Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Glycobiology ; 32(5): 391-403, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-34972864

RESUMO

The heat-labile enterotoxins of Escherichia coli and cholera toxin of Vibrio cholerae are related in structure and function. Each of these oligomeric toxins is comprised of one A polypeptide and five B polypeptides. The B-subunits bind to gangliosides, which are followed by uptake into the intoxicated cell and activation of the host's adenylate cyclase by the A-subunits. There are two antigenically distinct groups of these toxins. Group I includes cholera toxin and type I heat-labile enterotoxin of E. coli; group II contains the type II heat-labile enterotoxins of E. coli. Three variants of type II toxins, designated LT-IIa, LT-IIb and LT-IIc have been described. Earlier studies revealed the crystalline structure of LT-IIb. Herein the carbohydrate binding specificity of LT-IIc B-subunits was investigated by glycosphingolipid binding studies on thin-layer chromatograms and in microtiter wells. Binding studies using a large variety of glycosphingolipids showed that LT-IIc binds with high affinity to gangliosides with a terminal Neu5Acα3Gal or Neu5Gcα3Gal, e.g. the gangliosides GM3, GD1a and Neu5Acα3-/Neu5Gcα3--neolactotetraosylceramide and Neu5Acα3-/Neu5Gcα3-neolactohexaosylceramide. The crystal structure of LT-IIc B-subunits alone and with bound LSTd/sialyl-lacto-N-neotetraose d pentasaccharide uncovered the molecular basis of the ganglioside recognition. These studies revealed common and unique functional structures of the type II family of heat-labile enterotoxins.


Assuntos
Toxinas Bacterianas , Proteínas de Escherichia coli , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxina da Cólera/metabolismo , Enterotoxinas/química , Enterotoxinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Gangliosídeo G(M1)/metabolismo , Gangliosídeos/metabolismo , Temperatura Alta
2.
Int J Mol Sci ; 20(1)2018 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-30587795

RESUMO

Triple negative breast cancer (TNBC) remains a serious health problem with poor prognosis and limited therapeutic options. To discover novel approaches to treat TNBC, we screened cholera toxin (CT) and the members of the bacterial type II heat-labile enterotoxin family (LT-IIa, LT-IIb, and LT-IIc) for cytotoxicity in TNBC cells. Only LT-IIc significantly reduced viability of the TNBC cell lines BT549 and MDA-MB-231 (IC50 = 82.32 nM). LT-IIc had no significant cytotoxic effect on MCF10A (IC50 = 2600 nM), a non-tumorigenic breast epithelial cell line, and minimal effects on MCF7 and T47D, ER⁺ cells, or SKBR-3 cells, HER2⁺ cells. LT-IIc stimulated autophagy through inhibition of the mTOR pathway, while simultaneously inhibiting autophagic progression, as seen by accumulation of LC3B-II and p62. Morphologically, LT-IIc induced the formation of enlarged LAMP2+ autolysosomes, which was blocked by co-treatment with bafilomycin A1. LT-IIc induced apoptosis as demonstrated by the increase in caspase 3/7 activity and Annexin V staining. Co-treatment with necrostatin-1, however, demonstrated that the lethal response of LT-IIc is elicited, in part, by concomitant induction of necroptosis. Knockdown of ATG-5 failed to rescue LT-IIc-induced cytotoxicity, suggesting LT-IIc can exert its cytotoxic effects downstream or independently of autophagophore initiation. Collectively, these experiments demonstrate that LT-IIc acts bifunctionally, inducing autophagy, while simultaneously blocking autolysosomal progression in TNBC cells, inducing a specific cytotoxicity in this breast cancer subtype.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Enterotoxinas/toxicidade , Proteína 5 Relacionada à Autofagia/antagonistas & inibidores , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Temperatura Alta , Humanos , Imidazóis/metabolismo , Indóis/metabolismo , Lisossomos/metabolismo , Necrose , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
3.
J Antimicrob Chemother ; 72(1): 153-165, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27634916

RESUMO

OBJECTIVES: The pharmacodynamics of polymyxin/carbapenem combinations against carbapenem-resistant Acinetobacter baumannii (CRAB) are largely unknown. Our objective was to determine whether intensified meropenem regimens in combination with polymyxin B enhance killing and resistance suppression of CRAB. METHODS: Time-kill experiments for meropenem and polymyxin B combinations were conducted against three polymyxin B-susceptible (MIC of polymyxin B = 0.5 mg/L) CRAB strains with varying meropenem MICs (ATCC 19606, N16870 and 03-149-1; MIC of meropenem = 4, 16 and 64 mg/L, respectively) at 108 cfu/mL. A hollow-fibre infection model was then used to simulate humanized regimens of polymyxin B and meropenem (2, 4, 6 and 8 g prolonged infusions every 8 h) versus N16870 at 108 cfu/mL over 14 days. New mathematical mechanism-based models were developed using S-ADAPT. RESULTS: Time-kill experiments were well described by the mathematical mechanism-based models, with the presence of polymyxin B drastically decreasing the meropenem concentration needed for half-maximal activity against meropenem-resistant populations from 438 to 82.1 (ATCC 19606), 158 to 93.6 (N16870) and 433 to 76.0 mg/L (03-149-1). The maximum killing effect of combination treatment was similar among all three strains despite divergent meropenem MIC values (Emax = 2.13, 2.08 and 2.15; MIC of meropenem = 4, 16 and 64 mg/L, respectively). Escalating the dose of meropenem in hollow-fibre combination regimens from 2 g every 8 h to 8 g every 8 h resulted in killing that progressed from a >2.5 log10 cfu/mL reduction with regrowth by 72 h (2 g every 8 h) to complete eradication by 336 h (8 g every 8 h). CONCLUSION: Intensified meropenem dosing in combination with polymyxin B may offer a unique strategy to kill CRAB irrespective of the meropenem MIC.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Polimixina B/farmacologia , Tienamicinas/farmacologia , Resistência beta-Lactâmica , Antibacterianos/administração & dosagem , Humanos , Meropeném , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Modelos Teóricos , Polimixina B/administração & dosagem , Tienamicinas/administração & dosagem
4.
J Leukoc Biol ; 100(2): 361-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27059843

RESUMO

LT-IIb, a type II heat-labile enterotoxin produced by Escherichia coli, is a potent intradermal adjuvant that enhances immune responses to coadministered antigens. Although the immune mechanisms that promote this augmented immune response have not been well defined, prior intradermal immunization experiments suggested that early cellular and immunomodulatory events at the site of immunization modulated the augmentation of antigen-specific immune responses by LT-IIb. To investigate that hypothesis, mice were intradermally immunized with a recombinant ricin vaccine, a prospective toxin subunit antigen, in the presence and absence of LT-IIb. Analysis of tissue-fluid collection, coupled with histologic sections from the site of intradermal immunization, revealed that a single dose of LT-IIb induced local production of interleukin 6 and promoted a regional infiltration of neutrophils. The adjuvant effects of LT-IIb were abrogated in interleukin 6-deficient mice and when mice were depleted of neutrophils by pretreatment with anti-Ly6G. Overall, these data firmly demonstrated that LT-IIb, when used as an intradermal adjuvant, recruits neutrophils and is a potent rapid inducer of interleukin 6.


Assuntos
Toxinas Bacterianas/administração & dosagem , Enterotoxinas/administração & dosagem , Proteínas de Escherichia coli/administração & dosagem , Imunidade Humoral/imunologia , Inflamação/imunologia , Interleucina-6/fisiologia , Neutrófilos/imunologia , Dermatopatias/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/farmacologia , Enterotoxinas/farmacologia , Proteínas de Escherichia coli/farmacologia , Feminino , Imunidade Humoral/efeitos dos fármacos , Imunização , Inflamação/metabolismo , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Dermatopatias/metabolismo , Dermatopatias/prevenção & controle , Vacinas/administração & dosagem
5.
Infect Immun ; 84(6): 1693-1703, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27001538

RESUMO

Streptococcus pneumoniae commonly inhabits the nasopharynx as a member of the commensal biofilm. Infection with respiratory viruses, such as influenza A virus, induces commensal S. pneumoniae to disseminate beyond the nasopharynx and to elicit severe infections of the middle ears, lungs, and blood that are associated with high rates of morbidity and mortality. Current preventive strategies, including the polysaccharide conjugate vaccines, aim to eliminate asymptomatic carriage with vaccine-type pneumococci. However, this has resulted in serotype replacement with, so far, less fit pneumococcal strains, which has changed the nasopharyngeal flora, opening the niche for entry of other virulent pathogens (e.g., Streptococcus pyogenes, Staphylococcus aureus, and potentially Haemophilus influenzae). The long-term effects of these changes are unknown. Here, we present an attractive, alternative preventive approach where we subvert virus-induced pneumococcal disease without interfering with commensal colonization, thus specifically targeting disease-causing organisms. In that regard, pneumococcal surface protein A (PspA), a major surface protein of pneumococci, is a promising vaccine target. Intradermal (i.d.) immunization of mice with recombinant PspA in combination with LT-IIb(T13I), a novel i.d. adjuvant of the type II heat-labile enterotoxin family, elicited strong systemic PspA-specific IgG responses without inducing mucosal anti-PspA IgA responses. This response protected mice from otitis media, pneumonia, and septicemia and averted the cytokine storm associated with septic infection but had no effect on asymptomatic colonization. Our results firmly demonstrated that this immunization strategy against virally induced pneumococcal disease can be conferred without disturbing the desirable preexisting commensal colonization of the nasopharynx.


Assuntos
Anticorpos Antibacterianos/biossíntese , Proteínas de Bactérias/imunologia , Vacinas Pneumocócicas/administração & dosagem , Pneumonia Pneumocócica/prevenção & controle , Streptococcus pneumoniae/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/genética , Administração Intranasal , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Enterotoxinas/administração & dosagem , Enterotoxinas/genética , Enterotoxinas/imunologia , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Feminino , Expressão Gênica , Imunidade Humoral/efeitos dos fármacos , Imunização , Imunoglobulina G/biossíntese , Injeções Intradérmicas , Camundongos , Camundongos Endogâmicos BALB C , Nasofaringe/efeitos dos fármacos , Nasofaringe/imunologia , Nasofaringe/microbiologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/mortalidade , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Análise de Sobrevida , Simbiose/efeitos dos fármacos , Vacinas Conjugadas
6.
PLoS One ; 10(11): e0142942, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26565800

RESUMO

Poor immune responses elicited by vaccine antigens can be enhanced by the use of appropriate adjuvants. Type II heat-labile enterotoxins (HLT) produced by Escherichia coli are extremely potent adjuvants that augment both humoral and cellular immunity to co-administered antigens. Recent findings demonstrate that LT-IIb and LT-IIc, two type II HLT adjuvants, exhibit potent, yet distinguishable CD8(+) T cell adjuvant properties. While LT-IIc elicits a robust and rapid response at one week after administration, LT-IIb engenders a more gradual and slower expansion of antigen-specific CD8(+) T cells that correlates with improved immunity. The variations in immune effects elicited by the HLT adjuvants have been generally attributed to their highly divergent B subunits that mediate binding to various gangliosides on cell surfaces. Yet, HLT adjuvants with point mutations in the B subunit that significantly alter ganglioside binding retain similar adjuvant functions. Therefore, the contribution of the B subunits to adjuvanticity remains unclear. To investigate the influence of the B subunits on the enhancement of immune responses by LT-IIb and LT-IIc, chimeric HLT were engineered in which the B subunits of the two adjuvants were exchanged. Comparing the immune potentiating characteristics of both native and chimeric HLT adjuvants, it was found that not all the adjuvant characteristics of the HLT adjuvants were modulated by the respective B subunits. Specifically, the differences in the CD8(+) T cell kinetics and protective responses elicited by LT-IIb and LT-IIc did indeed followed their respective B subunits. However, induction of IL-1 from macrophages and the capacity to intoxicate cells in a mouse Y1 adrenal cell bioassay did not correlate with the B subunits. Therefore, it is likely that additional factors other than the B subunits contribute to the effects elicited by the HLT adjuvants.


Assuntos
Toxinas Bacterianas/química , Linfócitos T CD8-Positivos/imunologia , Enterotoxinas/química , Proteínas de Escherichia coli/química , Adjuvantes Imunológicos , Glândulas Suprarrenais/citologia , Animais , Bioensaio , Linfócitos T CD8-Positivos/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Escherichia coli/metabolismo , Feminino , Citometria de Fluxo , Gangliosídeos/química , Sistema Imunitário , Listeria monocytogenes , Macrófagos/metabolismo , Macrófagos Peritoneais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Linfócitos T/citologia
7.
PLoS One ; 9(12): e113978, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25536061

RESUMO

Vaccinations are extremely effective at combating infectious diseases. Many conserved antigen (Ag) targets, however, are poorly immunogenic. Protein subunit vaccines frequently elicit only humoral immune responses and fail to confer protection against serious intracellular pathogens. These barriers to vaccine development are often overcome by the use of appropriate adjuvants. Heat-labile enterotoxins (HLT) produced by enterotoxigenic strains of Escherichia coli are potent adjuvants when administered by mucosal or systemic routes. The efficacy of the type II HLT, however, has not been well-defined when administered by the intradermal (ID) route. Using a murine ID immunization model, the adjuvant properties of LT-IIb and LT-IIc, two type II HLTs, were compared with those of LT-I, a prototypical type I HLT. While all three HLT adjuvants enhanced Ag-specific humoral responses to similar levels, LT-IIb and LT-IIc, in contrast to LT-I, induced a more vigorous Ag-specific CD8+ T cell response and proffered faster clearance of Listeria monocytogenes in a challenge model. Additionally, LT-IIb and LT-IIc induced distinct differences in the profiles of the Ag-specific CD8+ T cell responses. While LT-IIc stimulated a robust and rapid primary CD8+ T cell response, LT-IIb exhibited slower CD8+ T cell expansion and contraction kinetics with the formation of higher percentages of effector memory cells. In comparison to LT-I and LT-IIc, LT-IIb evoked better long-term protection after immunization. Furthermore, LT-IIb and LT-IIc enhanced the total number of dendritic cells (DC) in the draining lymph node (DLN) and expression of costimulatory molecules CD80, CD86, and CD40 on DCs. In contrast to LT-I, LT-IIb and LT-IIc induced less edema, cellular infiltrates, and general inflammation at the site of ID injection. Thus, LT-IIb and LT-IIc are attractive comprehensive ID adjuvants with unique characteristic that enhance humoral and cellular immunity to a co-administered protein Ag.


Assuntos
Antígenos de Bactérias/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/farmacologia , Linfócitos T CD8-Positivos/imunologia , Escherichia coli Enterotoxigênica/química , Enterotoxinas/administração & dosagem , Enterotoxinas/farmacologia , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Escherichia coli/farmacologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Animais , Antígenos de Bactérias/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Imunização , Memória Imunológica/efeitos dos fármacos , Inflamação/patologia , Injeções Intradérmicas , Cinética , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Ovalbumina/imunologia
8.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 12): 1604-12, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23151625

RESUMO

The pentameric B subunit of the type II heat-labile enterotoxin of Escherichia coli (LT-IIb-B(5)) is a potent signaling molecule capable of modulating innate immune responses. It has previously been shown that LT-IIb-B(5), but not the LT-IIb-B(5) Ser74Asp variant [LT-IIb-B(5)(S74D)], activates Toll-like receptor (TLR2) signaling in macrophages. Consistent with this, the LT-IIb-B(5)(S74D) variant failed to bind TLR2, in contrast to LT-IIb-B(5) and the LT-IIb-B(5) Thr13Ile [LT-IIb-B(5)(T13I)] and LT-IIb-B(5) Ser74Ala [LT-IIb-B(5)(S74A)] variants, which displayed the highest binding activity to TLR2. Crystal structures of the Ser74Asp, Ser74Ala and Thr13Ile variants of LT-IIb-B(5) have been determined to 1.90, 1.40 and 1.90 Å resolution, respectively. The structural data for the Ser74Asp variant reveal that the carboxylate side chain points into the pore, thereby reducing the pore size compared with that of the wild-type or the Ser74Ala variant B pentamer. On the basis of these crystallographic data, the reduced TLR2-binding affinity of the LT-IIb-B(5)(S74D) variant may be the result of the pore of the pentamer being closed. On the other hand, the explanation for the enhanced TLR2-binding activity of the LT-IIb-B(5)(S74A) variant is more complex as its activity is greater than that of the wild-type B pentamer, which also has an open pore as the Ser74 side chain points away from the pore opening. Data for the LT-IIb-B(5)(T13I) variant show that four of the five variant side chains point to the outside surface of the pentamer and one residue points inside. These data are consistent with the lack of binding of the LT-IIb-B(5)(T13I) variant to GD1a ganglioside.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/farmacologia , Enterotoxinas/química , Enterotoxinas/farmacologia , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/farmacologia , Escherichia coli/química , Receptor 2 Toll-Like/metabolismo , Toxinas Bacterianas/metabolismo , Cristalização , Cristalografia por Raios X , Enterotoxinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Ligação Proteica , Transdução de Sinais , Eletricidade Estática , Relação Estrutura-Atividade
9.
Infect Immun ; 78(11): 4705-13, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20713622

RESUMO

Two families of bacterial heat-labile enterotoxins (HLTs) have been described: the type I HLTs are comprised of cholera toxin (CT) of Vibrio cholerae, LT-I of Escherichia coli, and several related HLTs; the type II HLTs are comprised of LT-IIa and LT-IIb. Herein, we report LT-IIc, a new type II HLT encoded from an enterotoxigenic E. coli (ETEC) strain isolated from an avian host. Using a mouse Y1 adrenal cell bioassay, LT-IIc was shown to be less cytotoxic than CT, LT-IIa, or LT-IIb. Cytotoxicity of LT-IIc was partially neutralized by antisera recognizing LT-IIa or LT-IIb but not by anti-CT antiserum. Genes encoding putative A polypeptide and B polypeptides of LT-IIc were arranged in an operon which was flanked by potential prophage sequences. Analysis of the nucleotide and predicted amino acid sequences demonstrated that the A polypeptide of LT-IIc has moderate homology to the A polypeptides of CT and LT-I and high homology to the A polypeptides of LT-IIa and LT-IIb. The B polypeptide of LT-IIc exhibited no significant homology to the B polypeptides of CT and LT-I and only moderate homology to the B polypeptides of LT-IIa and LT-IIb. The binding pattern of LT-IIc for gangliosides was distinctive from that of either LT-IIa or LT-IIb. The data suggest that other types of the type II HLT subfamily are circulating in the environment and that host specificity of type II HLT is likely governed by changes in the B polypeptide which mediate binding to receptors.


Assuntos
Toxinas Bacterianas/classificação , Toxinas Bacterianas/genética , Doenças das Aves/microbiologia , Diarreia/veterinária , Escherichia coli Enterotoxigênica/isolamento & purificação , Enterotoxinas/classificação , Enterotoxinas/genética , Proteínas de Escherichia coli/classificação , Proteínas de Escherichia coli/genética , Struthioniformes/microbiologia , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Diarreia/microbiologia , Escherichia coli Enterotoxigênica/genética , Escherichia coli Enterotoxigênica/metabolismo , Escherichia coli Enterotoxigênica/patogenicidade , Enterotoxinas/química , Enterotoxinas/metabolismo , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Macrófagos Peritoneais , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de DNA
10.
Infect Immun ; 78(3): 1147-62, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20008538

RESUMO

Iron (Fe) in soluble elemental form is found in the tissues and fluids of animals at concentrations insufficient for sustaining growth of bacteria. Consequently, to promote colonization and persistence, pathogenic bacteria evolved a myriad of scavenging mechanisms to acquire Fe from the host. Bordetella bronchiseptica, the etiologic agent of upper respiratory infections in a wide range of mammalian hosts, expresses a number of proteins for acquisition of Fe. Using proteomic and genomic approaches, three Fe-regulated genes were identified in the bordetellae: bfrH, a gene encoding a putative siderophore receptor; ecfI, a gene encoding a putative extracellular function (ECF) sigma factor; and ecfR, a gene encoding a putative EcfI modulator. All three genes are highly conserved in B. pertussis, B. parapertussis, and B. avium. Genetic analysis revealed that transcription of bfrH was coregulated by ecfI, ecfR, and fur1, one of two fur homologues carried by B. bronchiseptica. Overexpression of ecfI decoupled bfrH from Fe-dependent regulation. In contrast, expression of bfrH was significantly reduced in an ecfI deletion mutant. Deletion of ecfR, however, was correlated with a significant increase in expression of bfrH, due in part to a cis-acting nucleotide sequence within ecfR which likely reduces the frequency of readthrough transcription of bfrH from the Fe-dependent ecfIR promoter. Using a murine competition infection model, bfrH was shown to be required for optimal virulence of B. bronchiseptica. These experiments revealed ecfIR-bfrH as a locus encoding a new member of the growing family of Fe and ECF sigma factor-modulated regulons in the bordetellae.


Assuntos
Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas de Bactérias/fisiologia , Bordetella bronchiseptica/patogenicidade , Regulação Bacteriana da Expressão Gênica , Ferro/metabolismo , Receptores de Superfície Celular/biossíntese , Proteínas Repressoras/fisiologia , Fator sigma/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sequência Conservada , Feminino , Deleção de Genes , Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Dados de Sequência Molecular , Virulência
11.
J Bacteriol ; 189(17): 6266-75, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17586630

RESUMO

Expression of the hurIR bhuRSTUV heme utilization locus in Bordetella bronchiseptica is coordinately controlled by the global iron-dependent regulator Fur and the extracytoplasmic function sigma factor HurI. Activation of HurI requires transduction of a heme-dependent signal via HurI, HurR, and BhuR, a three-component heme-dependent regulatory system. In silico searches of the B. bronchiseptica genome to identify other genes that encode additional participants in this heme-dependent regulatory cascade revealed hurP, an open reading frame encoding a polypeptide with homology to (i) RseP, a site 2 protease (S2P) of Escherichia coli required for modifying the cytoplasmic membrane protein RseA, and (ii) YaeL, an S2P of Vibrio cholerae required for modification of the cytoplasmic membrane protein TcpP. A mutant of B. bronchiseptica defective for hurP was incapable of regulating expression of BhuR in a heme-dependent manner. Furthermore, the hurP mutant was unable to utilize hemin as a sole source of nutrient Fe. These defects in hemin utilization and heme-dependent induction of BhuR were restored when recombinant hurP (or recombinant rseP) was introduced into the mutant. Introduction of hurP into a yaeL mutant of V. cholerae also complemented its S2P defect. These data provided strong evidence that protease activity and cleavage site recognition was conserved in HurP, RseP, and YaeL. The data are consistent with a model in which HurP functionally modifies HurR, a sigma factor regulator that is essential for heme-dependent induction of bhuR.


Assuntos
Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas de Bactérias/biossíntese , Bordetella bronchiseptica/genética , Regulação Bacteriana da Expressão Gênica , Heme/metabolismo , Peptídeo Hidrolases/biossíntese , Receptores de Superfície Celular/biossíntese , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Bordetella bronchiseptica/metabolismo , Endopeptidases/genética , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/genética , Deleção de Genes , Teste de Complementação Genética , Hemina/metabolismo , Ferro/metabolismo , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutagênese Insercional , Peptídeo Hidrolases/genética , Receptores de Superfície Celular/genética , Alinhamento de Sequência , Vibrio cholerae/genética , Vibrio cholerae/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...