Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mar Pollut Bull ; 131(Pt A): 580-588, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29886985

ABSTRACT

The effects of simulated electromagnetic fields (EMF), emitted from sub-sea power cables, on the commercially important decapod, edible crab (Cancer pagurus), were assessed. Stress related parameters were measured (l-Lactate, d-Glucose, Haemocyanin and respiration rate) along with behavioural and response parameters (antennular flicking, activity level, attraction/avoidance, shelter preference and time spent resting/roaming) during 24-h periods. Exposure to EMF had no effect on Haemocyanin concentrations, respiration rate, activity level or antennular flicking rate. EMF exposure significantly disrupted haemolymph l-Lactate and d-Glucose natural circadian rhythms. Crabs showed a clear attraction to EMF exposed shelter (69%) compared to control shelter (9%) and significantly reduced their time spent roaming by 21%. Consequently, EMF emitted from Marine Renewable Energy Devices (MREDs) will likely affect edible crabs both behaviourally and physiologically, suggesting that the impact of EMF on crustaceans must be considered when planning MREDs.


Subject(s)
Brachyura/physiology , Circadian Rhythm/physiology , Electromagnetic Fields/adverse effects , Renewable Energy/adverse effects , Animals , Glucose/metabolism , Hemocyanins/metabolism , Hemolymph/metabolism , Lactates/metabolism
2.
Hum Mol Genet ; 27(15): 2614-2627, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29741614

ABSTRACT

Loss of excitatory amino acid transporters (EAATs) has been implicated in a number of human diseases including spinocerebellar ataxias, Alzhiemer's disease and motor neuron disease. EAAT4 and GLAST/EAAT1 are the two predominant EAATs responsible for maintaining low extracellular glutamate levels and preventing neurotoxicity in the cerebellum, the brain region essential for motor control. Here using genetically modified mice we identify new critical roles for EAAT4 and GLAST/EAAT1 as modulators of Purkinje cell (PC) spontaneous firing patterns. We show high EAAT4 levels, by limiting mGluR1 signalling, are essential in constraining inherently heterogeneous firing of zebrin-positive PCs. Moreover mGluR1 antagonists were found to restore regular spontaneous PC activity and motor behaviour in EAAT4 knockout mice. In contrast, GLAST/EAAT1 expression is required to sustain normal spontaneous simple spike activity in low EAAT4 expressing (zebrin-negative) PCs by restricting NMDA receptor activation. Blockade of NMDA receptor activity restores spontaneous activity in zebrin-negative PCs of GLAST knockout mice and furthermore alleviates motor deficits. In addition both transporters have differential effects on PC survival, with zebrin-negative PCs more vulnerable to loss of GLAST/EAAT1 and zebrin-positive PCs more vulnerable to loss of EAAT4. These findings reveal that glutamate transporter dysfunction through elevated extracellular glutamate and the aberrant activation of extrasynaptic receptors can disrupt cerebellar output by altering spontaneous PC firing. This expands our understanding of disease mechanisms in cerebellar ataxias and establishes EAATs as targets for restoring homeostasis in a variety of neurological diseases where altered cerebellar output is now thought to play a key role in pathogenesis.


Subject(s)
Cerebellum/metabolism , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 4/genetics , Purkinje Cells/physiology , Animals , Ataxia/genetics , Cell Survival/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 4/metabolism , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Purkinje Cells/cytology , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Hum Mol Genet ; 25(20): 4448-4461, 2016 10 15.
Article in English | MEDLINE | ID: mdl-28173092

ABSTRACT

Clinical phenotypes of spinocerebellar ataxia type-5 (SCA5) and spectrin-associated autosomal recessive cerebellar ataxia type-1 (SPARCA1) are mirrored in mice lacking ß-III spectrin (ß-III-/-). One function of ß-III spectrin is the stabilization of the Purkinje cell-specific glutamate transporter EAAT4 at the plasma membrane. In ß-III-/- mice EAAT4 levels are reduced from an early age. In contrast levels of the predominant cerebellar glutamate transporter GLAST, expressed in Bergmann glia, only fall progressively from 3 months onwards. Here we elucidated the roles of these two glutamate transporters in cerebellar pathogenesis mediated through loss of ß-III spectrin function by studying EAAT4 and GLAST knockout mice as well as crosses of both with ß-III-/- mice. Our data demonstrate that EAAT4 loss, but not abnormal AMPA receptor composition, in young ß-III-/- mice underlies early Purkinje cell hyper-excitability and that subsequent loss of GLAST, superimposed on the earlier deficiency of EAAT4, is responsible for Purkinje cell loss and progression of motor deficits. Yet the loss of GLAST appears to be independent of EAAT4 loss, highlighting that other aspects of Purkinje cell dysfunction underpin the pathogenic loss of GLAST. Finally, our results demonstrate that Purkinje cells in the posterior cerebellum of ß-III-/- mice are most susceptible to the combined loss of EAAT4 and GLAST, with degeneration of proximal dendrites, the site of climbing fibre innervation, most pronounced. This highlights the necessity for efficient glutamate clearance from these regions and identifies dysregulation of glutamatergic neurotransmission particularly within the posterior cerebellum as a key mechanism in SCA5 and SPARCA1 pathogenesis.


Subject(s)
Cerebellar Ataxia/metabolism , Disease Models, Animal , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 4/metabolism , Purkinje Cells/metabolism , Spectrin/metabolism , Spinocerebellar Ataxias/metabolism , Animals , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Excitatory Amino Acid Transporter 1/physiology , Excitatory Amino Acid Transporter 4/physiology , Female , Male , Mice , Mice, Knockout , Phenotype , Purkinje Cells/pathology , Spectrin/physiology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology
4.
Hum Mol Genet ; 23(14): 3875-82, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24603075

ABSTRACT

Beta III spectrin is present throughout the elaborate dendritic tree of cerebellar Purkinje cells and is required for normal neuronal morphology and cell survival. Spinocerebellar ataxia type 5 (SCA5) and spectrin associated autosomal recessive cerebellar ataxia type 1 are human neurodegenerative diseases involving progressive gait ataxia and cerebellar atrophy. Both disorders appear to result from loss of ß-III spectrin function. Further elucidation of ß-III spectrin function is therefore needed to understand disease mechanisms and identify potential therapeutic options. Here, we report that ß-III spectrin is essential for the recruitment and maintenance of ankyrin R at the plasma membrane of Purkinje cell dendrites. Two SCA5-associated mutations of ß-III spectrin both reduce ankyrin R levels at the cell membrane. Moreover, a wild-type ß-III spectrin/ankyrin-R complex increases sodium channel levels and activity in cell culture, whereas mutant ß-III spectrin complexes fail to enhance sodium currents. This suggests impaired ability to form stable complexes between the adaptor protein ankyrin R and its interacting partners in the Purkinje cell dendritic tree is a key mechanism by which mutant forms of ß-III spectrin cause ataxia, initially by Purkinje cell dysfunction and exacerbated by subsequent cell death.


Subject(s)
Ankyrins/metabolism , Purkinje Cells/metabolism , Sodium Channels/physiology , Spectrin/genetics , Spectrin/metabolism , Spinocerebellar Ataxias/genetics , Animals , Cell Membrane/metabolism , Cells, Cultured , HEK293 Cells , Humans , Mice , Mutation , Protein Stability , Purkinje Cells/ultrastructure , Rats , Rats, Sprague-Dawley
5.
J Neurosci ; 31(46): 16581-90, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090485

ABSTRACT

Mutations in the gene encoding ß-III spectrin give rise to spinocerebellar ataxia type 5, a neurodegenerative disease characterized by progressive thinning of the molecular layer, loss of Purkinje cells and increasing motor deficits. A mouse lacking full-length ß-III spectrin (ß-III⁻/⁻) displays a similar phenotype. In vitro and in vivo analyses of Purkinje cells lacking ß-III spectrin, reveal a critical role for ß-III spectrin in Purkinje cell morphological development. Disruption of the normally well ordered dendritic arborization occurs in Purkinje cells from ß-III⁻/⁻ mice, specifically showing a loss of monoplanar organization, smaller average dendritic diameter and reduced densities of Purkinje cell spines and synapses. Early morphological defects appear to affect distribution of dendritic, but not axonal, proteins. This study confirms that thinning of the molecular layer associated with disease pathogenesis is a consequence of Purkinje cell dendritic degeneration, as Purkinje cells from 8-month-old ß-III⁻/⁻ mice have drastically reduced dendritic volumes, surface areas and total dendritic lengths compared with 5- to 6-week-old ß-III⁻/⁻ mice. These findings highlight a critical role of ß-III spectrin in dendritic biology and are consistent with an early developmental defect in ß-III⁻/⁻ mice, with abnormal Purkinje cell dendritic morphology potentially underlying disease pathogenesis.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Dendrites/ultrastructure , Dendritic Spines/metabolism , Purkinje Cells/cytology , Spectrin/metabolism , Age Factors , Animals , Animals, Newborn , Calbindins , Excitatory Amino Acid Transporter 4/metabolism , Gene Expression Regulation, Developmental/genetics , Glucose Transporter Type 2/metabolism , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Phosphate Transport Proteins/metabolism , S100 Calcium Binding Protein G/metabolism , Silver Staining/methods , Sodium Channels/metabolism , Spectrin/deficiency , Vesicular Glutamate Transport Protein 1/metabolism
6.
Brain Res ; 1361: 140-5, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-20849835

ABSTRACT

Cystatin C (CC) is a cysteine protease inhibitor abundantly expressed in the central nervous system. Bunina bodies, small eosinophilic intraneuronal inclusions, are stain positive for CC and are the most specific histological hallmark of amyotrophic lateral sclerosis (ALS). In this study, employing a latex turbidimetric immunoassay, levels of CC in cerebrospinal fluid (CSF) were quantified in 130 age-matched individuals with either a neurological disorder [ALS, Alzheimer's disease (AD), Parkinson's disease (PD), tauopathy (TP), multiple system atrophy (MSA), chronic inflammatory demyelinating polyneuropathy (CIDP)] or no known neurological condition (normal control, NC). The CC level in CSF was found to be correlated with the age during the investigation but not the protein concentration. There was no difference in CC levels between NC and ALS or CIDP cases, whereas CC levels were significantly lower in MSA compared with NC. Of the 130 cases, 96 were genotyped, and G/A or A/A polymorphism at +73 within the CST3 gene was found in 28 individuals. The CC level was significantly lower in the combined group of G/A and A/A genotypes compared with G/G. The present data demonstrate that the level of CC in CSF should not be considered as a biomarker of ALS, but there is a correlation between CC levels and the CST3 genotype.


Subject(s)
Cystatin C/cerebrospinal fluid , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/genetics , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Alanine , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/genetics , Biomarkers , Case-Control Studies , Cystatin C/blood , Cystatin C/genetics , Female , Glycine , Humans , Immunoassay/methods , Latex , Male , Middle Aged , Multiple System Atrophy/cerebrospinal fluid , Multiple System Atrophy/genetics , Nephelometry and Turbidimetry/methods , Nervous System Diseases/blood , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/genetics , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/cerebrospinal fluid , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/genetics , Tauopathies/cerebrospinal fluid , Tauopathies/genetics
7.
Hum Mol Genet ; 19(18): 3634-41, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20603325

ABSTRACT

Spinocerebellar ataxia type 5 (SCA5) is an autosomal dominant neurodegenerative disorder caused by mutations in beta-III spectrin. A mouse lacking full-length beta-III spectrin has a phenotype closely mirroring symptoms of SCA5 patients. Here we report the analysis of heterozygous animals, which show no signs of ataxia or cerebellar degeneration up to 2 years of age. This argues against haploinsufficiency as a disease mechanism and points towards human mutations having a dominant-negative effect on wild-type (WT) beta-III spectrin function. Cell culture studies using beta-III spectrin with a mutation associated with SCA5 (L253P) reveal that mutant protein, instead of being found at the cell membrane, appears trapped in the cytoplasm associated with the Golgi apparatus. Furthermore, L253P beta-III spectrin prevents correct localization of WT beta-III spectrin and prevents EAAT4, a protein known to interact with beta-III spectrin, from reaching the plasma membrane. Interaction of beta-III spectrin with Arp1, a subunit of the dynactin-dynein complex, is also lost with the L253P substitution. Despite intracellular accumulation of proteins, this cellular stress does not induce the unfolded protein response, implying the importance of membrane protein loss in disease pathogenesis. Incubation at lower temperature (25 degrees C) rescues L253P beta-III spectrin interaction with Arp1 and normal protein trafficking to the membrane. These data provide evidence for a dominant-negative effect of an SCA5 mutation and show for the first time that trafficking of both beta-III spectrin and EAAT4 from the Golgi is disrupted through failure of the L253P mutation to interact with Arp1.


Subject(s)
Golgi Apparatus/metabolism , Microfilament Proteins/metabolism , Mutation, Missense , Spectrin/genetics , Spectrin/metabolism , Spinocerebellar Ataxias/metabolism , Animals , Disease Models, Animal , Female , Golgi Apparatus/genetics , Humans , Male , Mice , Mice, Knockout , Microfilament Proteins/genetics , Protein Binding , Protein Transport , Spinocerebellar Ataxias/genetics
8.
J Neurosci ; 30(14): 4857-67, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20371805

ABSTRACT

Mutations in SPTBN2, the gene encoding beta-III spectrin, cause spinocerebellar ataxia type 5 in humans (SCA5), a neurodegenerative disorder resulting in loss of motor coordination. How these mutations give rise to progressive ataxia and what the precise role beta-III spectrin plays in normal cerebellar physiology are unknown. We developed a mouse lacking full-length beta-III spectrin and found that homozygous mice reproduced features of SCA5 including gait abnormalities, tremor, deteriorating motor coordination, Purkinje cell loss, and cerebellar atrophy (molecular layer thinning). In vivo analysis reveals an age-related reduction in simple spike firing rate in surviving beta-III(-/-) Purkinje cells, whereas in vitro studies show these neurons to have reduced spontaneous firing, smaller sodium currents, and dysregulation of glutamatergic neurotransmission. Our data suggest an early loss of EAAT4- (protein interactor of beta-III spectrin) and a subsequent loss of GLAST-mediated uptake may play a role in neuronal pathology. These findings implicate a loss of beta-III spectrin function in SCA5 pathogenesis and indicate that there are at least two physiological effects of beta-III spectrin loss that underpin a progressive loss of inhibitory cerebellar output, namely an intrinsic Purkinje cell membrane defect due to reduced sodium currents and alterations in glutamate signaling.


Subject(s)
Motor Activity/genetics , Purkinje Cells/metabolism , Purkinje Cells/pathology , Spectrin/deficiency , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Action Potentials/genetics , Animals , Atrophy/genetics , Cerebellum/pathology , Gait/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Spectrin/genetics , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/physiopathology , Tremor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...