Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 98(1): 163-186, 2024.
Article in English | MEDLINE | ID: mdl-38393907

ABSTRACT

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.


Subject(s)
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.
Front Hum Neurosci ; 16: 836980, 2022.
Article in English | MEDLINE | ID: mdl-35431844

ABSTRACT

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.

3.
J Alzheimers Dis ; 74(1): 345-361, 2020.
Article in English | MEDLINE | ID: mdl-32039847

ABSTRACT

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.


Subject(s)
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
4.
Diab Vasc Dis Res ; 14(3): 200-213, 2017 05.
Article in English | MEDLINE | ID: mdl-28301218

ABSTRACT

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.


Subject(s)
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
5.
Biomaterials ; 115: 30-39, 2017 01.
Article in English | MEDLINE | ID: mdl-27886553

ABSTRACT

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.


Subject(s)
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
6.
Alzheimers Dement (Amst) ; 3: 51-62, 2016.
Article in English | MEDLINE | ID: mdl-27239548

ABSTRACT

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.

7.
Immunol Lett ; 168(1): 80-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26386375

ABSTRACT

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.


Subject(s)
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
8.
Brain Res ; 1620: 29-41, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-25960348

ABSTRACT

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.


Subject(s)
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
9.
PLoS One ; 8(4): e60726, 2013.
Article in English | MEDLINE | ID: mdl-23589757

ABSTRACT

The presence of self-reactive IgG autoantibodies in human sera is largely thought to represent a breakdown in central tolerance and is typically regarded as a harbinger of autoimmune pathology. In the present study, immune-response profiling of human serum from 166 individuals via human protein microarrays demonstrates that IgG autoantibodies are abundant in all human serum, usually numbering in the thousands. These IgG autoantibodies bind to human antigens from organs and tissues all over the body and their serum diversity is strongly influenced by age, gender, and the presence of specific diseases. We also found that serum IgG autoantibody profiles are unique to an individual and remarkably stable over time. Similar profiles exist in rat and swine, suggesting conservation of this immunological feature among mammals. The number, diversity, and apparent evolutionary conservation of autoantibody profiles suggest that IgG autoantibodies have some important, as yet unrecognized, physiological function. We propose that IgG autoantibodies have evolved as an adaptive mechanism for debris-clearance, a function consistent with their apparent utility as diagnostic indicators of disease as already established for Alzheimer's and Parkinson's diseases.


Subject(s)
Autoantibodies/metabolism , Immunoglobulin G/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Animals , Antigens/immunology , Antigens/metabolism , Autoantibodies/immunology , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Female , Humans , Immunoglobulin G/immunology , Male , Middle Aged , Protein Array Analysis , Rats , Sex Factors , Swine , Ubiquitination
10.
J Alzheimers Dis ; 35(1): 179-98, 2013.
Article in English | MEDLINE | ID: mdl-23388174

ABSTRACT

Diabetes mellitus (DM) and hypercholesterolemia (HC) have emerged as major risk factors for Alzheimer's disease, highlighting the importance of vascular health to normal brain functioning. Our previous study showed that DM and HC favor the development of advanced coronary atherosclerosis in a porcine model, and that treatment with darapladib, an inhibitor of lipoprotein-associated phospholipase A2, blocks atherosclerosis progression and improves animal alertness and activity levels. In the present study, we examined the effects of DM and HC on the permeability of the blood-brain barrier (BBB) using immunoglobulin G (IgG) as a biomarker. DMHC increased BBB permeability and the leak of microvascular IgG into the brain interstitium, which was bound preferentially to pyramidal neurons in the cerebral cortex. We also examined the effects of DMHC on the brain deposition of amyloid peptide (Aß42), a well-known pathological feature of Alzheimer's disease. Nearly all detectable Aß42 was contained within cortical pyramidal neurons and DMHC increased the density of Aß42-loaded neurons. Treatment of DMHC animals with darapladib reduced the amount of IgG-immunopositive material that leaked into the brain as well as the density of Aß42-containing neurons. Overall, these results suggest that a prolonged state of DMHC may have chronic deleterious effects on the functional integrity of the BBB and that, in this DMHC pig model, darapladib reduces BBB permeability. Also, the preferential binding of IgG and coincident accumulation of Aß42 in the same neurons suggests a mechanistic link between the leak of IgG through the BBB and intraneuronal deposition of Aß42 in the brain.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Amyloid beta-Peptides/metabolism , Benzaldehydes/therapeutic use , Blood-Brain Barrier/metabolism , Diabetes Mellitus/metabolism , Hypercholesterolemia/metabolism , Oximes/therapeutic use , Peptide Fragments/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Animals , Benzaldehydes/pharmacology , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Hypercholesterolemia/drug therapy , Hypercholesterolemia/pathology , Oximes/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Swine , Treatment Outcome
11.
PLoS One ; 7(7): e41845, 2012.
Article in English | MEDLINE | ID: mdl-22844530

ABSTRACT

Cancer is a leading cause of death of men and women worldwide. Tumor cell motility contributes to metastatic invasion that causes the vast majority of cancer deaths. Extracellular receptors modified by α2,3-sialic acids that promote this motility can serve as ideal chemotherapeutic targets. For example, the extracellular domain of the mucin receptor podoplanin (PDPN) is highly O-glycosylated with α2,3-sialic acid linked to galactose. PDPN is activated by endogenous ligands to induce tumor cell motility and metastasis. Dietary lectins that target proteins containing α2,3-sialic acid inhibit tumor cell growth. However, anti-cancer lectins that have been examined thus far target receptors that have not been identified. We report here that a lectin from the seeds of Maackia amurensis (MASL) with affinity for O-linked carbohydrate chains containing sialic acid targets PDPN to inhibit transformed cell growth and motility at nanomolar concentrations. Interestingly, the biological activity of this lectin survives gastrointestinal proteolysis and enters the cardiovascular system to inhibit melanoma cell growth, migration, and tumorigenesis. These studies demonstrate how lectins may be used to help develop dietary agents that target specific receptors to combat malignant cell growth.


Subject(s)
Cell Movement/drug effects , Cell Transformation, Neoplastic , Membrane Glycoproteins/metabolism , N-Acetylneuraminic Acid/metabolism , Plant Lectins/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Maackia/chemistry , Melanoma/blood supply , Melanoma/diet therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Molecular Sequence Data , Necrosis/chemically induced , Neovascularization, Pathologic/diet therapy , Plant Lectins/chemistry , Plant Lectins/metabolism , src-Family Kinases/metabolism
12.
J Autoimmun ; 38(4): 369-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22560840

ABSTRACT

Peptidyl arginine deiminases (PADs) catalyze a post-translational protein modification reaction called citrullination, where arginine is converted to citrulline. This modification has been linked to the pathogenesis of autoimmune diseases including rheumatoid arthritis (RA). More recently, several studies have suggested that Alzheimer's disease (AD), a devastating neurodegenerative disorder, may have an autoimmune component. In the present study, we have investigated the possibility that expression of PADs and protein citrullination plays a role in the production of brain-reactive autoantibodies that may contribute to Alzheimer's-related brain pathology. Here, we report the selective expression of the PAD isoforms, PAD2 and PAD4, in astrocytes and neurons, respectively, and the concomitant accumulation of citrullinated proteins within PAD4-expressing cells, including neurons of the hippocampus and cerebral cortex. Expression of PADs and citrullinated proteins is prominent in brain regions engaged in neurodegenerative changes typical for AD pathology. Furthermore, we also demonstrate that the pentatricopeptide repeat domain2 (PTCD2) protein, an antigen target of a prominent AD diagnostic autoantibody, is present in a citrullinated form in AD brains. Our results suggest that disease-associated neuronal loss results in the release of cellular contents, including citrullinated proteins, into the brain interstitium. We propose that these citrullinated proteins and their degradation fragments enter into the blood and lymphatic circulation, and some are capable of eliciting an immune response that results in the production of autoantibodies. The long-term and progressive nature of AD and other neurodegenerative diseases results in chronic exposure of the immune system to these citrullinated products and may drive the continual production of autoantibodies.


Subject(s)
Hydrolases/metabolism , Neurodegenerative Diseases/enzymology , Neurons/enzymology , Protein Processing, Post-Translational , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Astrocytes/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , Brain/metabolism , Brain/pathology , Citrulline/metabolism , Humans , Hydrolases/genetics , Isoenzymes/metabolism , Middle Aged , Mitochondrial Proteins/immunology , Mitochondrial Proteins/metabolism , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Protein Transport , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases
13.
PLoS One ; 6(9): e25019, 2011.
Article in English | MEDLINE | ID: mdl-21980374

ABSTRACT

Prenatal cocaine exposure causes sustained phosphorylation of the synaptic anchoring protein, glutamate receptor interacting protein (GRIP1/2), preventing synaptic targeting of the GluR2/3-containing alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs; J. Neurosci. 29: 6308-6319, 2009). Because overexpression of GRIP-associated neuronal rasGEF protein (GRASP-1) specifically reduces the synaptic targeting of AMPARs, we hypothesized that prenatal cocaine exposure enhances GRASP-1 synaptic membrane localization leading to hyper-activation of ras family proteins and heightened actin polymerization. Our results show a markedly increased GRIP1-associated GRASP-1 content with approximately 40% reduction in its rasGEF activity in frontal cortices (FCX) of 21-day-old (P21) prenatal cocaine-exposed rats. This cocaine effect is the result of a persistent protein kinase C (PKC)- and downstream Src tyrosine kinase-mediated GRIP phosphorylation. The hyperactivated PKC also increased membrane-associated GRASP-1 and activated small G-proteins RhoA, cdc42/Rac1 and Rap1 as well as filamentous actin (F-actin) levels without an effect on the phosphorylation state of actin. Since increased F-actin facilitates protein transport, our results suggest that increased GRASP-1 synaptic localization in prenatal cocaine-exposed brains is an adaptive response to restoring the synaptic expression of AMPA-GluR2/3. Our earlier data demonstrated that persistent PKC-mediated GRIP phosphorylation reduces GluR2/3 synaptic targeting in prenatal cocaine-exposed brains, we now show that the increased GRIP-associated GRASP-1 may contribute to the reduction in GluR2/3 synaptic expression and AMPAR signaling defects.


Subject(s)
Carrier Proteins/metabolism , Cocaine/toxicity , Maternal Exposure/adverse effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , Actins/metabolism , Animals , Brain/metabolism , Female , Immunohistochemistry , Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Male , Organ Culture Techniques , Phosphorylation , Pregnancy , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, AMPA , Synaptosomes/metabolism , rac GTP-Binding Proteins
14.
J Alzheimers Dis ; 25(4): 605-22, 2011.
Article in English | MEDLINE | ID: mdl-21483091

ABSTRACT

Previous studies have reported immunoglobulin-positive neurons in Alzheimer's disease (AD) brains, an observation indicative of blood-brain barrier (BBB) breakdown. Recently, we demonstrated the nearly ubiquitous presence of brain-reactive autoantibodies in human sera. The significance of these observations to AD pathology is unknown. Here, we show that IgG-immunopositive neurons are abundant in brain regions exhibiting AD pathology, including intraneuronal amyloid-ß(42) (Aß(42)) and amyloid plaques, and confirm by western analysis that brain-reactive autoantibodies are nearly ubiquitous in human serum. To investigate a possible interrelationship between neuronal antibody binding and Aß pathology, we tested the effects of human serum autoantibodies on the intraneuronal deposition of soluble Aß(42) peptide in adult mouse neurons in vitro (organotypic brain slice cultures). Binding of human autoantibodies to mouse neurons dramatically increased the rate and extent of intraneuronal Aß(42) accumulation in the mouse cerebral cortex and hippocampus. Additionally, individual sera exhibited variable potency related to their capacity to enhance intraneuronal Aß(42) peptide accumulation and immunolabel neurons in AD brain sections. Replacement of human sera with antibodies targeting abundant neuronal surface proteins resulted in a comparable enhancement of Aß(42) accumulation in mouse neurons. Overall, results suggest that brain-reactive autoantibodies are ubiquitous in the blood and that a defective BBB allows these antibodies to access the brain interstitium, bind to neuronal surfaces and enhance intraneuronal deposition of Aß(42) in AD brains. Thus, in the context of BBB compromise, brain-reactive autoantibodies may be an important risk factor for the initiation and/or progression of AD as well as other neurodegenerative diseases.


Subject(s)
Amyloid beta-Peptides/metabolism , Autoantibodies/blood , Brain/immunology , Neurons/metabolism , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Animals , Blood-Brain Barrier , Blotting, Western , Female , Humans , Image Processing, Computer-Assisted , Immunoglobulin G/metabolism , Immunohistochemistry , Male , Mice , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Pyramidal Cells/metabolism , Rats , Rats, Sprague-Dawley
15.
J Neurosci ; 29(19): 6308-19, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19439608

ABSTRACT

Prenatal cocaine exposure produces sustained neurobehavioral and brain synaptic changes closely resembling those of animals with defective AMPA receptors (AMPARs). We hypothesized that prenatal cocaine exposure attenuates AMPAR signaling by interfering with AMPAR synaptic targeting. AMPAR function is governed by receptor cycling on and off the synaptic membrane through its interaction with glutamate receptor-interacting protein (GRIP), a PDZ domain protein that is regulated by reversible phosphorylation. Our results show that prenatal cocaine exposure markedly reduces AMPAR synaptic targeting and attenuates AMPAR-mediated synaptic long-term depression in the frontal cortex of 21-d-old rats. This cocaine effect is the result of reduced GRIP-AMPAR interaction caused by persistent phosphorylation of GRIP by protein kinase C (PKC) and Src tyrosine kinase. These data support the restoration of AMPAR activation via suppressing excessive PKC-mediated GRIP phosphorylation as a novel therapeutic approach to treat the neurobehavioral consequences of prenatal cocaine.


Subject(s)
Carrier Proteins/metabolism , Central Nervous System Agents/toxicity , Cocaine/toxicity , Nerve Tissue Proteins/metabolism , Prenatal Exposure Delayed Effects , Receptors, AMPA/metabolism , Synapses/drug effects , Animals , Female , Frontal Lobe/drug effects , Frontal Lobe/growth & development , Gene Expression/drug effects , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Phosphorylation/drug effects , Pregnancy , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Synapses/metabolism , src-Family Kinases/metabolism
16.
Brain Res ; 1234: 158-71, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18708033

ABSTRACT

Deposition of beta-amyloid (Abeta) peptides in the walls of brain blood vessels, cerebral amyloid angiopathy (CAA), is common in patients with Alzheimer's disease (AD). Previous studies have demonstrated Abeta peptide deposition among vascular smooth muscle cells (VSMCs), but the source of the Abeta and basis for its selective deposition in VSMCs are unknown. In the present study, we examined the deposition patterns of Abeta peptides, Abeta40 and Abeta42, within the cerebrovasculature of AD and control patients using single- and double-label immunohistochemistry. Abeta40 and Abeta42 were abundant in VSMCs, especially in leptomeningeal arteries and their initial cortical branches; in later-stage AD brains this pattern extended into the microvasculature. Abeta peptide deposition was linked to loss of VSMC viability. Perivascular leak clouds of Abeta-positive material were associated primarily with arterioles. By contrast, control brains possessed far fewer Abeta42- and Abeta40-immunopositive blood vessels, with perivascular leak clouds of Abeta-immunopositive material rarely observed. We also demonstrate that VSMCs in brain blood vessels express the alpha7 nicotinic acetylcholine receptor (alpha7nAChR), which has high binding affinity for Abeta peptides, especially Abeta42. These results suggest that the blood and blood-brain barrier permeability provide a major source of the Abeta peptides that gradually deposit in brain VSMCs, and the presence and abundance of the alpha7nAChR on VSMCs may facilitate the selective accumulation of Abeta peptides in these cells.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Nicotinic/biosynthesis , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Arterioles/metabolism , Arterioles/pathology , Blood-Brain Barrier/physiology , Cell Survival , Cerebral Amyloid Angiopathy/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Entorhinal Cortex/pathology , Female , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , alpha7 Nicotinic Acetylcholine Receptor
17.
Brain Res ; 1142: 223-36, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17306234

ABSTRACT

We have investigated the possibility that soluble, blood-borne amyloid beta (Abeta) peptides can cross a defective blood-brain barrier (BBB) and interact with neurons in the brain. Immunohistochemical analyses revealed extravasated plasma components, including Abeta42 in 19 of 21 AD brains, but in only 3 of 13 age-matched control brains, suggesting that a defective BBB is common in AD. To more directly test whether blood-borne Abeta peptides can cross a defective BBB, we tracked the fate of fluorescein isothiocyanate (FITC)-labeled Abeta42 and Abeta40 introduced via tail vein injection into mice with a BBB rendered permeable by treatment with pertussis toxin. Both Abeta40 and Abeta42 readily crossed the permeabilized BBB and bound selectively to certain neuronal subtypes, but not glial cells. By 48 h post-injection, Abeta42-positive neurons were widespread in the brain. In the cerebral cortex, small fluorescent, Abeta42-positive granules were found in the perinuclear cytoplasm of pyramidal neurons, suggesting that these cells can internalize exogenous Abeta42. An intact BBB (saline-injected controls) blocked entry of blood-borne Abeta peptides into the brain. The neuronal subtype selectivity of Abeta42 and Abeta40 was most evident in mouse brains subjected to direct intracranial stereotaxic injection into the hippocampal region, thereby bypassing the BBB. Abeta40 was found to preferentially bind to a distinct subset of neurons positioned at the inner face of the dentate gyrus, whereas Abeta42 bound selectively to the population of large neurons in the hilus region of the dentate gyrus. Our results suggest that the blood may serve as a major, chronic source of soluble, exogenous Abeta peptides that can bind selectively to certain subtypes of neurons and accumulate within these cells.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/physiopathology , Brain/pathology , Capillary Permeability/physiology , Neurons/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Case-Control Studies , Collagen Type IV/metabolism , Fluorescent Antibody Technique/methods , Humans , Indoles , Mice , Pertussis Toxin/pharmacology , Postmortem Changes , Protein Transport/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...