Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
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
2.
J Comp Neurol ; 526(15): 2373-2387, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30014461

ABSTRACT

The corticospinal tract (CST) forms the major descending pathway mediating voluntary hand movements in primates, and originates from ∼nine cortical subdivisions in the macaque. While the terminals of spared motor CST axons are known to sprout locally within the cord in response to spinal injury, little is known about the response of the other CST subcomponents. We previously reported that following a cervical dorsal root lesion (DRL), the primary somatosensory (S1) CST terminal projection retracts to 60% of its original terminal domain, while the primary motor (M1) projection remains robust (Darian-Smith et al., J. Neurosci., 2013). In contrast, when a dorsal column lesion (DCL) is added to the DRL, the S1 CST, in addition to the M1 CST, extends its terminal projections bilaterally and caudally, well beyond normal range (Darian-Smith et al., J. Neurosci., 2014). Are these dramatic responses linked entirely to the inclusion of a CNS injury (i.e., DCL), or do the two components summate or interact? We addressed this directly, by comparing data from monkeys that received a unilateral DCL alone, with those that received either a DRL or a combined DRL/DCL. Approximately 4 months post-lesion, the S1 hand region was mapped electrophysiologically, and anterograde tracers were injected bilaterally into the region deprived of normal input, to assess spinal terminal labeling. Using multifactorial analyses, we show that following a DCL alone (i.e., cuneate fasciculus lesion), the S1 and M1 CSTs also sprout significantly and bilaterally beyond normal range, with a termination pattern suggesting some interaction between the peripheral and central lesions.


Subject(s)
Motor Cortex/pathology , Pyramidal Tracts/pathology , Somatosensory Cortex/pathology , Spinal Cord Injuries/pathology , Spinal Nerve Roots/pathology , Animals , Behavior, Animal , Brain Mapping , Gray Matter/pathology , Hand/innervation , Hand Strength , Macaca fascicularis , Male , Motor Cortex/injuries , Motor Skills Disorders/pathology , Pyramidal Tracts/injuries , Recovery of Function/physiology , Somatosensory Cortex/injuries , Spinal Cord Injuries/psychology , Spinal Nerve Roots/injuries , Touch
3.
Glia ; 66(4): 828-845, 2018 04.
Article in English | MEDLINE | ID: mdl-29288586

ABSTRACT

Microglia, the immune cells of the central nervous system, continuously survey the brain to detect alterations and maintain tissue homeostasis. The motility of microglial processes is indicative of their surveying capacity in normal and pathological conditions. The gold standard technique to study motility involves the use of two-photon microscopy to obtain time-lapse images from brain slices or the cortex of living animals. This technique generates four dimensionally-coded images which are analyzed manually using time-consuming, non-standardized protocols. Microglial process motility analysis is frequently performed using Z-stack projections with the consequent loss of three-dimensional (3D) information. To overcome these limitations, we developed ProMoIJ, a pack of ImageJ macros that perform automatic motility analysis of cellular processes in 3D. The main core of ProMoIJ is formed by two macros that assist the selection of processes, automatically reconstruct their 3D skeleton, and analyze their motility (process and tip velocity). Our results show that ProMoIJ presents several key advantages compared with conventional manual analysis: (1) reduces the time required for analysis, (2) is less sensitive to experimenter bias, and (3) is more robust to varying numbers of processes analyzed. In addition, we used ProMoIJ to demonstrate that commonly performed 2D analysis underestimates microglial process motility, to reveal that only cells adjacent to a laser injured area extend their processes toward the lesion site, and to demonstrate that systemic inflammation reduces microglial process motility. ProMoIJ is a novel, open-source, freely-available tool which standardizes and accelerates the time-consuming labor of 3D analysis of microglial process motility.


Subject(s)
Imaging, Three-Dimensional/methods , Microglia/cytology , Pattern Recognition, Automated/methods , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Lasers , Mice, Transgenic , Motor Cortex/cytology , Software , Somatosensory Cortex/cytology , Somatosensory Cortex/injuries , Somatosensory Cortex/pathology
4.
Neurosci Lett ; 665: 147-151, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29217256

ABSTRACT

OBJECTIVE: The parieto-insular vestibular cortex (PIVC) is a core region of vestibular input into regions of the cortex. The vestibular nuclei have reciprocal connections with the PIVC. However, little is known about injury of the core vestibular pathway to the PIVC in patients with dorsolateral medullary infarctions. In this study, using diffusion tensor tractography (DTT), we investigated injury of the neural connections between the vestibular nuclei and the PIVC in patients with typical central vestibular disorder. METHODS: Eight consecutive patients with lateral medullary syndrome and 10 control subjects were recruited for this study. To reconstruct the core vestibular pathway to the PIVC, we defined the seed region of interest (ROI) as the vestibular nuclei of the pons and the target ROI as the PIVC. Fractional anisotropy (FA), mean diffusivity (MD), and tract volume were measured. RESULT: The core vestibular pathway to the PIVC showed significantly lower tract volume in patients compared with the control group (p<0.05). By contrast, other DTI parameters did not show significant differences between the patient and control groups (p>0.05). CONCLUSION: In conclusion, injury of the core vestibular pathway to the PIVC was demonstrated in patients with lateral vestibular syndrome following dorsolateral medullary infarcts. We believe that analysis of the core vestibular pathway to the PIVC using DTT would be helpful in evaluating patients with lateral medullary syndrome.


Subject(s)
Brain Mapping , Cerebral Cortex/diagnostic imaging , Lateral Medullary Syndrome/diagnostic imaging , Somatosensory Cortex/diagnostic imaging , Vestibule, Labyrinth/diagnostic imaging , Adult , Aged , Brain Mapping/methods , Cerebral Cortex/injuries , Diffusion Tensor Imaging/methods , Female , Humans , Male , Middle Aged , Parietal Lobe/diagnostic imaging , Parietal Lobe/injuries , Physical Stimulation/methods , Somatosensory Cortex/injuries
5.
Brain Struct Funct ; 223(2): 851-872, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28993883

ABSTRACT

Massive corticothalamic afferents originating from layer 6a of primary sensory cortical areas modulate sensory responsiveness of thalamocortical neurons and are pivotal for shifting neuronal firing between burst and tonic modes. The influence of the corticothalamic pathways on the firing mode and sensory gain of thalamic neurons has only been extensively examined in anesthetized animals, but has yet to be established in the awake state. We made lesions of the rat barrel cortex and on the following day recorded responses of single thalamocortical and thalamic reticular neurons to a single vibrissal deflection in the somatosensory system during wakefulness. Our results showed that the cortical lesions shifted the response of thalamic neurons towards bursting, elevated the response probability and the gain of thalamocortical neurons, predominantly of recurring responses. In addition, after the lesions, the spontaneous activities of the vibrissa-responsive thalamic neurons, but not those of vibrissa-unresponsive cells, were typified by waxing-and-waning spindle-like rhythmic spiking with frequent bursting. In awake rats with intact cortex, identified layer 6a corticothalamic neurons responded to a single vibrissal deflection with short latencies that matched those of layer 4 neurons, strongly suggesting the existence of an immediate corticothalamic feedback. The present results show the importance of corticothalamic neurons in shaping thalamic activities during wakefulness.


Subject(s)
Neural Pathways/physiology , Neurons/physiology , Somatosensory Cortex/physiology , Thalamus/cytology , Wakefulness/physiology , Action Potentials/physiology , Animals , Electric Stimulation , Electrocardiography , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Physical Stimulation , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Rhodamines , Somatosensory Cortex/cytology , Somatosensory Cortex/injuries , Ventral Thalamic Nuclei/injuries , Vibrissae/innervation , Water Deprivation/physiology
6.
Neuroscience ; 362: 79-94, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-28827177

ABSTRACT

Trauma and tumor compressing the brain distort underlying cortical neurons. Compressed cortical neurons remodel their dendrites instantly. The effects on axons however remain unclear. Using a rat epidural bead implantation model, we studied the effects of unilateral somatosensory cortical compression on its transcallosal projection and the reversibility of the changes following decompression. Compression reduced the density, branching profuseness and boutons of the projection axons in the contralateral homotopic cortex 1week and 1month post-compression. Projection fiber density was higher 1-month than 1-week post-compression, suggesting adaptive temporal changes. Compression reduced contralateral cortical synaptophysin, vesicular glutamate transporter 1 (VGLUT1) and postsynaptic density protein-95 (PSD95) expressions in a week and the first two marker proteins further by 1month. ßIII-tubulin and kinesin light chain (KLC) expressions in the corpus callosum (CC) where transcallosal axons traveled were also decreased. Kinesin heavy chain (KHC) level in CC was temporarily increased 1week after compression. Decompression increased transcallosal axon density and branching profuseness to higher than sham while bouton density returned to sham levels. This was accompanied by restoration of synaptophysin, VGLUT1 and PSD95 expressions in the contralateral cortex of the 1-week, but not the 1-month, compression rats. Decompression restored ßIII-tubulin, but not KLC and KHC expressions in CC. However, KLC and KHC expressions in the cell bodies of the layer II/III pyramidal neurons partially recovered. Our results show cerebral compression compromised cortical axonal outputs and reduced transcallosal projection. Some of these changes did not recover in long-term decompression.


Subject(s)
Axonal Transport/physiology , Corpus Callosum/injuries , Corpus Callosum/metabolism , Neurons/metabolism , Somatosensory Cortex/injuries , Somatosensory Cortex/metabolism , Animals , Corpus Callosum/pathology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Functional Laterality , Kinesins/metabolism , Male , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuronal Plasticity/physiology , Neurons/pathology , Rats, Wistar , Somatosensory Cortex/pathology , Synaptophysin/metabolism , Tubulin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
7.
Cereb Cortex ; 27(8): 4213-4228, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28472290

ABSTRACT

Aging leads to adverse outcomes after traumatic brain injury. The mechanisms underlying these defects, however, are not yet clear. In this study, we found that astrocytes in the aged post-traumatic cerebral cortex develop a significantly reduced proliferative response, resulting in reduced astrocyte numbers in the penumbra. Moreover, experiments of reactive astrocytes in vitro reveal that their diminished proliferation is due to an age-related switch in the division mode with reduced cell-cycle re-entry rather than changes in cell-cycle length. Notably, reactive astrocytes in vivo and in vitro become refractory to stimuli increasing their proliferation during aging, such as Sonic hedgehog signaling. These data demonstrate for the first time that age-dependent, most likely intrinsic changes in the proliferative program of reactive astrocytes result in their severely hampered proliferative response to traumatic injury thereby affecting astrocyte homeostasis.


Subject(s)
Aging/physiology , Astrocytes/physiology , Brain Injuries/physiopathology , Cell Proliferation/physiology , Homeostasis/physiology , Somatosensory Cortex/physiopathology , Aging/pathology , Animals , Astrocytes/pathology , Brain Injuries/pathology , Cells, Cultured , Disease Models, Animal , Gliosis/pathology , Gliosis/physiopathology , Hedgehog Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Somatosensory Cortex/injuries , Somatosensory Cortex/pathology , Wounds, Stab
8.
Acta Neurobiol Exp (Wars) ; 77(1): 1-17, 2017.
Article in English | MEDLINE | ID: mdl-28379212

ABSTRACT

A number of neurological disorders such as epidural hematoma can cause compression of cerebral cortex. We here tested the hypothesis that sustained compression of primary somatosensory cortex may affect stellate neurons and thalamocortical afferent (TCA) fibers. A rat model with barrel cortex subjected to bead epidural compression was used. Golgi-Cox staining analyses showed the shrinkage of dendritic arbors and the stripping of dendritic spines of stellate neurons for at least 3 months post-lesion. Anterograde tracing analyses exhibited a progressive decline of TCA fiber density in barrel field for 6 months post-lesion. Due to the abrupt decrease of TCA fiber density at 3 days after compression, we further used electron microscopy to investigate the ultrastructure of TCA fibers at this time. Some TCA fiber terminal profiles with dissolved or darkened mitochondria and fewer synaptic vesicles were distorted and broken. Furthermore, the disruption of mitochondria and myelin sheath was observed in some myelinated TCA fibers. In addition, expressions of oxidative markers 3-nitrotyrosine and 4-hydroxynonenal were elevated in barrel field post-lesion. Treatment of antioxidant ascorbic acid or apocynin was able to reverse the increase of oxidative stress and the decline of TCA fiber density, rather than the shrinkage of dendrites and the stripping of dendritic spines of stellate neurons post-lesion. Together, these results indicate that sustained epidural compression of primary somatosensory cortex affects the TCA fibers and the dendrites of stellate neurons for a prolonged period. In addition, oxidative stress is responsible for the reduction of TCA fiber density in barrels rather than the shrinkage of dendrites and the stripping of dendritic spines of stellate neurons.


Subject(s)
Afferent Pathways/pathology , Brain Injuries/pathology , Epidural Space , Neurons/pathology , Somatosensory Cortex/pathology , Thalamus/pathology , Acetophenones/therapeutic use , Aldehydes/metabolism , Animals , Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Biotin/analogs & derivatives , Biotin/pharmacokinetics , Brain Injuries/drug therapy , Dendrites/pathology , Dendrites/ultrastructure , Dextrans/pharmacokinetics , Disease Models, Animal , Electron Transport Complex IV/metabolism , Epidural Space/physiology , Functional Laterality , Male , Neurons/ultrastructure , Oxidative Stress/physiology , Rats , Somatosensory Cortex/injuries , Thalamus/ultrastructure , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
J Neurotrauma ; 34(5): 1097-1110, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27650335

ABSTRACT

Focal neocortical brain injuries lead to functional alterations, which can spread beyond lesion-neighboring brain areas. The undamaged hemisphere and its associated disturbances after a unilateral lesion, so-called transhemispheric diaschisis, have been progressively disclosed over the last decades; they are strongly involved in the pathophysiology and, potentially, recovery of brain injuries. Understanding the temporal dynamics of these transhemispheric functional changes is crucial to decipher the role of the undamaged cortex in the processes of functional reorganization at different stages post-lesion. In this regard, little is known about the acute-subacute processes after 24-48 h in the brain hemisphere contralateral to injury. In the present study, we performed a controlled cortical impact to produce a unilateral traumatic brain injury (TBI) in the motor and somatosensory cortex of mice. In vitro extracellular multi-unit recordings from large neuronal populations, together with single-cell patch-clamp recordings in the cortical network contralateral to the lesion, revealed a strong, but transient, neuronal hyperactivity as early as 24-48 h post-TBI. This abnormal excitable state in the intact hemisphere was not accompanied by alterations in neuronal intrinsic properties, but it was associated with an impairment of the phasic gamma aminobutyric acid (GABA)ergic transmission and an increased expression of GABAA receptor subunits related to tonic inhibition exclusively in the contralateral hemisphere. These data unravel a series of early transhemispheric functional alterations after diffuse unilateral cortical injury, which may compensate and stabilize the disrupted brain functions. Therefore, our findings support the hypothesis that the undamaged hemisphere could play a significant role in early functional reorganization processes after a TBI.


Subject(s)
Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Motor Cortex/injuries , Motor Cortex/physiopathology , Somatosensory Cortex/injuries , Somatosensory Cortex/physiopathology , Animals , Disease Models, Animal , Electroencephalography , Mice, Inbred C57BL , Neuronal Plasticity , Patch-Clamp Techniques
10.
Neuroscience ; 334: 290-308, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27530700

ABSTRACT

Traumatic brain injury (TBI) is a major cause of morbidity and mortality world-wide and can result in persistent cognitive, sensory and behavioral dysfunction. Understanding the time course of TBI-induced pathology is essential to effective treatment outcomes. We induced TBI in rats using an impact acceleration method and tested for sensorimotor skill and sensory sensitivity behaviors for two weeks to find persistently poor outcomes post-injury. At two weeks post-injury we made high resolution extracellular recordings from barrel cortex neurons, to simple and complex whisker deflections. We found that the supragranular suppression of neural firing (compared to normal) previously seen in the immediate post-TBI aftermath had spread to include suppression of input and infragranular layers at two weeks post-injury; thus, there was suppression of whisker-driven firing rates in all cortical layers to both stimulus types. Further, there were abnormalities in temporal response patterns such that in layers 3-5 there was a temporal broadening of response patterns in response to both whisker deflection stimulus types and in L2 a narrowing of temporal patterns in response to the complex stimulus. Thus, at two weeks post-TBI, supragranular hypo-excitation has evolved to include deep cortical layers likely as a function of progressive atrophy and neurodegeneration. These results are consistent with the hypothesis that TBI alters the delicate excitatory/inhibitory balance in cortex and likely contributes to temporal broadening of responses and restricts the ability to code for complex sensory stimuli.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Neurons/physiology , Somatosensory Cortex/physiopathology , Action Potentials , Animals , Disease Models, Animal , Disease Progression , Male , Microelectrodes , Motor Activity/physiology , Rats, Sprague-Dawley , Somatosensory Cortex/injuries , Time Factors , Touch Perception/physiology , Vibrissae/physiology
11.
Neurosci Lett ; 628: 171-8, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27345383

ABSTRACT

Indwelling cannulas are often used to deliver pharmacological agents into the lateral ventricles of the brain to study their effects on memory and learning, yet little is known about the possible adverse effects of the cannulation itself. In this study, the effect of implanting an indwelling cannula into the right lateral ventricle was examined with respect to cognitive function and tissue damage in rats. Specifically, the cannula passed through sections of the primary motor (M1) and somatosensory hind limb (S1HL) cortices. One week following implantation, rats were impaired on the rotarod task, implying a deficit in fine motor control, likely caused by the passage of the cannula through the aforementioned cortical regions. Importantly, neither spatial working nor recognition memory was adversely affected. Histological examination showed immune cell activation only in the area immediately surrounding the cannulation site and not spreading to other brain regions. Both GFAP and CD-11b mRNA expression was elevated in the area immediately surrounding the cannulation site, but not in the contralateral hemisphere or the hippocampus. Neither of the inflammatory cytokines, TNF-α or IL-6, were upregulated in any region. These results show that cannulation into the lateral ventricle does not impair cognition and indicates that nootropic agents delivered via this method are enhancing normal memory rather than rescuing deficits caused by the surgery procedure.


Subject(s)
Catheterization/adverse effects , Lateral Ventricles/injuries , Memory, Short-Term , Recognition, Psychology , Spatial Memory , Animals , Cannula/adverse effects , Glial Fibrillary Acidic Protein/metabolism , Inflammation , Interleukin-6/metabolism , Male , Motor Cortex/injuries , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/injuries , Tumor Necrosis Factor-alpha/metabolism
12.
J Neurotrauma ; 33(2): 194-202, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-25961471

ABSTRACT

Traumatic brain injury (TBI) remains a primary cause of death and disability in both civilian and military populations worldwide. There is a critical need for the development of neuroprotective agents that can circumvent damage and provide functional recovery. We previously showed that methylene blue (MB), a U.S. Food and Drug Administration-grandfathered drug with energy-enhancing and antioxidant properties, given 1 and 3 h post-TBI, had neuroprotective effects in rats. This study aimed to further investigate the neuroprotection of delayed MB treatment (24 h postinjury) post-TBI as measured by lesion volume and functional outcomes. Comparisons were made with vehicle and acute MB treatment. Multi-modal magnetic resonance imaging and behavioral studies were performed at 1 and 3 h and 2, 7, and 14 days after an impact to the primary forelimb somatosensory cortex. We found that delaying MB treatment 24 h postinjury still minimized lesion volume and functional deficits, compared to vehicle-treated animals. The data further support the potential for MB as a neuroprotective treatment, especially when medical teatment is not readily available. MB has an excellent safety profile and is clinically approved for other indications. MB clinical trials on TBI can thus be readily explored.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries/drug therapy , Enzyme Inhibitors/pharmacology , Methylene Blue/pharmacology , Neuroprotective Agents/pharmacology , Somatosensory Cortex/injuries , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Forelimb , Magnetic Resonance Imaging , Male , Methylene Blue/administration & dosage , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
13.
Glia ; 63(12): 2340-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26250529

ABSTRACT

Astrocytes react to brain injury in a heterogeneous manner with only a subset resuming proliferation and acquiring stem cell properties in vitro. In order to identify novel regulators of this subset, we performed genomewide expression analysis of reactive astrocytes isolated 5 days after stab wound injury from the gray matter of adult mouse cerebral cortex. The expression pattern was compared with astrocytes from intact cortex and adult neural stem cells (NSCs) isolated from the subependymal zone (SEZ). These comparisons revealed a set of genes expressed at higher levels in both endogenous NSCs and reactive astrocytes, including two lectins-Galectins 1 and 3. These results and the pattern of Galectin expression in the lesioned brain led us to examine the functional significance of these lectins in brains of mice lacking Galectins 1 and 3. Following stab wound injury, astrocyte reactivity including glial fibrillary acidic protein expression, proliferation and neurosphere-forming capacity were found significantly reduced in mutant animals. This phenotype could be recapitulated in vitro and was fully rescued by addition of Galectin 3, but not of Galectin 1. Thus, Galectins 1 and 3 play key roles in regulating the proliferative and NSC potential of a subset of reactive astrocytes.


Subject(s)
Astrocytes/metabolism , Galectin 1/metabolism , Galectin 3/metabolism , Somatosensory Cortex/injuries , Somatosensory Cortex/metabolism , Animals , Astrocytes/pathology , Cell Proliferation/physiology , Cells, Cultured , Disease Models, Animal , Galectin 1/genetics , Galectin 3/genetics , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Gray Matter/injuries , Gray Matter/metabolism , Gray Matter/pathology , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Somatosensory Cortex/pathology , Stem Cell Niche/physiology
14.
Neurobiol Dis ; 71: 305-16, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25158291

ABSTRACT

Developmental cortical malformations are associated with a high incidence of drug-resistant epilepsy. The underlying epileptogenic mechanisms, however, are poorly understood. In rodents, cortical malformations can be modeled using neonatal freeze-lesion (FL), which has been shown to cause in vitro cortical hyperexcitability. Here, we investigated the therapeutic potential of gabapentin, a clinically used anticonvulsant and analgesic, in preventing FL-induced in vitro and in vivo hyperexcitability. Gabapentin has been shown to disrupt the interaction of thrombospondin (TSP) with α2δ-1, an auxiliary calcium channel subunit. TSP/α2δ-1 signaling has been shown to drive the formation of excitatory synapses during cortical development and following injury. Gabapentin has been reported to have neuroprotective and anti-epileptogenic effects in other models associated with increased TSP expression and reactive astrocytosis. We found that both TSP and α2δ-1 were transiently upregulated following neonatal FL. We therefore designed a one-week GBP treatment paradigm to block TSP/α2δ-1 signaling during the period of their upregulation. GBP treatment prevented epileptiform activity following FL, as assessed by both glutamate biosensor imaging and field potential recording. GBP also attenuated FL-induced increases in mEPSC frequency at both P7 and 28. Additionally, GBP treated animals had decreased in vivo kainic acid (KA)-induced seizure activity. Taken together these results suggest gabapentin treatment immediately after FL can prevent the formation of a hyperexcitable network and may have therapeutic potential to minimize epileptogenic processes associated with developmental cortical malformations.


Subject(s)
Amines/therapeutic use , Anticonvulsants/therapeutic use , Cyclohexanecarboxylic Acids/therapeutic use , Epilepsy/drug therapy , Epilepsy/etiology , Malformations of Cortical Development/complications , Somatosensory Cortex/injuries , gamma-Aminobutyric Acid/therapeutic use , Age Factors , Animals , Animals, Newborn , Calcium Channels/metabolism , Disease Models, Animal , Electric Stimulation , Evoked Potentials/drug effects , Excitatory Amino Acid Agonists/toxicity , Excitatory Postsynaptic Potentials/drug effects , Freezing/adverse effects , Gabapentin , Glial Fibrillary Acidic Protein , Glutamic Acid/metabolism , In Vitro Techniques , Kainic Acid/toxicity , Malformations of Cortical Development/etiology , Mice , Mice, Inbred C57BL , Neuroimaging , Patch-Clamp Techniques , Somatosensory Cortex/growth & development , Thrombospondins/metabolism
15.
Curr Biol ; 24(6): 677-80, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24613304

ABSTRACT

Studies in nonhuman primates have provided evidence of rapid neural reorganization in somatosensory cortex after brain damage [1] and amputation [2]. Furthermore, there is also evidence of experience-dependent plasticity in both human [3-5] and nonhuman primates [6] that is induced by repetitive tactile stimulation. Given the evidence of plasticity subsequent to both neural damage and tactile experience, we hypothesized that somatosensory damage could lead to increased levels of experience-dependent tactile plasticity. To examine this hypothesis, the tactile localization judgments of two individuals with left hemisphere somatosensory damage subsequent to stroke were examined. Suprathreshold tactile stimuli were presented to the hand or forearm, and the effect of the location of previous stimulation on localization judgments for subsequent stimuli was examined. Results showed that, only on the contralesional limb, even a single tactile stimulation could induce a significant perceptual shift in localization judgments for subsequent stimuli, with shifts occurring in the direction of the preceding stimulation. These results provide novel evidence of a very rapid time course for substantive perceptual changes in tactile location perception in response to simple stimulation, revealing a highly plastic and dynamic tactile system even many years after neural damage.


Subject(s)
Neuronal Plasticity/physiology , Somatosensory Cortex/injuries , Touch/physiology , Aged , Aged, 80 and over , Forearm , Hand , Humans , Judgment/physiology , Male , Middle Aged , Physical Stimulation , Somatosensory Cortex/physiopathology
16.
Headache ; 52(6): 966-84, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22568499

ABSTRACT

OBJECTIVE: This study tests the hypothesis that injury to the somatosensory cortex is associated with periorbital allodynia and increases in nociceptive neuropeptides in the brainstem in a mouse model of controlled cortical impact (CCI) injury. METHODS: Male C57BL/6 mice received either CCI or craniotomy-only followed by weekly periorbital von Frey (mechanical) sensory testing for up to 28 days post-injury. Mice receiving an incision only and naïve mice were included as control groups. Changes in calcitonin gene-related peptide (CGRP) and substance P (SP) within the brainstem were determined using enzyme-linked immunosorbent assay and immunohistochemistry, respectively. Activation of ionized calcium-binding adaptor molecule-1-labeled macrophages/microglia and glial fibrillary acidic protein (GFAP)-positive astrocytes were evaluated using immunohistochemistry because of their potential involvement in nociceptor sensitization. RESULTS: Incision-only control mice showed no changes from baseline periorbital von Frey mechanical thresholds. CCI significantly reduced mean periorbital von Frey thresholds (periorbital allodynia) compared with baseline and craniotomy-only at each endpoint, analysis of variance P < .0001. Craniotomy significantly reduced periorbital threshold at 14 days but not 7, 21, or 28 days compared with baseline threshold, P < .01. CCI significantly increased SP immunoreactivity in the brainstem at 7 and 14 days but not 28 days compared with craniotomy-only and controls, P < .001. CGRP levels in brainstem tissues were significantly increased in CCI groups compared with controls (incision-only and naïve mice) or craniotomy-only mice at each endpoint examined, P < .0001. There was a significant correlation between CGRP and periorbital allodynia (P < .0001, r = -0.65) but not for SP (r = 0.20). CCI significantly increased the number of macrophage/microglia in the injured cortex at each endpoint up to 28 days, although cell numbers declined over weeks post-injury, P < .001. GFAP(+) immunoreactivity was significantly increased at 7 but not 14 or 28 days after CCI, P < .001. Craniotomy resulted in transient periorbital allodynia accompanied by transient increases in SP, CGRP, and GFAP immunoreactivity compared with control mice. There was no increase in the number of macrophage/microglia cells compared with controls after craniotomy. CONCLUSION: Injury to the somatosensory cortex results in persistent periorbital allodynia and increases in brainstem nociceptive neuropeptides. Findings suggest that persistent allodynia and increased neuropeptides are maintained by mechanisms other than activation of macrophage/microglia or astrocyte in the injured somatosensory cortex.


Subject(s)
Brain Injuries/complications , Headache/etiology , Hyperalgesia/etiology , Neuropeptides/biosynthesis , Animals , Brain Injuries/metabolism , Brain Stem/chemistry , Brain Stem/metabolism , Calcitonin Gene-Related Peptide/biosynthesis , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Headache/metabolism , Hyperalgesia/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nociception/physiology , Somatosensory Cortex/injuries , Substance P/biosynthesis
17.
J Plast Surg Hand Surg ; 46(2): 106-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22471259

ABSTRACT

Injury to the peripheral nerves in the upper extremity results in changes in the nerve, and at multiple sites throughout the central nervous system (CNS). We studied the long-term effects of an injury to the median nerve in the forearm with a focus on changes in the CNS. Four patients with isolated injuries of the median nerve in their 20s were examined a mean of 14 years after the injury. Cortical activation was monitored during tactile stimulation of the fingers of the injured and healthy hand using functional magnetic resonance imaging at 3 Tesla. The neurophysiological state and clinical outcome were also examined. Activation in the primary somatosensory cortex was substantially larger during tactile stimulation of the injured hand than with stimulation of the uninjured hand. We also saw a redistribution of hemispheric dominance. Stimulation of the injured median nerve resulted in a substantially increased dominance of the contralateral hemisphere. However, stimulation of the healthy ulnar nerve resulted in a decreased dominance of the contralateral hemisphere. Neurophysiology showed low sensory amplitudes, velocity, and increased motor latency in the injured nerve. Clinically there were abnormalities predominately in the sensory domain. However, there was an overall improved mean result compared with a five year follow-up in the same subjects. The cortical changes could be the result of cortical reorganisation after a changed afferent signal pattern from the injured nerve. Even though the clinical function improved over time it did not return to normal, and neither did the cortical response.


Subject(s)
Magnetic Resonance Imaging/methods , Median Nerve/injuries , Median Neuropathy/diagnosis , Somatosensory Cortex/physiopathology , Somatosensory Disorders/diagnosis , Adolescent , Adult , Electric Stimulation/methods , Female , Follow-Up Studies , Humans , Incidence , Injury Severity Score , Male , Median Nerve/pathology , Median Nerve/surgery , Median Neuropathy/complications , Nerve Regeneration/physiology , Neuropsychological Tests , Neurosurgical Procedures/adverse effects , Neurosurgical Procedures/methods , Physical Examination/methods , Risk Assessment , Sensory Thresholds/physiology , Somatosensory Cortex/injuries , Somatosensory Disorders/epidemiology , Somatosensory Disorders/etiology , Time , Wrist Injuries/complications , Wrist Injuries/surgery , Young Adult
18.
Article in English | MEDLINE | ID: mdl-23366002

ABSTRACT

The long-term performance of chronic microelectrode array implants for neural ensemble recording is affected by temporal degradation in signal quality due to several factors including structural changes in the recording surface, electrical responses, and tissue immune reactivity. This study combines the information available from the temporal aggregation of both biotic and abiotic metrics to analyze and quantify the combined effects on long-term performance. Study of a 42-day implant showed there was a functional relationship between the measured impedance and the array neuronal yield. This was correlated with structural changes in the recording sites, microglial activation/degeneration, and elevation of a blood biochemical marker for axonal injury. We seek to elucidate the mechanisms of chronic microelectrode array failure through the study of the combined effects of these biotic and abiotic factors.


Subject(s)
Neural Prostheses , Tungsten , Animals , Axons/physiology , Biomarkers/blood , Electric Impedance , Electrophysiological Phenomena , Microelectrodes , Microscopy, Electron, Scanning , Neurofilament Proteins/blood , Prosthesis Failure , Rats , Somatosensory Cortex/injuries , Somatosensory Cortex/physiopathology , Somatosensory Cortex/surgery , Time Factors
19.
J Neurosci Methods ; 201(2): 296-306, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21871490

ABSTRACT

Traumatic brain injury (TBI) is a major cause of death and disability for which there is no cure. One of the issues inhibiting clinical trial success is the lack of targeting specific patient populations due to inconsistencies between clinical diagnostic tools and underlying pathophysiology. The development of reliable, noninvasive markers of TBI severity and injury mechanisms may better identify these populations, thereby improving clinical trial design. Magnetic resonance elastography (MRE), by assessing tissue mechanical properties, can potentially provide such marker. MRE synchronizes mechanical excitations with a phase contrast imaging pulse sequence to noninvasively register shear wave propagation, from which local values of tissue viscoelastic properties can be deduced. The working hypothesis of this study is that TBI involves a compression of brain tissue large enough to bring the material out of its elastic range, sufficiently altering mechanical properties to generate contrast on MRE measurements. To test this hypothesis, we combined microscopic MRE with brain tissue collected from adult male rats subjected to a controlled cortical impact injury. Measurements were made in different regions of interest (somatosensory cortex, hippocampus, and thalamus), and at different time points following the injury (immediate, 24 h, 7 days, 28 days). Values of stiffness in the somatosensory cortex were found to be 23-32% lower in the injured hemisphere than in the healthy one, when no significant difference was observed in the case of sham brains. A preliminary in vivo experiment is also presented, as well as alternatives to improve the faithfulness of stiffness recovery.


Subject(s)
Brain Injuries/physiopathology , Elasticity Imaging Techniques/methods , Miniaturization/methods , Animals , Brain Injuries/pathology , Disease Models, Animal , Elasticity Imaging Techniques/instrumentation , Hippocampus/injuries , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/injuries , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Thalamus/injuries , Thalamus/pathology , Thalamus/physiopathology
20.
J Neurotrauma ; 28(9): 1909-19, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21787180

ABSTRACT

Traumatic brain injury (TBI) induces transient or persistent dysfunction of gait and balance. Enhancement of cholinergic transmission has been reported to accelerate recovery of cognitive function after TBI, but the effects of this intervention on locomotor activity remain largely unexplored. The hypothesis that enhancement of cholinergic function by inhibition of acetylcholinesterase (AChE) improves locomotion following TBI was tested in Sprague-Dawley male rats after a unilateral controlled cortical impact (CCI) injury of the motor-sensory cortex. Locomotion was tested by time to fall on the constant speed and accelerating Rotarod, placement errors and time to cross while walking through a horizontal ladder, activity monitoring in the home cages, and rearing behavior. Assessments were performed the 1st and 2nd day and the 1st, 2nd, and 3rd week after TBI. The AChE inhibitor physostigmine hemisulfate (PHY) was administered continuously via osmotic minipumps implanted subcutaneously at the rates of 1.6-12.8 µmol/kg/day. All measures of locomotion were impaired by TBI and recovered to initial levels between 1 and 3 weeks post-TBI, with the exception of the maximum speed achievable on the accelerating Rotarod, as well as rearing in the open field. PHY improved performance in the accelerating Rotarod at 1.6 and 3.2 µmol/kg/day (AChE activity 95 and 78% of control, respectively), however, higher doses induced progressive deterioration. No effect or worsening of outcomes was observed at all PHY doses for home cage activity, rearing, and horizontal ladder walking. Potential benefits of cholinesterase inhibition on locomotor function have to be weighed against the evidence of the narrow range of useful doses.


Subject(s)
Brain Injuries/physiopathology , Cholinesterase Inhibitors/pharmacology , Motor Activity/drug effects , Motor Cortex/injuries , Physostigmine/pharmacology , Somatosensory Cortex/injuries , Animals , Locomotion/drug effects , Locomotion/physiology , Male , Motor Activity/physiology , Motor Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Rotarod Performance Test , Somatosensory Cortex/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...