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










Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 157(12): 4579-4587, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27779914

RESUMO

17ß-estradiol is a potent sex hormone synthesized primarily by gonads in females and males that regulates development and function of the reproductive system. Recent studies show that 17ß-estradiol is locally synthesized in nonreproductive tissues and regulates a myriad of events, including local inflammatory responses. In this study, we report that mesenteric lymph nodes (mLNs) and Peyer's patches (Pps) are novel sites of de novo synthesis of 17ß-estradiol. These secondary lymphoid organs are located within or close to the gastrointestinal tract, contain leukocytes, and function at the forefront of immune surveillance. 17ß-estradiol synthesis was initially identified using a transgenic mouse with red fluorescent protein coexpressed in cells that express aromatase, the enzyme responsible for 17ß-estradiol synthesis. Subsequent immunohistochemistry and tissue culture experiments revealed that aromatase expression was localized to high endothelial venules of these lymphoid organs, and these high endothelial venule cells synthesized 17ß-estradiol when isolated and cultured in vitro. Both mLNs and Pps contained 17ß-estradiol with concentrations that were significantly higher than those of peripheral blood. Furthermore, the total amount of 17ß-estradiol in these organs exceeded that of the gonads. Mice lacking either aromatase or estrogen receptor-ß had hypertrophic Pps and mLNs with more leukocytes than their wild-type littermates, demonstrating a role for 17ß-estradiol in leukocyte regulation. Importantly, we did not observe any sex-dependent differences in aromatase expression, 17ß-estradiol content, or steroidogenic capacity in these lymphoid organs.


Assuntos
Aromatase/metabolismo , Estradiol/biossíntese , Leucócitos/metabolismo , Linfonodos/metabolismo , Nódulos Linfáticos Agregados/metabolismo , Animais , Aromatase/genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Gônadas/metabolismo , Imuno-Histoquímica , Masculino , Mesentério/metabolismo , Camundongos , Camundongos Knockout , Baço/metabolismo
2.
PLoS Pathog ; 9(4): e1003247, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23592982

RESUMO

Cryptococcus neoformans is the most common cause of fungal meningitis, with high mortality and morbidity. The reason for the frequent occurrence of Cryptococcus infection in the central nervous system (CNS) is poorly understood. The facts that human and animal brains contain abundant inositol and that Cryptococcus has a sophisticated system for the acquisition of inositol from the environment suggests that host inositol utilization may contribute to the development of cryptococcal meningitis. In this study, we found that inositol plays an important role in Cryptococcus traversal across the blood-brain barrier (BBB) both in an in vitro human BBB model and in in vivo animal models. The capacity of inositol to stimulate BBB crossing was dependent upon fungal inositol transporters, indicated by a 70% reduction in transmigration efficiency in mutant strains lacking two major inositol transporters, Itr1a and Itr3c. Upregulation of genes involved in the inositol catabolic pathway was evident in a microarray analysis following inositol treatment. In addition, inositol increased the production of hyaluronic acid in Cryptococcus cells, which is a ligand known to binding host CD44 receptor for their invasion. These studies suggest an inositol-dependent Cryptococcus traversal of the BBB, and support our hypothesis that utilization of host-derived inositol by Cryptococcus contributes to CNS infection.


Assuntos
Barreira Hematoencefálica/microbiologia , Encéfalo/metabolismo , Encéfalo/microbiologia , Criptococose/microbiologia , Cryptococcus neoformans/patogenicidade , Inositol/metabolismo , Meningite Criptocócica/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Infecções do Sistema Nervoso Central/metabolismo , Infecções do Sistema Nervoso Central/microbiologia , Criptococose/metabolismo , Cryptococcus neoformans/metabolismo , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/biossíntese , Ácido Hialurônico/metabolismo , Masculino , Meningite Criptocócica/microbiologia , Camundongos , Camundongos Endogâmicos A , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Coelhos , Migração Transendotelial e Transepitelial
3.
Blood ; 115(23): 4951-62, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20354174

RESUMO

Cocaine abuse hastens the neurodegeneration often associated with advanced HIV-1 infection. The mechanisms, in part, revolve around the neuroinflammatory processes mediated by the chemokine monocyte chemotactic protein-1 (MCP-1/CCL2). Understanding factors that modulate MCP-1 and, in turn, facilitate monocyte extravasation in the brain is thus of paramount importance. We now demonstrate that cocaine induces MCP-1 in rodent microglia through translocation of the sigma receptor to the lipid raft microdomains of the plasma membrane. Sequential activation of Src, mitogen-activated protein kinases (MAPKs), and phosphatidylinositol-3' kinase (PI3K)/Akt and nuclear factor kappaB (NF-kappaB) pathways resulted in increased MCP-1 expression. Furthermore, conditioned media from cocaine-exposed microglia increased monocyte transmigration, and thus was blocked by antagonists for CCR2 or sigma receptor. These findings were corroborated by demonstrating increased monocyte transmigration in mice exposed to cocaine, which was attenuated by pretreatment of mice with the sigma receptor antagonist. Interestingly, cocaine-mediated transmigratory effects were not observed in CCR2 knockout mice. We conclude that cocaine-mediated induction of MCP-1 accelerates monocyte extravasation across the endothelium. Understanding the regulation of MCP-1 expression and functional changes by cocaine/sigma receptor system may provide insights into the development of potential therapeutic targets for HIV-1-associated neurocognitive disorders.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Infecções por HIV/metabolismo , HIV-1 , Monócitos/metabolismo , Doenças Neurodegenerativas/metabolismo , Receptores sigma/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Movimento Celular/genética , Quimiocina CCL2/genética , Cocaína/efeitos adversos , Transtornos Relacionados ao Uso de Cocaína/genética , Transtornos Relacionados ao Uso de Cocaína/patologia , Inibidores da Captação de Dopamina/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Infecções por HIV/genética , Infecções por HIV/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Camundongos , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Monócitos/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Receptores sigma/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
4.
J Neurosci ; 29(3): 630-7, 2009 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19158290

RESUMO

Progressive axonal degeneration follows demyelination in many neurological diseases, including multiple sclerosis and inherited demyelinating neuropathies, such as Charcot-Marie-Tooth disease. One glial molecule, the myelin-associated glycoprotein (MAG), located in the adaxonal plasmalemma of myelin-producing cells, is known to signal to the axon and to modulate axonal caliber through phosphorylation of axonal neurofilament proteins. This report establishes for the first time that MAG also promotes resistance to axonal injury and prevents axonal degeneration both in cell culture and in vivo. This effect on axonal stability depends on the RGD domain around arginine 118 in the extracellular portion of MAG, but it is independent of Nogo signaling in the axon. Exploiting this pathway may lead to therapeutic strategies for neurological diseases characterized by axonal loss.


Assuntos
Glicoproteína Associada a Mielina/fisiologia , Glicoproteína Associada a Mielina/uso terapêutico , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Acrilamida/toxicidade , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animais de Doenças , Proteínas Ligadas por GPI , Gânglios Espinais , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Mutagênese Sítio-Dirigida/métodos , Proteínas da Mielina/deficiência , Glicoproteína Associada a Mielina/deficiência , Degeneração Neural/etiologia , Degeneração Neural/genética , Degeneração Neural/patologia , Fibras Nervosas Mielinizadas/metabolismo , Condução Nervosa/genética , Condução Nervosa/fisiologia , Proteínas de Neurofilamentos/metabolismo , Receptor Nogo 1 , Fosfoinositídeo Fosfolipase C/toxicidade , Ratos , Receptores de Superfície Celular/deficiência , Traumatismos da Medula Espinal/complicações , Fatores de Tempo , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/uso terapêutico , Vincristina/uso terapêutico
5.
Infect Immun ; 76(4): 1379-89, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18227174

RESUMO

Host innate immune responses to many intracellular pathogens include the formation of inflammatory granulomas that are thought to provide a physical barrier between the microbe and host. Because two common features of infections with the live vaccine strain (LVS) of Francisella tularensis within the mouse liver are the formation of granulomas and the production of gamma interferon (IFN-gamma), we have asked what role IFN-gamma plays in hepatic granuloma formation and function. Francisella antigens were predominantly localized within granulomas of the livers of mice infected with F. tularensis LVS 4 days postinfection. Hepatic granulomas also contained large numbers of dying cells, some of which coexpressed the F4/80 macrophage antigen and activated caspase-3. IFN-gamma-deficient mice did not form normal numbers of hepatic granulomas and showed widely disseminated Francisella antigens within the liver. The incidence of cell death within hepatic granulomas also decreased significantly in the absence of IFN-gamma. Inducible NO synthase (iNOS) expression was restricted to the granulomas of wild-type mice but was not seen for IFN-gamma-deficient mice. Cell death within granulomas was also significantly decreased for iNOS-deficient mice. The predominant IFN-gamma-expressing cells in the liver were NK cells. Depleting NK cells resulted in the expression of bacterial antigens and iNOS outside the granulomas and the appearance of extensive hepatic focal necrosis. These findings indicate that IFN-gamma and hepatic NK cells that are activated during F. tularensis LVS infections regulate hepatic granuloma formation, the spatial containment of infection, the expression of iNOS, and the induction of cell death within the liver.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Granuloma/imunologia , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Fígado/imunologia , Vacinas Atenuadas/imunologia , Animais , Morte Celular , Deleção de Genes , Regulação Enzimológica da Expressão Gênica , Granuloma/microbiologia , Granuloma/patologia , Interferon gama/genética , Fígado/microbiologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Tularemia/imunologia , Tularemia/microbiologia , Tularemia/patologia
6.
J Neuroimmunol ; 193(1-2): 140-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18063113

RESUMO

Derivative myelin associated glycoprotein (dMAG) results from proteolysis of transmembrane MAG and can inhibit axonal growth. We have tested the ability of certain matrix metalloproteinases (MMPs) elevated with inflammatory and demyelinating diseases to cleave MAG. We show MMP-2, MMP-7 and MMP-9, but not MMP-1, cleave recombinant human MAG. Cleavage by MMP-7 occurs at Leu 509, just distal to the transmembrane domain and, to a lesser extent, at Met 234. We also show that MMP-7 cleaves MAG expressed on the external surface of CHO cells, releasing fragments that accumulate in the medium over periods of up to 48 h or more and that are able to inhibit outgrowth by dorsal root ganglion (DRG) neurons. We conclude that MMPs may have the potential both to disrupt MAG dependent axon-glia communication and to generate bioactive fragments that can inhibit neurite growth.


Assuntos
Metaloproteinases da Matriz/fisiologia , Glicoproteína Associada a Mielina/metabolismo , Sequência de Aminoácidos , Animais , Axônios/fisiologia , Células CHO , Cricetinae , Cricetulus , Gânglios Espinais/crescimento & desenvolvimento , Humanos , Metaloproteinase 7 da Matriz/fisiologia , Dados de Sequência Molecular , Esclerose Múltipla/enzimologia , Neuroglia/fisiologia , Fragmentos de Peptídeos/toxicidade , Proteínas Recombinantes/metabolismo , Transdução de Sinais
7.
J Neuroimmune Pharmacol ; 3(1): 52-6, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18046654

RESUMO

One of the hallmark features underlying the pathogenesis of HIV encephalitis is the disruption of blood-brain barrier (BBB). Cocaine, often abused by HIV-infected patients, has been suggested to worsen the HIV-associated dementia (HAD) via unknown mechanisms. The objective of the present study was to explore the effects of cocaine on BBB permeability using human brain microvascular endothelial cells (HBMECs). Additionally, because the chemokine CCL2 and its receptor CCR2 play a crucial role in the recruitment of inflammatory cells into the central nervous system in HAD brains, we tested for the effect of cocaine in modulating the CCL2/CCR2 axis. Our findings suggest that exposure of HBMECs to cocaine correlated with the breakdown of ZO-1 tight junction protein and reorganization of the cytoskeleton resulting in stress fiber formation. Furthermore, cocaine also modulated upregulation of the CCL2/CCR2 axis in monocytes. These findings conform to the multifaceted effects of cocaine leading to accelerated progression of HIV-1 neuropathogenesis.


Assuntos
Complexo AIDS Demência/fisiopatologia , Barreira Hematoencefálica/efeitos dos fármacos , Quimiocina CCL2/efeitos dos fármacos , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Junções Íntimas/efeitos dos fármacos , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Proteínas de Membrana/biossíntese , Proteínas de Membrana/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fosfoproteínas/biossíntese , Fosfoproteínas/efeitos dos fármacos , Receptores CCR2/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1
8.
Int J Parasitol ; 36(5): 513-9, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16620822

RESUMO

Using an in vitro model of the human blood-brain barrier consisting of human brain microvascular endothelial cells we recently demonstrated that Trypanosoma brucei gambiense bloodstream-forms efficiently cross these cells via a paracellular route while Trypanosoma brucei brucei crosses these cells poorly. Using a combination of techniques that include fluorescence activated cell sorting, confocal and electron microscopy, we now show that some T.b. gambiense blood stream form parasites have the capacity to enter human brain microvascular endothelial cells. The intracellular location of the trypanosomes was demonstrated in relation to the endothelial cell plasma membrane and to the actin cytoskeleton. These parasites may be a terminal stage within a lysosomal compartment or they may be viable trypanosomes that will be able to exit the brain microvascular endothelial cells. This process may provide an additional transcellular route by which the parasites cross the blood-brain barrier.


Assuntos
Barreira Hematoencefálica/parasitologia , Endotélio Vascular/parasitologia , Trypanosoma brucei gambiense/fisiologia , Tripanossomíase Africana/parasitologia , Animais , Barreira Hematoencefálica/ultraestrutura , Encéfalo/irrigação sanguínea , Células Cultivadas , Infecções Protozoárias do Sistema Nervoso Central/parasitologia , Infecções Protozoárias do Sistema Nervoso Central/patologia , Células Endoteliais/parasitologia , Células Endoteliais/ultraestrutura , Endotélio Vascular/ultraestrutura , Interações Hospedeiro-Parasita , Humanos , Microcirculação/parasitologia , Microscopia Confocal , Trypanosoma brucei gambiense/isolamento & purificação , Tripanossomíase Africana/patologia
9.
Infect Immun ; 73(5): 2923-31, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15845498

RESUMO

Escherichia coli K1 is a major gram-negative organism causing neonatal meningitis. E. coli K1 binding to and invasion of human brain microvascular endothelial cells (HBMEC) are a prerequisite for E. coli penetration into the central nervous system in vivo. In the present study, we showed using DNA microarray analysis that E. coli K1 associated with HBMEC expressed significantly higher levels of the fim genes compared to nonassociated bacteria. We also showed that E. coli K1 binding to and invasion of HBMEC were significantly decreased with its fimH deletion mutant and type 1 fimbria locked-off mutant, while they were significantly increased with its type 1 fimbria locked-on mutant. E. coli K1 strains associated with HBMEC were predominantly type 1 fimbria phase-on (i.e., fimbriated) bacteria. Taken together, we showed for the first time that type 1 fimbriae play an important role in E. coli K1 binding to and invasion of HBMEC and that type 1 fimbria phase-on E. coli is the major population interacting with HBMEC.


Assuntos
Encéfalo/irrigação sanguínea , Células Endoteliais/microbiologia , Escherichia coli/patogenicidade , Fímbrias Bacterianas/metabolismo , Microcirculação/microbiologia , Adesinas de Escherichia coli/genética , Adesinas de Escherichia coli/metabolismo , Animais , Aderência Bacteriana , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/microbiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Deleção de Genes , Humanos , Recém-Nascido , Análise de Sequência com Séries de Oligonucleotídeos , Coelhos
10.
Infect Immun ; 73(2): 1014-22, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15664945

RESUMO

Neurological manifestations of Lyme disease in humans are attributed in part to penetration of the blood-brain barrier (BBB) and invasion of the central nervous system (CNS) by Borrelia burgdorferi. However, how the spirochetes cross the BBB remains an unresolved issue. We examined the traversal of B. burgdorferi across the human BBB and systemic endothelial cell barriers using in vitro model systems constructed of human brain microvascular endothelial cells (BMEC) and EA.hy 926, a human umbilical vein endothelial cell (HUVEC) line grown on Costar Transwell inserts. These studies showed that B. burgdorferi differentially crosses human BMEC and HUVEC and that the human BMEC form a barrier to traversal. During the transmigration by the spirochetes, it was found that the integrity of the endothelial cell monolayers was maintained, as assessed by transendothelial electrical resistance measurements at the end of the experimental period, and that B. burgdorferi appeared to bind human BMEC by their tips near or at cell borders, suggesting a paracellular route of transmigration. Importantly, traversal of B. burgdorferi across human BMEC induces the expression of plasminogen activators, plasminogen activator receptors, and matrix metalloproteinases. Thus, the fibrinolytic system linked by an activation cascade may lead to focal and transient degradation of tight junction proteins that allows B. burgdorferi to invade the CNS.


Assuntos
Barreira Hematoencefálica/metabolismo , Borrelia burgdorferi/metabolismo , Doença de Lyme/metabolismo , Peptídeo Hidrolases/metabolismo , Barreira Hematoencefálica/microbiologia , Células Endoteliais/metabolismo , Humanos , Doença de Lyme/microbiologia
11.
J Biol Chem ; 280(2): 1360-8, 2005 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-15516338

RESUMO

Escherichia coli K1 is the most common Gram-negative organism causing meningitis, and its invasion of human brain microvascular endothelial cells (HBMEC) is a prerequisite for penetration into the central nervous system. We have reported previously that cytotoxic necrotizing factor 1 (CNF1) contributes to E. coli K1 invasion of HBMEC and interacts with 37-kDa laminin receptor precursor (37LRP) of HBMEC, which is a precursor of 67-kDa laminin receptor (67LR). In the present study, we examined the role of 67LR in the CNF1-expressing E. coli K1 invasion of HBMEC. Immunofluorescence microscopy and ligand overlay assays showed that 67LR is present on the HBMEC membrane and interacts with CNF1 protein as well as the CDPGYIGSR laminin peptide. 67LR was up-regulated and clustered at the sites of E. coli K1 on HBMEC in a CNF1-dependent manner. Pretreatment of CNF1+ E. coli K1 with recombinant 37-kDa laminin receptor precursor reduced the invasion rate to the level of Deltacnf1 mutant, and the invasion rate of CNF1+ E. coli K1 was enhanced in 67LR-overexpressing HBMEC, indicating 67LR is involved in the CNF1+ E. coli K1 invasion of HBMEC. Coimmunoprecipitation analysis showed that, upon incubation with CNF1+ E. coli K1 but not with Deltacnf1 mutant, focal adhesion kinase and paxillin were recruited and associated with 67LR. When immobilized onto polystyrene beads, CNF1 was sufficient to induce internalization of coupled beads into HBMEC through interaction with 67LR. Taken together, this is the first demonstration that E. coli K1 invasion of HBMEC occurs through the ligand-receptor (CNF1-67LR) interaction, and 67LR promotes CNF1-expressing E. coli K1 internalization of HBMEC.


Assuntos
Toxinas Bacterianas/metabolismo , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Receptores de Laminina/metabolismo , Toxinas Bacterianas/genética , Sítios de Ligação , Encéfalo/microbiologia , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/citologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Imunoprecipitação , Lisossomos/metabolismo , Lisossomos/microbiologia , Microcirculação , Microesferas , Peso Molecular , Paxilina , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores de Laminina/química , Receptores de Laminina/genética , Regulação para Cima
12.
J Parasitol ; 90(5): 970-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15562595

RESUMO

The neurological manifestations of sleeping sickness in man are attributed to the penetration of the blood-brain barrier (BBB) and invasion of the central nervous system by Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense. However, how African trypanosomes cross the BBB remains an unresolved issue. We have examined the traversal of African trypanosomes across the human BBB using an in vitro BBB model system constructed of human brain microvascular endothelial cells (BMECs) grown on Costar Transwell inserts. Human-infective T. b. gambiense strain IL 1852 was found to cross human BMECs far more readily than the animal-infective Trypanosoma brucei brucei strains 427 and TREU 927. Tsetse fly-infective procyclic trypomastigotes did not cross the human BMECs either alone or when coincubated with bloodstreamform T. b. gambiense. After overnight incubation, the integrity of the human BMEC monolayer measured by transendothelial electrical resistance was maintained on the inserts relative to the controls when the endothelial cells were incubated with T. b. brucei. However, decreases in electrical resistance were observed when the BMEC-coated inserts were incubated with T. b. gambiense. Light and electron microscopy studies revealed that T. b. gambiense initially bind at or near intercellular junctions before crossing the BBB paracellularly. This is the first demonstration of paracellular traversal of African trypanosomes across the BBB. Further studies are required to determine the mechanism of BBB traversal by these parasites at the cellular and molecular level.


Assuntos
Barreira Hematoencefálica/parasitologia , Células Endoteliais/parasitologia , Trypanosoma brucei brucei/fisiologia , Trypanosoma brucei gambiense/fisiologia , Animais , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/ultraestrutura , Cálcio/metabolismo , Linhagem Celular , Impedância Elétrica , Células Endoteliais/ultraestrutura , Humanos , Técnicas In Vitro , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Transfecção
13.
FEMS Immunol Med Microbiol ; 42(3): 271-9, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15477040

RESUMO

The mortality and morbidity associated with neonatal gram-negative meningitis have remained significant despite advances in antimicrobial chemotherapy. Escherichia coli K1 is the most common gram-negative organism causing neonatal meningitis. Our incomplete knowledge of the pathogenesis of this disease is one of the main reasons for this high mortality and morbidity. We have previously established both in vitro and in vivo models of the blood-brain barrier (BBB) using human brain microvascular endothelial cells (HBMEC) and hematogenous meningitis in neonatal rats, respectively. With these in vitro and in vivo models, we have shown that successful crossing of the BBB by circulating E. coli requires a high-degree of bacteremia, E. coli binding to and invasion of HBMEC, and E. coli traversal of the BBB as live bacteria. Our previous studies using TnphoA, signature-tagged mutagenesis and differential fluorescence induction identified several E. coli K1 determinants such as OmpA, Ibe proteins, AslA, TraJ and CNF1 contributing to invasion of HBMEC in vitro and traversal of the blood-brain barrier in vivo. We have shown that some of these determinants interact with specific receptors on HBMEC, suggesting E. coli translocation of the BBB is the result of specific pathogen-host cell interactions. Recent studies using functional genomics techniques have identified additional E. coli K1 factors that contribute to the high degree of bacteremia and HBMEC binding/invasion/transcytosis. In this review, we summarize the current knowledge on the mechanisms underlying the successful E. coli translocation of the BBB.


Assuntos
Translocação Bacteriana , Barreira Hematoencefálica/microbiologia , Escherichia coli/patogenicidade , Meningite devida a Escherichia coli/microbiologia , Animais , Bacteriemia , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Escherichia coli/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Humanos , Recém-Nascido , Fatores de Virulência/genética , Fatores de Virulência/fisiologia
14.
Cell Microbiol ; 6(8): 743-51, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15236641

RESUMO

Human granulocytic anaplasmosis (HGA) is caused by the obligate intracellular bacterium Anaplasma phagocytophilum. The bacterium infects, survives, propagates in, and alters neutrophil phenotype, indicating unique survival mechanisms. AnkA is the only known A. phagocytophilum component that gains access beyond neutrophil vacuoles and is transported to the infected host cell nucleus. The ability of native and recombinant AnkA to bind DNA and nuclear proteins from host HL-60 cells was assessed by the use of immunoprecipitation after cis-diamminedichloroplatinum (cis-DDP) DNA-protein crosslinking, by probing uninfected HL-60 cell nuclear lysates for AnkA binding, and by recovery and sequence analysis of immunoprecipitated DNA. AnkA binds HL-60 cell DNA as well as nuclear proteins of approximately 86, 53 and 25 kDa, whereas recombinant A. phagocytophilum Msp2 or control proteins do not. DNA immunoprecipitation reveals AnkA binding to a variety of target genes in the human genome, including genes that encode proteins with ATPase, tyrosine phosphatase and NADH dehydrogenase-like functions. These data indicate that AnkA could exert some effect on cells through binding to protein:DNA complexes in neutrophil nuclei. Whether AnkA binding leads to neutrophil functional alterations, and how such alterations might occur will depend upon definitive identification of binding partners and associated metabolic and biochemical pathways.


Assuntos
Anaplasma phagocytophilum/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , DNA/metabolismo , Granulócitos/microbiologia , Proteínas Nucleares/metabolismo , Adenosina Trifosfatases/genética , Núcleo Celular/metabolismo , Núcleo Celular/microbiologia , DNA/análise , Células HL-60 , Humanos , Peso Molecular , NADH Desidrogenase/genética , Testes de Precipitina , Proteínas Tirosina Fosfatases/genética
15.
FEMS Microbiol Lett ; 227(2): 243-7, 2003 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-14592715

RESUMO

Anaplasma phagocytophilum 44-kDa major surface protein-2 (Msp2) mediates partial neutrophil adhesion and interactions. Since A. phagocytophilum 44-kDa monoclonal antibodies also react with 160- and 100-kDa bands, a putative adhesin complex was studied. After separate excision/immunoprecipitation of these three bands, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) resolved each into three bands again with increased 44-kDa protein under reducing conditions suggesting oligomerization of Msp2 44-kDa monomers. With 9 M urea, each separately excised band was resolved only into 44-kDa monomers with three different pIs. With protein cross-linking, immunoblots showed four additional bands and increased high molecular mass band intensity, suggesting homo- and hetero-polymerization with other A. phagocytophilum proteins. Recognition of Msp2 complexes facilitates understanding of A. phagocytophilum-neutrophil adhesion.


Assuntos
Anaplasma phagocytophilum/metabolismo , Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Anaplasma phagocytophilum/imunologia , Técnicas de Cultura de Células , Glicoproteínas de Membrana/metabolismo , Neutrófilos/microbiologia
16.
Microb Pathog ; 35(1): 35-42, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12860457

RESUMO

Escherichia coli K1 invasion of brain microvascular endothelial cells (BMEC) is a prerequisite for penetration into the central nervous system. We previously have shown that outer membrane protein A (OmpA) and cytotoxic necrotizing factor-1 (CNF1) contribute to E. coli K1 invasion of BMEC. In this study we constructed a double-knockout mutant by deleting ompA and cnf1. We demonstrated that the double-knockout mutant was significantly less invasive in human BMEC as compared with its individual Delta ompA and Delta cnf1 mutants, suggesting that the contributions of OmpA and CNF1 to BMEC invasion are independent of each other. In addition, we showed that OmpA treatment of human BMEC resulted in phosphatidylinositol 3-kinase (PI3K) activation with no effect on RhoA, while CNF1 treatment resulted in RhoA activation with no effect on PI3K, supporting the concept that OmpA and CNF1 contribute to E. coli K1 invasion of BMEC using different mechanisms. This concept was further confirmed by using both PI3K inhibitor (LY294002) and Rho kinase inhibitor (Y27632), which exhibited additive effects on inhibiting E. coli K1 invasion of BMEC. We isolated a 96KD OmpA interacting human BMEC protein by affinity chromatography using purified OmpA, which was identified as gp96 protein, a member of the HSP90 family. This receptor differed from the CNF1 receptor (37LRP) identified from human BMEC. Taken together, these data indicate that OmpA and CNF1 contribute to E. coli K1 invasion of BMEC in an additive manner by interacting with different BMEC receptors and using diverse host cell signaling mechanisms.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Citotoxinas/fisiologia , Endotélio Vascular/microbiologia , Proteínas de Escherichia coli , Escherichia coli/patogenicidade , Meningite devida a Escherichia coli/microbiologia , Anticorpos/farmacologia , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Barreira Hematoencefálica/imunologia , Circulação Cerebrovascular/imunologia , Cromatografia de Afinidade , Cromonas/farmacologia , Citotoxinas/genética , Citotoxinas/imunologia , Inibidores Enzimáticos/farmacologia , Escherichia coli/genética , Escherichia coli/imunologia , Humanos , Morfolinas/farmacologia , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Cell Microbiol ; 5(4): 245-52, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12675682

RESUMO

Escherichia coli K1 has been shown to invade human brain microvascular endothelial cells (HBMEC) in vitro and translocate the blood-brain barrier in vivo, but it is unclear how E. coli K1 traverses HBMEC. We have previously shown that internalized E. coli K1 is localized within membrane-bound vacuole in HBMEC. The present study was carried out to understand intracellular trafficking of E. coli K1 containing vacuoles (ECVs) in HBMEC. ECVs initially acquired two early endosomal marker proteins, EEA1 and transferrin receptor. Rab7 and Lamp-1, markers for late endosome and late endosome/lysosome, respectively, were subsequently recruited on the ECVs, which was confirmed with flow cytometry analysis of ECVs. However, ECVs did not obtain cathepsin D, a lysosomal enzyme, even after 120 min incubation, suggesting that E. coli K1 avoids lysosomal fusion. In contrast, isogenic K1 capsule-deletion mutant obtained early and late endosomal markers on vacuolar membranes and allowed lysosomal fusion with subsequent degradation inside vacuoles. This observation was consistent with the decreased intracellular survival of K1 capsule-deletion mutant, even though the binding and internalization rates of the mutant were higher than those of the parent E. coli K1 strain. This is the first demonstration that E. coli K1, via the K1 capsule on the bacterial surface, modulates the maturation process of ECVs and prevents fusion with lysosomes, which is an event necessary for traversal of the blood-brain barrier as live bacteria.


Assuntos
Cápsulas Bacterianas/metabolismo , Transporte Biológico/fisiologia , Encéfalo/microbiologia , Endotélio Vascular/microbiologia , Escherichia coli/metabolismo , Polissacarídeos Bacterianos/metabolismo , Vacúolos/metabolismo , Antígenos de Bactérias/metabolismo , Cápsulas Bacterianas/ultraestrutura , Barreira Hematoencefálica/fisiologia , Encéfalo/citologia , Linhagem Celular , Endocitose/fisiologia , Endotélio Vascular/citologia , Epitopos , Escherichia coli/ultraestrutura , Humanos , Lisossomos/metabolismo
18.
J Biol Chem ; 278(19): 16857-62, 2003 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-12615923

RESUMO

Cytotoxic necrotizing factor 1 (CNF1) is a bacterial toxin known to activate Rho GTPases and induce host cell cytoskeleton rearrangements. The constitutive activation of Rho GTPases by CNF1 is shown to enhance bacterial uptake in epithelial cells and human brain microvascular endothelial cells. However, it is unknown how exogenous CNF1 exhibits such phenotypes in eukaryotic cells. Here, we identified 37-kDa laminin receptor precursor (LRP) as the receptor for CNF1 from screening the cDNA library of human brain microvascular endothelial cells by the yeast two-hybrid system using the N-terminal domain of CNF1 as bait. CNF1-mediated RhoA activation and bacterial uptake were inhibited by exogenous LRP or LRP antisense oligodeoxynucleotides, whereas they were increased in LRP-overexpressing cells. These findings indicate that the CNF1 interaction with LRP is the initial step required for CNF1-mediated RhoA activation and bacterial uptake in eukaryotic cells.


Assuntos
Citotoxinas/fisiologia , Endotélio Vascular/microbiologia , Endotélio Vascular/fisiologia , Proteínas de Escherichia coli , Escherichia coli/fisiologia , Precursores de Proteínas/fisiologia , Receptores de Laminina/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Toxinas Bacterianas , Linhagem Celular , Ativação Enzimática , Infecções por Escherichia coli/etiologia , Infecções por Escherichia coli/metabolismo , Oligonucleotídeos Antissenso , Ligação Proteica , Transdução de Sinais
19.
Cell Tissue Res ; 309(2): 281-6, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12172787

RESUMO

Escherichia coli K1 traversal of the human brain microvascular endothelial cells (HBMEC) that constitute the blood-brain barrier (BBB) is a complex process involving E. coli adherence to and invasion of HBMEC. In this study, we demonstrated that human transforming growth factor-beta-1 (TGF-beta1) increases E. coli K1 adherence, invasion, and transcytosis in HBMEC. In addition, TGF-beta1 increases RhoA activation and enhances actin condensation in HBMEC. We have previously shown that E. coli K1 invasion of HBMEC requires phosphatidylinositol-3 kinase (PI3K) and RhoA activation. TGF-beta1 increases E. coli K1 invasion in PI3K dominant-negative HBMEC, but not in RhoA dominant-negative HBMEC, indicating that TGF-beta1-mediated increase in E. coli K1 invasion is RhoA-dependent, but not PI3K-dependent. Our findings suggest that TGF-beta1 treatment of HBMEC increases E. coli K1 adherence, invasion, and transcytosis, which are probably dependent on RhoA.


Assuntos
Encéfalo/microbiologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Endotélio Vascular/microbiologia , Escherichia coli/patogenicidade , Fator de Crescimento Transformador beta/farmacologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Endotélio Vascular/citologia , Ativação Enzimática/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Humanos , Proteína rhoA de Ligação ao GTP/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
20.
J Biol Chem ; 277(18): 15607-12, 2002 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-11877402

RESUMO

Escherichia coli K1 invasion of brain microvascular endothelial cells (BMECs) is a prerequisite for penetration into the central nervous system and requires actin cytoskeletal rearrangements. Here, we demonstrate that E. coli K1 invasion of BMECs requires RhoA activation. In addition, we show that cytotoxic necrotizing factor-1 (CNF1) contributes to E. coli K1 invasion of brain endothelial cells in vitro and traversal of the blood-brain barrier in the experimental hematogenous meningitis animal model. These in vitro and in vivo effects of CNF1 were dependent upon RhoA activation as shown by (a) decreased invasion and RhoA activation with the Delta cnf1 mutant of E. coli K1 and (b) restoration of invasion frequency of the Delta cnf1 mutant to the level of the parent E. coli K1 strain in BMECs with constitutively active RhoA. In addition, CNF1-enhanced E. coli invasion of brain endothelial cells and stress fiber formation were independent of focal adhesion kinase and phosphatidylinositol 3-kinase activation. This is the first demonstration that CNF1 contributes to E. coli K1 invasion of BMECs.


Assuntos
Encéfalo/microbiologia , Citotoxinas/fisiologia , Proteínas de Escherichia coli , Escherichia coli/patogenicidade , Amidas/farmacologia , Animais , Toxinas Bacterianas/genética , Sequência de Bases , Barreira Hematoencefálica , Células Cultivadas , Circulação Cerebrovascular/fisiologia , Citotoxinas/genética , Primers do DNA , Modelos Animais de Doenças , Endotélio Vascular/microbiologia , Inibidores Enzimáticos/farmacologia , Escherichia coli/isolamento & purificação , Escherichia coli/fisiologia , Deleção de Genes , Humanos , Recém-Nascido , Meningites Bacterianas/microbiologia , Mutagênese Sítio-Dirigida , Piridinas/farmacologia , Ratos , Proteínas Recombinantes/metabolismo , Transfecção , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...