-specific protein signatures and to systemically uncover molecular pathways associated with AD progression. Our data revealed the existence of multiple proteomic signals in blood, indicative of the asymptomatic stages of AD. Comprehensive analysis of the nanoparticle-recovered blood proteome by label-free liquid chromatography-tandem mass spectrometry resulted in the discovery of AD-monitoring signatures that could discriminate the asymptomatic phase from amyloidopathy and cognitive deterioration. While the majority of differentially abundant plasma proteins were found to be upregulated at the initial asymptomatic stages, the abundance of these molecules was significantly reduced as a result of amyloidosis, suggesting a disease-stage-dependent fluctuation of the AD-specific blood proteome. The potential use of the proposed nano-omics approach to uncover information in the blood that is directly associated with brain neurodegeneration was further exemplified by the recovery of focal adhesion cascade proteins. We herein propose the integration of nanotechnology with already existing proteomic analytical tools in order to enrich the identification of blood-circulating signals of neurodegeneration, reinvigorating the potential clinical utility of the blood proteome at predicting the onset and kinetics of the AD progression trajectory.


Subject(s)
Alzheimer Disease , Nanoparticles , Alzheimer Disease/diagnosis , Animals , Biomarkers , Blood Proteins , Mice , Proteome , Proteomics
16.
Bioorg Med Chem Lett ; 19(23): 6770-4, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19854048

ABSTRACT

A novel series of isatin-based inhibitors of beta-secretase (BACE-1) have been identified using a virtual high-throughput screening approach. Structure-activity relationship studies revealed structural features important for inhibition. Docking studies suggest these inhibitors may bind within the BACE-1 active site through H-bonding interactions involving the catalytic aspartate residues.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Combinatorial Chemistry Techniques/methods , Drug Discovery , Enzyme Inhibitors/pharmacology , Isatin/pharmacology , Catalytic Domain , Computational Biology , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Isatin/chemical synthesis , Isatin/chemistry , Models, Molecular , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
17.
Sci Rep ; 9(1): 11416, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388055

ABSTRACT

The development of cardiovascular disease is intimately linked to elevated levels of low-density lipoprotein (LDL) cholesterol in the blood. Hepatic LDL receptor (LDLR) levels regulate the amount of plasma LDL. We identified the secreted zinc metalloproteinase, bone morphogenetic protein 1 (BMP1), as responsible for the cleavage of human LDLR within its extracellular ligand-binding repeats at Gly171↓Asp172. The resulting 120 kDa membrane-bound C-terminal fragment (CTF) of LDLR had reduced capacity to bind LDL and when expressed in LDLR null cells had compromised LDL uptake as compared to the full length receptor. Pharmacological inhibition of BMP1 or siRNA-mediated knockdown prevented the generation of the 120 kDa CTF and resulted in an increase in LDL uptake into cells. The 120 kDa CTF was detected in the livers from humans and mice expressing human LDLR. Collectively, these results identify that BMP1 regulates cellular LDL uptake and may provide a target to modulate plasma LDL cholesterol.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Lipoproteins, LDL/metabolism , Receptors, LDL/metabolism , Animals , Atherosclerosis/blood , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biopsy , Bone Morphogenetic Protein 1/antagonists & inhibitors , Bone Morphogenetic Protein 1/genetics , CHO Cells , Cricetulus , Gene Knockdown Techniques , Hep G2 Cells , Humans , Lipoproteins, LDL/blood , Liver/chemistry , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Oxadiazoles/pharmacology , Proteolysis/drug effects , RNA, Small Interfering/metabolism , Receptors, LDL/analysis , Receptors, LDL/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
J Alzheimers Dis ; 63(1): 13-33, 2018.
Article in English | MEDLINE | ID: mdl-29630551

ABSTRACT

With predictions showing that 131.5 million people worldwide will be living with dementia by 2050, an understanding of the molecular mechanisms underpinning disease is crucial in the hunt for novel therapeutics and for biomarkers to detect disease early and/or monitor disease progression. The metabolism of the microtubule-associated protein tau is altered in different dementias, the so-called tauopathies. Tau detaches from microtubules, aggregates into oligomers and neurofibrillary tangles, which can be secreted from neurons, and spreads through the brain during disease progression. Post-translational modifications exacerbate the production of both oligomeric and soluble forms of tau, with proteolysis by a range of different proteases being a crucial driver. However, the impact of tau proteolysis on disease progression has been overlooked until recently. Studies have highlighted that proteolytic fragments of tau can drive neurodegeneration in a fragment-dependent manner as a result of aggregation and/or transcellular propagation. Proteolytic fragments of tau have been found in the cerebrospinal fluid and plasma of patients with different tauopathies, providing an opportunity to develop these fragments as novel disease progression biomarkers. A range of therapeutic strategies have been proposed to halt the toxicity associated with proteolysis, including reducing protease expression and/or activity, selectively inhibiting protease-substrate interactions, and blocking the action of the resulting fragments. This review highlights the importance of tau proteolysis in the pathogenesis of tauopathies, identifies putative sites during tau fragment-mediated neurodegeneration that could be targeted therapeutically, and discusses the potential use of proteolytic fragments of tau as biomarkers for different tauopathies.


Subject(s)
Biomarkers/metabolism , Peptide Fragments/toxicity , Proteolysis , Tauopathies , tau Proteins/metabolism , Animals , Disease Progression , Humans , Tauopathies/chemically induced , Tauopathies/metabolism , Tauopathies/pathology
19.
Neurochem Int ; 118: 134-144, 2018 09.
Article in English | MEDLINE | ID: mdl-29649504

ABSTRACT

Cerebral ischemia is known to be a major cause of death and the later development of Alzheimer's disease and vascular dementia. However, ischemia induced cellular damage that initiates these diseases remain poorly understood. This is primarily due to lack of clinically relevant models that are highly reproducible. Here, we have optimised a murine model of global cerebral ischaemia with multiple markers to determine brain pathology, neurochemistry and correlated memory deficits in these animals. Cerebral ischaemia in mice was induced by bilateral common carotid artery occlusion. Following reperfusion, the mice were either fixed with 4% paraformaldehyde or decapitated under anaesthesia. Brains were processed for Western blotting or immunohistochemistry for glial (GLT1) and vesicular (VGLUT1, VGLUT2) glutamate transporters and paired helical filament (PHF1) tau. The PHF1 tau is the main component of neurofibrillary tangle, which is the pathological hallmark of Alzheimer's disease and vascular dementia. The novel object recognition behavioural assay was used to investigate the functional cognitive consequences in these mice. The results show consistent and selective neuronal and glial cell changes in the hippocampus and the cortex together with significant reductions in GLT1 (***P < 0.001), VGLUT1 (**P < 0.01) and VGLUT2 (***P < 0.001) expressions in the hippocampus in occluded mice as compared to sham-operated animals. These changes are associated with increased PHF1 (***P < 0.0001) protein and a significant impairment of performance (*p < 0.0006, N = 6/group) in the novel object recognition test. This model represents a useful tool for investigating cellular, biochemical and molecular mechanisms of global cerebral ischaemia and may be an ideal preclinical model for vascular dementia.


Subject(s)
Brain Chemistry/physiology , Brain Ischemia/metabolism , Dementia, Vascular/metabolism , Disease Models, Animal , Memory Disorders/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dementia, Vascular/genetics , Dementia, Vascular/pathology , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , tau Proteins/genetics
20.
PLoS One ; 11(7): e0159119, 2016.
Article in English | MEDLINE | ID: mdl-27447728

ABSTRACT

The cellular prion protein (PrPC) has been proposed to play an important role in the pathogenesis of Alzheimer's disease. In cellular models PrPC inhibited the action of the ß-secretase BACE1 on wild type amyloid precursor protein resulting in a reduction in amyloid-ß (Aß) peptides. Here we have assessed the effect of genetic ablation of PrPC in transgenic mice expressing human wild type amyloid precursor protein (line I5). Deletion of PrPC had no effect on the α- and ß-secretase proteolysis of the amyloid precursor protein (APP) nor on the amount of Aß38, Aß40 or Aß42 in the brains of the mice. In addition, ablation of PrPC did not alter Aß deposition or histopathology phenotype in this transgenic model. Thus using this transgenic model we could not provide evidence to support the hypothesis that PrPC regulates Aß production.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Prion Proteins/physiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/pathology , Female , Gene Deletion , Humans , Mice , Mice, Transgenic , Phenotype , Plaque, Amyloid/pathology , Prion Proteins/genetics , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL