Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Int J Mol Sci ; 23(15)2022 Jul 24.
Article in English | MEDLINE | ID: mdl-35897728

ABSTRACT

Recent technological development requires new approaches to address the problem of blindness. Such approaches need to be able to ensure that no cells with photosensitive capability remain in the retina. The presented model, Opn4-/- × Pde6brd10/rd10 (O×Rd) double mutant murine, is a combination of a mutation in the Pde6b gene (photoreceptor degeneration) together with a deletion of the Opn4 gene (responsible for the expression of melanopsin in the intrinsically photosensitive retinal ganglion cells). This model has been characterized and compared with those of WT mice and murine animal models displaying both mutations separately. A total loss of pupillary reflex was observed. Likewise, behavioral tests demonstrated loss of rejection to illuminated spaces and a complete decrease in visual acuity (optomotor test). Functional recordings showed an absolute disappearance of various wave components of the full-field and pattern electroretinogram (fERG, pERG). Likewise, visual evoked potential (VEP) could not be recorded. Immunohistochemical staining showed marked degeneration of the outer retinal layers and the absence of melanopsin staining. The combination of both mutations has generated an animal model that does not show any photosensitive element in its retina. This model is a potential tool for the study of new ophthalmological approaches such as optosensitive agents.


Subject(s)
Evoked Potentials, Visual , Retinal Degeneration , Animals , Blindness , Evoked Potentials, Visual/genetics , Mice , Mice, Inbred C57BL , Models, Genetic , Phenotype , Retina/metabolism , Retinal Degeneration/metabolism
2.
PLoS One ; 16(5): e0242394, 2021.
Article in English | MEDLINE | ID: mdl-34048428

ABSTRACT

While albino mice are widely used in research which includes the use of visually guided behavioral tests, information on their visual capability is scarce. We compared the spatial resolution (acuity) of albino mice (BALB/c) with that of pigmented mice (C57BL/6J). We used a high-throughput pattern electroretinogram (PERG) and pattern visual evoked potential (PVEP) method for objective assessment of retinal and cortical acuity, as well as optomotor head-tracking response/ reflex (OMR). We found that PERG, PVEP, and OMR acuities of C57BL/6J mice were all in the range of 0.5-0.6 cycles/degree (cyc/deg). BALB/c mice had PERG and PVEP acuities in the range of 0.1-0.2 cyc/deg but were unresponsive to OMR stimulus. Results indicate that retinal and cortical acuity can be reliably determined with electrophysiological methods in BALB/c mice, although PERG/PVEP acuities are lower than those of C57BL/6J mice. The reduced acuity of BALB/c mice appears to be primarily determined at retinal level.


Subject(s)
Evoked Potentials, Visual/physiology , Retina/physiology , Vision, Ocular/physiology , Visual Acuity/physiology , Animals , Electroretinography , Evoked Potentials, Visual/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Retina/diagnostic imaging , Vision, Ocular/genetics , Visual Acuity/genetics , Visual Pathways
3.
J Neurosci ; 40(27): 5214-5227, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32467358

ABSTRACT

The limitation of plasticity in the adult brain impedes functional recovery later in life from brain injury or disease. This pressing clinical issue may be resolved by enhancing plasticity in the adult brain. One strategy for triggering robust plasticity in adulthood is to reproduce one of the hallmark physiological events of experience-dependent plasticity observed during the juvenile critical period: to rapidly reduce the activity of parvalbumin (PV)-expressing interneurons and disinhibit local excitatory neurons. This may be achieved through the enhancement of local inhibitory inputs, particularly those of somatostatin (SST)-expressing interneurons. However, to date the means for manipulating SST interneurons for enhancing cortical plasticity in the adult brain are not known. We show that SST interneuron-selective overexpression of Lypd6, an endogenous nicotinic signaling modulator, enhances ocular dominance plasticity in the adult primary visual cortex (V1). Lypd6 overexpression mediates a rapid experience-dependent increase in the visually evoked activity of SST interneurons as well as a simultaneous reduction in PV interneuron activity and disinhibition of excitatory neurons. Recapitulating this transient activation of SST interneurons using chemogenetics similarly enhanced V1 plasticity. Notably, we show that SST-selective Lypd6 overexpression restores visual acuity in amblyopic mice that underwent early long-term monocular deprivation. Our data in both male and female mice reveal selective modulation of SST interneurons and a putative downstream circuit mechanism as an effective method for enhancing experience-dependent cortical plasticity as well as functional recovery in adulthood.SIGNIFICANCE STATEMENT The decline of cortical plasticity after closure of juvenile critical period consolidates neural circuits and behavior, but this limits functional recovery from brain diseases and dysfunctions in later life. Here we show that activation of cortical somatostatin (SST) interneurons by Lypd6, an endogenous modulator of nicotinic acetylcholine receptors, enhances experience-dependent plasticity and recovery from amblyopia in adulthood. This manipulation triggers rapid reduction of PV interneuron activity and disinhibition of excitatory neurons, which are known hallmarks of cortical plasticity during juvenile critical periods. Our study demonstrates modulation of SST interneurons by Lypd6 to achieve robust levels of cortical plasticity in the adult brain and may provide promising targets for restoring brain function in the event of brain trauma or disease.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , GPI-Linked Proteins/physiology , Interneurons/physiology , Neuronal Plasticity/physiology , Somatostatin/physiology , Visual Cortex/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Dominance, Ocular/genetics , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/physiology , Female , GPI-Linked Proteins/genetics , Immunohistochemistry , Male , Mice , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/genetics , Phosphatidylinositols/pharmacology , Receptors, Nicotinic/genetics , Recovery of Function/genetics , Vision, Monocular/genetics , Vision, Monocular/physiology , Visual Acuity/genetics
4.
Neurotherapeutics ; 17(3): 1300-1310, 2020 07.
Article in English | MEDLINE | ID: mdl-32462407

ABSTRACT

There is a lack of reliable, repeatable, and non-invasive clinical endpoints when investigating treatments for intellectual disability (ID). The aim of this study is to explore a novel approach towards developing new endpoints for neurodevelopmental disorders, in this case for ARID1B-related ID. In this study, twelve subjects with ARID1B-related ID and twelve age-matched controls were included in this observational case-control study. Subjects performed a battery of non-invasive neurobehavioral and neurophysiological assessments on two study days. Test domains included cognition, executive functioning, and eye tracking. Furthermore, several electrophysiological assessments were performed. Subjects wore a smartwatch (Withings® Steel HR) for 6 days. Tests were systematically assessed regarding tolerability, variability, repeatability, difference with control group, and correlation with traditional endpoints. Animal fluency, adaptive tracking, body sway, and smooth pursuit eye movements were assessed as fit-for-purpose regarding all criteria, while physical activity, heart rate, and sleep parameters show promise as well. The event-related potential waveform of the passive oddball and visual evoked potential tasks showed discriminatory ability, but EEG assessments were perceived as extremely burdensome. This approach successfully identified fit-for-purpose candidate endpoints for ARID1B-related ID and possibly for other neurodevelopmental disorders. Next, results could be replicated in different ID populations or the assessments could be included as exploratory endpoint in interventional trials in ARID1B-related ID.


Subject(s)
DNA-Binding Proteins/genetics , Endpoint Determination/methods , Genetic Variation/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Transcription Factors/genetics , Adolescent , Adult , Animals , Case-Control Studies , Child , Child, Preschool , Evoked Potentials, Visual/genetics , Executive Function/physiology , Female , Humans , Intellectual Disability/psychology , Male , Photic Stimulation/methods , Young Adult
5.
Curr Gene Ther ; 19(2): 134-138, 2019.
Article in English | MEDLINE | ID: mdl-31237206

ABSTRACT

BACKGROUND: During the first few trials of gene therapy for Leber's hereditary optic neuropathy performed by our group, the visual acuity of the patients increased gradually over several months, or even years. However, in the current round of gene therapy for Leber's hereditary optic neuropathy, we noted that the visual acuity of three patients increased rapidly, within a few days after treatment. CASE PRESENTATION: Three patients who were diagnosed with mitochondrial gene 11778 mutation (associated with a G-to-A transition at Mt-11778 in the ND4 subunit gene of complex I of mitochondrial DNA that changes an arginine to histidine at amino acid 340) by genetic diagnosis were followed up three times before gene therapy, which lasted for 1 year, without spontaneous improvement of vision. Visual acuity in one or both eyes of each of the three patients increased rapidly after the initial gene therapy treatment. CONCLUSION: We suspect that in some patients with Leber's hereditary optic neuropathy, a portion of the retinal ganglion cells might remain in a "dormant" state for a certain period of time; these may be activated, within an optimal timeframe, during gene therapy for Leber's hereditary optic neuropathy.


Subject(s)
DNA, Mitochondrial/genetics , Genetic Therapy , Optic Atrophy, Hereditary, Leber/therapy , Visual Acuity/genetics , Adolescent , Adult , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/physiology , Female , Humans , Male , Mutation/genetics , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/physiopathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Visual Fields/genetics , Young Adult
6.
Life Sci Alliance ; 2(1)2019 02.
Article in English | MEDLINE | ID: mdl-30760557

ABSTRACT

Apolipoprotein E (APOE) effects on brain function remain controversial. Removal of APOE not only impairs cognitive functions but also reduces neuritic amyloid plaques in mouse models of Alzheimer's disease (AD). Can APOE simultaneously protect and impair neural circuits? Here, we dissociated the role of APOE in AD versus aging to determine its effects on neuronal function and synaptic integrity. Using two-photon calcium imaging in awake mice to record visually evoked responses, we found that genetic removal of APOE improved neuronal responses in adult APP/PSEN1 mice (8-10 mo). These animals also exhibited fewer neuritic plaques with less surrounding synapse loss, fewer neuritic dystrophies, and reactive glia. Surprisingly, the lack of APOE in aged mice (18-20 mo), even in the absence of amyloid, disrupted visually evoked responses. These results suggest a dissociation in APOE's role in AD versus aging: APOE may be neurotoxic during early stages of amyloid deposition, although being neuroprotective in latter stages of aging.


Subject(s)
Aging/physiology , Apolipoproteins E/genetics , Regeneration/physiology , Visual Cortex/physiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloidosis , Animals , Disease Models, Animal , Evoked Potentials, Visual/genetics , Humans , Loss of Function Mutation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/metabolism , Neurons/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Synapses/metabolism
7.
Mult Scler Relat Disord ; 25: 73-76, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30053751

ABSTRACT

The coexistence of multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) in the same family is a rare event. We report a familial case originating from Sardinia of two siblings: one with NMOSD and one with MS. Human leukocyte antigen (HLA) typing showed that the two affected siblings were HLA-identical, sharing risk-increasing alleles, while a younger unaffected sister was haploidentical to her siblings but she also carried protective alleles. Our findings confirm the role of HLA in raising the risk to develop CNS inflammatory diseases and provide further knowledge on the relationship between NMOSD and MS.


Subject(s)
Family Health , Multiple Sclerosis , Neuromyelitis Optica , Adult , Brain/diagnostic imaging , Electroencephalography , Evoked Potentials, Visual/genetics , Female , Histocompatibility Antigens Class I/genetics , Humans , Italy/epidemiology , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/genetics , Multiple Sclerosis/physiopathology , Mutation/genetics , Neuromyelitis Optica/diagnostic imaging , Neuromyelitis Optica/genetics , Neuromyelitis Optica/physiopathology , Spinal Cord/diagnostic imaging
8.
J Alzheimers Dis ; 51(1): 21-6, 2016.
Article in English | MEDLINE | ID: mdl-26836173

ABSTRACT

Alzheimer's disease has been shown to affect vision in human patients and animal models. This may pose the risk of bias in behavior studies and therefore requires comprehensive investigation. We recorded electroretinography (ERG) under isoflurane anesthesia and visual evoked potentials (VEP) in awake amyloid expressing AßPPswe/PS1dE9 (AßPP/PS1) and wild-type littermate mice at a symptomatic age. The VEPs in response to patterned stimuli were normal in AßPP/PS1 mice. They also showed normal ERG amplitude but slightly shortened ERG latency in dark-adapted conditions. Our results indicate subtle changes in visual processing in aged male AßPP/PS1 mice specifically at a retinal level.


Subject(s)
Alzheimer Disease/physiopathology , Evoked Potentials, Visual/physiology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Contrast Sensitivity/genetics , Disease Models, Animal , Electroencephalography , Electroretinography , Evoked Potentials, Visual/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Photic Stimulation , Presenilin-1/genetics , Reaction Time/genetics
9.
Biol Psychol ; 114: 69-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26744236

ABSTRACT

Stimuli in simple oddball target detection paradigms cause evoked responses in brain potential. These responses are heritable traits, and potential endophenotypes for clinical phenotypes. These stimuli also cause responses in oscillatory activity, both evoked responses phase-locked to stimulus presentation and phase-independent induced responses. Here, we investigate whether phase-locked and phase-independent oscillatory responses are heritable traits. Oscillatory responses were examined in EEG recordings from 213 twin pairs (91 monozygotic and 122 dizygotic twins) performing a visual oddball task. After group Independent Component Analysis (group-ICA) and time-frequency decomposition, individual differences in evoked and induced oscillatory responses were compared between MZ and DZ twin pairs. Induced (phase-independent) oscillatory responses consistently showed the highest heritability (24-55%) compared to evoked (phase-locked) oscillatory responses and spectral energy, which revealed lower heritability at 1-35.6% and 4.5-32.3%, respectively. Since the phase-independent induced response encodes functional aspects of the brain response to target stimuli different from evoked responses, we conclude that the modulation of ongoing oscillatory activity may serve as an additional endophenotype for behavioral phenotypes and psychiatric genetics.


Subject(s)
Biological Clocks/genetics , Evoked Potentials, Visual/genetics , Task Performance and Analysis , Adolescent , Brain/physiology , Electroencephalography , Endophenotypes , Genetics, Behavioral , Humans , Individuality , Netherlands , Photic Stimulation/methods , Twins, Dizygotic/genetics
10.
Exp Neurol ; 274(Pt B): 145-55, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26277687

ABSTRACT

Proteoglycans play important roles in regulating the development and functions of the brain. They consist of a core protein and glycosaminoglycans, which are long sugar chains of repeating disaccharide units with sulfation. A recent study demonstrated that the sulfation pattern of chondroitin sulfate on proteoglycans contributes to regulation of the critical period of experience-dependent plasticity in the mouse visual cortex. In the present study, we investigated the role of keratan sulfate (KS), another glycosaminoglycan, in critical period plasticity in the mouse visual cortex. Immunohistochemical analyses demonstrated the presence of KS containing disaccharide units of N-acetylglucosamine (GlcNAc)-6-sulfate and nonsulfated galactose during the critical period, although KS containing disaccharide units of GlcNAc-6-sulfate and galactose-6-sulfate was already known to disappear before that period. The KS chains were distributed diffusely in the extracellular space and densely around the soma of a large population of excitatory and inhibitory neurons. Electron microscopic analysis revealed that the KS was localized within the perisynaptic spaces and dendrites but not in presynaptic sites. KS was mainly located on phosphacan. In mice deficient in GlcNAc-6-O-sulfotransferase 1, which is one of the enzymes necessary for the synthesis of KS chains, the expression of KS was one half that in wild-type mice. In the knockout mice, monocular deprivation during the critical period resulted in a depression of deprived-eye responses but failed to produce potentiation of nondeprived-eye responses. In addition, T-type Ca(2+) channel-dependent long-term potentiation (LTP), which occurs only during the critical period, was not observed. These results suggest that regulation by KS-phosphacan with a specific sulfation pattern is necessary for the generation of LTP and hence the potentiation of nondeprived-eye responses after monocular deprivation.


Subject(s)
Neuronal Plasticity/physiology , Neurons/physiology , Proteoglycans/metabolism , Sulfotransferases/genetics , Visual Cortex/growth & development , Age Factors , Animals , Animals, Newborn , Epitopes/metabolism , Evoked Potentials, Visual/genetics , Gene Expression Regulation/genetics , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/genetics , Neurons/ultrastructure , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Sensory Deprivation/physiology , Sulfotransferases/deficiency , Synaptophysin/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , Visual Cortex/cytology , Visual Cortex/metabolism , Carbohydrate Sulfotransferases
11.
Mol Ther ; 23(10): 1562-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26137852

ABSTRACT

Retinal disease is one of the most active areas of gene therapy, with clinical trials ongoing in the United States for five diseases. There are currently no treatments for patients with late-stage disease in which photoreceptors have been lost. Optogenetic gene therapies are in development, but, to date, have suffered from the low light sensitivity of microbial opsins, such as channelrhodopsin and halorhodopsin, and azobenzene-based photoswitches. Several groups have shown that photoreceptive G-protein-coupled receptors (GPCRs) can be expressed heterologously, and photoactivate endogenous Gi/o signaling. We hypothesized such a GPCR could increase sensitivity due to endogenous signal amplification. We targeted vertebrate rhodopsin to retinal ON-bipolar cells of blind rd1 mice and observed restoration of: (i) light responses in retinal explants, (ii) visually-evoked potentials in visual cortex in vivo, and (iii) two forms of visually-guided behavior: innate light avoidance and discrimination of temporal light patterns in the context of fear conditioning. Importantly, both the light responses of the retinal explants and the visually-guided behavior occurred reliably at light levels that were two to three orders of magnitude dimmer than required for channelrhodopsin. Thus, gene therapy with native light-gated GPCRs presents a novel approach to impart light sensitivity for visual restoration in a useful range of illumination.


Subject(s)
Optogenetics/methods , Rhodopsin/genetics , Vision, Ocular/genetics , Animals , Dependovirus/genetics , Ectopic Gene Expression , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/radiation effects , Genetic Therapy , Genetic Vectors/genetics , Light , Mice , Photic Stimulation , Retina/cytology , Retina/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Transduction, Genetic , Visual Perception
12.
Nat Commun ; 6: 6802, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25869033

ABSTRACT

The responses of visual cortical neurons to natural stimuli are both reliable and sparse. These properties require inhibition, yet the contribution of specific types of inhibitory neurons is not well understood. Here we demonstrate that optogenetic suppression of parvalbumin (PV)- but not somatostatin (SOM)-expressing interneurons reduces response reliability in the primary visual cortex of anaesthetized and awake mice. PV suppression leads to increases in the low firing rates and decreases in the high firing rates of cortical neurons, resulting in an overall reduction of the signal-to-noise ratio (SNR). In contrast, SOM suppression generally increases the overall firing rate for most neurons, without affecting the SNR. Further analysis reveals that PV, but not SOM, suppression impairs neural discrimination of natural stimuli. Together, these results reveal a critical role for PV interneurons in the formation of reliable visual cortical representations of natural stimuli.


Subject(s)
Evoked Potentials, Visual/genetics , Interneurons/metabolism , Neural Inhibition/genetics , Parvalbumins/genetics , Somatostatin/genetics , Visual Cortex/metabolism , Anesthesia , Animals , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interneurons/cytology , Mice , Mice, Transgenic , Optogenetics , Parvalbumins/metabolism , Photic Stimulation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Rhodopsins, Microbial/genetics , Rhodopsins, Microbial/metabolism , Signal-To-Noise Ratio , Somatostatin/metabolism , Synaptic Transmission/genetics , Visual Cortex/cytology , Wakefulness/physiology
13.
Neuron ; 86(1): 264-75, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25801703

ABSTRACT

Circadian rhythms control a variety of physiological processes, but whether they may also time brain development remains largely unknown. Here, we show that circadian clock genes control the onset of critical period plasticity in the neocortex. Within visual cortex of Clock-deficient mice, the emergence of circadian gene expression was dampened, and the maturation of inhibitory parvalbumin (PV) cell networks slowed. Loss of visual acuity in response to brief monocular deprivation was concomitantly delayed and rescued by direct enhancement of GABAergic transmission. Conditional deletion of Clock or Bmal1 only within PV cells recapitulated the results of total Clock-deficient mice. Unique downstream gene sets controlling synaptic events and cellular homeostasis for proper maturation and maintenance were found to be mis-regulated by Clock deletion specifically within PV cells. These data demonstrate a developmental role for circadian clock genes outside the suprachiasmatic nucleus, which may contribute mis-timed brain plasticity in associated mental disorders.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Nerve Net/physiology , Visual Cortex/cytology , Visual Cortex/growth & development , ARNTL Transcription Factors/genetics , Adenosine Triphosphatases/metabolism , Age Factors , Animals , Animals, Newborn , CLOCK Proteins/genetics , Evoked Potentials, Visual/genetics , Functional Laterality , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parvalbumins/genetics , Parvalbumins/metabolism , Period Circadian Proteins/metabolism , Sensory Deprivation/physiology , Visual Acuity/genetics , gamma-Aminobutyric Acid/metabolism
14.
J Hum Genet ; 60(3): 157-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25566755

ABSTRACT

A young Chinese male patient was identified as homozygous for Machado-Joseph disease (MJD)/spinocerebellar ataxia type 3. This patient had a 4-year medical history mainly presenting severe ataxia, abnormal eye movement and pyramidal signs. Magnetic resonance imaging of the brain and cervical spinal cord revealed no obvious abnormality despite the severe symptoms and the findings of an electromyogram. However, brainstem auditory evoked potential indicated peripheral impairment and visual evoked potential indicated central impairment of his visual pathways. Molecular diagnosis revealed the pattern of CAG repeat units of this patient was 71/71. This case demonstrates that homozygosity for MJD enhances the clinical severity of the disease, which suggests that genetic education is of great importance.


Subject(s)
Ataxin-3/genetics , Machado-Joseph Disease/genetics , Repressor Proteins/genetics , Trinucleotide Repeats/genetics , Asian People/genetics , Brain Stem/metabolism , Brain Stem/physiopathology , China , Consanguinity , DNA Mutational Analysis , Electromyography , Evoked Potentials, Auditory/genetics , Evoked Potentials, Auditory/physiology , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/physiology , Family Health , Female , Homozygote , Humans , Machado-Joseph Disease/pathology , Machado-Joseph Disease/physiopathology , Male , Middle Aged , Pedigree , Young Adult
15.
J Neuroimmunol ; 277(1-2): 176-85, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25468275

ABSTRACT

Apolipoprotein A1 (Apo A-I), the most abundant component of high-density lipoprotein (HDL), is an anti-inflammatory molecule, yet its potential role in the pathogenesis of multiple sclerosis (MS) has not been fully investigated. In this study, Western blot analyses of human plasma showed differential Apo A-I expression in healthy controls compared to MS patients. Further, primary progressive MS patients had less plasma Apo A-I than other forms of MS. Using experimental allergic encephalomyelitis (EAE) as a model for MS, Apo A-I deficient mice exhibited worse clinical disease and more neurodegeneration concurrent with increased levels of pro-inflammatory cytokines compared to wild-type animals. These data suggest that Apo A-I plays a role in the pathogenesis of EAE, a model for MS, creating the possibility for agents that increase Apo A-I levels as potential therapies for MS.


Subject(s)
Apolipoprotein A-I/genetics , Cytokines/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Adult , Animals , Apolipoprotein A-I/blood , Apolipoprotein A-I/deficiency , Case-Control Studies , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/physiology , Female , Fluoresceins , Freund's Adjuvant/toxicity , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/blood , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity
16.
Exp Neurol ; 261: 802-11, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25218043

ABSTRACT

Olfactomedin 2 (Olfm2) is a secretory glycoprotein belonging to the family of olfactomedin domain-containing proteins. A previous study has shown that a mutation in OLFM2 is associated with primary open angle glaucoma in Japanese patients. In the present study, we generated Olfm2 deficient mice by replacing the Olfm2 gene with the LacZ gene. The loss of Olfm2 resulted in no gross abnormalities. However, Olfm2 null mice showed reduced exploration, locomotion, olfactory sensitivity, abnormal motor coordination, and anxiety related behavior. The pattern of the Olfm2 gene expression was studied in the brain and eye using ß-galactosidase staining. In the brain, Olfm2 was mainly expressed in the olfactory bulb, cortex, piriform cortex, olfactory trabeculae, and inferior and superior colliculus. In the eye expression was detected mainly in retinal ganglion cells. In Olfm2 null mice, the amplitude of the first negative wave in the visual evoked potential test was significantly reduced as compared with wild-type littermates. Olfm2, similar to Olfm1, interacted with the GluR2 subunit of the AMPAR complexes and Olfm2 co-segregated with the AMPA receptor subunit GluR2 and other synaptic proteins in the synaptosomal membrane fraction upon biochemical fractionation of the adult mice cortex and retina. Immunoprecipitation from the synaptosomal membrane fraction of the Olfm2 null mouse brain cortex using the GluR2 antibody showed reduced levels of several components of the AMPAR complex in the immunoprecipitates including Olfm1, PSD95 and CNIH2. These results suggest that heterodimers of Olfm1 and Olfm2 interact with AMPAR more efficiently than Olfm2 homodimers and that Olfm2 plays a role in the organization of the AMPA receptor complexes.


Subject(s)
Extracellular Matrix Proteins/deficiency , Glycoproteins/deficiency , Movement Disorders/genetics , Olfaction Disorders/genetics , Receptors, AMPA/metabolism , Sequence Deletion/genetics , Vision Disorders/genetics , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Evoked Potentials, Visual/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Maze Learning/physiology , Mice , Mice, Knockout , Optic Nerve/pathology , Retina/metabolism , Vision Disorders/complications
17.
J Neurol ; 261(10): 1929-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25059390

ABSTRACT

Recessive mutations in GJC2, the gene-encoding connexin 47 (Cx47), cause Pelizaeus-Merzbacher-like disease type 1, a severe dysmyelinating disorder. One recessive mutation (p.Ile33Met) has been associated with a much milder phenotype--hereditary spastic paraplegia type 44. Here, we present evidence that a novel Arg98Leu mutation causes an even milder phenotype--a subclinical leukodystrophy. The Arg98Leu mutant forms gap junction plaques in HeLa cells comparable to wild-type Cx47, but electrical coupling was 20-fold lower in cell pairs expressing Arg98Leu than for cell pairs expressing wild-type Cx47. On the other hand, coupling between Cx47Arg98Leu and Cx43WT expressing cells did not show such reductions. Single channel conductance and normalized steady-state junctional conductance-junctional voltage (G(j)-V(j)) relations differed only slightly from those for wild-type Cx47. Our data suggest that the minimal phenotype in this patient results from a reduced efficiency of opening of Cx47 channels between oligodendrocyte and oligodendrocyte with preserved coupling between oligodendrocyte and astrocyte, and support a partial loss of function model for the mild Cx47 associated disease phenotypes.


Subject(s)
Connexins/genetics , Mutation/genetics , Pelizaeus-Merzbacher Disease/genetics , Adult , Arginine/genetics , Brain/pathology , Cell Line, Transformed , DNA Mutational Analysis , Electroencephalography , Evoked Potentials, Visual/genetics , Female , Green Fluorescent Proteins/genetics , Humans , Leucine/genetics , Magnetic Resonance Imaging , Membrane Potentials/genetics , Patch-Clamp Techniques , Pelizaeus-Merzbacher Disease/diagnosis , Transfection
18.
PLoS One ; 8(10): e76831, 2013.
Article in English | MEDLINE | ID: mdl-24130795

ABSTRACT

Mutations in PLA2G6 gene have variable phenotypic outcome including infantile neuroaxonal dystrophy, atypical neuroaxonal dystrophy, idiopathic neurodegeneration with brain iron accumulation and Karak syndrome. The cause of this phenotypic variation is so far unknown which impairs both genetic diagnosis and appropriate family counseling. We report detailed clinical, electrophysiological, neuroimaging, histologic, biochemical and genetic characterization of 11 patients, from 6 consanguineous families, who were followed for a period of up to 17 years. Cerebellar atrophy was constant and the earliest feature of the disease preceding brain iron accumulation, leading to the provisional diagnosis of a recessive progressive ataxia in these patients. Ultrastructural characterization of patients' muscle biopsies revealed focal accumulation of granular and membranous material possibly resulting from defective membrane homeostasis caused by disrupted PLA2G6 function. Enzyme studies in one of these muscle biopsies provided evidence for a relatively low mitochondrial content, which is compatible with the structural mitochondrial alterations seen by electron microscopy. Genetic characterization of 11 patients led to the identification of six underlying PLA2G6 gene mutations, five of which are novel. Importantly, by combining clinical and genetic data we have observed that while the phenotype of neurodegeneration associated with PLA2G6 mutations is variable in this cohort of patients belonging to the same ethnic background, it is partially influenced by the genotype, considering the age at onset and the functional disability criteria. Molecular testing for PLA2G6 mutations is, therefore, indicated in childhood-onset ataxia syndromes, if neuroimaging shows cerebellar atrophy with or without evidence of iron accumulation.


Subject(s)
Group VI Phospholipases A2/genetics , Mutation , Phenotype , Adolescent , Adult , Arabs , Child , Child, Preschool , Consanguinity , Electroencephalography , Evoked Potentials, Visual/genetics , Female , Follow-Up Studies , Genotype , Humans , Infant , Male , Muscles/pathology , Muscles/physiopathology , Neural Conduction/genetics , Neuroaxonal Dystrophies/ethnology , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/pathology , Neuroaxonal Dystrophies/physiopathology , Neuroimaging , Pedigree , Young Adult
19.
J Neurosci ; 33(38): 15220-5, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048851

ABSTRACT

Ocular dominance plasticity is a widely studied model of experience-dependent cortical plasticity. It has been shown that potentiation of open eye responses resulting from monocular deprivation relies on a homeostatic response to loss of input from the closed eye, but the mechanisms by which this occurs are not fully understood. The role of GluA1 in the homeostatic component of ocular dominance (OD) plasticity has not so far been tested. In this study, we tested the idea that the GluA1 subunit of the AMPA receptor is necessary for open eye potentiation. We found that open eye potentiation did not occur in GluA1 knock-out (GluA1(-/-)) mice but did occur in wild-type littermates when monocular deprivation was imposed during the critical period. We also found that depression of the closed eye response that normally occurs in the monocular as well as binocular zone is delayed, but only in the monocular zone in GluA1(-/-) mice and only in a background strain we have previously shown lacks synaptic scaling (C57BL/6OlaHsd). In adult mice, we found that OD plasticity and facilitation of OD plasticity by prior monocular experience were both present in GluA1(-/-) mice, suggesting that the GluA1-dependent mechanisms only operate during the critical period.


Subject(s)
Dominance, Ocular/physiology , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Visual Cortex/physiology , Age Factors , Analysis of Variance , Animals , Electroencephalography , Electroretinography , Evoked Potentials, Visual/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Photic Stimulation , Receptors, AMPA/genetics , Sensory Deprivation , Visual Pathways/physiology
20.
PLoS One ; 8(7): e68616, 2013.
Article in English | MEDLINE | ID: mdl-23844226

ABSTRACT

We previously reported that aged mice lacking complement factor H (CFH) exhibit visual defects and structural changes in the retina. However, it is not known whether this phenotype is age-related or is the consequence of disturbed development. To address this question we investigated the effect of Cfh gene deletion on the retinal phenotype of young and mid-age mice. Cfh(-/-) mouse eyes exhibited thickening of the retina and reduced nuclear density, but relatively normal scotopic and photopic electroretinograms. At 12 months there was evidence of subtle astroglial activation in the Cfh(-/-) eyes, and significant elevation of the complement regulator, decay-accelerating factor (DAF) in Müller cells. In the retinal pigment epithelium (RPE) of young control and Cfh(-/-) animals mitochondria and melanosomes were oriented basally and apically respectively, whereas the apical positioning of melanosomes was significantly perturbed in the mid-age Cfh(-/-) RPE. We conclude that deletion of Cfh in the mouse leads to defects in the retina that precede any marked loss of visual function, but which become progressively more marked as the animals age. These observations are consistent with a lifelong role for CFH in retinal homeostasis.


Subject(s)
Aging , Complement Factor H/physiology , Retina/physiopathology , Visual Acuity/physiology , Animals , CD55 Antigens/metabolism , Complement Factor H/deficiency , Complement Factor H/genetics , Electroretinography , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/physiology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Melanosomes/metabolism , Melanosomes/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Photic Stimulation , Retina/metabolism , Retina/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Visual Acuity/genetics , Visual Pathways/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...