Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.034
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Brain Mapp ; 44(10): 4120-4135, 2023 07.
Article in English | MEDLINE | ID: mdl-37195035

ABSTRACT

Late-stage macular degeneration (MD) often causes retinal lesions depriving an individual of central vision, forcing them to learn to use peripheral vision for daily tasks. To compensate, many patients develop a preferred retinal locus (PRL), an area of peripheral vision used more often than equivalent regions of spared vision. Thus, associated portions of cortex experience increased use, while portions of cortex associated with the lesion are deprived of sensory input. Prior research has not well examined the degree to which structural plasticity depends on the amount of use across the visual field. Cortical thickness, neurite density, and orientation dispersion were measured at portions of cortex associated with the PRL, the retinal lesion, and a control region in participants with MD as well as age-matched, gender-matched, and education-matched controls. MD participants had significantly thinner cortex in both the cortical representation of the PRL (cPRL) and the control region, compared with controls, but no significant differences in thickness, neurite density, or orientation dispersion were found between the cPRL and the control region as functions of disease or onset. This decrease in thickness is driven by a subset of early-onset participants whose patterns of thickness, neurite density, and neurite orientation dispersion are distinct from matched control participants. These results suggest that people who develop MD earlier in adulthood may undergo more structural plasticity than those who develop it late in life.


Subject(s)
Macular Degeneration , Visual Cortex , Humans , Neurites/pathology , Visual Cortex/diagnostic imaging , Visual Cortex/pathology , Visual Perception , Visual Fields , Retina/pathology , Macular Degeneration/pathology
2.
Cereb Cortex ; 31(5): 2345-2363, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33338196

ABSTRACT

The functional output of a cortical region is shaped by its complement of GABA neuron subtypes. GABA-related transcript expression differs substantially between the primate dorsolateral prefrontal cortex (DLPFC) and primary visual (V1) cortices in gray matter homogenates, but the laminar and cellular bases for these differences are unknown. Quantification of levels of GABA-related transcripts in layers 2 and 4 of monkey DLPFC and V1 revealed three distinct expression patterns: 1) transcripts with higher levels in DLPFC and layer 2 [e.g., somatostatin (SST)]; 2) transcripts with higher levels in V1 and layer 4 [e.g., parvalbumin (PV)], and 3) transcripts with similar levels across layers and regions [e.g., glutamic acid decarboxylase (GAD67)]. At the cellular level, these patterns reflected transcript- and cell type-specific differences: the SST pattern primarily reflected differences in the relative proportions of SST mRNA-positive neurons, the PV pattern primarily reflected differences in PV mRNA expression per neuron, and the GAD67 pattern reflected opposed patterns in the relative proportions of GAD67 mRNA-positive neurons and in GAD67 mRNA expression per neuron. These findings suggest that differences in the complement of GABA neuron subtypes and in gene expression levels per neuron contribute to the specialization of inhibitory neurotransmission across cortical circuits.


Subject(s)
Dorsolateral Prefrontal Cortex/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Visual Cortex/metabolism , Animals , Dorsolateral Prefrontal Cortex/pathology , Glutamate Decarboxylase/metabolism , Gray Matter/metabolism , Gray Matter/pathology , Haplorhini , Parvalbumins/metabolism , Visual Cortex/pathology
3.
Neurobiol Dis ; 159: 105483, 2021 11.
Article in English | MEDLINE | ID: mdl-34400304

ABSTRACT

Mitochondrial diseases are among the most prevalent groups of inherited neurological disorders, affecting up to 1 in 5000 adults. Despite the progress achieved on the identification of gene mutations causing mitochondrial pathologies, they cannot be cured so far. Harlequin mice, a relevant model of mitochondrial pathology due to apoptosis inducing factor depletion, suffer from progressive disappearance of retinal ganglion cells leading to optic neuropathy. In our previous work, we showed that administering adeno-associated virus encompassing the coding sequences for neuroglobin, (a neuroprotective molecule belonging to the globin family) or apoptosis-inducing factor, before neurodegeneration onset, prevented retinal ganglion cell loss and preserved visual function. One of the challenges to develop an effective treatment for optic neuropathies is to consider that by the time patients become aware of their handicap, a large amount of nerve fibers has already disappeared. Gene therapy was performed in Harlequin mice aged between 4 and 5 months with either a neuroglobin or an apoptosis-inducing factor vector to determine whether the increased abundance of either one of these proteins in retinas could preserve visual function at this advanced stage of the disease. We demonstrated that gene therapy, by preserving the connectivity of transduced retinal ganglion cells and optic nerve bioenergetics, results in the enhancement of visual cortex activity, ultimately rescuing visual impairment. This study demonstrates that: (a) An increased abundance of neuroglobin functionally overcomes apoptosis-inducing factor absence in Harlequin mouse retinas at a late stage of neuronal degeneration; (b) The beneficial effect for visual function could be mediated by neuroglobin localization to the mitochondria, thus contributing to the maintenance of the organelle homeostasis.


Subject(s)
Apoptosis Inducing Factor/genetics , Electron Transport Complex IV/metabolism , Electron Transport Complex I/metabolism , Neuroglobin/genetics , Optic Atrophy/metabolism , Optic Nerve/metabolism , Retinal Ganglion Cells/metabolism , Visual Acuity/genetics , Visual Cortex/metabolism , Animals , Disease Progression , Genetic Therapy , Mice , Optic Atrophy/pathology , Optic Atrophy/physiopathology , Optic Nerve/pathology , Optic Nerve/physiopathology , Retinal Ganglion Cells/pathology , Visual Cortex/pathology , Visual Pathways
4.
Br J Haematol ; 192(1): 151-157, 2021 01.
Article in English | MEDLINE | ID: mdl-32789861

ABSTRACT

The visual system is primarily affected in sickle cell disease (SCD), and eye examination is recommended starting in late childhood. So far, to our knowledge, all studies have focused on the retina, neglecting the changes that might be present in the cortical portion of the visual system. We performed a multimodal magnetic resonance imaging (MRI) evaluation of the visual cortex in 25 children with SCD (mean age: 12·3 ± 1·9 years) and 31 controls (mean age: 12·7 ± 1·6 years). At ophthalmologic examination, 3/25 SCD children had mild visual acuity deficits and 2/25 had mild tortuosity of the retinal vessels. None showed optic pathway infarcts at MRI or Transcranial Doppler abnormal blood velocities, and 6/25 disclosed posterior cerebral artery stenosis (five mild and one severe) at MR-angiography. Compared to controls, SCD children had increased posterior pericalcarine cortical thickness, with a different trajectory of cortical maturation and decreased connectivity within medial and ventral visual neural networks. Our findings suggest that SCD affects the development and the tuning of the visual cortex, leading to anatomical and functional changes in childhood even in the absence of retinopathy, and set the basis for future studies to determine if these changes can represent useful predictors of visual impairment in adulthood, biomarkers of disease progression or treatment response.


Subject(s)
Anemia, Sickle Cell/pathology , Visual Cortex/pathology , Adolescent , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/diagnostic imaging , Brain/diagnostic imaging , Brain/pathology , Child , Female , Humans , Magnetic Resonance Imaging , Male , Nerve Net/diagnostic imaging , Nerve Net/pathology , Visual Cortex/diagnostic imaging , Visual Pathways/diagnostic imaging , Visual Pathways/pathology
5.
J Neuroinflammation ; 18(1): 177, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399786

ABSTRACT

BACKGROUND: Synucleinopathies are characterized by neurodegeneration and deposition of the presynaptic protein α-synuclein in pathological protein inclusions. Growing evidence suggests the complement system not only has physiological functions in the central nervous system, but also is involved in mediating the pathological loss of synapses in Alzheimer's disease. However, it is not established whether the complement system has a similar role in the diseases Parkinson's disease, Dementia with Lewy bodies, and multiple system atrophy (MSA) that are associated with α-synuclein aggregate pathology. METHODS: To investigate if the complement system has a pathological role in synucleinopathies, we assessed the effect of the complement system on the viability of an α-synuclein expressing cell model and examined direct activation of the complement system by α-synuclein in a plate-based activation assay. Finally, we investigated the levels of the initiator of the classical pathway, C1q, in postmortem brain samples from MSA patients. RESULTS: We demonstrate that α-synuclein activates the classical complement pathway and mediates complement-dependent toxicity in α-synuclein expressing SH-SY5Y cells. The α-synuclein-dependent cellular toxicity was rescued by the complement inhibitors RaCI (inhibiting C5) and Cp20 (inhibiting C3). Furthermore, we observed a trend for higher levels of C1q in the putamen of MSA subjects than that of controls. CONCLUSION: α-Synuclein can activate the classical complement pathway, and the complement system is involved in α-synuclein-dependent cellular cytotoxicity suggesting the system could play a prodegenerative role in synucleinopathies.


Subject(s)
Complement Pathway, Classical/physiology , Inclusion Bodies/metabolism , Visual Cortex/metabolism , alpha-Synuclein/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Inclusion Bodies/pathology , Male , Middle Aged , Visual Cortex/pathology
6.
Arch Biochem Biophys ; 701: 108815, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33609537

ABSTRACT

Glaucoma is a neurodegenerative disease that affects eye structures and brain areas related to the visual system. Oxidative stress plays a key role in the development and progression of the disease. The aims of the present study were to evaluate the mitochondrial function and its participation in the brain redox metabolism in an experimental glaucoma model. 3-month-old female Wistar rats were subjected to cauterization of two episcleral veins of the left eye to elevate the intraocular pressure. Seven days after surgery, animals were sacrificed, the brain was carefully removed and the primary visual cortex was dissected. Mitochondrial bioenergetics and ROS production, and the antioxidant enzyme defenses from both mitochondrial and cytosolic fractions were evaluated. When compared to control, glaucoma decreased mitochondrial ATP production (23%, p < 0.05), with an increase in superoxide and hydrogen peroxide production (30%, p < 0.01 and 28%, p < 0.05, respectively), whereas no changes were observed in membrane potential and oxygen consumption rate. In addition, the glaucoma group displayed a decrease in complex II activity (34%, p < 0.01). Moreover, NOX4 expression was increased in glaucoma compared to the control group (27%, p < 0.05). Regarding the activity of enzymes associated with the regulation of the redox status, glaucoma showed an increase in mitochondrial SOD activity (34%, p < 0.05), mostly due to an increase in Mn-SOD (50%, p < 0.05). A decrease in mitochondrial GST activity was observed (11%, p < 0.05). GR and TrxR activity were decreased in both mitochondrial (16%, p < 0.05 and 20%, p < 0.05 respectively) and cytosolic (21%, p < 0.01 and 50%, p < 0.01 respectively) fractions in the glaucoma group. Additionally, glaucoma showed an increase in cytoplasmatic GPx (50%, p < 0.01). In this scenario, redox imbalance took place resulting in damage to mitochondrial lipids (39%, p < 0.01) and proteins (70%, p < 0.05). These results suggest that glaucoma leads to mitochondrial function impairment in brain visual targets, that is accompanied by an alteration in both mitochondrial and cytoplasmatic enzymatic defenses. As a consequence of redox imbalance, oxidative damage to macromolecules takes place and can further affect vital cellular functions. Understanding the role of the mitochondria in the development and progression of the disease could bring up new neuroprotective therapies.


Subject(s)
Glaucoma/metabolism , Mitochondria/metabolism , Visual Cortex/metabolism , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Female , Glaucoma/pathology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , NADPH Oxidase 4/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Visual Cortex/pathology
7.
FASEB J ; 34(5): 6950-6964, 2020 05.
Article in English | MEDLINE | ID: mdl-32246809

ABSTRACT

Dipeptidyl peptidase-4 inhibitors (or gliptins), a class of antidiabetic drugs, have recently been shown to have protective actions in the central nervous system. Their cellular and molecular mechanisms responsible for these effects are largely unknown. In the present study, two structurally different gliptins, sitagliptin and vildagliptin, were examined for their therapeutic actions in a controlled cortical impact (CCI) model of moderate traumatic brain injury (TBI) in mice. Early post-CCI treatment with sitagliptin, but not vildagliptin, significantly reduced body asymmetry, locomotor hyperactivity, and brain lesion volume. Sitagliptin attenuated post-CCI microglial deramification in the ipsilateral dorsolateral (DL) striatum, while vildagliptin had no effect. Sitagliptin also reduced striatal expression of galectin-3 and monocyte chemoattractant protein 1(MCP-1), and increased the cortical and striatal levels of the anti-inflammatory cytokine IL-10 on the ipsilateral side. These data support a differential protective effect of sitagliptin against TBI, possibly mediated by an anti-inflammatory effect in striatum to preserve connective network. Both sitagliptin and vildagliptin produced similar increases of active glucagon-like peptide-1 (GLP-1) in blood and brain. Increasing active GLP-1 may not be the sole molecular mechanisms for the neurotherapeutic effect of sitagliptin in TBI.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Sensorimotor Cortex/drug effects , Sensorimotor Cortex/injuries , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Chemokine CCL2/metabolism , Disease Models, Animal , Galectin 3/metabolism , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Interleukin-10 , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Sensorimotor Cortex/pathology , Sitagliptin Phosphate/pharmacology , Vildagliptin/pharmacology , Visual Cortex/drug effects , Visual Cortex/pathology
8.
Brain ; 143(10): 2945-2956, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32968778

ABSTRACT

Migraine is the most common neurological disorder worldwide and it has been shown to have complex polygenic origins with a heritability of estimated 40-70%. Both common and rare genetic variants are believed to underlie the pathophysiology of the prevalent types of migraine, migraine with typical aura and migraine without aura. However, only common variants have been identified so far. Here we identify for the first time a gene module with rare mutations through a systems genetics approach integrating RNA sequencing data from brain and vascular tissues likely to be involved in migraine pathology in combination with whole genome sequencing of 117 migraine families. We found a gene module in the visual cortex, based on single nuclei RNA sequencing data, that had increased rare mutations in the migraine families and replicated this in a second independent cohort of 1930 patients. This module was mainly expressed by interneurons, pyramidal CA1, and pyramidal SS cells, and pathway analysis showed association with hormonal signalling (thyrotropin-releasing hormone receptor and oxytocin receptor signalling pathways), Alzheimer's disease pathway, serotonin receptor pathway and general heterotrimeric G-protein signalling pathways. Our results demonstrate that rare functional gene variants are strongly implicated in the pathophysiology of migraine. Furthermore, we anticipate that the results can be used to explain the critical mechanisms behind migraine and potentially improving the treatment regime for migraine patients.


Subject(s)
Databases, Genetic , Family , Gene Regulatory Networks/physiology , Genetic Variation/physiology , Migraine Disorders/genetics , Protein Interaction Maps/physiology , Cohort Studies , Databases, Genetic/trends , Humans , Migraine Disorders/diagnosis , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , Trigeminal Ganglion/pathology , Visual Cortex/pathology
9.
Cereb Cortex ; 30(4): 2030-2041, 2020 04 14.
Article in English | MEDLINE | ID: mdl-31799618

ABSTRACT

Hemianopia can be rehabilitated by an auditory-visual "training" procedure, which restores visual responsiveness in midbrain neurons indirectly compromised by the cortical lesion and reinstates vision in contralesional space. Presumably, these rehabilitative changes are induced via mechanisms of multisensory integration/plasticity. If so, the paradigm should fail if the stimulus configurations violate the spatiotemporal principles that govern these midbrain processes. To test this possibility, hemianopic cats were provided spatially or temporally noncongruent auditory-visual training. Rehabilitation failed in all cases even after approximately twice the number of training trials normally required for recovery, and even after animals learned to approach the location of the undetected visual stimulus. When training was repeated with these stimuli in spatiotemporal concordance, hemianopia was resolved. The results identify the conditions needed to engage changes in remaining neural circuits required to support vision in the absence of visual cortex, and have implications for rehabilitative strategies in human patients.


Subject(s)
Acoustic Stimulation/methods , Hemianopsia/physiopathology , Hemianopsia/rehabilitation , Photic Stimulation/methods , Visual Cortex/physiopathology , Animals , Cats , Female , Hemianopsia/pathology , Male , Visual Cortex/pathology
10.
Neuroimage ; 222: 117216, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32745677

ABSTRACT

Susceptibility weighted magnetic resonance imaging (MRI) is sensitive to the local concentration of iron and myelin. Here, we describe a robust image processing pipeline for quantitative susceptibility mapping (QSM) and R2* mapping of fixed post-mortem, whole-brain data. Using this pipeline, we compare the resulting quantitative maps in brains from patients with amyotrophic lateral sclerosis (ALS) and controls, with validation against iron and myelin histology. Twelve post-mortem brains were scanned with a multi-echo gradient echo sequence at 7T, from which susceptibility and R2* maps were generated. Semi-quantitative histological analysis for ferritin (the principal iron storage protein) and myelin proteolipid protein was performed in the primary motor, anterior cingulate and visual cortices. Magnetic susceptibility and R2* values in primary motor cortex were higher in ALS compared to control brains. Magnetic susceptibility and R2* showed positive correlations with both myelin and ferritin estimates from histology. Four out of nine ALS brains exhibited clearly visible hyperintense susceptibility and R2* values in the primary motor cortex. Our results demonstrate the potential for MRI-histology studies in whole, fixed post-mortem brains to investigate the biophysical source of susceptibility weighted MRI signals in neurodegenerative diseases like ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Ferritins , Magnetic Resonance Imaging/methods , Myelin Sheath , Aged , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Diagnosis , Female , Ferritins/metabolism , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Humans , Male , Middle Aged , Motor Cortex/diagnostic imaging , Motor Cortex/metabolism , Motor Cortex/pathology , Myelin Sheath/metabolism , Visual Cortex/diagnostic imaging , Visual Cortex/metabolism , Visual Cortex/pathology
11.
Hum Brain Mapp ; 41(15): 4299-4313, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32648649

ABSTRACT

We have previously demonstrated that pediatric-onset multiple sclerosis (POMS) negatively impacts the visual pathway as well as motor processing speed. Relationships between MS-related diffuse structural damage of gray and white matter (WM) tissue and cortical responses to visual and motor stimuli remain poorly understood. We used magnetoencephalography in 14 POMS patients and 15 age- and sex-matched healthy controls to assess visual gamma (30-80 Hz), motor gamma (60-90 Hz), and motor beta (15-30 Hz) cortical oscillatory responses to a visual-motor task. Then, 3T MRI was used to: (a) calculate fractional anisotropy (FA) of the posterior visual and corticospinal motor WM pathways and (b) quantify volume and thickness of the cuneus and primary motor cortex. Visual gamma band power was reduced in POMS and was associated with reduced FA of the optic radiations but not with loss of cuneus volume or thickness. Activity in the primary motor cortex, as measured by postmovement beta rebound amplitude associated with peak latency, was decreased in POMS, although this reduction was not predicted by structural metrics. Our findings implicate loss of WM integrity as a contributor to reduced electrical responses in the visual cortex in POMS. Future work in larger cohorts will inform on the cognitive implications of this finding in terms of visual processing function and will determine whether the progressive loss of brain volume known to occur in POMS ultimately contributes to both progressive dysfunction in such tasks as well as progressive reduction in cortical electrical responses in the visual cortex.


Subject(s)
Beta Rhythm/physiology , Gamma Rhythm/physiology , Magnetic Resonance Imaging , Motor Cortex , Multiple Sclerosis, Relapsing-Remitting , Visual Cortex , Adolescent , Adult , Age of Onset , Child , Diffusion Tensor Imaging , Efferent Pathways/diagnostic imaging , Efferent Pathways/pathology , Efferent Pathways/physiopathology , Female , Humans , Magnetoencephalography , Male , Motor Cortex/diagnostic imaging , Motor Cortex/pathology , Motor Cortex/physiology , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/pathology , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Visual Cortex/diagnostic imaging , Visual Cortex/pathology , Visual Cortex/physiology , Visual Pathways/diagnostic imaging , Visual Pathways/pathology , Visual Pathways/physiopathology , Young Adult
12.
Eur Arch Psychiatry Clin Neurosci ; 270(5): 577-588, 2020 Aug.
Article in English | MEDLINE | ID: mdl-30937515

ABSTRACT

The concept of acute stress disorder (ASD) was introduced as a diagnostic entity to improve the identification of traumatized people who are likely to develop posttraumatic stress disorder (PTSD). Neuroanatomical models suggest that changes in the prefrontal cortex, amygdala, and hippocampus play a role in the development of PTSD. Using voxel-based morphometry, this study aimed to investigate the predictive power of gray matter volume (GMV) alterations for developing PTSD. The GMVs of ASD patients (n = 21) were compared to those of PTSD patients (n = 17) and healthy controls (n = 18) in whole-brain and region-of-interest analyses. The GMV alterations seen in ASD patients shortly after the traumatic event (T1) were also correlated with PTSD symptom severity and symptom clusters 4 weeks later (T2). Compared with healthy controls, the ASD patients had significantly reduced GMV in the left visual cortex shortly after the traumatic event (T1) and in the left occipital and prefrontal regions 4 weeks later (T2); no significant differences in GMV were seen between the ASD and PTSD patients. Furthermore, a significant negative association was found between the GMV reduction in the left lateral temporal regions seen after the traumatic event (T1) and PTSD hyperarousal symptoms 4 weeks later (T2). Neither amygdala nor hippocampus alterations were predictive for the development of PTSD. These data suggest that gray matter deficiencies in the left hemispheric occipital and temporal regions in ASD patients may predict a liability for developing PTSD.


Subject(s)
Gray Matter/pathology , Occipital Lobe/pathology , Prefrontal Cortex/pathology , Stress Disorders, Post-Traumatic/pathology , Stress Disorders, Traumatic, Acute/pathology , Temporal Lobe/pathology , Adult , Amygdala/diagnostic imaging , Amygdala/pathology , Disease Susceptibility/diagnostic imaging , Disease Susceptibility/pathology , Female , Gray Matter/diagnostic imaging , Hippocampus/diagnostic imaging , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Occipital Lobe/diagnostic imaging , Prefrontal Cortex/diagnostic imaging , Stress Disorders, Post-Traumatic/diagnostic imaging , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Traumatic, Acute/diagnostic imaging , Stress Disorders, Traumatic, Acute/physiopathology , Temporal Lobe/diagnostic imaging , Time Factors , Visual Cortex/diagnostic imaging , Visual Cortex/pathology , Young Adult
13.
Cereb Cortex ; 29(10): 4321-4333, 2019 09 13.
Article in English | MEDLINE | ID: mdl-30561529

ABSTRACT

Blindness early in life induces permanent alterations in brain anatomy, including reduced surface area of primary visual cortex (V1). Bilateral enucleation early in development causes greater reductions in primary visual cortex surface area than at later times. However, the time at which cortical surface area expansion is no longer sensitive to enucleation is not clearly established, despite being an important milestone for cortical development. Using histological and MRI techniques, we investigated how reductions in the surface area of V1 depends on the timing of blindness onset in rats, ferrets and humans. To compare data across species, we translated ages of all species to a common neuro-developmental event-time (ET) scale. Consistently, blindness during early cortical expansion induced large (~40%) reductions in V1 surface area, in rats and ferrets, while blindness occurring later had diminishing effects. Longitudinal measurements on ferrets confirmed that early enucleation disrupted cortical expansion, rather than inducing enhanced pruning. We modeled the ET associated with the conclusion of the effect of blindness on surface area at maturity (ETc), relative to the normal conclusion of visual cortex surface area expansion, (ETdev). A final analysis combining our data with extant published data confirmed that ETc occurred well before ETdev.


Subject(s)
Blindness/pathology , Blindness/physiopathology , Ferrets/anatomy & histology , Ferrets/growth & development , Sensory Deprivation/physiology , Visual Cortex/growth & development , Visual Cortex/pathology , Age of Onset , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Humans , Rats , Species Specificity
14.
Dev Med Child Neurol ; 62(11): 1324-1330, 2020 11.
Article in English | MEDLINE | ID: mdl-32770793

ABSTRACT

AIM: To investigate the utility of the Insight Inventory (a structured clinical inventory completed by caregivers) for assessment of children with cerebral visual impairment; and to investigate effectiveness of tailored habilitational strategies derived from the responses to the Insight Inventory. METHOD: Fifty-one eligible children (26 males, 25 females; mean age 9y 5mo, SD 3y, range 5-16y) were recruited from Great Ormond Street Hospital, London. They underwent baseline assessment including neuro-ophthalmological and neuropsychological evaluations, and parent- and child-reported ratings on a questionnaire-based measure of quality of life. Parents also completed the Insight Inventory. On the basis of responses to the Inventory, families received individualized habilitational strategies. Follow-up assessments 6 months later included repeating the Insight Inventory and quality of life questionnaires. RESULTS: Correlations were found between the Insight Inventory and the Wechsler Intelligence Scale for Children, Fourth Edition, the Beery-Buktenica Test of Visual-Motor Integration, and the Benton Facial Recognition Test, suggesting that the Insight Inventory is an effective tool to estimate visual-perceptual difficulties. At 6 months follow-up, caregiver reports indicated significant improvements in the quality of life of children below the age of 12 years. INTERPRETATION: The Insight Inventory is a simple questionnaire which covers practical aspects of cognitive visual function in everyday life. It provides in-depth information about the aspects that children struggle with. It can also guide programmes of individualized habilitation strategies, which may enhance the quality of life of younger children. WHAT THIS PAPER ADDS: Questionnaire scores demonstrate biologically plausible correlations with formal neuropsychological tests of visual function. After administration of matched practical habilitational strategies, younger children showed improvement in quality of life and functional vision scores.


Subject(s)
Attention/physiology , Motion Perception/physiology , Neurological Rehabilitation , Neuropsychological Tests , Pattern Recognition, Visual/physiology , Quality of Life , Vision Disorders/diagnosis , Vision Disorders/rehabilitation , Visual Cortex/pathology , Visual Fields/physiology , Visual Pathways/pathology , Adolescent , Child , Female , Follow-Up Studies , Humans , Male , Outcome Assessment, Health Care , Prospective Studies , Space Perception/physiology , Visual Acuity/physiology
15.
Cereb Cortex ; 29(11): 4863-4876, 2019 12 17.
Article in English | MEDLINE | ID: mdl-30843062

ABSTRACT

In the present combined DTI/fMRI study we investigated adaptive plasticity of neural networks involved in controlling spatial and nonspatial auditory working memory in the early blind (EB). In both EB and sighted controls (SC), fractional anisotropy (FA) within the right inferior longitudinal fasciculus correlated positively with accuracy in a one-back sound localization but not sound identification task. The neural tracts passing through the cluster of significant correlation connected auditory and "visual" areas in the right hemisphere. Activity in these areas during both sound localization and identification correlated with FA within the anterior corpus callosum, anterior thalamic radiation, and inferior fronto-occipital fasciculus. In EB, FA in these structures correlated positively with activity in both auditory and "visual" areas, whereas FA in SC correlated positively with activity in auditory and negatively with activity in visual areas. The results indicate that frontal white matter conveys cross-modal suppression of occipital areas in SC, while it mediates coactivation of auditory and reorganized "visual" cortex in EB.


Subject(s)
Auditory Cortex/pathology , Auditory Cortex/physiopathology , Auditory Perception/physiology , Blindness/pathology , Blindness/physiopathology , Visual Cortex/pathology , Visual Cortex/physiology , Adult , Brain Mapping , Diffusion Magnetic Resonance Imaging , Female , Humans , Male , Memory, Short-Term/physiology , Middle Aged , Neural Pathways/pathology , Neural Pathways/physiopathology , Neuronal Plasticity , Sound Localization/physiology , Space Perception/physiology , White Matter/pathology , White Matter/physiopathology
16.
Ecotoxicol Environ Saf ; 201: 110799, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32544743

ABSTRACT

Mercury (Hg) is an environmental contaminant that poses great risk to human health. However, it is still widely used in artisanal gold-mining enterprises around the world, especially in developing countries. Methylmercury (MeHg) is produced environmentally by biomethylation of inorganic Hg present in water sediments, leading to its subsequent accumulation in the aquatic food chain. Due to its high metabolic rate, the Central Nervous System (CNS) is one of the main targets of MeHg. In the present study, we investigate the impact of chronic MeHg intoxication on NADPH diaphorase (NADPH-d) activity and astrocyte mobilization in the visual cortex of the rat. After 60 days of MeHg administration by oral gavage (0.04 mg/kg/day), tissue samples containing the visual cortex were submitted to measurements of Hg levels, NADPH-d activity, and GFAP immunohistochemistry for identification of astrocytes. MeHg intoxication was associated with increased Hg deposits and with reduced NADPH-d neuropil reactivity in the visual cortex. A morphometric analysis suggested that NADPH-d-positive neurons were mostly spared from MeHg harmful action and intoxicated animals had astrocytic activation similar to the control group. The decrease in NADPH-d neuropil reactivity may be due to the negative effect of chronic MeHg poisoning on both the synthesis and transport of this enzyme in afferent pathways to the visual cortex. The relative resistance of NADPH-d-reactive neurons to chronic MeHg intoxication may be associated with peculiarities in cell metabolism or to a protective role of nitric oxide, safeguarding those neurons from Hg deleterious effects.


Subject(s)
Astrocytes/drug effects , Environmental Pollutants/toxicity , Methylmercury Compounds/toxicity , NADPH Dehydrogenase/metabolism , Neurons/drug effects , Visual Cortex/drug effects , Animals , Astrocytes/enzymology , Behavior, Animal/drug effects , Environmental Pollutants/metabolism , Gold , Humans , Male , Methylmercury Compounds/metabolism , Mining , Neurons/enzymology , Rats , Rats, Wistar , Visual Cortex/enzymology , Visual Cortex/pathology
17.
Neural Plast ; 2020: 8826087, 2020.
Article in English | MEDLINE | ID: mdl-33014034

ABSTRACT

The retina may serve as putative window into neuropathology of synaptic loss in Alzheimer's disease (AD). Here, we investigated synapse-rich layers versus layers composed by nuclei/cell bodies in an early stage of AD. In addition, we examined the associations between retinal changes and molecular and structural markers of cortical damage. We recruited 20 AD patients and 17 healthy controls (HC). Combining optical coherence tomography (OCT), magnetic resonance (MR), and positron emission tomography (PET) imaging, we measured retinal and primary visual cortex (V1) thicknesses, along with V1 amyloid ß (Aß) retention ([11C]-PiB PET tracer) and neuroinflammation ([11C]-PK11195 PET tracer). We found that V1 showed increased amyloid-binding potential, in the absence of neuroinflammation. Although thickness changes were still absent, we identified a positive association between the synapse-rich inner plexiform layer (IPL) and V1 in AD. This retinocortical interplay might reflect changes in synaptic function resulting from Aß deposition, contributing to early visual loss.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Gray Matter/pathology , Retina/pathology , Synapses/pathology , Visual Cortex/pathology , Aged , Female , Gray Matter/metabolism , Humans , Male , Middle Aged , Retina/metabolism , Synapses/metabolism , Visual Cortex/metabolism
18.
J Neurosci ; 38(49): 10454-10466, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30355624

ABSTRACT

Homeostatic synaptic plasticity is a synaptic mechanism through which the nervous system adjusts synaptic excitation and inhibition to maintain network stability. Retinoic acid (RA) and its receptor RARα have been established as critical mediators of homeostatic synaptic plasticity. In vitro studies reveal that RA signaling enhances excitatory synaptic strength and decreases inhibitory synaptic strength. However, it is unclear whether RA-mediated homeostatic synaptic plasticity occurs in vivo, and if so, whether it operates at specific types of synapses. Here, we examine the impact of RA/RARα signaling in the monocular zone of primary visual cortex (V1m) in mice of either sex. Exogenous RA treatment in acute cortical slices resulted in a reduction in mIPSCs of layer 2/3 pyramidal neurons, an effect mimicked by visual deprivation induced by binocular enucleation in postcritical period animals. Postnatal deletion of RARα blocked RA's effect on mIPSCs. Cell type-specific deletion of RARα revealed that RA acted specifically on parvalbumin (PV)-expressing interneurons. RARα deletion in PV+ interneurons blocked visual deprivation-induced changes in mIPSCs, demonstrating the critical involvement of RA signaling in PV+ interneurons in vivo Moreover, visual deprivation- or RA-induced downregulation of synaptic inhibition was absent in the visual cortical circuit of constitutive and PV-specific Fmr1 KO mice, strongly suggesting a functional interaction between fragile X mental retardation protein and RA signaling pathways. Together, our results demonstrate that RA/RARα signaling acts as a key component for homeostatic regulation of synaptic transmission at the inhibitory synapses of the visual cortex.SIGNIFICANCE STATEMENTIn vitro studies established that retinoic acid (RA) and its receptor RARα play key roles in homeostatic synaptic plasticity, a mechanism by which synaptic excitation/inhibition balance and network stability are maintained. However, whether synaptic RA signaling operates in vivo remains undetermined. Here, using a conditional RARα KO mouse and cell type-specific Cre-driver lines, we showed that RARα signaling in parvalbumin-expressing interneurons is crucial for visual deprivation-induced homeostatic synaptic plasticity at inhibitory synapses in visual cortical circuits. Importantly, this form of synaptic plasticity is absent when fragile X mental retardation protein is selectively deleted in parvalbumin-expressing interneurons, suggesting a functional connection between RARα and fragile X mental retardation protein signaling pathways in vivo Thus, dysfunction of RA-dependent homeostatic plasticity may contribute to cortical circuit abnormalities in fragile X syndrome.


Subject(s)
Homeostasis/physiology , Neuronal Plasticity/physiology , Receptors, Retinoic Acid/deficiency , Synapses/physiology , Visual Cortex/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Inhibition/physiology , Receptors, Retinoic Acid/genetics
19.
Glia ; 67(6): 1113-1121, 2019 06.
Article in English | MEDLINE | ID: mdl-30791140

ABSTRACT

Cortical spreading depression (CSD) is a slowly propagating wave of depolarization of gray matter. This phenomenon is believed to underlie the migraine aura and similar waves of depolarization may exacerbate injury in a number of neurological disease states. CSD is characterized by massive ion dyshomeostasis, cell swelling, and multiphasic blood flow changes. Recently, it was shown that CSD is associated with a closure of the paravascular space (PVS), a proposed exit route for brain interstitial fluid and solutes, including excitatory and inflammatory substances that increase in the wake of CSD. The PVS closure was hypothesized to rely on swelling of astrocytic endfeet due to their high expression of aquaporin-4 (AQP4) water channels. We investigated whether CSD is associated with swelling of endfeet around penetrating arterioles in the cortex of living mice. Endfoot cross-sectional area was assessed by two-photon microscopy of mice expressing enhanced green fluorescent protein in astrocytes and related to the degree of arteriolar constriction. In anesthetized mice CSD triggered pronounced endfoot swelling that was short-lasting and coincided with the initial arteriolar constriction. Mice lacking AQP4 displayed volume changes of similar magnitude. CSD-induced endfoot swelling and arteriolar constriction also occurred in awake mice, albeit with faster kinetics than in anesthetized mice. We conclude that swelling of astrocytic endfeet is a robust event in CSD. The early onset and magnitude of the endfoot swelling is such that it may significantly delay perivascular drainage of interstitial solutes in neurological conditions where CSD plays a pathophysiological role.


Subject(s)
Aquaporin 4/deficiency , Astrocytes/metabolism , Cell Size , Cortical Spreading Depression/physiology , Visual Cortex/metabolism , Animals , Aquaporin 4/genetics , Astrocytes/pathology , Mice , Mice, Transgenic , Visual Cortex/pathology
20.
Neuroimage ; 190: 254-268, 2019 04 15.
Article in English | MEDLINE | ID: mdl-29627591

ABSTRACT

Damage to the primary visual cortex (V1) leads to a visual field loss (scotoma) in the retinotopically corresponding part of the visual field. Nonetheless, a small amount of residual visual sensitivity persists within the blind field. This residual capacity has been linked to activity observed in the middle temporal area complex (V5/MT+). However, it remains unknown whether the organization of hV5/MT+ changes following early visual cortical lesions. We studied the organization of area hV5/MT+ of five patients with dense homonymous defects in a quadrant of the visual field as a result of partial V1+ or optic radiation lesions. To do so, we developed a new method, which models the boundaries of population receptive fields directly from the BOLD signal of each voxel in the visual cortex. We found responses in hV5/MT+ arising inside the scotoma for all patients and identified two possible sources of activation: 1) responses might originate from partially lesioned parts of area V1 corresponding to the scotoma, and 2) responses can also originate independent of area V1 input suggesting the existence of functional V1-bypassing pathways. Apparently, visually driven activity observed in hV5/MT+ is not sufficient to mediate conscious vision. More surprisingly, visually driven activity in corresponding regions of V1 and early extrastriate areas including hV5/MT+ did not guarantee visual perception in the group of patients with post-geniculate lesions that we examined. This suggests that the fine coordination of visual activity patterns across visual areas may be an important determinant of whether visual perception persists following visual cortical lesions.


Subject(s)
Scotoma , Vision Disorders , Visual Cortex , Visual Fields/physiology , Visual Pathways , Visual Perception/physiology , Adult , Echo-Planar Imaging , Female , Functional Neuroimaging , Humans , Male , Middle Aged , Scotoma/diagnostic imaging , Scotoma/physiopathology , Stroke/complications , Vision Disorders/diagnostic imaging , Vision Disorders/etiology , Vision Disorders/pathology , Vision Disorders/physiopathology , Visual Cortex/diagnostic imaging , Visual Cortex/pathology , Visual Cortex/physiopathology , Visual Pathways/diagnostic imaging , Visual Pathways/pathology , Visual Pathways/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL