Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(8): e0287997, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37590269

RESUMO

We identified a fragment (Domain 3-D3) of the immunodominant sporozoite surface glycoprotein of the zoonotic parasite Cryptosporidium gp900, which is absent C. hominis and C. parvum anthroponosum. The fragment is highly antigenic and is able to effectively differentiate between zoonotic C. parvum and species/genotypes that infect preferentially humans. D3 detection provides a serological tool to determine whether the source of human cryptosporidiosis is of animal or human origin. We demonstrate this in experimentally challenged piglets, mice, rats, and alpaca. We speculate that the absence of this fragment from the C. hominis and C. parvum anthroponosum gp900 protein may play a key role in their host restriction.


Assuntos
Camelídeos Americanos , Criptosporidiose , Cryptosporidium parvum , Cryptosporidium , Humanos , Animais , Camundongos , Ratos , Suínos , Glicoproteínas , Glicoproteínas de Membrana , Propionibacterium acnes
2.
Microbes Infect ; 20(4): 267-270, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29357304

RESUMO

Cryptosporidium hominis and Cryptosporidium parvum are the major Cryptosporidium species that infect humans. Earlier studies in gnotobiotic piglets, model susceptible to both, showed that piglets recovered from infection with C. hominis were fully protected against challenge with same species but incompletely protected against C. parvum challenge. In the present study, piglets were infected with C. parvum first, and after recovery were re-challenged with C. parvum or C. hominis. Again, full protection was only observed when piglets were challenged with the homologous parasite strain. Although the two species are genetically/antigenically almost identical, they do not confer complete protection against each other.


Assuntos
Criptosporidiose/imunologia , Cryptosporidium/imunologia , Suínos , Animais , Criptosporidiose/parasitologia , Criptosporidiose/prevenção & controle , Cryptosporidium parvum/imunologia , DNA de Protozoário/análise , Fezes/parasitologia , Vida Livre de Germes , Humanos , Intestinos/parasitologia , Especificidade da Espécie , Suínos/imunologia , Suínos/parasitologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-28095366

RESUMO

Human cryptosporidiosis, caused primarily by Cryptosporidium hominis and a subset of Cryptosporidium parvum, is a major cause of moderate-to-severe diarrhea in children under 5 years of age in developing countries and can lead to nutritional stunting and death. Cryptosporidiosis is particularly severe and potentially lethal in immunocompromised hosts. Biological and technical challenges have impeded traditional vaccinology approaches to identify novel targets for the development of vaccines against C. hominis, the predominant species associated with human disease. We deemed that the existence of genomic resources for multiple species in the genus, including a much-improved genome assembly and annotation for C. hominis, makes a reverse vaccinology approach feasible. To this end, we sought to generate a searchable online resource, termed C. hominis gene catalog, which registers all C. hominis genes and their properties relevant for the identification and prioritization of candidate vaccine antigens, including physical attributes, properties related to antigenic potential and expression data. Using bioinformatic approaches, we identified ∼400 C. hominis genes containing properties typical of surface-exposed antigens, such as predicted glycosylphosphatidylinositol (GPI)-anchor motifs, multiple transmembrane motifs and/or signal peptides targeting the encoded protein to the secretory pathway. This set can be narrowed further, e.g. by focusing on potential GPI-anchored proteins lacking homologs in the human genome, but with homologs in the other Cryptosporidium species for which genomic data are available, and with low amino acid polymorphism. Additional selection criteria related to recombinant expression and purification include minimizing predicted post-translation modifications and potential disulfide bonds. Forty proteins satisfying these criteria were selected from 3745 proteins in the updated C. hominis annotation. The immunogenic potential of a few of these is currently being tested.Database URL: http://cryptogc.igs.umaryland.edu.


Assuntos
Antígenos de Protozoários/genética , Criptosporidiose/genética , Cryptosporidium/genética , Genoma de Protozoário , Vacinas Protozoárias/genética , Animais , Criptosporidiose/prevenção & controle , Bases de Dados Genéticas , Humanos
4.
Infect Immun ; 83(1): 286-91, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368111

RESUMO

Hemolytic-uremic syndrome (HUS), caused by Shiga toxin (Stx)-producing Escherichia coli (STEC), remains untreatable. Production of human monoclonal antibodies against Stx, which are highly effective in preventing Stx sequelae in animal models, is languishing due to cost and logistics. We reported previously that the production and evaluation of a camelid heavy-chain-only VH domain (VHH)-based neutralizing agent (VNA) targeting Stx1 and Stx2 (VNA-Stx) protected mice from Stx1 and Stx2 intoxication. Here we report that a single intramuscular (i.m.) injection of a nonreplicating adenovirus (Ad) vector carrying a secretory transgene of VNA-Stx (Ad/VNA-Stx) protected mice challenged with Stx2 and protected gnotobiotic piglets infected with STEC from fatal systemic intoxication. One i.m. dose of Ad/VNA-Stx prevented fatal central nervous system (CNS) symptoms in 9 of 10 animals when it was given to piglets 24 h after bacterial challenge and in 5 of 9 animals when it was given 48 h after bacterial challenge, just prior to the onset of CNS symptoms. All 6 placebo animals died or were euthanized with severe CNS symptoms. Ad/VNA-Stx treatment had no impact on diarrhea. In conclusion, Ad/VNA-Stx treatment is effective in protecting piglets from fatal Stx2-mediated CNS complications following STEC challenge. With a low production cost and further development, this could presumably be an effective treatment for patients with HUS and/or individuals at high risk of developing HUS due to exposure to STEC.


Assuntos
Adenovírus Humanos/genética , Anticorpos Neutralizantes/uso terapêutico , Infecções por Escherichia coli/tratamento farmacológico , Escherichia coli O157/imunologia , Síndrome Hemolítico-Urêmica/tratamento farmacológico , Toxina Shiga I/antagonistas & inibidores , Toxina Shiga II/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/genética , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/genética , Feminino , Vetores Genéticos , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/microbiologia , Injeções Intramusculares , Camundongos , Toxina Shiga I/imunologia , Toxina Shiga II/imunologia , Análise de Sobrevida , Suínos , Fatores de Tempo
5.
Vaccine ; 29(27): 4431-4, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21549788

RESUMO

Leptospira interrogans serovar Copenhageni causes pulmonary hemorrhages with respiratory failure, a major cause of death in leptospirosis patients. Protective immunity to Leptospira is known to correlate with the production of leptospiral lipopolysaccharide (L-LPS)-specific agglutinating antibodies. We generated L-LPS-specific mouse monoclonal antibodies (MAbs) and investigated if these MAbs can protect guinea pigs against fatal pulmonary hemorrhages caused by serovar Copenhageni. The MAbs L8H4 and L9B11 against 22kDa L-LPS agglutinated leptospires and completely protected guinea pigs from the development of fatal pulmonary hemorrhages by serovar Copenhageni, whereas the MAb L4C1 against 8kDa L-LPS neither agglutinated the bacteria nor protected the animals against the fatal pulmonary hemorrhages.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Hemorragia/prevenção & controle , Imunização Passiva , Leptospira interrogans/imunologia , Leptospirose/prevenção & controle , Lipopolissacarídeos/imunologia , Pneumopatias/prevenção & controle , Aglutinação/imunologia , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Modelos Animais de Doenças , Cobaias , Hemorragia/imunologia , Hemorragia/microbiologia , Humanos , Immunoblotting , Leptospira interrogans/classificação , Leptospira interrogans/patogenicidade , Leptospirose/imunologia , Leptospirose/microbiologia , Pneumopatias/imunologia , Pneumopatias/microbiologia , Masculino , Camundongos , Sorotipagem , Doença de Weil
6.
J Infect Dis ; 201(7): 1081-3, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20196656

RESUMO

Escherichia coli strains that produce Shiga toxin 2 (Stx2) are isolated from hemolytic-uremic syndrome (HUS) cases more frequently than are strains that produce both Shiga toxin 1 (Stx1) and Stx2, whereas strains that produce only Stx1 are rarely isolated from HUS cases. Studies have implicated Stx2 as the sole contributor to acute kidney failure and other systemic complications in humans. The aim of the present study was to determine whether Stx2-specific antibody would be as effective against Shiga toxin-producing Escherichia coli (STEC) strains that produce both Stx1 and Stx2 as it is against strains that produce only Stx2, compared with Stx1-specific antibody. We found that Stx2-specific and Stx1-specific antibodies protected 100% and 0% of piglets, respectively, against oral challenge with a Stx1- and Stx2-producing STEC strain. We conclude that Stx2-specific antibody is sufficient to protect piglets, and possibly humans, against STEC strains that produce both toxins.


Assuntos
Anticorpos Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Escherichia coli Êntero-Hemorrágica/efeitos dos fármacos , Infecções por Escherichia coli/prevenção & controle , Toxina Shiga I/imunologia , Toxina Shiga II/imunologia , Animais , Especificidade de Anticorpos , Escherichia coli Êntero-Hemorrágica/imunologia , Escherichia coli Êntero-Hemorrágica/metabolismo , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Humanos , Toxina Shiga I/biossíntese , Toxina Shiga II/biossíntese , Suínos
7.
BMC Immunol ; 11: 16, 2010 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20334660

RESUMO

BACKGROUND: Shiga toxin 2 (Stx2), one of two Stx liberated by Stx-producing Escherichia coli, is composed of an A subunit monomer and a B subunit pentamer, and is directly linked with hemolytic uremic syndrome in children. The pentameric B subunit binds to its cell surface receptor Gb3 for toxin internalization, and the A subunit follows intracellular retrograde transport to the cytosol where its RNA N-glycosidase activity (RNA-NGA) shuts down the protein synthesis, and leads to cell death. The present study investigated the ability of 19 Stx2 A subunit-specific human monoclonal antibodies (HuMAbs) to neutralize the RNA-NGA, and the association this neutralizing activity with protection of HeLa cells and mice against Stx2-induced death. RESULTS: The HuMAbs that were stronger inhibitors of RNA-NGA were also better at neutralizing Stx2 mediated HeLa cell death, and those that were weaker inhibitors of RNA-NGA activity were also weaker in protecting HeLa cells. These results suggest that the ability of an A subunit-specific antibody to block the RNA-NGA of the toxin is directly related to its ability to neutralize Stx2-mediated HeLa cell death. However, with the exception of the best RNA-NGA blocking antibodies 5C12 and 2F10, the efficacies of antibody neutralization of RNA-NGA of Stx2 did not correlate with their in vivo protective efficacies. The HuMAb 6C3, which neutralized RNA N-glycosidase activity of Stx2 less effectively than the HuMAbs 6D8 and 6B7, protected 100% of the mice against Stx2 challenge at 50 microg/mouse dose. In contrast, the HuMAbs 6D8 and 6B7, which neutralized RNA N-glycosidase activity of Stx2 more effectively than 6C3, protected 20% and 0% mice at that dose, respectively. CONCLUSIONS: The neutralization efficiency of the RNA-NGA of Stx2 by A subunit-specific antibodies correlate strongly with their abilities to protect HeLa cells against Stx2-mediated toxicity but only the strongest RNA-NGA-neutralizing antibodies correlate very well with both protecting HeLa cells and mice against Stx2 challenge.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Proteínas Inativadoras de Ribossomos/antagonistas & inibidores , Toxina Shiga II/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Western Blotting , Células HeLa , Humanos , Camundongos
8.
Infect Immun ; 78(3): 1376-82, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20086088

RESUMO

5C12 HuMAb is a human monoclonal antibody against the A subunit of Shiga toxin 2 (Stx2). We have previously shown that 5C12 HuMAb effectively neutralizes the cytotoxic effects of this toxin by redirecting its transport within the cell and also by neutralizing the toxin's ability to inhibit protein synthesis. The 5C12 HuMAb and its recombinant IgG1 version protect mice at a dose of 0.6 microg against a lethal challenge of Stx2. The contribution of the Fc region to this observed neutralization activity of the 5C12 antibody against Stx2 was investigated in this study. Using recombinant DNA technology, 5C12 isotype variants (IgG1, IgG2, IgG3, and IgG4) and antibody fragments [Fab, F(ab')(2)] were expressed in Chinese hamster ovary cells and evaluated in vitro and in vivo. All four 5C12 isotype variants showed protection in vitro, with the IgG3 and IgG4 variants showing the highest protection in vivo. The Fab and F(ab')(2) fragments also showed protection in vitro but no protection in the mouse toxicity model. Similar results were obtained for a second HuMAb (5H8) against the B subunit of Stx2. The data suggest the importance of the Fc region for neutralization activity, but it is not clear if this is related to the stability of the full-length antibody or if the Fc region is required for effective elimination of the toxin from the body.


Assuntos
Anticorpos Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Antitoxinas/farmacologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Intoxicação/prevenção & controle , Toxina Shiga II/antagonistas & inibidores , Animais , Anticorpos Antibacterianos/genética , Anticorpos Monoclonais/genética , Anticorpos Neutralizantes/genética , Antitoxinas/genética , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Feminino , Células HeLa , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Camundongos , Proteínas Recombinantes/farmacologia , Análise de Sobrevida
9.
Mol Biochem Parasitol ; 168(1): 95-101, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19631240

RESUMO

The genus Cryptosporidium includes several species of intestinal protozoan parasites which multiply in intestinal epithelial cells. The impact of this infection on the transcriptome of cultured host cells was investigated using DNA microarray hybridizations. The expression of 14 genes found to be consistently up- or down-regulated in infected cell monolayers was validated with RT PCR. Using immunofluorescence we examined the expression of Protease Activated Receptor-2, which is encoded by one of the up-regulated genes. In infected cells this receptor localized to the host cell membrane which covers the intracellular trophozoites and meronts. This observation indicates that the composition of the host cell membrane is affected by the developing trophozoite, a phenomenon which has not been described previously.


Assuntos
Membrana Celular/química , Cryptosporidium parvum/fisiologia , Células Epiteliais/parasitologia , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita , Receptor PAR-2/análise , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Microscopia de Fluorescência , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Infect Immun ; 76(5): 1931-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18285498

RESUMO

Infection of children with Shiga toxin (Stx)-producing Escherichia coli (STEC) is the leading cause of hemolytic-uremic syndrome (HUS). Stx2, one of two toxins liberated by the bacteria, is directly linked with HUS. We have previously shown that Stx2-specific human monoclonal antibodies (HuMAbs) protect mice and piglets from fatal systemic complications of Stx2. The present study investigates the mechanisms by which our most efficacious A- and B-subunit-specific HuMAbs neutralize the cytotoxic effects of Stx2 in vitro. Whereas the B-subunit-specific HuMAb 5H8 blocked binding of Stx2 to its receptor on the cell surface, the A-subunit-specific HuMAb 5C12 did not interfere with the toxin-receptor binding. Further investigations revealed that 5C12 did not block endocytosis of Stx2 by HeLa cells as both Stx2 and 5C12 colocalized with early endosomes. However, 5C12 blocked the retrograde transport of the toxin into the Golgi and the endoplasmic reticulum, preventing the toxin from entering the cytosol where the toxin exerts its cytotoxic effect. The endocytosed 5C12/Stx2 complexes appear to be rapidly transported to the plasma membrane and/or to the slow recycling perinuclear compartments, followed by their slow recycling to the plasma membrane, and release into the extracellular environment.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/imunologia , Toxina Shiga II/antagonistas & inibidores , Membrana Celular/química , Citosol/química , Endocitose , Retículo Endoplasmático/química , Endossomos/química , Complexo de Golgi/química , Células HeLa , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Testes de Neutralização , Ligação Proteica
11.
Clin Diagn Lab Immunol ; 12(10): 1141-4, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16210474

RESUMO

Enterocytozoon bieneusi is clinically the most significant among the microsporidia causing chronic diarrhea, wasting, and cholangitis in individuals with human immunodeficiency virus/AIDS. Microscopy with either calcofluor or modified trichrome stains is the standard diagnostic test for microsporidiosis and does not allow species identification. Detection of E. bieneusi infection based on PCR is limited to a few reference laboratories, and thus it is not the standard diagnostic assay. We have recently reported the development and characterization of a panel of monoclonal antibodies against E. bieneusi, and in this publication we evaluated the specificity and sensitivity of an immunofluorescence assay (IFA), compared with PCR, in simian immunodeficiency virus-infected macaques. The IFA, which correlated with the primary PCR method, with a detection limit of 1.5 x 10(5) spores per gram of feces, will simplify considerably the detection of E. bieneusi spores in clinical and environmental specimens and in laboratory and epidemiological investigations.


Assuntos
Enterocytozoon/isolamento & purificação , Fezes/microbiologia , Imunofluorescência/normas , Microsporidiose/diagnóstico , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Animais , Anticorpos Monoclonais , Macaca , Sensibilidade e Especificidade , Síndrome de Imunodeficiência Adquirida dos Símios/microbiologia , Esporos Fúngicos/isolamento & purificação
12.
Clin Diagn Lab Immunol ; 12(9): 1109-13, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16148179

RESUMO

Enterocytozoon bieneusi is clinically the most significant among the microsporidia infecting humans, causing chronic diarrhea, wasting, and cholangitis in individuals with human immunodeficiency virus/AIDS. The lack of immune reagents is largely due to the absence of methods for laboratory propagation of E. bieneusi. We recently described a procedure for the concentration and purification of spores from diarrheic stool of infected humans. Purified spores were used to immunize mice for production and screening of monoclonal antibodies (MAbs) against E. bieneusi. The eight immunoglobulin M MAbs generated and fully characterized did not cross-react with other human microsporidia or with other microorganisms normally present in stool. One of the MAbs, 2G4, reacted with E. bieneusi spores in stools from monkeys and humans, without background fluorescence, which makes it an ideal diagnostic reagent. It also recognizes intracellular stages of the parasite and will be suitable for determining tissue distribution of E. bieneusi in infected hosts. At least two immunodominant antigens of E. bieneusi of 33,000 and 35,000 Da exist, which were recognized by rabbit and mouse antisera. The availability of MAbs against E. bieneusi will simplify considerably the diagnosis of this infection in humans and will provide tools for epidemiologic investigations regarding the true prevalence of the infection in various human and mammalian populations and the environmental sources of infection.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Enterocytozoon/imunologia , Microsporidiose/diagnóstico , Microsporidiose/imunologia , Animais , Especificidade de Anticorpos , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/isolamento & purificação , Reações Cruzadas , Diarreia/diagnóstico , Diarreia/imunologia , Diarreia/parasitologia , Enterocytozoon/crescimento & desenvolvimento , Fezes/parasitologia , Feminino , Humanos , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Esporos de Protozoários/imunologia
13.
Infect Immun ; 73(8): 4607-13, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16040972

RESUMO

Infection of children with Shiga toxin (Stx)-producing Escherichia coli (STEC) can lead to hemolytic-uremic syndrome (HUS) in 5 to 10% of patients. Stx2, one of two toxins liberated by the bacterium, is directly linked with HUS. We have previously shown that Stx-specific human monoclonal antibodies protect STEC-infected animals from fatal systemic complications. The present study defines the protective antibody dose in relation to the time of treatment after the onset of diarrhea in infected gnotobiotic piglets. Using the mouse toxicity model, we selected 5C12, an antibody specific for the A subunit, as the most effective Stx2 antibody for further characterization in the piglet model in which piglets developed diarrhea 16 to 40 h after bacterial challenge, followed by fatal neurological symptoms at 48 to 96 h. Seven groups of piglets received doses of 5C12 ranging from 6.0 mg/kg to 0.05 mg/kg of body weight, administered parenterally 48 h after bacterial challenge. The minimum fully protective antibody dose was 0.4 mg/kg, and the corresponding serum antibody concentration in these piglets was 0.7 mug (+/-0.5)/ml, measured 7 to 14 days after administration. Of 40 infected animals which received Stx2 antibody treatment of > or =0.4 mg/kg, 34 (85%) survived, while only 1 (2.5%) of 39 placebo-treated animals survived. We conclude that the administration of the Stx2-specific antibody was protective against fatal systemic complications even when it was administered well after the onset of diarrhea. These findings suggest that children treated with this antibody, even after the onset of bloody diarrhea, may be equally protected against the risk of developing HUS.


Assuntos
Anticorpos Monoclonais/farmacologia , Diarreia/tratamento farmacológico , Escherichia coli O157/efeitos dos fármacos , Toxina Shiga II/imunologia , Animais , Anticorpos Monoclonais/imunologia , Líquido Ascítico/imunologia , Diarreia/complicações , Diarreia/imunologia , Diarreia/veterinária , Relação Dose-Resposta Imunológica , Escherichia coli O157/imunologia , Vida Livre de Germes , Células HeLa , Humanos , Camundongos , Suínos
14.
J Clin Microbiol ; 43(1): 387-92, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15634999

RESUMO

Enterocytozoon bieneusi is clinically the most significant of the microsporidia in humans, causing chronic diarrhea wasting and cholangitis in individuals with human immunodeficiency virus infection and AIDS. Little progress on this infection has been made because of the inability to propagate E. bieneusi in vitro and in vivo, which limits the source of parasite spores to the stools of infected human patients. Given the size and shape of the E. bieneusi spores (1.1 to 1.6 by 0.7 to 1.0 microm) and the lack of specific immune reagents, the identification and purification of large quantities of spores from feces are technically challenging. Consequently, diagnosis relies entirely on PCR, a labor-intensive approach that requires highly skilled personnel. We describe a method for the purification of E. bieneusi spores from human stools and the production of rabbit-specific antisera. Spores were purified by a combination of isopycnic Percoll gradient centrifugation and continuous sucrose gradient centrifugation. Specific polyclonal antibodies raised in mice and rabbits reacted by indirect immunofluorescence with E. bieneusi but not with Encephalitozoon spp., Candida albicans, Staphylococcus aureus, Escherichia coli, or other forms present in human stools.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Especificidade de Anticorpos , Enterocytozoon/isolamento & purificação , Enterocytozoon/fisiologia , Fezes/parasitologia , Esporos de Protozoários/isolamento & purificação , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Centrifugação com Gradiente de Concentração/métodos , Enterocytozoon/imunologia , Humanos , Camundongos , Microsporidiose/parasitologia , Coelhos , Esporos de Protozoários/imunologia
15.
FEMS Microbiol Lett ; 223(2): 167-75, 2003 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12829282

RESUMO

Staphylococcus aureus are Gram-positive bacteria and cause diverse serious diseases in humans and animals through the production of toxins. The production of toxins is regulated by quorum sensing mechanisms, where proteins such as RNAIII activating protein (RAP) are secreted by the bacteria and induce virulence. Antibodies to RAP have been shown to protect mice from infection, but the molecular structure of RAP was not known and hindered vaccine development. To characterize RAP, recombinant protein was made and tested for its ability to induce genes important for pathogenesis (agr). In addition, monoclonal antibodies were produced to identify its cellular localization. Results shown here indicate that RAP is a 277-aa protein that is an ortholog of the ribosomal protein L2. Like the native molecule, recombinant RAP activates the production of RNAIII (encoded by agr). Using RAP specific monoclonal antibodies we demonstrate that RAP is continuously secreted and while RAP is expressed also in other bacteria (like Staphylococcus epidermidis, Staphylococcus xylosus and Escherichia coli), it is secreted to the culture medium only by S. aureus. Our results show that the ribosomal protein L2 has an extraribosomal function and that when secreted RAP acts as an autoinducer of virulence to regulate S. aureus pathogenesis.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Feminino , Regulação Bacteriana da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fosfoproteínas/metabolismo , RNA Antissenso/metabolismo , RNA Bacteriano/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Virulência
16.
Infect Immun ; 71(6): 3125-30, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12761090

RESUMO

Shiga toxin-producing Escherichia coli (STEC) strains are responsible for causing hemolytic-uremic syndrome (HUS), and systemic administration of Shiga toxin (Stx)-specific human monoclonal antibodies (HuMAbs) is considered a promising approach for prevention or treatment of the disease in children. The goal of the present study was to investigate the ability of Stx2-specific HuMAbs to protect against infections with STEC strains that produce Stx2 variants. Dose-response studies on five HuMAbs, using the mouse toxicity model, revealed that only the three directed against the A subunit were protective against Stx2 variants, and 5C12 was the most effective among the three tested. Two HuMAbs directed against the B subunit, while highly effective against Stx2, were ineffective against Stx2 variants. In a streptomycin-treated mouse model, parenteral administration of 5C12 significantly protected mice up to 48 h after oral bacterial challenge. We conclude that 5C12, reactive against the Stx2 A subunit, is an excellent candidate for immunotherapy against HUS and that antibodies directed against the A subunit of Stx2 have broad-spectrum activity that includes Stx2 variants, compared with those directed against the B subunit.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Infecções por Escherichia coli/prevenção & controle , Toxina Shiga II/imunologia , Animais , Feminino , Células HeLa , Humanos , Immunoblotting , Camundongos , Testes de Neutralização , Subunidades Proteicas , Toxina Shiga II/toxicidade , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA