Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Neuropathology ; 44(1): 76-82, 2024 Feb.
Article En | MEDLINE | ID: mdl-37403213

Intracranial capillary hemangiomas in adults are rare, and diagnosis can be challenging. Hemangiomas, in general (and particularly in the skin), are more often noted in the pediatric population. Due to the lack of imaging undertaken in the presymptomatic phase, the literature provides few clues on the rate of growth of these unusual tumors. Therefore, we report a case of a 64-year-old man with a medical history of Lyme disease who presented with exhaustion and confusion. Imaging demonstrated an intra-axial lesion with vascularity in the posterior right temporal lobe, raising the possibility of a glioma. Imaging two years prior revealed a very small lesion in the same location. The patient underwent a craniectomy, total resection of the lesion was completed, and his symptoms of confusion resolved. Biopsy revealed a capillary hemangioma composed of small vascular channels lined by endothelial cells and pericytes without smooth muscle. Features of glioma, vascular neoplasms or neuroborreliosis (cerebral Lyme disease) were not identified. Our case documents the growth over two years of a rare intracranial capillary hemangioma in an older adult male.


Brain Neoplasms , Glioma , Hemangioma, Capillary , Hemangioma , Lyme Disease , Humans , Male , Child , Aged , Middle Aged , Endothelial Cells/pathology , Hemangioma, Capillary/surgery , Hemangioma, Capillary/diagnosis , Hemangioma, Capillary/pathology , Hemangioma/pathology , Brain Neoplasms/pathology
2.
Case Rep Rheumatol ; 2019: 9563874, 2019.
Article En | MEDLINE | ID: mdl-30937208

Extraintestinal complications of ulcerative colitis include isolated case reports of cerebral vasculitis. In this case report, we describe autopsy findings in a 50-year-old female who died as a result of massive multifocal cerebral hemorrhage. Microscopic examination of the left colon showed findings typical for ulcerative colitis. Examination of the brain showed an extensive vasculitis. More affected vessels were noted in grey matter than in white matter. Many showed fibrinoid necrosis, invasion by neutrophils and thrombosis. There was extensive perivascular hemorrhage with associated infarction. Vessel analysis shows most of the vessels to have been venous rather than arterial. There were no perivascular sleeves of demyelination to suggest a primary demyelinating disorder, such as acute hemorrhagic leucoencephalitis. Our analysis shows that veins are the likely target of cerebral vasculitis in ulcerative colitis. This has clinical implications because venous occlusion generally causes massive intracerebral hemorrhage with a high mortality.

4.
Acta Neuropathol Commun ; 6(1): 4, 2018 01 04.
Article En | MEDLINE | ID: mdl-29301568

Proteoglycans are promising therapeutic targets in Multiple Sclerosis (MS), because they regulate many aspects of the immune response. This was studied using surfen, an agent that binds both heparan sulphate proteoglycans (HSPGs) and chondroitin sulphate proteoglycans (CSPGs). Initial cell culture work on bone marrow derived macrophages (BMDMs) found that surfen reduced concentrations of the chemokines CCL2, CCL4 and CCL5, with reduced messenger (m)RNA expression for Tumor Necrosis Factor, IL-6, IL-1ß and inducible nitric oxide synthase. These data were further explored using Experimental Autoimmune Encephalomyelitis (EAE) in mice. Surfen reduced clinical signs during EAE when administered from disease onset, and reduced infiltration by CD4 positive T cells and macrophages into the central nervous system. These mice also showed reduced mRNA expression for the chemokines CCL3 and CCL5, with reduced concentrations of CCL2, CCL3 and CCL5. During EAE, surfen treatment induced a persistent increase in Interleukin (IL)-4 concentrations which may enhance T helper 2 responses. During EAE, surfen treatment reduced mRNA expression for HSPGs (NDST1, agrin, syndecan-4, perlecan, serglycin, syndecan-1) and the CSPG versican. By contrast, surfen increased mRNA expression for the CSPG aggrecan, with no effect on neurocan. During EAE, significant positive correlations were found between mRNA expression and clinical score for syndecan-4, serglycin and syndecan-1 and a significant negative correlation for aggrecan. These correlations were absent in surfen treated mice. Repair in the later stages of MS involves remyelination, which was modeled by injecting lysolecithin (lysophosphatidylcholine, LPC) into mouse corpus callosum to create regions of demyelination. When surfen was injected 2 days after LPC, it delayed remyelination of the lesions, but had no effect when injected 7 days after LPC. The delayed remyelination was associated with local increases in CSPG expression. Therefore surfen suppresses inflammation but inhibits remyelination in these models. A mechanism in common may be increased CSPG expression.


Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/pharmacology , Inflammation/drug therapy , Remyelination/drug effects , Urea/analogs & derivatives , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/physiology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/physiology , Cells, Cultured , Chemokines/metabolism , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunologic Factors/administration & dosage , Inflammation/pathology , Inflammation/physiopathology , Macrophages/drug effects , Macrophages/pathology , Macrophages/physiology , Mice, Inbred C57BL , Proteoglycans/metabolism , RNA, Messenger/metabolism , Remyelination/physiology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , Urea/adverse effects , Urea/pharmacology
5.
Can J Neurol Sci ; 44(1): 96-104, 2017 Jan.
Article En | MEDLINE | ID: mdl-27832827

BACKGROUND: Sepsis is a systemic response to infection that can affect brain function by inducing resident cells (including astrocytes and microglia) to generate brain chemokines and cytokines. However, there are few studies on the human brain. Since this information may shed further light on pathogenesis, our study objective was to measure the expression of 36 chemokines and cytokines in autopsied brain from 3 cases of sepsis and 10 controls, and to relate this to astrocyte and microglial activation. METHODS: The right frontal pole was removed at autopsy and chemokine and cytokine expression measured by multiplexed enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction (qPCR). Immunohistochemistry and image analysis were carried out to determine the expression of glial fibrillary acidic protein (GFAP), a marker of activated astrocytes, and CD68 and CD45, markers of activated microglial cells. RESULTS: Concentrations of the chemokines CXCL8, CXCL10, CXCL12, CCL13 and CCL22 were increased in pooled data from the three cases of sepsis (p<0.05); however, their messenger RNA (mRNA) expression was unaltered. CXCL13, CXCL1, CXCL2, CCL1, CCL2, CCL8, CCL20, (interleukin) IL-16, IL-1ß and (tumour necrosis factor) TNF concentrations showed increases in two of three sepsis cases. Additionally, individual sepsis cases showed increases in mRNA expression for HDAC (histone deacetylase) 6 and EIF (eukaryotic translation initiation factor) 4A2. Brain GFAP expression was significantly increased (p<0.05) in pooled data from the three sepsis cases. Individual sepsis cases showed increases in CD68 or CD45 expression. CONCLUSIONS: These expression patterns add to our understanding of the pathogenesis of sepsis and its effects on the brain.


Brain/metabolism , Cytokines/metabolism , Gene Expression Regulation/physiology , Sepsis/pathology , Aged , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Male , Middle Aged , RNA, Messenger/metabolism
6.
PLoS One ; 9(2): e89770, 2014.
Article En | MEDLINE | ID: mdl-24587024

Angiogenesis is the formation of new blood vessels form pre-existing vasculature whose contribution to inflammatory conditions of the Central Nervous System is being studied in order to generate novel therapeutic targets. This study is the first to investigate the impact of two particular angiogenesis inhibitors on murine Experimental Autoimmune Encephalomyelitis (EAE), an inflammatory disease that mimics aspects of the human disease Multiple Sclerosis. The inhibitors were chosen to reduce angiogenesis by complimentary means. Extrinsic factors were targeted with B20-4.1.1 through its ability to bind to murine Vascular Endothelial Growth Factor (VEGF). Vascular processes connected to angiogenesis were targeted directly with K(1-3), the first three kringle domains of angiostatin. Mice treated with B20-4.1.1 and K(1-3) from onset of signs had reduced clinical scores 18-21 days after EAE induction. Both agents suppressed spinal cord angiogenesis without effect on local VEGF expression. B20-4.1.1 reduced spinal cord vascular permeability while K(1-3) had no effect. T cell infiltration into the spinal cord at day 21 was unaffected by either treatment. B20-4.1.1 reduced peripheral T cell proliferation while K(1-3) had no effect. Lymphoid cells from treated mice produced reduced levels of the T helper-17 (Th-17) cell cytokine interleukin (IL)-17 with no effect on the Th-1 cytokine interferon (IFN)-γ or Th-2 cytokine IL-4. However, when both drugs were added in vitro to naive T cells or to antigen stimulated T cells from mice with untreated EAE they had no effect on proliferation or levels of IL-17 or IFN-γ. We conclude that these angiogenesis inhibitors mitigate EAE by both suppressing spinal cord angiogenesis and reducing peripheral T cell activation.


Angiogenesis Inhibitors/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/therapeutic use , Angiostatins , Animals , Capillary Permeability/drug effects , Encephalomyelitis, Autoimmune, Experimental/complications , Immunohistochemistry , Lymphocyte Activation/drug effects , Mice , Neovascularization, Pathologic/etiology , Spinal Cord/blood supply , Spinal Cord/drug effects , T-Lymphocytes/drug effects , Vascular Endothelial Growth Factor A/metabolism
7.
Biochem Biophys Res Commun ; 443(2): 524-30, 2014 Jan 10.
Article En | MEDLINE | ID: mdl-24315874

Surfen (bis-2-methyl-4-amino-quinolyl-6-carbamide) binds to glycosaminoglycans (GAGs) and has been shown to influence their function, and the function of proteoglycans (complexes of GAGs linked to a core protein). T cells synthesize, secrete and express GAGs and proteoglycans which are involved in several aspects of T cell function. However, there are as yet no studies on the effect of GAG-binding agents such as surfen on T cell function. In this study, surfen was found to influence murine T cell activation. Doses between 2.5 and 20 µM produced a graduated reduction in the proliferation of T cells activated with anti-CD3/CD28 antibody-coated T cell expander beads. Surfen (20 mg/kg) was also administered to mice treated with anti-CD3 antibody to activate T cells in vivo. Lymphocytes from surfen-treated mice also showed reduced proliferation and lymph node cell counts were reduced. Surfen reduced labeling with a cell viability marker (7-ADD) but to a much lower extent than its effect on proliferation. Surfen also reduced CD25 (the α-subunit of the interleukin (IL)-2 receptor) expression with no effect on CD69 expression in T cells treated in vivo but not in vitro. When receptor activation was bypassed by treating T cells in vitro with phorbyl myristate acetate (10 ng/ml) and ionomycin (100 ng/ml), surfen treatment either increased proliferation (10 µM) or had no effect (2.5, 5 and 20 µM). In vitro treatment of T cells with surfen had no effect on IL-2 or interferon-γ synthesis and did not alter proliferation of the IL-2 dependent cell line CTLL-2. The effect of surfen was antagonized dose-dependently by co-treatment with heparin sulfate. We conclude that surfen inhibits T cell proliferation in vitro and in vivo. When T cell receptor-driven activation is bypassed surfen had a neutral or stimulatory effect on T cell proliferation. The results imply that endogenous GAGs and proteoglycans play a complex role in promoting or inhibiting different aspects of T cell activation.


Cytokines/metabolism , Glycosaminoglycans/metabolism , Lymphocyte Activation/physiology , T-Lymphocytes/metabolism , Urea/analogs & derivatives , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , Urea/pharmacology
9.
Int Immunopharmacol ; 17(4): 1218-25, 2013 Dec.
Article En | MEDLINE | ID: mdl-23827753

Neutrophils are first responders to injury in inflammatory diseases of the central nervous system (CNS) such as ischemic stroke, trauma and intracerebral hemorrhage. Studies carried out in the last three decades showed that neutrophils have mixed effects in animal models of stroke. Some studies correlated the presence of neutrophils to injury. When neutrophil infiltration was reduced by targeting CD18 or intercellular adhesion molecule-1 (ICAM-1) this generated improved outcomes. However other studies showed that when neutrophil infiltration was stimulated prior to stroke, this reduced the burden of disease. Clinical trials did not show a benefit in stroke patients from neutrophil blockade. Neutrophils may be subject to a threshold effect. When they reach a critical ratio relative to the volume of injury in the CNS, they adopt an anti-inflammatory phenotype that is able to reduce disease. When neutrophil infiltration was stimulated by injecting a stroke site in the rat with the chemokine CXCL1, this resulted in reductions in vascular permeability. Similar reductions in permeability were modeled in tissue culture models, in which neutrophils were applied to monolayers of brain endothelial cells. Neutrophils blocked the permeability increases associated with oxygen-glucose deprivation in human brain endothelial monolayers. The evidence suggests that neutrophils might adopt a pro-inflammatory N1 phenotype or an anti-inflammatory N2 phenotype in the CNS depending on environmental cues. The N2 phenotype may be adopted when neutrophil numbers exceed a critical threshold. This suggests that strategies that promote neutrophil infiltration into stroke, and other CNS inflammatory diseases, could result in improved outcomes.


Central Nervous System/immunology , Neutrophils/immunology , Stroke/immunology , Animals , Humans
10.
Int J Surg Pathol ; 21(4): 411-2, 2013 Aug.
Article En | MEDLINE | ID: mdl-23248340

We present a case of clear cell meningioma with unusual clinical and pathologic features. The patient was a 54-year-old man who underwent laminectomy and durotomy for an intradural tumor in the lumbar spinal canal. Sections showed a predominance of dense collagenous tissue with irregularly shaped and irregularly sized magenta-colored extracellular deposits. On electron microscopy, these deposits were osmiophilic and "petaloid." The final diagnosis of clear cell meningioma rested on relatively inconspicuous intervening nests of glycogen-containing clear cells that were positive for epithelial membrane antigen. The unusual extracellular deposits seen in this case have previously been characterized as tyrosine-rich crystals of the type most commonly seen in salivary gland tumors. Recognition of this tumor as a clear cell meningioma, despite misleading clinical features and initially challenging histologic findings, is not only a matter of diagnostic accuracy but also imparts important prognostic information.


Inclusion Bodies/ultrastructure , Meningeal Neoplasms/ultrastructure , Meningioma/ultrastructure , Tyrosine , Crystallization , Humans , Male , Middle Aged
11.
J Neuropathol Exp Neurol ; 71(11): 983-99, 2012 Nov.
Article En | MEDLINE | ID: mdl-23037326

Angiogenesis in the animal model of multiple sclerosis experimental autoimmune encephalomyelitis (EAE) is regulated by vascular endothelial growth factor (VEGF) and angiopoietin-2. We determined whether VEGF blockade with the anti-VEGF monoclonal antibody bevacizumab could inhibit angiogenesis and affect peripheral pathogenic immune responses in EAE. Mice treated with bevacizumab from the time of onset of clinical signs showed reduced clinical and pathologic scores. Bevacizumab suppressed angiogenesis and reduced angiopoietin-2 expression at Day 21 but had no effect on VEGF upregulation at Day 14. Messenger RNA levels for the angiogenesis-related protein CD105 were increased at Day 14. Bevacizumab reduced vascular permeability in the spinal cord at Day 14 and Day 21. In peripheral lymph nodes, it induced retention of CD4-positive T cells and inhibited T-cell proliferation. It also reduced mononuclear cell infiltration into spinal cord and the relative proportion of T cells. Isolated lymphoid cells showed reduced secretion of the T-helper 17 (Th-17) cell cytokine interleukin 17 and the Th-1 cytokine interferon-γ. When bevacizumab was added to naive T cells or to antigen-stimulated T cells from mice with untreated EAE in vitro, it had no effect on proliferation or the secretion of interleukin 17 or interferon-γ. These data indicate that bevacizumab ameliorates vascular and T-cell responses during EAE, but its effects on T cells may be indirect, possibly by suppressing angiogenesis.


Antibodies, Monoclonal, Humanized/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Epitopes, T-Lymphocyte/metabolism , Neovascularization, Pathologic/drug therapy , Spinal Cord/blood supply , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Severity of Illness Index , Spinal Cord/pathology
12.
Adv Exp Med Biol ; 763: 1-19, 2012.
Article En | MEDLINE | ID: mdl-23397617

The blood-brain barrier refers to the very low permeability across microvessels in the Central Nervous System (CNS), created by the interaction between vascular endothelial cells and surrounding cells of the neurovascular unit. Permeability can be modulated (increased and decreased) by a variety of factors including inflammatory mediators, inflammatory cells such as neutrophils and through alterations in the phenotype of blood vessels during angiogenesis and apoptosis. In this chapter, some of these factors are discussed as well as the challenge of treating harmful increases in permeability that result in brain swelling (vasogenic cerebral edema).


Blood-Brain Barrier/metabolism , Capillary Permeability , Endothelium, Vascular/metabolism , Angiopoietin-1/metabolism , Animals , Apoptosis , Aquaporin 4/metabolism , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Bradykinin/pharmacology , Brain Edema/metabolism , Brain Edema/pathology , Cell Movement , Endothelium, Vascular/physiology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neutrophils/metabolism , Rats , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Stroke/metabolism , Stroke/pathology , Vascular Endothelial Growth Factor A/metabolism
13.
J Neuropathol Exp Neurol ; 70(12): 1107-23, 2011 Dec.
Article En | MEDLINE | ID: mdl-22082662

The regulation of angiogenesis was studied over the course of the animal model of multiple sclerosis, acute experimental autoimmune encephalomyelitis (EAE) in mice using immunohistochemistry. During EAE, angiogenesis peaked 21 days after disease induction, with significant increases in gray matter and adjacent to the leptomeninges. Angiogenesis correlated with clinical and pathologic scores. Spinal cord expression of angiopoietin 1 (Ang-1) by neurons and glia was reduced at Day 14, but expression by inflammatory cells restored earlier levels at Day 21. Angiopoietin 2 expression increased markedly at Day 21 and was mostly associated with inflammatory cells. Levels of the angiopoietin receptor Tie-2 were reduced at Day 14, but recovered by day D21. Double labeling demonstrated Ang-1 expression on infiltrating CD3-positive T cells; Ang-2 was expressed by monocytes/macrophages. During EAE, the expression of vascular endothelial growth factor peaked at Day 14 and began to decrease by Day 21. Double labeling showed expression of Tie-2 and vascular endothelial growth factor receptor 2 but not Ang-2 in blood vessels at Day 21. Vascular permeability increased early in EAE, but was reduced by Day 21. Although individual values did not correlate with angiogenesis, the volume of permeable tissue showed a weak positive correlation with angiogenesis. These temporal changes in angiogenic factors suggest an integral role during EAE-related angiogenesis.


Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Capillary Permeability/physiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelium, Vascular/metabolism , Mice , Neovascularization, Pathologic/physiopathology , Neuroglia/metabolism , Neurons/metabolism , Receptor, TIE-2/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology
14.
Curr Neurovasc Res ; 8(1): 14-24, 2011 Feb.
Article En | MEDLINE | ID: mdl-21208159

Angiogenesis is implicated in diseases of the central nervous system, and its modulation represents an attractive therapeutic strategy. This is the first report of the effects of resveratrol (trans-3,4',5-trihydroxystilbene) on cerebral angiogenesis, using an in vitro model. Processes associated with angiogenesis were studied in cerebral vascular endothelial cells, using the human brain endothelial cell line hCMEC/D3 and primary bovine brain microvessel EC (BBMEC). Comparisons were made to human umbilical vein EC (HUVEC). In cerebral cultures, resveratrol (24h) induced a dose dependent reduction in BrdU incorporation and cell numbers (MTT assay) between 10-100 µM, while lower doses (100 nM-5 µM) had no effect. Cell migration (scratch assay) was inhibited between 10-100 µM depending on cell type. Doses between 10-100 µM reduced average tubule length on Matrigel, while higher doses (50,100 µM) also inhibited process formation around explanted rat aortic rings (ex vivo assay). Cell cycle analysis in hCMEC/D3 (propidium iodide) showed reduced progression to the G2/M phase, with a maximal effect at 25 µM. Resveratrol did not induce apoptosis in hCMEC/D3, based on caspase-3 activity. Cytotoxicity (LDH release) was induced by resveratrol (50,100 µM) in hCMEC/D3 and HUVEC, peaking at ~20% in hCMEC/D3 and ~35% in HUVEC. Cytotoxic effects were not detected in BBMEC. Resveratrol (10-50 µM) inhibited phosphorylation of the serine/threonine kinase Akt, by Western blot (15 min, 1h) with a prolonged inhibition (24h) for 25 µM. In conclusion, this study shows inhibitory effects of resveratrol on cerebral angiogenesis, using an in vitro model. This is discussed in terms of dosage, in vivo equivalence and therapeutic potential.


Angiogenesis Inhibitors/pharmacology , Cerebral Arteries/drug effects , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Stilbenes/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cattle , Cell Line , Cells, Cultured , Cerebral Arteries/enzymology , Cerebral Arteries/physiopathology , Endothelial Cells/enzymology , Endothelial Cells/pathology , Humans , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/physiopathology , Resveratrol , Stilbenes/therapeutic use
15.
Brain Res ; 1332: 20-31, 2010 May 21.
Article En | MEDLINE | ID: mdl-20346925

Experimentally, oxygen glucose deprivation (OGD) has been widely used to mimic the environmental conditions present during cerebral ischemia-reperfusion (IR) injury. OGD is known to increase permeability across cultured cerebral endothelial cells, which models the effect of IR on permeability across the blood-brain barrier (BBB); however, studies have yet to be performed in a human model. The effect of neutrophils on the increase in BBB permeability associated with IR injury has yet to be modeled in vitro. To address these questions, the human brain endothelial cell line hCMEC/D3 was exposed to OGD with reoxygenation, and permeability was measured for a range of OGD exposure times (1-24h). One hour of exposure to OGD induced a reversible increase in permeability, unassociated with cytotoxicity (assessed from lactate dehydrogenase release). However, 12-24h OGD exposures induced sustained increases in permeability associated with cytotoxicity. The 1h permeability increase was inhibited with the nitric oxide synthase inhibitors l-NAME (10(-)(7)mol/l) and 1400W (10(-)(7)mol/l). Neutrophils (5x10(6)/ml) blocked the permeability increase associated with 1h OGD, whether applied during or after OGD exposure. Permeability remained low if neutrophils were activated with leukotriene (Lt)B(4) (10(-)(7)mol/l) or exposed to a transendothelial gradient of LtB(4), while neutrophil activation with phorbyl myristate acetate (4x10(-)(8)mol/l) induced a small increase. Neutrophils had no effect on the permeability increase induced by 12h OGD exposure. This study finds that OGD induces reversible increases in permeability linked to nitric oxide synthesis in a human culture model of the BBB and shows that neutrophils mitigate permeability increases in this context.


Blood-Brain Barrier/physiology , Capillary Permeability/physiology , Cell Hypoxia/physiology , Endothelial Cells/physiology , Glucose/metabolism , Neutrophils/physiology , Brain/drug effects , Brain/physiology , Capillary Permeability/drug effects , Cell Count , Cell Line , Endothelial Cells/drug effects , Glucose/deficiency , Humans , L-Lactate Dehydrogenase/metabolism , Models, Biological , Reperfusion Injury/physiopathology , Time Factors
16.
Biochem Biophys Res Commun ; 391(1): 936-41, 2010 Jan 01.
Article En | MEDLINE | ID: mdl-19962958

Tumor necrosis factor (TNF) and its related ligands TNF-related apoptosis inducing ligand (TRAIL) and Fas ligand (FasL) play roles in the regulation of vascular responses, but their effect on the formation of new blood vessels (angiogenesis) is unclear. Therefore, we have examined the effects of these ligands on angiogenesis modeled with primary cultures of human umbilical vein endothelial cells (HUVEC). To examine angiogenesis in the context of the central nervous system, we have also modeled cerebral angiogenesis with the human brain endothelial cell line hCMEC/D3. Parameters studied were bromodeoxyuridine (BrdU) incorporation and cell number (MTT) assay (to assess endothelial proliferation), scratch assay (migration) and networks on Matrigel (tube formation). In our hands, neither TRAIL nor FasL (1, 10, and 100 ng/ml) had an effect on parameters of angiogenesis in the HUVEC model. In hCMEC/D3 cells by contrast, TRAIL inhibited all parameters (10-100 ng/ml, 24h). This was due to apoptosis, since its action was blocked by the pan-caspase inhibitor zVADfmk (5 x 10(-5) mol/l) and TRAIL increased caspase-3 activity 1h after application. However FasL (100 ng/ml) increased BrdU uptake without other effects. We conclude that TRAIL has different effects on in vitro angiogenesis depending on which model is used, but that FasL is generally ineffective when applied in vitro. The data suggest that TRAIL primarily influences angiogenesis by the induction of vascular endothelial apoptosis, leading to vessel regression.


Apoptosis , Brain/blood supply , Endothelium, Vascular/physiology , Neovascularization, Physiologic , TNF-Related Apoptosis-Inducing Ligand/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fas Ligand Protein/pharmacology , Fas Ligand Protein/physiology , Humans , TNF-Related Apoptosis-Inducing Ligand/pharmacology
17.
Brain Res ; 1298: 13-23, 2009 Nov 17.
Article En | MEDLINE | ID: mdl-19728990

The blood-brain barrier (BBB) restricts solute permeability across healthy cerebral endothelial cells. However, during inflammation, permeability is increased and can lead to deleterious cerebral edema. Neutrophils are early cellular participants in acute inflammation, but their effect on BBB permeability is unclear. To study this, neutrophils were applied in a resting and activated state to in vitro and in vivo models of the BBB. In vitro, human neutrophils (5 x 10(6)/ml) were activated with tumor necrosis factor (100 U/ml) and leukotriene B(4) (10(-7) mol/l). Untreated neutrophils reduced permeability across the human brain endothelial cell line hCMEC/D3. Activated neutrophils returned permeability to baseline, an effect blocked by the reactive oxygen scavengers superoxide dismutase (10 U/ml) and catalase (1000 U/ml). In vivo, human neutrophils (2.5 x1 0(5) in 4 microl) were injected into the striatum of anesthetized juvenile Wistar rats, and BBB permeability measured 30 min later. This was compared to control injections (4 microl) of vehicle (0.9% saline) and arachidonic acid (10(-3) mol/l). The injection generated a small hematoma around the injection tract (<3 microl). Untreated neutrophils induced significantly lower permeability in their vicinity than activated neutrophils, with a trend to lowered permeability compared to the vehicle control. Neither untreated nor activated neutrophils induced permeability increases, while arachidonic acid increased permeability as a positive control. This study further delineates the effect of neutrophils on the BBB, and demonstrates that resting neutrophils induce acute reductions in permeability while activated neutrophils have a neutral effect. The in vivo model reiterates some aspects of acute intracerebral hemorrhage.


Blood-Brain Barrier/metabolism , Capillary Permeability/physiology , Inflammation/metabolism , Neutrophils/metabolism , Animals , Blood-Brain Barrier/drug effects , Catalase/pharmacology , Cell Line , Chi-Square Distribution , Endothelial Cells/metabolism , Endothelium/metabolism , Humans , Neutrophil Activation , Rats , Rats, Wistar , Staining and Labeling , Superoxide Dismutase/pharmacology
19.
Br J Pharmacol ; 146(4): 576-84, 2005 Oct.
Article En | MEDLINE | ID: mdl-16056236

We have examined the role of TRPV1 activation in disrupting the blood-brain barrier by measuring the permeability of single pial venular capillaries in anaesthetized rats. Capsaicin application to the brain surface resulted in increased permeability, maximal 2.1+/-0.12 x 10(-6) cm s(-1) (mean+/-s.e.m.) with log EC50 -4.5+/-0.10. Substance P methyl ester gave a similar response (maximal 2.0+/-0.07, n = 6, log EC50 -4.8+/-0.07), but the selective NK2 agonist, beta-Ala8-NKA(4-10) peptide, had no effect. Although CGRP decreased the permeability of venules (log EC50 10.3+/-0.11), its receptor antagonist CGRP(8-37) had no effect on the response to capsaicin. The TRPV1 antagonist capsazepine (1 mM) reduced the response to capsaicin (100 microM), from 1.78+/-0.15 to 0.63+/-0.10 (n = 4). The NK1 receptor antagonists GR205171 (100 microM) and SDZ NKT 376 (1 mM) also reduced the response to capsaicin (from 1.75+/-0.14 to 0.46+/-0.08; n = 6, and from 1.85+/-0.13 to 0.48+/-0.05; n = 5, respectively), indicating that capsaicin acts via TRPV1 in series with NK(1). Starch microspheres were used to produce transient focal ischaemia. Permeability was increased on reperfusion to a greater extent and more rapidly in vessels with diameter greater than 40 microm than those less than 15 microm. Capsazepine given intraperitoneally during ischaemia reduced the permeability increase in small venules from 5.9+/-0.3 to 2.4+/-0.1, and from 11.4+/-0.8 to 5.1+/-0.9 in large venules. In conclusion, the TRPV1 receptor is active in the brain microvasculature and has its permeability-increasing effect via substance P. It also plays a role in the immediate blood-brain barrier disruption following ischaemia-reperfusion.


Blood-Brain Barrier/metabolism , Capillaries/metabolism , Capillary Permeability/drug effects , TRPV Cation Channels/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain Ischemia/chemically induced , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Capillaries/drug effects , Capillaries/pathology , Capillary Permeability/physiology , Capsaicin/administration & dosage , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Male , Microspheres , Piperidines/pharmacology , Rats , Rats, Wistar , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/metabolism , Starch , Substance P/analogs & derivatives , Substance P/pharmacology , TRPV Cation Channels/drug effects , Tetrazoles/pharmacology , Time Factors
20.
J Vasc Res ; 42(4): 337-47, 2005.
Article En | MEDLINE | ID: mdl-15985761

BACKGROUND: Apoptosis of vascular endothelial cells plays a central role in angiogenesis and atherosclerosis. This study investigates the molecular mechanisms of endothelial apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) following inhibition of phosphatidylinositol 3-kinase (PI3K). It examines downstream regulation and activation of the extrinsic and intrinsic pathways. METHODS AND RESULTS: By flow cytometry, TRAIL receptors 2 and 3 were present to a greater extent than receptors 1 and 4. TRAIL reduced cell numbers in combination with the PI3K inhibitor LY 294002. TRAIL (100 ng/ml) with LY 294002 (20 micromol/l) activated the extrinsic pathway, causing progressive cleavage of caspase-8 and caspase-3. Activation of the intrinsic pathway proceeded by release of mitochondrial factors Smac/DIABLO and cytochrome c, and caspase-9 cleavage. LY 294002 reduced phosphorylated Akt (p-Akt), with early loss of the short form of cellular FLIP (c-FLIP(S)) and concurrent reduction of Bcl-2. Treatment with small interfering RNA against PI3K also reduced c-FLIP(S) and Bcl-2, and cotreatment with TRAIL triggered caspase-3 cleavage. CONCLUSIONS: This study details the molecular regulation of TRAIL-induced apoptosis in vascular endothelium. Inhibition of PI3K reduces p-Akt, with concurrent reductions in c-FLIP(S) and Bcl-2, and so renders endothelium sensitive to TRAIL-induced apoptosis through the extrinsic and intrinsic pathways.


Apoptosis/drug effects , Endothelium, Vascular/drug effects , Intracellular Signaling Peptides and Proteins/analysis , Membrane Glycoproteins/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins/physiology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Cells, Cultured , Chromones/pharmacology , Endothelium, Vascular/pathology , Humans , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/analysis , TNF-Related Apoptosis-Inducing Ligand
...