Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
FASEB J ; 38(1): e23317, 2024 01.
Article in English | MEDLINE | ID: mdl-38095240

ABSTRACT

Alzheimer's disease (AD) is currently an incurable neurodegenerative disorder and is the most common etiological cause of dementia. Consequently, it has severe burden on its patients and on their caregivers and represents a global health concern. Clinical investigations have indicated that a dysregulation of peripheral T cell immune homeostasis may be involved in the pathogenesis of AD, as well as in the early stages of AD, characterized by mild cognitive impairment (MCI). However, the characteristics and concomitant feasibility of the use of T-cell receptor (TCR) typing for disease diagnosis remains largely unknown. We employed a high-throughput sequencing and multidimensional bioinformatics analyses for the identification of TCR repertoires present in peripheral blood samples of 10 patients with amnestic MCI (aMCI), 10 patients with AD, and 10 healthy controls (HCs). Based on the characteristics of the TCR repertoires in the amount and diversity of combinations of V-J, the spectrum of immune defense, and differentially expressed genes (DEGs), single and specific TCR profiles were observed in the patient samples of aMCI and AD compared to profiles of HCs. In particular, the diversity of TCR clonotypes manifested a pattern of "decreased first and then increased" pattern during the progression from aMCI to AD, a pattern that was not observed in HC samples. Additionally, a total of 46 and 35 amino acid CDR3 sequences with consistent and reverse expressive abundance with diversity of TCR clonotypes were identified, respectively. Taken together, we provide novel and essential preliminary evidence demonstrating the presence of diversity of T cell repertoires from differentially expressed V-J gene segments and amino acid clonotypes using peripheral blood samples from patients with AD, aMCI, and from HC. Such findings have the potential to reveal potential mechanisms through which aMCI progresses to AD and provide a reference for the future development of immune-related diagnoses and therapies for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , T-Lymphocytes , Cognitive Dysfunction/diagnosis , Receptors, Antigen, T-Cell , Amino Acids
2.
Am J Orthod Dentofacial Orthop ; 152(3): 348-354, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28863915

ABSTRACT

INTRODUCTION: The hypothesis of this study was that multiple factors are dominant in causing external apical root resorption (EARR). The objective of this investigation was to better understand the clinical factors that may lead to EARR. METHODS: Maxillary cone-beam computed tomography scans of 18 subjects who were treated with bilateral canine retractions during orthodontics were used to calculate EARR. The subjects were treated using well-calibrated segmental T-loops for delivering a 124-cN retraction force and the moment-to-force ratio suitable for moving the canine under either translation or controlled tipping. The subjects' age, sex, treatment duration, and genotype were collected. RESULTS: Six subjects of the 18 showed definite EARR, meaning that load was not the only causing factor. All 5 subjects with the genotype identified had GG genotype of IL-1ß rs11143634, indicating that people with this genotype may be at high risk. Longer treatment duration, female sex, and older age may also contribute to EARR, although the findings were not statistically significant. CONCLUSIONS: EARR appears to be related to multiple factors. The orthodontic load and the genotype should be the focuses for future studies.


Subject(s)
Cuspid , Root Resorption/etiology , Tooth Movement Techniques/adverse effects , Adolescent , Adult , Age Factors , Child , Cone-Beam Computed Tomography , Cuspid/diagnostic imaging , Female , Genotype , Humans , Interleukin-1beta/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , Root Resorption/diagnostic imaging , Root Resorption/genetics , Sex Factors , Young Adult
3.
Metallomics ; 16(5)2024 05 02.
Article in English | MEDLINE | ID: mdl-38664065

ABSTRACT

Mercury is a well-recognized environmental contaminant and neurotoxin, having been associated with a number of deleterious neurological conditions including neurodegenerative diseases, such as Alzheimer's disease. To investigate how mercury and other metals behave in the brain, we used synchrotron micro-X-ray fluorescence to map the distribution pattern and quantify concentrations of metals in human brain. Brain tissue was provided by the Rush Alzheimer's Disease Center and samples originated from individuals diagnosed with Alzheimer's disease and without cognitive impairment. Data were collected at the 2-ID-E beamline at the Advanced Photon Source at Argonne National Laboratory with an incident beam energy of 13 keV. Course scans were performed at low resolution to determine gross tissue features, after which smaller regions were selected to image at higher resolution. The findings revealed (1) the existence of mercury particles in the brain samples of two subjects; (2) co-localization and linear correlation of mercury and selenium in all particles; (3) co-localization of these particles with zinc structures; and (4) association with sulfur in some of these particles. These results suggest that selenium and sulfur may play protective roles against mercury in the brain, potentially binding with the metal to reduce the induced toxicity, although at different affinities. Our findings call for further studies to investigate the relationship between mercury, selenium, and sulfur, as well as the potential implications in Alzheimer's disease and related dementias.


Subject(s)
Alzheimer Disease , Brain , Mercury , Selenium , Spectrometry, X-Ray Emission , Synchrotrons , Humans , Mercury/analysis , Mercury/metabolism , Selenium/analysis , Selenium/metabolism , Brain/metabolism , Spectrometry, X-Ray Emission/methods , Alzheimer Disease/metabolism , Aged , Male , Female , Zinc/analysis , Zinc/metabolism
4.
J Biol Chem ; 287(16): 12858-66, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22362783

ABSTRACT

Prion diseases are a group of rare, fatal neurodegenerative disorders associated with a conformational transformation of the cellular prion protein (PrP(C)) into a self-replicating and proteinase K-resistant conformer, termed scrapie PrP (PrP(Sc)). Aggregates of PrP(Sc) deposited around neurons lead to neuropathological alterations. Currently, there is no effective treatment for these fatal illnesses; thus, the development of an effective therapy is a priority. PrP peptide-based ELISA assay methods were developed for detection and immunoaffinity chromatography capture was developed for purification of naturally occurring PrP peptide autoantibodies present in human CSF, individual donor serum, and commercial preparations of pooled intravenous immunoglobulin (IVIg). The ratio of anti-PrP autoantibodies (PrP-AA) to total IgG was ∼1:1200. The binding epitope of purified PrP-AA was mapped to an N-terminal region comprising the PrP amino acid sequence KTNMK. Purified PrP-AA potently blocked fibril formation by a toxic 21-amino acid fragment of the PrP peptide containing the amino acid alanine to valine substitution corresponding to position 117 of the full-length peptide (A117V). Furthermore, PrP-AA attenuated the neurotoxicity of PrP(A117V) and wild-type peptides in rat cerebellar granule neuron (CGN) cultures. In contrast, IgG preparations depleted of PrP-AA had little effect on PrP fibril formation or PrP neurotoxicity. The specificity of PrP-AA was demonstrated by immunoprecipitating PrP protein in brain tissues of transgenic mice expressing the human PrP(A117V) epitope and Sc237 hamster. Based on these intriguing findings, it is suggested that human PrP-AA may be useful for interfering with the pathogenic effects of pathogenic prion proteins and, thereby has the potential to be an effective means for preventing or attenuating human prion disease progression.


Subject(s)
Amyloid/immunology , Antibodies, Blocking/pharmacology , Autoantibodies/pharmacology , PrPC Proteins/immunology , PrPSc Proteins/immunology , Prion Diseases , Animals , Antibodies, Blocking/immunology , Antibody Specificity , Autoantibodies/immunology , Cricetinae , Epitope Mapping , Epitopes , Heterozygote , Humans , Immunoglobulins, Intravenous/pharmacology , Mice , Mice, Transgenic , Neuroglia/cytology , Neuroglia/immunology , Neuroglia/pathology , Neurons/cytology , Neurons/immunology , Neurons/pathology , PrPC Proteins/genetics , PrPC Proteins/metabolism , PrPSc Proteins/genetics , PrPSc Proteins/metabolism , Primary Cell Culture , Prion Diseases/immunology , Prion Diseases/prevention & control , Prion Diseases/therapy , Rats , Rats, Sprague-Dawley
6.
Fluids Barriers CNS ; 20(1): 32, 2023 May 01.
Article in English | MEDLINE | ID: mdl-37122007

ABSTRACT

Lead (Pb) is a known environmental risk factor in the etiology of Alzheimer's disease (AD). The existing reports suggest that Pb exposure increases beta-amyloid (Aß) levels in brain tissues and cerebrospinal fluid (CSF) and facilitates the formation of amyloid plaques, which is a pathological hallmark for AD. Pb exposure has long been associated with cerebral vasculature injury. Yet it remained unclear if Pb exposure caused excessive Ab buildup in cerebral vasculature, which may damage the blood-brain barrier and cause abnormal Ab accumulation. This study was designed to investigate the impact of chronic Pb exposure on Aß accumulation in cerebral capillary and the expression of low-density lipoprotein receptor protein-1 (LRP1), a critical Aß transporter, in brain capillary and parenchyma. Sprague-Dawley rats received daily oral gavage at doses of 0, 14 (low-dose), and 27 (high-dose) mg Pb/kg as Pb acetate, 5 d/wk, for 4 or 8 wks. At the end of Pb exposure, a solution containing Aß40 was infused into the brain via the cannulated internal carotid artery. Data by ELISA showed a strikingly high affinity of Ab to cerebral vasculature, which was approximately 7-14 times higher than that to the parenchymal fractions collected from control brains. Pb exposure further aggravated the Aß accumulation in cerebral vasculature in a dose-dependent manner. Western blot analyses revealed that Pb exposure decreased LRP1 expression in cortical capillaries and hippocampal parenchyma. Immunohistochemistry (IHC) studies further revealed a disrupted distribution of LRP1 alongside hippocampal vasculature accompanied with a decreased expression in hippocampal neurons by Pb exposure. Taken together, the current study demonstrated that the cerebral vasculature naturally possessed a high affinity to Aß present in circulating blood. Pb exposure significantly increased Aß accumulation in cerebral vasculature; such an increased Aß accumulation was due partly to the diminished expression of LRP1 in response to Pb in tested brain regions. Perceivably, Pb-facilitated Ab aggravation in cerebral vasculature may contribute to Pb-associated amyloid alterations.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Lead , Animals , Rats , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Capillaries/metabolism , Lead/toxicity , Lead/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Rats, Sprague-Dawley
7.
J Trace Elem Med Biol ; 77: 127146, 2023 May.
Article in English | MEDLINE | ID: mdl-36871432

ABSTRACT

BACKGROUND: The iron concentration increases during normal brain development and is identified as a risk factor for many neurodegenerative diseases, it is vital to monitor iron content in the brain non-invasively. PURPOSE: This study aimed to quantify in vivo brain iron concentration with a 3D rosette-based ultra-short echo time (UTE) magnetic resonance imaging (MRI) sequence. METHODS: A cylindrical phantom containing nine vials of different iron concentrations (iron (II) chloride) from 0.5 millimoles to 50 millimoles and six healthy subjects were scanned using 3D high-resolution (0.94 ×0.94 ×0.94 mm3) rosette UTE sequence at an echo time (TE) of 20 µs. RESULTS: Iron-related hyperintense signals (i.e., positive contrast) were detected based on the phantom scan, and were used to establish an association between iron concentration and signal intensity. The signal intensities from in vivo scans were then converted to iron concentrations based on the association. The deep brain structures, such as the substantia nigra, putamen, and globus pallidus, were highlighted after the conversion, which indicated potential iron accumulations. CONCLUSION: This study suggested that T1-weighted signal intensity could be used for brain iron mapping.


Subject(s)
Iron , Magnetic Resonance Imaging , Humans , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Brain Mapping/methods , Contrast Media
8.
J Neurosci ; 31(15): 5847-54, 2011 Apr 13.
Article in English | MEDLINE | ID: mdl-21490226

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder primarily affecting regions of the brain responsible for higher cognitive functions. Immunization against ß-amyloid (Aß) in animal models of AD has been shown to be effective on the molecular level but also on the behavioral level. Recently, we reported naturally occurring autoantibodies against Aß (NAbs-Aß) being reduced in Alzheimer's disease patients. Here, we further investigated their physiological role: in epitope mapping studies, NAbs-Aß recognized the mid-/C-terminal end of Aß and preferentially bound to oligomers but failed to bind to monomers/fibrils. NAbs-Aß were able to interfere with Aß peptide toxicity, but NAbs-Aß did not readily clear senile plaques although early fleecy-like plaques were reduced. Administration of NAbs-Aß in transgenic mice improved the object location memory significantly, almost reaching performance levels of wild-type control mice. These findings suggest a novel physiological mechanism involving NAbs-Aß to dispose of proteins or peptides that are prone to forming toxic aggregates.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Autoantibodies/immunology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Animals, Genetically Modified , Antibody Formation , Behavior, Animal , Brain/pathology , Cells, Cultured , Chromatography, Gel , Disease Models, Animal , Epitopes , Female , Humans , Immunization , Immunoglobulin G/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Models, Molecular , Plaque, Amyloid/pathology , Surface Plasmon Resonance
9.
Chemosphere ; 301: 134667, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35460676

ABSTRACT

A synergetic system of water falling film dielectric barrier discharge (DBD) plasma and persulfate (PS) was established and applied to enhance the enrofloxacin (EFA) degradation in this study. The simultaneous existence of electrons, reactive species, heat and UV-visible light in the DBD plasma system were utilized together to activate the PS to form SO4-· and other reactive oxygen species (ROS), and then worked in synergy with the DBD plasma to oxidize the EFA. The obtained results verified that there was a significant increase in the degradation percentages of EFA (20 mg L-1) in the DBD/PS system, and the trend was more obvious under the condition of larger discharge power input. When 0.8 mM PS was added into the DBD system with 0.8 kW discharge power, the degradation percentage of EFA could reach 99.35% after 60 min treatment, the corresponding synergetic factor (SF) was 7.94. Analysis of the O3 and the H2O2 concentrations in the DBD plasma system before and after the PS addition explained the activation of the PS by the HO·. The quenching experiments on reactive species suggested that SO4-·, HO·, and 1O2 were all important reactive species for EFA degradation. The intermediates formed by the EFA degradation were detected and the degradation pathways were speculated. Results of toxicity analysis illustrated that the toxicity of the initial EFA solution decreased after degradation in the synergetic system of DBD/PS.


Subject(s)
Water Pollutants, Chemical , Water , Enrofloxacin/analysis , Hydrogen Peroxide , Oxidation-Reduction , Water Pollutants, Chemical/analysis
10.
Biology (Basel) ; 11(2)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35205174

ABSTRACT

Lead (Pb) is an environmental element that has been implicated in the development of dementia and Alzheimer's disease (AD). Additionally, innate immune activation contributes to AD pathophysiology. However, the mechanisms involved remain poorly understood. The choroid plexus (CP) is not only the site of cerebrospinal fluid (CSF) production, but also an important location for communication between the circulation and the CSF. In this study, we investigated the involvement of the CP during Pb exposure by evaluating the expression of the monocyte chemoattractant protein-1 (MCP-1). MCP-1 is highly expressed in the CP compared to other CNS tissues. MCP-1 regulates macrophage infiltration and is upregulated in AD brains. Our study revealed that Pb exposure stimulated MCP-1 expression, along with a significantly increased macrophage infiltration into the CP. By using cultured Z310 rat CP cells, Pb exposure stimulated MCP-1 expression in a dose-related fashion and markedly activated both NF-κB and p38 MAP kinase. Interestingly, both SB 203580, a p38 inhibitor, and BAY 11-7082, an NF-κB p65 inhibitor, significantly blocked Pb-induced MCP-1 expression. However, SB203580 did not directly inhibit NF-κB p65 phosphorylation. In conclusion, Pb exposure stimulates MCP-1 expression via the p38 and NF-κB p65 pathways along with macrophage infiltration into the CP.

11.
Neurotoxicology ; 88: 106-115, 2022 01.
Article in English | MEDLINE | ID: mdl-34793780

ABSTRACT

Lead (Pb) is a well-known neurotoxicant and environmental hazard. Recent experimental evidence has linked Pb exposure with neurological deterioration leading to neurodegenerative diseases, such as Alzheimer's disease. To understand brain regional distribution of Pb and its interaction with other metal ions, we used synchrotron micro-x-ray fluorescence technique (µ-XRF) to map the metal distribution pattern and to quantify metal concentrations in mouse brains. Lead-exposed mice received oral gavage of Pb acetate once daily for 4 weeks; the control mice received sodium acetate. Brain tissues were cut into slices and subjected for analysis. Synchrotron µ-XRF scans were run on the PETRA III P06 beamline (DESY). Coarse scans of the entire brain were performed to locate the cortex and hippocampus, after which scans with higher resolution were run in these areas. The results showed that: a) the total Pb intensity in Pb-exposed brain slices was significantly higher than in control brain; b) Pb typically deposited in localized particles of <10 um2 in both the Pb-exposed and control brain slices, with more of these particles in Pb-exposed samples; c) selenium (Se) was significantly correlated with Pb in these particles in the cortex and hippocampus/corpus callosum regions in the Pb-exposed samples, and the molar ratio of the Se and Pb in these particles is close to 1:1. These results indicated that Se may play a crucial role in Pb-induced neurotoxicity. Our findings call for further studies to investigate the relationship between Pb exposure and possible Se detoxification responses, and the implication in the etiology of Alzheimer's disease.


Subject(s)
Brain Chemistry/drug effects , Lead Poisoning, Nervous System/metabolism , Lead/analysis , Selenium/analysis , Animals , Lead/administration & dosage , Male , Mice , Spectrometry, X-Ray Emission , Synchrotrons
12.
Dement Geriatr Cogn Disord ; 32(3): 198-201, 2011.
Article in English | MEDLINE | ID: mdl-22122982

ABSTRACT

BACKGROUND/AIMS: Some studies have implicated the role of apolipoprotein J [clusterin (CLU), apoJ] in the pathogenesis of Alzheimer's disease (AD). In this study, we investigated the polymorphisms rs11136000 and rs9331888 within CLU in late-onset sporadic AD (LOAD) patients and nondemented subjects. METHODS: LOAD patients and control subjects were analyzed. Genotyping of rs11136000 and rs9331888 was performed using standard PCR following different restriction endonuclease digestion. RESULTS: Although there were no significant differences in genotype frequencies of these two polymorphisms, the haplotype CG was associated with a statistically significantly increased LOAD risk. CONCLUSION: The rs11136000 and rs9331888 polymorphisms of the CLU gene are associated with LOAD.


Subject(s)
Alzheimer Disease/genetics , Clusterin/genetics , Polymorphism, Genetic/genetics , Age Factors , Age of Onset , Aged , Aged, 80 and over , Female , Gene Frequency , Genetic Association Studies , Genotype , Haplotypes , Humans , Linkage Disequilibrium , Male , Real-Time Polymerase Chain Reaction , Sex Factors
13.
Neuroreport ; 32(1): 38-43, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33252477

ABSTRACT

Minocycline and doxycycline, two semisynthetic second-generation tetracyclines, are reported to provide neuroprotection against brain injury and glutamate-induced neurotoxicity in neuronal cultures. Doxycycline has been postulated as the potential ideal candidate for further therapeutic development as it has fewer adverse effects than minocycline. In this study, we determined whether minocycline and doxycycline could similarly protect neurons against excitotoxic insults. We treated cultured rat cortical neurons and cerebellar granule neurons (CGN) with excitotoxic concentrations of NMDA or glutamate in the presence or absence of minocycline or doxycycline. Intracellular Ca concentration ([Ca]i) was also measured using a Fluorescent Light Imaging Plate Reader (FLIPR; Molecular Devices) with the calcium sensitive dye Fluo-3 AM. We found that minocycline and tetracycline markedly protected neurons against NMDA- and glutamate-induced neuronal death. In contrast, the structurally related tetracycline, doxycycline, was ineffective at concentrations up to 100 µM. Furthermore, minocycline, but not doxycycline, also significantly attenuated NMDA- or glutamate-induced [Ca]i in both cortical neurons and CGN. Our results suggest that minocycline but not doxycycline is able to directly block NMDA- or glutamate-induced excitotoxicity in neurons most likely by inhibiting NMDA- and glutamate-induced [Ca]i. This finding may contribute to our understanding of the molecular mechanisms underlying doxycycline- and minocycline-induced neuroprotection.


Subject(s)
Doxycycline/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Minocycline/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , N-Methylaspartate/toxicity , Neurons/metabolism , Rats , Rats, Sprague-Dawley
14.
Stem Cells ; 27(2): 478-88, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19023032

ABSTRACT

Adipose tissue stroma contains a population of mesenchymal stem cells, which support repair when administered to damaged tissues, in large part through secreted trophic factors. We directly tested the ability of media collected from cultured adipose-derived stem cells (ASCs) to protect neurons in a rat model of brain hypoxic-ischemic (HI) injury. Concentrated conditioned medium from cultured rat ASCs (ASC-CM) or control medium was infused through the jugular vein of neonatal Sprague-Dawley rats subjected to HI injury. The ASC-CM was administered either 1 hour before or 24 hours after induction of injury. Analysis at 1 week indicated that administration at both time points significantly protected against hippocampal and cortical volume loss. Analysis of parallel groups for behavioral and learning changes at 2 months postischemia demonstrated that both treated groups performed significantly better than the controls in Morris water maze functional tests. Subsequent post-mortem evaluation of brain damage at the 2-month time point confirmed neuronal loss to be similar to that observed at 1 week for all groups. We have identified several neurotrophic factors in ASC-CM, particularly insulin-like growth factor-1 and brain-derived neurotrophic factor, which are important factors that could contribute to the protective effects of ASCs observed in studies with both in vitro and in vivo neuronal injury models. These data suggest that delivery of the milieu of factors secreted by ASCs may be a viable therapeutic option for treatment of HI, as well as other brain injuries.


Subject(s)
Adipose Tissue/cytology , Brain/drug effects , Culture Media, Conditioned/pharmacology , Hypoxia-Ischemia, Brain/prevention & control , Stromal Cells/physiology , Animals , Animals, Newborn , Cells, Cultured , Female , Humans , Hypoxia-Ischemia, Brain/pathology , Maze Learning , Pregnancy , Rats , Rats, Sprague-Dawley , Stromal Cells/metabolism
15.
Mov Disord ; 25(1): 97-107, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20014118

ABSTRACT

The aim of the study was to investigate the efficacy of the antibiotic minocycline as a drug treatment in patients with Multiple-System-Atrophy Parkinson-type (MSA-P). Sixty-three patients were randomized to minocycline 200 mg/d (n = 32) or a matching placebo (n = 31). The primary outcome variable was the change in the value of the motor score of the Unified Multiple-System-Atrophy Rating-Scale (UMSARSII) from baseline to 48 weeks. Secondary outcome variables included subscores and individual Parkinsonian symptoms as determined by the UMSARS and the Unified-Parkinson's-Disease Rating-Scale (UPDRS). Health-related quality of life (HrQoL) was assessed using the EQ-5D and SF-12. "Progression rate" was assumed to be reflected in the change in motor function over 48 weeks. At 24 weeks and 48 weeks of follow-up, there was a significant deterioration in motor scores in both groups, but neither the change in UMSARSII nor in UPDRSIII differed significantly between treatment groups, i.e. "progression rate" was considered to be similar in both treatment arms. HrQoL did not differ among the two treatment arms. In a small subgroup of patients (n = 8; minocycline = 3, placebo = 5)[(11)C](R)-PK11195-PET was performed. The three patients in the minocycline group had an attenuated mean increase in microglial activation as compared to the placebo group (P = 0.07) and in two of them individually showed decreased [11C](R)-PK11195 binding actually decreased. These preliminary PET-data suggest that minocycline may interfere with microglial activation. The relevance of this observation requires further investigation. This prospective, 48 week, randomized, double-blind, multinational study failed to show a clinical effect of minocycline on symptom severity as assessed by clinical motor function.


Subject(s)
Isoquinolines , Minocycline/therapeutic use , Multiple System Atrophy/diagnostic imaging , Multiple System Atrophy/drug therapy , Neuroprotective Agents/therapeutic use , Adult , Aged , Double-Blind Method , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Multiple System Atrophy/psychology , Positron-Emission Tomography/methods , Quality of Life , Severity of Illness Index , Statistics, Nonparametric , Time Factors , Treatment Outcome
16.
J Trace Elem Med Biol ; 61: 126520, 2020 Apr 08.
Article in English | MEDLINE | ID: mdl-32325398

ABSTRACT

Formation of amyloid plaques is the hallmark of Alzheimer's disease. Our early studies show that lead (Pb) exposure in PDAPP transgenic mice increases ß-amyloid (Aß) levels in the cerebrospinal fluid (CSF) and hippocampus, leading to the formation of amyloid plaques in mouse brain. Aß in the CSF is regulated by the blood-CSF barrier (BCB) in the choroid plexus. However, the questions as to whether and how Pb exposure affected the influx and efflux of Aß in BCB remained unknown. This study was conducted to investigate whether Pb exposure altered the Aß efflux in the choroid plexus from the CSF to blood, and how Pb may affect the expression and subcellular translocation of two major Aß transporters, i.e., the receptor for advanced glycation end-products (RAGE) and the low density lipoprotein receptor protein-1 (LRP1) in the choroid plexus. Sprague-Dawley rats received daily oral gavage at doses of 0, 14 (low-dose), and 27 (high-dose) mg Pb/kg as Pb acetate, 5 d/wk, for 4 or 8 wks. At the end of Pb exposure, a solution containing Aß40 (2.5 µg/mL) was infused to rat brain via a cannulated internal carotid artery. Subchronic Pb exposure at both dose levels significantly increased Aß levels in the CSF and choroid plexus (p < 0.05) by ELISA. Confocal data showed that 4-wk Pb exposures prompted subcellular translocation of RAGE from the choroidal cytoplasm toward apical microvilli. Furthermore, it increased the RAGE expression in the choroid plexus by 34.1 % and 25.1 % over the controls (p < 0.05) in the low- and high- dose groups, respectfully. Subchronic Pb exposure did not significantly affect the expression of LRP1; yet the high-dose group showed LRP1 concentrated along the basal lamina. The data from the ventriculo-cisternal perfusion revealed a significantly decreased efflux of Aß40 from the CSF to blood via the blood-CSF barrier. Incubation of freshly dissected plexus tissues with Pb in artificial CSF supported a Pb effect on increased RAGE expression. Taken together, these data suggest that Pb accumulation in the choroid plexus after subchronic exposure reduces the clearance of Aß from the CSF to blood by the choroid plexus, which, in turn, leads to an increase of Aß in the CSF. Interaction of Pb with RAGE and LRP1 in choroidal epithelial cells may contribute to the altered Aß transport by the blood-CSF barrier in brain ventricles.

17.
J Trace Elem Med Biol ; 62: 126648, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32980769

ABSTRACT

BACKGROUND: Lead (Pb) is an environmental factor has been suspected of contributing to the dementia including Alzheimer's disease (AD). Our previous studies have shown that Pb exposure at the subtoxic dose increased brain levels of beta-amyloid (Aß) and amyloid plaques, a pathological hallmark for AD, in amyloid precursor protein (APP) transgenic mice, and is hypothesized to inhibit Aß clearance in the blood- cerebrospinal fluid (CSF) barrier. However, it remains unclear how different levels of Pb affect Aß clearance in the whole blood-brain barrier system. This study was designed to investigate whether chronic exposure of Pb affected the permeability of the blood-brain barrier system by using the Dynamic Contrast-Enhanced Computerized Tomography (DCE-CT) method. METHODS: DEC-CT was used to investigate whether chronic exposure of toxic Pb affected the permeability of the real-time blood brain barrier system. RESULTS: Data showed that Pb exposure increased permeability surface area product, and also significantly induced brain perfusion. However, Pb exposure did not alter extracellular volumes or fractional blood volumes of mouse brain. CONCLUSION: Our data suggest that Pb exposure at subtoxic and toxic levels directly targets the brain vasculature and damages the blood brain barrier system.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Lead/toxicity , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , Mice , Mice, Transgenic
18.
Brain Res Bull ; 154: 102-105, 2020 01.
Article in English | MEDLINE | ID: mdl-31733348

ABSTRACT

Unconjugated bilirubin, the end product of heme catabolism and antioxidant, induced brain damage in human neonates is a well-recognized clinical syndrome. However, the cellular and molecular mechanisms underlying bilirubin neurotoxicity remain unclear. To characterize the sequence of events leading to bilirubin-induced neurotoxicity, we investigated whether bilirubin-induced glial activation was involved in bilirubin neurotoxicity by exposing co-cultured rat glial cells and cerebellar granule neurons (CGN) to bilirubin. We found that bilirubin could markedly induce the expression of TNF-α and iNOS in glial cells, and even at low concentrations, the co-culture of glial cells with neurons significantly enhances neurotoxicity of bilirubin. Pretreatment of the co-cultured cells with minocycline protected CGN from glia-mediated bilirubin neurotoxicity and inhibited overexpression of TNF-α and iNOS in glia. Furthermore, we found that high doses of bilirubin were able to induce glial injury, and minocycline attenuated bilirubin-induced glial cell death. Our data suggest that glial cells play an important role in brain damage caused by bilirubin, and minocycline blocks bilirubin-induced encephalopathy possibly by directly and indirectly inhibiting neuronal death pathways.


Subject(s)
Bilirubin/metabolism , Minocycline/pharmacology , Neuroglia/metabolism , Animals , Bilirubin/toxicity , Cell Death/drug effects , Cerebellum/cytology , Minocycline/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Neurosci Lett ; 449(3): 240-5, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-18786612

ABSTRACT

Active as well as passive immunization against beta-amlyoid (Abeta) has been proposed as a treatment to lower cerebral amyloid burden and stabilize cognitive decline in Alzheimer's disease (AD). To clarify the mechanism of action underlying passive immunization, the in vivo distribution (and sites of degradation) of peripherally administered radiolabeled human and mouse anti-Abeta antibodies were analyzed in a transgenic mouse model of AD. In APP23 mice, a model in which mutated human amyloid precursor protein is overexpressed, the biodistribution of intravenously applicated (111)indium-conjugated affinity-purified human polyclonal autoantibodies (NAbs-Abeta) was compared to that of monoclonal anti-Abeta(1-17) (6E10), anti-Abeta(17-24) antibodies (4G8) and anti-CD-20 (Rituximab), a non-Abeta targeting control. Blood clearance half-lives were 50+/-6h for Rituximab, 20-30h for NAbs-Abeta, 29+/-5h for 4G8 and 27+/-3h for 6E10. Blood activity was higher for 6E10 at 4h as compared to 4G8, Rituximab and NAbs-Abeta. At the 96h time point, Rituximab had the highest blood activity among the antibodies tested. As expected, all antibodies displayed hepatobiliary clearance. Additionally, NAbs-Abeta was excreted in the urinary tract. Liver and kidney uptake of NAbs-Abeta increased over time and was higher than in the monoclonal antibodies at 48h/96h. The brain-to-blood radioactivity ratio for NAbs-Abeta at later time points (>48h) was higher than that of 6E10, 4G8 and Rituximab. In addition, the distribution varied, with highest values found in the hippocampus. Our data indicate a cerebral accumulation of human NAbs-Abeta in the APP23 model. Further studies with human immunoglobulins and particularly with those that recognize different Abeta-epitopes are required in order to delineate in more detail the mode of action of NAbs-Abeta.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Autoantibodies/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Murine-Derived , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Humans , Indium/pharmacokinetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Rituximab , Time Factors
20.
Neurol Res ; 31(5): 483-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19500451

ABSTRACT

Cerebral hypoxia-ischemia during the perinatal period is the single most important cause of acute newborn mortality and chronic disability. Despite our increasing understanding of the mechanisms of neuronal injury, an effective clinical therapy has yet to be established to mitigate brain damage and improve the prognosis and well-being of these newborn patients. Insulin-like growth factor 1 (IGF-1) is a well-known neurotrophic factor, essential for the survival and functional maturation of immature neurons. This study demonstrated that subcutaneous administration of IGF-1 at 24 and 48 hours of recovery significantly reduced hypoxia-ischemia-induced injury to immature rat brains and improved long-term memory and cognitive behavior. IGF-1's therapeutic effects likely involve its ability to prevent delayed apoptosis, as we demonstrated in primary cortical neuronal cultures under oxygen and glucose deprivation. IGF-1's neuroprotective effects parallel the activities of phosphatidylinositol-3/Akt and its down-stream signaling pathway, suggesting a potential mechanistic link. Overall, evidence from this investigation strongly supports IGF-1's potential therapeutic use in the treatment of hypoxic-ischemic encephalopathy in newborn patients.


Subject(s)
Cell Survival/physiology , Cytoprotection/physiology , Hypoxia-Ischemia, Brain/physiopathology , Insulin-Like Growth Factor I/pharmacology , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Brain/pathology , Brain Injuries/etiology , Brain Injuries/physiopathology , Cell Survival/drug effects , Cells, Cultured , Cytoprotection/drug effects , Female , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/prevention & control , Maze Learning/drug effects , Memory , Neurons/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL