Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Biol Chem ; 290(30): 18316-31, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25963144

RESUMO

Airway epithelia express sialylated receptors that recognize exogenous danger signals. Regulation of receptor responsiveness to these signals remains incompletely defined. Here, we explore the mechanisms through which the human sialidase, neuraminidase-1 (NEU1), promotes the interaction between the sialoprotein, mucin 1 (MUC1), and the opportunistic pathogen, Pseudomonas aeruginosa. P. aeruginosa flagellin engaged the MUC1 ectodomain (ED), increasing NEU1 association with MUC1. The flagellin stimulus increased the association of MUC1-ED with both NEU1 and its chaperone/transport protein, protective protein/cathepsin A. Scatchard analysis demonstrated NEU1-dependent increased binding affinity of flagellin to MUC1-expressing epithelia. NEU1-driven MUC1-ED desialylation rapidly increased P. aeruginosa adhesion to and invasion of the airway epithelium. MUC1-ED desialylation also increased its shedding, and the shed MUC1-ED competitively blocked P. aeruginosa adhesion to cell-associated MUC1-ED. Levels of desialylated MUC1-ED were elevated in the bronchoalveolar lavage fluid of mechanically ventilated patients with P. aeruginosa airway colonization. Preincubation of P. aeruginosa with these same ex vivo fluids competitively inhibited bacterial adhesion to airway epithelia, and MUC1-ED immunodepletion completely abrogated their inhibitory activity. These data indicate that a prokaryote, P. aeruginosa, in a ligand-specific manner, mobilizes eukaryotic NEU1 to enhance bacterial pathogenicity, but the host retaliates by releasing MUC1-ED into the airway lumen as a hyperadhesive decoy receptor.


Assuntos
Flagelina/metabolismo , Pulmão/metabolismo , Mucina-1/metabolismo , Neuraminidase/metabolismo , Pseudomonas aeruginosa/metabolismo , Adesinas Bacterianas/metabolismo , Aderência Bacteriana , Líquido da Lavagem Broncoalveolar , Linhagem Celular , Humanos , Pulmão/microbiologia , Pulmão/patologia , Ácido N-Acetilneuramínico/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/patogenicidade
2.
Am J Physiol Lung Cell Mol Physiol ; 306(9): L876-86, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24658138

RESUMO

Sialic acids on glycoconjugates play a pivotal role in many biological processes. In the airways, sialylated glycoproteins and glycolipids are strategically positioned on the plasma membranes of epithelia to regulate receptor-ligand, cell-cell, and host-pathogen interactions at the molecular level. We now demonstrate, for the first time, sialidase activity for ganglioside substrates in human airway epithelia. Of the four known mammalian sialidases, NEU3 has a substrate preference for gangliosides and is expressed at mRNA and protein levels at comparable abundance in epithelia derived from human trachea, bronchi, small airways, and alveoli. In small airway and alveolar epithelia, NEU3 protein was immunolocalized to the plasma membrane, cytosolic, and nuclear subcellular fractions. Small interfering RNA-induced silencing of NEU3 expression diminished sialidase activity for a ganglioside substrate by >70%. NEU3 immunostaining of intact human lung tissue could be localized to the superficial epithelia, including the ciliated brush border, as well as to nuclei. However, NEU3 was reduced in subepithelial tissues. These results indicate that human airway epithelia express catalytically active NEU3 sialidase.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Epitélio/metabolismo , Neuraminidase/metabolismo , Sistema Respiratório/metabolismo , Biotinilação , Western Blotting , Catálise , Células Cultivadas , Citometria de Fluxo , Gangliosídeos/metabolismo , Humanos , Técnicas Imunoenzimáticas , Neuraminidase/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácidos Siálicos/metabolismo , Frações Subcelulares
3.
J Biol Chem ; 289(13): 9121-35, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24550400

RESUMO

The highly sialylated vascular endothelial surface undergoes changes in sialylation upon adopting the migratory/angiogenic phenotype. We recently established endothelial cell (EC) expression of NEU1 sialidase (Cross, A. S., Hyun, S. W., Miranda-Ribera, A., Feng, C., Liu, A., Nguyen, C., Zhang, L., Luzina, I. G., Atamas, S. P., Twaddell, W. S., Guang, W., Lillehoj, E. P., Puché, A. C., Huang, W., Wang, L. X., Passaniti, A., and Goldblum, S. E. (2012) NEU1 and NEU3 sialidase activity expressed in human lung microvascular endothelia. NEU1 restrains endothelial cell migration whereas NEU3 does not. J. Biol. Chem. 287, 15966-15980). We asked whether NEU1 might regulate EC capillary-like tube formation on a Matrigel substrate. In human pulmonary microvascular ECs (HPMECs), prior silencing of NEU1 did not alter tube formation. Infection of HPMECs with increasing multiplicities of infection of an adenovirus encoding for catalytically active WT NEU1 dose-dependently impaired tube formation, whereas overexpression of either a catalytically dead NEU1 mutant, NEU1-G68V, or another human sialidase, NEU3, did not. NEU1 overexpression also diminished EC adhesion to the Matrigel substrate and restrained EC migration in a wounding assay. In HPMECs, the adhesion molecule, CD31, also known as platelet endothelial cell adhesion molecule-1, was sialylated via α2,6-linkages, as shown by Sambucus nigra agglutinin lectin blotting. NEU1 overexpression increased CD31 binding to Arachis hypogaea or peanut agglutinin lectin, indicating CD31 desialylation. In the postconfluent state, when CD31 ectodomains are homophilically engaged, NEU1 was recruited to and desialylated CD31. In postconfluent ECs, CD31 was desialylated compared with subconfluent cells, and prior NEU1 silencing completely protected against CD31 desialylation. Prior CD31 silencing and the use of CD31-null ECs each abrogated the NEU1 inhibitory effect on EC tube formation. Sialyltransferase 6 GAL-I overexpression increased α2,6-linked CD31 sialylation and dose-dependently counteracted NEU1-mediated inhibition of EC tube formation. These combined data indicate that catalytically active NEU1 inhibits in vitro angiogenesis through desialylation of its substrate, CD31.


Assuntos
Capilares/citologia , Células Endoteliais/metabolismo , Pulmão/irrigação sanguínea , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Animais , Antígenos CD/genética , Capilares/fisiologia , Adesão Celular , Movimento Celular , Células Endoteliais/citologia , Humanos , Camundongos , Neovascularização Fisiológica , Transporte Proteico , Sialiltransferases/genética
4.
J Biol Chem ; 287(19): 15966-80, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22403397

RESUMO

The microvascular endothelial surface expresses multiple molecules whose sialylation state regulates multiple aspects of endothelial function. To better regulate these sialoproteins, we asked whether endothelial cells (ECs) might express one or more catalytically active sialidases. Human lung microvascular EC lysates contained heat-labile sialidase activity for a fluorogenic substrate, 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MU-NANA), that was dose-dependently inhibited by the competitive sialidase inhibitor, 2,3-dehydro-2-deoxy-N-acetylneuraminic acid but not its negative control. The EC lysates also contained sialidase activity for a ganglioside mixture. Using real time RT-PCR to detect mRNAs for the four known mammalian sialidases, NEU1, -2, -3, and -4, NEU1 mRNA was expressed at levels 2700-fold higher that those found for NEU2, -3, or -4. Western analyses indicated NEU1 and -3 protein expression. Using confocal microscopy and flow cytometry, NEU1 was immunolocalized to both the plasma membrane and the perinuclear region. NEU3 was detected both in the cytosol and nucleus. Prior siRNA-mediated knockdown of NEU1 and NEU3 each decreased EC sialidase activity for 4-MU-NANA by >65 and >17%, respectively, and for the ganglioside mixture by 0 and 40%, respectively. NEU1 overexpression in ECs reduced their migration into a wound by >40%, whereas NEU3 overexpression did not. Immunohistochemical studies of normal human tissues immunolocalized NEU1 and NEU3 proteins to both pulmonary and extrapulmonary vascular endothelia. These combined data indicate that human lung microvascular ECs as well as other endothelia express catalytically active NEU1 and NEU3. NEU1 restrains EC migration, whereas NEU3 does not.


Assuntos
Movimento Celular , Células Endoteliais/enzimologia , Neuraminidase/metabolismo , Aorta/enzimologia , Artérias Carótidas/enzimologia , Linhagem Celular , Membrana Celular/enzimologia , Núcleo Celular/enzimologia , Artérias Cerebrais/enzimologia , Citosol/enzimologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Humanos , Himecromona/análogos & derivados , Himecromona/farmacologia , Immunoblotting , Rim/enzimologia , Pulmão/enzimologia , Microscopia Confocal , Ácido N-Acetilneuramínico/análogos & derivados , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/antagonistas & inibidores , Neuraminidase/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade por Substrato
5.
J Biol Chem ; 287(11): 8214-31, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22247545

RESUMO

Epithelial cells (ECs) lining the airways provide a protective barrier between the external environment and the internal host milieu. These same airway epithelia express receptors that respond to danger signals and initiate repair programs. Because the sialylation state of a receptor can influence its function and is dictated in part by sialidase activity, we asked whether airway epithelia express catalytically active sialidase(s). Human primary small airway and A549 ECs expressed NEU1 sialidase at the mRNA and protein levels, and NEU1 accounted for >70% of EC sialidase activity. Blotting with Maackia amurensis and peanut agglutinin lectins established epidermal growth factor receptor (EGFR) and MUC1 as in vivo substrates for NEU1. NEU1 associated with EGFR and MUC1, and NEU1-EGFR association was regulated by EGF stimulation. NEU1 overexpression diminished EGF-stimulated EGFR Tyr-1068 autophosphorylation by up to 44% but enhanced MUC1-dependent Pseudomonas aeruginosa adhesion by 1.6-1.7-fold and flagellin-stimulated ERK1/2 activation by 1.7-1.9-fold. In contrast, NEU1 depletion increased EGFR activation (1.5-fold) and diminished MUC1-mediated bacterial adhesion (38-56%) and signaling (73%). These data indicate for the first time that human airway epithelia express catalytically active NEU1 sialidase that regulates EGFR- and MUC1-dependent signaling and bacterial adhesion. NEU1 catalytic activity may offer an additional level of regulation over the airway epithelial response to ligands, pathogens, and injurious stimuli.


Assuntos
Receptores ErbB/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Mucina-1/metabolismo , Neuraminidase/biossíntese , Mucosa Respiratória/metabolismo , Linhagem Celular Transformada , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mucina-1/genética , Neuraminidase/genética , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Mucosa Respiratória/microbiologia
6.
Am J Physiol Lung Cell Mol Physiol ; 291(6): L1232-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16891393

RESUMO

Tumor necrosis factor (TNF)-alpha is a key mediator of sepsis-associated multiorgan failure, including the acute respiratory distress syndrome. We examined the role of protein tyrosine phosphorylation in TNF-alpha-induced pulmonary vascular permeability. Postconfluent human lung microvascular and pulmonary artery endothelial cell (EC) monolayers exposed to human recombinant TNF-alpha displayed a dose- and time-dependent increase in transendothelial [(14)C]albumin flux in the absence of EC injury. TNF-alpha also increased tyrosine phosphorylation of EC proteins, and several substrates were identified as the zonula adherens proteins vascular endothelial (VE)-cadherin, and beta-catenin, gamma-catenin, and p120 catenin (p120(ctn)). Prior protein tyrosine kinase (PTK) inhibition protected against the TNF-alpha effect. TNF-alpha activated multiple PTKs, including src family PTKs. Prior PTK inhibition with the src-selective agents PP1 and PP2 each protected against approximately 60% of the TNF-alpha-induced increment in [(14)C]albumin flux. PP2 also blocked TNF-alpha-induced tyrosine phosphorylation of VE-cadherin, gamma-catenin, and p120(ctn). To identify which src family kinase(s) was required for TNF-alpha-induced vascular permeability, small interfering RNA (siRNA) targeting each of the three src family PTKs expressed in human EC, c-src, fyn, and yes, were introduced into the barrier function assay. Only fyn siRNA protected against the TNF-alpha effect, whereas the c-src and yes siRNAs did not. These combined data suggest that TNF-alpha regulates the pulmonary vascular endothelial paracellular pathway, in part, through fyn activation.


Assuntos
Caderinas/metabolismo , Endotélio Vascular/fisiologia , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-fyn/fisiologia , Mucosa Respiratória/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Caderinas/efeitos dos fármacos , Linhagem Celular , Endotélio Vascular/efeitos dos fármacos , Humanos , Pulmão , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-fyn/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Mucosa Respiratória/efeitos dos fármacos
7.
Am J Pathol ; 166(4): 1247-58, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793303

RESUMO

The pulmonary vascular endothelial paracellular pathway and zonula adherens (ZA) integrity are regulated, in part, through protein tyrosine phosphorylation. ZA-associated protein tyrosine phosphatase (PTP)s are thought to counterregulate tyrosine phosphorylation events within the ZA multiprotein complex. One such receptor PTP, PTPmu, is highly expressed in lung tissue and is almost exclusively restricted to the endothelium. We therefore studied whether PTPmu, in pulmonary vascular endothelia, associates with and/or regulates both the tyrosine phosphorylation state of vascular endothelial (VE)-cadherin and the paracellular pathway. PTPmu was expressed in postconfluent human pulmonary artery and lung microvascular endothelial cells (ECs) where it was almost exclusively restricted to EC-EC boundaries. In human lung microvascular ECs, knockdown of PTPmu through RNA interference dramatically impaired barrier function. In immortalized human microvascular ECs, overexpression of wild-type PTPmu enhanced barrier function. PTPmu-VE-cadherin interactions were demonstrated through reciprocal co-immunoprecipitation assays and co-localization with double-label fluorescence microscopy. When glutathione S-transferase-PTPmu was incubated with purified recombinant VE-cadherin, and when glutathione S-transferase-VE-cadherin was incubated with purified recombinant PTPmu, PTPmu directly bound to VE-cadherin. Overexpression of wild-type PTPmu decreased tyrosine phosphorylation of VE-cadherin. Therefore, PTPmu is expressed in human pulmonary vascular endothelia where it directly binds to VE-cadherin and regulates both the tyrosine phosphorylation state of VE-cadherin and barrier integrity.


Assuntos
Barreira Alveolocapilar/metabolismo , Permeabilidade Capilar/fisiologia , Endotélio Vascular/metabolismo , Pulmão/irrigação sanguínea , Proteínas Tirosina Fosfatases/metabolismo , Caderinas/metabolismo , Células Cultivadas , Glutationa Transferase/metabolismo , Humanos , Immunoblotting , Microcirculação/metabolismo , Fosforilação , Artéria Pulmonar/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Anticancer Res ; 24(1): 235-40, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15015602

RESUMO

Muc1 is the cell surface glycoprotein abundantly expressed in cancer cells and has been shown to be involved in tumor metastasis and promotion. Recently, we identified a 37 bp segment on the hamster Muc1 promoter with the ability to suppress Muc1 transcription. This 37 bp putative negative regulatory element (NRE) binds to a transcriptional regulator Yin Yang 1 (YY1). In the present study, we examined whether binding of YY1 is responsible for the negative regulatory effect by the 37 bp segment using a hamster pancreatic cancer cell line, HP-1 cells, transfected with various expression plasmid constructs. Our results showed that: (1) overexpression of YY1 up-regulated the transcriptional activity of the full-length hamster Muc1 promoter in a dose-dependent manner; (2) the mutation of the YY1 binding site did not affect either the basal transcriptional activity or the increased transcriptional activity by YY1; and (3) even the deletion of the 37 bp NRE segment could not abrogate the increased transcriptional activity by YY1. We conclude that the NRE acts in a YY1-independent manner and that YY1 instead enhances Muc1 transcriptional activity. Further study of the precise mechanism by which YY1 augments Muc1 gene expression should be worthwhile.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Mucina-1/genética , Neoplasias Pancreáticas/genética , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular Tumoral , Cricetinae , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Regulação Neoplásica da Expressão Gênica , Genes Reguladores , Mutação , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Fator de Transcrição YY1
9.
Am J Physiol Lung Cell Mol Physiol ; 285(1): L63-75, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12626337

RESUMO

Protein tyrosine phosphorylation is tightly regulated through the actions of both protein tyrosine kinases and protein tyrosine phosphatases. In this study, we demonstrate that protein tyrosine phosphatase inhibition promotes tyrosine phosphorylation of endothelial cell-cell adherens junction proteins, opens an endothelial paracellular pathway, and increases both transendothelial albumin flux and neutrophil migration. Tyrosine phosphatase inhibition with sodium orthovanadate or phenylarsine oxide induced dose- and time-dependent increases in [14C]bovine serum albumin flux across postconfluent bovine pulmonary artery endothelial cell monolayers. These increases in albumin flux were coincident with actin reorganization and intercellular gap formation in both postconfluent monolayers and preformed endothelial cell capillary tubes. Vanadate (25 microM) increased tyrosine phosphorylation of endothelial cell proteins 12-fold within 1 h. Tyrosine phosphorylated proteins were immunolocalized to the intercellular boundaries, and several were identified as the endothelial cell-cell adherens junction proteins, vascular-endothelial cadherin, and beta-, gamma-, and p120-catenin as well as platelet endothelial cell adhesion molecule-1. Of note, these tyrosine phosphorylation events were not associated with disassembly of the adherens junction complex or its uncoupling from the actin cytoskeleton. The dose and time requirements for vanadate-induced increases in phosphorylation were comparable with those defined for increments in transendothelial [14C]albumin flux and neutrophil migration, and pretreatment with the tyrosine kinase inhibitor herbimycin A protected against these effects. These data suggest that protein tyrosine phosphatases and their substrates, which localize to the endothelial cell-cell boundaries, regulate adherens junctional integrity, the movement of macromolecules and cells through the endothelial paracellular pathway, and capillary tube stability.


Assuntos
Endotélio Vascular/enzimologia , Proteínas Tirosina Fosfatases/metabolismo , Artéria Pulmonar/enzimologia , Junções Aderentes/enzimologia , Animais , Antígenos CD , Caderinas/metabolismo , Capilares/enzimologia , Radioisótopos de Carbono , Bovinos , Movimento Celular/imunologia , Radioisótopos de Cromo , Proteínas do Citoesqueleto/metabolismo , Desmoplaquinas , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Neovascularização Fisiológica/fisiologia , Neutrófilos/citologia , Neutrófilos/enzimologia , Fosforilação , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Artéria Pulmonar/citologia , Soroalbumina Bovina/farmacocinética , Transativadores/metabolismo , Tirosina/metabolismo , Vanadatos/farmacologia , beta Catenina
10.
Am J Physiol Lung Cell Mol Physiol ; 284(1): L160-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12388348

RESUMO

The mucin gene Muc1 is expressed in glandular epithelial cells and is involved in lubricative and protective functions. It is also overexpressed in many carcinomas including breast and lung cancer cells. To study the transcriptional regulation of Muc1, we cloned a 2.4-kb fragment containing the promoter region of the hamster Muc1 gene and analyzed it for its ability to mediate transcription. Transcriptional initiation was localized to 22 base pairs downstream of the TATA box. We performed functional analysis of the Muc1 promoter in hamster (HP-1 and Chinese hamster ovary) and human cells (MCF-7, A549, and BEAS-2B) using deletion/reporter constructs. A positive regulatory region between bases -555 and -252 and a putative negative regulatory element (P-NRE) between nucleotides -1,652 and -1,614 were found to be active in transfected cells. The P-NRE contains a yin yang 1 (YY1) transcription factor binding site, and electrophoretic mobility shift assays with HP-1 cell nuclear extract revealed the binding of YY1 to this site. Our data suggest that YY1 may play an inhibitory role in the transcription of the Muc1 gene.


Assuntos
Regulação da Expressão Gênica/fisiologia , Genes Reguladores/fisiologia , Mucina-1/genética , Transcrição Gênica/fisiologia , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Sítios de Ligação , Cricetinae , Fatores de Ligação de DNA Eritroide Específicos , Humanos , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Fator de Transcrição YY1
11.
Korean Journal of Medicine ; : 352-358, 1997.
Artigo em Coreano | WPRIM (Pacífico Ocidental) | ID: wpr-56217

RESUMO

OBJECTIVE: Stenotrophomonas maltophilia has been emerging as an important nosocomial pathogen in recent years in patients with impaired host- defense mechanism or who has been exposed to large amount of inocula. This organism is usually resistant to multiple (commonly used) antimicrobial agents, particularly to those of the beta-lactam class. To evaluate the clinical feature of Stenotrophomonas maltophilia infection and in vitro anti- microbial susceptibility, we performed a retrospective study. METHODS: We analyzed the result of in vitro antimicrobial susceptibility test for 200 isolates of S. maltophilia and the annual isolation rate during the period between January 1990 and December 1994 in our institution, and performed a retrospective study for the available records of 165 cases among them. The data were obtained with only the first isolation of the organism for each patients. RESULTS: Total of 165 initial isolates, the isolates were from wounds in 50(30.3%), urine in 47(28.5%), the respiratory tract in 37(22.4%), blood in 9(5.5%), bile in 6(3.6%), and miscellaneous sources in 16(9.7%). The 84.2% of isolates were hospital-acquired isolate and 58.3% of these patients had received antecedent antibiotic therapy: polymicrobial growth was demonstrated in 61.9% of the cases. In vitro antimicrobial susceptibiiity test, ofloxacin was active against the isolates in 89.2%, moxalactam in 85.9%, ciprofloxacin in 83.9%, TMP-SMX(trimethoprim-sulfamethoxazole) in 64.2%, As expected, S. maltophilia isolates were, in general, not susceptible to cephalosporins, penicillins. The annual isolation rate at Kyung Hee University hospital was not increased significantly from 1990 to 1994, 19.53 per 10,000 patients dismissals in 1990, 13.56 in 1994. The major underlying diseases of patients were malignancy(17.6%), cerebrovascular disorder(17%), diabetic mellitus(13.3%). Mortality rate is 10.3%. CONCLUSION: S. maltophilia has been emerging as an important nosocomial pathogen in immunocompromised patients, especially those receiving broad-spectrum antimicrobial therapy. And this organism is resistant to multiple antimicrobial agents, particularly to those of the beta-lactam class. When antimicrobial treatment is necessary, the clinician should be guided by results of in vitro susceptibility testing because of the notable in vitro resistance of S. maltophilia to commonly used antibiotics. And when S. maltophilia has been recovered from a patient, wound and contact isolation is warranted.


Assuntos
Humanos , Antibacterianos , Anti-Infecciosos , Bile , Cefalosporinas , Ciprofloxacina , Hospedeiro Imunocomprometido , Mortalidade , Moxalactam , Ofloxacino , Penicilinas , Sistema Respiratório , Estudos Retrospectivos , Stenotrophomonas maltophilia , Stenotrophomonas , Ferimentos e Lesões
12.
Korean Journal of Medicine ; : 360-366, 1997.
Artigo em Coreano | WPRIM (Pacífico Ocidental) | ID: wpr-188075

RESUMO

OBJECTIVES: Aeromonas species is a gram-negative, facultative anaerobe of the family Vibrionaceae. The organism has been recognized as a pathogen associated with illness in human, such as acute gastroenteritis, cellulitis, septicemia, and other rare diseases. METHODS: To evaluate the clinical significance of Aeromonas bacteremia in Korea and it's susceptibility of antibiotics, we evaluated the 17patients with Aeromonas bacteremia. Identification was done by use of API 20E system and antibiotic susceptibility was tested with disk diffusion method. RESULTS: Male to female ratio was 11:6, and mean age was 54.1years(8-85years) old. Liver cirrhosis was the most common underlying disease(10cases of 17patients, 58.8%a). Other underlying diseases were as follows: gallstone in 2cases, cholangiocarcinoma in 2cases, and aplastic anemia in 1case, cerebral infarction in 1case. But one had no underlying disease. So Aeromonas bacteremia were occurred in 14immunocompromised patients(82.3%), and in 10patients with hepatobiliary diseases, A. hydrophila was most commonly isolated(13cases, 764%), and the A sobria(4cases, 23.5%) was infrequently isolated. The overall fatality was 47%, and there had no significant difference in fatality between A. hydrophila and A sobria All Aeromonas species had resistance to ampicillin and carbenicillin. CONCLUSION: Because Aeromonas bacteremia may occur through water-borne route, especially in immunocompromised host. We should pay attention to immunocompromised patients, espacially having hepatobiliary disease.


Assuntos
Feminino , Humanos , Masculino , Aeromonas , Ampicilina , Anemia Aplástica , Antibacterianos , Bacteriemia , Carbenicilina , Celulite (Flegmão) , Infarto Cerebral , Colangiocarcinoma , Difusão , Cálculos Biliares , Gastroenterite , Hospedeiro Imunocomprometido , Coreia (Geográfico) , Cirrose Hepática , Doenças Raras , Sepse , Vibrionaceae
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA