Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
J Alzheimers Dis ; 98(1): 163-186, 2024.
Article En | MEDLINE | ID: mdl-38393907

Background: Increased blood-brain barrier (BBB) permeability and amyloid-ß (Aß) peptides (especially Aß1-42) (Aß42) have been linked to Alzheimer's disease (AD) pathogenesis, but the nature of their involvement in AD-related neuropathological changes leading to cognitive changes remains poorly understood. Objective: To test the hypothesis that chronic extravasation of bloodborne Aß42 peptide and brain-reactive autoantibodies and their entry into the brain parenchyma via a permeable BBB contribute to AD-related pathological changes and cognitive changes in a mouse model. Methods: The BBB was rendered chronically permeable through repeated injections of Pertussis toxin (PT), and soluble monomeric, fluorescein isothiocyanate (FITC)-labeled or unlabeled Aß42 was injected into the tail-vein of 10-month-old male CD1 mice at designated intervals spanning ∼3 months. Acquisition of learned behaviors and long-term retention were assessed via a battery of cognitive and behavioral tests and linked to neuropathological changes. Results: Mice injected with both PT and Aß42 demonstrated a preferential deficit in the capacity for long-term retention and an increased susceptibility to interference in selective attention compared to mice exposed to PT or saline only. Immunohistochemical analyses revealed increased BBB permeability and entry of bloodborne Aß42 and immunoglobulin G (IgG) into the brain parenchyma, selective neuronal binding of IgG and neuronal accumulation of Aß42 in animals injected with both PT and Aß42 compared to controls. Conclusion: Results highlight the potential synergistic role of BBB compromise and the influx of bloodborne Aß42 into the brain in both the initiation and progression of neuropathologic and cognitive changes associated with AD.


Alzheimer Disease , Blood-Brain Barrier , Male , Mice , Animals , Blood-Brain Barrier/metabolism , Alzheimer Disease/pathology , Peptide Fragments/toxicity , Peptide Fragments/metabolism , Brain/pathology , Amyloid beta-Peptides/metabolism , Cognition , Immunoglobulin G/metabolism
2.
J Alzheimers Dis ; 92(3): 1077-1091, 2023.
Article En | MEDLINE | ID: mdl-36847005

BACKGROUND: Evidence for the universal presence of IgG autoantibodies in blood and their potential utility for the diagnosis of Alzheimer's disease (AD) and other neurodegenerative diseases has been extensively demonstrated by our laboratory. The fact that AD-related neuropathological changes in the brain can begin more than a decade before tell-tale symptoms emerge has made it difficult to develop diagnostic tests useful for detecting the earliest stages of AD pathogenesis. OBJECTIVE: To determine the utility of a panel of autoantibodies for detecting the presence of AD-related pathology along the early AD continuum, including at pre-symptomatic [an average of 4 years before the transition to mild cognitive impairment (MCI)/AD)], prodromal AD (MCI), and mild-moderate AD stages. METHODS: A total of 328 serum samples from multiple cohorts, including ADNI subjects with confirmed pre-symptomatic, prodromal, and mild-moderate AD, were screened using Luminex xMAP® technology to predict the probability of the presence of AD-related pathology. A panel of eight autoantibodies with age as a covariate was evaluated using randomForest and receiver operating characteristic (ROC) curves. RESULTS: Autoantibody biomarkers alone predicted the probability of the presence of AD-related pathology with 81.0% accuracy and an area under the curve (AUC) of 0.84 (95% CI = 0.78-0.91). Inclusion of age as a parameter to the model improved the AUC (0.96; 95% CI = 0.93-0.99) and overall accuracy (93.0%). CONCLUSION: Blood-based autoantibodies can be used as an accurate, non-invasive, inexpensive, and widely accessible diagnostic screener for detecting AD-related pathology at pre-symptomatic and prodromal AD stages that could aid clinicians in diagnosing AD.


Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/pathology , Cognitive Dysfunction/pathology , Biomarkers , ROC Curve , Autoantibodies
3.
Front Hum Neurosci ; 16: 836980, 2022.
Article En | MEDLINE | ID: mdl-35431844

Though hippocampal volume reduction is a pathological hallmark of schizophrenia, the molecular pathway(s) responsible for this degeneration remains unknown. Recent reports have suggested the potential role of impaired blood-brain barrier (BBB) function in schizophrenia pathogenesis. However, direct evidence demonstrating an impaired BBB function is missing. In this preliminary study, we used immunohistochemistry and serum immunoglobulin G (IgG) antibodies to investigate the state of BBB function in formalin-fixed postmortem samples from the hippocampus and surrounding temporal cortex of patients with schizophrenia (n = 25) and controls without schizophrenia (n = 27) matched for age, sex, and race. Since a functional BBB prevents the extravasation of IgGs, detection of IgGs in the parenchyma is used as direct evidence of BBB breakdown. We also developed a semi-quantitative approach to quantify the extent of IgG leak and therein BBB breach. Analysis of our immunohistochemistry data demonstrated a significantly higher incidence of IgG leak in patients with schizophrenia compared to controls. Further, BBB permeability was significantly higher in advanced-age patients with schizophrenia than both advanced-age controls and middle-aged patients with schizophrenia. Male patients with schizophrenia also demonstrated a significant increase in IgG permeability compared to control males. Interestingly, the extravasated IgGs also demonstrated selective immunoreactivity for neurons. Based on these observations, we suggest that BBB dysfunction and IgG autoantibodies could be two key missing pathoetiological links underwriting schizophrenia hippocampal damage.

4.
J Alzheimers Dis ; 74(1): 345-361, 2020.
Article En | MEDLINE | ID: mdl-32039847

Blood-brain barrier (BBB) permeability is a recognized early feature of Alzheimer's disease (AD). In the present study, we examined consequences of increased BBB permeability on the development of AD-related pathology by tracking selected leaked plasma components and their interactions with neurons in vivo and in vitro. Histological sections of cortical regions of postmortem AD brains were immunostained to determine the distribution of amyloid-ß1-42 (Aß42), cathepsin D, IgG, GluR2/3, and alpha7 nicotinic acetylcholine receptor (α7nAChR). Results revealed that chronic IgG binding to pyramidal neurons coincided with internalization of Aß42, IgG, GluR2/3, and α7nAChR as well as lysosomal compartment expansion in these cells in regions of AD pathology. To test possible mechanistic interrelationships of these phenomena, we exposed differentiated SH-SY5Y neuroblastoma cells to exogenous, soluble Aß42 peptide and serum from AD and control subjects. The rate and extent of Aß42 internalization in these cells was enhanced by serum containing neuron-binding IgG autoantibodies. This was confirmed by treating cells with individual antibodies specific for α7nAChR, purified IgG from AD or non-AD sera, and sera devoid of IgG, in the presence of 100 nM Aß42. Initial co-localization of IgG, α7nAChR, and Aß42 was temporally and spatially linked to early endosomes (Rab11) and later to lysosomes (LAMP-1). Aß42 internalization was attenuated by treatment with monovalent F(ab) antibody fragments generated from purified IgG from AD serum and then rescued by coupling F(ab) fragments with divalent human anti-Fab. Overall, results suggest that cross-linking of neuron-binding autoantibodies targeting cell surface proteins can accelerate intraneuronal Aß42 deposition in AD.


Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Autoantibodies/immunology , Brain/immunology , Neurons/metabolism , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Blood-Brain Barrier , Cell Line, Tumor , Female , Humans , Immunoglobulin G/metabolism , Lysosomes/metabolism , Male , Membrane Proteins/metabolism , Pyramidal Cells/metabolism
5.
Brain Behav Immun Health ; 2: 100032, 2020 Feb.
Article En | MEDLINE | ID: mdl-38377421

The present study demonstrates, using human protein microarrays and plasma and cerebrospinal fluid samples obtained pre-surgically and simultaneously from 46 hip fracture repair patients, that CSF exhibits an extraordinarily complex IgG autoantibody profile composed of thousands of autoantibodies. We show that the pattern of expression levels of individual autoantibodies in CSF closely mimics that in the blood, regardless of age, gender or the presence or absence of disease, indicative of a blood-based origin for CSF autoantibodies. In addition, using five longitudinal serum samples obtained from one healthy individual over a span of nine years, we found that blood autoantibody profiles are remarkably stable over a long period of time, and that autoantibody profiles in both blood and CSF show features that are common among different individuals as well as individual-specific. Lastly, we demonstrate that an elevated CSF/plasma autoantibody ratio is more common in elderly hip fracture repair patients that experienced post-operative delirium than in non-delirium subjects, thus highlighting the crucial role that blood-brain and/or blood-CSF barrier compromise may play in the development of post-operative delirium.

6.
PLoS One ; 14(11): e0225178, 2019.
Article En | MEDLINE | ID: mdl-31730624

Post-operative delirium (POD) is the most common complication following major surgery in non-demented older (>65 y/o) patients. Patients experiencing POD show increased risk for future cognitive decline, including mild cognitive impairment (MCI) and Alzheimer's disease (AD) and, conversely, patients with cognitive decline at surgery show increased risk for POD. Here, we demonstrate that a previously established panel of AD-driven MCI (ADMCI) autoantibody (aAB) biomarkers can be used to detect prodromal AD pre-surgically in individuals admitted into the hospital for hip fracture repair (HFR) surgery. Plasma from 39 STRIDE (STRIDE: A Strategy to Reduce the Incidence of Postoperative Delirium in Elderly Patients) HFR patients and sera from 25 age- and sex-matched non-demented and non-surgical controls were screened using human protein microarrays to measure expression of a panel of 44 previously identified MCI aAB biomarkers. The predictive classification accuracy of the aAB biomarker panel was evaluated using Random Forest (RF). The ADMCI aAB biomarkers successfully distinguished 21 STRIDE HFR patients (CDR = 0.5) from 25 matched non-surgical controls with an overall accuracy of 91.3% (sensitivity = 95.2%; specificity = 88.0%). The ADMCI aAB panel also correctly identified six patients with preoperative CDR = 0 who later converted to CDR = 0.5 or >1 at one-year follow-up. Lastly, the majority of cognitively normal (CDR = 0) STRIDE HFR subjects that were positive for CSF AD biomarkers based on the A/T/N classification system were likewise classified as ADMCI aAB-positive using the biomarker panel. Results suggest that pre-surgical detection of ADMCI aAB biomarkers can readily identify HFR patients with likely early-stage AD pathology using pre-surgery blood samples, opening up the potential for early, blood-based AD detection and improvements in peri- and postoperative patient management.


Alzheimer Disease/diagnosis , Alzheimer Disease/etiology , Autoantibodies/immunology , Biomarkers , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Autoantibodies/blood , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Female , Hip Fractures/complications , Hip Fractures/therapy , Humans , Male , Protein Array Analysis , Sensitivity and Specificity , Severity of Illness Index
7.
Sci Rep ; 9(1): 2190, 2019 02 18.
Article En | MEDLINE | ID: mdl-30778117

Regulating the intrinsic interactions between blood vessels and nerve cells has the potential to enhance repair and regeneration of the central nervous system. Here, we evaluate the efficacy of aligned microvessels to induce and control directional axon growth from neural progenitor cells in vitro and host axons in a rat spinal cord injury model. Interstitial fluid flow aligned microvessels generated from co-cultures of cerebral-derived endothelial cells and pericytes in a three-dimensional scaffold. The endothelial barrier function was evaluated by immunostaining for tight junction proteins and quantifying the permeability coefficient (~10-7 cm/s). Addition of neural progenitor cells to the co-culture resulted in the extension of Tuj-positive axons in the direction of the microvessels. To validate these findings in vivo, scaffolds were transplanted into an acute spinal cord hemisection injury with microvessels aligned with the rostral-caudal direction. At three weeks post-surgery, sagittal sections indicated close alignment between the host axons and the transplanted microvessels. Overall, this work demonstrates the efficacy of exploiting neurovascular interaction to direct axon growth in the injured spinal cord and the potential to use this strategy to facilitate central nervous system regeneration.


Axon Guidance/physiology , Nerve Regeneration/physiology , Animals , Coculture Techniques , Disease Models, Animal , Endothelial Cells/physiology , Female , Guided Tissue Regeneration , In Vitro Techniques , Microvessels/growth & development , Microvessels/physiology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Rats , Rats, Sprague-Dawley , Spinal Cord/blood supply , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Tissue Scaffolds
8.
Schizophr Bull ; 45(1): 233-246, 2019 01 01.
Article En | MEDLINE | ID: mdl-29474698

Blood-based biomarker discovery for psychotic disorders has yet to impact upon routine clinical practice. In physical disorders antibodies have established roles as diagnostic, prognostic and predictive (theranostic) biomarkers, particularly in disorders thought to have a substantial autoimmune or infective aetiology. Two approaches to antibody biomarker identification are distinguished: a "top-down" approach, in which antibodies to specific antigens are sought based on the known function of the antigen and its putative role in the disorder, and emerging "bottom-up" or "omics" approaches that are agnostic as to the significance of any one antigen, using high-throughput arrays to identify distinctive components of the antibody repertoire. Here we review the evidence for antibodies (to self-antigens as well as infectious organism and dietary antigens) as biomarkers of diagnosis, prognosis, and treatment response in psychotic disorders. Neuronal autoantibodies have current, and increasing, clinical utility in the diagnosis of organic or atypical psychosis syndromes. Antibodies to selected infectious agents show some promise in predicting cognitive impairment and possibly other symptom domains (eg, suicidality) within psychotic disorders. Finally, infectious antibodies and neuronal and other autoantibodies have recently emerged as potential biomarkers of response to anti-infective therapies, immunotherapies, or other novel therapeutic strategies in psychotic disorders, and have a clear role in stratifying patients for future clinical trials. As in nonpsychiatric disorders, combining biomarkers and large-scale use of "bottom-up" approaches to biomarker identification are likely to maximize the eventual clinical utility of antibody biomarkers in psychotic disorders.


Antibodies/immunology , Biomarkers , Psychotic Disorders/diagnosis , Humans , Psychotic Disorders/immunology , Psychotic Disorders/physiopathology
9.
J Neuroimmunol ; 309: 51-57, 2017 08 15.
Article En | MEDLINE | ID: mdl-28601288

The goal of this preliminary proof-of-concept study was to use human protein microarrays to identify blood-based autoantibody biomarkers capable of diagnosing multiple sclerosis (MS). Using sera from 112 subjects, including 51 MS subjects, autoantibody biomarkers effectively differentiated MS subjects from age- and gender-matched normal and breast cancer controls with 95.0% and 100% overall accuracy, but not from subjects with Parkinson's disease. Autoantibody biomarkers were also useful in distinguishing subjects with the relapsing-remitting form of MS from those with the secondary progressive subtype. These results demonstrate that autoantibodies can be used as noninvasive blood-based biomarkers for the detection and subtyping of MS.


Autoantibodies/metabolism , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Chronic Progressive/diagnosis , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Adult , Aged , Biomarkers/blood , Female , Humans , Male , Middle Aged , Random Allocation
10.
Diab Vasc Dis Res ; 14(3): 200-213, 2017 05.
Article En | MEDLINE | ID: mdl-28301218

Using a porcine model of diabetes mellitus and hypercholesterolaemia, we previously showed that diabetes mellitus and hypercholesterolaemia is associated with a chronic increase in blood-brain barrier permeability in the cerebral cortex, leading to selective binding of immunoglobulin G and deposition of amyloid-beta1-42 peptide in pyramidal neurons. Treatment with Darapladib (GlaxoSmithKline, SB480848), an inhibitor of lipoprotein-associated phospholipase-A2, alleviated these effects. Here, investigation of the effects of chronic diabetes mellitus and hypercholesterolaemia on the pig retina revealed a corresponding increased permeability of the blood-retina barrier coupled with a leak of plasma components into the retina, alterations in retinal architecture, selective IgG binding to neurons in the ganglion cell layer, thinning of retinal layers due to cell loss and increased glial fibrillary acidic protein expression in Müller cells, all of which were curtailed by treatment with Darapladib. These findings suggest that chronic diabetes mellitus and hypercholesterolaemia induces increased blood-retina barrier permeability that may be linked to altered expression of blood-retina barrier-associated tight junction proteins, claudin and occludin, leading to structural changes in the retina consistent with diabetic retinopathy. Additionally, results suggest that drugs with vascular anti-inflammatory properties, such as Darapladib, may have beneficial effects on eye diseases strongly linked to vascular abnormalities such as diabetic retinopathy and age-related macular degeneration.


1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Benzaldehydes/pharmacology , Blood-Retinal Barrier/drug effects , Capillary Permeability/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/prevention & control , Hypercholesterolemia/drug therapy , Oximes/pharmacology , Phospholipase A2 Inhibitors/pharmacology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Blood-Retinal Barrier/enzymology , Blood-Retinal Barrier/pathology , Blood-Retinal Barrier/physiopathology , Claudin-5/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/physiopathology , Gliosis , Hypercholesterolemia/complications , Hypercholesterolemia/enzymology , Hypercholesterolemia/physiopathology , Immunoglobulin G/metabolism , Male , Occludin/metabolism , Protein Binding , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/enzymology , Sus scrofa
11.
Biomaterials ; 115: 30-39, 2017 01.
Article En | MEDLINE | ID: mdl-27886553

Transport of fluid and solutes is tightly controlled within the brain, where vasculature exhibits a blood-brain barrier and there is no organized lymphatic network facilitating waste transport from the interstitial space. Here, using a compliant, three-dimensional co-culture model of the blood-brain barrier, we show that mechanical stimuli exerted by blood flow mediate both the permeability of the endothelial barrier and waste transport along the basement membrane. Application of both shear stress and cyclic strain facilitates tight junction formation in the endothelial monolayer, with and without the presence of astrocyte endfeet in the surrounding matrix. We use both dextran perfusion and TEER measurements to assess the initiation and maintenance of the endothelial barrier, and microparticle image velocimetry to characterize the fluid dynamics within the in vitro vessels. Application of pulsatile flow to the in vitro vessels induces pulsatile strain to the vascular wall, providing an opportunity to investigate stretch-induced transport along the basement membrane. We find that a pulsatile wave speed of approximately 1 mm/s with Womersley number of 0.004 facilitates retrograde transport of high molecular weight dextran along the basement membrane between the basal endothelium and surrounding astrocytes. Together, these findings indicate that the mechanical stress exerted by blood flow is an important regulator of transport both across and along the walls of cerebral microvasculature.


Biological Transport, Active/physiology , Blood-Brain Barrier/physiology , Capillary Permeability/physiology , Endothelium, Vascular/physiology , Mechanotransduction, Cellular/physiology , Stress, Physiological/physiology , Tissue Engineering/methods , Batch Cell Culture Techniques/methods , Blood Flow Velocity/physiology , Cells, Cultured , Elastic Modulus/physiology , Humans , Printing, Three-Dimensional , Pulsatile Flow/physiology , Stress, Mechanical , Tight Junctions/physiology
12.
J Alzheimers Dis ; 54(2): 445-56, 2016 09 06.
Article En | MEDLINE | ID: mdl-27497477

Alzheimer's disease (AD) and diabetes mellitus (DM) are among the most pervasive and devastating disorders that afflict people throughout the world. Although typically associated with older demographics, recent epidemiologic studies have reported parallel trends in decreasing age of onset and increasing incidence of these conditions. Promising research continues to implicate the cerebrovasculature and blood-brain barrier (BBB) as playing key roles in AD pathoetiology. Similarly, complications accompanying DM, such as diabetic nephropathy/retinopathy, cardiovascular disease, and stroke, have been rooted in vascular compromise. Not surprisingly, DM is now considered a major risk factor for AD. The purpose of this review is to highlight investigations into the role of the cerebrovasculature in the development and progression of AD. We give particular attention to studies on humans and a variety of animal model systems that have demonstrated a link between BBB dysfunction and pathological changes in the brain consistent with aging and AD. Together, these studies suggest that the vascular complications associated with chronic, poorly managed DM can lead to subclinical BBB breakdown that precedes and drives the pathological changes progressing to symptomatic AD, providing a common mechanistic thread connecting these two disorders. Furthermore, this emphasizes the need to focus on the vasculature as a potential therapeutic target with the intent of limiting BBB breakdown involved in disease initiation and progression. In conclusion, AD may be more than just an associated comorbidity of DM, and instead another manifestation of the underlying vascular pathology that is common to both.


Alzheimer Disease/metabolism , Blood-Brain Barrier/metabolism , Cerebrovascular Circulation/physiology , Diabetes Mellitus/metabolism , Aging/metabolism , Aging/pathology , Alzheimer Disease/epidemiology , Alzheimer Disease/pathology , Animals , Blood-Brain Barrier/pathology , Capillary Permeability/physiology , Diabetes Mellitus/epidemiology , Diabetes Mellitus/pathology , Humans , Risk Factors
13.
Alzheimers Dement (Amst) ; 3: 51-62, 2016.
Article En | MEDLINE | ID: mdl-27239548

INTRODUCTION: There is an urgent need to identify biomarkers that can accurately detect and diagnose Alzheimer's disease (AD). Autoantibodies are abundant and ubiquitous in human sera and have been previously demonstrated as disease-specific biomarkers capable of accurately diagnosing mild-moderate stages of AD and Parkinson's disease. METHODS: Sera from 236 subjects, including 50 mild cognitive impairment (MCI) subjects with confirmed low CSF Aß42 levels, were screened with human protein microarrays to identify potential biomarkers for MCI. Autoantibody biomarker performance was evaluated using Random Forest and Receiver Operating Characteristic curves. RESULTS: Autoantibody biomarkers can differentiate MCI patients from age-matched and gender-matched controls with an overall accuracy, sensitivity, and specificity of 100.0%. They were also capable of differentiating MCI patients from those with mild-moderate AD and other neurologic and non-neurologic controls with high accuracy. DISCUSSION: Autoantibodies can be used as noninvasive and effective blood-based biomarkers for early diagnosis and staging of AD.

14.
Brain Res ; 1637: 154-167, 2016 Apr 15.
Article En | MEDLINE | ID: mdl-26907191

S100B is a calcium-sensor protein that impacts multiple signal transduction pathways. It is widely considered to be an important biomarker for several neuronal diseases as well as blood-brain barrier (BBB) breakdown. In this report, we demonstrate a BBB deficiency in mice that lack S100B through detection of leaked Immunoglobulin G (IgG) in the brain parenchyma. IgG leaks and IgG-binding to selected neurons were observed in S100B knockout (S100BKO) mice at 6 months of age but not at 3 months. By 9 months, IgG leaks persisted and the density of IgG-bound neurons increased significantly. These results reveal a chronic increase in BBB permeability upon aging in S100BKO mice for the first time. Moreover, coincident with the increase in IgG-bound neurons, autoantibodies targeting brain proteins were detected in the serum via western blots. These events were concurrent with compromise of neurons, increase of activated microglia and lack of astrocytic activation as evidenced by decreased expression of microtubule-associated protein type 2 (MAP2), elevated number of CD68 positive cells and unaltered expression of glial fibrillary acidic protein (GFAP) respectively. Results suggest a key role for S100B in maintaining BBB functional integrity and, further, propose the S100BKO mouse as a valuable model system to explore the link between chronic functional compromise of the BBB, generation of brain-reactive autoantibodies and neuronal dysfunctions.


Autoantibodies/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Neurons/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Age Factors , Animals , Immunoglobulin G/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neurons/immunology , Permeability , S100 Calcium Binding Protein beta Subunit/deficiency , S100 Calcium Binding Protein beta Subunit/immunology
15.
Biomed Res Int ; 2015: 937148, 2015.
Article En | MEDLINE | ID: mdl-26697497

Among the top ten causes of death in the United States, Alzheimer's disease (AD) is the only one that cannot be cured, prevented, or even slowed down at present. Significant efforts have been exerted in generating model systems to delineate the mechanism as well as establishing platforms for drug screening. In this study, a promising candidate model utilizing primary mouse brain organotypic (MBO) cultures is reported. For the first time, we have demonstrated that the MBO cultures exhibit increased blood brain barrier (BBB) permeability as shown by IgG leakage into the brain parenchyma, astrocyte activation as evidenced by increased expression of glial fibrillary acidic protein (GFAP), and neuronal damage-response as suggested by increased vimentin-positive neurons occur upon histamine treatment. Identical responses-a breakdown of the BBB, astrocyte activation, and neuronal expression of vimentin-were then demonstrated in brains from AD patients compared to age-matched controls, consistent with other reports. Thus, the histamine-treated MBO culture system may provide a valuable tool in combating AD.


Alzheimer Disease/metabolism , Blood-Brain Barrier/drug effects , Brain/pathology , Histamine/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/pathology , Brain/drug effects , Brain/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Histamine/pharmacology , Histamine/toxicity , Humans , Male , Mice , Neurons/metabolism , Neurons/pathology , Organ Culture Techniques , Vimentin/metabolism
16.
Immunol Lett ; 168(1): 80-8, 2015 Nov.
Article En | MEDLINE | ID: mdl-26386375

INTRODUCTION: There is a great need to identify readily accessible, blood-based biomarkers for Parkinson's disease (PD) that are useful for accurate early detection and diagnosis. This advancement would allow early patient treatment and enrollment into clinical trials, both of which would greatly facilitate the development of new therapies for PD. METHODS: Sera from a total of 398 subjects, including 103 early-stage PD subjects derived from the Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism (DATATOP) study, were screened with human protein microarrays containing 9,486 potential antigen targets to identify autoantibodies potentially useful as biomarkers for PD. A panel of selected autoantibodies with a higher prevalence in early-stage PD was identified and tested using Random Forest for its ability to distinguish early-stage PD subjects from controls and from individuals with other neurodegenerative and non-neurodegenerative diseases. RESULTS: Results demonstrate that a panel of selected, blood-borne autoantibody biomarkers can distinguish early-stage PD subjects (90% confidence in diagnosis) from age- and sex-matched controls with an overall accuracy of 87.9%, a sensitivity of 94.1% and specificity of 85.5%. These biomarkers were also capable of differentiating patients with early-stage PD from those with more advanced (mild-moderate) PD with an overall accuracy of 97.5%, and could distinguish subjects with early-stage PD from those with other neurological (e.g., Alzheimer's disease and multiple sclerosis) and non-neurological (e.g., breast cancer) diseases. CONCLUSION: These results demonstrate, for the first time, that a panel of selected autoantibodies may prove to be useful as effective blood-based biomarkers for the diagnosis of early-stage PD.


Autoantibodies/blood , Biomarkers/blood , Early Diagnosis , Parkinson Disease/blood , Parkinson Disease/diagnosis , Adult , Aged , Aged, 80 and over , Autoantibodies/immunology , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Parkinson Disease/immunology , Sensitivity and Specificity
17.
Int Rev Neurobiol ; 122: 1-51, 2015.
Article En | MEDLINE | ID: mdl-26358889

Autoantibodies are self-reactive antibodies that have been widely implicated as causal agents of autoimmune diseases. They are found in the blood of all human sera, regardless of age, gender, or the presence or absence of disease. While the underlying reason for their ubiquity remains unknown, it has been hypothesized that they participate in the clearance of blood-borne cell and tissue debris generated in both healthy and diseased individuals on a daily basis. Although much evidence supports this debris clearance role, recent studies also suggest a causal role for autoantibodies in disease. This chapter first presents well-known examples of autoimmune diseases that emphasize a direct causal role for autoantibodies and then discusses the veritable explosion of evidence now supporting their involvement in a wide variety of other diseases, including cancers and several types of neurological and neurodegenerative diseases. Lastly, translational strategies that take advantage of the "cause and/or effect" role of autoantibodies and recent technological advancements in their detection to exploit autoantibodies as sensitive and specific biomarkers useful for the detection and diagnosis of disease are outlined. Their use in the diagnosis and staging of Alzheimer's and Parkinson's diseases is presented, and future applications in clinical medicine and basic science are highlighted.


Autoantibodies/blood , Autoimmune Diseases/diagnosis , Neurodegenerative Diseases/diagnosis , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Biomarkers/blood , Humans , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/immunology , Severity of Illness Index
18.
Brain Res ; 1620: 29-41, 2015 Sep 16.
Article En | MEDLINE | ID: mdl-25960348

A large percentage of patients subjected to general anesthesia at 65 years and older exhibit postoperative delirium (POD). Here, we test the hypothesis that inhaled anesthetics (IAs), such as Sevoflurane and Isoflurane, act directly on brain vascular endothelial cells (BVECs) to increase blood-brain barrier (BBB) permeability, thereby contributing to POD. Rats of young (3-5 months), middle (10-12 months) and old (17-19 months) ages were anesthetized with Sevoflurane or Isoflurane for 3h. After exposure, some were euthanized immediately; others were allowed to recover for 24h before sacrifice. Immunohistochemistry was employed to monitor the extent of BBB breach, and scanning electron microscopy (SEM) was used to examine changes in the luminal surfaces of BVECs. Quantitative immunohistochemistry revealed increased BBB permeability in older animals treated with Sevoflurane, but not Isoflurane. Extravasated immunoglobulin G showed selective affinity for pyramidal neurons. SEM demonstrated marked flattening of the luminal surfaces of BVECs in anesthetic-treated rats. Results suggest an aging-linked BBB compromise resulting from exposure to Sevoflurane. Changes in the luminal surface topology of BVECs indicate a direct effect on the plasma membrane, which may weaken or disrupt their BBB-associated tight junctions. Disruption of brain homeostasis due to plasma influx into the brain parenchyma and binding of plasma components (e.g., immunoglobulins) to neurons may contribute to POD. We propose that, in the elderly, exposure to some IAs can cause BBB compromise that disrupts brain homeostasis, perturbs neuronal function and thereby contributes to POD. If unresolved, this may progress to postoperative cognitive decline and later dementia.


Anesthetics, Inhalation/toxicity , Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Isoflurane/toxicity , Methyl Ethers/toxicity , Aging/drug effects , Aging/metabolism , Aging/pathology , Animals , Blood-Brain Barrier/growth & development , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Capillary Permeability/physiology , Cognition Disorders/chemically induced , Cognition Disorders/etiology , Delirium/chemically induced , Delirium/etiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Immunoglobulin G/metabolism , Immunohistochemistry , Microscopy, Electron, Scanning , Postoperative Complications/chemically induced , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Rats, Sprague-Dawley , Sevoflurane , Time Factors
19.
Dev Cell ; 28(5): 561-572, 2014 Mar 10.
Article En | MEDLINE | ID: mdl-24582807

The control of germline quality is critical to reproductive success and survival of a species; however, the mechanisms underlying this process remain unknown. Here, we demonstrate that elongation factor 2 kinase (eEF2K), an evolutionarily conserved regulator of protein synthesis, functions to maintain germline quality and eliminate defective oocytes. We show that disruption of eEF2K in mice reduces ovarian apoptosis and results in the accumulation of aberrant follicles and defective oocytes at advanced reproductive age. Furthermore, the loss of eEF2K in Caenorhabditis elegans results in a reduction of germ cell death and significant decline in oocyte quality and embryonic viability. Examination of the mechanisms by which eEF2K regulates apoptosis shows that eEF2K senses oxidative stress and quickly downregulates short-lived antiapoptotic proteins, XIAP and c-FLIPL by inhibiting global protein synthesis. These results suggest that eEF2K-mediated inhibition of protein synthesis renders cells susceptible to apoptosis and functions to eliminate suboptimal germ cells.


Apoptosis , Caenorhabditis elegans/physiology , Elongation Factor 2 Kinase/physiology , Germ Cells/pathology , Oocytes/physiology , Quality Control , Animals , Blotting, Western , Caenorhabditis elegans/cytology , Caspases/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Immunoenzyme Techniques , In Situ Nick-End Labeling , Male , Mice , Mice, Knockout , NIH 3T3 Cells , Oocytes/cytology , Ovary/cytology , Ovary/physiology , Phosphorylation
...