Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cureus ; 12(1): e6748, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-32133270

ABSTRACT

Background Neuroanatomists have long been fascinated by the complex topographic organization of the cerebrum. We examined historical and modern phylogenetic theories pertaining to microneurosurgical anatomy and intrinsic brain tumor development. Methods Literature and history related to the study of anatomy, evolution, and tumor predilection of the limbic and paralimbic regions were reviewed. We used vertebrate histological cross-sections, photographs from Albert Rhoton Jr.'s dissections, and original drawings to demonstrate the utility of evolutionary temporal causality in understanding anatomy. Results Phylogenetic neuroanatomy progressed from the substantial works of Alcmaeon, Herophilus, Galen, Vesalius, von Baer, Darwin, Felsenstein, Klingler, MacLean, and many others. We identified two major modern evolutionary theories: "triune brain" and topological phylogenetics. While the concept of "triune brain" is speculative and highly debated, it remains the most popular in the current neurosurgical literature. Phylogenetics inspired by mathematical topology utilizes computational, statistical, and embryological data to analyze the temporal transformations leading to three-dimensional topographic anatomy. These transformations have shaped well-defined surgical planes, which can be exploited by the neurosurgeon to access deep cerebral targets. The microsurgical anatomy of the cerebrum and the limbic system is redescribed by incorporating the dimension of temporal causality. Yasargil's anatomical classification of glial tumors can be revisited in light of modern phylogenetic cortical categorization. Conclusion Historical and modern topological phylogenetic notions provide a deeper understanding of neurosurgical anatomy and approaches to the limbic and paralimbic regions. However, many questions remain unanswered and further research is needed to elucidate the anatomical pathology of intrinsic brain tumors.

3.
Exp Eye Res ; 182: 109-124, 2019 05.
Article in English | MEDLINE | ID: mdl-30922891

ABSTRACT

Mild TBI is often accompanied by visual system dysfunction and injury, which is at least partly caused by microglial neuroinflammatory processes initiated by the injury. Using our focal cranial blast mouse model of closed-skull mild TBI, we evaluated the ability of the cannabinoid type-2 (CB2) receptor inverse agonist SMM-189, which biases microglia from the harmful M1 state to the beneficial M2 state, to mitigate visual system dysfunction and injury after TBI. Male C57BL/6 or Thy1-EYFP reporter mice received a closed-head blast of either 0-psi (sham) or 50-psi to the left side of the cranium. Blast mice received vehicle or 6 mg/kg SMM-189 daily beginning 2 h after blast. Sham mice received vehicle. In some mice, retina and optic nerve/tract were assessed morphologically at 3-7 days after blast, while other mice were assessed functionally by Optomotry 30 days after blast and morphologically at ≥30 days after blast. Mice sacrificed at 3-7 days were treated daily until sacrificed, while those assessed ≥30 days after blast were treated daily for 2 weeks post blast. Axon damage was evident in the left optic nerve and its continuation as the right optic tract at 3 days post blast in vehicle-treated blast mice in the form of swollen axon bulbs, and was accompanied by a significant increase in the abundance of microglia. Testing at 30 days post blast revealed that the contrast sensitivity function was significantly reduced in both eyes in vehicle-treated blast mice compared to vehicle-treated sham blast mice, and axon counts at ≥30 days after blast revealed a ∼10% loss in left optic nerve in vehicle-treated blast mice. Left optic nerve axon loss was highly correlated with the left eye deficit in contrast sensitivity. Immunolabeling at 30 days post blast showed a significant increase in the abundance of microglia in the retinas of both eyes and in GFAP + Müller cell processes traversing the inner plexiform layer in the left eye of vehicle-treated blast mice. SMM-189 treatment reduced axon injury and microglial abundance at 3 days, and mitigated axon loss, contrast sensitivity deficits, microglial abundance, and Müller cell GFAP upregulation at ≥30 days after blast injury. Analysis of right optic tract microglia at 3 days post blast for M1 versus M2 markers revealed that SMM-189 biased microglia toward the M2 state, with this action of SMM-189 being linked to reduced axonal injury. Taken together, our results show that focal left side cranial blast resulted in impaired contrast sensitivity and retinal pathology bilaterally and optic nerve loss ipsilaterally. The novel cannabinoid drug SMM-189 significantly mitigated the functional deficit and the associated pathologies. Our findings suggest the value of combatting visual system injury after TBI by using CB2 inverse agonists such as SMM-189, which appear to target microglia and bias them away from the pro-inflammatory M1 state, toward the protective M2 state.


Subject(s)
Benzophenones/pharmacology , Brain Injuries, Traumatic/complications , Microglia/pathology , Optic Nerve/pathology , Optic Tract/pathology , Vision Disorders/drug therapy , Visual Acuity , Animals , Axons/pathology , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Vision Disorders/etiology , Vision Disorders/pathology
4.
eNeuro ; 4(4)2017.
Article in English | MEDLINE | ID: mdl-28828401

ABSTRACT

Mild traumatic brain injury (mTBI) can cause severe long-term cognitive and emotional deficits, including impaired memory, depression, and persevering fear, but the neuropathological basis of these deficits is uncertain. As medial prefrontal cortex (mPFC) and hippocampus play important roles in memory and emotion, we used multi-site, multi-electrode recordings of oscillatory neuronal activity in local field potentials (LFPs) in awake, head-fixed mice to determine if the functioning of these regions was abnormal after mTBI, using a closed-skull focal cranial blast model. We evaluated mPFC, hippocampus CA1, and primary somatosensory/visual cortical areas (S1/V1). Although mTBI did not alter the power of oscillations, it did cause increased coherence of θ (4-10 Hz) and ß (10-30 Hz) oscillations within mPFC and S1/V1, reduced CA1 sharp-wave ripple (SWR)-evoked LFP activity in mPFC, downshifted SWR frequencies in CA1, and enhanced θ-γ phase-amplitude coupling (PAC) within mPFC. These abnormalities might be linked to the impaired memory, depression, and persevering fear seen after mTBI. Treatment with the cannabinoid type-2 (CB2) receptor inverse agonist SMM-189 has been shown to mitigate functional deficits and neuronal injury after mTBI in mice. We found that SMM-189 also reversed most of the observed neurophysiological abnormalities. This neurophysiological rescue is likely to stem from the previously reported reduction in neuron loss and/or the preservation of neuronal function and connectivity resulting from SMM-189 treatment, which appears to stem from the biasing of microglia from the proinflammatory M1 state to the prohealing M2 state by SMM-189.


Subject(s)
Benzophenones/therapeutic use , Brain Concussion/drug therapy , Brain Concussion/pathology , Brain/drug effects , Cannabinoid Receptor Agonists/therapeutic use , Action Potentials/drug effects , Animals , Brain/pathology , Brain Mapping , Brain Waves/drug effects , Disease Models, Animal , Electroencephalography , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Principal Component Analysis , Receptor, Cannabinoid, CB2/metabolism , Time Factors
5.
Front Neurosci ; 10: 449, 2016.
Article in English | MEDLINE | ID: mdl-27766068

ABSTRACT

We have previously reported that mild TBI created by focal left-side cranial blast in mice produces widespread axonal injury, microglial activation, and a variety of functional deficits. We have also shown that these functional deficits are reduced by targeting microglia through their cannabinoid type-2 (CB2) receptors using 2-week daily administration of the CB2 inverse agonist SMM-189. CB2 inverse agonists stabilize the G-protein coupled CB2 receptor in an inactive conformation, leading to increased phosphorylation and nuclear translocation of the cAMP response element binding protein (CREB), and thus bias activated microglia from a pro-inflammatory M1 to a pro-healing M2 state. In the present study, we showed that SMM-189 boosts nuclear pCREB levels in microglia in several brain regions by 3 days after TBI, by using pCREB/CD68 double immunofluorescent labeling. Next, to better understand the basis of motor deficits and increased fearfulness after TBI, we used unbiased stereological methods to characterize neuronal loss in cortex, striatum, and basolateral amygdala (BLA) and assessed how neuronal loss was affected by SMM-189 treatment. Our stereological neuron counts revealed a 20% reduction in cortical and 30% reduction in striatal neurons bilaterally at 2-3 months post blast, with SMM-189 yielding about 50% rescue. Loss of BLA neurons was restricted to the blast side, with 33% of Thy1+ fear-suppressing pyramidal neurons and 47% of fear-suppressing parvalbuminergic (PARV) interneurons lost, and Thy1-negative fear-promoting pyramidal neurons not significantly affected. SMM-189 yielded 50-60% rescue of Thy1+ and PARV neuron loss in BLA. Thus, fearfulness after mild TBI may result from the loss of fear-suppressing neuron types in BLA, and SMM-189 may reduce fearfulness by their rescue. Overall, our findings indicate that SMM-189 rescues damaged neurons and thereby alleviates functional deficits resulting from TBI, apparently by selectively modulating microglia to the beneficial M2 state. CB2 inverse agonists thus represent a promising therapeutic approach for mitigating neuroinflammation and neurodegeneration.

6.
J Neurotrauma ; 33(4): 403-22, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26414413

ABSTRACT

Mild traumatic brain injury (TBI) from focal head impact is the most common form of TBI in humans. Animal models, however, typically use direct impact to the exposed dura or skull, or blast to the entire head. We present a detailed characterization of a novel overpressure blast system to create focal closed-head mild TBI in mice. A high-pressure air pulse limited to a 7.5 mm diameter area on the left side of the head overlying the forebrain is delivered to anesthetized mice. The mouse eyes and ears are shielded, and its head and body are cushioned to minimize movement. This approach creates mild TBI by a pressure wave that acts on the brain, with minimal accompanying head acceleration-deceleration. A single 20-psi blast yields no functional deficits or brain injury, while a single 25-40 psi blast yields only slight motor deficits and brain damage. By contrast, a single 50-60 psi blast produces significant visual, motor, and neuropsychiatric impairments and axonal damage and microglial activation in major fiber tracts, but no contusive brain injury. This model thus reproduces the widespread axonal injury and functional impairments characteristic of closed-head mild TBI, without the complications of systemic or ocular blast effects or head acceleration that typically occur in other blast or impact models of closed-skull mild TBI. Accordingly, our model provides a simple way to examine the biomechanics, pathophysiology, and functional deficits that result from TBI and can serve as a reliable platform for testing therapies that reduce brain pathology and deficits.


Subject(s)
Air Pressure , Brain Concussion/pathology , Brain Concussion/physiopathology , Disease Models, Animal , Explosions , Skull/injuries , Animals , Brain Concussion/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
Exp Neurol ; 271: 53-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25957630

ABSTRACT

Diffuse axonal injury is thought to be the basis of the functional impairments stemming from mild traumatic brain injury. To examine how axons are damaged by traumatic events, such as motor vehicle accidents, falls, sports activities, or explosive blasts, we have taken advantage of the spinal cord with its extensive white matter tracts. We developed a closed-body model of spinal cord injury in mice whereby high-pressure air blasts targeted to lower thoracic vertebral levels produce tensile, compressive, and shear forces within the parenchyma of the spinal cord and thereby cause extensive axonal injury. Markers of cytoskeletal integrity showed that spinal cord axons exhibited three distinct pathologies: microtubule breakage, neurofilament compaction, and calpain-mediated spectrin breakdown. The dorsally situated axons of the corticospinal tract primarily exhibited microtubule breakage, whereas all three pathologies were common in the lateral and ventral white matter. Individual axons typically demonstrated only one of the three pathologies during the first 24h after blast injury, suggesting that the different perturbations are initiated independently of one another. For the first few days after blast, neurofilament compaction was frequently accompanied by autophagy, and subsequent to that, by the fragmentation of degenerating axons. TuJ1 immunolabeling and mice with YFP-reporter labeling each revealed more extensive microtubule breakage than did ßAPP immunolabeling, raising doubts about the sensitivity of this standard approach for assessing axonal injury. Although motor deficits were mild and largely transient, some aspects of motor function gradually worsened over several weeks, suggesting that a low level of axonal degeneration continued past the initial wave. Our model can help provide further insight into how to intervene in the processes by which initial axonal damage culminates in axonal degeneration, to improve outcomes after traumatic injury. Importantly, our findings of extensive axonal injury also caution that repeated trauma is likely to have cumulative adverse consequences for both brain and spinal cord.


Subject(s)
Blast Injuries/complications , Diffuse Axonal Injury/etiology , Psychomotor Disorders/etiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/etiology , Amyloid beta-Protein Precursor/metabolism , Animals , Autophagy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Female , Gait/physiology , Gene Expression Regulation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neurofilament Proteins/metabolism , Protein Kinase C/metabolism , Time Factors , Tubulin/metabolism
8.
Front Neurol ; 5: 2, 2014.
Article in English | MEDLINE | ID: mdl-24478749

ABSTRACT

Emotional disorders are a common outcome from mild traumatic brain injury (TBI) in humans, but their pathophysiological basis is poorly understood. We have developed a mouse model of closed-head blast injury using an air pressure wave delivered to a small area on one side of the cranium, to create mild TBI. We found that 20-psi blasts in 3-month-old C57BL/6 male mice yielded no obvious behavioral or histological evidence of brain injury, while 25-40 psi blasts produced transient anxiety in an open field arena but little histological evidence of brain damage. By contrast, 50-60 psi blasts resulted in anxiety-like behavior in an open field arena that became more evident with time after blast. In additional behavioral tests conducted 2-8 weeks after blast, 50-60 psi mice also demonstrated increased acoustic startle, perseverance of learned fear, and enhanced contextual fear, as well as depression-like behavior and diminished prepulse inhibition. We found no evident cerebral pathology, but did observe scattered axonal degeneration in brain sections from 50 to 60 psi mice 3-8 weeks after blast. Thus, the TBI caused by single 50-60 psi blasts in mice exhibits the minimal neuronal loss coupled to "diffuse" axonal injury characteristic of human mild TBI. A reduction in the abundance of a subpopulation of excitatory projection neurons in basolateral amygdala enriched in Thy1 was, however, observed. The reported link of this neuronal population to fear suppression suggests their damage by mild TBI may contribute to the heightened anxiety and fearfulness observed after blast in our mice. Our overpressure air blast model of concussion in mice will enable further studies of the mechanisms underlying the diverse emotional deficits seen after mild TBI.

9.
Int J Mol Sci ; 16(1): 758-87, 2014 Dec 31.
Article in English | MEDLINE | ID: mdl-25561230

ABSTRACT

We have developed a focal blast model of closed-head mild traumatic brain injury (TBI) in mice. As true for individuals that have experienced mild TBI, mice subjected to 50-60 psi blast show motor, visual and emotional deficits, diffuse axonal injury and microglial activation, but no overt neuron loss. Because microglial activation can worsen brain damage after a concussive event and because microglia can be modulated by their cannabinoid type 2 receptors (CB2), we evaluated the effectiveness of the novel CB2 receptor inverse agonist SMM-189 in altering microglial activation and mitigating deficits after mild TBI. In vitro analysis indicated that SMM-189 converted human microglia from the pro-inflammatory M1 phenotype to the pro-healing M2 phenotype. Studies in mice showed that daily administration of SMM-189 for two weeks beginning shortly after blast greatly reduced the motor, visual, and emotional deficits otherwise evident after 50-60 psi blasts, and prevented brain injury that may contribute to these deficits. Our results suggest that treatment with the CB2 inverse agonist SMM-189 after a mild TBI event can reduce its adverse consequences by beneficially modulating microglial activation. These findings recommend further evaluation of CB2 inverse agonists as a novel therapeutic approach for treating mild TBI.


Subject(s)
Benzophenones/pharmacology , Brain Injuries/drug therapy , Motor Activity/drug effects , Receptor, Cannabinoid, CB2/agonists , Animals , Brain Injuries/complications , Brain Injuries/pathology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Depression/etiology , Depression/pathology , Disease Models, Animal , Drug Inverse Agonism , Humans , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Phenotype , Receptor, Cannabinoid, CB2/metabolism , Vision Disorders/etiology , Vision Disorders/pathology
10.
Mov Disord ; 28(12): 1691-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24014043

ABSTRACT

Although dystonia represents a major source of motor disability in Huntington's disease (HD), its pathophysiology remains unknown. Because recent animal studies indicate that loss of parvalbuminergic (PARV+) striatal interneurons can cause dystonia, we investigated if loss of PARV+ striatal interneurons occurs during human HD progression, and thus might contribute to dystonia in HD. We used immunolabeling to detect PARV+ interneurons in fixed sections, and corrected for disease-related striatal atrophy by expressing PARV+ interneuron counts in ratio to interneurons co-containing somatostatin and neuropeptide Y (whose numbers are unaffected in HD). At all symptomatic HD grades, PARV+ interneurons were reduced to less than 26% of normal abundance in rostral caudate. In putamen rostral to the level of globus pallidus, loss of PARV+ interneurons was more gradual, not dropping off to less than 20% of control until grade 2. Loss of PARV+ interneurons was even more gradual in motor putamen at globus pallidus levels, with no loss at grade 1, and steady grade-wise decline thereafter. A large decrease in striatal PARV+ interneurons, thus, occurs in HD with advancing disease grade, with regional variation in the loss per grade. Given the findings of animal studies and the grade-wise loss of PARV+ striatal interneurons in motor striatum in parallel with the grade-wise appearance and worsening of dystonia, our results raise the possibility that loss of PARV+ striatal interneurons is a contributor to dystonia in HD.


Subject(s)
Corpus Striatum/pathology , Dystonia/pathology , Huntington Disease/pathology , Nerve Degeneration/pathology , Neurons/pathology , Parvalbumins/metabolism , Adult , Aged , Aged, 80 and over , Corpus Striatum/metabolism , Dystonia/metabolism , Female , Humans , Huntington Disease/metabolism , Male , Middle Aged , Nerve Degeneration/metabolism , Neurons/metabolism
11.
J Comp Neurol ; 521(6): 1354-77, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23047588

ABSTRACT

We examined thalamic input to striatum in rats using immunolabeling for the vesicular glutamate transporter (VGLUT2). Double immunofluorescence viewed with confocal laser scanning microscopy (CLSM) revealed that VGLUT2+ terminals are distinct from VGLUT1+ terminals. CLSM of Phaseolus vulgaris-leucoagglutinin (PHAL)-labeled cortical or thalamic terminals revealed that VGLUT2 is rare in corticostriatal terminals but nearly always present in thalamostriatal terminals. Electron microscopy revealed that VGLUT2+ terminals made up 39.4% of excitatory terminals in striatum (with VGLUT1+ corticostriatal terminals constituting the rest), and 66.8% of VGLUT2+ terminals synapsed on spines and the remainder on dendrites. VGLUT2+ axospinous terminals had a mean diameter of 0.624 µm, while VGLUT2+ axodendritic terminals a mean diameter of 0.698 µm. In tissue in which we simultaneously immunolabeled thalamostriatal terminals for VGLUT2 and striatal neurons for D1 (with about half of spines immunolabeled for D1), 54.6% of VGLUT2+ terminals targeted D1+ spines (i.e., direct pathway striatal neurons), and 37.3% of D1+ spines received VGLUT2+ synaptic contacts. By contrast, 45.4% of VGLUT2+ terminals targeted D1-negative spines (i.e., indirect pathway striatal neurons), and only 25.8% of D1-negative spines received VGLUT2+ synaptic contacts. Similarly, among VGLUT2+ axodendritic synaptic terminals, 59.1% contacted D1+ dendrites, and 40.9% contacted D1-negative dendrites. VGLUT2+ terminals on D1+ spines and dendrites tended to be slightly smaller than those on D1-negative spines and dendrites. Thus, thalamostriatal terminals contact both direct and indirect pathway striatal neurons, with a slight preference for direct. These results are consistent with physiological studies indicating slightly different effects of thalamic input on the two types of striatal projection neurons.


Subject(s)
Corpus Striatum/ultrastructure , Neurons/ultrastructure , Thalamus/ultrastructure , Vesicular Glutamate Transport Protein 2/ultrastructure , Animals , Corpus Striatum/chemistry , Corpus Striatum/cytology , Excitatory Postsynaptic Potentials/physiology , Male , Microscopy, Confocal/methods , Neural Pathways/chemistry , Neural Pathways/cytology , Neural Pathways/ultrastructure , Neurons/chemistry , Rats , Rats, Sprague-Dawley , Thalamus/chemistry , Thalamus/cytology , Vesicular Glutamate Transport Protein 2/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...