Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Microbiol ; 15: 211, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26467858

RESUMO

BACKGROUND: Neonatal meningitis-causing Escherichia coli (NMEC) is the predominant Gram-negative bacterial pathogen associated with meningitis in newborn infants. High levels of heterogeneity and diversity have been observed in the repertoire of virulence traits and other characteristics among strains of NMEC making it difficult to define the NMEC pathotype. The objective of the present study was to identify genotypic and phenotypic characteristics of NMEC that can be used to distinguish them from commensal E. coli. METHODS: A total of 53 isolates of NMEC obtained from neonates with meningitis and 48 isolates of fecal E. coli obtained from healthy individuals (HFEC) were comparatively evaluated using five phenotypic (serotyping, serum bactericidal assay, biofilm assay, antimicorbial susceptibility testing, and in vitro cell invasion assay) and three genotypic (phylogrouping, virulence genotyping, and pulsed-field gel electrophoresis) methods. RESULTS: A majority (67.92%) of NMEC belonged to B2 phylogenetic group whereas 59% of HFEC belonged to groups A and D. Serotyping revealed that the most common O and H types present in NMEC tested were O1 (15%), O8 (11.3%), O18 (13.2%), and H7 (25.3%). In contrast, none of the HFEC tested belonged to O1 or O18 serogroups. The most common serogroup identified in HFEC was O8 (6.25%). The virulence genotyping reflected that more than 70% of NMEC carried kpsII, K1, neuC, iucC, sitA, and vat genes with only less than 27% of HFEC possessing these genes. All NMEC and 79% of HFEC tested were able to invade human cerebral microvascular endothelial cells. No statistically significant difference was observed in the serum resistance phenotype between NMEC and HFEC. The NMEC strains demonstrated a greater ability to form biofilms in Luria Bertani broth medium than did HFEC (79.2% vs 39.9%). CONCLUSION: The results of our study demonstrated that virulence genotyping and phylogrouping may assist in defining the potential NMEC pathotype.


Assuntos
Infecções por Escherichia coli/microbiologia , Escherichia coli/classificação , Escherichia coli/isolamento & purificação , Genótipo , Meningites Bacterianas/microbiologia , Fenótipo , Biofilmes/crescimento & desenvolvimento , Atividade Bactericida do Sangue , Endocitose , Escherichia coli/genética , Escherichia coli/fisiologia , Humanos , Recém-Nascido , Testes de Sensibilidade Microbiana , Tipagem Molecular , Sorogrupo , Fatores de Virulência/genética
2.
Br J Pharmacol ; 172(22): 5390-402, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26362823

RESUMO

BACKGROUND AND PURPOSE: Stroke and Alzheimer's disease (AD) are related pathologies in which the cerebrovascular system is involved. Plasma levels of semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1, also known as Primary Amine Oxidase -PrAO) are increased in both stroke and AD patients and contribute to the vascular damage. During inflammation, its enzymatic activity mediates leukocyte recruitment to the injured tissue, inducing damage in the blood-brain barrier (BBB) and neuronal tissue. We hypothesized that by altering cerebrovascular function, SSAO/VAP-1 might play a role in the stroke-AD transition. Therefore, we evaluated the protective effect of the novel multitarget-directed ligand DPH-4, initially designed for AD therapy, on the BBB. EXPERIMENTAL APPROACH: A human microvascular brain endothelial cell line expressing human SSAO/VAP-1 was generated, as the expression of SSAO/VAP-1 is lost in cultured cells. To simulate ischaemic damage, these cells were subjected to oxygen and glucose deprivation (OGD) and re-oxygenation conditions. The protective role of DPH-4 was then evaluated in the presence of methylamine, an SSAO substrate, and/or ß-amyloid (Aß). KEY RESULTS: Under our conditions, DPH-4 protected brain endothelial cells from OGD and re-oxygenation-induced damage, and also decreased SSAO-dependent leukocyte adhesion. DPH-4 was also effective at preventing the damage induced by OGD and re-oxygenation in the presence of Aß as a model of AD pathology. CONCLUSIONS AND IMPLICATIONS: From these results, we concluded that the multitarget compound DPH-4 might be of therapeutic benefit to delay the onset and/or progression of the neurological pathologies associated with stroke and AD, which appear to be linked.


Assuntos
Amina Oxidase (contendo Cobre)/metabolismo , Isquemia Encefálica/metabolismo , Moléculas de Adesão Celular/metabolismo , Hidroxiquinolinas/farmacologia , Fármacos Neuroprotetores/farmacologia , Piperidinas/farmacologia , Hipóxia Celular/fisiologia , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Glucose/metabolismo , Humanos , Microvasos/citologia , Oxigênio/metabolismo
3.
Biomed Microdevices ; 15(1): 145-50, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22955726

RESUMO

The blood-brain barrier (BBB) is a unique feature of the human body, preserving brain homeostasis and preventing toxic substances to enter the brain. However, in various neurodegenerative diseases, the function of the BBB is disturbed. Mechanisms of the breakdown of the BBB are incompletely understood and therefore a realistic model of the BBB is essential. We present here the smallest model of the BBB yet, using a microfluidic chip, and the immortalized human brain endothelial cell line hCMEC/D3. Barrier function is modulated both mechanically, by exposure to fluid shear stress, and biochemically, by stimulation with tumor necrosis factor alpha (TNF-α), in one single device. The device has integrated electrodes to analyze barrier tightness by measuring the transendothelial electrical resistance (TEER). We demonstrate that hCMEC/D3 cells could be cultured in the microfluidic device up to 7 days, and that these cultures showed comparable TEER values with the well-established Transwell assay, with an average (± SEM) of 36.9 Ω.cm(2) (± 0.9 Ω.cm(2)) and 28.2 Ω.cm(2) (± 1.3 Ω.cm(2)) respectively. Moreover, hCMEC/D3 cells on chip expressed the tight junction protein Zonula Occludens-1 (ZO-1) at day 4. Furthermore, shear stress positively influenced barrier tightness and increased TEER values with a factor 3, up to 120 Ω.cm(2). Subsequent addition of TNF-α decreased the TEER with a factor of 10, down to 12 Ω.cm(2). This realistic microfluidic platform of the BBB is very well suited to study barrier function in detail and evaluate drug passage to finally gain more insight into the treatment of neurodegenerative diseases.


Assuntos
Barreira Hematoencefálica/metabolismo , Fenômenos Mecânicos , Técnicas Analíticas Microfluídicas/instrumentação , Fenômenos Biomecânicos , Barreira Hematoencefálica/citologia , Linhagem Celular , Impedância Elétrica , Células Endoteliais/metabolismo , Humanos , Microscopia Confocal
4.
Rev Neurol (Paris) ; 166(3): 284-8, 2010 Mar.
Artigo em Francês | MEDLINE | ID: mdl-19699499

RESUMO

Over the last few years, the blood-brain barrier has come to be considered as the main limitation for the treatment of neurological diseases caused by inflammatory, tumor or neurodegenerative disorders. In the blood-brain barrier, the close intercellular contact between cerebral endothelial cells due to tight junctions prevents the passive diffusion of hydrophilic components from the bloodstream into the brain. Several specific transport systems (via transporters expressed on cerebral endothelial cells) are implicated in the delivery of nutriments, ions and vitamins to the brain; other transporters expressed on cerebral endothelial cells extrude endogenous substances or xenobiotics, which have crossed the cerebral endothelium, out of the brain and into the bloodstream. Recently, several strategies have been proposed to target the brain, (i) by by-passing the blood-brain barrier by central drug administration, (ii) by increasing permeability of the blood-brain barrier, (iii) by modulating the expression and/or the activity of efflux transporters, (iv) by using the physiological receptor-dependent blood-brain barrier transport, and (v) by creating new viral or chemical vectors to cross the blood-brain barrier. This review focuses on the illustration of these different approaches.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Preparações Farmacêuticas/metabolismo , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Barreira Hematoencefálica/fisiologia , Sistemas de Liberação de Medicamentos , Humanos , Preparações Farmacêuticas/administração & dosagem
5.
FASEB J ; 24(1): 229-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19770225

RESUMO

Cerebral amyloid angiopathy (CAA) is an age-associated condition and a common finding in Alzheimer's disease in which amyloid-beta (Abeta) vascular deposits are featured in >80% of the cases. Familial Abeta variants bearing substitutions at positions 21-23 are primarily associated with CAA, although they manifest with strikingly different clinical phenotypes: cerebral hemorrhage or dementia. The recently reported Piedmont L34V Abeta mutant, located outside the hot spot 21-23, shows a similar hemorrhagic phenotype, albeit less aggressive than the widely studied Dutch E22Q variant. We monitored the apoptotic events occurring after stimulation of human brain microvascular endothelial and smooth muscle cells with nonfibrillar structures of both variants and wild-type Abeta40. Induction of analogous caspase-mediated mitochondrial pathways was elicited by all peptides, although within different time frames and intensity. Activated pathways were susceptible to pharmacological modulation either through direct inhibition of mitochondrial cytochrome c release or by the action of pan- and pathway-specific caspase inhibitors, giving a clear indication of the independent or synergistic engagement of both extrinsic and intrinsic mechanisms. Structural analyses of the Abeta peptides showed that apoptosis preceded fibril formation, correlating with the presence of oligomers and/or protofibrils. The data support the notion that rare genetic mutations constitute unique paradigms to understand the molecular pathogenesis of CAA.


Assuntos
Peptídeos beta-Amiloides/genética , Encéfalo/irrigação sanguínea , Angiopatia Amiloide Cerebral Familiar/genética , Angiopatia Amiloide Cerebral Familiar/patologia , Substituição de Aminoácidos , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Apoptose , Encéfalo/metabolismo , Encéfalo/patologia , Caspases/metabolismo , Linhagem Celular , Angiopatia Amiloide Cerebral Familiar/metabolismo , Citocromos c/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Variação Genética , Humanos , Mitocôndrias/metabolismo , Mutação , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
Cytogenet Genome Res ; 126(4): 313-7, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19864871

RESUMO

The immortalized human cerebral microvessel endothelial cell line hCMEC/D3 has been repeatedly used as a model of human blood-brain barrier (BBB). hCMEC/D3 cells between passage 25 and 35 are most often applied in research, remained phenotypically nontransformed, and cells maintained many characteristics of human brain endothelial cells. Also hCMEC/D3 was thought to have conserved a normal diploid karyotype over all these passages. Here we characterized the cell line using high-resolution multicolor fluorescence in situ hybridization (FISH) approaches and revealed a complex karyotype in the 30th passage. Clonal cryptic unbalanced structural rearrangements and numerical aberrations were discovered and described as follows: 45 approximately 48,XX, -X,del(5)(q11)[2],del(9)(q11)[3],+9[3],del(11)(q13 approximately 14)[2], der(14)t(14;21)(q32.33;q22.3)[28],der(15)t(9;15)(p11;p11)[13], dup(15)(p11q11)[5],der(21)t(17;21)(p12;q22)[9],-22[6][cp28]. In summary, a complex karyotype with clonal unbalanced chromosomal rearrangements is present in hCMEC/D3. Thus, we solicit to include molecular cytogenetics in the testing of all cell lines prior to application of their use in complex studies.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/citologia , Células Endoteliais/citologia , Microvasos/citologia , Linhagem Celular , Aberrações Cromossômicas , Humanos , Cariotipagem
7.
Rev Neurol (Paris) ; 165(12): 1010-22, 2009 Dec.
Artigo em Francês | MEDLINE | ID: mdl-19487004

RESUMO

The main characteristic of the blood-brain-barrier (BBB) is its extremely low permeability, due to tight intercellular endothelial junctions and a variety of transporters, which provides the brain with a unique protection against the potential toxicity of several xenobiotics, but also constitutes a major limitation to drug delivery to the central nervous system. Several dysfunctions of the BBB have been recently implicated in the pathophysiology of neurological diseases: inflammatory, vascular, tumoral, infectious and neurodegenerative diseases. Based on a better knowledge of the BBB biology, new therapeutic strategies are emerging, which by-pass the BBB or take advantage of the selective expression of membrane proteins by brain endothelial cells or circulating leucocytes to target new drugs, such as the anti-VLA4 antibody recently approved for multiple sclerosis treatment. This review will focus on the recently described BBB dysfunctions presumably involved in various neurological diseases.


Assuntos
Barreira Hematoencefálica/fisiologia , Doenças do Sistema Nervoso/fisiopatologia , Isquemia Encefálica/fisiopatologia , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/fisiopatologia , Permeabilidade da Membrana Celular , Difusão , Endotélio Vascular/fisiopatologia , Epilepsia/sangue , Epilepsia/fisiopatologia , Humanos , Esclerose Múltipla/sangue , Esclerose Múltipla/fisiopatologia , Degeneração Neural/sangue , Degeneração Neural/fisiopatologia , Doenças do Sistema Nervoso/sangue , Acidente Vascular Cerebral/fisiopatologia
8.
Rev Neurol (Paris) ; 165(11): 863-74, 2009 Nov.
Artigo em Francês | MEDLINE | ID: mdl-19427009

RESUMO

The blood-brain barrier provides the central nervous system with a unique protection against the toxic effects of many xenobiotics. This protection results from the unique anatomic and biological structure of the endothelium of blood vessels in the brain. The main features of the blood-brain barrier are the presence of tight intercellular junctions which strictly limit the diffusion of blood-borne solutes and cells into the brain and the polarized expression of transporters which specifically control the cerebral availability of nutrients, drugs or xenobiotics. Recent findings in molecular and cellular biology improved our knowledge of blood-brain barrier permeability and its regulation. The importance of these findings has been recently highlighted by the description of dysfunctions of the blood-brain barrier which could have an impact on the pathophysiology of several neurological diseases. This review focuses on recent advances in our understanding of blood-brain barrier biology and physiology, presenting the structural organization of the blood-brain barrier and the functional regulation of solute permeability and cellular transendothelial migration.


Assuntos
Barreira Hematoencefálica/fisiologia , Astrócitos/fisiologia , Transporte Biológico , Capilares/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/fisiologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Endotélio Vascular/fisiologia , Metabolismo Energético , Humanos , Neurônios/fisiologia , Xenobióticos/toxicidade
9.
Arthritis Rheum ; 58(11): 3550-61, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18975312

RESUMO

OBJECTIVE: To assess angiogenesis and explore the expression and regulation of vascular endothelial growth factor (VEGF), VEGF receptor 1 (VEGFR-1), and VEGFR-2, the leading mediators of angiogenesis, in SSc patients and controls. METHODS: Late-outgrowth endothelial progenitor cells (EPCs), isolated from the peripheral blood of systemic sclerosis (SSc) patients and controls, and human umbilical vein endothelial cells (HUVECs) were assessed under normal and hypoxic conditions. Genomic background was evaluated in a large case-control study (including 659 patients with SSc and 511 controls) using tag single-nucleotide polymorphisms on VEGFR1 and VEGFR2 genes. RESULTS: EPCs from SSc patients had the phenotype of genuine endothelial cells and displayed in vitro angiogenic properties similar to those of HUVECs and control EPCs under basal conditions, as determined by flow cytometry, tube formation, and migration assay. However, after 6 hours of hypoxic exposure, EPCs from SSc patients exhibited lower induced expression of VEGFR-1 at the messenger RNA and protein levels, but similar VEGF and VEGFR-2 expression, compared with HUVECs or EPCs from healthy controls. There was no evidence of defective expression of hypoxia-inducible factor 1alpha. These results were supported by the lower serum levels of soluble VEGFR-1 found in SSc patients (n = 187) compared with healthy controls (n = 48) (mean +/- SD 163.7 +/- 98.5 versus 210.4 +/- 109.5 pg/ml; P = 0.0042). These abnormalities did not seem to be related to genomic background. CONCLUSION: Our findings shed new light on the possible role of VEGFR-1 in the main vascular disturbances that occur in SSc and lead to more severe disease.


Assuntos
Hipóxia Celular/fisiologia , Endotélio Vascular/citologia , Neovascularização Patológica/fisiopatologia , Escleroderma Sistêmico/fisiopatologia , Células-Tronco/química , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Fator A de Crescimento do Endotélio Vascular/análise , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/análise , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
10.
Ann Rheum Dis ; 67(10): 1455-60, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18174219

RESUMO

BACKGROUND: Heterogeneous data have been reported regarding the detection and number of circulating endothelial progenitor cells (EPCs) in systemic sclerosis (SSc). OBJECTIVE: We investigated the number of circulating EPCs using recent recommendations and we quantified their late outgrowth in patients with SSc and healthy controls. PATIENTS AND METHODS: EPCs, defined as Lin-/7AAD-/CD34+/CD133+/VEGFR-2+ cells, were quantified in 50 patients with SSc (mean age: 55 (16) years, disease duration: 9 (9) years) and 26 controls (mean age: 53 (19) years) by cell sorting/flow cytometry and by counting late outgrowth colony-forming units (CFU). RESULTS: Patients with SSc displayed higher circulating EPC counts than controls (median 86 (5-282) vs 49 (5-275)) EPCs for 1 million Lin- mononuclear cells; p = 0.01). Lower EPC counts were associated with the higher Medsger's severity score (p = 0.01) and with the presence of past and/or current digital ulcers (p = 0.026). There was no difference for the number of late outgrowth EPC-CFUs between patients with SSc and controls in cell culture evaluation. The formation of colonies was associated with higher levels of circulating EPCs (p = 0.02) and the number of colonies correlated with levels of EPCs (R = 0.73, p = 0.0004), validating our combination of fluorescence-activated cell sorter surface markers. CONCLUSIONS: We quantified circulating EPCs with an accurate combination of markers herein validated. Our data demonstrate increased circulating EPC levels in SSc, supporting their mobilisation from bone marrow. Furthermore, the subset of patients with digital vascular lesions and high severity score displayed low EPC counts, suggesting increased homing at this stage. The predictive value of this biomarker now warrants further evaluation.


Assuntos
Endotélio Vascular/patologia , Escleroderma Sistêmico/sangue , Células-Tronco/patologia , Adulto , Idoso , Separação Celular/métodos , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo/métodos , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Índice de Gravidade de Doença
11.
Gene ; 380(2): 127-36, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16887298

RESUMO

The Mitochondrial Tumor suppressor 1 (MTUS1) gene is a newly identified candidate tumor suppressor gene at chromosomal position 8p22. We report here that MTUS1 encodes a family of proteins whose leader member (ATIP1) was previously isolated in our laboratory as a novel interacting partner of the angiotensin II AT2 receptor involved in growth inhibition (Nouet, JBC 279: 28989-97, 2004). The MTUS1 gene contains 17 coding exons distributed over 112 kb of genomic DNA. Alternative exon usage generates three major transcripts (ATIP1, ATIP3 and ATIP4), each showing different tissue distribution. ATIP polypeptides are identical in their carboxy-terminal region carrying four coiled-coil domains. In their amino-terminal portion, ATIP polypeptides exhibit distinct motifs for localisation in the cytosol, nucleus or cell membrane, suggesting that MTUS1 gene products may be involved in a variety of intracellular functions in an AT2-dependent and independent manner. ATIP1 is ubiquitous and highly expressed in the brain. ATIP3 is the major transcript in tissues (prostate, bladder, breast, ovary, colon) corresponding to cancer types with frequent loss of heterozygosity at 8p22. Interestingly, ATIP4 is a brain-specific transcript highly abundant in the cerebellum and fetal brain. High evolutionary conservation of ATIP amino-acid sequences suggests important biological roles for this new family of proteins in tumor suppression and/or brain function.


Assuntos
Genes Supressores de Tumor , Receptor Tipo 2 de Angiotensina/metabolismo , Proteínas Supressoras de Tumor/genética , Processamento Alternativo , Sequência de Bases , Northern Blotting/métodos , Sistema Nervoso Central/metabolismo , Mapeamento Cromossômico , Cromossomos Humanos Par 8 , Evolução Molecular , Éxons , Feminino , Expressão Gênica , Variação Genética , Humanos , Íntrons , Masculino , Família Multigênica/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Análise de Sequência , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
12.
Mol Cell Endocrinol ; 252(1-2): 207-15, 2006 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-16650523

RESUMO

A high frequency of allelic loss affecting chromosome 8p and a minimal region of deletion at p21-22 have been previously reported in hepatocellular carcinoma (HCC), suggesting that at least one tumor suppressor gene is present in this region. In this study, we assessed whether the angiotensin II AT2 receptor interacting protein (ATIP)/mitochondrial tumor suppressor gene (MTUS1), a gene newly identified at position 8p22, may be a candidate tumor suppressor gene mutated in HCC. We searched for alterations in the 17 coding exons of ATIP/MTUS1 by means of denaturating high-performance liquid chromatography and sequencing, in 51 HCC tumors and 58 cell lines for which loss of heterozygosity status was known. Five major nucleotide substitutions were identified, all located in exons used by the ATIP3 transcript which is the only ATIP transcript variant expressed in liver. These nucleotide variations result in amino-acid substitution or deletion of conserved structural motifs (nuclear localisation signal, polyproline motif, leucine zipper) and also affect exonic splicing enhancer motifs and physiological splice sites, suggesting potential deleterious effects on ATIP3 function and/or expression.


Assuntos
Carcinoma Hepatocelular/genética , Cromossomos Humanos Par 8 , Genes Supressores de Tumor , Neoplasias Hepáticas/genética , Proteínas Supressoras de Tumor/genética , Substituição de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Mapeamento Cromossômico , Análise Mutacional de DNA , DNA de Neoplasias/genética , Variação Genética , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Polimorfismo Genético , Splicing de RNA
13.
FASEB J ; 19(13): 1872-4, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16141364

RESUMO

Establishment of a human model of the blood-brain barrier has proven to be a difficult goal. To accomplish this, normal human brain endothelial cells were transduced by lentiviral vectors incorporating human telomerase or SV40 T antigen. Among the many stable immortalized clones obtained by sequential limiting dilution cloning of the transduced cells, one was selected for expression of normal endothelial markers, including CD31, VE cadherin, and von Willebrand factor. This cell line, termed hCMEC/D3, showed a stable normal karyotype, maintained contact-inhibited monolayers in tissue culture, exhibited robust proliferation in response to endothelial growth factors, and formed capillary tubes in matrix but no colonies in soft agar. hCMEC/D3 cells expressed telomerase and grew indefinitely without phenotypic dedifferentiation. These cells expressed chemokine receptors, up-regulated adhesion molecules in response to inflammatory cytokines, and demonstrated blood-brain barrier characteristics, including tight junctional proteins and the capacity to actively exclude drugs. hCMEC/D3 are excellent candidates for studies of blood-brain barrier function, the responses of brain endothelium to inflammatory and infectious stimuli, and the interaction of brain endothelium with lymphocytes or tumor cells. Thus, hCMEC/D3 represents the first stable, fully characterized, well-differentiated human brain endothelial cell line and should serve as a widely usable research tool.


Assuntos
Barreira Hematoencefálica , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Resistência a Múltiplos Medicamentos , Células Endoteliais/citologia , Ágar/química , Animais , Antígenos CD , Antígenos Transformantes de Poliomavirus/biossíntese , Antígenos Transformantes de Poliomavirus/genética , Barreira Hematoencefálica/efeitos dos fármacos , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Caderinas/biossíntese , Capilares/patologia , Bovinos , Adesão Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Clonagem Molecular , Colágeno/farmacologia , Citocinas/metabolismo , Combinação de Medicamentos , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Cariotipagem , Laminina/farmacologia , Lentivirus/genética , Linfócitos/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Perfusão , Permeabilidade , Fenótipo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Proteoglicanas/farmacologia , RNA/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Telomerase/genética , Telomerase/metabolismo , Fatores de Tempo , Regulação para Cima , Fator de von Willebrand/biossíntese
14.
Brain Res Mol Brain Res ; 137(1-2): 77-88, 2005 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-15950764

RESUMO

Endothelin-1 (ET-1), a vasoactive and mitogenic peptide mainly produced by vascular endothelial cells, may be involved in the progression of several human tumors. Here, we present an immunohistochemical analysis of the expression pattern of ET-1 receptor subtypes (ET(A)-R and ET(B)-R) and a functional study of their potential role in human oligodendrogliomas and oligoastrocytomas. By comparison, we assessed the corresponding expression patterns of glioblastomas. Interestingly, a nuclear localization of ET-1 receptor subtypes (associated or not with a cytoplasmic labeling) was constantly observed in tumor cells from all three glioma types. Moreover, we noted a distinct receptor distribution in the different gliomas: a nuclear expression of ET(B)-R by tumor cells was found to be restricted to oligodendrogliomas and oligoastrocytomas, while a nuclear expression of ET(A)-R was only detected in tumor cells from some glioblastomas. Using primary cultures of oligodendroglial tumor cells, we confirmed the selective expression of nuclear ET(B)-R, together with a plasma membrane expression, and further demonstrated that this receptor was functionally coupled to intracellular signaling pathways known to be involved in cell survival and/or proliferation: extracellular signal-regulated kinase and focal adhesion kinase activation, actin cytoskeleton reorganization. In addition, impairment of ET(B)-R activation in these cells by in vitro treatment with an ET(B)-R-specific antagonist induced cell death. These data point to ET-1 as a possible survival factor for oligodendrogliomas via ET(B)-R activation and suggest that ET(B)-R-specific antagonists might constitute a potential therapeutic alternative for oligodendrogliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Endotelina-1/metabolismo , Oligodendroglioma/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Citoesqueleto de Actina/metabolismo , Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico , Astrocitoma/tratamento farmacológico , Astrocitoma/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citoplasma/metabolismo , Antagonistas do Receptor de Endotelina B , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Imuno-Histoquímica , Oligodendroglioma/tratamento farmacológico , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Proteínas Tirosina Quinases/metabolismo , Células Tumorais Cultivadas
15.
J Neurochem ; 93(2): 279-89, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15816851

RESUMO

One of the main difficulties with primary rat brain endothelial cell (RBEC) cultures is obtaining pure cultures. The variation in purity limits the achievement of in vitro models of the rat blood-brain barrier. As P-glycoprotein expression is known to be much higher in RBECs than in any contaminating cells, we have tested the effect of five P-glycoprotein substrates (vincristine, vinblastine, colchicine, puromycin and doxorubicin) on RBEC cultures, assuming that RBECs would resist the treatment with these toxic compounds whereas contaminating cells would not. Treatment with either 4 microg/mL puromycin for the first 2 days of culture or 3 microg/mL puromycin for the first 3 days showed the best results without causing toxicity to the cells. Transendothelial electrical resistance was significantly increased in cell monolayers treated with puromycin compared with untreated cell monolayers. When cocultured with astrocytes in the presence of cAMP, the puromycin-treated RBEC monolayer showed a highly reduced permeability to sodium fluorescein (down to 0.75 x 10(-6) cm/s) and a high electrical resistance (up to 500 Omega x cm(2)). In conclusion, this method of RBEC purification will allow the production of in vitro models of the rat blood-brain barrier for cellular and molecular biology studies as well as pharmacological investigations.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Puromicina/farmacologia , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Técnicas de Cocultura/métodos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/fisiologia , Puromicina/metabolismo , Ratos , Ratos Wistar
16.
J Neurochem ; 88(1): 23-31, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14675146

RESUMO

The blood-brain barrier (BBB) plays an important role in controlling the passage of molecules from blood to brain extracellular fluid. The multidrug efflux pump P-glycoprotein (P-gp) is highly expressed in the luminal membrane of brain endothelium and contributes to the formation of a functional barrier to lipid-soluble drugs such as anticancer agents. The mdr1a P-gp-encoding gene is exclusively expressed in the rodent BBB. Primary cultures of rat brain endothelial cells and GP8.3 cells showed a dramatic decrease in mdr1a mRNA level and some expression of mdr1b mRNA. GPNT cells, derived from GP8.3 cells after transfection with a puromycin resistance gene, were chronically treated with 5 microg/mL puromycin, a P-gp substrate. Compared with rat brain endothelial cells and GP8.3 cells, GPNT cells exhibited a very high level of expression of mdr1a mRNA together with a moderate level of mdr1b mRNA expression. Accordingly, P-gp expression and activity were strongly increased. When GP8.3 and puromycin-starved GPNT cells were treated with puromycin, mdr1a expression was selectively increased. High expression of mdr1a mRNA in GPNT cells may thus be related to the chronic treatment with puromycin. We conclude that GPNT cells may be used as a valuable rat in vitro model for studying the regulation of mdr1a expression at the BBB level.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/genética , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Puromicina/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Capilares/citologia , Capilares/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Expressão Gênica/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Vincristina/farmacocinética , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
17.
Pharmacogenomics J ; 3(4): 215-26, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12931135

RESUMO

The clinical efficacy of oral hydroxyurea (HU) in adults and children with sickle cell anemia (SCA) cannot solely be explained by its ability to enhance fetal hemoglobin (HbF) expression. Since increased adherence of sickle red blood cells to vascular endothelium is a possible contributing factor to vaso-occlusive crisis (VOC), we explored the effect of HU on human endothelial cell (EC) lines (TrHBMEC and EA-hy 926). We demonstrated that HU, in a dose-dependent and reversible manner, significantly decreased (up to three-fold) the release of endothelin-1 (ET-1), a vasoconstrictor peptide through downregulation (up to three-fold) of ET-1 gene expression. This finding is of therapeutic relevance as SCA patients exhibit elevated serum levels of ET-1 during episodes of VOC and levels correlate with disease severity. Unexpectedly, HU upregulated (up to three-fold) the expression of membrane-bound intercellular cell adhesion molecule 1 (mbICAM-1) and its soluble form (sICAM-1) with a parallel increase in ICAM-1 mRNA expression. Although ICAM-1 does not appear to be involved in the sickle cell adhesion to vascular endothelium, it may exacerbate vaso-occlusion by promoting leukocyte adhesion. The HU-induced increase in mbICAM-1 may appear inconsistent with the clinical benefits confered by HU. However, both the increase in sICAM-1- and HU-induced leukocyte reduction in patients, may counteract the potentially detrimental effect of elevated mbICAM-1 expression. Also HU reduces the expression of vascular cell adhesion molecule (VCAM-1) on EC. Since HU reduces the very late antigen 4-positive reticulocytes in SCA patients, a ligand for VCAM-1, HU-induced downregulation of VCAM-1 on EC will very likely decrease the reticulocyte-endothelium adhesion. Thus, HU, apart from inducing HbF expression in the red cell, also affects the expression profile of EC compartment.


Assuntos
Antidrepanocíticos/farmacologia , Células Endoteliais/efeitos dos fármacos , Endotelina-1/genética , Expressão Gênica/efeitos dos fármacos , Hidroxiureia/farmacologia , Molécula 1 de Adesão Intercelular/genética , Anemia Falciforme/tratamento farmacológico , Linhagem Celular , Relação Dose-Resposta a Droga , Regulação para Baixo , Células Endoteliais/metabolismo , Endotelina-1/biossíntese , Citometria de Fluxo , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
18.
J Cell Biol ; 155(1): 133-43, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11581290

RESUMO

ErbB2 is a receptor tyrosine kinase belonging to the family of epidermal growth factor (EGF) receptors which is generally involved in cell differentiation, proliferation, and tumor growth, and activated by heterodimerization with the other members of the family. We show here that type IV pilus-mediated adhesion of Neisseria meningitidis onto endothelial cells induces tyrosyl phosphorylation and massive recruitment of ErbB2 underneath the bacterial colonies. However, neither the phosphorylation status nor the cellular localization of the EGF receptors, ErbB3 or ErbB4, were affected in infected cells. ErbB2 phosphorylation induced by N. meningitidis provides docking sites for the kinase src and leads to its subsequent activation. Specific inhibition of either ErbB2 and/or src activity reduces bacterial internalization into endothelial cells without affecting bacteria-induced actin cytoskeleton reorganization or ErbB2 recruitment. Moreover, inhibition of both actin polymerization and the ErbB2/src pathway totally prevents bacterial entry. Altogether, our results provide new insight into ErbB2 function by bringing evidence of a bacteria-induced ErbB2 clustering leading to src kinase phosphorylation and activation. This pathway, in cooperation with the bacteria-induced reorganization of the actin cytoskeleton, is required for the efficient internalization of N. meningitidis into endothelial cells, an essential process enabling this pathogen to cross host cell barriers.


Assuntos
Aderência Bacteriana , Endotélio/microbiologia , Neisseria meningitidis/fisiologia , Receptor ErbB-2/metabolismo , Actinas/metabolismo , Linhagem Celular , Cortactina , Proteínas do Citoesqueleto , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fímbrias Bacterianas/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Microscopia de Fluorescência , Neisseria meningitidis/genética , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Quinazolinas , Transdução de Sinais/fisiologia , Tirfostinas/farmacologia , Quinases da Família src/metabolismo
19.
Proc Natl Acad Sci U S A ; 98(17): 9942-7, 2001 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-11504951

RESUMO

Royal College of Surgeons rats are genetically predisposed to undergo significant visual loss caused by a primary dysfunction of retinal pigment epithelial (RPE) cells. By using this model, we have examined the efficacy of subretinal transplantation of two independent human RPE cell lines each exhibiting genetic modifications that confer long-term stability in vitro. The two cell lines, a spontaneously derived cell line (ARPE19) and an extensively characterized genetically engineered human RPE cell line (h1RPE7), which expresses SV40 large T (tumor) antigen, were evaluated separately. Both lines result in a significant preservation of visual function as assessed by either behavioral or physiological techniques. This attenuation of visual loss correlates with photoreceptor survival and the presence of donor cells in the areas of rescued photoreceptors at 5 months postgrafting (6 months of age). These results demonstrate the potential of genetically modified human RPE cells for ultimate application in therapeutic transplantation strategies for retinal degenerative diseases caused by RPE dysfunction.


Assuntos
Proteínas do Olho/fisiologia , Epitélio Pigmentado Ocular/transplante , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases/deficiência , Degeneração Retiniana/terapia , Animais , Antígenos Transformantes de Poliomavirus/genética , Linhagem Celular Transformada/transplante , Sobrevivência Celular , Transformação Celular Viral , Proteínas do Olho/genética , Movimentos da Cabeça/fisiologia , Humanos , Fagocitose , Epitélio Pigmentado Ocular/citologia , Ratos , Ratos Mutantes , Receptores Proteína Tirosina Quinases/genética , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Segmento Externo da Célula Bastonete/metabolismo , Segmento Externo da Célula Bastonete/patologia , Limiar Sensorial , Vírus 40 dos Símios/genética , Colículos Superiores/fisiopatologia , Transplante Heterólogo , Testes Visuais , Campos Visuais , Percepção Visual , c-Mer Tirosina Quinase
20.
J Immunol ; 165(6): 3375-83, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10975856

RESUMO

Endothelium of the cerebral blood vessels, which constitutes the blood-brain barrier, controls adhesion and trafficking of leukocytes into the brain. Investigating signaling pathways triggered by the engagement of adhesion molecules expressed on brain endothelial cells using two rat brain endothelial cell lines (RBE4 and GP8), we report in this paper that ICAM-1 cross-linking induces a sustained tyrosine phosphorylation of the phosphatidylinositol-phospholipase C (PLC)gamma1, with a concomitant increase in both inositol phosphate production and intracellular calcium concentration. Our results suggest that PLC are responsible, via a calcium- and protein kinase C (PKC)-dependent pathway, for p60Src activation and tyrosine phosphorylation of the p60Src substrate, cortactin. PKCs are also required for tyrosine phosphorylation of the cytoskeleton-associated proteins, focal adhesion kinase and paxillin, but not for ICAM-1-coupled p130Cas phosphorylation. PKC's activation is also necessary for stress fiber formation induced by ICAM-1 cross-linking. Finally, cell pretreatment with intracellular calcium chelator or PKC inhibitors significantly diminishes transmonolayer migration of activated T lymphocytes, without affecting their adhesion to brain endothelial cells. In summary, our data demonstrate that ICAM-1 cross-linking induces calcium signaling which, via PKCs, mediates phosphorylation of actin-associated proteins and cytoskeletal rearrangement in brain endothelial cell lines. Our results also indicate that these calcium-mediated intracellular events are essential for lymphocyte migration through the blood-brain barrier.


Assuntos
Encéfalo/fisiologia , Sinalização do Cálcio/fisiologia , Citoesqueleto/metabolismo , Endotélio Linfático/fisiologia , Molécula 1 de Adesão Intercelular/metabolismo , Líquido Intracelular/fisiologia , Linfócitos/fisiologia , Actinas/metabolismo , Animais , Cálcio/metabolismo , Cálcio/fisiologia , Linhagem Celular Transformada , Movimento Celular/fisiologia , Cortactina , Citoesqueleto/fisiologia , Endotélio Linfático/citologia , Ativação Enzimática , Fosfatos de Inositol/biossíntese , Molécula 1 de Adesão Intercelular/fisiologia , Proteínas dos Microfilamentos/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Fosforilação , Proteína Quinase C/fisiologia , Ratos , Fosfolipases Tipo C/metabolismo , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...