Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-27928512

ABSTRACT

Alzheimer's Disease (AD) is an age-related neurodegenerative disorder in which aggregation-prone neurotoxic amyloid ß-peptide (Aß) accumulates in the brain. Extracellular vesicles (EVs) are small 50-150 nanometer membrane vesicles that have recently been implicated in the prion-like spread of self-aggregating proteins. Here we report that EVs isolated from AD patient CSF and plasma, from the plasma of two AD mouse models, and from the medium of neural cells expressing familial AD presenilin 1 mutations, destabilize neuronal Ca2+ homeostasis, impair mitochondrial function, and sensitize neurons to excitotoxicity. EVs contain a relatively low amount of Aß but have an increased Aß42/ Aß40 ratio; the majority of Aß is located on the surface of the EVs. Impairment of lysosome function results in increased generation EVs with elevated Aß42 levels. EVs may mediate transcellular spread of pathogenic Aß species and that impair neuronal Ca2+ handling and mitochondrial function, and may thereby render neurons vulnerable to excitotoxicity.

3.
Cell Rep ; 15(5): 926-934, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117401

ABSTRACT

During neuronal differentiation, use of an alternative splice site on the rat telomere repeat-binding factor 2 (TRF2) mRNA generates a short TRF2 protein isoform (TRF2-S) capable of derepressing neuronal genes. However, the RNA-binding proteins (RBPs) controlling this splicing event are unknown. Here, using affinity pull-down analysis, we identified heterogeneous nuclear ribonucleoproteins H1 and H2(HNRNPH) as RBPs specifically capable of interacting with the spliced RNA segment (exon 7) of Trf2 pre-mRNA. HNRNPH proteins prevent the production of the short isoform of Trf2 mRNA, as HNRNPH silencing selectively elevates TRF2-S levels. Accordingly, HNRNPH levels decline while TRF2-S levels increase during neuronal differentiation. In addition, CRISPR/Cas9-mediated deletion of hnRNPH2 selectively accelerates the NGF-triggered differentiation of rat pheochromocytoma cells into neurons. In sum, HNRNPH is a splicing regulator of Trf2 pre-mRNA that prevents the expression of TRF2-S, a factor implicated in neuronal differentiation.


Subject(s)
Alternative Splicing/genetics , Cell Differentiation/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Neurons/cytology , Telomeric Repeat Binding Protein 2/metabolism , Animals , Base Sequence , Exons/genetics , Neurons/metabolism , PC12 Cells , Protein Binding , Proteomics , RNA/metabolism , RNA Precursors/genetics , Rats , Telomeric Repeat Binding Protein 2/genetics
4.
Cerebellum ; 15(4): 509-17, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26374457

ABSTRACT

Telomerase reverse transcriptase (TERT) is the catalytic subunit of telomerase, an enzyme that elongates telomeres at the ends of chromosomes during DNA replication. Recently, it was shown that TERT has additional roles in cell survival, mitochondrial function, DNA repair, and Wnt signaling, all of which are unrelated to telomeres. Here, we demonstrate that TERT is enriched in Purkinje neurons, but not in the granule cells of the adult mouse cerebellum. TERT immunoreactivity in Purkinje neurons is present in the nucleus, mitochondria, and cytoplasm. Furthermore, TERT co-localizes with mitochondrial markers, and immunoblot analysis of protein extracts from isolated mitochondria and synaptosomes confirmed TERT localization in mitochondria. TERT expression in Purkinje neurons increased significantly in response to two stressors: a sub-lethal dose of X-ray radiation and exposure to a high glutamate concentration. While X-ray radiation increased TERT levels in the nucleus, glutamate exposure elevated TERT levels in mitochondria. Our findings suggest that in mature Purkinje neurons, TERT is present both in the nucleus and in mitochondria, where it may participate in adaptive responses of the neurons to excitotoxic and radiation stress.


Subject(s)
Cytosol/enzymology , Glutamic Acid/toxicity , Mitochondria/enzymology , Purkinje Cells/enzymology , Radiation Injuries, Experimental/enzymology , Telomerase/metabolism , Animals , Cell Nucleus/enzymology , Cell Nucleus/pathology , Cell Nucleus/radiation effects , Cytosol/pathology , Cytosol/radiation effects , DNA Damage/physiology , DNA Damage/radiation effects , Electron Transport Complex IV/metabolism , Fluorescent Antibody Technique , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Mitochondria/radiation effects , Purkinje Cells/pathology , Purkinje Cells/radiation effects , Radiation Injuries, Experimental/pathology , Stress, Physiological/physiology , Stress, Physiological/radiation effects , Telomerase/genetics , Tissue Culture Techniques , X-Rays/adverse effects
5.
Exp Neurol ; 273: 151-60, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26277686

ABSTRACT

OBJECTIVE: Multiple sclerosis (MS) is a debilitating neurological disorder involving an autoimmune reaction to oligodendrocytes and degeneration of the axons they ensheath in the CNS. Because the damage to oligodendrocytes and axons involves local inflammation and associated oxidative stress, we tested the therapeutic efficacy of combined treatment with a potent anti-inflammatory thalidomide analog (lenalidomide) and novel synthetic anti-oxidant cerium oxide nanoparticles (nanoceria) in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS: C57BL/6 mice were randomly assigned to a control (no EAE) group, or one of the four myelin oligodendrocyte glycoprotein-induced EAE groups: vehicle, lenalidomide, nanoceria, or lenalidomide plus nanoceria. During a 23 day period, clinical EAE symptoms were evaluated daily, and MRI brain scans were performed at 11-13 days and 20-22 days. Histological and biochemical analyses of brain tissue samples were performed to quantify myelin loss and local inflammation. RESULTS: Lenalidomide treatment alone delayed symptom onset, while nanoceria treatment had no effect on symptom onset or severity, but did promote recovery; lenalidomide and nanoceria each significantly attenuated white matter pathology and associated inflammation. Combined treatment with lenalidomide and nanoceria resulted in a near elimination of EAE symptoms, and reduced white matter pathology and inflammatory cell responses to a much greater extent than either treatment alone. INTERPRETATION: By suppressing inflammation and oxidative stress, combined treatment with lenalidomide and nanoceria can reduce demyelination and associated neurological symptoms in EAE mice. Our preclinical data suggest a potential application of this combination therapy in MS.


Subject(s)
Autoimmunity/drug effects , Central Nervous System/drug effects , Cerium/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/therapeutic use , Thalidomide/analogs & derivatives , Analysis of Variance , Animals , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Lenalidomide , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , RNA, Messenger , Thalidomide/therapeutic use , Time Factors
6.
Biogerontology ; 15(6): 643-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25305051

ABSTRACT

The term cellular senescence was introduced more than five decades ago to describe the state of growth arrest observed in aging cells. Since this initial discovery, the phenotypes associated with cellular senescence have expanded beyond growth arrest to include alterations in cellular metabolism, secreted cytokines, epigenetic regulation and protein expression. Recently, senescence has been shown to play an important role in vivo not only in relation to aging, but also during embryonic development. Thus, cellular senescence serves different purposes and comprises a wide range of distinct phenotypes across multiple cell types. Whether all cell types, including post-mitotic neurons, are capable of entering into a senescent state remains unclear. In this review we examine recent data that suggest that cellular senescence plays a role in brain aging and, notably, may not be limited to glia but also neurons. We suggest that there is a high level of similarity between some of the pathological changes that occur in the brain in Alzheimer's and Parkinson's diseases and those phenotypes observed in cellular senescence, leading us to propose that neurons and glia can exhibit hallmarks of senescence previously documented in peripheral tissues.


Subject(s)
Aging/pathology , Brain/pathology , Cellular Senescence , Neurodegenerative Diseases/pathology , Aged , Aging/genetics , Aging/metabolism , Animals , Brain/metabolism , Cellular Senescence/genetics , Cellular Senescence/physiology , Epigenesis, Genetic , Humans , Mice , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology
7.
Cell Rep ; 7(5): 1401-1409, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24857657

ABSTRACT

The primarily neuronal RNA-binding protein HuD is implicated in learning and memory. Here, we report the identification of several HuD target transcripts linked to Alzheimer's disease (AD) pathogenesis. HuD interacted with the 3' UTRs of APP mRNA (encoding amyloid precursor protein) and BACE1 mRNA (encoding ß-site APP-cleaving enzyme 1) and increased the half-lives of these mRNAs. HuD also associated with and stabilized the long noncoding (lnc)RNA BACE1AS, which partly complements BACE1 mRNA and enhances BACE1 expression. Consistent with HuD promoting production of APP and APP-cleaving enzyme, the levels of APP, BACE1, BACE1AS, and Aß were higher in the brain of HuD-overexpressing mice. Importantly, cortex (superior temporal gyrus) from patients with AD displayed significantly higher levels of HuD and, accordingly, elevated APP, BACE1, BACE1AS, and Aß than did cortical tissue from healthy age-matched individuals. We propose that HuD jointly promotes the production of APP and the cleavage of its amyloidogenic fragment, Aß.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , ELAV Proteins/metabolism , RNA, Long Noncoding/metabolism , 3' Untranslated Regions , Aged , Aged, 80 and over , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Case-Control Studies , Cell Line, Tumor , Cerebral Cortex/metabolism , ELAV Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA Stability , RNA, Long Noncoding/genetics
8.
Trends Neurosci ; 37(5): 256-63, 2014 May.
Article in English | MEDLINE | ID: mdl-24698125

ABSTRACT

Telomeres, ribonucleoprotein complexes that cap eukaryotic chromosomes, typically shorten in leukocytes with aging. Aging is a primary risk factor for neurodegenerative disease (ND), and a common assumption has arisen that leukocyte telomere length (LTL) can serve as a predictor of neurological disease. However, the evidence for shorter LTL in Alzheimer's and Parkinson's patients is inconsistent. The diverse causes of telomere shortening may explain variability in LTL between studies and individuals. Additional research is needed to determine whether neuronal and glial telomeres shorten during aging and in neurodegenerative disorders, if and how LTL is related to brain cell telomere shortening, and whether telomere shortening plays a causal role in or exacerbates neurological disorders.


Subject(s)
Nervous System Diseases/genetics , Telomere Shortening/genetics , Animals , Humans
9.
Glia ; 61(7): 1018-28, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23650073

ABSTRACT

Inflammation is a common component of acute injuries of the central nervous system (CNS) such as ischemia, and degenerative disorders such as Alzheimer's disease. Glial cells play important roles in local CNS inflammation, and an understanding of the roles for microRNAs in glial reactivity in injury and disease settings may therefore lead to the development of novel therapeutic interventions. Here, we show that the miR-181 family is developmentally regulated and present in high amounts in astrocytes compared to neurons. Overexpression of miR-181c in cultured astrocytes results in increased cell death when exposed to lipopolysaccharide (LPS). We show that miR-181 expression is altered by exposure to LPS, a model of inflammation, in both wild-type and transgenic mice lacking both receptors for the inflammatory cytokine TNF-α. Knockdown of miR-181 enhanced LPS-induced production of pro-inflammatory cytokines (TNF-α, IL-6, IL-1ß, IL-8) and HMGB1, while overexpression of miR-181 resulted in a significant increase in the expression of the anti-inflammatory cytokine IL-10. To assess the effects of miR-181 on the astrocyte transcriptome, we performed gene array and pathway analysis on astrocytes with reduced levels of miR-181b/c. To examine the pool of potential miR-181 targets, we employed a biotin pull-down of miR-181c and gene array analysis. We validated the mRNAs encoding MeCP2 and X-linked inhibitor of apoptosis as targets of miR-181. These findings suggest that miR-181 plays important roles in the molecular responses of astrocytes in inflammatory settings. Further understanding of the role of miR-181 in inflammatory events and CNS injury could lead to novel approaches for the treatment of CNS disorders with an inflammatory component.


Subject(s)
Astrocytes/metabolism , MicroRNAs/metabolism , Neuroimmunomodulation/immunology , Animals , Astrocytes/drug effects , Biotinylation , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cytokines/metabolism , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Male , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Knockout , Neuroimmunomodulation/drug effects , Neurons/drug effects , Neurons/metabolism , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type II/deficiency , Transfection , X-Linked Inhibitor of Apoptosis Protein/genetics
10.
Free Radic Biol Med ; 61: 61-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23542375

ABSTRACT

Eukaryotic elongation factor 2 (eEF-2) is an important regulator of the protein translation machinery whereby it controls the movement of the ribosome along the mRNA. The activity of eEF-2 is regulated by changes in cellular energy status and nutrient availability and by posttranslational modifications such as phosphorylation and mono-ADP-ribosylation. However, the mechanisms regulating protein translation under conditions of cellular stress in neurons are unknown. Here we show that when rat hippocampal neurons experience oxidative stress (lipid peroxidation induced by exposure to cumene hydroperoxide; CH), eEF-2 is hyperphosphorylated and ribosylated, resulting in reduced translational activity. The degradation of eEF-2 requires calpain proteolytic activity and is accompanied by accumulation of eEF-2 in the nuclear compartment. The subcellular localization of both native and phosphorylated forms of eEF-2 is influenced by CRM1 and 14.3.3, respectively. In hippocampal neurons p53 interacts with nonphosphorylated (active) eEF-2, but not with its phosphorylated form. The p53-eEF-2 complexes are present in cytoplasm and nucleus, and their abundance increases when neurons experience oxidative stress. The nuclear localization of active eEF-2 depends upon its interaction with p53, as cells lacking p53 contain less active eEF-2 in the nuclear compartment. Overexpression of eEF-2 in hippocampal neurons results in increased nuclear levels of eEF-2 and decreased cell death after exposure to CH. Our results reveal novel molecular mechanisms controlling the differential subcellular localization and activity state of eEF-2 that may influence the survival status of neurons during periods of elevated oxidative stress.


Subject(s)
Neurons/metabolism , Oxidative Stress , Peptide Elongation Factor 2/physiology , 14-3-3 Proteins/physiology , Adenosine Diphosphate Ribose/metabolism , Animals , Benzene Derivatives/pharmacology , Cell Survival/drug effects , Cells, Cultured , HCT116 Cells , Humans , Karyopherins/physiology , Lipid Peroxidation , Peptide Elongation Factor 2/analysis , Phosphorylation , Rats , Receptors, Cytoplasmic and Nuclear/physiology , Tumor Suppressor Protein p53/physiology , Exportin 1 Protein
11.
Mol Cell Biol ; 31(4): 626-38, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21135128

ABSTRACT

Adipose tissue development is tightly regulated by altering gene expression. MicroRNAs are strong posttranscriptional regulators of mammalian differentiation. We hypothesized that microRNAs might influence human adipogenesis by targeting specific adipogenic factors. We identified microRNAs that showed varying abundance during the differentiation of human preadipocytes into adipocytes. Among them, miR-130 strongly affected adipocyte differentiation, as overexpressing miR-130 impaired adipogenesis and reducing miR-130 enhanced adipogenesis. A key effector of miR-130 actions was the protein peroxisome proliferator-activated receptor γ (PPARγ), a major regulator of adipogenesis. Interestingly, miR-130 potently repressed PPARγ expression by targeting both the PPARγ mRNA coding and 3' untranslated regions. Adipose tissue from obese women contained significantly lower miR-130 and higher PPARγ mRNA levels than that from nonobese women. Our findings reveal that miR-130 reduces adipogenesis by repressing PPARγ biosynthesis and suggest that perturbations in this regulation is linked to human obesity.


Subject(s)
Adipogenesis/genetics , Adipogenesis/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA Primers/genetics , Female , Gene Expression Regulation, Developmental , Humans , In Vitro Techniques , Mice , Obesity/genetics , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Species Specificity , Thinness/genetics , Thinness/metabolism
12.
Mol Cell Biol ; 30(17): 4197-210, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20584986

ABSTRACT

Neuronal development and plasticity are maintained by tightly regulated gene expression programs. Here, we report that the developmentally regulated microRNA miR-375 affects dendrite formation and maintenance. miR-375 overexpression in mouse hippocampus potently reduced dendrite density. We identified the predominantly neuronal RNA-binding protein HuD as a key effector of miR-375 influence on dendrite maintenance. Heterologous reporter analysis verified that miR-375 repressed HuD expression through a specific, evolutionarily conserved site on the HuD 3' untranslated region. miR-375 overexpression lowered both HuD mRNA stability and translation and recapitulated the effects of HuD silencing, which reduced the levels of target proteins with key functions in neuronal signaling and cytoskeleton organization (N-cadherin, PSD-95, RhoA, NCAM1, and integrin alpha1). Moreover, the increase in neurite outgrowth after brain-derived neurotrophic factor (BDNF) treatment was diminished by miR-375 overexpression; this effect was rescued by reexpression of miR-375-refractory HuD. Our findings indicate that miR-375 modulates neuronal HuD expression and function, in turn affecting dendrite abundance.


Subject(s)
Dendrites/metabolism , ELAV Proteins/genetics , MicroRNAs/metabolism , Neurogenesis , Animals , Cell Line , Dendrites/ultrastructure , ELAV Proteins/metabolism , ELAV-Like Protein 4 , Gene Expression Regulation , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neurites/metabolism , Neurites/ultrastructure , RNA Stability , RNA, Messenger/genetics
13.
Neuromolecular Med ; 11(3): 153-61, 2009.
Article in English | MEDLINE | ID: mdl-19763905

ABSTRACT

MicroRNAS (miRNAs) have been suggested to play important roles in the central nervous system during development as well as disease. miRNAs appear to be dysregulated in a number of neurodegenerative diseases, developmental disorders, and as a result of stroke. Each miRNA has the ability to regulate hundreds of messenger RNA transcripts, both by causing degradation of the mRNA and by inhibition of protein translation. Recent findings suggest that it may eventually be possible to treat some neurological disorders by restoring or inhibiting miRNAs altered by disease pathology. Both viral delivery and administration of modified oligonucleotides mimicking or inhibiting specific miRNAs have been effective in model systems. Artificial miRNAs have also been generated for the repression of specific transcripts. Alteration of miRNA expression by disease and insult also holds the potential for improved diagnostic tools. Finally, miRNAs have been shown to control cellular proliferation and specification, suggesting that manipulation of miRNAs in cultured cells could result in more convenient generation of pure cell populations for transplantation.


Subject(s)
Central Nervous System , MicroRNAs , Nerve Regeneration/physiology , Nervous System Diseases , Neurodegenerative Diseases , Trauma, Nervous System , Animals , Central Nervous System/pathology , Central Nervous System/physiology , Central Nervous System/physiopathology , Gene Expression Regulation , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/therapeutic use , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology , Nervous System Diseases/therapy , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/therapy , Transduction, Genetic , Trauma, Nervous System/pathology , Trauma, Nervous System/physiopathology , Trauma, Nervous System/therapy
14.
Dev Neurosci ; 31(1-2): 159-66, 2009.
Article in English | MEDLINE | ID: mdl-19372696

ABSTRACT

This study investigated the potential long-term effects of cocaine exposure on brain functioning using fMRI in school-aged children. The sample included 12 children with prenatal cocaine exposure and 12 non-exposed children (8-9 years old). Groups did not differ on IQ, socioeconomic status, or perinatal risk factors. A response inhibition task was administered during an fMRI scan using a 1.5-T MRI system. Task performance did not differentiate groups, but groups were differentiated by patterns of task-related brain activity. Cocaine-exposed children showed greater activation in the right inferior frontal cortex and caudate during response inhibition, whereas non-exposed children showed greater activations in temporal and occipital regions. These preliminary findings suggest that prenatal cocaine may affect the development of brain systems involved in the regulation of attention and response inhibition.


Subject(s)
Cocaine-Related Disorders/complications , Cocaine/adverse effects , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology , Behavior/drug effects , Child , Cognition/drug effects , Data Interpretation, Statistical , Female , Functional Laterality/drug effects , Gyrus Cinguli/pathology , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Prefrontal Cortex/pathology , Pregnancy , Psychomotor Performance/drug effects
15.
Neuroimage ; 40(1): 342-52, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18248740

ABSTRACT

The discrimination of voice-onset time, an acoustic-phonetic cue to voicing in stop consonants, was investigated to explore the neural systems underlying the perception of a rapid temporal speech parameter. Pairs of synthetic stimuli taken from a [da] to [ta] continuum varying in voice-onset time (VOT) were presented for discrimination judgments. Participants exhibited categorical perception, discriminating 15-ms and 30-ms between-category comparisons and failing to discriminate 15-ms within-category comparisons. Contrastive analysis with a tone discrimination task demonstrated left superior temporal gyrus activation in all three VOT conditions with recruitment of additional regions, particularly the right inferior frontal gyrus and middle frontal gyrus for the 15-ms between-category stimuli. Hemispheric differences using anatomically defined regions of interest showed two distinct patterns with anterior regions showing more activation in the right hemisphere relative to the left hemisphere and temporal regions demonstrating greater activation in the left hemisphere relative to the right hemisphere. Activation in the temporal regions appears to reflect initial acoustic-perceptual analysis of VOT. Greater activation in the right hemisphere anterior regions may reflect increased processing demands, suggesting involvement of the right hemisphere when the acoustic distance between the stimuli are reduced and when the discrimination judgment becomes more difficult.


Subject(s)
Cerebral Cortex/physiology , Cues , Discrimination, Psychological/physiology , Magnetic Resonance Imaging/methods , Psychomotor Performance/physiology , Time Perception/physiology , Adult , Auditory Perception/physiology , Brain/physiology , Cerebral Cortex/anatomy & histology , Data Interpretation, Statistical , Female , Functional Laterality/physiology , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Reaction Time/physiology , Regression Analysis , Voice
SELECTION OF CITATIONS
SEARCH DETAIL
...