Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Exp Eye Res ; 241: 109818, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38422787

ABSTRACT

Down syndrome (DS) is the most common chromosomal disorder in humans. DS is associated with increased prevalence of several ocular sequelae, including characteristic blue-dot cerulean cataract. DS is accompanied by age-dependent accumulation of Alzheimer's disease (AD) amyloid-ß (Aß) peptides and amyloid pathology in the brain and comorbid early-onset Aß amyloidopathy and colocalizing cataracts in the lens. Quasi-elastic light scattering (QLS) is an established optical technique that noninvasively measures changes in protein size distributions in the human lens in vivo. In this cross-sectional study, lenticular QLS correlation time was decreased in adolescent subjects with DS compared to age-matched control subjects. Clinical QLS was consistent with alterations in relative particle hydrodynamic radius in lenses of adolescents with DS. These correlative results suggest that noninvasive QLS can be used to evaluate molecular changes in the lenses of individuals with DS.


Subject(s)
Alzheimer Disease , Cataract/congenital , Down Syndrome , Lens, Crystalline , Humans , Adolescent , Down Syndrome/complications , Down Syndrome/pathology , Cross-Sectional Studies , Alzheimer Disease/metabolism , Lens, Crystalline/metabolism , Amyloid beta-Peptides/metabolism
2.
Radiology ; 306(1): 244-251, 2023 01.
Article in English | MEDLINE | ID: mdl-36125373

ABSTRACT

Background T1-weighted MRI and quantitative longitudinal relaxation rate (R1) mapping have been used to evaluate gadolinium retention in the brain after gadolinium-based contrast agent (GBCA) administration. Whether MRI measures accurately reflect gadolinium regional distribution and concentration in the brain remains unclear. Purpose To compare gadolinium retention in rat forebrain measured with in vivo quantitative MRI R1 and ex vivo laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) mapping after gadobenate, gadopentetate, gadodiamide, or gadobutrol administration. Materials and Methods Adult female Sprague-Dawley rats were randomly assigned to one of five groups (eight per group) and administered gadobenate, gadopentetate, gadodiamide, gadobutrol (2.4 mmol/kg per week for 5 weeks), or saline (4.8 mL/kg per week for 5 weeks). MRI R1 mapping was performed at baseline and 1 week after the final injection to determine R1 and ΔR1. Postmortem brains from the same rats were analyzed with LA-ICP-MS elemental mapping to determine regional gadolinium concentrations. Student t tests were performed to compare results between GBCA and saline groups. Results Rats that were administered gadobenate showed gadolinium-related MRI ΔR1 in 39.5% of brain volume (ΔR1 = 0.087 second-1 ± 0.051); gadopentetate, 20.6% (ΔR1 = 0.069 second-1 ± 0.018); gadodiamide, 5.4% (ΔR1 = 0.055 second-1 ± 0.019); and gadobutrol, 2.2% (ΔR1 = 0.052 second-1 ± 0.041). Agent-specific gadolinium-related ΔR1 was detected in multiple forebrain regions (neocortex, hippocampus, dentate gyrus, thalamus, and caudate-putamen) in rats treated with gadobenate or gadopentetate, whereas rats treated with gadodiamide showed gadolinium-related ΔR1 in caudate-putamen. By contrast, LA-ICP-MS elemental mapping showed a similar regional distribution pattern of heterogeneous retained gadolinium in the forebrain of rats treated with gadobenate, gadopentetate, or gadodiamide, with the average gadolinium concentration of 0.45 µg · g-1 ± 0.07, 0.50 µg · g-1 ± 0.10, and 0.60 µg · g-1 ± 0.11, respectively. Low levels (0.01 µg · g-1 ± 0.00) of retained gadolinium were detected in the forebrain of gadobutrol-treated rats. Conclusion Differences in in vivo MRI longitudinal relaxation rate versus ex vivo elemental mass spectrometry measures of retained gadolinium in rat forebrains suggest that some forms of retained gadolinium may escape detection with MRI. © RSNA, 2022 Online supplemental material is available for this article.


Subject(s)
Gadolinium , Organometallic Compounds , Rats , Female , Animals , Rats, Sprague-Dawley , Gadolinium DTPA , Contrast Media , Meglumine , Magnetic Resonance Imaging/methods , Brain , Mass Spectrometry
3.
Exp Eye Res ; 221: 108974, 2022 08.
Article in English | MEDLINE | ID: mdl-35202705

ABSTRACT

Neuropathological hallmarks of Alzheimer's disease (AD) include pathogenic accumulation of amyloid-ß (Aß) peptides and age-dependent formation of amyloid plaques in the brain. AD-associated Aß neuropathology begins decades before onset of cognitive symptoms and slowly progresses over the course of the disease. We previously reported discovery of Aß deposition, ß-amyloidopathy, and co-localizing supranuclear cataracts (SNC) in lenses from people with AD, but not other neurodegenerative disorders or normal aging. We confirmed AD-associated Aß molecular pathology in the lens by immunohistopathology, amyloid histochemistry, immunoblot analysis, epitope mapping, immunogold electron microscopy, quantitative immunoassays, and tryptic digest mass spectrometry peptide sequencing. Ultrastructural analysis revealed that AD-associated Aß deposits in AD lenses localize as electron-dense microaggregates in the cytoplasm of supranuclear (deep cortex) fiber cells. These Aß microaggregates also contain αB-crystallin and scatter light, thus linking Aß pathology and SNC phenotype expression in the lenses of people with AD. Subsequent research identified Aß lens pathology as the molecular origin of the distinctive cataracts associated with Down syndrome (DS, trisomy 21), a chromosomal disorder invariantly associated with early-onset Aß accumulation and Aß amyloidopathy in the brain. Investigation of 1249 participants in the Framingham Eye Study found that AD-associated quantitative traits in brain and lens are co-heritable. Moreover, AD-associated lens traits preceded MRI brain traits and cognitive deficits by a decade or more and predicted future AD. A genome-wide association study of bivariate outcomes in the same subjects identified a new AD risk factor locus in the CTNND2 gene encoding δ-catenin, a protein that modulates Aß production in brain and lens. Here we report identification of AD-related human Aß (hAß) lens pathology and age-dependent SNC phenotype expression in the Tg2576 transgenic mouse model of AD. Tg2576 mice express Swedish mutant human amyloid precursor protein (APP-Swe), accumulate hAß peptides and amyloid pathology in the brain, and exhibit cognitive deficits that slowly progress with increasing age. We found that Tg2576 trangenic (Tg+) mice, but not non-transgenic (Tg-) control mice, also express human APP, accumulate hAß peptides, and develop hAß molecular and ultrastructural pathologies in the lens. Tg2576 Tg+ mice exhibit age-dependent Aß supranuclear lens opacification that recapitulates lens pathology and SNC phenotype expression in human AD. In addition, we detected hAß in conditioned medium from lens explant cultures prepared from Tg+ mice, but not Tg- control mice, a finding consistent with constitutive hAß generation in the lens. In vitro studies showed that hAß promoted mouse lens protein aggregation detected by quasi-elastic light scattering (QLS) spectroscopy. These results support mechanistic (genotype-phenotype) linkage between Aß pathology and AD-related phenotypes in lens and brain. Collectively, our findings identify Aß pathology as the shared molecular etiology of two age-dependent AD-related cataracts associated with two human diseases (AD, DS) and homologous murine cataracts in the Tg2576 transgenic mouse model of AD. These results represent the first evidence of AD-related Aß pathology outside the brain and point to lens Aß as an optically-accessible AD biomarker for early detection and longitudinal monitoring of this devastating neurodegenerative disease.


Subject(s)
Alzheimer Disease , Cataract , Neurodegenerative Diseases , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/pathology , Cataract/pathology , Disease Models, Animal , Genome-Wide Association Study , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/pathology
4.
Sci Rep ; 11(1): 4292, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619310

ABSTRACT

Galactic cosmic radiation (GCR) composed of high-energy, heavy particles (HZE) poses potentially serious hazards to long-duration crewed missions in deep space beyond earth's magnetosphere, including planned missions to Mars. Chronic effects of GCR exposure on brain structure and cognitive function are poorly understood, thereby limiting risk reduction and mitigation strategies to protect against sequelae from exposure during and after deep-space travel. Given the selective vulnerability of the hippocampus to neurotoxic insult and the importance of this brain region to learning and memory, we hypothesized that GCR-relevant HZE exposure may induce long-term alterations in adult hippocampal neurogenesis, synaptic plasticity, and hippocampal-dependent learning and memory. To test this hypothesis, we irradiated 3-month-old male and female mice with a single, whole-body dose of 10, 50, or 100 cGy 56Fe ions (600 MeV, 181 keV/µm) at Brookhaven National Laboratory. Our data reveal complex, dynamic, time-dependent effects of HZE exposure on the hippocampus. Two months post exposure, neurogenesis, synaptic plasticity and learning were impaired compared to sham-irradiated, age-matched controls. By six months post-exposure, deficits in spatial learning were absent in irradiated mice, and synaptic potentiation was enhanced. Enhanced performance in spatial learning and facilitation of synaptic plasticity in irradiated mice persisted 12 months post-exposure, concomitant with a dramatic rebound in adult-born neurons. Synaptic plasticity and spatial learning remained enhanced 20 months post-exposure, indicating a life-long influence on plasticity and cognition from a single exposure to HZE in young adulthood. These findings suggest that GCR-exposure can persistently alter brain health and cognitive function during and after long-duration travel in deep space.


Subject(s)
Brain/metabolism , Brain/radiation effects , Cosmic Radiation/adverse effects , Astronauts , Biomarkers , Brain/physiopathology , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Dentate Gyrus/radiation effects , Environmental Exposure/adverse effects , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Hippocampus/radiation effects , Humans , Male , Neurogenesis/radiation effects , Radiation Exposure/adverse effects , Space Flight , Spatial Learning/radiation effects , Time Factors
5.
J Gerontol A Biol Sci Med Sci ; 75(9): e53-e62, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32515825

ABSTRACT

The absence of clinical tools to evaluate individual variation in the pace of aging represents a major impediment to understanding aging and maximizing health throughout life. The human lens is an ideal tissue for quantitative assessment of molecular aging in vivo. Long-lived proteins in lens fiber cells are expressed during fetal life, do not undergo turnover, accumulate molecular alterations throughout life, and are optically accessible in vivo. We used quasi-elastic light scattering (QLS) to measure age-dependent signals in lenses of healthy human subjects. Age-dependent QLS signal changes detected in vivo recapitulated time-dependent changes in hydrodynamic radius, protein polydispersity, and supramolecular order of human lens proteins during long-term incubation (~1 year) and in response to sustained oxidation (~2.5 months) in vitro. Our findings demonstrate that QLS analysis of human lens proteins provides a practical technique for noninvasive assessment of molecular aging in vivo.


Subject(s)
Aging/physiology , Crystallins/physiology , Dynamic Light Scattering , Lens, Crystalline/physiology , Adolescent , Adult , Child , Child, Preschool , Cross-Sectional Studies , Crystallins/chemistry , Dynamic Light Scattering/methods , Electrophoresis, Polyacrylamide Gel , Female , Humans , Male , Microscopy, Electron, Transmission , Middle Aged , Oxidation-Reduction , Young Adult
6.
Radiology ; 294(2): 377-385, 2020 02.
Article in English | MEDLINE | ID: mdl-31769744

ABSTRACT

Background Gadolinium retention after repeated gadolinium-based contrast agent (GBCA) exposure has been reported in subcortical gray matter. However, gadolinium retention in the cerebral cortex has not been systematically investigated. Purpose To determine whether and where gadolinium is retained in rat and human cerebral cortex. Materials and Methods The cerebral cortex in Sprague-Dawley rats treated with gadopentetate dimeglumine (three doses over 4 weeks; cumulative gadolinium dose, 7.2 mmol per kilogram of body weight; n = 6) or saline (n = 6) was examined with antemortem MRI. Two human donors with repeated GBCA exposure (three and 15 doses; 1 and 5 months after exposure), including gadopentetate dimeglumine, and two GBCA-naive donors were also evaluated. Elemental brain maps (gadolinium, phosphorus, zinc, copper, iron) for rat and human brains were constructed by using laser ablation inductively coupled plasma mass spectrometry. Results Gadopentetate dimeglumine-treated rats showed region-, subregion-, and layer-specific gadolinium retention in the neocortex (anterior cingulate cortex: mean gadolinium concentration, 0.28 µg ∙ g-1 ± 0.04 [standard error of the mean]) that was comparable (P > .05) to retention in the allocortex (mean gadolinium concentration, 0.33 µg ∙ g-1 ± 0.04 in piriform cortex, 0.24 µg ∙ g-1 ± 0.04 in dentate gyrus, 0.17 µg ∙ g-1 ± 0.04 in hippocampus) and subcortical structures (0.47 µg ∙ g-1 ± 0.10 in facial nucleus, 0.39 µg ∙ g-1 ± 0.10 in choroid plexus, 0.29 µg ∙ g-1 ± 0.05 in caudate-putamen, 0.26 µg ∙ g-1 ± 0.05 in reticular nucleus of the thalamus, 0.24 µg ∙ g-1 ± 0.04 in vestibular nucleus) and significantly greater than that in the cerebellum (0.17 µg ∙ g-1 ± 0.03, P = .01) and white matter tracts (anterior commissure: 0.05 µg ∙ g-1 ± 0.01, P = .002; corpus callosum: 0.05 µg ∙ g-1 ± 0.02, P = .001; cranial nerve: 0.02 µg ∙ g-1 ± 0.01, P = .004). Retained gadolinium colocalized with parenchymal iron. T1-weighted MRI signal intensification was not observed. Gadolinium retention was detected in the cerebral cortex, pia mater, and pia-ensheathed leptomeningeal vessels in two GBCA-exposed human brains but not in two GBCA-naive human brains. Conclusion Repeated gadopentetate dimeglumine exposure is associated with gadolinium retention in specific regions, subregions, and layers of cerebral cortex that are critical for higher cognition, affect, and behavior regulation, sensorimotor coordination, and executive function. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Kanal in this issue.


Subject(s)
Cerebral Cortex/metabolism , Contrast Media/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Administration, Intravenous , Adult , Animals , Contrast Media/administration & dosage , Female , Gadolinium DTPA/administration & dosage , Humans , Male , Mass Spectrometry/methods , Middle Aged , Models, Animal , Rats , Rats, Sprague-Dawley
7.
Aging (Albany NY) ; 11(24): 12497-12531, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844034

ABSTRACT

Life-long eye lens function requires an appropriate gradient refractive index, biomechanical integrity and transparency. We conducted an extensive study of wild-type mouse lenses 1-30 months of age to define common age-related changes. Biomechanical testing and morphometrics revealed an increase in lens volume and stiffness with age. Lens capsule thickness and peripheral fiber cell widths increased between 2 to 4 months of age but not further, and thus, cannot account for significant age-dependent increases in lens stiffness after 4 months. In lenses from mice older than 12 months, we routinely observed cataracts due to changes in cell structure, with anterior cataracts due to incomplete suture closure and a cortical ring cataract corresponding to a zone of compaction in cortical lens fiber cells. Refractive index measurements showed a rapid growth in peak refractive index between 1 to 6 months of age, and the area of highest refractive index is correlated with increases in lens nucleus size with age. These data provide a comprehensive overview of age-related changes in murine lenses, including lens size, stiffness, nuclear fraction, refractive index, transparency, capsule thickness and cell structure. Our results suggest similarities between murine and primate lenses and provide a baseline for future lens aging studies.


Subject(s)
Aging/pathology , Lens, Crystalline/ultrastructure , Aging/physiology , Animals , Biomechanical Phenomena , Cataract/etiology , Female , Lens, Crystalline/physiology , Male , Mice, Inbred C57BL , Refraction, Ocular
8.
Brain ; 141(2): 422-458, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29360998

ABSTRACT

The mechanisms underpinning concussion, traumatic brain injury, and chronic traumatic encephalopathy, and the relationships between these disorders, are poorly understood. We examined post-mortem brains from teenage athletes in the acute-subacute period after mild closed-head impact injury and found astrocytosis, myelinated axonopathy, microvascular injury, perivascular neuroinflammation, and phosphorylated tau protein pathology. To investigate causal mechanisms, we developed a mouse model of lateral closed-head impact injury that uses momentum transfer to induce traumatic head acceleration. Unanaesthetized mice subjected to unilateral impact exhibited abrupt onset, transient course, and rapid resolution of a concussion-like syndrome characterized by altered arousal, contralateral hemiparesis, truncal ataxia, locomotor and balance impairments, and neurobehavioural deficits. Experimental impact injury was associated with axonopathy, blood-brain barrier disruption, astrocytosis, microgliosis (with activation of triggering receptor expressed on myeloid cells, TREM2), monocyte infiltration, and phosphorylated tauopathy in cerebral cortex ipsilateral and subjacent to impact. Phosphorylated tauopathy was detected in ipsilateral axons by 24 h, bilateral axons and soma by 2 weeks, and distant cortex bilaterally at 5.5 months post-injury. Impact pathologies co-localized with serum albumin extravasation in the brain that was diagnostically detectable in living mice by dynamic contrast-enhanced MRI. These pathologies were also accompanied by early, persistent, and bilateral impairment in axonal conduction velocity in the hippocampus and defective long-term potentiation of synaptic neurotransmission in the medial prefrontal cortex, brain regions distant from acute brain injury. Surprisingly, acute neurobehavioural deficits at the time of injury did not correlate with blood-brain barrier disruption, microgliosis, neuroinflammation, phosphorylated tauopathy, or electrophysiological dysfunction. Furthermore, concussion-like deficits were observed after impact injury, but not after blast exposure under experimental conditions matched for head kinematics. Computational modelling showed that impact injury generated focal point loading on the head and seven-fold greater peak shear stress in the brain compared to blast exposure. Moreover, intracerebral shear stress peaked before onset of gross head motion. By comparison, blast induced distributed force loading on the head and diffuse, lower magnitude shear stress in the brain. We conclude that force loading mechanics at the time of injury shape acute neurobehavioural responses, structural brain damage, and neuropathological sequelae triggered by neurotrauma. These results indicate that closed-head impact injuries, independent of concussive signs, can induce traumatic brain injury as well as early pathologies and functional sequelae associated with chronic traumatic encephalopathy. These results also shed light on the origins of concussion and relationship to traumatic brain injury and its aftermath.awx350media15713427811001.


Subject(s)
Athletic Injuries/complications , Brain Concussion/etiology , Craniocerebral Trauma/complications , Craniocerebral Trauma/etiology , Tauopathies/etiology , Vascular System Injuries/etiology , Action Potentials/physiology , Adolescent , Animals , Athletes , Brain/pathology , Calcium-Binding Proteins , Cohort Studies , Computer Simulation , Craniocerebral Trauma/diagnostic imaging , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/physiology , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Models, Neurological , Prefrontal Cortex/physiopathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Young Adult
9.
FASEB J ; 29(6): 2628-39, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25757569

ABSTRACT

FE65 and FE65L1 are cytoplasmic adaptor proteins that bind a variety of proteins, including the amyloid precursor protein, and that mediate the assembly of multimolecular complexes. We previously reported that FE65/FE65L1 double knockout (DKO) mice display disorganized laminin in meningeal fibroblasts and a cobblestone lissencephaly-like phenotype in the developing cortex. Here, we examined whether loss of FE65 and FE65L1 causes ocular and muscular deficits, 2 phenotypes that frequently accompany cobblestone lissencephaly. Eyes of FE65/FE65L1 DKO mice develop normally, but lens degeneration becomes apparent in young adult mice. Abnormal lens epithelial cell migration, widespread small vacuole formation, and increased laminin expression underneath lens capsules suggest impaired interaction between epithelial cells and capsular extracellular matrix in DKO lenses. Cortical cataracts develop in FE65L1 knockout (KO) mice aged 16 months or more but are absent in wild-type or FE65 KO mice. FE65 family KO mice show attenuated grip strength, and the nuclei of DKO muscle cells frequently locate in the middle of muscle fibers. These findings reveal that FE65 and FE65L1 are essential for the maintenance of lens transparency, and their loss produce phenotypes in brain, eye, and muscle that are comparable to the clinical features of congenital muscular dystrophies in humans.


Subject(s)
Carrier Proteins/genetics , Cataract/genetics , Muscle Weakness/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis , Blotting, Western , Carrier Proteins/metabolism , Cataract/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunohistochemistry , Laminin/metabolism , Lens Capsule, Crystalline/metabolism , Lens Capsule, Crystalline/pathology , Lens Diseases/genetics , Lens Diseases/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle Weakness/metabolism , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Nerve Tissue Proteins/deficiency , Nuclear Proteins/deficiency
10.
PLoS One ; 7(9): e43728, 2012.
Article in English | MEDLINE | ID: mdl-22984439

ABSTRACT

Multiple lines of evidence suggest that specific subtypes of age-related cataract (ARC) and Alzheimer disease (AD) are related etiologically. To identify shared genetic factors for ARC and AD, we estimated co-heritability of quantitative measures of cataract subtypes with AD-related brain MRI traits among 1,249 members of the Framingham Eye Study who had a brain MRI scan approximately ten years after the eye exam. Cortical cataract (CC) was found to be co-heritable with future development of AD and with several MRI traits, especially temporal horn volume (THV, ρ = 0.24, P<10(-4)). A genome-wide association study using 187,657 single nucleotide polymorphisms (SNPs) for the bivariate outcome of CC and THV identified genome-wide significant association with CTNND2 SNPs rs17183619, rs13155993 and rs13170756 (P<2.6 × 10(-7)). These SNPs were also significantly associated with bivariate outcomes of CC and scores on several highly heritable neuropsychological tests (5.7 × 10(-9) ≤ P<3.7 × 10(-6)). Statistical interaction was demonstrated between rs17183619 and APP SNP rs2096488 on CC (P = 0.0015) and CC-THV (P = 0.038). A rare CTNND2 missense mutation (G810R) 249 base pairs from rs17183619 altered δ-catenin localization and increased secreted amyloid-ß(1-42) in neuronal cell culture. Immunohistopathological analysis of lens tissue obtained from two autopsy-confirmed AD subjects and two non-AD controls revealed elevated expression of δ-catenin in epithelial and cortical regions of lenses from AD subjects compared to controls. Our findings suggest that genetic variation in delta catenin may underlie both cortical lens opacities in mid-life and subsequent MRI and cognitive changes that presage the development of AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/pathology , Brain/physiopathology , Cataract/genetics , Catenins/genetics , Catenins/metabolism , Aged , Alzheimer Disease/complications , Amyloid beta-Peptides/genetics , Base Pairing/genetics , Brain/metabolism , Case-Control Studies , Cataract/complications , Cataract/pathology , Computational Biology , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , HEK293 Cells , Humans , Immunohistochemistry , Lateral Ventricles/pathology , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Male , Mutation/genetics , Neuropsychological Tests , Organ Size , Polymorphism, Single Nucleotide/genetics , Delta Catenin
11.
Sci Transl Med ; 4(134): 134ra60, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22593173

ABSTRACT

Blast exposure is associated with traumatic brain injury (TBI), neuropsychiatric symptoms, and long-term cognitive disability. We examined a case series of postmortem brains from U.S. military veterans exposed to blast and/or concussive injury. We found evidence of chronic traumatic encephalopathy (CTE), a tau protein-linked neurodegenerative disease, that was similar to the CTE neuropathology observed in young amateur American football players and a professional wrestler with histories of concussive injuries. We developed a blast neurotrauma mouse model that recapitulated CTE-linked neuropathology in wild-type C57BL/6 mice 2 weeks after exposure to a single blast. Blast-exposed mice demonstrated phosphorylated tauopathy, myelinated axonopathy, microvasculopathy, chronic neuroinflammation, and neurodegeneration in the absence of macroscopic tissue damage or hemorrhage. Blast exposure induced persistent hippocampal-dependent learning and memory deficits that persisted for at least 1 month and correlated with impaired axonal conduction and defective activity-dependent long-term potentiation of synaptic transmission. Intracerebral pressure recordings demonstrated that shock waves traversed the mouse brain with minimal change and without thoracic contributions. Kinematic analysis revealed blast-induced head oscillation at accelerations sufficient to cause brain injury. Head immobilization during blast exposure prevented blast-induced learning and memory deficits. The contribution of blast wind to injurious head acceleration may be a primary injury mechanism leading to blast-related TBI and CTE. These results identify common pathogenic determinants leading to CTE in blast-exposed military veterans and head-injured athletes and additionally provide mechanistic evidence linking blast exposure to persistent impairments in neurophysiological function, learning, and memory.


Subject(s)
Blast Injuries/complications , Blast Injuries/pathology , Brain Injury, Chronic/complications , Brain Injury, Chronic/pathology , Military Personnel/psychology , Veterans/psychology , Acceleration , Adolescent , Adult , Animals , Athletes , Axons/pathology , Behavior, Animal , Blast Injuries/physiopathology , Brain Concussion/complications , Brain Concussion/pathology , Brain Concussion/physiopathology , Brain Injury, Chronic/physiopathology , Disease Models, Animal , Head/pathology , Head/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/ultrastructure , Humans , Intracranial Pressure , Long-Term Potentiation , Male , Mice , Middle Aged , Phosphorylation , Postmortem Changes , Synaptic Transmission , Young Adult , tau Proteins/metabolism
12.
J Biol Chem ; 286(38): 33203-12, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21768092

ABSTRACT

The 15-kDa selenoprotein (Sep15) is a thioredoxin-like, endoplasmic reticulum-resident protein involved in the quality control of glycoprotein folding through its interaction with UDP-glucose:glycoprotein glucosyltransferase. Expression of Sep15 is regulated by dietary selenium and the unfolded protein response, but its specific function is not known. In this study, we developed and characterized Sep15 KO mice by targeted removal of exon 2 of the Sep15 gene coding for the cysteine-rich UDP-glucose:glycoprotein glucosyltransferase-binding domain. These KO mice synthesized a mutant mRNA, but the shortened protein product could be detected neither in tissues nor in Sep15 KO embryonic fibroblasts. Sep15 KO mice were viable and fertile, showed normal brain morphology, and did not activate endoplasmic reticulum stress pathways. However, parameters of oxidative stress were elevated in the livers of these mice. We found that Sep15 mRNA was enriched during lens development. Further phenotypic characterization of Sep15 KO mice revealed a prominent nuclear cataract that developed at an early age. These cataracts did not appear to be associated with severe oxidative stress or glucose dysregulation. We suggest that the cataracts resulted from an improper folding status of lens proteins caused by Sep15 deficiency.


Subject(s)
Cataract/metabolism , Cataract/pathology , Homeostasis , Selenoproteins/deficiency , Selenoproteins/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Brain/pathology , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Male , Mice , Mice, Knockout , Molecular Sequence Data , Molecular Weight , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , NIH 3T3 Cells , Oxidation-Reduction , Oxidative Stress , Prostate/metabolism , Prostate/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Selenoprotein P/metabolism , Selenoproteins/chemistry , Selenoproteins/genetics , Unfolded Protein Response
13.
PLoS One ; 6(6): e19638, 2011.
Article in English | MEDLINE | ID: mdl-21698273

ABSTRACT

BACKGROUND AND AIMS: Recent work has suggested that Zn(2+) plays a critical role in regulating acidity within the secretory compartments of isolated gastric glands. Here, we investigate the content, distribution and demand for Zn(2+) in gastric mucosa under baseline conditions and its regulation during secretory stimulation. METHODS AND FINDINGS: Content and distribution of zinc were evaluated in sections of whole gastric mucosa using X-ray fluorescence microscopy. Significant stores of Zn(2+) were identified in neural elements of the muscularis, glandular areas enriched in parietal cells, and apical regions of the surface epithelium. In in vivo studies, extraction of the low abundance isotope, (70)Zn(2+), from the circulation was demonstrated in samples of mucosal tissue 24 hours or 72 hours after infusion (250 µg/kg). In in vitro studies, uptake of (70)Zn(2+) from media was demonstrated in isolated rabbit gastric glands following exposure to concentrations as low as 10 nM. In additional studies, demand of individual gastric parietal cells for Zn(2+) was monitored using the fluorescent zinc reporter, fluozin-3, by measuring increases in free intracellular concentrations of Zn(2+) {[Zn(2+)](i)} during exposure to standard extracellular concentrations of Zn(2+) (10 µM) for standard intervals of time. Under resting conditions, demand for extracellular Zn(2+) increased with exposure to secretagogues (forskolin, carbachol/histamine) and under conditions associated with increased intracellular Ca(2+) {[Ca(2+)](i)}. Uptake of Zn(2+) was abolished following removal of extracellular Ca(2+) or depletion of intracellular Ca(2+) stores, suggesting that demand for extracellular Zn(2+) increases and depends on influx of extracellular Ca(2+). CONCLUSIONS: This study is the first to characterize the content and distribution of Zn(2+) in an organ of the gastrointestinal tract. Our findings offer the novel interpretation, that Ca(2+) integrates basolateral demand for Zn(2+) with stimulation of secretion of HCl into the lumen of the gastric gland. Similar connections may be detectable in other secretory cells and tissues.


Subject(s)
Calcium/metabolism , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Zinc/metabolism , Animals , Fluorescent Dyes , Mass Spectrometry , Microscopy, Fluorescence , Polycyclic Compounds , Rabbits
14.
PLoS One ; 5(5): e10659, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20502642

ABSTRACT

Down syndrome (DS, trisomy 21) is the most common chromosomal disorder and the leading genetic cause of intellectual disability in humans. In DS, triplication of chromosome 21 invariably includes the APP gene (21q21) encoding the Alzheimer's disease (AD) amyloid precursor protein (APP). Triplication of the APP gene accelerates APP expression leading to cerebral accumulation of APP-derived amyloid-beta peptides (Abeta), early-onset AD neuropathology, and age-dependent cognitive sequelae. The DS phenotype complex also includes distinctive early-onset cerulean cataracts of unknown etiology. Previously, we reported increased Abeta accumulation, co-localizing amyloid pathology, and disease-linked supranuclear cataracts in the ocular lenses of subjects with AD. Here, we investigate the hypothesis that related AD-linked Abeta pathology underlies the distinctive lens phenotype associated with DS. Ophthalmological examinations of DS subjects were correlated with phenotypic, histochemical, and biochemical analyses of lenses obtained from DS, AD, and normal control subjects. Evaluation of DS lenses revealed a characteristic pattern of supranuclear opacification accompanied by accelerated supranuclear Abeta accumulation, co-localizing amyloid pathology, and fiber cell cytoplasmic Abeta aggregates (approximately 5 to 50 nm) identical to the lens pathology identified in AD. Peptide sequencing, immunoblot analysis, and ELISA confirmed the identity and increased accumulation of Abeta in DS lenses. Incubation of synthetic Abeta with human lens protein promoted protein aggregation, amyloid formation, and light scattering that recapitulated the molecular pathology and clinical features observed in DS lenses. These results establish the genetic etiology of the distinctive lens phenotype in DS and identify the molecular origin and pathogenic mechanism by which lens pathology is expressed in this common chromosomal disorder. Moreover, these findings confirm increased Abeta accumulation as a key pathogenic determinant linking lens and brain pathology in both DS and AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Down Syndrome/pathology , Lens, Crystalline/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Brain/ultrastructure , Cataract/pathology , Child , Child, Preschool , Down Syndrome/metabolism , Eye Proteins/chemistry , Eye Proteins/metabolism , Female , Humans , Lens, Crystalline/ultrastructure , Light , Male , Middle Aged , Molecular Sequence Data , Protein Structure, Quaternary , Scattering, Radiation , Young Adult
15.
Clin Ophthalmol ; 3: 235-42, 2009.
Article in English | MEDLINE | ID: mdl-19668572

ABSTRACT

PURPOSE: Interleukin-1beta (IL-1beta) and S100B calcium binding protein B (S100B) have been implicated in the pathogenesis of Alzheimer's disease. Both are present in and around senile plaques and have been shown to increase levels of amyloid precursor protein (APP) mRNA in vitro. However, it is not known how either of these substances affects APP in vivo. METHODS: We have studied the effects of IL-1beta and S100B on the expression and processing of APP using a retinal-vitreal model. We have also investigated the effect of amyloid beta peptide (Abeta) on APP in the same system and the regulation of S100B production by Abeta and IL-1beta from retinal glial cells. RESULTS: Retinal ganglion cells constitutively express APP. However, after intravitreal injection of IL-1beta or Abeta there was a marked reduction in APP levels as detected by Western blotting and IL-1beta produced a decrease in APP immunoreactivity (IR). Nissl staining showed that the integrity of the injected retinas was unchanged after injection. Two days after S100B injection, there was a small reduction in APP-IR but this was accompanied by the appearance of some intensely stained large ganglion cells and there was some up-regulation in APP holoprotein levels on Western blot. Seven days post-S100B injection, these large, highly stained cells had increased in number throughout the retina. Injection of Abeta and IL-1beta also caused an increase in S100B production within the retinal Müller glial cells. CONCLUSION: These results support the hypothesis that S100B (a glial-derived neurotrophic factor) and IL-1beta (a pro-inflammatory cytokine) can modulate the expression and processing of APP in vivo and so may contribute to the progression of Alzheimer's disease.

16.
Neuroreport ; 14(17): 2271-4, 2003 Dec 02.
Article in English | MEDLINE | ID: mdl-14625461

ABSTRACT

Using the novel rat retinal-vitreal model we have investigated the effect of metabotropic glutamate receptor activation on amyloid precursor protein (APP) metabolism. The release of low mol. wt fragments of APP, at 15-23 kDa in particular, was markedly up-regulated by the metabotropic glutamate receptor agonist (1S,3R)-1-amino-1,3-cyclopentane dicarboxylic acid ((1S,3R)-ACPD) in a concentration- and time-dependent manner, and this response was blocked by the receptor antagonist (S)-alpha-methyl-4-caboxyphenylglycine ((S)-MCPG). These results, together with the observation of a lack of deleterious effects of (1S,3R)-ACPD on the retinal neurons, support a physiological role of metabotropic glutamate receptors in mediating the release of soluble APP fragments, an action which may have important functional and therapeutic implications for Alzheimer's disease.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/physiology , Retina/metabolism , Up-Regulation/physiology , Amyloid beta-Protein Precursor/agonists , Amyloid beta-Protein Precursor/biosynthesis , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Retina/drug effects , Up-Regulation/drug effects
17.
Free Radic Res ; 37(1): 91-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12653222

ABSTRACT

Injection of the glutamate agonist N-methyl-D-aspartate (NMDA) into the vitreous body of rats resulted in severe degeneration of neurons in the retina, with a loss of 81% of ganglion cells and 43% of non-ganglion cells. The cocktail EM-X is a novel antioxidant drink derived from ferment of unpolished rice, papaya and sea-weeds with effective microorganisms (EM-X). In animals treated with an intraperitoneal injection of EM-X, the loss of ganglion cells was reduced to 55% and that of non-ganglion cells to 34% when compared to untreated NMDA-injected retinas. Cell degeneration resulting from NMDA excitotoxicity, is thought to be mediated via oxidative stress mechanisms. The neuroprotective effect of the EM-X in this system is therefore likely to be due, at least in part, to its flavonoids, saponins, vitamin E and ascorbic content.


Subject(s)
Antioxidants/administration & dosage , N-Methylaspartate/toxicity , Probiotics/administration & dosage , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Animals , Female , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Ganglion Cells/pathology
18.
Neurosci Lett ; 328(1): 55-9, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12123858

ABSTRACT

Injection of the glutamate agonist N-methyl-D-aspartate into the vitreous body of the rat eye resulted in a number of morphological changes in the retina. Most apparent was a dramatic reduction in the density and sizes of neurons accompanied by a decrease in amyloid precursor protein and glial fibrillary acidic protein immunoreactivity. Cell counts revealed that 81% of ganglion cells and 43% of non-ganglion cells were lost as a result of the treatment. However, in animals treated with the antioxidant ergothioneine, these figures dropped to 44 and 31%, respectively. Thus, ergothioneine appears to be neuroprotective in this system and the data suggest that antioxidants may provide a useful means of modulating glutamate-based toxicity.


Subject(s)
Antioxidants/pharmacology , Cell Death/drug effects , Ergothioneine/pharmacology , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Amyloid beta-Protein Precursor/drug effects , Amyloid beta-Protein Precursor/metabolism , Animals , Antioxidants/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Cell Count , Cell Death/physiology , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Ergothioneine/therapeutic use , Female , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , N-Methylaspartate/antagonists & inhibitors , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Neurotoxins/antagonists & inhibitors , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Retina/drug effects , Retina/metabolism , Retina/physiopathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...