Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 786
Filter
1.
Exp Neurol ; 376: 114756, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508482

ABSTRACT

Overexpression of the Ube3a gene and the resulting increase in Ube3a protein are linked to autism spectrum disorder (ASD). However, the cellular and molecular processes underlying Ube3a-dependent ASD remain unclear. Using both male and female mice, we find that neurons in the somatosensory cortex of the Ube3a 2× Tg ASD mouse model display reduced dendritic spine density and increased immature filopodia density. Importantly, the increased gene dosage of Ube3a in astrocytes alone is sufficient to confer alterations in neurons as immature dendritic protrusions, as observed in primary hippocampal neuron cultures. We show that Ube3a overexpression in astrocytes leads to a loss of astrocyte-derived spinogenic protein, thrombospondin-2 (TSP2), due to a suppression of TSP2 gene transcription. By neonatal intraventricular injection of astrocyte-specific virus, we demonstrate that Ube3a overexpression in astrocytes in vivo results in a reduction in dendritic spine maturation in prelimbic cortical neurons, accompanied with autistic-like behaviors in mice. These findings reveal an astrocytic dominance in initiating ASD pathobiology at the neuronal and behavior levels. SIGNIFICANCE STATEMENT: Increased gene dosage of Ube3a is tied to autism spectrum disorders (ASDs), yet cellular and molecular alterations underlying autistic phenotypes remain unclear. We show that Ube3a overexpression leads to impaired dendritic spine maturation, resulting in reduced spine density and increased filopodia density. We find that dysregulation of spine development is not neuron autonomous, rather, it is mediated by an astrocytic mechanism. Increased gene dosage of Ube3a in astrocytes leads to reduced production of the spinogenic glycoprotein thrombospondin-2 (TSP2), leading to abnormalities in spines. Astrocyte-specific Ube3a overexpression in the brain in vivo confers dysregulated spine maturation concomitant with autistic-like behaviors in mice. These findings indicate the importance of astrocytes in aberrant neurodevelopment and brain function in Ube3a-depdendent ASD.


Subject(s)
Autism Spectrum Disorder , Dendritic Spines , Ubiquitin-Protein Ligases , Animals , Mice , Female , Dendritic Spines/pathology , Dendritic Spines/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Male , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Neurons/metabolism , Neurons/pathology , Thrombospondins/metabolism , Thrombospondins/genetics , Thrombospondins/biosynthesis , Neuroglia/metabolism , Neuroglia/pathology , Mice, Transgenic , Somatosensory Cortex/metabolism , Somatosensory Cortex/pathology , Cells, Cultured , Neurogenesis/physiology , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology
2.
Nat Commun ; 13(1): 534, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35087056

ABSTRACT

Behavioral experience and flexibility are crucial for survival in a constantly changing environment. Despite evolutionary pressures to develop adaptive behavioral strategies in a dynamically changing sensory landscape, the underlying neural correlates have not been well explored. Here, we use genetically encoded voltage imaging to measure signals in primary somatosensory cortex (S1) during sensory learning and behavioral adaptation in the mouse. In response to changing stimulus statistics, mice adopt a strategy that modifies their detection behavior in a context dependent manner as to maintain reward expectation. Surprisingly, neuronal activity in S1 shifts from simply representing stimulus properties to transducing signals necessary for adaptive behavior in an experience dependent manner. Our results suggest that neuronal signals in S1 are part of an adaptive framework that facilitates flexible behavior as individuals gain experience, which could be part of a general scheme that dynamically distributes the neural correlates of behavior during learning.


Subject(s)
Adaptation, Psychological/physiology , Somatosensory Cortex/physiology , Animals , Brain , Learning , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Perception , Reward , Somatosensory Cortex/pathology
3.
Sci Rep ; 12(1): 114, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997092

ABSTRACT

Microglia are subject to change in tandem with the endogenously generated biological oscillations known as our circadian rhythm. Studies have shown microglia harbor an intrinsic molecular clock which regulates diurnal changes in morphology and influences inflammatory responses. In the adult brain, microglia play an important role in the regulation of condensed extracellular matrix structures called perineuronal nets (PNNs), and it has been suggested that PNNs are also regulated in a circadian and diurnal manner. We sought to determine whether microglia mediate the diurnal regulation of PNNs via CSF1R inhibitor dependent microglial depletion in C57BL/6J mice, and how the absence of microglia might affect cortical diurnal gene expression rhythms. While we observe diurnal differences in microglial morphology, where microglia are most ramified at the onset of the dark phase, we do not find diurnal differences in PNN intensity. However, PNN intensity increases across many brain regions in the absence of microglia, supporting a role for microglia in the regulation of PNNs. Here, we also show that cortical diurnal gene expression rhythms are intact, with no cycling gene changes without microglia. These findings demonstrate a role for microglia in the maintenance of PNNs, but not in the maintenance of diurnal rhythms.


Subject(s)
Brain Waves , Circadian Rhythm , Microglia/pathology , Nerve Net/pathology , Somatosensory Cortex/pathology , Animals , Brain Waves/drug effects , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Nerve Net/drug effects , Nerve Net/metabolism , Nerve Net/physiopathology , Organic Chemicals/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Somatosensory Cortex/physiopathology , Time Factors
4.
World Neurosurg ; 159: 108, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34968755

ABSTRACT

Intraaxial tumors of the central lobe are challenging lesions to deal with because of the high eloquence of this anatomic area.1,2 Diffusion tensor imaging magnetic resonance imaging and fluorescein (F) have proven to be useful in the planning and execution, respectively of glioma surgery.3-9 Nevertheless, the advantages of intraoperative use of augmented reality (AR) with diffusion tensor imaging-based high-definition fiber tractography (HDFT) are still underestimated. In the AR HDFT-F technique reported by our group, the integration of AR into the microscope comes through the BrainLAB Curve navigation platform (BrainLAB AG, Munich Germany), Smartbrush software (BrainLAB AG), KINEVO 900 surgical microscope (Carl Zeiss, Oberkochen, Germany), and YELLOW 560 filter (Carl Zeiss).9 The microscope establishes a wired autodetection of the navigation platform, and the eyepiece functions as a "see-through display" of the AR images, which are overlapped onto the surgical field. Video 1 shows the technical key aspects of the intraoperative use of the AR HDFT-F technique in the maximal safe anatomic resection of a postcentral gyrus high-grade glioma.


Subject(s)
Augmented Reality , Brain Neoplasms , Glioma , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Diffusion Tensor Imaging/methods , Fluorescein , Glioma/diagnostic imaging , Glioma/surgery , Humans , Neuronavigation/methods , Somatosensory Cortex/pathology
5.
J Neurosci ; 42(4): 532-551, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34848499

ABSTRACT

Mutations in some cell adhesion molecules (CAMs) cause abnormal synapse formation and maturation, and serve as one of the potential mechanisms of autism spectrum disorders (ASDs). Recently, DSCAM (Down syndrome cell adhesion molecule) was found to be a high-risk gene for autism. However, it is still unclear how DSCAM contributes to ASD. Here, we show that DSCAM expression was downregulated following synapse maturation, and that DSCAM deficiency caused accelerated dendritic spine maturation during early postnatal development. Mechanistically, the extracellular domain of DSCAM interacts with neuroligin1 (NLGN1) to block the NLGN1-neurexin1ß (NRXN1ß) interaction. DSCAM extracellular domain was able to rescue spine overmaturation in DSCAM knockdown neurons. Precocious spines in DSCAM-deficient mice showed increased glutamatergic transmission in the developing cortex and induced autism-like behaviors, such as social novelty deficits and repetitive behaviors. Thus, DSCAM might be a repressor that prevents premature spine maturation and excessive glutamatergic transmission, and its deficiency could lead to autism-like behaviors. Our study provides new insight into the potential pathophysiological mechanisms of ASDs.SIGNIFICANCE STATEMENTDSCAM is not only associated with Down syndrome but is also a strong autism risk gene based on large-scale sequencing analysis. However, it remains unknown exactly how DSCAM contributes to autism. In mice, either neuron- and astrocyte-specific or pyramidal neuron-specific DSCAM deficiencies resulted in autism-like behaviors and enhanced spatial memory. In addition, DSCAM knockout or knockdown in pyramidal neurons led to increased dendritic spine maturation. Mechanistically, the extracellular domain of DSCAM binds to NLGN1 and inhibits NLGN1-NRXN1ß interaction, which can rescue abnormal spine maturation induced by DSCAM deficiency. Our research demonstrates that DSCAM negatively modulates spine maturation, and that DSCAM deficiency leads to excessive spine maturation and autism-like behaviors, thus providing new insight into a potential pathophysiological mechanism of autism.


Subject(s)
Autism Spectrum Disorder/metabolism , Cell Adhesion Molecules/deficiency , Dendritic Spines/metabolism , Neurogenesis/physiology , Somatosensory Cortex/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , COS Cells , Cell Adhesion Molecules/genetics , Cells, Cultured , Chlorocebus aethiops , Dendritic Spines/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/pathology
6.
Sci Rep ; 11(1): 15738, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344918

ABSTRACT

The aim of this study was to investigate central pain representations during loading of the periodontium induced by orthodontic and occlusal stress. Nineteen healthy male volunteers (25.7 ± 2.8 years) were tested on two consecutive days: after phenotyping (questionnaires) and determination of warmth (WPT) and heat (HPT) pain thresholds, functional magnetic resonance imaging was performed as event-related paradigm including 36 tooth clenchings of 3 s duration, alternating with rest periods varying between 20-30 s. The task was performed in absence (T1) and 24 h after placement of an elastic separator between the second bicuspid and the first molar on the right side of the lower jaw (T2). No significant changes in WPT and HPT were observed but pain ratings were significantly elevated at T2. Significantly elevated activation at T2, as compared to T1, was found in bilateral sensorimotor cortex, bilateral secondary sensory cortex, supplementary motor area, right rolandic operculum, and bilateral insula. Our data show for the first time in humans that periodontal stimulation, as tested by tooth clenching in the presence of an elastic separator, goes along with specific expressions of pain at behavioral and neuronal network levels. Findings supplement the existing neuroimaging literature on odontogenic pain.


Subject(s)
Magnetic Resonance Imaging/methods , Pain, Postoperative/pathology , Periodontal Pocket/surgery , Somatosensory Cortex/pathology , Adult , Healthy Volunteers , Humans , Male , Young Adult
7.
Int J Mol Sci ; 22(12)2021 Jun 13.
Article in English | MEDLINE | ID: mdl-34199241

ABSTRACT

Traumatic brain injury (TBI) disrupts thalamic and cortical integrity. The effect of post-injury reorganization and plasticity in thalamocortical pathways on the functional outcome remains unclear. We evaluated whether TBI causes structural changes in the thalamocortical axonal projection terminals in the primary somatosensory cortex (S1) that lead to hyperexcitability. TBI was induced in adult male Sprague Dawley rats with lateral fluid-percussion injury. A virus carrying the fluorescent-tagged opsin channel rhodopsin 2 transgene was injected into the ventroposterior thalamus. We then traced the thalamocortical pathways and analyzed the reorganization of their axonal terminals in S1. Next, we optogenetically stimulated the thalamocortical relays from the ventral posterior lateral and medial nuclei to assess the post-TBI functionality of the pathway. Immunohistochemical analysis revealed that TBI did not alter the spatial distribution or lamina-specific targeting of projection terminals in S1. TBI reduced the axon terminal density in the motor cortex by 44% and in S1 by 30%. A nematic tensor-based analysis revealed that in control rats, the axon terminals in layer V were orientated perpendicular to the pial surface (60.3°). In TBI rats their orientation was more parallel to the pial surface (5.43°, difference between the groups p < 0.05). Moreover, the level of anisotropy of the axon terminals was high in controls (0.063) compared with TBI rats (0.045, p < 0.05). Optical stimulation of the sensory thalamus increased alpha activity in electroencephalography by 312% in controls (p > 0.05) and 237% (p > 0.05) in TBI rats compared with the baseline. However, only TBI rats showed increased beta activity (33%) with harmonics at 5 Hz. Our findings indicate that TBI induces reorganization of thalamocortical axonal terminals in the perilesional cortex, which alters responses to thalamic stimulation.


Subject(s)
Brain Injuries, Traumatic/pathology , Motor Cortex/pathology , Somatosensory Cortex/pathology , Thalamus/pathology , Animals , Anisotropy , Beta Rhythm/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Male , Optogenetics , Photic Stimulation , Rats, Sprague-Dawley
8.
J Neurosci Res ; 99(6): 1550-1564, 2021 06.
Article in English | MEDLINE | ID: mdl-33675112

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE) causes significant morbidity despite treatment with therapeutic hypothermia. Mitochondrial dysfunction may drive the mechanisms underlying neuronal cell death, thereby making mitochondria prime targets for neuroprotection. The mitochondrial permeability transition pore (mPTP) is one such target within mitochondria. In adult animal models, mPTP inhibition is neuroprotective. However, evidence for mPTP inhibition in neonatal models of neurologic disease is less certain. We tested the therapeutic efficacy of the mPTP small molecule inhibitor GNX-4728 and examined the developmental presence of brain mPTP proteins for drug targeting in a neonatal piglet model of hypoxic-ischemic brain injury. Male neonatal piglets were randomized to hypoxia-ischemia (HI) or sham procedure with GNX-4728 (15 mg/kg, IV) or vehicle (saline/cyclodextrin/DMSO, IV). GNX-4728 was administered as a single dose within 5 min after resuscitation from bradycardic arrest. Normal, ischemic, and injured neurons were counted in putamen and somatosensory cortex using hematoxylin and eosin staining. In separate neonatal and juvenile pigs, western blots of putamen mitochondrial-enriched fractions were used to evaluate mitochondrial integrity and the presence of mPTP proteins. We found that a single dose of GNX-4728 did not protect putamen and cortical neurons from cell death after HI. However, loss of mitochondrial matrix integrity occurred within 6h after HI, and while mPTP components are present in the neonatal brain their levels were significantly different compared to that of a mature juvenile brain. Thus, the neonatal brain mPTP may not be a good target for current neurotherapeutic drugs that are developed based on adult mitochondria.


Subject(s)
Asphyxia Neonatorum/prevention & control , Hypoxia-Ischemia, Brain/prevention & control , Mitochondrial Permeability Transition Pore , Neuroprotective Agents/therapeutic use , Animals , Animals, Newborn , Cell Death , Heart Arrest , Male , Putamen/pathology , Somatosensory Cortex/pathology , Swine
9.
J Clin Endocrinol Metab ; 106(6): 1566-1575, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33711158

ABSTRACT

CONTEXT: Although diabetic peripheral neuropathy (DPN) is predominantly considered a disorder of the peripheral nerves, some evidence for central nervous system involvement has recently emerged. However, whether or to what extent the microstructure of central somatosensory tracts may be injured remains unknown. OBJECTIVE: This work aimed to detect the microstructure of central somatosensory tracts in type 2 diabetic patients and to correlate it with the severity of DPN. METHODS: A case-control study at a tertiary referral hospital took place with 57 individuals with type 2 diabetes (25 with DPN, 32 without DPN) and 33 nondiabetic controls. The fractional anisotropy (FA) values of 2 major somatosensory tracts (the spinothalamic tract and its thalamocortical [spino-thalamo-cortical, STC] pathway, the medial lemniscus and its thalamocortical [medial lemnisco-thalamo-cortical, MLTC] pathway) were assessed based on diffusion tensor tractography. Regression models were further applied to detect the association of FA values with the severity of DPN in diabetic patients. RESULTS: The mean FA values of left STC and left MLTC pathways were significantly lower in patients with DPN than those without DPN and controls. Moreover, FA values of left STC and left MLTC pathways were significantly associated with the severity of DPN (expressed as Toronto Clinical Scoring System values) in patients after adjusting for multiple confounders. CONCLUSION: Our findings demonstrated the axonal degeneration of central somatosensory tracts in type 2 diabetic patients with DPN. The parallel disease progression of the intracranial and extracranial somatosensory system merits further attention to the central nerves in diabetic patients with DPN.


Subject(s)
Diabetic Neuropathies/pathology , Gray Matter/ultrastructure , Somatosensory Cortex/ultrastructure , Adult , Case-Control Studies , China , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/psychology , Diabetic Neuropathies/diagnosis , Diabetic Neuropathies/psychology , Female , Gray Matter/diagnostic imaging , Gray Matter/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Prognosis , Severity of Illness Index , Somatosensory Cortex/diagnostic imaging , Somatosensory Cortex/pathology
10.
J Neurotrauma ; 38(12): 1689-1701, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33427032

ABSTRACT

Traumatic brain injury (TBI) often induces structural damage, disruption of the blood-brain barrier (BBB), neurodegeneration, and dysfunctions of surviving neuronal networks. Nitric oxide (NO) signaling has been suggested to affect brain functions after TBI. The NO exhibits most of its biological effects by activation of the primary targets-guanylyl cyclases (NO-GCs), which exists in two isoforms (NO-GC1 and NO-GC2), and the subsequently produced cyclic guanosine monophosphate (cGMP). However, the specific function of the NO-NO-GCs-cGMP pathway in the context of brain injury is not fully understood. To investigate the specific role of the isoform NO-GC1 early after brain injuries, we performed an in vivo unilateral controlled cortical impact (CCI) in the somatosensory cortex of knockout mice lacking NO-GC1 and their wild-type (WT) littermates. Morphological and electrophysiological changes of cortical neurons located 500 µm distant from the lesion border were studied early (24 h) after TBI. The CCI-operated WT mice exhibited significant BBB disruption, an impairment of dendritic spine morphology, a reduced pre-synaptic glutamate release, and less neuronal activity in the ipsilateral cortical network. The impaired ipsilateral neuronal excitability was associated with increased A-type K+ currents (IA) in the WT mice early after TBI. Interestingly, NO-GC1 KO mice revealed relatively less BBB rupture and a weaker brain edema formation early after TBI. Further, lack of NO-GC1 also prevented the impaired synaptic transmission and network function that were observed in TBI-treated WT mice. These data suggest that NO-GC1 signaling mediates early brain damage and the strength of ipsilateral cortical network in the early phase after TBI.


Subject(s)
Brain Edema/pathology , Brain Injuries, Traumatic/pathology , Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Receptors, Cell Surface/metabolism , Synaptic Transmission/physiology , Animals , Brain Edema/etiology , Brain Injuries, Traumatic/complications , Cyclic GMP/metabolism , Isoenzymes/metabolism , Mice , Mice, Knockout , Signal Transduction/physiology , Somatosensory Cortex/injuries , Somatosensory Cortex/pathology
11.
Stroke ; 52(2): 687-698, 2021 01.
Article in English | MEDLINE | ID: mdl-33412903

ABSTRACT

BACKGROUND AND PURPOSE: Stroke disrupts neuronal functions in both local and remotely connected regions, leading to network-wide deficits that can hinder recovery. The thalamus is particularly affected, with progressive development of neurodegeneration accompanied by inflammatory responses. However, the complexity of the involved inflammatory responses is poorly understood. Herein we investigated the spatiotemporal changes in the secondary degenerative thalamus after cortical stroke, using targeted transcriptome approach in conjunction with histology and flow cytometry. METHODS: Cortical ischemic stroke was generated by permanent occlusion of the left middle cerebral artery in male C57BL6J mice. Neurodegeneration, neuroinflammatory responses, and microglial activation were examined in naive and stroke mice at from poststroke days (PD) 1 to 84, in both ipsilesional somatosensory cortex and ipsilesional thalamus. NanoString neuropathology panel (780 genes) was used to examine transcriptome changes at PD7 and PD28. Fluorescence activated cell sorting was used to collect CD11c+ microglia from ipsilesional thalamus, and gene expressions were validated by quantitative real-time polymerase chain reaction. RESULTS: Neurodegeneration in the thalamus was detected at PD7 and progressively worsened by PD28. This was accompanied by rapid microglial activation detected as early as PD1, which preceded the neurodegenerative changes. Transcriptome analysis showed higher number of differentially expressed genes in ipsilesional thalamus at PD28. Notably, neuroinflammation was the top activated pathway, and microglia was the most enriched cell type. Itgax (CD11c) was the most significantly increased gene, and its expression was highly detected in microglia. Flow-sorted CD11c+ microglia from degenerative thalamus indicated molecular signatures similar to neurodegenerative disease-associated microglia; these included downregulated Tmem119 and CX3CR1 and upregulated ApoE, Axl, LpL, CSF1, and Cst7. CONCLUSIONS: Our findings demonstrate the dynamic changes of microglia after stroke and highlight the importance of investigating stroke network-wide deficits. Importantly, we report the existence of a unique subtype of microglia (CD11c+) with neurodegenerative disease-associated microglia features in the degenerative thalamus after stroke.


Subject(s)
Cerebral Cortex/pathology , Microglia/pathology , Neurodegenerative Diseases/pathology , Stroke/complications , Stroke/pathology , Thalamic Diseases/etiology , Thalamic Diseases/pathology , Animals , CD11 Antigens/chemistry , Cerebrovascular Circulation , Encephalitis/pathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Somatosensory Cortex/pathology , Thalamus/pathology , Transcriptome
12.
Neuroimage ; 225: 117512, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33130274

ABSTRACT

Lower midlife physical activity is associated with higher risk of neurodegenerative disease in late life. However, it remains unknown whether physical exercise and fitness are associated with brain structural integrity during midlife. The purpose of this study was to compare brain structures between middle-aged aerobically trained adults (MA), middle-aged sedentary (MS), and young sedentary (YS) adults. Thirty MA (54±4 years), 30 MS (54±4 years), and 30 YS (32±6 years) participants (50% women) underwent measurements of brain volume, cortical thickness, and white matter (WM) fiber integrity using MRI. MA participants had aerobic training for 24.8±9.6 years and the highest cardiorespiratory fitness level (i.e., peak oxygen uptake: VO2peak) among all groups. Global WM integrity, as assessed with fractional anisotropy (FA) from diffusion tensor imaging, was lower in the MS compared with the YS group. However, global FA in the MA group was significantly higher than that in the MS group (P<0.05) and at a similar level to the YS group. Furthermore, tract-based spatial statistical analysis demonstrated that FA in the anterior, superior, and limbic WM tracts (e.g., the genu of the corpus callosum, superior longitudinal fasciculus, uncinate fasciculus) was higher in the MA compared with MS groups, and positively associated with VO2peak, independently from age and sex. From cortical thickness analysis, MS and MA participants showed thinner prefrontal and parieto-temporal areas than the YS group. On the other hand, the MA group exhibited thicker precentral, postcentral, pericalcarine, and lateral occipital cortices than the MS and YS groups. But, the insula and right superior frontal gyrus showed thinner cortical thickness in the MA compared with the MS groups. Collectively, these findings suggest that midlife aerobic exercise is associated with higher WM integrity and greater primary motor and somatosensory cortical thickness.


Subject(s)
Brain Cortical Thickness , Brain/diagnostic imaging , Cardiorespiratory Fitness , Exercise , Sedentary Behavior , White Matter/diagnostic imaging , Adult , Age Factors , Anisotropy , Brain/pathology , Diffusion Tensor Imaging , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Motor Cortex/diagnostic imaging , Motor Cortex/pathology , Neurodegenerative Diseases , Neuropsychological Tests , Organ Size , Oxygen Consumption , Risk Factors , Somatosensory Cortex/diagnostic imaging , Somatosensory Cortex/pathology , White Matter/pathology
13.
Epileptic Disord ; 22(4): 482-488, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32782231

ABSTRACT

We report a patient with reflex tooth-brushing-triggered epilepsy, associated with a post-central lesion within the right somatosensory face area. Contralateral facial sensory and motor phenomena, associated with contralateral upper limb extension, were present at seizure onset after gingival stimulation, but seizures could also be induced by contact with solid food or liquids. Spontaneous seizures also were recorded. Secondary generalization was infrequent. Stereoelectroencephalography implantation was performed, with seizure recording and cortical/subcortical stimulation for mapping, to identify the precise extent of surgical resection. Complete postoperative control of epilepsy was achieved, accompanied by a mild and transient neurological deficit. [Published with video sequence].


Subject(s)
Epilepsy, Reflex/diagnosis , Epilepsy, Reflex/surgery , Somatosensory Cortex , Adult , Electrocorticography , Female , Gingiva/physiopathology , Humans , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Somatosensory Cortex/surgery , Stereotaxic Techniques
14.
Neurobiol Aging ; 94: 89-100, 2020 10.
Article in English | MEDLINE | ID: mdl-32593032

ABSTRACT

An increasing number of neuroimaging studies addressing patients with corticobasal syndrome use macroscopic definitions of brain regions. As a closer link to functionally relevant units, we aimed at identifying magnetic resonance-based atrophy patterns in regions defined by probability maps of cortical microstructure. For this purpose, three analyses were conducted: (1) Whole-brain cortical thickness was compared between 36 patients with corticobasal syndrome and 24 controls. A pattern of pericentral atrophy was found, covering primary motor area 4, premotor area 6, and primary somatosensory areas 1, 2, and 3a. Within the central region, only area 3b was without atrophy. (2) In 18 patients, longitudinal measures with follow-ups of up to 59 months (mean 21.3 ± 15.4) were analyzed. Areas 1, 2, and 6 showed significantly faster atrophy rates than primary somatosensory area 3b. (3) In an individual autopsy case, longitudinal in vivo morphometry and postmortem pathohistology were conducted. The rate of magnetic resonance-based atrophy was significantly correlated with tufted-astrocyte load in those cytoarchitectonically defined regions also seen in the group study, with area 3b being selectively unaffected.


Subject(s)
Magnetic Resonance Imaging , Neurodegenerative Diseases/pathology , Parkinson Disease/pathology , Somatosensory Cortex/diagnostic imaging , Somatosensory Cortex/pathology , Aged , Astrocytes/pathology , Atrophy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Somatosensory Cortex/cytology , Syndrome
15.
Proc Natl Acad Sci U S A ; 117(16): 9094-9100, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32253308

ABSTRACT

Stem cell transplantation can improve behavioral recovery after stroke in animal models but whether stem cell-derived neurons become functionally integrated into stroke-injured brain circuitry is poorly understood. Here we show that intracortically grafted human induced pluripotent stem (iPS) cell-derived cortical neurons send widespread axonal projections to both hemispheres of rats with ischemic lesions in the cerebral cortex. Using rabies virus-based transsynaptic tracing, we find that at 6 mo after transplantation, host neurons in the contralateral somatosensory cortex receive monosynaptic inputs from grafted neurons. Immunoelectron microscopy demonstrates myelination of the graft-derived axons in the corpus callosum and that their terminals form excitatory, glutamatergic synapses on host cortical neurons. We show that the stroke-induced asymmetry in a sensorimotor (cylinder) test is reversed by transplantation. Light-induced inhibition of halorhodopsin-expressing, grafted neurons does not recreate the impairment, indicating that its reversal is not due to neuronal activity in the graft. However, we find bilateral decrease of motor performance in the cylinder test after light-induced inhibition of either grafted or endogenous halorhodopsin-expressing cortical neurons, located in the same area, and after inhibition of endogenous halorhodopsin-expressing cortical neurons by exposure of their axons to light on the contralateral side. Our data indicate that activity in the grafted neurons, probably mediated through transcallosal connections to the contralateral hemisphere, is involved in maintaining normal motor function. This is an example of functional integration of efferent projections from grafted neurons into the stroke-affected brain's neural circuitry, which raises the possibility that such repair might be achievable also in humans affected by stroke.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Infarction, Middle Cerebral Artery/therapy , Motor Activity/physiology , Neurons/transplantation , Somatosensory Cortex/physiopathology , Action Potentials/physiology , Animals , Behavior Observation Techniques , Behavior, Animal/physiology , Cell Differentiation/physiology , Cell Line , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/etiology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Neurons/physiology , Optogenetics , Patch-Clamp Techniques , Rats , Recovery of Function , Somatosensory Cortex/cytology , Somatosensory Cortex/pathology
16.
Genes (Basel) ; 11(4)2020 04 01.
Article in English | MEDLINE | ID: mdl-32244845

ABSTRACT

Impaired function of GABAergic interneurons, and the subsequent alteration of excitation/inhibition balance, is thought to contribute to autism spectrum disorders (ASD). Altered numbers of GABAergic interneurons and reduced expression of GABA receptors has been detected in the brain of ASD subjects and mouse models of ASD. We previously showed a reduced expression of GABAergic interneuron markers parvalbumin (PV) and somatostatin (SST) in the forebrain of adult mice lacking the Engrailed2 gene (En2-/- mice). Here, we extended this analysis to postnatal day (P) 30 by using in situ hybridization, immunohistochemistry, and quantitative RT-PCR to study the expression of GABAergic interneuron markers in the hippocampus and somatosensory cortex of En2-/- and wild type (WT) mice. In addition, GABA receptor subunit mRNA expression was investigated by quantitative RT-PCR in the same brain regions of P30 and adult En2-/- and WT mice. As observed in adult animals, PV and SST expression was decreased in En2-/- forebrain of P30 mice. The expression of GABA receptor subunits (including the ASD-relevant Gabrb3) was also altered in young and adult En2-/- forebrain. Our results suggest that GABAergic neurotransmission deficits are already evident at P30, confirming that neurodevelopmental defects of GABAergic interneurons occur in the En2 mouse model of ASD.


Subject(s)
Autism Spectrum Disorder/pathology , GABAergic Neurons/pathology , Gene Expression Regulation, Developmental , Hippocampus/pathology , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Receptors, GABA/metabolism , Somatosensory Cortex/pathology , Animals , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , Disease Models, Animal , Female , GABAergic Neurons/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Interneurons/pathology , Male , Mice , Mice, Knockout , Parvalbumins/metabolism , Somatosensory Cortex/metabolism , Somatostatin/metabolism
17.
Phys Ther ; 100(6): 946-962, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32201890

ABSTRACT

BACKGROUND: Pain is one of the main symptoms associated with spinal cord injury (SCI) and can be associated with changes to the central nervous system (CNS). PURPOSE: This article provides an overview of the evidence relating to CNS changes (structural and functional) associated with pain in SCIs. DATA SOURCES: A systematic review was performed, according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) recommendations, on PubMed, Embase, and Web of Science in March 2018. STUDY SELECTION: Studies were selected if they concerned changes in the CNS of patients with SCI, regardless of the type of imagery. DATA EXTRACTION: Data were extracted by 2 blinded reviewers. DATA SYNTHESIS: There is moderate evidence for impaired electroencephalographic function and metabolic abnormalities in the anterior cingulate in patients experiencing pain. There is preliminary evidence that patients with pain have morphological and functional changes to the somatosensory cortex and alterations to thalamic metabolism. There are conflicting data regarding the relationships between lesion characteristics and pain. In contrast, patients without pain can display protective neuroplasticity. LIMITATIONS AND CONCLUSION: Further studies are required to elucidate fully the relationships between pain and neuroplasticity in patients with SCIs. However, current evidence might support the use of physical therapist treatments targeting CNS plasticity in patients with SCI pain.


Subject(s)
Central Nervous System/pathology , Central Nervous System/physiopathology , Chronic Pain/etiology , Neuralgia/etiology , Spinal Cord Injuries/complications , Adult , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Brain/physiopathology , Central Nervous System/diagnostic imaging , Central Nervous System/metabolism , Chronic Pain/physiopathology , Electroencephalography/methods , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/physiopathology , Humans , Magnetic Resonance Imaging/methods , Neuralgia/physiopathology , Neuroimaging/methods , Neuronal Plasticity/physiology , Selection Bias , Somatosensory Cortex/diagnostic imaging , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Spinal Cord/diagnostic imaging , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/diagnostic imaging , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Thalamus/diagnostic imaging , Thalamus/metabolism
18.
Sci Rep ; 10(1): 3771, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111927

ABSTRACT

Primary motor cortex (M1) infarctions sometimes cause sensory impairment. Because sensory signals play a vital role in motor control, sensory impairment compromises the recovery and rehabilitation of motor disability. However, the neural mechanism of the sensory impairment is poorly understood. We show that sensory processing in mouse primary somatosensory cortex (S1) was impaired in the acute phase of M1 infarctions and recovered in a layer-specific manner in the subacute phase. This layer-dependent recovery process and the anatomical connection pattern from M1 to S1 suggested that functional connectivity from M1 to S1 plays a key role in the sensory processing impairment. A simulation study demonstrated that the loss of inhibition from M1 to S1 in the acute phase of M1 infarctions could impair sensory processing in S1, and compensation for the inhibition could recover the temporal coding. Consistently, the optogenetic activation of M1 suppressed the sustained response in S1. Taken together, we revealed how focal stroke in M1 alters the cortical network activity of sensory processing, in which inhibitory input from M1 to S1 may be involved.


Subject(s)
Brain Infarction/physiopathology , Sensation , Somatosensory Cortex/physiopathology , Stroke/physiopathology , Animals , Brain Infarction/pathology , Male , Mice , Optogenetics , Somatosensory Cortex/pathology , Stroke/pathology
19.
Brain Cogn ; 139: 105518, 2020 03.
Article in English | MEDLINE | ID: mdl-31954233

ABSTRACT

Excessive video gaming has a number of psychological and social consequences. In this study, we looked at possible changes in gray and white matter and asked whether these changes are correlated to psychological measures. Twentynine players of violent videogames (mean daily playing time 4.7 h) and age matched controls were subjected to a battery of questionnaires assessing aggression, empathy, hostility, internet addiction and psychological well-being. Diffusion tensor and 3D T1-weighted MR images were obtained to examine gray (via voxel-based morphometry) and white (via tract-based spatial statistics) matter changes. Widespread regions of decreased gray matter in the players were found but no region showed increased intensity of gray matter. Density of gray matter showed a negative correlation with the total length of playing in years in the right posterior cingulate gyrus, left pre- and postcentral gyrus, right thalamus, among others. Furthermore, fractional anisotropy, a marker for white matter structure, was decreased in the left and right cingulum in the players. Both, gray and white matter changes correlated with measures of aggression, hostility, self esteem, and the degree of internet addiction. This study thus shows profound changes of brain structure as a function of excessive playing of violent video games.


Subject(s)
Behavior, Addictive/diagnostic imaging , Brain/diagnostic imaging , Internet , Video Games , Adolescent , Adult , Anisotropy , Behavior, Addictive/psychology , Brain/pathology , Case-Control Studies , Frontal Lobe/diagnostic imaging , Frontal Lobe/pathology , Gray Matter/diagnostic imaging , Gray Matter/pathology , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/pathology , Humans , Magnetic Resonance Imaging , Male , Organ Size , Self Concept , Somatosensory Cortex/diagnostic imaging , Somatosensory Cortex/pathology , Thalamus/diagnostic imaging , Thalamus/pathology , White Matter/diagnostic imaging , White Matter/pathology , Young Adult
20.
Mol Psychiatry ; 25(5): 1112-1129, 2020 05.
Article in English | MEDLINE | ID: mdl-31431686

ABSTRACT

In mood disorders, psychomotor and sensory abnormalities are prevalent, disabling, and intertwined with emotional and cognitive symptoms. Corticostriatal neurons in motor and somatosensory cortex are implicated in these symptoms, yet mechanisms of their vulnerability are unknown. Here, we demonstrate that S100a10 corticostriatal neurons exhibit distinct serotonin responses and have increased excitability, compared with S100a10-negative neurons. We reveal that prolonged social isolation disrupts the specific serotonin response which gets restored by chronic antidepressant treatment. We identify cell-type-specific transcriptional signatures in S100a10 neurons that contribute to serotonin responses and strongly associate with psychomotor and somatosensory function. Our studies provide a strong framework to understand the pathogenesis and create new avenues for the treatment of mood disorders.


Subject(s)
Annexin A2/metabolism , Antidepressive Agents/pharmacology , Neurons/drug effects , Neurons/metabolism , S100 Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/metabolism , Animals , Biomarkers/metabolism , Male , Mice , Motor Cortex/pathology , Serotonin/metabolism , Somatosensory Cortex/pathology , Stress, Psychological/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...