Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Alzheimers Dement ; 17(7): 1109-1120, 2021 07.
Article in English | MEDLINE | ID: mdl-33656270

ABSTRACT

We compared the efficacy of neurovascular coupling and substrate supply in cerebral cortex during severe metabolic challenges in transgenic Alzheimer's [CVN-AD] and control [C57Bl/6] mice, to evaluate the hypothesis that metabolic insufficiency is a critical component of degeneration leading to dementia. We analyzed cerebral blood flow and metabolic responses to spreading depression (induced by K+ applied to the cortex) and anoxia across aging in CVN-AD + C57Bl/6 genotypes. In the CVN-AD genotype progression to histological and cognitive hallmarks of dementia is a stereotyped function of age. We correlated physiology and imaging of the cortex with the blood flow responses measured with laser doppler probes. The results show that spreading depression resulted in a hyperemic blood flow response that was dramatically reduced (24% in amplitude, 70% in area) in both middle-aged and aged CVN-AD mice compared to C57Bl/6 age-matched controls. However, spreading depression amplitude and conduction velocity (≈6 mm/min) did not differ among groups. Anoxia (100% N2 ) showed significantly decreased (by 62%) reactive blood flow and autoregulation in aged AD-CVN mice compared to aged control animals. Significantly reduced neurovascular coupling occurred prematurely with aging in CVN-AD mice. Abbreviated physiological hyperemia and decreased resilience to anoxia may enhance early-onset metabolic deficiency through decreased substrate supply to the brain. Metabolic deficiency may contribute significantly to the degeneration associated with dementia as a function of aging and regions of the brain involved.


Subject(s)
Cerebrovascular Circulation/physiology , Depression/physiopathology , Disease Models, Animal , Hypoxia/physiopathology , Mice, Inbred C57BL , Neurovascular Coupling , Aging , Alzheimer Disease/pathology , Animals , Female , Hemodynamics/physiology , Humans , Male , Mice
2.
Alzheimers Dement ; 17(4): 561-573, 2021 04.
Article in English | MEDLINE | ID: mdl-33480182

ABSTRACT

INTRODUCTION: The study of Alzheimer's disease (AD) has revealed biological pathways with implications for disease neuropathology and pathophysiology. These pathway-level effects may also be mediated by individual characteristics or covariates such as age or sex. Evaluation of AD biological pathways in the context of interactions with these covariates is critical to the understanding of AD as well as the development of model systems used to study the disease. METHODS: Gene set enrichment methods are powerful tools used to interpret gene-level statistics at the level of biological pathways. We introduce a method for quantifying gene set enrichment using likelihood ratio-derived test statistics (gsLRT), which accounts for sample covariates like age and sex. We then use our method to test for age and sex interactions with protein expression levels in AD and to compare the pathway results between human and mouse species. RESULTS: Our method, based on nested logistic regressions is competitive with the existing standard for gene set testing in the context of linear models and complex experimental design. The gene sets we identify as having a significant association with AD-both with and without additional covariate interactions-are validated by previous studies. Differences between gsLRT results on mouse and human datasets are observed. DISCUSSION: Characterizing biological pathways involved in AD builds on the important work involving single gene drivers. Our gene set enrichment method finds pathways that are significantly related to AD while accounting for covariates that may be relevant to disease development. The method highlights commonalities and differences between human AD and mouse models, which may inform the development of higher fidelity models for the study of AD.


Subject(s)
Alzheimer Disease/pathology , Disease Models, Animal , Gene Expression Regulation , Models, Statistical , Age Factors , Animals , Humans , Mice , Sex Factors
3.
J Proteome Res ; 19(4): 1447-1458, 2020 04 03.
Article in English | MEDLINE | ID: mdl-31984744

ABSTRACT

Vendor-independent software tools for quantification of small molecules and metabolites are lacking, especially for targeted analysis workflows. Skyline is a freely available, open-source software tool for targeted quantitative mass spectrometry method development and data processing with a 10 year history supporting six major instrument vendors. Designed initially for proteomics analysis, we describe the expansion of Skyline to data for small molecule analysis, including selected reaction monitoring, high-resolution mass spectrometry, and calibrated quantification. This fundamental expansion of Skyline from a peptide-sequence-centric tool to a molecule-centric tool makes it agnostic to the source of the molecule while retaining Skyline features critical for workflows in both peptide and more general biomolecular research. The data visualization and interrogation features already available in Skyline, such as peak picking, chromatographic alignment, and transition selection, have been adapted to support small molecule data, including metabolomics. Herein, we explain the conceptual workflow for small molecule analysis using Skyline, demonstrate Skyline performance benchmarked against a comparable instrument vendor software tool, and present additional real-world applications. Further, we include step-by-step instructions on using Skyline for small molecule quantitative method development and data analysis on data acquired with a variety of mass spectrometers from multiple instrument vendors.


Subject(s)
Metabolomics , Proteomics , Amino Acid Sequence , Mass Spectrometry , Software
4.
Alzheimers Dement ; 16(5): 734-749, 2020 05.
Article in English | MEDLINE | ID: mdl-32291962

ABSTRACT

OBJECTIVE: The present work evaluates the relationship between postoperative immune and neurovascular changes and the pathogenesis of surgery-induced delirium superimposed on dementia. BACKGROUND AND RATIONALE: Postoperative delirium is a common complication in many older adults and in patients with dementia including Alzheimer's disease (AD). The course of delirium can be particularly debilitating, while its pathophysiology remains poorly defined. HISTORICAL EVOLUTION: As of 2019, an estimated 5.8 million people of all ages have been diagnosed with AD, 97% of whom are >65 years of age. Each year, many of these patients require surgery. However, anesthesia and surgery can increase the risk for further cognitive decline. Surgery triggers neuroinflammation both in animal models and in humans, and a failure to resolve this inflammatory state may contribute to perioperative neurocognitive disorders as well as neurodegenerative pathology. UPDATED HYPOTHESIS: We propose an immunovascular hypothesis whereby dysregulated innate immunity negatively affects the blood-brain interface, which triggers delirium and thereby exacerbates AD neuropathology. EARLY EXPERIMENTAL DATA: We have developed a translational model to study delirium superimposed on dementia in APPSwDI/mNos2-/- AD mice (CVN-AD) after orthopedic surgery. At 12 months of age, CVN-AD showed distinct neuroimmune and vascular impairments after surgery, including acute microgliosis and amyloid-ß deposition. These changes correlated with attention deficits, a core feature of delirium-like behavior. FUTURE EXPERIMENTS AND VALIDATION STUDIES: Future research should determine the extent to which prevention of surgery-induced microgliosis and/or neurovascular unit dysfunction can prevent or ameliorate postoperative memory and attention deficits in animal models. Translational human studies should evaluate perioperative indices of innate immunity and neurovascular integrity and assess their potential link to perioperative neurocognitive disorders. MAJOR CHALLENGES FOR THE HYPOTHESIS: Understanding the complex relationships between delirium and dementia will require mechanistic studies aimed at evaluating the role of postoperative neuroinflammation and blood-brain barrier changes in the setting of pre-existing neurodegenerative and/or aging-related pathology. LINKAGE TO OTHER MAJOR THEORIES: Non-resolving inflammation with vascular disease that leads to cognitive impairments and dementia is increasingly important in risk stratification for AD in the aging population. The interdependence of these factors with surgery-induced neuroinflammation and cognitive dysfunction is also becoming apparent, providing a strong platform for assessing the relationship between postoperative delirium and longer term cognitive dysfunction in older adults.


Subject(s)
Delirium/physiopathology , Dementia/complications , Inflammation , Postoperative Complications , Animals , Blood-Brain Barrier , Brain/pathology , Cognition Disorders/etiology , Disease Models, Animal , Humans , Mice , Neurocognitive Disorders
5.
J Stroke Cerebrovasc Dis ; 29(4): 104646, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32067855

ABSTRACT

BACKGROUND: African-Americans (AA) are 3 times more likely to have small-vessel-type ischemic strokes (SVS) than Whites. Small vessel strokes are associated with cognitive impairment, a relationship incompletely explained by white matter hyperintensity (WMH) burden. We examined whether inflammatory/endothelial dysfunction biomarkers are associated with cognition after SVS in AAs. METHODS: Biomarkers were obtained in 24 subjects (median age 56.5 years, 54% women, median 12 years education). Cognition was assessed more than 6 weeks poststroke using the memory composite score (MCS), which was generated using recall from the Hopkins Verbal Learning Test-II and Brief Visuospatial Memory Test-Revised. A semi-automated, volumetric protocol was used to quantify WMH volume (WMHv) on clinical MRI scans. Potential biomarkers including vascular cell adhesion molecule-1 (VCAM-1), interleukin-1 receptor antagonist, interleukin-6, interleukin-8, interleukin-10, interferon gamma, and thrombin-antithrombin (TAT) were log-transformed and correlated with MCS with adjustment for potential confounders. RESULTS: Among serum biomarkers, only VCAM-1-correlated with poorer memory based on the MCS (r = -.659; P = .0006). VCAM-1 (r = .554; P = .005) and age (r = .479; P = .018) correlated with WMHv; VCAM-1 was independently associated with MCS after adjustment for WMHv, age, and education (P = .023). CONCLUSIONS: The findings of this exploratory analysis suggest that endothelial dysfunction and inflammation as reflected by VCAM-1 levels may play a role in poststroke cognitive impairment. Additional studies are needed to validate this observation and to evaluate this relationship in non-AAs and with other stroke types and compare this finding to cognitive impairment in nonstroke populations.


Subject(s)
Black or African American/psychology , Cerebral Small Vessel Diseases/blood , Memory Disorders/blood , Memory , Stroke/blood , Vascular Cell Adhesion Molecule-1/blood , Biomarkers/blood , Cerebral Small Vessel Diseases/diagnosis , Cerebral Small Vessel Diseases/ethnology , Cerebral Small Vessel Diseases/psychology , Female , Humans , Male , Memory Disorders/diagnosis , Memory Disorders/ethnology , Memory Disorders/psychology , Middle Aged , Neuropsychological Tests , Risk Factors , Stroke/diagnosis , Stroke/ethnology , Stroke/psychology , United States/epidemiology
6.
J Neurosci ; 35(15): 5969-82, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878270

ABSTRACT

The pathogenesis of Alzheimer's disease (AD) is a critical unsolved question; and although recent studies have demonstrated a strong association between altered brain immune responses and disease progression, the mechanistic cause of neuronal dysfunction and death is unknown. We have previously described the unique CVN-AD mouse model of AD, in which immune-mediated nitric oxide is lowered to mimic human levels, resulting in a mouse model that demonstrates the cardinal features of AD, including amyloid deposition, hyperphosphorylated and aggregated tau, behavioral changes, and age-dependent hippocampal neuronal loss. Using this mouse model, we studied longitudinal changes in brain immunity in relation to neuronal loss and, contrary to the predominant view that AD pathology is driven by proinflammatory factors, we find that the pathology in CVN-AD mice is driven by local immune suppression. Areas of hippocampal neuronal death are associated with the presence of immunosuppressive CD11c(+) microglia and extracellular arginase, resulting in arginine catabolism and reduced levels of total brain arginine. Pharmacologic disruption of the arginine utilization pathway by an inhibitor of arginase and ornithine decarboxylase protected the mice from AD-like pathology and significantly decreased CD11c expression. Our findings strongly implicate local immune-mediated amino acid catabolism as a novel and potentially critical mechanism mediating the age-dependent and regional loss of neurons in humans with AD.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Arginine/metabolism , Brain/metabolism , Immunologic Factors/metabolism , Age Factors , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Antigens, CD/metabolism , Disease Models, Animal , Disease Progression , Eflornithine/pharmacology , Eflornithine/therapeutic use , Humans , Immunologic Factors/genetics , Maze Learning/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Microglia/metabolism , Mutation/genetics , Nitric Oxide Synthase Type II/genetics , Ornithine Decarboxylase Inhibitors/pharmacology , Ornithine Decarboxylase Inhibitors/therapeutic use
7.
Neuroimage ; 142: 498-511, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27521741

ABSTRACT

Multivariate biomarkers are needed for detecting Alzheimer's disease (AD), understanding its etiology, and quantifying the effect of therapies. Mouse models provide opportunities to study characteristics of AD in well-controlled environments that can help facilitate development of early interventions. The CVN-AD mouse model replicates multiple AD hallmark pathologies, and we identified multivariate biomarkers characterizing a brain circuit disruption predictive of cognitive decline. In vivo and ex vivo magnetic resonance imaging (MRI) revealed that CVN-AD mice replicate the hippocampal atrophy (6%), characteristic of humans with AD, and also present changes in subcortical areas. The largest effect was in the fornix (23% smaller), which connects the septum, hippocampus, and hypothalamus. In characterizing the fornix with diffusion tensor imaging, fractional anisotropy was most sensitive (20% reduction), followed by radial (15%) and axial diffusivity (2%), in detecting pathological changes. These findings were strengthened by optical microscopy and ultrastructural analyses. Ultrastructual analysis provided estimates of axonal density, diameters, and myelination-through the g-ratio, defined as the ratio between the axonal diameter, and the diameter of the axon plus the myelin sheath. The fornix had reduced axonal density (47% fewer), axonal degeneration (13% larger axons), and abnormal myelination (1.5% smaller g-ratios). CD68 staining showed that white matter pathology could be secondary to neuronal degeneration, or due to direct microglial attack. In conclusion, these findings strengthen the hypothesis that the fornix plays a role in AD, and can be used as a disease biomarker and as a target for therapy.


Subject(s)
Alzheimer Disease/pathology , Diffusion Tensor Imaging/methods , Fornix, Brain/pathology , Hippocampus/pathology , Microscopy, Electron/methods , White Matter/pathology , Alzheimer Disease/diagnostic imaging , Animals , Atrophy/pathology , Biomarkers , Disease Models, Animal , Fornix, Brain/diagnostic imaging , Hippocampus/diagnostic imaging , Mice , Mice, Transgenic , White Matter/diagnostic imaging
8.
Exp Eye Res ; 145: 17-25, 2016 04.
Article in English | MEDLINE | ID: mdl-26554939

ABSTRACT

Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor for Alzheimer's disease (AD), is associated with neuronal and vascular impairments. The retina, which is as an extension of the central nervous system (CNS), is a particularly suitable model for studying developmental and functional aspects of the neuronal and vascular systems. This study investigates the apoE4-dependent developmental effects on the retinal vasculature and neuronal systems and on the levels of apoE and the vascular endothelial growth factor (VEGF) in the retina. This was performed utilizing retinas of 4, 7, 12, and of 120-day-old human-apoE4-targeted replacement mice and of corresponding mice that express the AD benign isoform, apoE3. The results obtained revealed retinal vascular pathology in the apoE4 mice, which started on the early post-natal days. This includes transient increase in vascular branching, and vascular buds which are round vascular elements representing sprouting or retracting vessels. These effects peaked and ended during the neonatal period. Examination of the synaptic system utilizing the pre-synaptic marker synaptophysin revealed a significant decrease of retinal synaptic density in the apoE4 mice, which was detectable by post-natal day 12 (P12). These morphological changes are associated with neonatal age-dependent elevation in the apoE levels in both apoE3 and apoE4 retinas which is more profound in the apoE4 mice and a corresponding increase in VEGF levels, which is less profound in the apoE4 mice. Additionally, we observed lower levels of retinal VEGF in the apoE4 mice compared to the apoE3 mice retinas on P12. These results show that apoE4 has a transient vascular effect during retinal development that ends in the neonatal period, which is accompanied by a synaptic effect that begins at the end of the neonatal period. These findings show that the apoE4 genotype can have distinct developmental effects on both the retinal vasculature and on neurons and suggest that the vascular effects of apoE4 may be related to reduced levels of VEGF.


Subject(s)
Apolipoprotein E4/genetics , Retina/growth & development , Retinal Vessels/growth & development , Animals , Animals, Newborn , Apolipoprotein E4/metabolism , Blotting, Western , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Models, Animal , Retina/cytology , Retina/metabolism , Retinal Vessels/cytology , Retinal Vessels/metabolism
9.
J Proteome Res ; 12(10): 4462-77, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24006891

ABSTRACT

Alzheimer's disease (AD) is a complex neurodegenerative process that involves altered brain immune, neuronal and metabolic functions. Understanding the underlying mechanisms has relied on mouse models that mimic components of AD pathology. We used gel-free, label-free LC-MS/MS to quantify protein and phosphopeptide levels in brains of APPSwDI/NOS2-/- (CVN-AD) mice. CVN-AD mice show a full spectrum of AD-like pathology, including amyloid deposition, hyperphosphorylated and aggregated tau, and neuronal loss that worsens with age. Tryptic digests, with or without phosphopeptide enrichment on an automated titanium dioxide LC system, were separated by online two-dimensional LC and analyzed on a Waters Synapt G2 HDMS, yielding relative expression for >950 proteins and >1100 phosphopeptides. Among differentially expressed proteins were known markers found in humans with AD, including GFAP and C1Q. Phosphorylation of connexin 43, not previously described in AD, was increased at 42 weeks, consistent with dysregulation of gap junctions and activation of astrocytes. Additional alterations in phosphoproteins suggests dysregulation of mitochondria, synaptic transmission, vesicle trafficking, and innate immune pathways. These data validate the CVN-AD mouse model of AD, identify novel disease and age-related changes in the brain during disease progression, and demonstrate the utility of integrating unbiased and phosphoproteomics for understanding disease processes in AD.


Subject(s)
Alzheimer Disease/metabolism , Nitric Oxide Synthase Type II/genetics , Phosphopeptides/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Chromatography, Ion Exchange , Chromatography, Reverse-Phase , Humans , Longitudinal Studies , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nitric Oxide Synthase Type II/deficiency , Phosphopeptides/isolation & purification , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , Proteome/isolation & purification , Proteome/metabolism , Receptors, Complement/metabolism , Tandem Mass Spectrometry , tau Proteins/metabolism
10.
J Immunol ; 186(4): 2535-42, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21289314

ABSTRACT

The molecular mechanism by which apolipoprotein E (apoE) suppresses inflammatory cytokine and NO production is unknown. Using an affinity purification approach, we found that peptide mimetics of apoE, derived from its receptor binding domain residues 130-150, bound to the SET protein, which is a potent physiological inhibitor of protein phosphatase 2A (PP2A). Both holo-apoE protein and apoE-mimetic peptides bound to the C-terminal region of SET, which is then associated with an increase in PP2A-mediated phosphatase activity. As physiological substrates for PP2A, the LPS-induced phosphorylation status of signaling MAPK and Akt kinase is reduced following treatment with apoE-mimetic peptides. On the basis of our previous report, in which apoE-mimetic peptides reduced I-κB kinase and NF-κB activation, we also demonstrate a mechanism for reduced production of inducible NO synthase protein and its NO product. These data provide evidence for a novel molecular mechanism by which apoE and apoE-mimetic peptides antagonize SET, thereby enhancing endogenous PP2A phosphatase activity, which reduces levels of phosphorylated kinases, signaling, and inflammatory response.


Subject(s)
Apolipoproteins E/physiology , Histone Chaperones/metabolism , Inflammation Mediators/physiology , Molecular Mimicry/immunology , Oncogene Proteins/metabolism , Peptide Fragments/physiology , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Apolipoproteins E/metabolism , Cell Line , DNA-Binding Proteins , Down-Regulation/immunology , Enzyme Activation/immunology , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/physiology , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/physiology , Peptide Fragments/metabolism , Protein Binding/immunology , Protein Phosphatase 2/physiology , Signal Transduction/immunology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology , Up-Regulation/immunology
11.
Article in English | MEDLINE | ID: mdl-37650081

ABSTRACT

The principles governing genotype-phenotype relationships are still emerging(1-3), and detailed translational as well as transcriptomic information is required to understand complex phenotypes, such as the pathogenesis of Alzheimer's disease. For this reason, the proteomics of Alzheimer disease (AD) continues to be studied extensively. Although comparisons between data obtained from humans and mouse models have been reported, approaches that specifically address the between-species statistical comparisons are understudied. Our study investigated the performance of two statistical methods for identification of proteins and biological pathways associated with Alzheimer's disease for cross-species comparisons, taking specific data analysis challenges into account, including collinearity, dimensionality reduction and cross-species protein matching. We used a human dataset from a well-characterized cohort followed for over 22 years with proteomic data available. For the mouse model, we generated proteomic data from whole brains of CVN-AD and matching control mouse models. We used these analyses to determine the reliability of a mouse model to forecast significant proteomic-based pathological changes in the brain that may mimic pathology in human Alzheimer's disease. Compared with LASSO regression, partial least squares discriminant analysis provided better statistical performance for the proteomics analysis. The major biological finding of the study was that extracellular matrix proteins and integrin-related pathways were dysregulated in both the human and mouse data. This approach may help inform the development of mouse models that are more relevant to the study of human late-onset Alzheimer's disease.

12.
J Neurochem ; 123(5): 736-49, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23016931

ABSTRACT

Fibrillar amyloid plaques are largely composed of amyloid-beta (Aß) peptides that are metabolized into products, including Aß1-16, by proteases including matrix metalloproteinase 9 (MMP-9). The balance between production and degradation of Aß proteins is critical to amyloid accumulation and resulting disease. Regulation of MMP-9 and its endogenous inhibitor tissue inhibitor of metalloproteinase (TIMP)-1 by nitric oxide (NO) has been shown. We hypothesize that nitric oxide synthase (NOS2) protects against Alzheimer's disease pathology by increasing amyloid clearance through NO regulation of MMP-9/TIMP-1 balance. We show NO-mediated increased MMP-9/TIMP-1 ratios enhanced the degradation of fibrillar Aß in vitro, which was abolished when silenced for MMP-9 protein translation. The in vivo relationship between MMP-9, NO and Aß degradation was examined by comparing an Alzheimer's disease mouse model that expresses NOS2 with a model lacking NOS2. To quantitate MMP-9 mediated changes, we generated an antibody recognizing the Aß1-16 fragment, and used mass spectrometry multi-reaction monitoring assay for detection of immunoprecipitated Aß1-16 peptides. Aß1-16 levels decreased in brain lysates lacking NOS2 when compared with strains that express human amyloid precursor protein on the NOS2 background. TIMP-1 increased in the APPSwDI/NOS2(-/-) mice with decreased MMP activity and increased amyloid burden, thereby supporting roles for NO in the regulation of MMP/TIMP balance and plaque clearance.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Matrix Metalloproteinase 9/metabolism , Nitric Oxide/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Chromatography, Liquid , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoprecipitation , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
13.
Front Neurosci ; 16: 848654, 2022.
Article in English | MEDLINE | ID: mdl-35784847

ABSTRACT

Spatial navigation and orientation are emerging as promising markers for altered cognition in prodromal Alzheimer's disease, and even in cognitively normal individuals at risk for Alzheimer's disease. The different APOE gene alleles confer various degrees of risk. The APOE2 allele is considered protective, APOE3 is seen as control, while APOE4 carriage is the major known genetic risk for Alzheimer's disease. We have used mouse models carrying the three humanized APOE alleles and tested them in a spatial memory task in the Morris water maze. We introduce a new metric, the absolute winding number, to characterize the spatial search strategy, through the shape of the swim path. We show that this metric is robust to noise, and works for small group samples. Moreover, the absolute winding number better differentiated APOE3 carriers, through their straighter swim paths relative to both APOE2 and APOE4 genotypes. Finally, this novel metric supported increased vulnerability in APOE4 females. We hypothesized differences in spatial memory and navigation strategies are linked to differences in brain networks, and showed that different genotypes have different reliance on the hippocampal and caudate putamen circuits, pointing to a role for white matter connections. Moreover, differences were most pronounced in females. This departure from a hippocampal centric to a brain network approach may open avenues for identifying regions linked to increased risk for Alzheimer's disease, before overt disease manifestation. Further exploration of novel biomarkers based on spatial navigation strategies may enlarge the windows of opportunity for interventions. The proposed framework will be significant in dissecting vulnerable circuits associated with cognitive changes in prodromal Alzheimer's disease.

15.
J Neuroinflammation ; 8: 115, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21906275

ABSTRACT

BACKGROUND: Anti-Aß immunotherapy is a promising approach to the prevention and treatment of Alzheimer's disease (AD) currently in clinical trials. There is extensive evidence, both in mice and humans that a significant adverse event is the occurrence of microhemorrhages. Also, vasogenic edema was reported in phase 2 of a passive immunization clinical trial. In order to overcome these vascular adverse effects it is critical that we understand the mechanism(s) by which they occur. METHODS: We have examined the matrix metalloproteinase (MMP) protein degradation system in two previously published anti-Aß immunotherapy studies. The first was a passive immunization study in which we examined 22 month old APPSw mice that had received anti-Aß antibodies for 1, 2 or 3 months. The second is an active vaccination study in which we examined 16 month old APPSw/NOS2-/- mice treated with Aß vaccination for 4 months. RESULTS: There is a significant activation of the MMP2 and MMP9 proteinase degradation systems by anti-Aß immunotherapy, regardless of whether this is delivered through active vaccination or passive immunization. We have characterized this activation by gene expression, protein expression and zymography assessment of MMP activity. CONCLUSIONS: Since the MMP2 and MMP9 systems are heavily implicated in the pathophysiology of intracerbral hemorrhage, these data may provide a potential mechanism of microhemorrhage due to immunotherapy. Increased activity of the MMP system, therefore, is likely to be a major factor in increased microhemorrhage occurrence.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/therapeutic use , Cerebral Hemorrhage , Immunotherapy/methods , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/adverse effects , Amyloid beta-Peptides/immunology , Animals , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/immunology , Cerebrovascular Circulation , Disease Models, Animal , Enzyme Activation , Humans , Mice , Mice, Transgenic , Microcirculation , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism
16.
ASN Neuro ; 13: 17590914211019443, 2021.
Article in English | MEDLINE | ID: mdl-34121475

ABSTRACT

Metabolic adaptations in the brain are critical to the establishment and maintenance of normal cellular functions and to the pathological responses to disease processes. Here, we have focused on specific metabolic pathways that are involved in immune-mediated neuronal processes in brain using isolated neurons derived from human autopsy brain sections of normal individuals and individuals diagnosed as Alzheimer's disease (AD). Laser capture microscopy was used to select specific cell types in immune-stained thin brain sections followed by NanoString technology to identify and quantify differences in mRNA levels between age-matched control and AD neuronal samples. Comparisons were also made between neurons isolated from AD brain sections expressing pathogenic hyperphosphorylated AT8- positive (AT8+) tau and non-AT8+ AD neurons using double labeling techniques. The mRNA expression data showed unique patterns of metabolic pathway expression between the subtypes of captured neurons that involved membrane based solute transporters, redox factors, and arginine and methionine metabolic pathways. We also identified the expression levels of a novel metabolic gene, Radical-S-Adenosyl Domain1 (RSAD1) and its corresponding protein, Rsad1, that impact methionine usage and radical based reactions. Immunohistochemistry was used to identify specific protein expression levels and their cellular location in NeuN+ and AT8+ neurons. APOE4 vs APOE3 genotype-specific and sex-specific gene expression differences in these metabolic pathways were also observed when comparing neurons from individuals with AD to age-matched individuals.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Apolipoprotein E4 , Female , Humans , Male , Neurons/metabolism , Phosphorylation , tau Proteins/genetics , tau Proteins/metabolism
17.
J Am Soc Mass Spectrom ; 32(6): 1448-1458, 2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34028275

ABSTRACT

Immune-based metabolic reprogramming of arginine utilization in the brain contributes to the neuronal pathology associated with Alzheimer's disease (AD). To enable our long-term goals of differentiation of AD mouse model genotypes, ages, and sexes based on activity of this pathway, we describe here the novel dosing (using uniformly labeled (13C615N4) arginine) and analysis methods using capillary electrophoresis high-resolution accurate-mass mass spectrometry for isotope tracing of metabolic products of arginine. We developed a pseudoprimed infusion-dosing regimen, using repeated injections, to achieve a steady state of uniformly labeled arginine in 135-195 min post bolus dose. Incorporation of stable isotope labeled carbon and nitrogen from uniformly labeled arginine into a host of downstream metabolites was measured in vivo in mice using serially sampled dried blood spots from the tail. In addition to the dried blood spot time course samples, total isotope incorporation into arginine-related metabolites was measured in the whole brain and plasma after 285 min. Preliminary demonstration of the technique identified differences isotope incorporation in arginine metabolites between male and female mice in a mouse-model of sporadic Alzheimer's disease (APOE4/huNOS2). The technique described herein will permit arginine pathway activity differentiation between mouse genotypes, ages, sexes, or drug treatments in order to elucidate the contribution of this pathway to Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Arginine/analysis , Electrophoresis, Capillary/methods , Mass Spectrometry/methods , Alzheimer Disease/blood , Animals , Apolipoprotein E4/genetics , Arginine/blood , Arginine/chemistry , Brain/metabolism , Carbon Isotopes/analysis , Carbon Isotopes/pharmacokinetics , Disease Models, Animal , Female , Humans , Isotope Labeling , Male , Mice, Transgenic , Nitric Oxide Synthase Type II/genetics , Nitrogen Isotopes/analysis , Nitrogen Isotopes/pharmacokinetics , Proof of Concept Study
18.
J Neurosci ; 29(25): 7957-65, 2009 Jun 24.
Article in English | MEDLINE | ID: mdl-19553436

ABSTRACT

Shown to lower amyloid deposits and improve cognition in APP transgenic mouse models, immunotherapy appears to be a promising approach for the treatment of Alzheimer's disease (AD). Due to limitations in available animal models, however, it has been unclear whether targeting amyloid is sufficient to reduce the other pathological hallmarks of AD-namely, accumulation of pathological, nonmutated tau and neuronal loss. We have now developed two transgenic mouse models (APPSw/NOS2(-/-) and APPSwDI/NOS2(-/-)) that more closely model AD. These mice show amyloid pathology, hyperphosphorylated and aggregated normal mouse tau, significant neuron loss, and cognitive deficits. A beta(1-42) or KLH vaccinations were started in these animals at 12 months, when disease progression and cognitive decline are well underway, and continued for 4 months. Vaccinated APPSwDI/NOS2(-/-) mice, which have predominantly vascular amyloid pathology, showed a 30% decrease in brain A beta and a 35-45% reduction in hyperphosphorylated tau. Neuron loss and cognitive deficits were partially reduced. In APPSw/NOS2(-/-) vaccinated mice, brain A beta was reduced by 65-85% and hyperphosphorylated tau by 50-60%. Furthermore, neurons were completely protected, and memory deficits were fully reversed. Microhemorrhage was observed in all vaccinated APPSw/NOS2(-/-) mice and remains a significant adverse event associated with immunotherapy. Nevertheless, by providing evidence that reducing amyloid pathology also reduces nonmutant tau pathology and blocks neuron loss, these data support the development of amyloid-lowering therapies for disease-modifying treatment of AD.


Subject(s)
Alzheimer Disease/therapy , Alzheimer Vaccines/pharmacology , Amyloid beta-Peptides/therapeutic use , Amyloid/metabolism , Cognition Disorders/therapy , Nerve Degeneration/therapy , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Vaccines/administration & dosage , Amyloid/biosynthesis , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Analysis of Variance , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunotherapy, Active/methods , Maze Learning , Memory Disorders/therapy , Mice , Mice, Transgenic , Neuropsychological Tests , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Phosphorylation
19.
Glia ; 58(4): 410-22, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19780197

ABSTRACT

Spinal cord contusion produces a central lesion surrounded by a peripheral rim of residual white matter. Despite stimulation of NG2(+) progenitor cell proliferation, the lesion remains devoid of normal glia chronically after spinal cord injury (SCI). To investigate potential cell-cell interactions of the predominant cells in the lesion at 3 days after injury, we used magnetic activated cell sorting to purify NG2(+) progenitors and OX42(+) microglia/macrophages from contused rat spinal cord. Purified NG2(+) cells from the injured cord grew into spherical masses when cultured in defined medium with FGF2 plus GGF2. The purified OX42(+) cells did not form spheroids and significantly reduced sphere growth by NG2(+) cells in co-cultures. Conditioned medium from these OX42(+) cells, unlike that from normal peritoneal macrophages or astrocytes also inhibited growth of NG2(+) cells, suggesting inhibition by secreted factors. Expression analysis of freshly purified OX42(+) cells for a panel of six genes for secreted factors showed expression of several that could contribute to inhibition of NG2(+) cells. Further, the pattern of expression of four of these, TNFalpha, TSP1, TIMP1, MMP9, in sequential coronal tissue segments from a 2 cm length of cord centered on the injury epicenter correlated with the expression of Iba1, a marker gene for OX42(+) cells, strongly suggesting a potential regional influence by activated microglia/macrophages on NG2(+) cells in vivo after SCI. Thus, the nonreplacement of lost glial cells in the central lesion zone may involve, at least in part, inhibitory factors produced by microglia/macrophages that are concentrated within the lesion.


Subject(s)
Antigens/metabolism , Macrophages/physiology , Microglia/physiology , Neuroglia/physiology , Proteoglycans/metabolism , Spinal Cord Injuries/physiopathology , Stem Cells/physiology , Animals , Astrocytes/physiology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Female , Matrix Metalloproteinase 9/metabolism , Microfilament Proteins , Rats , Thrombospondin 1/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Stroke ; 41(10 Suppl): S135-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20876489

ABSTRACT

BACKGROUND AND PURPOSE: Cerebral amyloid angiopathy Type 1 is characterized by amyloid ß protein deposition along cerebral capillaries and is accompanied by perivascular neuroinflammation and accumulation of phospho-tau protein. Tg-SwDI mice recapitulate capillary amyloid deposition and associated neuroinflammation but lack accumulation of perivascular phospho-tau protein. METHODS: Tg-SwDI mice were bred onto a nitric oxide synthase 2 gene knockout background and aged for 1 year. Brains were harvested and analyzed using immunohistochemical and quantitative stereological methods to determine the extent of capillary amyloid deposition, perivascular activated microglia, and cell-specific accumulation of phospho-tau protein. Similar methods were also used to compare Tg-SwDI/NOS2(-/-) and human cerebral amyloid angiopathy Type 1 brain tissues. RESULTS: The absence of nitric oxide synthase 2 gene had no effect on the regional pattern or frequency of capillary cerebral amyloid angiopathy or the numbers of perivascular activated microglia in Tg-SwDI mice. On the other hand, Tg-SwDI/NOS2(-/-) mice accumulated phospho-tau protein in perivascular neurons and activated microglia. Tg-SwDI/NOS2(-/-) mice exhibited a very similar distribution of capillary amyloid, activated microglia, and perivascular phospho-tau protein as seen in human cerebral amyloid angiopathy Type 1. CONCLUSIONS: These findings indicate that Tg-SwDI/NOS2(-/-) mice more fully recapitulate the pathological changes observed with capillary amyloid in human cerebral amyloid angiopathy Type 1.


Subject(s)
Brain/pathology , Capillaries/pathology , Cerebral Amyloid Angiopathy/pathology , Nitric Oxide Synthase Type II/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Capillaries/metabolism , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Nitric Oxide Synthase Type II/genetics , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL