51
|
Miners JS, Clarke P, Love S. Clusterin levels are increased in Alzheimer's disease and influence the regional distribution of Aβ. Brain Pathol 2016; 27:305-313. [PMID: 27248362 DOI: 10.1111/bpa.12392] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 12/22/2022] Open
Abstract
Clusterin, also known as apoJ, is a lipoprotein abundantly expressed within the CNS. It regulates Aβ fibril formation and toxicity and facilitates amyloid-β (Aβ) transport across the blood-brain barrier. Genome-wide association studies have shown variations in the clusterin gene (CLU) to influence the risk of developing sporadic Alzheimer's disease (AD). To explore whether clusterin modulates the regional deposition of Aβ, we measured levels of soluble (NP40-extracted) and insoluble (guanidine-HCl-extracted) clusterin, Aβ40 and Aβ42 by sandwich ELISA in brain regions with a predilection for amyloid pathology-mid-frontal cortex (MF), cingulate cortex (CC), parahippocampal cortex (PH), and regions with little or no pathology-thalamus (TH) and white matter (WM). Clusterin level was highest in regions with plaque pathology (MF, CC, PH and PC), approximately mirroring the regional distribution of Aβ. It was significantly higher in AD than controls, and correlated positively with Aβ42 and insoluble Aβ40. Soluble clusterin level rose significantly with severity of cerebral amyloid angiopathy, and in MF and PC regions was highest in APOE ɛ4 homozygotes. In the TH and WM (areas with little amyloid pathology) clusterin was unaltered in AD and did not correlate with Aβ level. There was a significant positive correlation between the concentration of clusterin and the regional levels of insoluble Aβ42; however, the molar ratio of clusterin : Aβ42 declined with insoluble Aβ42 level in a region-dependent manner, being lowest in regions with predilection for Aβ plaque pathology. Under physiological conditions, clusterin reduces aggregation and promotes clearance of Aβ. Our findings indicate that in AD, clusterin increases, particularly in regions with most abundant Aβ, but because the increase does not match the rising level of Aβ42, the molar ratio of clusterin : Aβ42 in those regions falls, probably contributing to Aβ deposition within the tissue.
Collapse
Affiliation(s)
- J Scott Miners
- Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, United Kingdom
| | - Polly Clarke
- Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, United Kingdom
| | - Seth Love
- Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, United Kingdom
| |
Collapse
|
52
|
Troakes C, Smyth R, Noor F, Maekawa S, Killick R, King A, Al-Sarraj S. Clusterin expression is upregulated following acute head injury and localizes to astrocytes in old head injury. Neuropathology 2016; 37:12-24. [PMID: 27365216 DOI: 10.1111/neup.12320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 01/20/2023]
Abstract
There is mounting evidence linking traumatic brain injury (TBI) to neurodegeneration. Clusterin (apolipoprotein J or ApoJ) is a complement inhibitor that appears to have a neuroprotective effect in response to tissue damage and has been reported to be upregulated in Alzheimer's disease. Here we investigated the time course and cellular expression pattern of clusterin in human TBI. Tissue from 32 patients with TBI of varying survival times (from under 30 min to 10 months) was examined using immunohistochemistry for clusterin alongside other markers of neurodegeneration and neuroinflammation. TBI cases were compared to ischemic brain damage, Alzheimer's disease and controls. Double immunofluorescence was carried out in order to examine cellular expression. Clusterin was initially expressed in an axonal location less than 30 min following TBI and increased in intensity and the frequency of deposits with increasing survival time up to 24 h, after which it appeared to reduce in intensity but was still evident several weeks after injury. Clusterin was first evident in astrocytes after 45 min, being increasingly seen up to 48 h but remaining intense in TBI cases with long survival times. Our results suggest clusterin plays a role in modulating the inflammatory response of acute and chronic TBI and that it is a useful marker for TBI, particularly in cases with short survival times. Its prominent accumulation in astrocytes, alongside a mounting inflammatory response and activation of microglial cells supports a potential role in the neurodegenerative changes that occur as a result of TBI.
Collapse
Affiliation(s)
- Claire Troakes
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Rachel Smyth
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Farzana Noor
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Satomi Maekawa
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Richard Killick
- Old Age Psychiatry Department, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Andrew King
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Clinical Neuropathology Department, King's College Hospital NHS Foundation Trust, London, UK
| | - Safa Al-Sarraj
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Clinical Neuropathology Department, King's College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
53
|
Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M. TREM2 Binds to Apolipoproteins, Including APOE and CLU/APOJ, and Thereby Facilitates Uptake of Amyloid-Beta by Microglia. Neuron 2016; 91:328-40. [DOI: 10.1016/j.neuron.2016.06.015] [Citation(s) in RCA: 466] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/14/2016] [Accepted: 06/07/2016] [Indexed: 11/30/2022]
|
54
|
Mungenast AE, Siegert S, Tsai LH. Modeling Alzheimer's disease with human induced pluripotent stem (iPS) cells. Mol Cell Neurosci 2016; 73:13-31. [PMID: 26657644 PMCID: PMC5930170 DOI: 10.1016/j.mcn.2015.11.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 11/05/2015] [Accepted: 11/25/2015] [Indexed: 02/08/2023] Open
Abstract
In the last decade, induced pluripotent stem (iPS) cells have revolutionized the utility of human in vitro models of neurological disease. The iPS-derived and differentiated cells allow researchers to study the impact of a distinct cell type in health and disease as well as performing therapeutic drug screens on a human genetic background. In particular, clinical trials for Alzheimer's disease (AD) have been failing. Two of the potential reasons are first, the species gap involved in proceeding from initial discoveries in rodent models to human studies, and second, an unsatisfying patient stratification, meaning subgrouping patients based on the disease severity due to the lack of phenotypic and genetic markers. iPS cells overcome this obstacles and will improve our understanding of disease subtypes in AD. They allow researchers conducting in depth characterization of neural cells from both familial and sporadic AD patients as well as preclinical screens on human cells. In this review, we briefly outline the status quo of iPS cell research in neurological diseases along with the general advantages and pitfalls of these models. We summarize how genome-editing techniques such as CRISPR/Cas9 will allow researchers to reduce the problem of genomic variability inherent to human studies, followed by recent iPS cell studies relevant to AD. We then focus on current techniques for the differentiation of iPS cells into neural cell types that are relevant to AD research. Finally, we discuss how the generation of three-dimensional cell culture systems will be important for understanding AD phenotypes in a complex cellular milieu, and how both two- and three-dimensional iPS cell models can provide platforms for drug discovery and translational studies into the treatment of AD.
Collapse
Affiliation(s)
- Alison E Mungenast
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Sandra Siegert
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| |
Collapse
|
55
|
Del Valle E, Navarro A, Martínez-Pinilla E, Torices S, Tolivia J. Apo J and Apo D: Complementary or Antagonistic Roles in Alzheimer's Disease? J Alzheimers Dis 2016; 53:639-50. [PMID: 27197790 DOI: 10.3233/jad-160032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Apolipoprotein D (Apo D) and Apolipoprotein J (Apo J) are among the only nine apolipoproteins synthesized in the nervous system. Apart from development, these apolipoproteins are implicated in the normal aging process as well as in different neuropathologies as Alzheimer's disease (AD), where a neuroprotective role has been postulated. Different authors have proposed that Apo D and Apo J could be biomarkers for AD but as far as we know, there are no studies about the relationship between them as well as their expression pattern along the progression of the disease. In this paper, using double immunohistochemistry techniques, we have demonstrated that Apo D is mainly located in glial cells while Apo J expression preferentially occurs in neurons; both proteins are also present in AD diffuse and mature senile plaques but without signal overlap. In addition, we have observed that Apo J and Apo D immunostaining shows a positive correlation with the progression of the disease and the Braak's stages. These results suggest complementary and cell-dependent neuroprotective roles for each apolipoprotein during AD progress.
Collapse
|
56
|
Fini ME, Bauskar A, Jeong S, Wilson MR. Clusterin in the eye: An old dog with new tricks at the ocular surface. Exp Eye Res 2016; 147:57-71. [PMID: 27131907 DOI: 10.1016/j.exer.2016.04.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/22/2016] [Accepted: 04/23/2016] [Indexed: 12/30/2022]
Abstract
The multifunctional protein clusterin (CLU) was first described in 1983 as a secreted glycoprotein present in ram rete testis fluid that enhanced aggregation ('clustering') of a variety of cells in vitro. It was also independently discovered in a number of other systems. By the early 1990s, CLU was known under many names and its expression had been demonstrated throughout the body, including in the eye. Its homeostatic activities in proteostasis, cytoprotection, and anti-inflammation have been well documented, however its roles in health and disease are still not well understood. CLU is prominent at fluid-tissue interfaces, and in 1996 it was demonstrated to be the most highly expressed transcript in the human cornea, the protein product being localized to the apical layers of the mucosal epithelia of the cornea and conjunctiva. CLU protein is also present in human tears. Using a preclinical mouse model for desiccating stress that mimics human dry eye disease, the authors recently demonstrated that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration in the tears. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to LGALS3 (galectin-3), a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. CLU depletion from the ocular surface epithelia is seen in a variety of inflammatory conditions in humans and mice that lead to squamous metaplasia and a keratinized epithelium. This suggests that CLU might have a specific role in maintaining mucosal epithelial differentiation, an idea that can now be tested using the mouse model for desiccating stress. Most excitingly, the new findings suggest that CLU could serve as a novel biotherapeutic for dry eye disease.
Collapse
Affiliation(s)
- M Elizabeth Fini
- USC Institute for Genetic Medicine and Departments of Cell & Neurobiology and Ophthalmology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Aditi Bauskar
- USC Institute for Genetic Medicine and Graduate Program in Medical Biology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Shinwu Jeong
- USC Institute for Genetic Medicine and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 2250 Alcatraz St., Suite 240, Los Angeles, CA 90089-9037, USA.
| | - Mark R Wilson
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Northfields Avenue, Wollongong, New South Wales, 2522 Australia.
| |
Collapse
|
57
|
Algotar AM, Behnejad R, Singh P, Thompson PA, Hsu CH, Stratton SP. EFFECT OF SELENIUM SUPPLEMENTATION ON PROTEOMIC SERUM BIOMARKERS IN ELDERLY MEN. J Frailty Aging 2016; 4:107-10. [PMID: 26366377 DOI: 10.14283/jfa.2015.48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVES To determine the effect of selenium supplementation on the human proteomic profile. DESIGN Serum samples were collected in this pilot study from a randomized placebo controlled Phase 2 clinical trial (Watchful Waiting (WW)). SETTING Subjects were followed every three months for up to five years at the University of Arizona Prostate Cancer Prevention Program. PARTICIPANTS One hundred and forty men (age < 85 years) had biopsy-proven prostate cancer, a Gleason sum score less than eight, no metastatic cancer, and no prior treatment for prostate cancer. INTERVENTION As part of the WW trial, men were randomized to placebo, selenium 200 μg/day or selenium 800 μg/day. For the purpose of the current study, 40 subjects enrolled in the WW study (20 from the placebo group and 20 from Se 800 μg/day group) were selected. MEASUREMENTS Baseline serum samples were collected at each follow-up visit and stored at -80 degrees Celsius. A multiplexed proteomic panel investigated changes in 120 proteins markers simultaneously. RESULTS Thirteen proteins (Apolipoprotein J, IL-10, IL-1 alpha, MMP-3, IL-12p70, IL-2 receptor alpha, cathepsin B, eotaxin, EGFR, FGF-basic, myeloperoxidase, RANTES, TGF-beta) were determined to be either statistically (p-value < 0.05) or marginally significantly (0.05 < p-value <0.1) changed in the selenium supplemented group as compared to placebo. CONCLUSION Although independent validation of these results is needed, this study is the first of its kind to utilize high throughput fluorescence based protein multiplex panel in analyzing changes in the proteomic profile due to selenium supplementation. Results from this study provide insight into the ability of selenium to modulate numerous protein markers and thus impact various biological processes in humans.
Collapse
Affiliation(s)
- A M Algotar
- Department of Preventive Medicine, Loma Linda University Medical Center, Loma Linda, CA ; University of Arizona Cancer Center, Tucson, AZ
| | - R Behnejad
- University of Arizona Cancer Center, Tucson, AZ
| | - P Singh
- Department of Hematology-Oncology, University of Arizona, Tucson, AZ
| | - P A Thompson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - C H Hsu
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ
| | | |
Collapse
|
58
|
Yoon SY, Kim DH. Alzheimer's disease genes and autophagy. Brain Res 2016; 1649:201-209. [PMID: 27016058 DOI: 10.1016/j.brainres.2016.03.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 03/09/2016] [Accepted: 03/13/2016] [Indexed: 11/15/2022]
Abstract
Autophagy is a process to degrade and recycle cellular constituents via the lysosome for regulating cellular homeostasis. Its dysfunction is now considered to be involved in many diseases, including neurodegenerative diseases. Many features reflecting autophagy impairment, such as autophagosome accumulation and lysosomal dysfunction, have been also revealed to be involved in Alzheimer's disease (AD). Recent genetic studies such as genome-wide association studies in AD have identified a number of novel genes associated with AD. Some of the identified genes have demonstrated dysfunction in autophagic processes in AD, while others remain under investigation. Since autophagy is strongly regarded to be one of the major pathogenic mechanisms of AD, it is necessary to review how the AD-associated genes are related to autophagy. We anticipate our current review to be a starting point for future studies regarding AD-associated genes and autophagy. This article is part of a Special Issue entitled SI:Autophagy.
Collapse
Affiliation(s)
- Seung-Yong Yoon
- Alzheimer's Disease Experts Lab (ADEL), Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Brain Science, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Dong-Hou Kim
- Alzheimer's Disease Experts Lab (ADEL), Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Brain Science, University of Ulsan College of Medicine, Seoul, Republic of Korea; Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Republic of Korea; Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
59
|
Cervera-Carles L, Clarimón J. Genetic and Epigenetic Architecture of Alzheimer’s Dementia. CURRENT GENETIC MEDICINE REPORTS 2016. [DOI: 10.1007/s40142-016-0086-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
60
|
Han SH, Park JC, Mook-Jung I. Amyloid β-interacting partners in Alzheimer's disease: From accomplices to possible therapeutic targets. Prog Neurobiol 2016; 137:17-38. [DOI: 10.1016/j.pneurobio.2015.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022]
|
61
|
Lehallier B, Essioux L, Gayan J, Alexandridis R, Nikolcheva T, Wyss-Coray T, Britschgi M. Combined Plasma and Cerebrospinal Fluid Signature for the Prediction of Midterm Progression From Mild Cognitive Impairment to Alzheimer Disease. JAMA Neurol 2015; 73:203-212. [PMID: 26659895 DOI: 10.1001/jamaneurol.2015.3135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Importance A reliable method of detecting Alzheimer disease (AD) in its prodromal state is needed for patient stratification in clinical trials or for personalizing existing or potential upcoming therapies. Current cerebrospinal fluid (CSF)- or imaging-based single biomarkers for AD offer reliable identification of patients with underlying AD but insufficient prediction of the rate of AD progression. Objective To optimize prediction of progression from mild cognitive impairment (MCI) to AD dementia by combining information from diverse patient variables. Design, Setting, and Participants This cohort study from the Alzheimer Disease Neuroimaging Initiative (ADNI) enrolled 928 patients with MCI at baseline and 249 selected variables available in the ADNI data set. Variables included clinical and demographic data, cognitive scores, magnetic resonance imaging-based brain volumetric data, the apolipoprotein E (APOE) and translocase of outer mitochondrial membrane 40 homolog (TOMM40) genotypes, and analyte levels measured in the CSF and plasma. Data were collected in July 2012 and analyzed from July 1, 2012, to June 1, 2015. Main Outcomes and Measures Progression from MCI to AD within 1 to 6 years. To determine whether combinations of markers could predict progression from MCI to AD within 1 to 6 years, the elastic net algorithm was used in an iterative resampling of a training- and test-based variable selection and modeling approach. Results Among the 928 patients with MCI in the ADNI database, 94 had 224 of the required variables available for the modeling. The results showed the contributions of age, Clinical Dementia Rating Sum of Boxes composite test score, hippocampal volume, and multiple plasma and CSF factors in modeling progression to AD. A combination of apolipoprotein A-II and cortisol levels in plasma and fibroblast growth factor 4, heart-type fatty acid binding protein, calcitonin, and tumor necrosis factor-related apoptosis-inducing ligand receptor 3 (TRAIL-R3) in CSF allowed for reliable prediction of disease status 3 years from the time of sample collection (80% classification accuracy, 88% sensitivity, and 70% specificity). Conclusions and Relevance These study findings suggest that a combination of markers measured in plasma and CSF, distinct from β-amyloid and tau, could prove useful in predicting midterm progression from MCI to AD dementia. Such a large-scale, multivariable-based analytical approach could be applied to other similar large data sets involving AD and beyond.
Collapse
Affiliation(s)
- Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Laurent Essioux
- Translational Technologies and Bioinformatics, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd, Basel, Switzerland
| | - Javier Gayan
- Translational Technologies and Bioinformatics, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd, Basel, Switzerland
| | - Roxana Alexandridis
- Translational Technologies and Bioinformatics, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd, Basel, Switzerland
| | - Tania Nikolcheva
- Roche Pharma Development, F. Hoffmann-La Roche, Ltd, Basel, Switzerland
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California4Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Markus Britschgi
- Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Areas, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd, Basel, Switzerland
| | | |
Collapse
|
62
|
Montañola A, de Retana SF, López-Rueda A, Merino-Zamorano C, Penalba A, Fernández-Álvarez P, Rodríguez-Luna D, Malagelada A, Pujadas F, Montaner J, Hernández-Guillamon M. ApoA1, ApoJ and ApoE Plasma Levels and Genotype Frequencies in Cerebral Amyloid Angiopathy. Neuromolecular Med 2015; 18:99-108. [PMID: 26661731 DOI: 10.1007/s12017-015-8381-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/17/2015] [Indexed: 01/26/2023]
Abstract
The involvement of apolipoproteins, such as the ApoE4 isoform, in Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) highlights the fact that certain lipid carriers may participate in soluble β-amyloid (Aβ) transport. Our general aim was to characterize the soluble levels of the apolipoproteins apoE, apoA1 and apoJ/clusterin and their genotype status in patients with CAA. We analyzed the genotypes frequency of APOA1 (rs5069, rs670), CLU (rs11136000, rs1532278, rs7012010, rs9331888) and APOE (rs429358, rs7412) in a cohort of patients with CAA-associated intracerebral hemorrhage (ICH) (n = 59) and compared the results with those from hypertension-associated ICH (n = 42), AD patients (n = 73) and controls (n = 88). In a subgroup of patients, we also determined the plasma concentrations of apoE, apoA1 and apoJ/clusterin. We found increased plasma apoJ/clusterin levels in CAA patients compared to AD patients or controls after adjusting for sex and age (CAA vs. controls, p = 0.033; CAA vs. AD, p = 0.013). ApoA1 levels were not altered between groups, although a strong correlation was observed between plasma Aβ(1-40) and apoA1 among CAA patients (r = 0.583, p = 0.007). Regarding plasma apoE concentration, a robust association between circulating levels and genotype status was confirmed (p < 0.001). Whereas the APOE4 frequency was higher in AD (p < 0.001) and CAA (p = 0.013), the APOA1 and CLU genotypes were not different among groups. In the CAA cohort, the risk-linked CLU variant (C) rs11136000 was associated with white matter hyperintensities (p = 0.045) and the presence of lobar microbleeds (p = 0.023) on MRI. In summary, our findings suggest that apoA1 may act as a physiological transporter of Aβ(1-40) and that apoJ/clusterin appears to be a chaperone related to distinctive lesions in CAA brains.
Collapse
Affiliation(s)
- Alex Montañola
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Sofía Fernández de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Antonio López-Rueda
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Cristina Merino-Zamorano
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Penalba
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | | | - David Rodríguez-Luna
- Neurovascular Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Ana Malagelada
- Dementia Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Francesc Pujadas
- Dementia Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Neurovascular Unit, Neurology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
63
|
Vanhooren V, Navarrete Santos A, Voutetakis K, Petropoulos I, Libert C, Simm A, Gonos ES, Friguet B. Protein modification and maintenance systems as biomarkers of ageing. Mech Ageing Dev 2015; 151:71-84. [PMID: 25846863 DOI: 10.1016/j.mad.2015.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 12/22/2022]
|
64
|
A correlativity study of plasma APL1β28 and clusterin levels with MMSE/MoCA/CASI in aMCI patients. Sci Rep 2015; 5:15546. [PMID: 26503441 PMCID: PMC4621490 DOI: 10.1038/srep15546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/28/2015] [Indexed: 01/20/2023] Open
Abstract
Amnestic mild cognitive impairment (aMCI) is a sub-clinical condition characterized by memory deficits that are not severe enough to affect daily functioning. Here we investigated two potential biomarkers found in the cerebrospinal fluid of AD patients, APLP1-derived Aβ-like peptides 28 (APL1β28) and clusterin plasma levels, in terms of their relationship to cognitive function, as reflected in the Mini-Mental State Examination (MMSE), the Montreal Cognitive Assessment (MoCA) and the Cognitive Assessment Screening Instrument (CASI) in aMCI patients. Forty-seven aMCI patients and thirty-five age- and gender-matched healthy adult controls were recruited for this study. Using the ELISA method, we found that the mean concentrations of both APL1β28 and clusterin were not significantly different between the control and aMCI groups. The APL1β28 levels were positively correlated with clusterin and that both were negatively correlated with the MMSE scores of the aMCI patients. Clusterin levels were negatively correlated with the MoCA and CASI scores of the aMCI patients. Using multivariate analysis, the correlation between clusterin and MMSE/MoCA/CASI was independent of other AD risk factors including age, education, sex, body mass index and ApoE genotype. The data presented here demonstrate that plasma clusterin levels reflect cognitive function in aMCI patients.
Collapse
|
65
|
Bauskar A, Mack WJ, Mauris J, Argüeso P, Heur M, Nagel BA, Kolar GR, Gleave ME, Nakamura T, Kinoshita S, Moradian-Oldak J, Panjwani N, Pflugfelder SC, Wilson MR, Fini ME, Jeong S. Clusterin Seals the Ocular Surface Barrier in Mouse Dry Eye. PLoS One 2015; 10:e0138958. [PMID: 26402857 PMCID: PMC4581869 DOI: 10.1371/journal.pone.0138958] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/04/2015] [Indexed: 12/02/2022] Open
Abstract
Dry eye is a common disorder caused by inadequate hydration of the ocular surface that results in disruption of barrier function. The homeostatic protein clusterin (CLU) is prominent at fluid-tissue interfaces throughout the body. CLU levels are reduced at the ocular surface in human inflammatory disorders that manifest as severe dry eye, as well as in a preclinical mouse model for desiccating stress that mimics dry eye. Using this mouse model, we show here that CLU prevents and ameliorates ocular surface barrier disruption by a remarkable sealing mechanism dependent on attainment of a critical all-or-none concentration. When the CLU level drops below the critical all-or-none threshold, the barrier becomes vulnerable to desiccating stress. CLU binds selectively to the ocular surface subjected to desiccating stress in vivo, and in vitro to the galectin LGALS3, a key barrier component. Positioned in this way, CLU not only physically seals the ocular surface barrier, but it also protects the barrier cells and prevents further damage to barrier structure. These findings define a fundamentally new mechanism for ocular surface protection and suggest CLU as a biotherapeutic for dry eye.
Collapse
Affiliation(s)
- Aditi Bauskar
- USC Institute for Genetic Medicine and Graduate Program in Medical Biology, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Wendy J. Mack
- Southern California Clinical & Translational Science Institute and Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Jerome Mauris
- The Schepens Eye Research Institute, Massachusetts Eye & Ear and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Pablo Argüeso
- The Schepens Eye Research Institute, Massachusetts Eye & Ear and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Martin Heur
- USC Eye Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Barbara A. Nagel
- Research Microscopy and Histology Core, Department of Pathology, Saint Louis University School of Medicine, St Louis, Missouri, United States of America
| | - Grant R. Kolar
- Department of Pathology and Department of Ophthalmology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Martin E. Gleave
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Takahiro Nakamura
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Janet Moradian-Oldak
- Center for Craniofacial Molecular Biology, Division of Biomedical Sciences, University of Southern California, Herman Ostrow School of Dentistry of USC, Los Angeles, California, United States of America
| | - Noorjahan Panjwani
- New England Eye Center/Department of Ophthalmology and Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Stephen C. Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mark R. Wilson
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | - M. Elizabeth Fini
- USC Institute for Genetic Medicine and Departments of Cell & Neurobiology and Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| | - Shinwu Jeong
- USC Institute for Genetic Medicine and Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
66
|
Optimization of specific multiplex DNA primers to detect variable CLU genomic lesions in patients with Alzheimer’s disease. BIOCHIP JOURNAL 2015. [DOI: 10.1007/s13206-015-9306-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
67
|
Magnetic Bead-Based Serum Peptidome Profiling in Patients with Gestational Diabetes Mellitus. BIOMED RESEARCH INTERNATIONAL 2015; 2015:586309. [PMID: 26090425 PMCID: PMC4450277 DOI: 10.1155/2015/586309] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/06/2014] [Accepted: 08/28/2014] [Indexed: 12/18/2022]
Abstract
Gestational diabetes mellitus (GDM) is a frequent medical condition during pregnancy. Early diagnosis and treatment of GDM are crucial for both the mother and the baby. In the present study, we aimed to identify specific biomarkers to assist in the early detection of GDM and give some clues to the possible causes of GDM by comparing serum peptide profile differences between GDM patients and healthy controls. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was used in combination with weak cation exchange magnetic bead (WCX-MB). Levels of four peptides (4418.9, 2219.7, 2211.5, and 1533.4 Da) were significantly different. Interestingly, three of them (4418.9, 2211.5, and 1533.4 Da) were identified when GDM patients with two degrees of glucose intolerance were compared. Additionally, peptides 2211.5 and 1533.4 Da showed a decreasing trend as glucose intolerance increased, while peptide 4418.9 Da exhibited the reverse tendency. In conclusion, our study provides novel insights into the altered serum peptide profile of GDM patients. The specific candidate biomarkers may contribute to the development of GDM.
Collapse
|
68
|
Shuai P, Liu Y, Lu W, Liu Q, Li T, Gong B. Genetic associations of CLU rs9331888 polymorphism with Alzheimer's disease: A meta-analysis. Neurosci Lett 2015; 591:160-165. [PMID: 25703218 DOI: 10.1016/j.neulet.2015.02.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 02/06/2015] [Accepted: 02/14/2015] [Indexed: 11/29/2022]
Abstract
Recent studies showed the Clusterin gene (CLU) is associated with Alzheimer's disease (AD). However, studies investigating the association of CLU single-nucleotide polymorphism (SNP) rs9331888 with AD are controversial. We then performed a meta-analysis to assess the association between CLU SNP rs9331888 and AD. Computerized bibliographic searches of PUBMED and AlzGene database were conducted for the period up to July, 2014. The strength of the association between SNP rs9331888 and AD was estimated by odds ratios (ORs) and OR 95% confidence intervals (CIs). A total of 11 studies composed of 8766 AD cases and 11,366 controls were included in this study. Significant association of SNP rs9331888 with AD was found in Caucasian population among allelic model (C vs. G: OR=1.12, 95%CI=1.06-1.17, P<0.001), additive model (CC vs. GG: OR=1.25, 95%CI=1.12-1.40, P<0.001), recessive model (CC vs. CG+GG: OR=1.20, 95%CI=1.07-1.34, P=0.001), and dominant model (CC+CG vs. GG: OR=1.13, 95%CI=1.06-1.21, P<0.001). No significant association among these models was found in Asian and overall populations. Sensitivity analysis found that one study caused the distinct heterogeneity in Asian subgroup. Our analysis demonstrated that CLU SNP rs9331888 might be associated with an increased AD risk in Caucasian population, but not in Asian population.
Collapse
Affiliation(s)
- Ping Shuai
- Health Management Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; Sichuan Provincial Key Laboratory for Disease Gene Study, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuping Liu
- Health Management Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wenxue Lu
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Qiaolan Liu
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Tinxin Li
- Health Management Center, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Gong
- Sichuan Provincial Key Laboratory for Disease Gene Study, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
69
|
Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry 2015; 77:43-51. [PMID: 24951455 PMCID: PMC4234692 DOI: 10.1016/j.biopsych.2014.05.006] [Citation(s) in RCA: 905] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 01/18/2023]
Abstract
We review the genetic risk factors for late-onset Alzheimer's disease (AD) and their role in AD pathogenesis. More recent advances in understanding of the human genome-technologic advances in methods to analyze millions of polymorphisms in thousands of subjects-have revealed new genes associated with AD risk, including ABCA7, BIN1, CASS4, CD33, CD2AP, CELF1, CLU, CR1, DSG2, EPHA1, FERMT2, HLA-DRB5-DBR1, INPP5D, MS4A, MEF2C, NME8, PICALM, PTK2B, SLC24H4-RIN3, SORL1, and ZCWPW1. Emerging technologies to analyze the entire genome in large data sets have also revealed coding variants that increase AD risk: PLD3 and TREM2. We review the relationship between these AD risk genes and the cellular and neuropathologic features of AD. Understanding the mechanisms underlying the association of these genes with risk for disease will provide the most meaningful targets for therapeutic development to date.
Collapse
Affiliation(s)
| | - Alison M. Goate
- Corresponding author Contact information: Department of Psychiatry, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8134, St. Louis, MO 63110, phone: 314-362-8691, fax: 314-747-2983,
| |
Collapse
|
70
|
Abstract
Besides their well-documented function of reverse transport of cholesterol, high-density lipoproteins (HDLs) display pleiotropic effects due to their antioxidant, antithrombotic, anti-inflammatory and antiapoptotic properties that may play a major protective role in acute stroke, in particular by limiting the deleterious effects of ischaemia on the blood-brain barrier (BBB) and on the parenchymal cerebral compartment. HDLs may also modulate leukocyte and platelet activation, which may also represent an important target that would justify the use of HDL-based therapy in acute stroke. In this review, we will present an update of all the recent findings in HDL biology that could support a potential clinical use of HDL therapy in ischaemic stroke.
Collapse
|
71
|
Liu J, Huang P, He Y, Hong WX, Ren X, Yang X, He Y, Wang W, Zhang R, Yang H, Zhao Z, Huang H, Chen L, Zhao D, Xian H, Yang F, Ma D, Yang L, Yin Y, Zhou L, Chen X, Cheng J. Serum amyloid A and clusterin as potential predictive biomarkers for severe hand, foot and mouth disease by 2D-DIGE proteomics analysis. PLoS One 2014; 9:e108816. [PMID: 25268271 PMCID: PMC4182520 DOI: 10.1371/journal.pone.0108816] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 09/02/2014] [Indexed: 01/23/2023] Open
Abstract
Hand, foot, and mouth disease (HFMD) affects more than one million children, is responsible for several hundred child deaths every year in China and is the cause of widespread concerns in society. Only a small fraction of HFMD cases will develop further into severe HFMD with neurologic complications. A timely and accurate diagnosis of severe HFMD is essential for assessing the risk of progression and planning the appropriate treatment. Human serum can reflect the physiological or pathological states, which is expected to be an excellent source of disease-specific biomarkers. In the present study, a comparative serological proteome analysis between severe HFMD patients and healthy controls was performed via a two-dimensional difference gel electrophoresis (2D-DIGE) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) strategy. Fifteen proteins were identified as differentially expressed in the sera of the severe HFMD patients compared with the controls. The identified proteins were classified into different groups according to their molecular functions, biological processes, protein classes and physiological pathways by bioinformatics analysis. The up-regulations of two identified proteins, serum amyloid A (SAA) and clusterin (CLU), were confirmed in the sera of the HFMD patients by ELISA assay. This study not only increases our background knowledge about and scientific insight into the mechanisms of HFMD, but also reveals novel potential biomarkers for the clinical diagnosis of severe HFMD.
Collapse
Affiliation(s)
- Jianjun Liu
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Peiwu Huang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Wen-Xu Hong
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xiaohu Ren
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xifei Yang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yanxia He
- Shenzhen Children’s Hospital, Shenzhen, China
| | | | - Renli Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Hong Yang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Zhiguang Zhao
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Haiyan Huang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Long Chen
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Dejian Zhao
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Huixia Xian
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Fang Yang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Dongli Ma
- Shenzhen Children’s Hospital, Shenzhen, China
| | - Linqing Yang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yundong Yin
- Shenzhen Children’s Hospital, Shenzhen, China
| | - Li Zhou
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | | | - Jinquan Cheng
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- * E-mail:
| |
Collapse
|
72
|
Maarouf CL, Kokjohn TA, Walker DG, Whiteside CM, Kalback WM, Whetzel A, Sue LI, Serrano G, Jacobson SA, Sabbagh MN, Reiman EM, Beach TG, Roher AE. Biochemical assessment of precuneus and posterior cingulate gyrus in the context of brain aging and Alzheimer's disease. PLoS One 2014; 9:e105784. [PMID: 25166759 PMCID: PMC4148328 DOI: 10.1371/journal.pone.0105784] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/24/2014] [Indexed: 12/12/2022] Open
Abstract
Defining the biochemical alterations that occur in the brain during “normal” aging is an important part of understanding the pathophysiology of neurodegenerative diseases and of distinguishing pathological conditions from aging-associated changes. Three groups were selected based on age and on having no evidence of neurological or significant neurodegenerative disease: 1) young adult individuals, average age 26 years (n = 9); 2) middle-aged subjects, average age 59 years (n = 5); 3) oldest-old individuals, average age 93 years (n = 6). Using ELISA and Western blotting methods, we quantified and compared the levels of several key molecules associated with neurodegenerative disease in the precuneus and posterior cingulate gyrus, two brain regions known to exhibit early imaging alterations during the course of Alzheimer’s disease. Our experiments revealed that the bioindicators of emerging brain pathology remained steady or decreased with advancing age. One exception was S100B, which significantly increased with age. Along the process of aging, neurofibrillary tangle deposition increased, even in the absence of amyloid deposition, suggesting the presence of amyloid plaques is not obligatory for their development and that limited tangle density is a part of normal aging. Our study complements a previous assessment of neuropathology in oldest-old subjects, and within the limitations of the small number of individuals involved in the present investigation, it adds valuable information to the molecular and structural heterogeneity observed along the course of aging and dementia. This work underscores the need to examine through direct observation how the processes of amyloid deposition unfold or change prior to the earliest phases of dementia emergence.
Collapse
Affiliation(s)
- Chera L. Maarouf
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Tyler A. Kokjohn
- Department of Microbiology, Midwestern University, Glendale, Arizona, United States of America
| | - Douglas G. Walker
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Charisse M. Whiteside
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Walter M. Kalback
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Alexis Whetzel
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Lucia I. Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Geidy Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Sandra A. Jacobson
- Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Marwan N. Sabbagh
- Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, Arizona, United States of America
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Alex E. Roher
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
- * E-mail:
| |
Collapse
|
73
|
Genetic markers for diagnosis and pathogenesis of Alzheimer's disease. Gene 2014; 545:185-93. [DOI: 10.1016/j.gene.2014.05.031] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/07/2014] [Accepted: 05/13/2014] [Indexed: 02/06/2023]
|
74
|
Algamal M, Milojevic J, Jafari N, Zhang W, Melacini G. Mapping the interactions between the Alzheimer's Aβ-peptide and human serum albumin beyond domain resolution. Biophys J 2014; 105:1700-9. [PMID: 24094411 DOI: 10.1016/j.bpj.2013.08.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/14/2013] [Accepted: 08/20/2013] [Indexed: 01/14/2023] Open
Abstract
Human serum albumin (HSA) is a potent inhibitor of Aβ self-association and this novel, to our knowledge, function of HSA is of potential therapeutic interest for the treatment of Alzheimer's disease. It is known that HSA interacts with Aβ oligomers through binding sites evenly partitioned across the three albumin domains and with comparable affinities. However, as of this writing, no information is available on the HSA-Aβ interactions beyond domain resolution. Here, we map the HSA-Aβ interactions at subdomain and peptide resolution. We show that each separate subdomain of HSA domain 3 inhibits Aβ self-association. We also show that fatty acids (FAs) compete with Aβ oligomers for binding to domain 3, but the determinant of the HSA/Aβ oligomer interactions are markedly distinct from those of FAs. Although salt bridges with the FA carboxylate determine the FA binding affinities, hydrophobic contacts are pivotal for Aβ oligomer recognition. Specifically, we identified a site of Aβ oligomer recognition that spans the HSA (494-515) region and aligns with the central hydrophobic core of Aβ. The HSA (495-515) segment includes residues affected by FA binding and this segment is prone to self-associate into β-amyloids, suggesting that sites involved in fibrilization may provide a lead to develop inhibitors of Aβ self-association.
Collapse
Affiliation(s)
- Moustafa Algamal
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | |
Collapse
|
75
|
Doudevski I, Rostagno A, Cowman M, Liebmann J, Ritch R, Ghiso J. Clusterin and complement activation in exfoliation glaucoma. Invest Ophthalmol Vis Sci 2014; 55:2491-9. [PMID: 24550356 DOI: 10.1167/iovs.13-12941] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE The study was done to better understand the biological significance of clusterin co-localization with the exfoliation deposits (XF deposits), and provide insight into a pathogenic mechanism involving activation of the complement system and its pro-inflammatory consequences in patients with exfoliation glaucoma. METHODS Exfoliation lens deposits were analyzed by high resolution atomic force microscopy imaging and confocal immunofluorescence. Levels of clusterin and vitronectin, as well as of the complement activation products C3a and soluble C5b-9, were assessed via ELISA. RESULTS Atomic-force microscopy examination of lenses with exfoliation syndrome (XFS) revealed a dense fibrillar network on the anterior, aqueous-bathed surface of the lens, while the epithelial side displayed no discernible structural features at the same resolution. Clusterin colocalized with XF deposits, demonstrating integral association with the fibrils. Levels of activation-derived complement components C3a and soluble C5b-9, as well as the complement inhibitors clusterin and vitronectin, were found significantly elevated (1.7-fold, P < 0.05; 4.1-fold, P < 0.05; 1.8-fold, P < 0.01; and 3.0-fold, P < 0.01, respectively) in aqueous humor from glaucoma patients with XFS compared to non-XFS glaucoma controls. CONCLUSIONS The data provide compelling evidence for the activation of the complement system in XFS, highlighting the generation of subproducts with potent proinflammatory activity, which are capable of triggering and chronically maintaining levels of subclinical inflammation, suggesting novel targets for therapeutic intervention. The colocalization of clusterin in exfoliation fibrils suggests a failed attempt to prevent tissue accumulation of protein aggregates, as seen in other protein folding disorders, likely due to the abnormal high levels of misfolded proteins overwhelming its chaperone capacity.
Collapse
Affiliation(s)
- Ivo Doudevski
- Department of Pathology, New York University School of Medicine, New York, New York, United States
| | | | | | | | | | | |
Collapse
|
76
|
Abstract
Alzheimer’s disease (AD) is a complex and heterogeneous neurodegenerative disorder, classified as either early onset (under 65 years of age), or late onset (over 65 years of age). Three main genes are involved in early onset AD: amyloid precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2). The apolipoprotein E (APOE) E4 allele has been found to be a main risk factor for late-onset Alzheimer’s disease. Additionally, genome-wide association studies (GWASs) have identified several genes that might be potential risk factors for AD, including clusterin (CLU), complement receptor 1 (CR1), phosphatidylinositol binding clathrin assembly protein (PICALM), and sortilin-related receptor (SORL1). Recent studies have discovered additional novel genes that might be involved in late-onset AD, such as triggering receptor expressed on myeloid cells 2 (TREM2) and cluster of differentiation 33 (CD33). Identification of new AD-related genes is important for better understanding of the pathomechanisms leading to neurodegeneration. Since the differential diagnoses of neurodegenerative disorders are difficult, especially in the early stages, genetic testing is essential for diagnostic processes. Next-generation sequencing studies have been successfully used for detecting mutations, monitoring the epigenetic changes, and analyzing transcriptomes. These studies may be a promising approach toward understanding the complete genetic mechanisms of diverse genetic disorders such as AD.
Collapse
Affiliation(s)
- Eva Bagyinszky
- Department of BioNano Technology Gachon University, Gyeonggi-do, South Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Seong Soo A An
- Department of BioNano Technology Gachon University, Gyeonggi-do, South Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Budang Hospital, Gyeonggi-do, South Korea
| |
Collapse
|
77
|
Carrano A, Snkhchyan H, Kooij G, van der Pol S, van Horssen J, Veerhuis R, Hoozemans J, Rozemuller A, de Vries HE. ATP-binding cassette transporters P-glycoprotein and breast cancer related protein are reduced in capillary cerebral amyloid angiopathy. Neurobiol Aging 2014; 35:565-75. [DOI: 10.1016/j.neurobiolaging.2013.09.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 08/30/2013] [Accepted: 09/10/2013] [Indexed: 12/12/2022]
|
78
|
Wang H, Eckel RH. What are lipoproteins doing in the brain? Trends Endocrinol Metab 2014; 25:8-14. [PMID: 24189266 PMCID: PMC4062975 DOI: 10.1016/j.tem.2013.10.003] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 09/30/2013] [Accepted: 10/02/2013] [Indexed: 12/25/2022]
Abstract
Lipoproteins in plasma transport lipids between tissues, however, only high-density lipoproteins (HDL) appear to traverse the blood-brain barrier (BBB); thus, lipoproteins found in the brain must be produced within the central nervous system. Apolipoproteins E (ApoE) and ApoJ are the most abundant apolipoproteins in the brain, are mostly synthesized by astrocytes, and are found on HDL. In the hippocampus and other brain regions, lipoproteins help to regulate neurobehavioral functions by processes that are lipoprotein receptor-mediated. Moreover, lipoproteins and their receptors also have roles in the regulation of body weight and energy balance, acting through lipoprotein lipase (LPL) and the low-density lipoprotein (LDL) receptor-related protein (LRP). Thus, understanding lipoproteins and their metabolism in the brain provides a new opportunity with potential therapeutic relevance.
Collapse
Affiliation(s)
- Hong Wang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
79
|
Li X, Ma Y, Wei X, Li Y, Wu H, Zhuang J, Zhao Z. Clusterin in Alzheimer's disease: a player in the biological behavior of amyloid-beta. Neurosci Bull 2013; 30:162-8. [PMID: 24353014 DOI: 10.1007/s12264-013-1391-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/29/2013] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) remains a major killer, and although its pathogenesis varies, one dominant feature is an increase in the expression, formation, and sedimentation of senile plaques of amyloid-beta (Aβ) peptides in the brain. The chaperone protein clusterin has, since its first discovery at the end of the 20(th) century, been labeled as a cytoprotector. However, epigenetic studies showing that clusterin is associated with the severity and risk of AD, especially in the hippocampus, triggered studies to clarify its role in the pathogenesis of AD. It is true that clusterin can inhibit the aggregation of Aβ and therefore prevent further formation of senile plaques in the AD brain, yet it induces the formation of soluble forms of Aβ which are toxic to neurons. Another problematic finding is that clusterin is involved in a pathway through which Aβ has neurodegenerative effects intracellularly. Although the role of clusterin in the pathogenesis of AD is still not clear, this review specifically discusses the interactions between clusterin and Aβ, to open up the possibility of a potential therapeutic approach for treating AD.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | | | | | | | | | | | | |
Collapse
|
80
|
Nizamutdinov II, Andreeva TV, Stepanov VA, Marusin AV, Rogaev EI, Zasedatelev AS, Nasedkina TV. Biochip for determination of genetic markers of sporadic Alzheimer’s disease risk in the Russian Slavic population. Mol Biol 2013. [DOI: 10.1134/s0026893313060101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
81
|
Marques F, Sousa JC, Sousa N, Palha JA. Blood-brain-barriers in aging and in Alzheimer's disease. Mol Neurodegener 2013; 8:38. [PMID: 24148264 PMCID: PMC4015275 DOI: 10.1186/1750-1326-8-38] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/23/2013] [Indexed: 12/26/2022] Open
Abstract
The aging process correlates with a progressive failure in the normal cellular and organ functioning; these alterations are aggravated in Alzheimer's disease (AD). In both aging and AD there is a general decrease in the capacity of the body to eliminate toxic compounds and, simultaneously, to supply the brain with relevant growth and nutritional factors. The barriers of the brain are targets of this age related dysfunction; both the endothelial cells of the blood-brain barrier and the choroid plexus epithelial cells of the blood-cerebrospinal fluid barrier decrease their secretory capacity towards the brain and their ability to remove toxic compounds from the brain. Additionally, during normal aging and in AD, the permeability of the brain barriers increase. As such, a greater contact of the brain parenchyma with the blood content alters the highly controlled neural environment, which impacts on neural function. Of interest, the brain barriers are more than mere obstacles to the passage of molecules and cells, and therefore active players in brain homeostasis, which is still to be further recognized and investigated in the context of health and disease. Herein, we provide a review on how the brain barriers change during aging and in AD and how these processes impact on brain function.
Collapse
Affiliation(s)
- Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga 4710-057, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimaraes, Portugal
| | - João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga 4710-057, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimaraes, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga 4710-057, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimaraes, Portugal
| | - Joana Almeida Palha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga 4710-057, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimaraes, Portugal
| |
Collapse
|
82
|
Abstract
Adenocarcinoma of the prostate is the most common cancer in men in the Western Hemisphere. This diagnosis includes a clinicopathologically diverse collection of disease entities, encompassing a spectrum from early localized disease to advanced-stage castration-sensitive and ultimately metastatic, castration-resistant states. Although early-stage disease is treatable and potentially curable, treatment options for castration-resistant prostate cancer, the common pathway to prostate cancer death, remain limited and palliative in nature. Therapeutic resistance to androgen blockade, cytotoxic chemotherapy, and radiotherapy is underpinned by a number of cellular mechanisms. The upregulation of protective, antiapoptotic chaperone proteins is one of these mechanisms, and is exemplified by the protein clusterin in castration-resistant prostate cancer. Antisense oligonucleotide technology provides the potential to inhibit specific genes in cancer cells and with this the possibility of a vast impact in oncology, but no antisense drugs have been approved for use in cancer patients to date. Custirsen (OGX-011) is a novel antisense oligonucleotide drug which targets clusterin expression, and its application in prostate cancer is reviewed in this article.
Collapse
Affiliation(s)
- Sohaib Al-Asaaed
- Division of Medical Oncology, Department of Oncology, Western University, London, ON, Canada
| | | |
Collapse
|
83
|
Giménez-Llort L, Rivera-Hernández G, Marin-Argany M, Sánchez-Quesada JL, Villegas S. Early intervention in the 3xTg-AD mice with an amyloid β-antibody fragment ameliorates first hallmarks of Alzheimer disease. MAbs 2013; 5:665-77. [PMID: 23884018 DOI: 10.4161/mabs.25424] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The single-chain variable fragment, scFv-h3D6, has been shown to prevent in vitro toxicity induced by the amyloid β (Aβ) peptide in neuroblastoma cell cultures by withdrawing Aβ oligomers from the amyloid pathway. Present study examined the in vivo effects of scFv-h3D6 in the triple-transgenic 3xTg-AD mouse model of Alzheimer disease. Prior to the treatment, five-month-old female animals, corresponding to early stages of the disease, showed the first behavioral and psychological symptoms of dementia -like behaviors. Cognitive deficits included long- and short-term learning and memory deficits and high swimming navigation speed. After a single intraperitoneal dose of scFv-h3D6, the swimming speed was reversed to normal levels and the learning and memory deficits were ameliorated. Brain tissues of these animals revealed a global decrease of Aβ oligomers in the cortex and olfactory bulb after treatment, but this was not seen in the hippocampus and cerebellum. In the untreated 3xTg-AD animals, we observed an increase of both apoJ and apoE concentrations in the cortex, as well as an increase of apoE in the hippocampus. Treatment significantly recovered the non-pathological levels of these apolipoproteins. Our results suggest that the benefit of scFv-h3D6 occurs at both behavioral and molecular levels.
Collapse
Affiliation(s)
- Lydia Giménez-Llort
- Institut de Neurociències; Unitat de Biociències; Universitat Autònoma de Barcelona; Barcelona, Spain; Departament de Psiquiatria i Medicina Legal; Unitat de Biociències; Universitat Autònoma de Barcelona; Barcelona, Spain
| | | | | | | | | |
Collapse
|
84
|
Affiliation(s)
- Amy R. Wyatt
- School of Biological Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia;
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Justin J. Yerbury
- School of Biological Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia;
| | - Heath Ecroyd
- School of Biological Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia;
| | - Mark R. Wilson
- School of Biological Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia;
| |
Collapse
|
85
|
Advances in blood-based protein biomarkers for Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2013; 5:18. [PMID: 23659521 PMCID: PMC3706757 DOI: 10.1186/alzrt172] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that accounts for the majority of dementia cases. While research over the past decades has made advances into understanding disease pathology, definite AD diagnosis currently relies on confirmation by autopsy. The anticipated dramatic rise in affected individuals over the next decades necessitates the development of diagnostic tests applicable to living individuals, which depends on identification of disease biomarkers. Diagnostics based on blood protein biomarkers are particularly desired since these would allow for economical, rapid and non-invasive analysis of individual biomarker profiles. Research is actively ongoing in this field and has led to the identification of autoantibodies and various proteins in the blood that may represent a disease-specific blood signature of AD. This review provides an overview on the progress in the field of identification of AD-specific blood protein biomarkers.
Collapse
|
86
|
Xu G, Stevens SM, Moore BD, McClung S, Borchelt DR. Cytosolic proteins lose solubility as amyloid deposits in a transgenic mouse model of Alzheimer-type amyloidosis. Hum Mol Genet 2013; 22:2765-74. [PMID: 23512986 DOI: 10.1093/hmg/ddt121] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The extracellular accumulation of β-amyloid peptide is a key trigger in the pathogenesis of Alzheimer's disease (AD). In humans, amyloid deposition precedes the appearance of intracellular inclusion pathology formed by cytosolic proteins such as Tau, α-synuclein and TDP-43. These secondary pathologies have not been observed in mice that model Alzheimer-type amyloidosis by expressing mutant amyloid precursor protein, with or without mutant presenilin 1. The lack of secondary pathology in these models has made it difficult to establish how amyloid deposition initiates the cascade of events that leads to secondary intracellular pathology that characterizes human AD. In transgenic mice that model Alzheimer-type amyloidosis, we sought to determine whether there is evidence of altered cytosolic protein folding by assessing whether amyloid deposition causes normally soluble proteins to misfold. Using a method that involved detergent extraction and sedimentation coupled with proteomic approaches, we identified numerous cytosolic proteins that show specific losses in solubility as amyloid accumulates. The proteins identified included glycolytic enzymes and members of the 14-3-3 chaperone family. A substantial accumulation of lysine 48-linked polyubiquitin was also detected. Overall, the data demonstrate that the accumulation of amyloid by some manner causes the loss of solubility intracellular cytosolic proteins.
Collapse
Affiliation(s)
- Guilian Xu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
87
|
Elias-Sonnenschein LS, Bertram L, Visser PJ. Relationship between genetic risk factors and markers for Alzheimer's disease pathology. Biomark Med 2013; 6:477-95. [PMID: 22917148 DOI: 10.2217/bmm.12.56] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by neuritic plaques (main constituent: β-amyloid [Aβ]) and neurofibrillary tangles (hyperphosphorylated tau protein) in the brain. Abnormalities in Aβ and tau can be measured upon neuropathological examination, in cerebrospinal fluid or by PET. Etiologically, a growing body of evidence suggests that susceptibility to AD is genetically controlled. However, the precise nature of the underlying risk genes and their relation to AD biomarkers remains largely elusive. To this end, we performed a qualitative review of 17 studies (covering 47 polymorphisms in 26 genes) and investigated the potential relation between the most compelling AD risk genes and markers for Aβ and tau in cerebrospinal fluid, PET imaging and neuropathological examination. Of all covered genes, only APOE and PICALM showed consistent effects on Aβ but not on tau, while no obvious effects were observed for CLU, CR1, ACE, SORL and MAPT.
Collapse
Affiliation(s)
- Lyzel S Elias-Sonnenschein
- School for Mental Health & Neuroscience, Alzheimer Centrum Limburg, Department of Psychiatry & Neuropsychology, Maastricht University, Maastricht, The Netherlands.
| | | | | |
Collapse
|
88
|
Barber RC. The genetics of Alzheimer's disease. SCIENTIFICA 2012; 2012:246210. [PMID: 24278680 PMCID: PMC3820554 DOI: 10.6064/2012/246210] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/28/2012] [Indexed: 06/02/2023]
Abstract
Alzheimer's disease is a progressive, neurodegenerative disease that represents a growing global health crisis. Two major forms of the disease exist: early onset (familial) and late onset (sporadic). Early onset Alzheimer's is rare, accounting for less than 5% of disease burden. It is inherited in Mendelian dominant fashion and is caused by mutations in three genes (APP, PSEN1, and PSEN2). Late onset Alzheimer's is common among individuals over 65 years of age. Heritability of this form of the disease is high (79%), but the etiology is driven by a combination of genetic and environmental factors. A large number of genes have been implicated in the development of late onset Alzheimer's. Examples that have been confirmed by multiple studies include ABCA7, APOE, BIN1, CD2AP, CD33, CLU, CR1, EPHA1, MS4A4A/MS4A4E/MS4A6E, PICALM, and SORL1. Despite tremendous progress over the past three decades, roughly half of the heritability for the late onset of the disease remains unidentified. Finding the remaining genetic factors that contribute to the development of late onset Alzheimer's disease holds the potential to provide novel targets for treatment and prevention, leading to the development of effective strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Robert C. Barber
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| |
Collapse
|
89
|
Resenberger UK, Müller V, Munter LM, Baier M, Multhaup G, Wilson MR, Winklhofer KF, Tatzelt J. The heat shock response is modulated by and interferes with toxic effects of scrapie prion protein and amyloid β. J Biol Chem 2012; 287:43765-76. [PMID: 23115236 PMCID: PMC3527961 DOI: 10.1074/jbc.m112.389007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 10/27/2012] [Indexed: 12/19/2022] Open
Abstract
The heat shock response (HSR) is an evolutionarily conserved pathway designed to maintain proteostasis and to ameliorate toxic effects of aberrant protein folding. We have studied the modulation of the HSR by the scrapie prion protein (PrP(Sc)) and amyloid β peptide (Aβ) and investigated whether an activated HSR or the ectopic expression of individual chaperones can interfere with PrP(Sc)- or Aβ-induced toxicity. First, we observed different effects on the HSR under acute or chronic exposure of cells to PrP(Sc) or Aβ. In chronically exposed cells the threshold to mount a stress response was significantly increased, evidenced by a decreased expression of Hsp72 after stress, whereas an acute exposure lowered the threshold for stress-induced expression of Hsp72. Next, we employed models of PrP(Sc)- and Aβ-induced toxicity to demonstrate that the induction of the HSR ameliorates the toxic effects of both PrP(Sc) and Aβ. Similarly, the ectopic expression of cytosolic Hsp72 or the extracellular chaperone clusterin protected against PrP(Sc)- or Aβ-induced toxicity. However, toxic signaling induced by a pathogenic PrP mutant located at the plasma membrane was prevented by an activated HSR or Hsp72 but not by clusterin, indicating a distinct mode of action of this extracellular chaperone. Our study supports the notion that different pathological protein conformers mediate toxic effects via similar cellular pathways and emphasizes the possibility to exploit the heat shock response therapeutically.
Collapse
Affiliation(s)
- Ulrike K. Resenberger
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
| | - Veronika Müller
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
| | - Lisa M. Munter
- Institut für Chemie und Biochemie, Freie Universität, 14195 Berlin, Germany
- the Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3A0G4, Canada
| | | | - Gerd Multhaup
- Institut für Chemie und Biochemie, Freie Universität, 14195 Berlin, Germany
- the Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3A0G4, Canada
| | - Mark R. Wilson
- the School of Biological Sciences, University of Wollongong, Wollongong, New South Wales 2522, Australia, and
| | - Konstanze F. Winklhofer
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
- the German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany
| | - Jörg Tatzelt
- From the Neurobiochemistry, Adolf-Butenandt-Institute, Ludwig-Maximilians-University Munich, D-80336 Munich, Germany
- the German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany
| |
Collapse
|
90
|
Karch CM, Jeng AT, Nowotny P, Cady J, Cruchaga C, Goate AM. Expression of novel Alzheimer's disease risk genes in control and Alzheimer's disease brains. PLoS One 2012; 7:e50976. [PMID: 23226438 PMCID: PMC3511432 DOI: 10.1371/journal.pone.0050976] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 10/29/2012] [Indexed: 01/18/2023] Open
Abstract
Late onset Alzheimer’s disease (LOAD) etiology is influenced by complex interactions between genetic and environmental risk factors. Large-scale genome wide association studies (GWAS) for LOAD have identified 10 novel risk genes: ABCA7, BIN1, CD2AP, CD33, CLU, CR1, EPHA1, MS4A6A, MS4A6E, and PICALM. We sought to measure the influence of GWAS single nucleotide polymorphisms (SNPs) and gene expression levels on clinical and pathological measures of AD in brain tissue from the parietal lobe of AD cases and age-matched, cognitively normal controls. We found that ABCA7, CD33, and CR1 expression levels were associated with clinical dementia rating (CDR), with higher expression being associated with more advanced cognitive decline. BIN1 expression levels were associated with disease progression, where higher expression was associated with a delayed age at onset. CD33, CLU, and CR1 expression levels were associated with disease status, where elevated expression levels were associated with AD. Additionally, MS4A6A expression levels were associated with Braak tangle and Braak plaque scores, with elevated expression levels being associated with more advanced brain pathology. We failed to detect an association between GWAS SNPs and gene expression levels in our brain series. The minor allele of rs3764650 in ABCA7 is associated with age at onset and disease duration, and the minor allele of rs670139 in MS4A6E was associated with Braak tangle and Braak plaque score. These findings suggest that expression of some GWAS genes, namely ABCA7, BIN1, CD33, CLU, CR1 and the MS4A family, are altered in AD brains.
Collapse
Affiliation(s)
- Celeste M Karch
- Department of Psychiatry and Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
91
|
Narayan P, Meehan S, Carver JA, Wilson MR, Dobson CM, Klenerman D. Amyloid-β oligomers are sequestered by both intracellular and extracellular chaperones. Biochemistry 2012; 51:9270-6. [PMID: 23106396 PMCID: PMC4981287 DOI: 10.1021/bi301277k] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The aberrant aggregation of the amyloid-β peptide into β-sheet rich, fibrillar structures proceeds via a heterogeneous ensemble of oligomeric intermediates that have been associated with neurotoxicity in Alzheimer's disease (AD). Of particular interest in this context are the mechanisms by which molecular chaperones, part of the primary biological defenses against protein misfolding, influence Aβ aggregation. We have used single-molecule fluorescence techniques to compare the interactions between distinct aggregation states (monomers, oligomers, and amyloid fibrils) of the AD-associated amyloid-β(1-40) peptide, and two molecular chaperones, both of which are upregulated in the brains of patients with AD and have been found colocalized with Aβ in senile plaques. One of the chaperones, αB-crystallin, is primarily found inside cells, while the other, clusterin, is predominantly located in the extracellular environment. We find that both chaperones bind to misfolded oligomeric species and form long-lived complexes, thereby preventing both their further growth into fibrils and their dissociation. From these studies, we conclude that these chaperones have a common mechanism of action based on sequestering Aβ oligomers. This conclusion suggests that these chaperones, both of which are ATP-independent, are able to inhibit potentially pathogenic Aβ oligomer-associated processes whether they occur in the extracellular or intracellular environment.
Collapse
Affiliation(s)
- Priyanka Narayan
- Department of Chemistry, University of Cambridge, Lensfield Road,
Cambridge, UK CB2 1EW
| | - Sarah Meehan
- Department of Chemistry, University of Cambridge, Lensfield Road,
Cambridge, UK CB2 1EW
| | - John A. Carver
- School of Chemistry and Physics, University of Adelaide, Adelaide,
South Australia 5005, Australia
| | - Mark R. Wilson
- School of Biological Sciences, University of Wollongong, Wollongong,
New South Wales 2522, Australia
| | - Christopher M. Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road,
Cambridge, UK CB2 1EW
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Lensfield Road,
Cambridge, UK CB2 1EW
| |
Collapse
|
92
|
Begum N, Wang B, Mori M, Vares G. Does ionizing radiation influence Alzheimer's disease risk? JOURNAL OF RADIATION RESEARCH 2012; 53:815-22. [PMID: 22872779 PMCID: PMC3483841 DOI: 10.1093/jrr/rrs036] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD) is a human neurodegenerative disease, and its global prevalence is predicted to increase dramatically in the following decades. There is mounting evidence describing the effects of ionizing radiation (IR) on the brain, suggesting that exposure to IR might ultimately favor the development of AD. Therefore better understanding the possible connections between exposure to IR and AD pathogenesis is of utmost importance. In this review, recent developments in the research on the biological and cognitive effects of IR in the brain will be explored. Because AD is largely an age-related pathology, the effects of IR on ageing will be investigated.
Collapse
Affiliation(s)
- Nasrin Begum
- Center for Nuclear Medicine and Ultrasound, Rajshahi Medical College Hospital Campus, GPO Box No. 35, Rajshahi, Bangladesh
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Masahiko Mori
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Guillaume Vares
- National Institute of Radiological Sciences, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- Corresponding author. Tel: +81-(0)43-206-4730;
| |
Collapse
|
93
|
Sagare AP, Deane R, Zlokovic BV. Low-density lipoprotein receptor-related protein 1: a physiological Aβ homeostatic mechanism with multiple therapeutic opportunities. Pharmacol Ther 2012; 136:94-105. [PMID: 22820095 PMCID: PMC3432694 DOI: 10.1016/j.pharmthera.2012.07.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/03/2012] [Indexed: 11/29/2022]
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is the main cell surface receptor involved in brain and systemic clearance of the Alzheimer's disease (AD) toxin amyloid-beta (Aβ). In plasma, a soluble form of LRP1 (sLRP1) is the major transport protein for peripheral Aβ. LRP1 in brain endothelium and mural cells mediates Aβ efflux from brain by providing a transport mechanism for Aβ across the blood-brain barrier (BBB). sLRP1 maintains a plasma 'sink' activity for Aβ through binding of peripheral Aβ which in turn inhibits re-entry of free plasma Aβ into the brain. LRP1 in the liver mediates systemic clearance of Aβ. In AD, LRP1 expression at the BBB is reduced and Aβ binding to circulating sLRP1 is compromised by oxidation. Cell surface LRP1 and circulating sLRP1 represent druggable targets which can be therapeutically modified to restore the physiological mechanisms of brain Aβ homeostasis. In this review, we discuss how increasing LRP1 expression at the BBB and liver with lifestyle changes, statins, plant-based active principles and/or gene therapy on one hand, and how replacing dysfunctional plasma sLRP1 on the other regulate Aβ clearance from brain ultimately controlling the onset and/or progression of AD.
Collapse
Affiliation(s)
- Abhay P. Sagare
- Department of Physiology and Biophysics, and Center for Neurodegeneration and Regeneration at the Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, 1501 San Pablo Street, Los Angeles, CA 90089, United States
| | - Rashid Deane
- Department of Neurosurgery, Arthur Kornberg Medical Research Building, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Berislav V. Zlokovic
- Department of Physiology and Biophysics, and Center for Neurodegeneration and Regeneration at the Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, 1501 San Pablo Street, Los Angeles, CA 90089, United States
| |
Collapse
|
94
|
Ferrari R, Moreno JH, Minhajuddin AT, O'Bryant SE, Reisch JS, Barber RC, Momeni P. Implication of common and disease specific variants in CLU, CR1, and PICALM. Neurobiol Aging 2012; 33:1846.e7-18. [DOI: 10.1016/j.neurobiolaging.2012.01.110] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/25/2012] [Accepted: 01/30/2012] [Indexed: 11/30/2022]
|
95
|
Roles of Extracellular Chaperones in Amyloidosis. J Mol Biol 2012; 421:499-516. [DOI: 10.1016/j.jmb.2012.01.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 01/24/2023]
|
96
|
Hamley IW. The Amyloid Beta Peptide: A Chemist’s Perspective. Role in Alzheimer’s and Fibrillization. Chem Rev 2012; 112:5147-92. [DOI: 10.1021/cr3000994] [Citation(s) in RCA: 670] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- I. W. Hamley
- Department
of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD,
U.K
| |
Collapse
|
97
|
Rodrigues R, Smith MA, Wang X, Perry G, Lee HG, Zhu X, Petersen RB. Molecular neuropathogenesis of Alzheimer's disease: an interaction model stressing the central role of oxidative stress. FUTURE NEUROLOGY 2012; 7:287-305. [PMID: 23086377 DOI: 10.2217/fnl.12.27] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) exhibits a complex etiology that simultaneously manifests as a complex cellular, neurobiological, molecular, anatomic-physiological and clinical entity. Other significant psychiatric conditions, such as depression and schizophrenia, may also present with complex and concurrent clinical and/or molecular phenotypes. These neuropsychiatric pathologies also originate from both environmental and genetic factors. We analyzed the molecular phenotypes of AD and discuss them with respect to the classical theories, which we integrated into mechanisms that share molecular and/or anatomical connections. Based on these mechanisms, we propose an interaction model and discuss the model in light of studies that refute or support it. Given the spectrum of AD phenotypes, we limit the scope of our discussion to a few, which facilitates concrete analysis. In addition, the study of specific, individual pathogenic phenotypes may be critical to defining the complex mechanisms leading to AD, thereby improving strategies for developing novel therapies.
Collapse
Affiliation(s)
- Roberto Rodrigues
- Ave. Icaraí Cristal 74 (Clinic), 90.810-000 Porto Alegre, Rio Grande do Sul (RS), Brazil
| | | | | | | | | | | | | |
Collapse
|
98
|
Ling IF, Bhongsatiern J, Simpson JF, Fardo DW, Estus S. Genetics of clusterin isoform expression and Alzheimer's disease risk. PLoS One 2012; 7:e33923. [PMID: 22506010 PMCID: PMC3323613 DOI: 10.1371/journal.pone.0033923] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 02/21/2012] [Indexed: 01/13/2023] Open
Abstract
The minor allele of rs11136000 within CLU is strongly associated with reduced Alzheimer's disease (AD) risk. The mechanism underlying this association is unclear. Here, we report that CLU1 and CLU2 are the two primary CLU isoforms in human brain; CLU1 and CLU2 share exons 2-9 but differ in exon 1 and proximal promoters. The expression of both CLU1 and CLU2 was increased in individuals with significant AD neuropathology. However, only CLU1 was associated with the rs11136000 genotype, with the minor "protective" rs11136000T allele being associated with increased CLU1 expression. Since CLU1 and CLU2 are predicted to encode intracellular and secreted proteins, respectively, we compared their expression; for both CLU1 and CLU2 transfected cells, clusterin is present in the secretory pathway, accumulates in the extracellular media, and is similar in size to clusterin in human brain. Overall, we interpret these results as indicating that the AD-protective minor rs11136000T allele is associated with increased CLU1 expression. Since CLU1 and CLU2 appear to produce similar proteins and are increased in AD, the AD-protection afforded by the rs11136000T allele may reflect increased soluble clusterin throughout life.
Collapse
Affiliation(s)
- I-Fang Ling
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jiraganya Bhongsatiern
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - James F. Simpson
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - David W. Fardo
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
99
|
Gu H, Wei X, Chen S, Kurz A, Müller U, Gasser T, Dodel RC, Farlow MR, Du Y. Association of clusterin gene polymorphisms with late-onset Alzheimer's disease. Dement Geriatr Cogn Disord 2012; 32:198-201. [PMID: 22122982 DOI: 10.1159/000331276] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Some studies have implicated the role of apolipoprotein J [clusterin (CLU), apoJ] in the pathogenesis of Alzheimer's disease (AD). In this study, we investigated the polymorphisms rs11136000 and rs9331888 within CLU in late-onset sporadic AD (LOAD) patients and nondemented subjects. METHODS LOAD patients and control subjects were analyzed. Genotyping of rs11136000 and rs9331888 was performed using standard PCR following different restriction endonuclease digestion. RESULTS Although there were no significant differences in genotype frequencies of these two polymorphisms, the haplotype CG was associated with a statistically significantly increased LOAD risk. CONCLUSION The rs11136000 and rs9331888 polymorphisms of the CLU gene are associated with LOAD.
Collapse
Affiliation(s)
- Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Clusterin, also known as apolipoprotein J, is a ubiquitous multifunctional glycoprotein. Following its identification in 1983, clusterin was found to be clearly increased in Alzheimer's disease (AD). Later research demonstrated that clusterin could bind amyloid-beta (Abeta) peptides and prevent fibril formation, a hallmark of AD pathology. In addition to preventing excessive inflammation, intracellular clusterin was found to reduce apoptosis and oxidative stress. Although early studies were inconclusive, two recent large-scale genome-wide association studies (GWAS) independently identified variants within the clusterin gene as risk factors for developing AD. This review focuses on the characteristics of clusterin and possible mechanisms of its relationship to AD.
Collapse
Affiliation(s)
- Zhong-Chen Wu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, Shandong Province, China
| | | | | | | |
Collapse
|