Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Adv ; 6(32): eaaz6892, 2020 08.
Article in English | MEDLINE | ID: mdl-32821816

ABSTRACT

Neuroimaging biomarkers that can detect white matter (WM) pathology after mild traumatic brain injury (mTBI) and predict long-term outcome are needed to improve care and develop therapies. We used diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) to investigate WM microstructure cross-sectionally and longitudinally after mTBI and correlate these with neuropsychological performance. Cross-sectionally, early decreases of fractional anisotropy and increases of mean diffusivity corresponded to WM regions with elevated free water fraction on NODDI. This elevated free water was more extensive in the patient subgroup reporting more early postconcussive symptoms. The longer-term longitudinal WM changes consisted of declining neurite density on NODDI, suggesting axonal degeneration from diffuse axonal injury for which NODDI is more sensitive than DTI. Therefore, NODDI is a more sensitive and specific biomarker than DTI for WM microstructural changes due to mTBI that merits further study for mTBI diagnosis, prognosis, and treatment monitoring.

2.
J Neurosurg Sci ; 58(4): 249-59, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25418276

ABSTRACT

Traumatic brain injury (TBI) is a significant contributing factor to injury-related deaths worldwide. Despite the medical and fiscal importance of this subject, guidelines for the surgical management of closed and penetrating TBI are largely based on Level III evidence. Furthermore, the results of a large, costly trial designed to further elucidate the role of decompressive craniectomy, an important surgical intervention in patients with severe TBI and medically-refractory intracranial hypertension, have limited applicability. In this review, we summarize the existing guidelines for the surgical management of TBI, present an overview of the underlying pathophysiologic principles and neurophysiologic consequences relating to decompressive hemicraniectomy, highlight the history, relevant studies, and outcomes pertaining to decompressive craniectomy for patients with severe TBI, and discuss some of the current controversies in the surgical management of traumatic brain injury. Despite the varied outcomes seen in the literature, DC is indeed an important intervention in the management of TBI, as it is highly effective at reducing ICP and thus, an important higher-treatment strategy for patients with medically-refractory intracranial hypertension. There will continue to be unresolved controversies regarding decisions pertaining to defining an "optimal" surgical candidate, specific timing, techniques, and post-operative management of TBI patients who undergo surgery. New guidelines for the surgical management of TBI are forthcoming. Regardless, for neurosurgeons involved in the care of individuals with TBI, understanding the pathophysiologic and neurophysiologic consequences of surgical interventions, and gaining an understanding of the extant literature is imperative.


Subject(s)
Brain Injuries/surgery , Decompressive Craniectomy/methods , Brain Injuries/physiopathology , Humans , Practice Guidelines as Topic
3.
Libyan J Med ; 4(3): 97-103, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-21483522

ABSTRACT

Traumatic injury/hemorrhagic shock (T/HS) elicits an acute inflammatory response that may result in death. Inflammation describes a coordinated series of molecular, cellular, tissue, organ, and systemic responses that drive the pathology of various diseases including T/HS and traumatic brain injury (TBI). Inflammation is a finely tuned, dynamic, highly-regulated process that is not inherently detrimental, but rather required for immune surveillance, optimal post-injury tissue repair, and regeneration. The inflammatory response is driven by cytokines and chemokines and is partially propagated by damaged tissue-derived products (Damage-associated Molecular Patterns; DAMP's). DAMPs perpetuate inflammation through the release of pro-inflammatory cytokines, but may also inhibit anti-inflammatory cytokines. Various animal models of T/HS in mice, rats, pigs, dogs, and non-human primates have been utilized in an attempt to move from bench to bedside. Novel approaches, including those from the field of systems biology, may yield therapeutic breakthroughs in T/HS and TBI in the near future.

4.
Libyan j. med ; 4(3): 97-103, 2009. figures
Article in English | AIM (Africa) | ID: biblio-1265095

ABSTRACT

Traumatic injury/hemorrhagic shock (T/HS) elicits an acute inflammatory response that may result in death. Inflammation describes a coordinated series of molecular; cellular; tissue; organ; and systemic responses that drive the pathology of various diseases including T/HS and traumatic brain injury (TBI). Inflammation is a finely tuned; dynamic; highly-regulated process that is not inherently detrimental; but rather required for immune surveillance; optimal post-injury tissue repair; and regeneration. The inflammatory response is driven by cytokines and chemokines and is partially propagated by damaged tissue-derived products (Damage-associated Molecular Patterns; DAMP's). DAMPs perpetuate inflammation through the release of pro-inflammatory cytokines; but may also inhibit anti-inflammatory cytokines. Various animal models of T/HS in mice; rats; pigs; dogs; and non-human primates have been utilized in an attempt to move from bench to bedside. Novel approaches; including those from the field of systems biology; may yield therapeutic breakthroughs in T/HS and TBI in the near future. Key words: Trauma; Hemorrhagic Shock; Taumatic Brain Injury; Inflammation; Systems Biology


Subject(s)
Humans , Shock, Hemorrhagic , Brain Hemorrhage, Traumatic , Encephalitis , Shock , Systems Biology , Wounds and Injuries
5.
Acta Neurochir (Wien) ; 149(9): 969-72; discussion 972, 2007.
Article in English | MEDLINE | ID: mdl-17558459

ABSTRACT

Hemangioblastomas are rarely found in a supratentorial location and are commonly associated with the von Hippel-Lindau complex. Therefore, patients with such tumors must be evaluated for both other hemangioblastomas within the central nervous system as well as for this complex via physical examination, radiographic examination, and genetic testing. We report the seventh case of a patient with an isolated supratentorial dural based hemangioblastoma not associated with the von Hippel-Lindau complex.


Subject(s)
Dura Mater , Hemangioblastoma/diagnosis , Magnetic Resonance Imaging , Meningeal Neoplasms/diagnosis , Female , Hemangioblastoma/pathology , Hemangioblastoma/surgery , Humans , Meningeal Neoplasms/pathology , Meningeal Neoplasms/surgery , Middle Aged , Neurosurgical Procedures , von Hippel-Lindau Disease
6.
Clin Neuropharmacol ; 24(5): 254-64, 2001.
Article in English | MEDLINE | ID: mdl-11586110

ABSTRACT

Spinal cord injury (SCI) is a devastating and common neurologic disorder that has profound influences on modern society from physical, psychosocial, and socioeconomic perspectives. Accordingly, the present decade has been labeled the Decade of the Spine to emphasize the importance of SCI and other spinal disorders. Spinal cord injury may be divided into both primary and secondary mechanisms of injury. The primary injury, in large part, determines a given patient's neurologic grade on admission and thereby is the strongest prognostic indicator. However, secondary mechanisms of injury can exacerbate damage and limit restorative processes, and hence, contribute to overall morbidity and mortality. A burgeoning body of evidence has facilitated our understanding of these secondary mechanisms of injury that are amenable to pharmacological interventions, unlike the primary injury itself. Secondary mechanisms of injury encompass an array of perturbances and include neurogenic shock, vascular insults such as hemorrhage and ischemia-reperfusion, excitotoxicity, calcium-mediated secondary injury and fluid-electrolyte disturbances, immunologic injury, apoptosis, disturbances in mitochondrion function, and other miscellaneous processes. Comprehension of secondary mechanisms of injury serves as a basis for the development and application of targeted pharmacological strategies to confer neuroprotection and restoration while mitigating ongoing neural injury. The first article in this series will comprehensively review the pathophysiology of SCI while emphasizing those mechanisms for which pharmacologic therapy has been developed, and the second article reviews the pharmacologic interventions for SCI.


Subject(s)
Spinal Cord Injuries/physiopathology , Acute Disease , Animals , Apoptosis , Cell Death , Hemorrhage/pathology , Hemorrhage/physiopathology , Humans , Ischemia/pathology , Ischemia/physiopathology , Spinal Cord Injuries/pathology
7.
Clin Neuropharmacol ; 24(5): 265-79, 2001.
Article in English | MEDLINE | ID: mdl-11586111

ABSTRACT

Spinal cord injury (SCI) remains a common and devastating problem of modern society. Through an understanding of underlying pathophysiologic mechanisms involved in the evolution of SCI, treatments aimed at ameliorating neural damage may be developed. The possible pharmacologic treatments for acute spinal cord injury are herein reviewed. Myriad treatment modalities, including corticosteroids, 21-aminosteroids, opioid receptor antagonists, gangliosides, thyrotropin-releasing hormone (TRH) and TRH analogs, antioxidants and free radical scavengers, calcium channel blockers, magnesium replacement therapy, sodium channel blockers, N -methyl-D-aspartate receptor antagonists, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid-kainate receptor antagonists, modulators of arachadonic acid metabolism, neurotrophic growth factors, serotonin antagonists, antibodies against inhibitors of axonal regeneration, potassium channel blockers (4-aminopyridine), paclitaxel, clenbuterol, progesterone, gabexate mesylate, activated protein C, caspase inhibitors, tacrolimus, antibodies against adhesion molecules, and other immunomodulatory therapy have been studied to date. Although most of these agents have shown promise, only one agent, methylprednisolone, has been shown to provide benefit in large clinical trials. Given these data, many individuals consider methylprednisolone to be the standard of care for the treatment of acute SCI. However, this has not been established definitively, and questions pertaining to methodology have emerged regarding the National Acute Spinal Cord Injury Study trials that provided these conclusions. Additionally, the clinical significance (in contrast to statistical significance) of recovery after methylprednisolone treatment is unclear and must be considered in light of the potential adverse effects of such treatment. This first decade of the new millennium, now touted as the Decade of the Spine, will hopefully witness the emergence of universal and efficacious pharmacologic therapy and ultimately a cure for SCI.


Subject(s)
Drug Therapy/methods , Spinal Cord Injuries/drug therapy , Acute Disease , Animals , Clinical Trials as Topic/methods , Clinical Trials as Topic/statistics & numerical data , Humans
8.
J Neurotrauma ; 18(6): 607-14, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11437083

ABSTRACT

The immunophilin ligand, cyclosporin A (CsA), is effective in reducing the axonal damage associated with traumatic brain injury (TBI). Based upon extensive ultrastructural and immunohistochemical studies, the neuroprotection afforded by CsA appeared to be mediated via mitochondrial protection, specifically, the prevention of mitochondrial swelling and inhibition of mitochondrial permeability transition (MPT). However, the potential that CsA could also be neuroprotective via the immunophilin-mediated inhibition of the protein phosphatase, calcineurin (CN) has not been directly assessed. To address this issue, the current study assessed the ability of FK506, another immunophilin ligand that inhibits CN with no effect on MPT, to attenuate axonal damage in a rat impact-acceleration model of TBI. Traumatic axonal injury (TAI), detected via an antibody against beta-amyloid precursor protein (APP), a specific marker of axonal injury, was significantly reduced at 24 hr postinjury in Sprague-Dawley rats receiving intravenous FK506 (2 mg/kg; n = 5) 30 min prior to injury compared to vehicle controls (n = 3). While not rejecting the established efficacy of CsA in providing neuroprotection via its targeting of MPT, this study does underscore the potential importance of CN in the progressive pathobiology of TAI, suggesting that CN may constitute another important therapeutic target.


Subject(s)
Axons/pathology , Brain Injuries/drug therapy , Immunosuppressive Agents/therapeutic use , Tacrolimus/therapeutic use , Acceleration , Animals , Blood Gas Analysis , Brain/metabolism , Brain Injuries/pathology , Image Processing, Computer-Assisted , Immunohistochemistry , Immunosuppressive Agents/pharmacokinetics , Injections, Intravenous , Male , Rats , Rats, Sprague-Dawley , Tacrolimus/pharmacokinetics
9.
J Neurosci ; 20(8): 2825-34, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10751434

ABSTRACT

Axonal injury is a feature of traumatic brain injury (TBI) contributing to both morbidity and mortality. The traumatic axon injury (TAI) results from focal perturbations of the axolemma, allowing for calcium influx triggering local intraaxonal cytoskeletal and mitochondrial damage. This mitochondrial damage has been posited to cause local bioenergetic failure, leading to axonal failure and disconnection; however, this mitochondrial damage may also lead to the release of cytochrome c (cyto-c), which then activates caspases with significant adverse intraaxonal consequences. In the current communication, we examine this possibility. Rats were subjected to TBI, perfused with aldehydes at 15-360 min after injury, and processed for light microscopic (LM) and electron microscopic (EM) single-labeling immunohistochemistry to detect extramitochondrially localized cytochrome c (cyto-c) and the signature protein of caspase-3 activation (120 kDa breakdown product of alpha-spectrin) in TAI. Combinations of double-labeling fluorescent immunohistochemistry (D-FIHC) were also used to demonstrate colocalization of calpain activation with cyto-c release and caspase-3-induction. In foci of TAI qualitative-quantitative LM demonstrated a parallel, significant increase in cyto-c release and caspase-3 activation over time after injury. EM analysis demonstrated that cyto-c and caspase-3 immunoreactivity were associated with mitochondrial swelling-disruption in sites of TAI. Furthermore, D-IFHC revealed a colocalization of calpain activation, cyto-c release, and caspase-3 induction in these foci, which also revealed progressive TAI. The results demonstrate that cyto-c and caspase-3 participate in the terminal processes of TAI. This suggests that those factors that play a role in the apoptosis in the neuronal soma are also major contributors to the demise of the axonal appendage.


Subject(s)
Brain Injuries/enzymology , Calpain/metabolism , Caspases/metabolism , Cytochrome c Group/metabolism , Diffuse Axonal Injury/enzymology , Animals , Brain Injuries/pathology , Caspase 3 , Diffuse Axonal Injury/pathology , Enzyme Activation , Rats , Rats, Sprague-Dawley
10.
Acta Neurochir Suppl ; 73: 15-20, 1999.
Article in English | MEDLINE | ID: mdl-10494336

ABSTRACT

To gain better insight into the initiating factors involved in traumatically induced axonal injury cats and rats were subjected to various forms of traumatic brain injury. Following injury at intervals ranging from 10 min. to 3 hours, the animals were sacrificed and prepared in accordance with multiple immunocytochemical strategies capable of detecting focal changes in the axolemma, the subaxolemmal spectrin network, the underlying cytoskeleton as well as any related abnormalities in axoplasmic transport. Through these approaches it was recognized that the most severe forms of injury resulted in focal abnormalities of axonal permeability which were observed together with calpain-mediated spectrin proteolysis in the subaxolemmal network. These events were associated with compaction of the underlying neurofilaments and some microtubular loss which occurred without any direct evidence of overt axoplasmic proteolysis with the exception of the most severely injured fibers. In addition to these severely injured axonal profiles, other injured axons did not manifest overt changes in axolemmal permeability or early calpain-mediated spectrin proteolysis but demonstrated dramatic neurofilament and microtubular misalignment and impaired axoplasmic transport. Lastly, other small caliber axons showed another form of intraaxonal change manifested in the local pooling of organelles in the nodal and paranodal regions, with the suggestion that some of these changes may be reversible. In relation to these axonal responses the efficacy of various therapeutic investigations were assessed. The use of calcium chelators showed a trend for protection in those axons manifesting altered axolemmal permeability. However, the use of early and delayed hypothermia demonstrated dramatic protection resulting in significant reduction in the number of damaged axonal profiles. These studies illustrate the diversity and complexity of those axonal responses evoked by traumatic brain injury, suggesting that multiple forms of therapy may be needed to blunt these multifaceted forms of progression.


Subject(s)
Axons/pathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Animals , Brain Injuries/drug therapy , Calcium/physiology , Calpain/metabolism , Cats , Chelating Agents/therapeutic use , Disease Progression , Microscopy, Electron , Nerve Fibers/physiology , Rats , Spectrin/metabolism
11.
J Neurotrauma ; 16(6): 511-21, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10391367

ABSTRACT

Recent observations concerning presumed calcium-induced mitochondrial damage and focal intraaxonal proteolysis in the pathogenesis of traumatic axonal injury (TAI) have opened new perspectives for therapeutic intervention. Studies from our laboratory demonstrated that cyclosporin A (CsA), a potent inhibitor of Ca2+-induced mitochondrial damage, administered 30 min prior to traumatic brain injury preserved mitochondrial integrity in those axonal foci destined to undergo delayed disconnection. We attributed this neuroprotection to the inhibition by CsA of mitochondrial permeability transition (MPT). Additional experiments proved that CsA pretreatment also significantly reduced calcium-induced, calpain-mediated spectrin proteolysis (CMSP) and neurofilament compaction (NFC), pivotal events in the pathogenesis of axonal failure and disconnection. Given these provocative findings the goal of the current study was to evaluate the potential of CsA to inhibit calcium-induced axonal damage in a more clinically relevant postinjury treatment paradigm. To this end, cyclosporin A was administered intrathecally to Sprague Dawley rats 30 min following impact acceleration traumatic brain injury. The first group of animals were sacrificed 120 min postinjury and the density of CMSP and NFC immunoreactive damaged axonal segments of CsA-treated and vehicle-treated injured animals were quantitatively analyzed. A second group of CsA- versus vehicle-treated rats was sacrificed at 24 h postinjury to compare the density of damaged axons displaying beta amyloid precursor protein (APP) immunoreactivity, a signature protein of axonal perturbation and disconnection. Postinjury CsA administration resulted in a significant decrease (>60%) in CMSP/NFC immunoreactivity in corticospinal tracts and medial longitudinal fasciculi. A similar decrease was detected in the density of APP immunoreactive damaged axons, indicating an attenuation of axonal disconnection at 24 h postinjury in CsA-treated animals. These results once again suggest that the maintenance of the functional integrity of the mitochondria can prevent TAI, presumably via the preservation of the local energy homeostasis of the axon. Moreover and perhaps more importantly, these studies also demonstrate the efficacy of CsA administration when given in the early posttraumatic period. Collectively, our findings suggest that a therapeutic window exists for the use of drugs targeting mitochondria and energy regulation in traumatic brain injury.


Subject(s)
Axons/drug effects , Brain Injuries/drug therapy , Cyclosporine/pharmacology , Neuroprotective Agents/pharmacology , Amyloid beta-Protein Precursor/analysis , Animals , Axons/pathology , Biomarkers/analysis , Brain Stem/drug effects , Brain Stem/metabolism , Brain Stem/pathology , Calcium/physiology , Calpain/drug effects , Calpain/physiology , Disease Models, Animal , Disease Progression , Male , Neural Pathways/injuries , Neural Pathways/physiopathology , Neurofilament Proteins/drug effects , Neurofilament Proteins/metabolism , Rats , Rats, Sprague-Dawley , Spectrin/drug effects , Spectrin/metabolism
12.
Neuroreport ; 10(2): 353-8, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-10203334

ABSTRACT

In traumatic axonal injury, Ca2+ influx across a focally damaged axolemma precipitates local mitochondrial failure, degradation of the subaxolemmal spectrin network and compaction of neurofilaments, which collectively contribute to axonal failure. In previous studies, cyclosporin A pretreatment preserved mitochondrial integrity and attenuated axonal failure following trauma. Here we investigate whether this CsA-linked protection was related to the concomitant blunting of intra-axonal, Ca2+-induced cytoskeletal changes in traumatic axonal injury, assessed with antibodies targeting spectrin proteolysis and neurofilament compaction. CsA pretreatment dramatically reduced Ca2+-induced cytoskeletal damage following injury; CsA-treated rats, compared with vehicle-treated rats, displayed a 70% decrease in immunoreactive/damaged profiles. We suggest that CsA-mediated preservation of mitochondrial integrity enables the restoration of ionic and metabolic homeostasis thereby short-circuiting Ca2+-induced proteolysis in injured axons.


Subject(s)
Axons/drug effects , Brain Injuries/pathology , Calcium/physiology , Cyclosporine/pharmacology , Animals , Axons/pathology , Biomarkers , Brain/pathology , Calpain/physiology , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Peptide Hydrolases/metabolism , Pyramidal Tracts/metabolism , Rats , Rats, Sprague-Dawley , Spectrin/metabolism
13.
J Cereb Blood Flow Metab ; 19(4): 443-51, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10197514

ABSTRACT

Traumatic brain injury evokes multiple axonal pathologies that contribute to the ultimate disconnection of injured axons. In severe traumatic brain injury, the axolemma is perturbed focally, presumably allowing for the influx of Ca2+ and initiation of Ca2+ -sensitive, proaxotomy processes. Mitochondria in foci of axolemmal failure may act as Ca2+ sinks that sequester Ca2+ to preserve low cytoplasmic calcium concentrations. This Ca2+ load within mitochondria, however, may cause colloid osmotic swelling and loss of function by a Ca2+ -induced opening of the permeability transition pore. Local failure of mitochondria, in turn, can decrease production of high-energy phosphates necessary to maintain membrane pumps and restore ionic balance in foci of axolemmal permeability change. The authors evaluated the ability of the permeability transition pore inhibitor cyclosporin A (CsA) to prevent mitochondrial swelling in injured axonal segments demonstrating altered axolemmal permeability after impact acceleration injury in rat. At the electron microscopic level, statistically fewer abnormal mitochondria were seen in traumatically injured axons from CsA-pretreated injured animals. Further, this mitochondrial protection translated into axonal protection in a second group of injured rats, whose brains were reacted with antibodies against amyloid precursor protein, a known marker of injured axons. Pretreatment with CsA significantly reduced the number of axons undergoing delayed axotomy, as evidenced by a decrease in the density of amyloid precursor protein-immunoreactive axons. Collectively, these studies demonstrate that CsA protects both mitochondria and the related axonal shaft, suggesting that this agent may be of therapeutic use in traumatic brain injury.


Subject(s)
Axons/drug effects , Brain Injuries/drug therapy , Cyclosporine/therapeutic use , Mitochondria/drug effects , Neuroprotective Agents/therapeutic use , Amyloid beta-Protein Precursor/immunology , Animals , Antibodies, Monoclonal , Axons/pathology , Brain Injuries/pathology , Immunohistochemistry , Injections, Spinal , Male , Mitochondria/pathology , Permeability/drug effects , Rats , Rats, Sprague-Dawley
14.
Brain Res ; 784(1-2): 1-6, 1998 Feb 16.
Article in English | MEDLINE | ID: mdl-9518527

ABSTRACT

Traumatic injury evokes two characteristic forms of focal axonal injury, one of which involves focal perturbation of axolemmal permeability associated with rapid compaction of the underlying axonal neurofilament lattice and microtubular loss. In this process, the neurofilament sidearms have been the subject of intense scrutiny in relation to their role in this NF compaction, with the suggestion that the sidearms, thought to maintain interfilament distance, are proteolytically cleaved and degraded at the time of injury. The current communication addresses the fate of the NF sidearms in such injured axons. Adult cats were subjected to moderate/severe fluid percussion brain injury after intrathecal administration of horseradish peroxidase (HRP). This tracer, excluded by the intact axolemma of uninjured axons, was used to recognize injured axons via HRP intra-axonal uptake/flooding with HRP. Animals were perfused and processed for light microscopic and electron microscopic study of both HRP-containing and non-HRP-containing axons from the same field. HRP-containing axons consistently displayed evidence of traumatically-induced (NF) cytoskeletal collapse. Electron micrographs of HRP-containing axons as well as uninjured, non-HRP-containing axons from the same fields were videographically captured, digitized, enlarged and analysed for NF sidearm length and NF density. HRP-containing axons were found to have increased NF density. Surprisingly, this increased NF density occurred despite the retention of the NF sidearms, which now, however, were reduced in height in comparison to the non-HRP-containing uninjured axons. These observations are not consistent with previously published reports suggesting that overt proteolytic degradation of sidearms was responsible for NF compaction. Based on our findings, we suggest that the NF compaction associated with traumatically-induced axolemmal permeability changes may have its genesis in more subtle sidearm modification, perhaps involving a change in phosphorylation state.


Subject(s)
Axons/physiology , Axons/ultrastructure , Brain Injuries/pathology , Neurites/physiology , Neurites/ultrastructure , Animals , Cats , Histocytochemistry , Horseradish Peroxidase , Image Processing, Computer-Assisted , Male , Microscopy, Electron
15.
Neuroreport ; 7(4): 966-8, 1996 Mar 22.
Article in English | MEDLINE | ID: mdl-8724684

ABSTRACT

The anatomical relationship between nitric oxide synthase (NOS)-positive neurons and blood vessels was examined in the hippocampus of the rat. NADPH-diaphorase histochemistry was used to identify NOS-positive neurons by light-microscopy. A close association of somatic, dendritic and axonal processes of NOS-positive neurons with cerebral blood vessels was observed. These findings suggest the possibility of neurovascular signaling by local NOS-containing neurons, through direct vascular innervation by terminals generating nitric oxide, and paracrine signaling from closely apposed somatic and dendritic neuronal elements.


Subject(s)
Hippocampus/enzymology , Neurons/enzymology , Nitric Oxide Synthase/analysis , Animals , Hippocampus/blood supply , Hippocampus/cytology , Rats , Rats, Sprague-Dawley
16.
J Neurochem ; 65(5): 2072-9, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7595492

ABSTRACT

The A2a adenosine receptor agonist 2(-)[2-(4-amino-3- iodophenyl)ethylamino]adenosine is a potent coronary vasodilator. The corresponding radioiodinated ligand, [125I]APE, discriminates between high- and low-affinity conformations of A2a adenosine receptors. In this study, [125I]APE was used for rapid (24-h) autoradiography in rat brain sections. The pattern of [125I]APE binding is consistent with that expected of an A2a-selective radioligand. It is highest in striatum, nucleus accumbens, and olfactory tubercle, with little binding to cortex and septal nuclei. Specific [125I]APE binding to these brain regions is abolished by 1 microM 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine (CGS-21680) but is little affected by 100 nM 8-cyclopentyl-1,3-dipropylxanthine. Conversion of [125I]APE to the corresponding arylazide results in [125I]AzPE. The rank-order potency of compounds to compete for [125I]AzPE binding in the dark is CGS-21680 > D-(R)-N6-phenylisopropyladenosine > N6- cyclopentyladenosine, indicating that it also is an A2a-selective ligand. Specific photoaffinity labeling by [125I]AzPE of a single polypeptide (42 kDa) corresponding to A2a adenosine receptors is reduced 55 +/- 4% by 100 microM guanosine 5'-O-(3-thiotriphosphate) and 91 +/- 1.3% by 100 nM CGS-21680. [125I]APE and [125I]AzPE are valuable new tools for characterizing A2a adenosine receptors and their coupling to GTP-binding proteins by autoradiography and photoaffinity labeling.


Subject(s)
Adenosine/analogs & derivatives , Brain/metabolism , Receptors, Purinergic P1/metabolism , Adenosine/metabolism , Affinity Labels , Animals , Autoradiography , In Vitro Techniques , Iodine Radioisotopes , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...