Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Plast Reconstr Aesthet Surg ; 75(1): 217-225, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34266803

RESUMO

INTRODUCTION: In many patients with cutaneous melanoma that affects the trunk area, there is lymphatic drainage to multiple basins (MLBD). This study aimed to examine whether MLBD is associated with disease outcomes. METHODS: Lymphatic mapping and sentinel lymph node (SLN) biopsy were performed in 161 patients with truncal melanoma. The number and location of draining nodal basins were established during the preoperative lymphoscintigraphy using technetium-99 m rhenium sulphide nanocolloid. RESULTS: MLBD was present in 59 (37%) patients, and single lymphatic basin drainage (SLBD) in 102 (63%) patients. Patients with MLBD showed no increased risk for SLN metastasis compared to patients with SLBD (27% versus 29%, respectively). There was no significant difference in disease-free survival (DFS) between patients with MLBD and those with SLBD. Five-year DFS was 64% for patients with MLBD and SLBD. Multivariate analysis showed that the presence of ulceration (p = 0.01) was an independent predictor of SLN metastasis, while melanoma thickness (p = 0.01) and SLN metastasis (p = 0.01) were independent predictors of DFS. In patients with a negative SLN, five-year DFS was 74% for patients with MLBD and 73% for those with SLBD. Multivariate analysis showed that melanoma thickness (p = 0.00) was an independent predictor of DFS. CONCLUSION: MLBD does not negatively impact the disease outcome in patients with truncal melanoma.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Excisão de Linfonodo , Linfonodos/patologia , Metástase Linfática/patologia , Melanoma/diagnóstico por imagem , Melanoma/patologia , Melanoma/cirurgia , Prognóstico , Biópsia de Linfonodo Sentinela , Neoplasias Cutâneas/diagnóstico por imagem , Neoplasias Cutâneas/cirurgia , Síndrome
2.
Oral Dis ; 21(4): 536-43, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25639284

RESUMO

OBJECTIVES: This study examined the single nucleotide polymorphisms (SNPs) in high-mobility group box 1 (HMGB1) gene in patients with oral squamous cell carcinoma (OSCC) and oral lichen planus (OLP). MATERIALS AND METHODS: The study was conducted on 93 patients with OSCC, 53 patients with OLP, and 100 controls, all Caucasians of the same ethnicity, matched by age. HMGB1 genotypes for 4 SNPs, 2262G/A (rs1045411), 1177G/C (rs3742305), 3814C/G (rs2249825), and rs4540927, were assessed using TaqMan SNP Genotyping Assays, Applied Biosystems. RESULTS: The HMGB1 1177GG genotype was associated with lymph-node metastasis and tumor stage in OSCCs (P = 0.016 and P = 0.030, respectively). Genotype 1177GG resulted in poorer recurrence-free survival (RFS), P = 0.000. The 1177G/C polymorphism was an independent predictor of RFS compared to GG genotype, P = 0.001. The three polymorphisms were in linkage disequilibrium (LD). The AGC and GGC haplotypes were associated with an increased oral cancer risk, determined over the haplotype odds ratios (HOR = 13.316, P = 0.015, and HOR = 5.769, P = 0.029, respectively). The AGC haplotype was related to erosive OLP progression to OSCC (HOR = 12.179, P = 0.001). CONCLUSIONS: HMGB1 polymorphism 1177G/C could be associated with tumor progression and recurrence-free survival in patients with OSCC. The haplotypes of HMGB1 gene might be associated with susceptibility to OSCC and OLP progression to OSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Proteína HMGB1/genética , Líquen Plano Bucal/genética , Neoplasias Bucais/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/patologia , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/patologia , Polimorfismo de Nucleotídeo Único
3.
Br J Cancer ; 105(11): 1697-707, 2011 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22027709

RESUMO

BACKGROUND: ANG1005 consists of three molecules of paclitaxel conjugated via ester bonds to the 19-amino-acid peptide Angiopep-2. The new chemical agent has been shown to cross the blood-brain barrier (BBB) by receptor-mediated transcytosis via low-density lipoprotein receptor-related protein 1 (LRP1). The experiments here examined the role of LRP1 in the subsequent endocytosis of drug into cancer cells. METHODS: Localisation of ANG1005 and Angiopep-2 was examined by immunohistochemistry and in-vivo near-infrared fluorescence imaging in mice carrying orthotopic glioma tumours. Transport of ANG1005 and Angiopep-2 was examined in U87 glioblastoma cell lines. RESULTS: Systemically administered ANG1005 and Cy5.5Angiopep-2 localised to orthotopic glioma tumours in mice. The glioma transplants correlated with high expression levels of LRP1. Decreasing LRP1 activity, by RNA silencing or LRP1 competitors, decreased uptake of ANG1005 and Angiopep-2 into U87 glioblastoma cells. Conversely, LRP1 expression and endocytosis rates for ANG1005 and Angiopep-2 increased in U87 cells under conditions that mimicked the microenvironment near aggressive tumours, that is, hypoxic and acidic conditions. CONCLUSION: ANG1005 might be a particularly effective chemotherapeutic agent for the wide array of known LRP1-expressing brain and non-brain cancers, in particular those with an aggressive phenotype.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Paclitaxel/farmacocinética , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Endocitose , Glioma/tratamento farmacológico , Glioma/patologia , Células Hep G2 , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos , Camundongos Nus , Paclitaxel/farmacologia , Peptídeos/farmacocinética , Peptídeos/farmacologia , Fenótipo , Interferência de RNA , Receptores de LDL/genética , Microambiente Tumoral , Proteínas Supressoras de Tumor/genética
4.
Br J Cancer ; 103(10): 1606-16, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20959824

RESUMO

BACKGROUND: Insulin-like growth factor-binding protein 7 (IGFBP7) is an abundant, selective and accessible biomarker of glioblastoma multiforme (GBM) tumour vessels. In this study, an anti-IGFBP7 single-domain antibody (sdAb) was developed to target GBM vessels for molecular imaging applications. METHODS: Human GBM was modelled in mice by intracranial implantation of U87MG.EGFRvIII cells. An anti-IGFBP7 sdAb, isolated from an immune llama library by panning, was assessed in vitro for its binding affinity using surface plasmon resonance and by ex vivo immunobinding on mouse and human GBM tissue. Tumour targeting by Cy5.5-labelled anti-IGFBP7 sdAb as well as by anti-IGFBP7 sdAb conjugated to PEGylated Fe3O4 nanoparticles (NPs)-Cy5.5 were assessed in U87MG.EGFRvIII tumour-bearing mice in vivo using optical imaging and in brain sections using fluorescent microscopy. RESULTS: Surface plasmon resonance analyses revealed a medium affinity (K(D)=40-50 nM) binding of the anti-IGFBP7 sdAb to the purified antigen. The anti-IGFBP7 sdAb also selectively bound to both mouse and human GBM vessels, but not normal brain vessels in tissue sections. In vivo, intravenously injected anti-IGFBP7 sdAb-Cy5.5 bound to GBM vessels creating high imaging signal in the intracranial tumour. Similarly, the anti-IGFBP7 sdAb-functionalised PEGylated Fe3O4 NP-Cy5.5 demonstrated enhanced tumour signal compared with non-targeted NPs. Fluorescent microscopy confirmed the presence of anti-IGFBP7 sdAb and anti-IGFBP7 sdAb-PEGylated Fe3O4 NPs selectively in GBM vessels. CONCLUSIONS: Anti-IGFBP7 sdAbs are novel GBM vessel-targeting moieties suitable for molecular imaging.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/imunologia , Animais , Anticorpos , Encéfalo/imunologia , Encéfalo/patologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/ultraestrutura , Primers do DNA , DNA Complementar/genética , DNA Complementar/imunologia , Amplificação de Genes , Glioblastoma/imunologia , Glioblastoma/ultraestrutura , Humanos , Imunoglobulina G/imunologia , Imuno-Histoquímica , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Camundongos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Distribuição Tecidual
5.
Br J Pharmacol ; 160(4): 1016-28, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20590596

RESUMO

BACKGROUND AND PURPOSE: The overexpression of epidermal growth factor receptor (EGFR) and its mutated variant EGFRvIII occurs in 50% of glioblastoma multiforme. We developed antibody fragments against EGFR/EGFRvIII for molecular imaging and/or therapeutic targeting applications. EXPERIMENTAL APPROACH: An anti-EGFR/EGFRvIII llama single-domain antibody (EG(2)) and two higher valency format constructs, bivalent EG(2)-hFc and pentavalent V2C-EG(2) sdAbs, were analysed in vitro for their binding affinities using surface plasmon resonance and cell binding studies, and in vivo using pharmacokinetic, biodistribution, optical imaging and fluorescent microscopy studies. KEY RESULTS: Kinetic binding analyses by surface plasmon resonance revealed intrinsic affinities of 55 nM and 97 nM for the monovalent EG(2) to immobilized extracellular domains of EGFR and EGFRvIII, respectively, and a 10- to 600-fold increases in apparent affinities for the multivalent binders, V2C-EG(2) and EG(2)-hFc, respectively. In vivo pharmacokinetic and biodistribution studies in mice revealed plasma half-lives for EG(2), V2C-EG(2) and EG(2)-hFc of 41 min, 80 min and 12.5 h, respectively, as well as a significantly higher retention of EG(2)-hFc compared to the other two constructs in EGFR/EGFRvIII-expressing orthotopic brain tumours, resulting in the highest signal in the tumour region in optical imaging studies. Time domain volumetric optical imaging fusion with high-resolution micro-computed tomography of microvascular brain network confirmed EG(2)-hFc selective accumulation/retention in anatomically defined tumour regions. CONCLUSIONS: Single domain antibodies can be optimized for molecular imaging applications by methods that improve their apparent affinity and prolong plasma half-life and, at the same time, preserve their ability to penetrate tumour parenchyma.


Assuntos
Neoplasias Encefálicas/diagnóstico , Receptores ErbB/metabolismo , Glioblastoma/diagnóstico , Cadeias Pesadas de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/metabolismo , Animais , Anticorpos/metabolismo , Afinidade de Anticorpos , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/ultraestrutura , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Glioblastoma/irrigação sanguínea , Glioblastoma/metabolismo , Glioblastoma/ultraestrutura , Meia-Vida , Humanos , Proteínas Imobilizadas/metabolismo , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Cinética , Masculino , Camundongos , Camundongos Nus , Imagem Molecular/métodos , Peso Molecular , Tomografia Óptica/métodos , Imagem Corporal Total/métodos , Microtomografia por Raio-X/métodos
6.
J Microsc ; 232(2): 225-34, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19017221

RESUMO

The localization of asialo-GM1 in ordered membrane raft domains in HeLa cells has been examined using a combination of membrane fractionation and fluorescence imaging. The glycolipid is enriched in Triton X-100 insoluble membrane fractions that contain high concentrations of cholesterol and caveolin-1 but is also found in detergent soluble membrane fractions. Near-field fluorescence microscopy shows that a fraction of the asialo-GM1 is localized in small nanoscale clusters that have an upper limit for the average diameter of approximately 90 nm and are partially colocalized with caveolae membrane domains. In addition to clusters, a diffuse, non-clustered population of asialo-GM1 is observed and is hypothesized to correspond to glycolipid isolated in detergent soluble membrane fractions.


Assuntos
Membrana Celular/química , Gangliosídeo G(M1)/análise , Microscopia de Fluorescência/métodos
7.
Oncogene ; 27(54): 6834-44, 2008 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18711401

RESUMO

Insulin-like growth factor-binding protein 7 (IGFBP7) is a selective biomarker of glioblastoma (GBM) vessels, strongly expressed in tumor endothelial cells and vascular basement membrane. IGFBP7 gene regulation and its potential role in tumor angiogenesis remain unclear. Mechanisms of IGFBP7 induction and its angiogenic capacity were examined in human brain endothelial cells (HBECs) exposed to tumor-like conditions. HBEC treated with GBM cell (U87MG)-conditioned media (-CM) exhibited fourfold upregulation of IGFBP7 mRNA and protein compared to control cells. IGFBP7 gene regulation in HBEC was methylation independent. U87MG-CM analysed by enzyme-linked immunosorbent assay contained approximately 5 pM transforming growth factor (TGF)-beta1, a concentration sufficient to stimulate IGFBP7 in HBEC to similar levels as U87MG-CM. Both pan-TGF-beta-neutralizing antibody (1D11) and the TGF-beta1 receptor (activin receptor-like kinase 5, ALK5) antagonist, SB431542, blocked U87MG-CM-induced IGFBP7 expression in HBEC, indicating that TGF-beta1 is an important tumor-secreted effector capable of IGFBP7 induction in endothelial cells. HBEC exposed to either U87MG-CM or IGFBP7 protein exhibited increased capillary-like tube (CLT) formation in Matrigel. Both TGF-beta1- and U87MG-CM-induced Smad-2 phosphorylation and U87MG-CM-induced CLT formation in HBEC were inhibited by the ALK5 antagonist, SB431542. These data suggest that proangiogenic IGFBP7 may be induced in brain endothelial cells by TGF-betas secreted by GBM, most likely through TGF-beta1/ALK5/Smad-2 pathway.


Assuntos
Endotélio Vascular/fisiologia , Glioblastoma/irrigação sanguínea , Glioblastoma/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Neovascularização Patológica/genética , RNA Mensageiro/genética , Transdução de Sinais/fisiologia , Proteína Smad2/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/fisiologia , Capilares/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Circulação Cerebrovascular/fisiologia , Meios de Cultivo Condicionados , Endotélio Vascular/citologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/efeitos dos fármacos
8.
Toxicol In Vitro ; 19(3): 299-334, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15713540

RESUMO

The aim of the present study was to identify a model for the blood-brain barrier based on the use of a continuous cell line, and to investigate the specificity of this model. A set of test compounds, reflecting different transport mechanisms and different degrees of permeability, as well as different physiochemical properties was selected. In vivo data for transport across the blood-brain barrier of this set of test compounds was generated as part of the study using two different in vivo models. A computational prediction model was also developed, based on 74 proprietary Pharmacia compounds, previously tested in one of the in vivo models. Molsurf descriptors were calculated and 21 descriptors were correlated with log(Brain(conc.)/Plasma(conc.)) using partial least squares projection to latent structures (PLS). However, the correlation between predicted and measured values was found to be rather low and differed between one and two log units for several of the compounds. The test compounds were analyzed in vitro using primary bovine and human brain endothelial cells co-cultured with astrocytes, and also using two different immortalized brain endothelial cell lines, one originating from rat and one from mouse. Cell models using cells not derived from the blood-brain barrier, ECV/C6, MDCK and Caco-2 cell lines, were also used. No linear correlation between in vivo and in vitro permeability was found for any of the in vitro models when all compounds were included in the analysis. The highest r2 values were seen in the bovine brain endothelial cells (r2=0.43) and MDCKwt (r2=0.46) cell models. Higher correlations were seen when only passively transported compounds were included in the analysis, bovine brain endothelial cells (r2=0.74), MDCKwt (r2=0.65) and Caco-2 (r2=0.86). By plotting in vivo Papp values against logDpH7.4 it was possible to classify compounds into four different classes: (1) compounds crossing the blood-brain barrier by passive diffusion, (2) compounds crossing the blood-brain barrier by blood-flow limited passive diffusion, (3) compounds crossing the blood-brain barrier by carrier mediated influx, and (4) compounds being actively excreted from the brain by active efflux. Papp and Pe values obtained using the different in vitro models were also plotted against logDpH7.4 and compared to the plot obtained when in vivo Papp values were used. Several of the in vitro models could distinguish between passively distributed compounds and efflux substrates. Of the cell lines included in the present study, the MDCKmdr-1 cell line gave the best separation of passively and effluxed compounds. Ratios between AUC in brain and AUC in blood were also calculated for six of the compounds and compared to ratios between Pe or Papp for transport in the apical to basolateral and basolateral to apical direction. Again the MDCKmdr-1 cell line gave the best correlation with only one compound (AZT) giving large discrepancy between in vitro and in vivo data. None of the in vitro models could identify compounds known to be substrates for carrier mediated influxed as such, and the results indicate that a tighter in vitro blood-brain barrier model probably is needed in order to facilitate studies on carrier mediated influx. The findings presented also indicate that identification of "batteries" of in vitro tests are likely to be necessary in order to improve in vitro-in vivo correlations and to make it possible to perform acceptable predictions of in vivo brain distributions from in vitro data.


Assuntos
Barreira Hematoencefálica/citologia , Células Cultivadas/metabolismo , Endotélio Vascular/citologia , Modelos Biológicos , Xenobióticos/farmacocinética , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Bovinos , Cães , Endotélio Vascular/metabolismo , Humanos , Camundongos , Permeabilidade , Ratos , Reprodutibilidade dos Testes
9.
Brain Res ; 1003(1-2): 151-8, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-15019574

RESUMO

The two inducible enzymes, manganese superoxide dismutase (MnSOD) and heme-oxygenase-1 (HO-1) may participate in the cellular defense of brain endothelium against oxidative stress. The time-dependent expression of MnSOD and HO-1 mRNAs and proteins was investigated in vitro in rat cerebral endothelial cells (CEC) subjected to sublethal mild or moderate hydroxyl radical-induced oxidative stress. Mild oxidative stress induced increases in both MnSOD and HO-1 mRNA and protein expression. Moderate oxidative stress resulted in a significant reduction in HO-1 mRNA and protein expression, whereas MnSOD expression pattern was similar to that observed after mild oxidative stress. A profound protein loss of both MnSOD and HO-1 was detected 24 h after exposure of CEC to a moderate oxidative stress. The data indicate that cerebral endothelial cells respond by increasing the expression of antioxidant defense enzymes in a manner dependent on the oxidative stress intensity.


Assuntos
Córtex Cerebral/enzimologia , Células Endoteliais/enzimologia , Heme Oxigenase (Desciclizante)/biossíntese , Estresse Oxidativo/fisiologia , Superóxido Dismutase/biossíntese , Animais , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Compostos de Ferro/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Superóxido Dismutase/genética
10.
J Cereb Blood Flow Metab ; 21(11): 1310-9, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11702046

RESUMO

SUMMARY: Although the neuroprotective effects of hypothermia have been known for a long time, the molecular correlates of this neuroprotection are poorly understood. In this study, the authors investigated how hypothermia affects inflammatory responses in the brain elicited by systemic injection of IL-1 beta. Leukocyte rolling and adhesion were quantified in pial venules (20 to 50 microm) of C57/Bl6 mice 4 hours after intraperitoneal injection of IL-1 beta (5 microg/kg) using an open cranial window and intravital microscopy. Animals were subjected to moderate hypothermia (32 degrees C) or normothermia (37 degrees C) for 1 or 4 hours after IL-1 beta injection. Significant increases in leukocyte rolling and adhesion were observed in IL-1 beta-injected animals as compared with sham controls. Whereas 1-hour hypothermia did not affect IL-1 beta-induced leukocyte rolling and adhesion, 4-hour hypothermia caused a reduction in both rolling and adhesion. Molecular mechanisms of hypothermic effects were investigated in cultured human cerebral endothelial cells exposed to IL-1 beta (50 U/mL) for 4 hours at 37 degrees C or 32 degrees C followed by 18 hours at 37 degrees C. Human cerebral endothelial cells exposed to IL-1 beta at 32 degrees C showed attenuated NF-kappa B activation determined by the Luciferase yellow reporter gene assay and reduced expression of IL-8 and IL-1 beta measured by reverse transcriptase-polymerase chain reaction and enzyme-linked immunosorbent assay. Intracellular adhesion molecule-1 was induced to similar levels (threefold over control) at both temperatures. The expression of CD18 on neutrophils in vitro was not affected by either IL-1 beta or hypothermia. These findings suggest that mechanisms by which hypothermia reduces leukocyte rolling and adhesion include suppression of inflammatory gene transcription in brain endothelial cells.


Assuntos
Barreira Hematoencefálica/imunologia , Antígenos CD18/genética , Hipotermia Induzida , Interleucina-1/farmacologia , Neutrófilos/citologia , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Endotélio Vascular/citologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Interleucina-1/genética , Interleucina-8/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Neutrófilos/fisiologia , Pia-Máter/irrigação sanguínea , Pia-Máter/imunologia
11.
J Neuroimmunol ; 119(2): 365-76, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11585641

RESUMO

Mechanisms of hypoxia-induced activation of nuclear factor-kappaB (NF-kappaB) and inflammatory genes were investigated in fetal human astrocytes in culture. Astrocytes were subjected to interleukin-1beta (IL-1beta; 50-100 u/ml; 4-24 h), or to a 4-h hypoxia (<2% O2) followed by a 4-24-h reoxygenation. NF-kappaB binding and transcriptional activity increased up to 10-fold in astrocytes exposed to IL-1beta, and up to 3-fold in astrocytes subjected to hypoxia followed by reoxygenation. Both IL-1beta- mRNAs and proteins hypoxia-induced NF-kappaB activation were blocked by the proteasome inhibitor, MG-132. MG-132 inhibited IL-1beta-induced up-regulation of IL-1beta and IL-8 mRNA and protein but increased hypoxia-stimulated expression/release of IL-1beta and IL-8. IL-1 receptor antagonist (IL-1Ra) blocked both hypoxic astrocyte-conditioned media-induced NF-kappaB activation and the expression/release of IL-1beta and IL-8. Astrocytes subjected to hypoxia in the presence of IL-1Ra failed to activate NF-kappaB, but expressed elevated levels of IL-1beta and IL-8. The data suggest that hypoxia/reoxygenation-induced up-regulation of IL-1beta and IL-8 in human astrocytes has two components, a NF-kappaB independent up-regulation during hypoxia, followed by amplification through autocrine IL-1beta-induced NF-kappaB activation during reoxygenation.


Assuntos
Astrócitos/fisiologia , Comunicação Autócrina/imunologia , Hipóxia Encefálica/imunologia , NF-kappa B/genética , Transcrição Gênica/imunologia , Astrócitos/citologia , Hipóxia Celular/imunologia , Células Cultivadas , Cisteína Endopeptidases , Inibidores de Cisteína Proteinase/farmacologia , Primers do DNA , Feto/citologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/genética , Interleucina-8/genética , Leupeptinas/farmacologia , Luciferases/genética , Complexos Multienzimáticos/antagonistas & inibidores , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma , Sialoglicoproteínas/farmacologia
12.
J Cereb Blood Flow Metab ; 20(6): 967-78, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10894180

RESUMO

Leukocyte infiltration into the brain contributes to the development of ischemic brain damage and is mediated by endothelial/leukocyte adhesion molecules, cytokines, and chemokines released by ischemic brain cells. In this study, we provide evidence that human astrocytes (FHAs) subjected to in vitro hypoxia produce proinflammatory mediator(s) capable of up-regulating inflammatory genes, including intercellular adhesion molecule-1, interleukin (IL)-1beta, tumor necrosis factor-alpha, IL-8, and monocyte chemotactic protein-1 (MCP-1) in human cerebromicrovascular endothelial cells (HCECs). FHAS were exposed to hypoxia in an anaerobic chamber for 4 hours, followed by reoxygenation for 24 hours. Astrocyte-conditioned media (ACM) collected from normoxic FHAS or FHAS subjected to hypoxia/reoxygenation were applied to HCEC cultures for 4 to 24 hours. Semiquantitative reverse transcription-polymerase chain reaction, immunocytochemistry, and enzyme-linked immunosorbent assay demonstrated up-regulation of intercellular adhesion molecule-1 in HCECs exposed to hypoxic ACM. A pronounced elevation in cytokine IL-1beta and tumor necrosis factor-alpha, and chemokine IL-8 and MCP-1 mRNA, accompanied by increased release of immunoreactive cytokines and chemokines into cell media was observed in HCECs exposed to hypoxic ACM. Hypoxia/reoxygenation induced a transient (4 to 18 hours of reoxygenation) up-regulation of IL-1beta mRNA in FHAS and a two- to threefold increase in IL-1beta levels secreted into ACM. Pretreatment of FHAS with 10 micromol/L dexamethasone inhibited both hypoxia-induced expression/secretion of IL-1beta and the ability of hypoxic ACM to induce inflammatory phenotype in HCECs. The ability of hypoxic ACM to up-regulate inflammatory genes in HCECs was inhibited in the presence of IL-1 receptor antagonist (IL-1Ra) and by pretreating ACM with the blocking anti-IL-1beta antibody. These findings strongly implicate IL-1beta secreted by hypoxic astrocytes in triggering inflammatory activation of HCECs and thereby influencing inflammatory responses at the site of the blood-brain barrier.


Assuntos
Astrócitos/imunologia , Encéfalo/irrigação sanguínea , Endotélio Vascular/imunologia , Interleucina-1/imunologia , Elementos Antissenso (Genética) , Astrócitos/citologia , Astrócitos/enzimologia , Caspase 1/metabolismo , Hipóxia Celular/imunologia , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Meios de Cultivo Condicionados/farmacologia , Dexametasona/farmacologia , Endotélio Vascular/química , Endotélio Vascular/citologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Glucocorticoides/farmacologia , Humanos , Técnicas In Vitro , Molécula 1 de Adesão Intercelular/análise , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/genética , Interleucina-8/genética , Interleucina-8/imunologia , Comunicação Parácrina/imunologia , RNA Mensageiro/análise , Sialoglicoproteínas/farmacologia
13.
Brain Pathol ; 10(1): 113-26, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10668901

RESUMO

Brain inflammation has been implicated in the development of brain edema and secondary brain damage in ischemia and trauma. Adhesion molecules, cytokines and leukocyte chemoattractants released/presented at the site of blood-brain barrier (BBB) play an important role in mobilizing peripheral inflammatory cells into the brain. Cerebral endothelial cells (CEC) are actively engaged in processes of microvascular stasis and leukocyte infiltration by producing a plethora of pro-inflammatory mediators. When challenged by external stimuli including cytokines and hypoxia, CEC have been shown to release/express various products of arachidonic acid cascade with both vasoactive and pro-inflammatory properties, including prostaglandins, leukotrienes, and platelet-activating factor (PAF). These metabolites induce platelet and neutrophil activation and adhesion, changes in local cerebral blood flow and blood rheology, and increases in BBB permeability. Ischemic CEC have also been shown to express and release bioactive inflammatory cytokines and chemokines, including IL-1beta, IL-8 and MCP-1. Many of these mediators and ischemia in vitro and in vivo have been shown to up-regulate the expression of both selectin and Ig-families of adhesion molecules in CEC and to facilitate leukocyte adhesion and transmigration into the brain. Collectively, these studies demonstrate a pivotal role of CEC in initiating and regulating inflammatory responses in cerebral ischemia.


Assuntos
Encéfalo/fisiopatologia , Encefalite/fisiopatologia , Endotélio Vascular/fisiopatologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Mediadores da Inflamação/metabolismo , Animais , Circulação Cerebrovascular/fisiologia , Humanos
14.
Acta Neurochir Suppl ; 76: 29-34, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11450027

RESUMO

Ischemic alterations in the glutathione (GSH) redox system of the blood-brain barrier (BBB) may facilitate oxidative injury and formation of vasogenic brain edema. In this study, both the intra- and extracellular GSH contents of human cerebromicrovascular endothelial cells (HCEC) were reduced by 35% after exposing the cells to 4 h in vitro ischemia and 24 h-recovery. The intracellular/extracellular GSH ratio was not affected, indicating a constant rate of GSH efflux. The activities of the peroxide detoxifying enzymes, glutathione peroxidase and glutathione S-transferase, increased by 35%-50%, whereas the GSH regenerating enzyme, glutathione reductase, remained unchanged in ischemic HCEC. gamma-glutamyl transpeptidase (GGTP), a GSH catabolizing enzyme enriched in brain capillaries, was reduced by 30-50% in ischemic HCEC. The effect of in vitro ischemia on HCEC permeability was assessed by measuring sodium fluorescein clearance across a compartmentalized in vitro BBB model. Sodium fluorescein clearance across HCEC monolayers exposed to leukotriene C4 in the presence of the GGTP inhibitor, acivicin (1 microM), or after in vitro ischemia was increased by 60% and 30%, respectively, suggesting that oxidative stress and loss of GGTP may 'unmask' BBB permeabilizing actions of leukotrienes. These results indicate that oxidative stress and loss of GGTP activity in HCEC contribute to ischemic BBB disruption and vasogenic brain edema.


Assuntos
Barreira Hematoencefálica/fisiologia , Edema Encefálico/fisiopatologia , Hipóxia Celular/fisiologia , Endotélio Vascular/fisiopatologia , Glutationa/metabolismo , Homeostase/fisiologia , Leucotrienos/fisiologia , Permeabilidade Capilar/fisiologia , Células Cultivadas , Humanos , Estresse Oxidativo/fisiologia , Lobo Temporal/fisiopatologia
15.
Acta Neurochir Suppl ; 76: 47-53, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11450070

RESUMO

Brain inflammation has been implicated in the development of brain edema and secondary brain damage in ischemia and trauma. Mechanisms involved in leukocyte infiltration across the blood-brain barrier are still unknown. In this study, we show that human cere-bromicrovascular endothelial cells (HCEC) subjected to a 4 h in vitro ischemia (hypoxia + glucose deprivation) followed by a 4-24 h recovery express elevated levels of ICAM-1, IL-8, and MCP-1 mRNAs (semi-quantitative RT-PCR) and secrete increased amounts of the immunoreactive chemokines IL-8 and MCP-1 (ELISA). The ischemia-induced expression of ICAM-1 in HCEC, and the expression/release of IL-8 and MCP-1 in HCEC were abolished by the non-steroid anti-inflammatory drug, indomethacin (100-300 microM). The immunosuppressant cyclosporin A (50 microM) partially reduced the ischemia-stimulated IL-8 and MCP-1 secretion by HCEC. Both indomethacin and cyclosporin A also inhibited the ischemia-induced neutrophil chemotaxis elicited by HCEC media. The study indicates that in vitro ischemia augments the expression of adhesion molecules and leukocyte chemoattractants at the site of the BBB. This ischemic pro-inflammatory activation of HCEC may constitute a key event in initiating post-ischemic inflammation, and it can be suppressed by the anti-inflammatory drugs, indomethacin and cyclosporin A.


Assuntos
Edema Encefálico/imunologia , Isquemia Encefálica/imunologia , Quimiocinas/antagonistas & inibidores , Ciclosporina/farmacologia , Endotélio Vascular/efeitos dos fármacos , Indometacina/farmacologia , Molécula 1 de Adesão Intercelular/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Quimiocinas/metabolismo , Endotélio Vascular/imunologia , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia
16.
J Neuroimmunol ; 101(2): 148-60, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10580798

RESUMO

Leukocyte infiltration into the brain has been implicated in the development of ischemic brain damage. In this study, simulated in vitro ischemia/reperfusion and IL-1beta were found to up-regulate both the expression of intercellular adhesion molecule- (ICAM-1) in cultured human cerebromicrovascular endothelial cells (HCEC) and the adhesion of allogenic neutrophils to HCEC. Both HCEC and human fetal astrocytes (FHAS) also responded to IL-1beta and to in vitro ischemia/reperfusion by a pronounced up-regulation of IL-8 and MCP-1 mRNA and by increased release of IL-8 and MCP-1 in cell culture media. FHAS were found to release 30-times higher levels of MCP-1 than HCEC under both basal and ischemic conditions. However, 100 u/ml IL-1beta induced greater stimulation of both IL-8 and MCP-1 secretion in HCEC (50 and 20 times above controls, respectively) than in FHAS (three and two times above controls, respectively). IL-8 was the principal neutrophil chemoattractant released from IL-1beta-treated HCEC, since IL-8 antibody completely inhibited neutrophil chemotaxis enticed by HCEC media. However, the IL-8 antibody neutralized only 50% of IL-1beta-stimulated neutrophil chemoattractants released from FHAS, and 40%-60% of ischemia-stimulated chemotactic activity released by either HCEC or FHAS. These results suggest that simulated in vitro ischemia, in addition to IL-8 and MCP-1, stimulates secretion of other bioactive chemokines from HCEC and FHAS.


Assuntos
Astrócitos/metabolismo , Isquemia Encefálica/imunologia , Encéfalo/irrigação sanguínea , Quimiocinas/biossíntese , Endotélio Vascular/metabolismo , Barreira Hematoencefálica , Isquemia Encefálica/metabolismo , Células Cultivadas , Quimiocina CCL2/biossíntese , Quimiotaxia de Leucócito , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Interleucina-1/farmacologia , Interleucina-8/biossíntese , Neutrófilos/imunologia
17.
J Cereb Blood Flow Metab ; 19(11): 1270-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10566974

RESUMO

Calcitonin gene-related peptide (CGRP) and adrenomedullin (ADM) are potent dilators of human brain arteries, and they have been implicated in the neurogenic inflammation underlying migraine headache and in the evolution of stroke, respectively. However, little is known about the presynaptic and postsynaptic distribution of their respective receptors in the human cerebrovascular bed and trigeminovascular system. In the current study, the expression of mRNA for ADM and the two cloned human CGRP1 receptors (identified here as A-CGRP1 receptors [Aiyar et al., 1996] and K-CGRP1 receptors) [Kapas and Clark, 1995] were evaluated in human brain vessels and trigeminal ganglia. Further, the ability of CGRP and ADM to activate adenylate cyclase in cerebromicrovascular and astroglial cell cultures was determined, and the receptors involved were characterized pharmacologically. Isolated human pial vessels, intracortical microvessels, and capillaries, as well as cultures of brain endothelial (EC), smooth muscle (SMC), and astroglial (AST) cells, all expressed mRNA for the two cloned CGRP1 receptors; however, message for the K-CGRP1 receptor was barely detectable in microvascular tissues and cells. In contrast, only isolated capillaries and cultured AST exhibited message for the ADM receptor. In human trigeminal ganglia, mRNA for ADM and the two CGRP1 receptors was systematically present. The CGRP dose-dependently increased (up to 50-fold) cAMP formation in cell cultures, an effect significantly blocked by 0.1 to 10 micromol/L of the CGRP1 receptor antagonist CGRP8-37. The ADM receptor agonist, ADM13-52 (1 micromol/L), similarly increased cAMP production in all cell types, and this response was virtually abolished by 1 micromol/L CGRP8-37. Low concentrations (1 to 10 micromol/L) of the ADM receptor antagonist ADM22-52 blocked the ADM13-52-induced cAMP formation in AST (26% at 10 micromol/L, P < 0.05), whereas they potentiated this response in brain EC and SMC (40% and 100%, P < 0.001, respectively). Even at a higher dose (50 micromol/L), ADM22-52 inhibited the ADM13-52 effect in vascular cells (45%) much less effectively than in AST (95%). These results indicate that both CGRP and ADM can affect human brain vessels through a CGRP1 receptor, and they further suggest the presence of functional ADM receptors in human astroglial cells.


Assuntos
Astrócitos/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Capilares/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Peptídeos , Gânglio Trigeminal/metabolismo , Células Cultivadas , Circulação Cerebrovascular , Humanos , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Receptores de Adrenomedulina
18.
J Cereb Blood Flow Metab ; 19(8): 908-17, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10458598

RESUMO

Physiologic and anatomic evidence suggest that 5-hydroxytryptamine (5-HT) neurons regulate local cerebral blood flow and blood-brain barrier permeability. To evaluate the possibility that some of these effects occur directly on the blood vessels, molecular and/or pharmacologic approaches were used to assess the presence of 5-HT receptors in human brain microvascular fractions, endothelial and smooth muscle cell cultures, as well as in astroglial cells which intimately associate with intraparenchymal blood vessels. Isolated microvessels and capillaries consistently expressed messages for the h5-HT1B, h5-HT1D, 5-HT1F, 5-HT2A but not 5-HT7 receptors. When their distribution within the vessel wall was studied in more detail, it was found that capillary endothelial cells exhibited mRNA for the h5-HT1D and for the 5-HT7 receptors whereas microvascular smooth muscle cells, in addition to h5-HT1D and 5-HT7, also showed polymerase chain reaction products for h5-HT1B receptors. Expression of 5-HT1F and 5-HT2A receptor mRNAs was never detected in any of the microvascular cell cultures. In contrast, messages for all 5-HT receptors tested were detected in human brain astrocytes with a predominance of the 5-HT2A and 5-HT7 subtypes. In all cultures, sumatriptan inhibited (35-58%, P < .05) the forskolin-stimulated production of cyclic AMP, an effect blocked by the 5-HT1B/1D receptor antagonists GR127935 and GR55562. In contrast, 5-carboxamidotryptamine induced strong increases (> or = 400%, P < .005) in basal cyclic AMP levels that were abolished by mesulergine, a nonselective 5-HT7 receptor antagonist. Only astroglial cells showed a ketanserin-sensitive increase (177%, P < .05) in IP3 formation when exposed to 5-HT. These results show that specific populations of functional 5-HT receptors are differentially distributed within the various cellular compartments of the human cortical microvascular bed, and that human brain astroglial cells are endowed with multiple 5-HT receptors. These findings emphasize the complex interactions between brain serotonergic pathways and non-neuronal cells within the CNS and, further, they raise the possibility that some of these receptors may be activated by antimigraine compounds such as brain penetrant triptan derivatives.


Assuntos
Astrócitos/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Circulação Cerebrovascular/fisiologia , Endotélio Vascular/metabolismo , Microcirculação/metabolismo , Músculo Liso Vascular/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , Humanos , Neurônios/metabolismo , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Serotonina/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistemas do Segundo Mensageiro/efeitos dos fármacos
19.
Int J Dev Neurosci ; 17(3): 173-84, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10452361

RESUMO

Glutamate plays an important role in brain development, physiological function, and neurodegeneration. Astrocytes control synaptic concentration of glutamate via the high affinity glutamate transporters, GLT-1 and GLAST, and the glutamate catabolizing enzyme, glutamine synthetase. In this study we show that astrocytes cultured from rat brain in various stages of development including embryonic (E18), postnatal (P1-P21) and mature (P50), show distinct patterns of GLT-1 and GLAST expression, glutamine synthetase activity, and phenotypic changes induced by dibutyryl-cyclic adenosine monophosphate. The transcripts for GLT-1 message were detectable in embryonic astrocytes only, whereas the GLAST message was highly expressed in E18 and P1-P4 astrocyte cultures, declined in P10-P21, and was undetectable in P50 astrocytes. Uptake of 3H-glutamate correlated well with GLAST expression in astrocyte cultures of all developmental stages. Glutamine synthetase activity significantly declined from high embryonic levels in P4 astrocytes and remained low throughout postnatal maturation. Exposure of astrocyte cultures to the differentiating agent, db-cAMP (250-500 microM; 6 days), resulted in a pronounced stellation, up-regulation of GLT-1 and GLAST in E18, and GLAST in P4 cultures, while it was ineffective in P10 astrocytes. By contrast, db-cAMP induced a more pronounced stimulation of glutamine synthetase activity (up to 10-fold above basal) in P10 than in E18 cultures (up to 2 times above basal). The differences in expression/inducibility of glutamate transporters and glutamine synthetase observed in astrocyte cultures derived from various stages of fetal and postnatal development suggest that astrocytes in vivo might also respond differently to environmental or injurious stimuli during development and maturation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Glutamato-Amônia Ligase/metabolismo , Sistema X-AG de Transporte de Aminoácidos , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Biomarcadores , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Bucladesina/farmacologia , Diferenciação Celular/fisiologia , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Desenvolvimento Embrionário e Fetal/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Z Gerontol Geriatr ; 32(1): 33-40, 1999 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-10408023

RESUMO

These studies were performed in an attempt to clarify some of the pathophysiologic mechanisms which occur during and after global ischemia. Both nitric oxide and endothelin were demonstrated in gerbils to participate in responses to ischemia. It was shown that endogenous nitric oxide influences early postischemic reperfusion, systemic blood pressure and postischemic dopamine metabolism. Furthermore, the results indicated that nitric oxide played a role in dopamine release and that preischemic intracerebral nitric oxide formation significantly decreased ischemic dopamine release. In addition, ischemic release of endothelin-1 was detected; participation of nitric oxide in this release was observed. Further indication of functional interactions between nitric oxide and endothelin-1 in postischemic reperfusion were indicated by observations that endothelin-1 antagonists inhibited early hypoperfusion caused by Nitro-L-arginin and late hypoperfusion caused by endogenous endothelin-1. Nitric oxide was shown to decrease edema formation during the early postischemic period but contribute to edema formation during the late postischemic period. The findings indicate the importance of nitric oxide in stroke and ischemia.


Assuntos
Infarto Cerebral/fisiopatologia , Endotelina-1/fisiologia , Óxido Nítrico/fisiologia , Animais , Pressão Sanguínea/fisiologia , Encéfalo/fisiopatologia , Edema Encefálico/fisiopatologia , Circulação Cerebrovascular/fisiologia , Dopamina/metabolismo , Gerbillinae , Traumatismo por Reperfusão/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA