Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Med Microbiol ; 306(7): 529-540, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27476047

RESUMO

There is a broad interest in adapting live vaccine strains (LVS) for use as recombinant vaccines that can deliver heterologous antigens. The Salmonella enterica serovar Typhimurium SL1344 ΔwecA LVS contains a mutation in wecA that abrogates production of Enterobacterial common antigen. This ΔwecA strain is attenuated in vivo, persistently colonizes the host, and protects against both wild type and cross-Salmonella serovar lethal challenge in a murine model of salmonellosis. Given these characteristics, we hypothesized that the SL1344 ΔwecA strain could be used as a carrier for heterologous antigen expression. To test this hypothesis, SL1344 ΔwecA was engineered to express the Pseudomonas aeruginosa O11 O-antigen gene cluster. Intraperitoneal (IP) but not oral immunization of BALB/c mice with the heterologous expression strain protected against lethal P. aeruginosa intranasal (IN) challenge. Furthermore, IP immunization resulted in P. aeruginosa O11-specific Ig and IgG antibody production. Functional analysis of sera collected from the IP immunized mice showed antibody-mediated agglutination and opsonophagocytic activity against P. aeruginosa. En masse, these results indicate that the S. Typhimurium SL1344 ΔwecA strain expressing the P. aeruginosa O11 O-antigen gene cluster is able to induce a humoral immune response and to protect against lethal P. aeruginosa challenge. As such, the S. Typhimurium SL1344 ΔwecA LVS can likely serve as a vehicle for expression of a wide variety of heterologous antigens as a means to create recombinant vaccines.


Assuntos
Vacinas Bacterianas/imunologia , Portadores de Fármacos , Antígenos O/genética , Antígenos O/metabolismo , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/genética , Salmonella typhimurium/genética , Administração Oral , Animais , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Modelos Animais de Doenças , Feminino , Injeções Intraperitoneais , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
2.
Antimicrob Agents Chemother ; 56(1): 378-90, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22064541

RESUMO

Helicobacter pylori has developed antimicrobial resistance to virtually all current antibiotics. Thus, there is a pressing need to develop new anti-H. pylori therapies. We recently described a novel oligo-acyl-lysyl (OAK) antimicrobial peptidomimetic, C(12)K-2ß(12), that shows potent in vitro bactericidal activity against H. pylori. Herein, we define the mechanism of action and evaluate the in vivo efficacy of C(12)K-2ß(12) against H. pylori after experimental infection of Mongolian gerbils. We demonstrate using a 1-N-phenylnaphthylamine (fluorescent probe) uptake assay and electron microscopy that C(12)K-2ß(12) rapidly permeabilizes the bacterial membrane and creates pores that cause bacterial cell lysis. Furthermore, using nucleic acid binding assays, Western blots, and confocal microscopy, we show that C(12)K-2ß(12) can cross the bacterial membranes into the cytoplasm and tightly bind to bacterial DNA, RNA, and proteins, a property that may result in inhibition of enzymatic activities and macromolecule synthesis. To define the in vivo efficacy of C(12)K-2ß(12), H. pylori-infected gerbils were orogastrically treated with increasing doses and concentrations of C(12)K-2ß(12) 1 day or 1 week postinfection. The efficacy of C(12)K-2ß(12) was strongest in animals that received the largest number of doses at the highest concentration, indicating dose-dependent activity of the peptide (P < 0.001 by analysis of variance [ANOVA]) regardless of the timing of the treatment with C(12)K-2ß(12). Overall, our results demonstrate a dual mode of action of C(12)K-2ß(12) against the H. pylori membrane and cytoplasmic components. Moreover, and consistent with the previously reported in vitro efficacy, C(12)K-2ß(12) shows significant in vivo efficacy against H. pylori when used as monotherapy. Therefore, OAK peptides may be a valuable resource for therapeutic treatment of H. pylori infection.


Assuntos
Antibacterianos/administração & dosagem , Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Permeabilidade da Membrana Celular/efeitos dos fármacos , Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori/efeitos dos fármacos , Peptidomiméticos/administração & dosagem , 1-Naftilamina/análogos & derivados , Administração Oral , Animais , Antibacterianos/síntese química , Antibacterianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/síntese química , Peptídeos Catiônicos Antimicrobianos/metabolismo , Western Blotting , DNA Bacteriano/metabolismo , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana Múltipla , Corantes Fluorescentes , Gerbillinae , Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/ultraestrutura , Testes de Sensibilidade Microbiana , Microscopia Eletrônica , Peptidomiméticos/síntese química , Peptidomiméticos/metabolismo , Ligação Proteica , RNA Bacteriano/metabolismo , Estômago/efeitos dos fármacos , Estômago/microbiologia
3.
Nature ; 435(7045): 1117-21, 2005 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-15973412

RESUMO

Haemoglobin C, which carries a glutamate-to-lysine mutation in the beta-globin chain, protects West African children against Plasmodium falciparum malaria. Mechanisms of protection are not established for the heterozygous (haemoglobin AC) or homozygous (haemoglobin CC) states. Here we report a marked effect of haemoglobin C on the cell-surface properties of P. falciparum-infected erythrocytes involved in pathogenesis. Relative to parasite-infected normal erythrocytes (haemoglobin AA), parasitized AC and CC erythrocytes show reduced adhesion to endothelial monolayers expressing CD36 and intercellular adhesion molecule-1 (ICAM-1). They also show impaired rosetting interactions with non-parasitized erythrocytes, and reduced agglutination in the presence of pooled sera from malaria-immune adults. Abnormal cell-surface display of the main variable cytoadherence ligand, PfEMP-1 (P. falciparum erythrocyte membrane protein-1), correlates with these findings. The abnormalities in PfEMP-1 display are associated with markers of erythrocyte senescence, and are greater in CC than in AC erythrocytes. Haemoglobin C might protect against malaria by reducing PfEMP-1-mediated adherence of parasitized erythrocytes, thereby mitigating the effects of their sequestration in the microvasculature.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/parasitologia , Hemoglobina C/metabolismo , Malária/sangue , Malária/prevenção & controle , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Animais , Anticorpos/imunologia , Antígenos CD36/metabolismo , Adesão Celular , Agregação Eritrocítica , Eritrócitos/patologia , Citometria de Fluxo , Hemeproteínas/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Malária/parasitologia , Plasmodium falciparum/patogenicidade
4.
J Exp Med ; 195(7): 881-92, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11927632

RESUMO

It is thought that both helper and effector functions of CD4(+) T cells contribute to protective immunity to blood stage malaria infection. However, malaria infection does not induce long-term immunity and its mechanisms are not defined. In this study, we show that protective parasite-specific CD4(+) T cells were depleted after infection with both lethal and nonlethal species of rodent PLASMODIUM: It is further shown that the depletion is confined to parasite-specific T cells because (a) ovalbumin (OVA)-specific CD4(+) T cells are not depleted after either malaria infection or direct OVA antigen challenge, and (b) the depletion of parasite-specific T cells during infection does not kill bystander OVA-specific T cells. A significant consequence of the depletion of malaria parasite-specific CD4(+) T cells is impaired immunity, demonstrated in mice that were less able to control parasitemia after depletion of transferred parasite-specific T cells. Using tumor necrosis factor (TNF)-RI knockout- and Fas-deficient mice, we demonstrate that the depletion of parasite-specific CD4(+) T cells is not via TNF or Fas pathways. However, in vivo administration of anti-interferon (IFN)-gamma antibody blocks depletion, suggesting that IFN-gamma is involved in the process. Taken together, these data suggest that long-term immunity to malaria infection may be affected by an IFN-gamma-mediated depletion of parasite-specific CD4(+) T cells during infection. This study provides further insight into the nature of immunity to malaria and may have a significant impact on approaches taken to develop a malaria vaccine.


Assuntos
Linfócitos T CD4-Positivos/parasitologia , Malária/imunologia , Plasmodium/imunologia , Linfócitos T/imunologia , Animais , Biomarcadores/análise , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Linfonodos/imunologia , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Plasmodium/classificação , Plasmodium berghei/imunologia , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia
5.
PLoS Pathog ; 4(9): e1000147, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18773118

RESUMO

Plasmodium falciparum malaria parasites, living in red blood cells, express proteins of the erythrocyte membrane protein-1 (PfEMP1) family on the red blood cell surface. The binding of PfEMP1 molecules to human cell surface receptors mediates the adherence of infected red blood cells to human tissues. The sequences of the 60 PfEMP1 genes in each parasite genome vary greatly from parasite to parasite, yet the variant PfEMP1 proteins maintain receptor binding. Almost all parasites isolated directly from patients bind the human CD36 receptor. Of the several kinds of highly polymorphic cysteine-rich interdomain region (CIDR) domains classified by sequence, only the CIDR1alpha domains bind CD36. Here we describe the CD36-binding portion of a CIDR1alpha domain, MC179, as a bundle of three alpha-helices that are connected by a loop and three additional helices. The MC179 structure, containing seven conserved cysteines and 10 conserved hydrophobic residues, predicts similar structures for the hundreds of CIDR sequences from the many genome sequences now known. Comparison of MC179 with the CIDR domains in the genome of the P. falciparum 3D7 strain provides insights into CIDR domain structure. The CIDR1alpha three-helix bundle exhibits less than 20% sequence identity with the three-helix bundles of Duffy-binding like (DBL) domains, but the two kinds of bundles are almost identical. Despite the enormous diversity of PfEMP1 sequences, the CIDR1alpha and DBL protein structures, taken together, predict that a PfEMP1 molecule is a polymer of three-helix bundles elaborated by a variety of connecting helices and loops. From the structures also comes the insight that DBL1alpha domains are approximately 100 residues larger and that CIDR1alpha domains are approximately 100 residues smaller than sequence alignments predict. This new understanding of PfEMP1 structure will allow the use of better-defined PfEMP1 domains for functional studies, for the design of candidate vaccines, and for understanding the molecular basis of cytoadherence.


Assuntos
Cisteína , Plasmodium falciparum/química , Proteínas de Protozoários/química , Animais , Sítios de Ligação , Antígenos CD36/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas de Protozoários/metabolismo
6.
Antimicrob Agents Chemother ; 53(10): 4231-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19620333

RESUMO

The gastric pathogen Helicobacter pylori has developed resistance to virtually all current antibiotics; thus, there is a pressing need to develop new anti-H. pylori therapies. The goal of this work was to evaluate the antibacterial effect of oligo-acyl-lysyl (OAK) antimicrobial peptidomimetics to determine if they might represent alternatives to conventional antibiotic treatment of H. pylori infection. A total of five OAK sequences were screened for growth-inhibitory and/or bactericidal effects against H. pylori strain G27; four of these sequences had growth-inhibitory and bactericidal effects. The peptide with the highest efficacy against strain G27, C12K-2beta12, was selected for further characterization against five additional H. pylori strains (26695, J99, 7.13, SS1, and HPAG1). C12K-2beta12 displayed MIC and minimum bactericidal concentration (MBC) ranges of 6.5 to 26 microM and 14.5 to 90 microM, respectively, across the six strains after 24 h of exposure. G27 was the most sensitive H. pylori strain (MIC = 6.5 to 7 microM; MBC = 15 to 20 microM), whereas 26695 was the least susceptible strain (MIC = 25 to 26 microM; MBC = 70 to 90 microM). H. pylori was completely killed after 6 to 8 h of incubation in liquid cultures containing two times the MBC of C12K-2beta12. The OAK demonstrated strong in vitro stability, since efficacy was maintained after incubation at extreme temperatures (4 degrees C, 37 degrees C, 42 degrees C, 50 degrees C, 55 degrees C, 60 degrees C, and 95 degrees C) and at low pH, although reduced killing kinetics were observed at pH 4.5. Additionally, upon transient exposure to the bacteria, C12K-2beta12 showed irreversible and significant antibacterial effects and was also nonhemolytic. Our results show a significant in vitro effect of C12K-2beta12 against H. pylori and suggest that OAKs may be a valuable resource for the treatment of H. pylori infection.


Assuntos
Antibacterianos/farmacologia , Helicobacter pylori/efeitos dos fármacos , Polímeros/farmacologia , Animais , Hemólise/efeitos dos fármacos , Cavalos , Concentração de Íons de Hidrogênio , Cinética , Testes de Sensibilidade Microbiana , Temperatura
7.
J Microbiol ; 52(4): 259-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24682990

RESUMO

The Gram-negative pathogen Helicobacter pylori is increasingly more resistant to the three major antibiotics (metronidazole, clarithromycin and amoxicillin) that are most commonly used to treat infection. As a result, there is an increased rate of treatment failure; this translates into an overall higher cost of treatment due to the need for increased length of treatment and/or the requirement for combination or sequential therapy. Given the rise in antibiotic resistance, the complicated treatment regime, and issues related to patient compliance that stem from the duration and complexity of treatment, there is clearly a pressing need for the development of novel therapeutic strategies to combat H. pylori infection. As such, researchers are actively investigating the utility of antimicrobial peptides, small molecule inhibitors and naturopathic therapies. Herein we review and discuss each of these novel approaches as a means to target this important gastric pathogen.


Assuntos
Antibacterianos/uso terapêutico , Produtos Biológicos/uso terapêutico , Infecções por Helicobacter/tratamento farmacológico , Farmacorresistência Bacteriana , Helicobacter pylori/efeitos dos fármacos , Humanos
8.
PLoS One ; 8(7): e68917, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935905

RESUMO

The most evident challenge to treatment of Helicobacter pylori, a bacterium responsible for gastritis, peptic ulcers and gastric cancer, is the increasing rate of resistance to all currently used therapeutic antibiotics. Thus, the development of novel therapies is urgently required. N-geranyl-N'-(2-adamantyl) ethane-1, 2-diamine (SQ109) is an ethylene diamine-based antitubercular drug that is currently in clinical trials for the treatment of tuberculosis (TB). Previous pharmacokinetic studies of SQ109 revealed that persistently high concentrations of SQ109 remain in the stomach 4 hours post oral administration in rats. This finding, combined with the need for new anti-Helicobacter therapies, prompted us to define the in vitro efficacy of SQ109 against H. pylori. Liquid broth micro-dilution was used for susceptibility studies to determine the antimicrobial activity of SQ109 against a total of 6 laboratory strains and 20 clinical isolates of H. pylori; the clinical isolates included a multi-drug resistant strain. All strains tested were susceptible to SQ109 with MIC and MBC ranges of 6-10 µM and 50-60 µM, respectively. SQ109 killing kinetics were concentration- and time-dependent. SQ109 killed H. pylori in 8-10 h at 140 µM (2MBCs) or 4-6 h at 200 µM (~3MBCs). Importantly, though the kinetics of killing were altered, SQ109 retained potent bactericidal activity against H. pylori at low pH. Additionally, SQ109 demonstrated robust thermal stability and was effective at killing slow growing or static bacteria. In fact, pretreatment of cultures with a bacteriostatic concentration of chloramphenicol (Cm) synergized the effects of typically bacteriostatic concentrations of SQ109 to the level of five-logs of bacterial killing. A molar-to-molar comparison of the efficacy of SQ109 as compared to metronidazole (MTZ), amoxicillin (AMX), rifampicin (RIF) and clarithromycin (CLR), revealed that SQ109 was superior to MTZ, AMX and RIF but not to CLR. Finally, the frequency of resistance to SQ109 was low and electron microscopy studies revealed that SQ109 interacted with bacterial inner membrane and cytoplasmic content(s). Collectively, our in vitro data demonstrate that SQ109 is an effective monotherapy against susceptible and multi-drug resistant strains of H. pylori and may be useful alone or in combination with other antibiotics for development as a new class of anti-Helicobacter drugs.


Assuntos
Adamantano/análogos & derivados , Antituberculosos/farmacologia , Membrana Celular/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Etilenodiaminas/farmacologia , Helicobacter pylori/efeitos dos fármacos , Adamantano/farmacologia , Amoxicilina/farmacologia , Membrana Celular/ultraestrutura , Cloranfenicol/farmacologia , Claritromicina/farmacologia , Sinergismo Farmacológico , Úlcera Duodenal/complicações , Úlcera Duodenal/tratamento farmacológico , Úlcera Duodenal/microbiologia , Duodenite/complicações , Duodenite/tratamento farmacológico , Duodenite/microbiologia , Gastrite/complicações , Gastrite/tratamento farmacológico , Gastrite/microbiologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/isolamento & purificação , Helicobacter pylori/ultraestrutura , Humanos , Concentração de Íons de Hidrogênio , Metronidazol/farmacologia , Testes de Sensibilidade Microbiana , Microscopia Eletrônica , Úlcera Gástrica/complicações , Úlcera Gástrica/tratamento farmacológico , Úlcera Gástrica/microbiologia
9.
J Infect Dis ; 193(5): 731-40, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16453270

RESUMO

BACKGROUND: After continuous exposure to malarial infections in regions of Africa where malaria is hyperendemic, children attain clinical immunity. This immunity results, in part, from the acquisition of antibodies against a large repertoire of variant antigens expressed on the surface of infected erythrocytes, such as the Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1). We determined whether a subunit vaccine to a portion of PfEMP1 could induce protection in nonhuman primates. METHODS: We immunized Aotus nancymai monkeys with PfEMP1 recombinant (r) cysteine-rich interdomain region 1 alpha (CIDR1 alpha ) and infected them twice with P. falciparum Vietnam Oak Knoll strain, the most virulent strain of P. falciparum in Aotus monkeys--each infection expressed a different PfEMP1. Anti-PfEMP1 antibodies were analyzed by enzyme-linked immunosorbent assay against rCIDR1 alpha and by flow cytometry against infected erythrocytes. RESULTS: Immunization with rCIDR1 alpha was not protective, despite delayed patency during the first infection, but it protected monkeys against severe anemia during reinfection. Protection against anemia is associated with a more rapid increase in antibodies to PfEMP1. CONCLUSION: The findings of reduced severe disease in rCIDR1 alpha -vaccinated Aotus monkeys provide experimental support for a PfEMP1-based vaccine to protect African children against severe malarial disease. Such vaccination may function by priming for the accelerated acquisition of immunity to new PfEMP1 variants.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Anemia , Animais , Anticorpos Antiprotozoários/sangue , Aotus trivirgatus , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Eritrócitos/química , Eritrócitos/parasitologia , Citometria de Fluxo , Hematócrito , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/fisiopatologia , Parasitemia , Estrutura Terciária de Proteína , Recidiva , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
10.
Proc Natl Acad Sci U S A ; 100(5): 2628-33, 2003 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-12594331

RESUMO

Although there is good evidence that immunity to the blood stages of malaria parasites can be mediated by different effector components of the adaptive immune system, target antigens for a principal component, effector CD4(+) T cells, have never been defined. We generated CD4(+) T cell lines to fractions of native antigens from the blood stages of the rodent parasite, Plasmodium yoelii, and identified fraction-specific T cells that had a Th1 phenotype (producing IL-2, IFN-gamma, and tumor necrosis factor-alpha, but not IL-4, after antigenic stimulation). These T cells could inhibit parasite growth in recipient severe combined immunodeficient mice. N-terminal sequencing of the fraction showed identity with hypoxanthine guanine xanthine phosphoribosyl transferase (HGXPRT). Recombinant HGXPRT from the human malaria parasite, Plasmodium falciparum, activated the T cells in vitro, and immunization of normal mice with recombinant HGXPRT reduced parasite growth rates in all mice after challenge.


Assuntos
Malária/imunologia , Malária/prevenção & controle , Pentosiltransferases/química , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Divisão Celular , Citocinas/metabolismo , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Epitopos , Citometria de Fluxo , Interferon gama/metabolismo , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Focalização Isoelétrica , Ativação Linfocitária , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Dados de Sequência Molecular , Pentosiltransferases/genética , Fenótipo , Plasmodium yoelii/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Células Th1 , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA