Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Cell Mol Biol (Noisy-le-grand) ; 60(1): 45-52, 2014 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-24857383

RESUMO

The Vacuolar H+-ATPases (V-ATPases), a multi-subunits nanomotor present in all eukaryotic cells resides in the endomembranes of exocytotic and endocytotic pathways. Plasmalemmal V-ATPases have been shown to be involved in tumor cell metastasis. Pigment epithelium-derived factor (PEDF), a potent endogenous inhibitor of angiogenesis, is down-regulated in prostate cancer cells. We hypothesized that the transduction of PEDF in prostate cancer cells will down-regulate V-ATPase function; that in turn will decrease the expression of the V-ATPase accessory protein ATP6ap2 and a-subunit isoforms that target V-ATPase to the cell surface. To test these hypotheses, we used the human androgen-sensitive prostate cancer cells LNCaP, and its castration-refractory-derivative CL1 that were engineered to stably co-express the DsRed Express Fluorescent Protein with or without PEDF. To determine if PEDF down-regulates the function of V-ATPase, we measured the rate of proton fluxes (JH+) of the cytosolic and endosome/lysosome compartments. The mRNA levels for subunit-a isoforms and the ATP6ap2 were measured using quantitative reverse transcription-PCR. The results showed that PEDF expression decreased the rate of JH+ in metastatic CL1 cells without affecting JH+ in non-metastatic LNCaP cells, when studying pH(cyt). Interestingly, PEDF did not affect JH+ in endosomes/lysosomes either in metastatic cells or in non-metastatic cells. We also showed that PEDF significantly decreases the levels of a4 isoform and ATP6ap2 in metastatic CL1 cells, without affecting the levels of a4 isoform in the non-metastatic LNCaP cells. These data identify PEDF as a novel regulator of V-ATPase suggesting a new way by which PEDF may inhibit prostate tumor growth.


Assuntos
Regulação para Baixo , Proteínas do Olho/fisiologia , Neovascularização Patológica/genética , Fatores de Crescimento Neural/fisiologia , Neoplasias da Próstata/genética , Serpinas/fisiologia , ATPases Vacuolares Próton-Translocadoras/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Proteínas do Olho/metabolismo , Humanos , Masculino , Fatores de Crescimento Neural/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Serpinas/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
2.
Clin Microbiol Infect ; 17(5): 697-703, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20636426

RESUMO

Mycobacterium avium ssp. hominissuis, hereafter referred to as M. avium, forms biofilm, a property that, in mice, is associated with lung infection via aerosol. As M. avium might co-inhabit the respiratory tract with other pathogens, treatment of the co-pathogen-associated infections, such as in bronchiectasis, would expose M. avium to therapeutic compounds that may have their origin in other organisms sharing the natural environments. Incubation of M. avium with two compounds produced by environmental organisms, streptomycin and tetracycline, in vitro at subinhibitory concentrations increased biofilm formation in a number of M. avium strains, although exposure to ampicillin, moxifloxacin, rifampin and trimethoprim-sulphamethoxazole had no effect on biofilm formation. No selection of genotypically resistant clones was observed. Although incubation of bacteria in the presence of streptomycin upregulates the expression of biofilm-associated genes, the response to the antibiotics had no association with the expression of a regulator (LysR) linked to the formation of biofilm in M. avium. Biofilms are composed of planktonic and sessile bacteria. Whereas planktonic M. avium is susceptible to clarithromycin and ethambutol (clinically used antimicrobials), sessile bacteria are at least three-fold to four-fold more resistant to antibiotics. The sessile phenotype, however, is reversible, and no selection of resistant clones was observed. Mice infected through the airway with both phenotypes were infected with a similar number of bacteria, demonstrating no phenotype advantage. M. avium biofilm formation is enhanced by commonly used compounds and, in the sessile bacterial phenotype, is resistant to clarithromycin and ethambutol, in a reversible manner.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Mycobacterium avium/efeitos dos fármacos , Mycobacterium avium/fisiologia , Animais , Bronquiectasia/tratamento farmacológico , Bronquiectasia/microbiologia , Proteínas de Ligação a DNA , Farmacorresistência Bacteriana , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Mycobacterium avium/genética , Fenótipo , Reação em Cadeia da Polimerase
3.
J Fish Dis ; 30(10): 587-600, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17850575

RESUMO

The natural route by which fish become infected with mycobacteria is unknown. Danio rerio (Hamilton) were exposed by bath immersion and intubation to Mycobacterium marinum and Mycobacterium peregrinum isolates obtained from diseased zebrafish. Exposed fish were collected over the course of 8 weeks and examined for the presence of mycobacteriosis. Mycobacteria were consistently cultured from the intestines, and often from the livers and spleens of fish exposed by both methods. Mycobacteria were not observed in the gills. Histological analysis revealed that fish infected with M. marinum often developed granulomas accompanied by clinical signs of mycobacteriosis, while infection with M. peregrinum infrequently led to clinical signs of disease. Passage of the bacteria through environmental amoebae (Acanthamoeba castellani) was associated with increased growth of M. peregrinum over the course of 8 weeks, when compared to infection with the bacteria not passed through amoebae. The results provide evidence that zebrafish acquire mycobacteria primarily through the intestinal tract, resulting in mycobacterial dissemination.


Assuntos
Doenças dos Peixes/microbiologia , Intestinos/microbiologia , Infecções por Mycobacterium/veterinária , Mycobacterium/patogenicidade , Peixe-Zebra , Acanthamoeba castellanii/microbiologia , Animais , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Doenças dos Peixes/patologia , Fígado/microbiologia , Infecções por Mycobacterium/microbiologia , Infecções por Mycobacterium/patologia , Mycobacterium marinum/patogenicidade , Baço/microbiologia , Fatores de Tempo , Virulência , Microbiologia da Água
4.
Clin Exp Immunol ; 136(3): 490-500, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15147351

RESUMO

The pathogenic mycobacteria are an insidious group of bacterial pathogens that cause the deaths of millions of people every year. One of the reasons these pathogens are so successful is that they are able to invade and replicate within host macrophages, one of the first lines of defence against intruding pathogens. In contrast, non-pathogenic mycobacteria, such as Mycobacterium smegmatis are killed rapidly by macrophages. In order to understand better the series of events that allow pathogenic mycobacteria to survive and replicate within macrophages, while the non-pathogenic mycobacteria are killed rapidly, we inoculated the human monocytic cell line U937 with pathogenic (M. tuberculosis and M. avium) and non-pathogenic (M. smegmatis) mycobacteria and monitored the expression of over 3500 genes at 4, 12 and 24 h post-inoculation using a commercially available gene array system. We observed multiple differences in the gene expression patterns of monocytes infected with pathogenic and non-pathogenic mycobacteria including genes involved in cytokine, lymphokine and chemokine production, adhesion, apoptosis, signal transduction, transcription, protein cleavage, actin polymerization and growth. We also observed differences in gene expression profiles in monocytes infected with M. tuberculosis or M. avium, indicating that there are differences in the host pathogen interactions of mononuclear phagocytes infected with different pathogenic mycobacterial species. These results increase the understanding of the mechanisms used by pathogenic mycobacteria to cause disease, the host response to these organisms, and provide new insights for antimycobacterial intervention strategies.


Assuntos
Macrófagos/imunologia , Macrófagos/microbiologia , Infecções por Mycobacterium/imunologia , Mycobacterium/patogenicidade , Regulação da Expressão Gênica , Humanos , Ativação de Macrófagos/genética , Mycobacterium avium/patogenicidade , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células U937
5.
Infect Immun ; 69(12): 7242-9, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11705893

RESUMO

Mycobacterium avium and Mycobacterium intracellulare are closely related organisms and comprise the Mycobacterium avium complex. These organisms share many common characteristics, including the ability to cause life-threatening respiratory infections in people with underlying lung pathology or immunological defects and occasionally in those with no known predisposing conditions. However, the ability to invade the mucosa of the gastrointestinal tract and cause disseminated disease in AIDS patients has not been epidemiologically linked to M. intracellulare and appears to be unique to M. avium. We compared the abilities of M. avium and M. intracellulare to tolerate the acidic conditions of the stomach, to resist the membrane-disrupting activity of cationic peptides, and to invade intestinal epithelial cells in vitro and in vivo. We observed that M. avium and M. intracellulare were both tolerant to the acidic conditions encountered in the stomach and resistant to cationic peptides. However, when strains of M. avium and M. intracellulare were examined for their ability to enter cultured human intestinal cells or mouse intestinal mucosa, we observed that M. avium could invade more efficiently than M. intracellulare. To elucidate the basis of this pathogenic difference and identify genes involved in the invasion of the intestinal mucosa, we performed chromosomal DNA subtractive hybridization using M. avium and M. intracellulare chromosomal DNAs. In all, 21 genes that were present in M. avium but absent in M. intracellulare were identified, including some that may be associated with the ability of M. avium to invade the intestinal mucosa.


Assuntos
Complexo Mycobacterium avium/genética , Complexo Mycobacterium avium/patogenicidade , Tuberculose Gastrointestinal/etiologia , Animais , Antibacterianos , Células Cultivadas , Farmacorresistência Bacteriana , Células Epiteliais/microbiologia , Feminino , Genes Bacterianos , Genoma Bacteriano , Ácido Clorídrico/farmacologia , Íleo/microbiologia , Íleo/ultraestrutura , Mucosa Intestinal/microbiologia , Camundongos , Dados de Sequência Molecular , Hibridização de Ácido Nucleico/métodos , Fenótipo , Polimixina B/farmacologia
6.
Antimicrob Agents Chemother ; 45(8): 2210-4, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11451676

RESUMO

The activity of telithromycin, a new ketolide, was evaluated in vitro and in vivo against Mycobacterium avium complex (MAC) strains. The MIC of telithromycin for several M. avium isolates obtained from the blood of AIDS patients ranged from 16 to >128 microg/ml (MIC at which 90% of isolates are inhibited, >128 microg/ml), and the compound did show activity in the macrophage system at concentrations greater than 8 or 16 microg/ml, but this was dependent on the MAC strain used. Telithromycin was then administered to mice infected with MAC strain 101 for 4 weeks at doses of 100, 200, or 400 mg/kg of body weight/day. Treatment with 100 and 200 mg/kg/day was bacteriostatic, but at 400 mg/kg/day telithromycin was bactericidal for MAC strains. The frequency of the emergence of resistance to telithromycin was low despite prolonged usage (12 weeks). This study demonstrates that telithromycin is active in vivo against MAC and warrants further evaluation.


Assuntos
Antibacterianos/farmacologia , Resistência a Medicamentos , Cetolídeos , Macrolídeos , Macrófagos/microbiologia , Complexo Mycobacterium avium/efeitos dos fármacos , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Complexo Mycobacterium avium/isolamento & purificação , Infecção por Mycobacterium avium-intracellulare/sangue
7.
Clin Immunol ; 99(3): 378-86, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11358435

RESUMO

Mycobacterial infection is associated with granuloma formation in which the presence of apoptosis has been recognized. The role of CD4+ T and CD8+ T cells in host protection against mycobacterial infections has been demonstrated. Previous studies, however, have shown that CD8+ T cells have a limited role in host defense against Mycobacterium avium infection, and we hypothesize that M. avium infection could lead to T cell apoptosis. To investigate this hypothesis, C57BL/6 mice were infected with M. avium strain 101, and the rate of apoptosis of splenic lymphocytes cultured ex vivo with peritoneal macrophages was determined and compared with that of controls. When exposed to infected macrophages ex vivo, splenic lymphocytes from M. avium-infected mice underwent apoptosis, as determined by the TUNEL assay. This increased T cell apoptosis above the control level was observed after 3 weeks but not after only 1 week of infection in mice. No splenic T cell apoptosis was observed when lymphocytes from Mycobacterium smegmatis-infected mice were cultured in the presence of M. smegmatis-infected peritoneal macrophages. Likewise, macrophages infected in vitro with heat-killed M. avium did not trigger T cell apoptosis. Culture of macrophages in different chamber from lymphocytes, separated by a transwell membrane, was not associated with increase of apoptosis compared with uninfected control, suggesting a requirement for direct cell-cell interactions to trigger lymphocyte apoptosis. Using a double staining TUNEL followed by anti-mouse CD4 or anti-mouse CD8 monoclonal antibodies, it was observed that only CD8+ T cells but not CD4+ T cells underwent apoptosis at 3 weeks of infection. In conclusion, M. avium infection in C57/BL6 mice for 3 weeks renders CD8+ T cells prone to apoptosis when exposed ex vivo to macrophages infected with M. avium.


Assuntos
Apoptose , Linfócitos T CD8-Positivos/fisiologia , Macrófagos/fisiologia , Mycobacterium avium , Tuberculose/imunologia , Animais , Comunicação Celular , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Coelhos , Ratos
8.
FEMS Microbiol Lett ; 197(2): 151-7, 2001 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-11313128

RESUMO

Mycobacterium avium is both a pathogen that infects several hosts such as humans, pigs, and birds, as well as a microorganism that is encountered in environmental sources (soil and water). Protein secretion by the bacterium is likely to influence its ability to overcome adverse and competitive conditions both within or outside the host. Using a combination of cloning and information available in the databank, we characterized the secA gene from M. avium, encoding for a major preprotein translocase subunit associated with the secretion system of prokaryotics. In addition, we cloned the secA promoter sequence in a reporter construct upstream of a promoterless gfp. It was determined that the secA of M. avium shares large homology with the secA of Mycobacterium tuberculosis but not with secA of Mycobacterium leprae. secA expression was determined to be greater at logarithmic growth phase although it was also expressed at low levels during the stationary phase. secA expression was also observed when the bacteria were incubated in water as well as within human monocyte-derived macrophages and in conditions that are associated with biofilm formation. Future evaluation of the sec pathway in M. avium might provide important information about secreted proteins that are required for survival in different environments.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Proteínas de Escherichia coli , Proteínas de Membrana Transportadoras , Mycobacterium avium/genética , Adenosina Trifosfatases/química , Proteínas de Bactérias/química , Sequência de Bases , Biofilmes/crescimento & desenvolvimento , Proteínas de Transporte/química , Clonagem Molecular , Meios de Cultura , Humanos , Macrófagos/microbiologia , Dados de Sequência Molecular , Mycobacterium avium/química , Mycobacterium avium/crescimento & desenvolvimento , Regiões Promotoras Genéticas , Canais de Translocação SEC , Proteínas SecA , Água
9.
Microbes Infect ; 3(1): 37-42, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11226852

RESUMO

Mycobacteria are intracellular pathogens capable of invading mononuclear phagocytes, mucosal epithelial cells (including M cells) and Schwann cells. To enter cells, mycobacteria have been shown to interact with several molecules on macrophage and epithelial cell surfaces. This suggests adaptation to the host environment. In this review we address the strategies used by pathogenic mycobacteria to gain access to the intracellular environment.


Assuntos
Mucosa/microbiologia , Mycobacterium/fisiologia , Animais , Células Epiteliais/microbiologia , Humanos , Mucosa Intestinal/microbiologia , Macrófagos/microbiologia , Mucosa/citologia , Mucosa/metabolismo , Mycobacterium/metabolismo , Fagócitos/microbiologia , Mucosa Respiratória/microbiologia , Células de Schwann/microbiologia
10.
Infect Immun ; 69(1): 508-17, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11119544

RESUMO

Successful parasitism of host cells by intracellular pathogens involves adherence, entry, survival, intracellular replication, and cell-to-cell spread. Our laboratory has been examining the role of early events, adherence and entry, in the pathogenesis of the facultative intracellular pathogen Legionella pneumophila. Currently, the mechanisms used by L. pneumophila to gain access to the intracellular environment are not well understood. We have recently isolated three loci, designated enh1, enh2, and enh3, that are involved in the ability of L. pneumophila to enter host cells. One of the genes present in the enh1 locus, rtxA, is homologous to repeats in structural toxin genes (RTX) found in many bacterial pathogens. RTX proteins from other bacterial species are commonly cytotoxic, and some of them have been shown to bind to beta(2) integrin receptors. In the current study, we demonstrate that the L. pneumophila rtxA gene is involved in adherence, cytotoxicity, and pore formation in addition to its role in entry. Furthermore, an rtxA mutant does not replicate as well as wild-type L. pneumophila in monocytes and is less virulent in mice. Thus, we conclude that the entry gene rtxA is an important virulence determinant in L. pneumophila and is likely to be critical for the production of Legionnaires' disease in humans.


Assuntos
Toxinas Bacterianas/genética , Genes Bacterianos , Legionella pneumophila/genética , Animais , Aderência Bacteriana , Células Epiteliais/microbiologia , Legionella pneumophila/patogenicidade , Pulmão/microbiologia , Camundongos , Monócitos/microbiologia , Virulência
11.
Antimicrob Agents Chemother ; 45(1): 217-22, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11120969

RESUMO

Moxifloxacin activity against Mycobacterium avium complex (MAC) was evaluated in vitro against 25 strains. The MIC was determined to range from 0.125 to 2.0 microg/ml. In addition, U937 macrophage monolayers infected with MAC strain 101 (serovar 1) were treated with moxifloxacin (0.25 to 8 microg/ml) daily, and the number of intracellular bacteria was quantitated after 4 days. Moxifloxacin showed inhibitory activity at 0.5 microg/ml and higher. To assess the activity of moxifloxacin containing regimens in vivo, we infected C57BL bg(+)/bg(+) mice with 3 x 10(7) MAC strain 101 bacteria intravenously. One week later treatment was begun with the following: (i) moxifloxacin (50 mg/kg/day or 100 mg/kg/day), ethambutol (100 mg/kg/day), or a combination of moxifloxacin and ethambutol; or (ii) moxifloxacin (100 mg/kg/day), azithromycin (200 mg/kg/day), or rifabutin (40 mg/kg/day) as oral monotherapy; or (iii) all permutations of two-drug therapy or all three drugs in combination. All groups contained at least 14 animals, and the control group received the drug vehicle. After 4 weeks, quantitative blood cultures were obtained and the number of bacteria in liver and spleen was quantitated. Moxifloxacin, ethambutol, and azithromycin were active as single agents in liver, spleen, and blood. Rifabutin showed inhibitory activity only in the blood. Two-drug combinations containing azithromycin were no more active than azithromycin alone. Similarly, the three-drug combination was not more active than azithromycin alone in the spleen. Rifabutin did not add to the activity of any other single agent or two-drug combination. Moxifloxacin at both concentrations in combination with ethambutol was significantly more active than each drug alone.


Assuntos
Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia , Antituberculosos/farmacologia , Compostos Aza , Azitromicina/farmacologia , Etambutol/farmacologia , Fluoroquinolonas , Complexo Mycobacterium avium/efeitos dos fármacos , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Quinolinas , Rifabutina/farmacologia , Animais , Antibacterianos/uso terapêutico , Anti-Infecciosos/uso terapêutico , Antituberculosos/uso terapêutico , Azitromicina/uso terapêutico , Interações Medicamentosas , Quimioterapia Combinada , Etambutol/uso terapêutico , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Moxifloxacina , Infecção por Mycobacterium avium-intracellulare/microbiologia , Rifabutina/uso terapêutico
12.
Infect Immun ; 68(10): 5824-9, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10992491

RESUMO

The mechanism by which mycobacteria elicit class I-restricted T-cell responses remains undefined because these organisms have been shown to reside exclusively within membrane-bound vesicles in macrophages (Mphi), their primary host cells. We studied the interaction of M. avium with dendritic cells (DC) because they are the most potent antigen-presenting cells and are abundant at M. avium infection sites. We observed that both DC and Mphi, generated from human peripheral blood monocytes by short-term culture, internalized M. avium. The onset of programmed cell death and the percentage of apoptotic cells in infected DC and Mphi were comparable. However, following infection, DC secreted significantly larger amounts of interleukin-12, but not interleukin-1beta, than infected autologous Mphi. Further analysis of infected cells showed that while phagosomes failed to acidify in both M. avium-infected DC and Mphi, bacilli grew more slowly in DC. Electron microscopy studies revealed that M. avium resided within endocytic vacuoles in both cell types. The vacuolar membrane surrounding some bacilli in approximately 10% of the vacuoles in DC possessed several breaks. The importance of this finding will have to be addressed in future studies.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Monócitos/citologia , Complexo Mycobacterium avium/fisiologia , Apoptose , Células Cultivadas , Células Dendríticas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Interleucina-12/biossíntese , Lisossomos/fisiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Microscopia Eletrônica , Complexo Mycobacterium avium/imunologia , Fagossomos/fisiologia , Vacúolos/microbiologia
13.
Antimicrob Agents Chemother ; 44(10): 2619-22, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10991834

RESUMO

Resistance to clarithromycin in breakthrough Mycobacterium avium complex (MAC) isolates typically occurs 3 to 4 months after the initiation of monotherapy in bacteremic AIDS patients. It has been suggested that continuation of clarithromycin therapy still results in clinical and microbiological improvement. To study this paradox, C57BL/6 beige mice were infected with a clarithromycin-resistant (MIC, > or =128 microg/ml) strain of MAC 101 (CLA-R MAC 101) and treated with 200 mg of clarithromycin per kg of body weight/day alone or in combination with ethambutol (100 mg/kg/day) for 2 weeks. Mice infected with a clarithromycin-susceptible strain of MAC 101 had bacterial loads reduced by 90% in the liver and 91% in the spleen (P<0.05, compared with the control). Clarithromycin treatment of CLA-R MAC 101 resulted in a 65% reduction of bacterial loads in the liver (P = 0.009) and a 71% reduction in the spleen (P = 0.009), compared with the results for the untreated control. CLA-R MAC 101 and MAC 101 (isogenic strains) had comparable growth rates in murine tissue, ruling out a loss of virulence of CLA-R MAC 101. Strains of MAC currently defined as macrolide resistant may still respond to treatment with an agent such as clarithromycin within infected tissues.


Assuntos
Antibacterianos/farmacologia , Claritromicina/farmacologia , Complexo Mycobacterium avium/efeitos dos fármacos , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Infecções Oportunistas Relacionadas com a AIDS/microbiologia , Animais , Antibacterianos/uso terapêutico , Antituberculosos/uso terapêutico , Claritromicina/uso terapêutico , Resistência Microbiana a Medicamentos , Etambutol/uso terapêutico , Humanos , Técnicas In Vitro , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Complexo Mycobacterium avium/patogenicidade , Infecção por Mycobacterium avium-intracellulare/microbiologia , RNA Ribossômico 23S/metabolismo
15.
Infect Immun ; 68(5): 2979-84, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10768998

RESUMO

Central nervous system (CNS) infections caused by nontuberculous mycobacteria have been described previously, especially in patients with AIDS. To investigate specific aspects of the pathogenesis of this entity, C57BL bg(+)/bg(-) mice were infected intravenously with Mycobacterium avium, and cultures of blood and brain as well as histopathology examination of brain tissue were carried out at several time points up to 6 months after infection. Low-grade inflammatory changes with small aggregates of lymphocytes and macrophages as well as perivascular cuffing were seen early in the infection. A small number of bacteria could be observed in the parenchyma of the choroid plexus. Six months after infection, numerous bacteria were present within the foamy macrophage of the granulomatous lesions along the ventricle and meninges. None of the mice developed clinical signs of meningitis or encephalitis or even died spontaneously during the period of observation. Use of CD18(-/-) knockout mice indicated that transport of the bacterium within neutrophils or monocytes into the brain is unlikely. Mild chronic CNS infection developed in the mice during sustained systemic M. avium infection, similar to what has been reported in most human cases.


Assuntos
Encéfalo/microbiologia , Doenças do Sistema Nervoso Central/microbiologia , Complexo Mycobacterium avium/isolamento & purificação , Tuberculose/microbiologia , Animais , Encéfalo/patologia , Antígenos CD18/genética , Doenças do Sistema Nervoso Central/patologia , Doença Crônica , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tuberculose/patologia
16.
Microb Pathog ; 28(3): 135-44, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10702355

RESUMO

Mycobacterium avium is a common pathogen in AIDS patients. The extracellular environment within the granuloma shown to support mycobacterial growth is in the caseous fluid. Previous work demonstrated that the presence of human epidermal growth factor (EGF), which is found in the tissue of chronic granulomous lesions, increases the growth rate of M. avium and Mycobacterium tuberculosis. Previously, a protein capable of binding recombinant human EGF (rEGF) in a western blot was identified with homology to glyceraldehyde-3-phosphate dehydrogenase (GAP) in both M. avium and M. tuberculosis but not Mycobacterium smegmatis. Surface GAPs have been identified in group A Streptococcus, enteropathogenic Escherichia coli, Candida albicans and Schistosoma mansoni. We have cloned the gap gene of M. avium. M. avium GAP has high homology with M. tuberculosis GAP. The protein was also expressed in M. smegmatis, conveying the ability to bind rEGF, but no growth increase was observed in 7H9 broth in the presence of rEGF up to 500 ng/ml. Only one copy of the GAP gene was identified in M. avium These results contribute to the understanding of M. avium pathogenesis by characterizing its interaction with a host protein present in the site of infection.


Assuntos
Receptores ErbB/genética , Genes Bacterianos , Gliceraldeído-3-Fosfato Desidrogenases/genética , Mycobacterium avium/genética , Sequência de Aminoácidos , Sequência de Bases , Southern Blotting , Clonagem Molecular , Primers do DNA/genética , DNA Bacteriano/análise , Biblioteca Genômica , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Mycobacterium avium/enzimologia , Proteínas Recombinantes/genética , Análise de Sequência
18.
Arch Immunol Ther Exp (Warsz) ; 48(6): 521-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11197607

RESUMO

Infections caused by Mycobacterium avium are common in AIDS patients and patients with chronic lung diseases. The bacterium can be acquired both through the intestinal route and respiratory route. M. avium is capable of invading mucosal epithelial cells and translocating across the mucosa. The bacterium can infect macrophages, interfering with several functions of the host cell. The host defense against M. avium is primarily dependent on CD4+ T lymphocytes and natural killer cells. Activated macrophages can inhibit or kill intracellular bacteria by mechanisms that are currently unknown, but M. avium can invade resting macrophages and suppress key aspects of their function by triggering the release of transforming growth factor beta and interleukin 10. Co-infection with HIV-1 appears to be mutually beneficial, with both organisms growing faster.


Assuntos
Complexo Mycobacterium avium/patogenicidade , Infecção por Mycobacterium avium-intracellulare/etiologia , Infecções Oportunistas Relacionadas com a AIDS/etiologia , Animais , Humanos , Técnicas In Vitro , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium avium/patogenicidade , Infecção por Mycobacterium avium-intracellulare/complicações , Infecção por Mycobacterium avium-intracellulare/imunologia
19.
Cell Microbiol ; 2(6): 561-8, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11207608

RESUMO

Mycobacterium avium is a common pathogen in AIDS patients that is primarily (but not exclusively) acquired through the gastrointestinal tract, leading to the development of bacteraemia and disseminated disease. To cause infection through the gut, binding and invasion of the intestinal epithelial barrier are required. To characterize this process further, we determined the cell surface(s) (basolateral vs. apical membrane) that M. avium interacts with in intestinal mucosal cells in vitro. The level of binding and invasion of both HT-29 and Caco-2 intestinal cell monolayers by M. avium were similar when the assay was performed with control medium in the presence of Ca2+ (when only the apical surface was exposed), with Ca2+-depleted medium or with Ca2+-depleted medium + 1 mM EGTA (exposure of both apical and basolateral membranes), suggesting that the bacterium enters the apical surface of the epithelial lining. These observations were confirmed by assays in a transwell system and by using fluorescent microscopy. Real-time video microscopy showed that M. avium entry was not associated with membrane ruffling and the use of pharmacological inhibitors of the small GTPases demonstrated that M. avium invasion is dependent on the activation of the small GTPases Rho, but not on Rac or Cdc42. Passage of M. avium through HT-29 cells led to a phenotypic change (intracellular growth; IG) that was associated with a significantly greater (between five- and ninefold) ability to bind to and invade new monolayers of epithelial cells or macrophages when compared with the invasion by M. avium grown on agar (extracellular growth; EG). IG phenotype invasion of HT-29 cells also takes place only by the apical surface. M. avium enters intestinal epithelial cells by the apical surface and, once within the cells, changes phenotype, becoming more invasive towards both macrophages and other epithelial cells.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/microbiologia , Membrana Celular/microbiologia , Células Epiteliais/microbiologia , Mucosa Intestinal/citologia , Complexo Mycobacterium avium/patogenicidade , Aderência Bacteriana , Células CACO-2/microbiologia , Linhagem Celular , Humanos , Mucosa Intestinal/microbiologia , Microscopia Eletrônica , Microscopia de Vídeo , Complexo Mycobacterium avium/fisiologia , Infecção por Mycobacterium avium-intracellulare/microbiologia , Proteínas rho de Ligação ao GTP/metabolismo
20.
Infect Immun ; 67(10): 5069-75, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10496879

RESUMO

Mycobacterium avium is an opportunistic pathogen in AIDS patients, who acquire the infection mainly through the gastrointestinal tract. Previous studies in vitro have shown that M. avium invades epithelial cells of both intestinal and laryngeal origin. In addition, M. avium enters the intestinal mucosa of healthy mice. Because M. avium invasion of the intestinal mucosa in vivo initially is not accompanied by significant influx of inflammatory cells, we sought to determine whether M. avium would trigger chemokine release upon entry into epithelial cells by using HT-29 intestinal and HEp-2 laryngeal epithelial cell lines. Chemokine synthesis was measured both by the presence of specific mRNA and protein secretion in the cell culture supernatant as determined by enzyme-linked immunosorbent assay. Infection of HT-29 intestinal cells with M. avium did not induce the release of interleukin-8 (IL-8) or RANTES for up to 7 days postinfection. However, infection of HEp-2 cells resulted in the release of IL-8 and RANTES at 72 h. Similar findings were observed with other AIDS M. avium isolates belonging to different serovars. Secretion of IL-8 by HEp-2 cells was dependent upon bacterial uptake. In addition, prior infection with M. avium suppressed IL-8 production by HT-29 cells infected with Salmonella typhimurium. Our results suggest that M. avium infection of epithelial cells is associated with a delay in IL-8 and RANTES production which, in the case of HT-29, is prolonged up to 1 week. These findings may explain the weak inflammatory response after intestinal mucosa invasion in mice and are probably related with the ability of the bacterium to evade the host's immune response.


Assuntos
Quimiocina CCL5/biossíntese , Interleucina-8/biossíntese , Mycobacterium avium/fisiologia , Quimiocina CCL5/genética , Células Epiteliais/microbiologia , Células HT29 , Humanos , Interleucina-8/genética , RNA Mensageiro/análise , Salmonella/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA