Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
J Bacteriol ; 205(12): e0032423, 2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-37971230

RESUMO

IMPORTANCE: This study is the first example of C. difficile growing with siderophores as the sole iron source and describes the characterization of the ferric hydroxamate uptake ABC transporter (FhuDBGC). This transporter shows specificity to the siderophore ferrichrome. While not required for pathogenesis, this transporter highlights the redundancy in iron acquisition mechanisms that C. difficile uses to compete for iron during an infection.


Assuntos
Clostridioides difficile , Sideróforos , Ferro/metabolismo , Ferricromo/metabolismo , Clostridioides difficile/metabolismo , Clostridioides , Proteínas de Membrana Transportadoras
2.
Microbiol Spectr ; 11(3): e0049623, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37199664

RESUMO

The opportunistic fungal pathogen Aspergillus fumigatus utilizes two high-affinity iron uptake mechanisms, termed reductive iron assimilation (RIA) and siderophore-mediated iron acquisition (SIA). The latter has been shown to be crucial for virulence of this fungus and is a target for development of novel strategies for diagnosis and treatment of fungal infections. So far, research on SIA in this mold focused mainly on the hyphal stage, revealing the importance of extracellular fusarinine-type siderophores in iron acquisition as well as of the siderophore ferricrocin in intracellular iron handling. The current study aimed to characterize iron acquisition during germination. High expression of genes involved in biosynthesis and uptake of ferricrocin in conidia and during germination, independent of iron availability, suggested a role of ferricrocin in iron acquisition during germination. In agreement, (i) bioassays indicated secretion of ferricrocin during growth on solid media during both iron sufficiency and limitation, (ii) ferricrocin was identified in the supernatant of conidia germinating in liquid media during both iron sufficiency and limitation, (iii) in contrast to mutants lacking all siderophores, mutants synthesizing ferricrocin but lacking fusarinine-type siderophores were able to grow under iron limitation in the absence of RIA, and (iv) genetic inactivation of the ferricrocin transporter Sit1 decreased germination in the absence of RIA. Taken together, this study revealed that ferricrocin has not only an intracellular role but also functions as an extracellular siderophore to support iron acquisition. The iron availability-independent ferricrocin secretion and uptake during early germination indicate developmental, rather than iron regulation. IMPORTANCE Aspergillus fumigatus is one of the most common airborne fungal pathogens for humans. Low-molecular-mass iron chelators, termed siderophores, have been shown to play a central role in iron homeostasis and, consequently, virulence of this mold. Previous studies demonstrated the crucial role of secreted fusarinine-type siderophores, such as triacetylfusarinine C, in iron acquisition, as well as of the ferrichrome-type siderophore ferricrocin in intracellular iron storage and transport. Here, we demonstrate that ferricrocin is also secreted to mediate iron acquisition during germination together with reductive iron assimilation. During early germination, ferricrocin secretion and uptake were not repressed by iron availability, indicating developmental regulation of this iron acquisition system in this growth phase.


Assuntos
Ferricromo , Sideróforos , Humanos , Ferricromo/metabolismo , Aspergillus fumigatus/metabolismo , Ferro/metabolismo
3.
Mol Microbiol ; 119(3): 340-349, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36648393

RESUMO

In Bradyrhizobium japonicum, iron uptake from ferric siderophores involves selective outer membrane proteins and non-selective periplasmic and cytoplasmic membrane components that accommodate numerous structurally diverse siderophores. Free iron traverses the cytoplasmic membrane through the ferrous (Fe2+ ) transporter system FeoAB, but the other non-selective components have not been described. Here, we identify fsrB as an iron-regulated gene required for growth on iron chelates of catecholate- and hydroxymate-type siderophores, but not on inorganic iron. Utilization of the non-physiological iron chelator EDDHA as an iron source was also dependent on fsrB. Uptake activities of 55 Fe3+ bound to ferrioxamine B, ferrichrome or enterobactin were severely diminished in the fsrB mutant compared with the wild type. Growth of the fsrB or feoB strains on ferrichrome were rescued with plasmid-borne E. coli fhuCDB ferrichrome transport genes, suggesting that FsrB activity occurs in the periplasm rather than the cytoplasm. Whole cells of an fsrB mutant are defective in ferric reductase activity. Both whole cells and spheroplasts catalyzed the demetallation of ferric siderophores that were defective in an fsrB mutant. Collectively, the data support a model whereby FsrB is required for reduction of iron and its dissociation from the siderophore in the periplasm, followed by transport of the ferrous ion into the cytoplasm by FeoAB.


Assuntos
Ferro , Sideróforos , Sideróforos/metabolismo , Ferro/metabolismo , Ferricromo/metabolismo , Escherichia coli/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Compostos Férricos/metabolismo
4.
Environ Microbiol ; 25(4): 811-831, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36571575

RESUMO

Pseudomonas aeruginosa is a ubiquitous bacterium found in many natural and man-made environments. It is also a pathogen for plants, animals, and humans. As for almost all living organisms, iron is an essential nutrient for the growth of P. aeruginosa. The bacterium has evolved complex systems to access iron and maintain its homeostasis to survive in diverse natural and dynamic host environments. To access ferric iron, P. aeruginosa is able to produce two siderophores (pyoverdine and pyochelin), as well as use a variety of siderophores produced by other bacteria (mycobactins, enterobactin, ferrioxamine, ferrichrome, vibriobactin, aerobactin, rhizobactin and schizokinen). Furthermore, it can also use citrate, in addition to catecholamine neuromediators and plant-derived mono catechols, as siderophores. The P. aeruginosa genome also encodes three heme-uptake pathways (heme being an iron source) and one ferrous iron acquisition pathway. This review aims to summarize current knowledge concerning the molecular mechanisms involved in all the iron and heme acquisition strategies used by P. aeruginosa.


Assuntos
Ferro , Sideróforos , Humanos , Ferro/metabolismo , Sideróforos/metabolismo , Pseudomonas aeruginosa/metabolismo , Ferricromo/metabolismo , Transporte Biológico
5.
Sci Rep ; 11(1): 19624, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34608174

RESUMO

The putative ferricrocin synthetase gene ferS in the fungal entomopathogen Beauveria bassiana BCC 2660 was identified and characterized. The 14,445-bp ferS encodes a multimodular nonribosomal siderophore synthetase tightly clustered with Fusarium graminearum ferricrocin synthetase. Functional analysis of this gene was performed by disruption with the bar cassette. ΔferS mutants were verified by Southern and PCR analyses. HPLC and TLC analyses of crude extracts indicated that biosynthesis of ferricrocin was abolished in ΔferS. Insect bioassays surprisingly indicated that ΔferS killed the Spodoptera exigua larvae faster (LT50 59 h) than wild type (66 h). Growth and developmental assays of the mutant and wild type demonstrated that ΔferS had a significant increase in germination under iron depletion and radial growth and a decrease in conidiation. Mitotracker staining showed that the mitochondrial activity was enriched in ΔferS under both iron excess and iron depletion. Comparative transcriptomes between wild type and ΔferS indicated that the mutant was increased in the expression of eight cytochrome P450 genes and those in iron homeostasis, ferroptosis, oxidative stress response, ergosterol biosynthesis, and TCA cycle, compared to wild type. Our data suggested that ΔferS sensed the iron excess and the oxidative stress and, in turn, was up-regulated in the antioxidant-related genes and those in ergosterol biosynthesis and TCA cycle. These increased biological pathways help ΔferS grow and germinate faster than the wild type and caused higher insect mortality than the wild type in the early phase of infection.


Assuntos
Beauveria/crescimento & desenvolvimento , Beauveria/metabolismo , Ferricromo/análogos & derivados , Interações Hospedeiro-Patógeno , Insetos/microbiologia , Ferro/metabolismo , Animais , Beauveria/classificação , Beauveria/patogenicidade , Biologia Computacional , Ferricromo/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Regulação Fúngica da Expressão Gênica , Homeostase , Mutação , Estresse Oxidativo , Filogenia , Virulência/genética
6.
Int J Oncol ; 57(3): 721-732, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32705165

RESUMO

Pancreatic cancer is associated with a poor prognosis due to challenges in early detection, severe progression of the primary tumor, metastatic lesions, and resistance to antitumor agents. However, previous studies have indicated a relationship between the microbiome and pancreatic cancer outcomes. Our previous study demonstrated that ferrichrome derived from Lactobacillus casei, a probiotic bacteria, exhibited tumor­suppressive effects in colorectal and gastric cancer, and that the suppressive effects were stronger than conventional antitumor agents, such as 5­fluorouracil (5­FU) and cisplatin, suggesting that certain probiotics exert antitumorigenic effects. However, whether or not probiotic­derived molecules, including ferrichrome, exert a tumor­suppressive effect in other gastrointestinal tumors, such as pancreatic cancer, remains unclear. In the present study, it was demonstrated that probiotic­derived ferrichrome inhibited the growth of pancreatic cancer cells, and its tumor­suppressive effects were further revealed in 5­FU­resistant pancreatic cancer cells in vitro and in vivo in a mouse xenograft model. Ferrichrome inhibited the progression of cancer cells via dysregulation of the cell cycle by activating p53. DNA fragmentation and cleavage of poly (ADP­ribose) polymerase were induced by ferrichrome treatment, suggesting that ferrichrome induced apoptosis in pancreatic cancer cells. A transcriptome analysis revealed that the expression p53­associated mRNAs was significantly altered by ferrichrome treatment. Thus, the tumor­suppressive effects of probiotics may mediated by probiotic­derived molecules, such as ferrichrome, which may have applications as an antitumor drug, even in refractory and 5­FU­resistant pancreatic cancer.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ferricromo/farmacologia , Lacticaseibacillus casei/química , Neoplasias Pancreáticas/tratamento farmacológico , Probióticos/química , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ferricromo/metabolismo , Ferricromo/uso terapêutico , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Humanos , Injeções Intravenosas , Masculino , Camundongos , Neoplasias Pancreáticas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Angew Chem Int Ed Engl ; 59(9): 3558-3562, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-31863717

RESUMO

Albomycin δ2 is a sulfur-containing sideromycin natural product that shows potent antibacterial activity against clinically important pathogens. The l-serine-thioheptose dipeptide partial structure, known as SB-217452, has been found to be the active seryl-tRNA synthetase inhibitor component of albomycin δ2 . Herein, it is demonstrated that AbmF catalyzes condensation between the 6'-amino-4'-thionucleoside with the d-ribo configuration and seryl-adenylate supplied by the serine adenylation activity of AbmK. Formation of the dipeptide is followed by C3'-epimerization to produce SB-217452 with the d-xylo configuration, which is catalyzed by the radical S-adenosyl-l-methionine enzyme AbmJ. Gene deletion suggests that AbmC is involved in peptide assembly linking SB-217452 with the siderophore moiety. This study establishes how the albomycin biosynthetic machinery generates its antimicrobial component SB-217452.


Assuntos
Antibacterianos/biossíntese , Ferricromo/análogos & derivados , Pirimidinonas/metabolismo , Serina-tRNA Ligase/metabolismo , Tiofenos/metabolismo , Antibacterianos/química , Biocatálise , Ferricromo/química , Ferricromo/metabolismo , Peptídeo Sintases/metabolismo , Pirimidinonas/química , Serina-tRNA Ligase/antagonistas & inibidores , Serina-tRNA Ligase/genética , Streptomyces/química , Streptomyces/metabolismo , Tiofenos/química
8.
J Am Chem Soc ; 141(6): 2211-2214, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30673214

RESUMO

Albomycins are peptidyl thionucleoside natural products that display antimicrobial activity against clinically important pathogens. Their structures are characterized by a thioheptose with atypical stereochemistry including a d-xylofuranose ring modified with a d-amino acid moiety. Herein it is demonstrated that AbmH is a pyridoxal 5'-phosphate (PLP)-dependent transaldolase that catalyzes a threo-selective aldol-type reaction to generate the thioheptose core with a d-ribofuranose ring and an l-amino acid moiety. The conversion of l-to d-amino acid configuration is catalyzed by the PLP-dependent epimerase AbmD. The d- ribo to d- xylo conversion of the thiofuranose ring appears according to gene deletion experiments to be mediated by AbmJ, which is annotated as a radical S-adenosyl-l-methionine (SAM) enzyme. These studies establish several key steps in the assembly of the thioheptose core during the biosynthesis of albomycins.


Assuntos
Antibacterianos/biossíntese , Antibacterianos/química , Ferricromo/análogos & derivados , Heptoses/química , Nucleosídeos/química , Biocatálise , Ferricromo/química , Ferricromo/metabolismo , Estereoisomerismo , Transaldolase/metabolismo
9.
Funct Integr Genomics ; 19(1): 137-150, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30251029

RESUMO

Aureobasidium melanogenum HN6.2 is a unique yeast strain who can produce the siderophore of fusigen under iron starvation to guarantee its survival. However, a comprehensive understanding of mechanisms involved in iron acquisition and homeostasis for it is still vacant. In this study, genome sequencing and mining revealed that A. melanogenum HN6.2 strain was the first yeast species that exclusively possessed all the four known mechanisms for the iron acquisition: (i) the siderophore-mediated iron uptake; (ii) reductive iron assimilation; (iii) low-affinity ferrous uptake; and (iv) heme utilization, which suggested its stronger adaptability than Aspergillus fumigatus and Saccharomyces cerevisiae. This HN6.2 strain also employed the vacuolar iron storage for immobilizing the excessive iron to avoid its cellular toxicity. Specially, genome mining indicated that A. melanogenum HN6.2 strain could also synthesize ferricrocin siderophore. Further HPLC and Q-Tof-MS analysis confirmed that the siderophores synthesized by this strain consisted of cyclic fusigen, linear fusigen, ferricrocin, and hydroxyferricrocin and they played parallel roles as both intracellular and extracellular siderophores. Also, the heme utilization for this strain was experimentally verified by the knock-out of heme oxygenase gene. For iron homeostasis, the transcriptome analysis revealed that this strain mainly employed two central regulators of SreA/HapX to tune iron uptake and storage at the transcriptional level. It was also noted that mitogen-activated protein kinase C gene (MpkC) exhibited a transcriptional up-regulation under iron sufficiency, suggesting that it may serve as another factor involved in the repression of siderophore biosynthesis. This is the first genetic blueprint of iron acquisition and homeostasis for A. melanogenum.


Assuntos
Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Genoma Fúngico , Ferro/metabolismo , Saccharomycetales/metabolismo , Sideróforos/metabolismo , Transcriptoma , Aspergillus fumigatus/genética , Aspergillus fumigatus/metabolismo , Mineração de Dados , Compostos Férricos/metabolismo , Ferricromo/análogos & derivados , Ferricromo/metabolismo , Compostos Ferrosos/metabolismo , Proteínas Fúngicas/metabolismo , Ontologia Genética , Heme/metabolismo , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase (Desciclizante)/metabolismo , Homeostase/genética , Ácidos Hidroxâmicos/metabolismo , Transporte de Íons , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Anotação de Sequência Molecular , Oxirredução , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomycetales/genética , Vacúolos/metabolismo
10.
Sci Rep ; 6: 35306, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27748436

RESUMO

Ergothioneine (EGT; 2-mercaptohistidine trimethylbetaine) is a trimethylated and sulphurised histidine derivative which exhibits antioxidant properties. Here we report that deletion of Aspergillus fumigatus egtA (AFUA_2G15650), which encodes a trimodular enzyme, abrogated EGT biosynthesis in this opportunistic pathogen. EGT biosynthetic deficiency in A. fumigatus significantly reduced resistance to elevated H2O2 and menadione, respectively, impaired gliotoxin production and resulted in attenuated conidiation. Quantitative proteomic analysis revealed substantial proteomic remodelling in ΔegtA compared to wild-type under both basal and ROS conditions, whereby the abundance of 290 proteins was altered. Specifically, the reciprocal differential abundance of cystathionine γ-synthase and ß-lyase, respectively, influenced cystathionine availability to effect EGT biosynthesis. A combined deficiency in EGT biosynthesis and the oxidative stress response regulator Yap1, which led to extreme oxidative stress susceptibility, decreased resistance to heavy metals and production of the extracellular siderophore triacetylfusarinine C and increased accumulation of the intracellular siderophore ferricrocin. EGT dissipated H2O2 in vitro, and elevated intracellular GSH levels accompanied abrogation of EGT biosynthesis. EGT deficiency only decreased resistance to high H2O2 levels which suggests functionality as an auxiliary antioxidant, required for growth at elevated oxidative stress conditions. Combined, these data reveal new interactions between cellular redox homeostasis, secondary metabolism and metal ion homeostasis.


Assuntos
Aspergillus fumigatus/metabolismo , Ergotioneína/biossíntese , Proteínas Fúngicas/metabolismo , Antioxidantes/química , Aspergillus fumigatus/genética , Carbono-Oxigênio Liases/metabolismo , Compostos Férricos/química , Ferricromo/análogos & derivados , Ferricromo/metabolismo , Proteínas Fúngicas/genética , Deleção de Genes , Teste de Complementação Genética , Gliotoxina/química , Glutationa/metabolismo , Histidina/química , Peróxido de Hidrogênio/química , Ácidos Hidroxâmicos/química , Liases/metabolismo , Metais Pesados/química , Oxirredução , Estresse Oxidativo , Proteômica/métodos , Espécies Reativas de Oxigênio/metabolismo , Sideróforos/química , Vitamina K 3/química
11.
Biochem J ; 473(9): 1203-13, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26929401

RESUMO

Aspergillus fumigatus is an opportunistic fungal pathogen for immunocompromised patients, and genes involved in siderophore metabolism have been identified as virulence factors. Recently, we identified the membrane transporters sit1 and sit2, which are putative virulence factors of A. fumigatus; sit1 and sit2 are homologous to yeast Sit1, and sit1 and sit2 gene expression was up-regulated after iron depletion. When expressed heterologously in Saccharomyces cerevisiae, sit1 and sit2 were localized to the plasma membrane; sit1 efficiently complemented ferrichrome (FC) and ferrioxamine B (FOB) uptake in yeast cells, whereas sit2 complemented only FC uptake. Deletion of sit1 resulted in a decrease in FOB and FC uptake, and deletion of sit2 resulted in a decrease in FC uptake in A. fumigatus It is of interest that a sit1 and sit2 double-deletion mutant resulted in a synergistic decrease in FC uptake activity. Both sit1 and sit2 were localized to the plasma membrane in A. fumigatus The expression levels of the sit1 and sit2 genes were dependent on hapX under low-but not high-iron conditions. Furthermore, mirB, and sidA gene expression was up-regulated and sreA expression down-regulated when sit1 and sit2 were deleted. Although sit1 and sit2 failed to affect mouse survival rate, these genes affected conidial killing activity. Taken together, our results suggest that sit1 and sit2 are siderophore transporters and putative virulence factors localized to the plasma membrane.


Assuntos
Aspergillus fumigatus/metabolismo , Membrana Celular/metabolismo , Desferroxamina/metabolismo , Compostos Férricos/metabolismo , Ferricromo/metabolismo , Ferro/metabolismo , Fatores de Virulência/metabolismo , Animais , Aspergillus fumigatus/genética , Aspergillus fumigatus/patogenicidade , Membrana Celular/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Fatores de Virulência/genética
12.
Appl Environ Microbiol ; 82(2): 467-77, 2016 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-26519385

RESUMO

Albomycin (ABM), also known as grisein, is a sulfur-containing metabolite produced by Streptomyces griseus ATCC 700974. Genes predicted to be involved in the biosynthesis of ABM and ABM-like molecules are found in the genomes of other actinomycetes. ABM has potent antibacterial activity, and as a result, many attempts have been made to develop ABM into a drug since the last century. Although the productivity of S. griseus can be increased with random mutagenesis methods, understanding of Streptomyces sulfur amino acid (SAA) metabolism, which supplies a precursor for ABM biosynthesis, could lead to improved and stable production. We previously characterized the gene cluster (abm) in the genome-sequenced S. griseus strain and proposed that the sulfur atom of ABM is derived from either cysteine (Cys) or homocysteine (Hcy). The gene product, AbmD, appears to be an important link between primary and secondary sulfur metabolic pathways. Here, we show that propargylglycine or iron supplementation in growth media increased ABM production by significantly changing the relative concentrations of intracellular Cys and Hcy. An SAA metabolic network of S. griseus was constructed. Pathways toward increasing Hcy were shown to positively impact ABM production. The abmD gene and five genes that increased the Hcy/Cys ratio were assembled downstream of hrdBp promoter sequences and integrated into the chromosome for overexpression. The ABM titer of one engineered strain, SCAK3, in a chemically defined medium was consistently improved to levels ∼400% of the wild type. Finally, we analyzed the production and growth of SCAK3 in shake flasks for further process development.


Assuntos
Aminoácidos Sulfúricos/metabolismo , Antibacterianos/biossíntese , Ferricromo/análogos & derivados , Streptomyces griseus/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Ferricromo/metabolismo , Família Multigênica , Streptomyces griseus/genética , Streptomyces griseus/crescimento & desenvolvimento
13.
FEMS Microbiol Lett ; 362(2): 1-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25670702

RESUMO

Iron is an essential element for life. However, the iron overload can be toxic. Here, we investigated the significant increase of tenellin and iron-tenellin complex production in ferricrocin-deficient mutants of Beauveria bassiana. Our chemical analysis indicated that the ferricrocin-deficient mutants T1, T3 and T5 nearly abolished ferricrocin production. In turn, these mutants had significant accumulation of iron-tenellin complex in their mycelia at 247-289 mg g(-1) cell dry weight under iron-replete condition. Both tenellin and iron-tenellin complex were not detected in the wild-type under such condition. Mass analysis of the mutants' crude extracts demonstrated that tenellin formed a 3:1 complex with iron in the absence of ferricrocin. The unexpected link between ferricrocin and tenellin biosynthesis in ferricrocin-deficient mutants could be a survival strategy during iron-mediated oxidative stress.


Assuntos
Beauveria/metabolismo , Ferricromo/análogos & derivados , Ferro/metabolismo , Piridonas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sideróforos/metabolismo , Beauveria/química , Beauveria/genética , Beauveria/ultraestrutura , Cromatografia Líquida de Alta Pressão , Ferricromo/química , Ferricromo/metabolismo , Espectrometria de Massas , Mutação , Piridonas/química , Interferência de RNA
14.
PLoS One ; 9(8): e105953, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170896

RESUMO

Iron is an essential nutrient for the growth of most bacteria. To obtain iron, bacteria have developed specific iron-transport systems located on the membrane surface to uptake iron and iron complexes such as ferrichrome. Interference with the iron-acquisition systems should be therefore an efficient strategy to suppress bacterial growth and infection. Based on the chemical similarity of iron and ruthenium, we used a Ru(II) complex R-825 to compete with ferrichrome for the ferrichrome-transport pathway in Streptococcus pneumoniae. R-825 inhibited the bacterial growth of S. pneumoniae and stimulated the expression of PiuA, the iron-binding protein in the ferrichrome-uptake system on the cell surface. R-825 treatment decreased the cellular content of iron, accompanying with the increase of Ru(II) level in the bacterium. When the piuA gene (SPD_0915) was deleted in the bacterium, the mutant strain became resistant to R-825 treatment, with decreased content of Ru(II). Addition of ferrichrome can rescue the bacterial growth that was suppressed by R-825. Fluorescence spectral quenching showed that R-825 can bind with PiuA in a similar pattern to the ferrichrome-PiuA interaction in vitro. These observations demonstrated that Ru(II) complex R-825 can compete with ferrichrome for the ferrichrome-transport system to enter S. pneumoniae, reduce the cellular iron supply, and thus suppress the bacterial growth. This finding suggests a novel antimicrobial approach by interfering with iron-uptake pathways, which is different from the mechanisms used by current antibiotics.


Assuntos
Ferro/metabolismo , Compostos Organometálicos/farmacologia , Rutênio/química , Streptococcus pneumoniae/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Complexos de Coordenação/farmacologia , Relação Dose-Resposta a Droga , Ferricromo/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Mutação , Compostos Organometálicos/química , Compostos Organometálicos/metabolismo , Ligação Proteica , Rutênio/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crescimento & desenvolvimento
15.
PLoS One ; 8(8): e71451, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951167

RESUMO

Iron scarcity is one of the nutrition limitations that the Gram-positive infectious pathogens Streptococcus pneumoniae encounter in the human host. To guarantee sufficient iron supply, the ATP binding cassette (ABC) transporter Pia is employed to uptake iron chelated by hydroxamate siderophore, via the membrane-anchored substrate-binding protein PiaA. The high affinity towards ferrichrome enables PiaA to capture iron at a very low concentration in the host. We presented here the crystal structures of PiaA in both apo and ferrichrome-complexed forms at 2.7 and 2.1 Å resolution, respectively. Similar to other class III substrate binding proteins, PiaA is composed of an N-terminal and a C-terminal domain bridged by an α-helix. At the inter-domain cleft, a molecule of ferrichrome is stabilized by a number of highly conserved residues. Upon ferrichrome binding, two highly flexible segments at the entrance of the cleft undergo significant conformational changes, indicating their contribution to the binding and/or release of ferrichrome. Superposition to the structure of Escherichia coli ABC transporter BtuF enabled us to define two conserved residues: Glu119 and Glu262, which were proposed to form salt bridges with two arginines of the permease subunits. Further structure-based sequence alignment revealed that the ferrichrome binding pattern is highly conserved in a series of PiaA homologs encoded by both Gram-positive and negative bacteria, which were predicted to be sensitive to albomycin, a sideromycin antibiotic derived from ferrichrome.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Bactérias/metabolismo , Ferricromo/metabolismo , Streptococcus pneumoniae/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
16.
Metallomics ; 4(12): 1262-70, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23151814

RESUMO

Iron is an essential element for all eukaryotes but its excess has deleterious effects. Aspergillus fumigatus produces extracellular siderophores for iron uptake and the intracellular siderophore ferricrocin (FC) for distribution and storage of iron. Iron excess has previously been shown to increase the content of ferric FC and the expression of the putative vacuolar iron importer CccA (AFUA_4G12530), indicating a role of both the vacuole and FC in iron detoxification. In this study, we show that CccA-deficiency decreases iron resistance in particular in combination with derepressed iron uptake, while overproduction of CccA increases iron resistance. Green fluorescence protein-tagging confirmed localization of CccA in the vacuolar membrane. In contrast to CccA-deficiency, inactivation of FC biosynthesis did not affect iron resistance, which indicates that vacuolar rather than FC-mediated iron storage is the major iron detoxifying mechanism. After uptake, extracellular siderophore backbones are hydrolyzed and recycled. Lack of FC, CccA, and in particular lack of both increased the cellular content of iron chelated by siderophore breakdown products. These data indicate that the transfer of iron from extracellular siderophores to the metabolism, FC or the vacuole precedes recycling of siderophore breakdown products. Furthermore, this study indicates that CccA does not play an exclusive role in vacuolar iron storage for nutritional reuse.


Assuntos
Aspergillus fumigatus/metabolismo , Ferro/metabolismo , Sideróforos/metabolismo , Aspergillus fumigatus/genética , Transporte Biológico Ativo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Ferricromo/análogos & derivados , Ferricromo/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Filogenia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade da Espécie , Vacúolos/metabolismo
17.
Arch Microbiol ; 192(11): 899-908, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20737137

RESUMO

Nitrosomonas europaea has a single three-gene operon (nitABC) encoding an iron ABC transporter system (NitABC). Phylogenetic analysis clustered the subunit NitB with Fe(3+)-ABC transporter permease components from other organisms. The N. europaea strain deficient in nitB (nitB::kan) grew well in either Fe-replete or Fe-limited media and in Fe-limited medium containing the catecholate-type siderophore, enterobactin or the citrate-based dihydroxamate-type siderophore, aerobactin. However, the nitB::kan mutant strain was unable to grow in Fe-limited media containing either the hydroxamate-type siderophores, ferrioxamine and ferrichrome or the mixed-chelating type siderophore, pyoverdine. Exposure of N. europaea cells to a ferrichrome analog coupled to the fluorescent moiety naphthalic diimide (Fhu-NI) led to increase in fluorescence in the wild type but not in nitB::kan mutant cells. Spheroplasts prepared from N. europaea wild type exposed to Fhu-NI analog retained the fluorescence, while spheroplasts of the nitB::kan mutant were not fluorescent. NitABC transports intact Fe(3+)-ferrichrome complex into the cytoplasm and is an atypical ABC type iron transporter for Fe(3+) bound to ferrioxamine, ferrichrome or pyoverdine siderophores into the cytoplasm. The mechanisms to transport iron in either the Fe(3+) or Fe(2+) forms or Fe(3+) associated with enterobactin or aerobactin siderophores into the cell across the cytoplasmic membrane are as yet undetermined.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Nitrosomonas europaea/metabolismo , Sideróforos/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Proteínas da Membrana Bacteriana Externa/genética , Transporte Biológico , Membrana Celular/metabolismo , Desferroxamina/metabolismo , Enterobactina/metabolismo , Compostos Férricos/metabolismo , Ferricromo/metabolismo , Genes Bacterianos , Ácidos Hidroxâmicos/metabolismo , Mutagênese , Mutação , Nitrosomonas europaea/genética , Nitrosomonas europaea/crescimento & desenvolvimento , Oligopeptídeos/metabolismo , Óperon , Filogenia , RNA Bacteriano/genética
18.
Appl Environ Microbiol ; 76(12): 3806-17, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20435771

RESUMO

The fission yeast Schizosaccharomyces pombe excretes and accumulates the hydroxamate-type siderophore ferrichrome. The sib1(+) and sib2(+) genes encode, respectively, a siderophore synthetase and an l-ornithine N(5)-oxygenase that participate in ferrichrome biosynthesis. In the present report, we demonstrate that sib1(+) and sib2(+) are repressed by the GATA-type transcriptional repressor Fep1 in response to high levels of iron. We further found that the loss of Fep1 results in increased ferrichrome production. We showed that a sib1Delta sib2Delta mutant strain exhibits a severe growth defect on iron-poor media. We determined that two metabolic pathways are involved in biosynthesis of ornithine, an obligatory precursor of ferrichrome. Ornithine is produced by hydrolysis of arginine by the Car1 and Car3 proteins. Although car3(+) was constitutively expressed, car1(+) transcription levels were repressed upon exposure to iron, with a concomitant decrease of Car1 arginase activity. Ornithine is also generated by transformation of glutamate, which itself is produced by two separate biosynthetic pathways which are transcriptionally regulated by iron in an opposite fashion. In one pathway, the glutamate dehydrogenase Gdh1, which produces glutamate from 2-ketoglutarate, was repressed under iron-replete conditions in a Fep1-dependent manner. The other pathway involves two coupled enzymes, glutamine synthetase Gln1 and Fe-S cluster-containing glutamate synthase Glt1, which were both repressed under iron-limiting conditions but were expressed under iron-replete conditions. Collectively, these results indicate that under conditions of iron deprivation, yeast remodels metabolic pathways linked to ferrichrome synthesis in order to limit iron utilization without compromising siderophore production and its ability to sequester iron from the environment.


Assuntos
Ferricromo/metabolismo , Regulação Fúngica da Expressão Gênica , Ferro/metabolismo , Redes e Vias Metabólicas/genética , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Arginina/metabolismo , Meios de Cultura/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Deleção de Genes , Ácido Glutâmico/metabolismo , Modelos Biológicos , Ornitina/metabolismo , Schizosaccharomyces/crescimento & desenvolvimento
19.
J Bacteriol ; 192(5): 1212-20, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20047910

RESUMO

The uptake of iron into Pseudomonas aeruginosa is mediated by two major siderophores produced by the bacterium, pyoverdine and pyochelin. The bacterium is also able of utilize several heterologous siderophores of bacterial or fungal origin. In this work, we have investigated the iron uptake in P. aeruginosa PAO1 by the heterologous ferrichrome siderophore. (55)Fe uptake assays showed that ferrichrome is transported across the outer membrane primarily (80%) by the FiuA receptor and to a lesser extent (20%) by a secondary transporter. Moreover, we demonstrate that like in the uptake of ferripyoverdine and ferripyochelin, the energy required for both pathways of ferrichrome uptake is provided by the inner membrane protein TonB1. Desferrichrome-(55)Fe uptake in P. aeruginosa was also dependent on the expression of the permease FiuB, suggesting that this protein is the inner membrane transporter of the ferrisiderophore. A biomimetic fluorescent analogue of ferrichrome, RL1194, was used in vivo to monitor the kinetics of iron release from ferrichrome in P. aeruginosa in real time. This dissociation involves acylation of ferrichrome and its biomimetic analogue RL1194 and recycling of both modified siderophores into the extracellular medium. FiuC, an N-acetyltransferase, is certainly involved in this mechanism of iron release, since its mutation abolished desferrichrome-(55)Fe uptake. The acetylated derivative reacts with iron in the extracellular medium and is able to be taken up again by the cells. All these observations are discussed in light of the current knowledge concerning ferrichrome uptake in P. aeruginosa and in Escherichia coli.


Assuntos
Ferricromo/metabolismo , Ferro/metabolismo , Pseudomonas aeruginosa/metabolismo , Acilação , Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Redes e Vias Metabólicas
20.
Fungal Genet Biol ; 46(9): 707-13, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19460452

RESUMO

Zinc plays a critical role in a diverse array of biochemical processes. However, excess of zinc is deleterious to cells. Therefore, cells require finely tuned homeostatic mechanisms to balance uptake and storage of zinc. Here we show that iron starvation affects zinc metabolism by downregulating expression of the plasma membrane zinc importer encoding zrfB and upregulating the putative vacuolar zinc transporter-encoding zrcA in Aspergillus fumigatus. Nevertheless, the zinc content of iron-starved mycelia exceeded that of iron replete mycelia, possibly due to unspecific metal uptake induced by iron starvation. In agreement with increased zinc excess and zinc toxicity during iron starvation, deficiency in siderophore-mediated high-affinity iron uptake caused hypersensitivity to zinc. Moreover, an increase of zinc uptake by conditional overexpression of zrfB was more toxic under iron depleted compared to iron replete conditions. This deregulated zinc uptake under iron starvation caused a decrease in heme production and an increase in protoporphyrin IX accumulation. Furthermore, zinc excess impaired production of the extracellular siderophore triacetylfusarinine C but not the intracellular siderophore ferricrocin. Taken together, these data demonstrate a fine tuned coordination of zinc and iron metabolism in A. fumigatus.


Assuntos
Aspergillus fumigatus/metabolismo , Ferro/metabolismo , Zinco/metabolismo , Aspergillus fumigatus/genética , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Compostos Férricos/metabolismo , Ferricromo/análogos & derivados , Ferricromo/metabolismo , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Heme/biossíntese , Homeostase , Ácidos Hidroxâmicos/metabolismo , Micélio/metabolismo , Protoporfirinas/metabolismo , RNA Fúngico/análise , RNA Fúngico/biossíntese , RNA Fúngico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA