Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Synth Biol ; 9(8): 1968-1977, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32786921

RESUMO

Glarea lozoyensis is an important industrial fungus that produces the pneumocandin B0, which is used for the synthesis of antifungal drug caspofungin. However, because of the limitations and complications of traditional genetic tools, G. lozoyensis strain engineering has been hindered. In this study, we established an efficient CRISPR/Cas9-based gene editing tool in G. lozoyensis SIPI1208. With this method, gene mutagenesis efficiency in the target locus can be up to 80%, which enables the rapid gene knockout. According to the reports, GloF and Ap-HtyE, proline hydroxylases involved in pneumocandin and Echinocandin B biosynthesis, respectively, can catalyze the proline to generate different ratios of trans-3-hydroxy-l-proline to trans-4-hydroxy-l-proline. Heterologous expression of Ap-HtyE in G. lozoyensis decreased the ratio of pneumocandin C0 to (pneumocandin B0 + pneumocandin C0) from 33.5% to 11% without the addition of proline to the fermentation medium. Furthermore, the gloF was replaced by ap-htyE to study the production of pneumocandin C0. However, the gene replacement has been hampered by traditional gene tools since gloF and gloG, two contiguous genes indispensable in the biosynthesis of pneumocandins, are cotranscribed into one mRNA. With the CRISPR/Cas9 strategy, ap-htyE was knocked in and successfully replaced gloF, and results showed that the knock-in strain retained the ability to produce pneumocandin B0, but the production of pneumocandin C0 was abolished. Thus, this strain displayed a competitive advantage in the industrial production of pneumocandin B0. In summary, this study showed that the CRISPR/Cas9-based gene editing tool is efficient for manipulating genes in G. lozoyensis.


Assuntos
Ascomicetos/genética , Sistemas CRISPR-Cas/genética , Proteínas Fúngicas/genética , Edição de Genes/métodos , Equinocandinas/biossíntese , Equinocandinas/química , Proteínas Fúngicas/metabolismo , Mutagênese Sítio-Dirigida , Prolil Hidroxilases/genética , Prolil Hidroxilases/metabolismo , RNA Guia de Cinetoplastídeos/metabolismo
2.
Environ Microbiol ; 22(6): 2292-2311, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32239586

RESUMO

The antifungal echinocandin lipopeptide, acrophiarin, was circumscribed in a patent in 1979. We confirmed that the producing strain NRRL 8095 is Penicillium arenicola and other strains of P. arenicola produced acrophiarin and acrophiarin analogues. Genome sequencing of NRRL 8095 identified the acrophiarin gene cluster. Penicillium arenicola and echinocandin-producing Aspergillus species belong to the family Aspergillaceae of the Eurotiomycetes, but several features of acrophiarin and its gene cluster suggest a closer relationship with echinocandins from Leotiomycete fungi. These features include hydroxy-glutamine in the peptide core instead of a serine or threonine residue, the inclusion of a non-heme iron, α-ketoglutarate-dependent oxygenase for hydroxylation of the C3 of the glutamine, and a thioesterase. In addition, P. arenicola bears similarity to Leotiomycete echinocandin-producing species because it exhibits self-resistance to exogenous echinocandins. Phylogenetic analysis of the genes of the echinocandin biosynthetic family indicated that most of the predicted proteins of acrophiarin gene cluster exhibited higher similarity to the predicted proteins of the pneumocandin gene cluster of the Leotiomycete Glarea lozoyensis than to those of the echinocandin B gene cluster from A. pachycristatus. The fellutamide gene cluster and related gene clusters are recognized as relatives of the echinocandins. Inclusion of the acrophiarin gene cluster into a comprehensive phylogenetic analysis of echinocandin gene clusters indicated the divergent evolutionary lineages of echinocandin gene clusters are descendants from a common ancestral progenitor. The minimal 10-gene cluster may have undergone multiple gene acquisitions or losses and possibly horizontal gene transfer after the ancestral separation of the two lineages.


Assuntos
Anti-Infecciosos/metabolismo , Ascomicetos , Aspergillus , Equinocandinas , Lipopeptídeos , Penicillium , Ascomicetos/genética , Aspergillus/genética , Equinocandinas/biossíntese , Equinocandinas/genética , Lipopeptídeos/biossíntese , Lipopeptídeos/genética , Família Multigênica , Penicillium/genética
3.
World J Microbiol Biotechnol ; 35(7): 109, 2019 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-31280382

RESUMO

Echinocandin B (ECB) is an important lipohexapeptide used for chemical manufacture of the antifungal agent anidulafungin. Sterigmatocystin (ST) is a polyketide mycotoxin produced by certain species of Aspergillus such as Aspergillus delacroxii SIPIW15, which could produce both ECB and ST. However, the presence of the potent carcinogen ST will greatly affect the quality and safety of ECB production. Therefore, it is essential to eliminate the ST biosynthesis and increase ECB titers in Asp. delacroxii SIPIW15. In this study, the polyketide synthase gene (stcA) required for biosynthesis of ST and its flanking region in Asp. delacroxii SIPIW15 were cloned, sequenced and analyzed firstly. Based on Agrobacterium-mediated transformation, the ΔstcA mutant AMT-1 was obtained and its yield of ECB was increased by 40% without ST detected at the same time as compared to the original strain. The results of the fed-batch experiments showed that the ECB yield of the ΔstcA strain AMT-1 was increased to 2163 ± 31 mg/l and no ST was detected in the 50 l bioreactor. This work suggested that the ΔstcA strain AMT-1 has the potential for application in ECB production improvement, and more importantly, to eliminate ST-related environmental pollution in ECB fermentation industry.


Assuntos
Aspergillus/genética , Aspergillus/metabolismo , Equinocandinas/biossíntese , Equinocandinas/genética , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Genes Fúngicos/genética , Policetídeo Sintases/genética , Esterigmatocistina/biossíntese , Agrobacterium/genética , Anidulafungina , Antifúngicos , Sequência de Bases , Técnicas de Cultura Celular por Lotes , Reatores Biológicos , DNA Fúngico/isolamento & purificação , Fermentação , Engenharia Metabólica , Redes e Vias Metabólicas/genética , Metabolismo Secundário/genética , Transformação Genética
4.
Biotechnol Appl Biochem ; 66(4): 626-633, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31069846

RESUMO

The ecdB is a transcription factor, located in the echinocandin B biosynthetic gene cluster of Emericella rugulosa NRRL11440. Here, we validated the ecdB mRNA sequence for functional expression and to explore the role of EcdB protein in the echinocandin B regulation. The sequence alignment study revealed that the ecdB coding sequence was found 75 bp shorter than the reference mRNA sequence. This coding sequence encodes for EcdB protein and comprises three conserved domains; DNA binding domain (DBD), coiled-coil domain, and signature middle homology region. The full-length and DBD (truncated) DNA sequences were expressed in Escherichia coli BL21(DE3) under different tested conditions. The expression of EcdB protein was found to be toxic, which curbs the cell growth. In contrast to truncated protein (GST:EcdB1-54), the full-length (GST:EcdB) protein was expressed at very low titer and not detectable in SDS-PAGE under the varying isopropyl ß-d-1-thiogalactopyranoside (IPTG), temperature, and media conditions. However, GST:EcdB1-54 was successfully purified under standard conditions (0.5 mM IPTG at 0.5OD) with 33 kDa expected size. The functionality of GST:EcdB1-54 was attained by electrophoretic mobility shift assay study as a clear band shifting showed with ecdA promoter. Taken together, we conclude that EcdB interacts with the ecdA promoter that reflected to require for echinocandin B regulation.


Assuntos
Aspergillus nidulans/metabolismo , Equinocandinas/biossíntese , Proteínas Fúngicas/biossíntese , Família Multigênica , Aspergillus nidulans/química , Equinocandinas/genética , Equinocandinas/isolamento & purificação , Proteínas Fúngicas/genética , Proteínas Fúngicas/isolamento & purificação , Família Multigênica/genética
5.
Appl Microbiol Biotechnol ; 102(23): 9881-9891, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30255232

RESUMO

Fungal diseases are a global public health problem. Invasive fungal infections pose a serious threat to patients with compromised immune systems, such as those undergoing organ or bone marrow transplants, cancer, or HIV/AIDS. Pneumocandins are antifungal lipohexapeptides of the echinocandin family that noncompetitively inhibit of 1,3-ß-glucan synthase of fungal cell wall and provide the precursor for the semisynthesis of caspofungin, which is widely used as first-line therapy for invasive fungal infections. Recently, the biosynthetic steps leading to formation of pneumocandin B0 and echinocandin B have been elucidated, and thus, provide a framework and attractive model for further design new antifungal therapeutics around natural variations in echinocandin structural diversities via genetic and chemical tools. In this article, we analyze the biosynthetic pathway of pneumocandins and other echinocandins, provide an update on the array of pneumocandin analogues generated by genetic manipulation, and summarize advances in the enhancement of pneumocandin B0 production by random mutagenesis and fermentation optimization. We also give offer advice on the development of improved pneumocandin drug candidates and more efficient production of pneumocandin B0.


Assuntos
Equinocandinas/biossíntese , Equinocandinas/farmacologia , Fungos/metabolismo , Antifúngicos/química , Antifúngicos/farmacologia , Vias Biossintéticas , Equinocandinas/química , Fermentação , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/química , Proteínas Fúngicas/farmacologia , Microbiologia Industrial
6.
Appl Environ Microbiol ; 84(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29352089

RESUMO

Echinocandins are antifungal nonribosomal hexapeptides produced by fungi. Two of the amino acids are hydroxy-l-prolines: trans-4-hydroxy-l-proline and, in most echinocandin structures, (trans-2,3)-3-hydroxy-(trans-2,4)-4-methyl-l-proline. In the case of echinocandin biosynthesis by Glarea lozoyensis, both amino acids are found in pneumocandin A0, while in pneumocandin B0 the latter residue is replaced by trans-3-hydroxy-l-proline (3-Hyp). We have recently reported that all three amino acids are generated by the 2-oxoglutarate-dependent proline hydroxylase GloF. In echinocandin B biosynthesis by Aspergillus species, 3-Hyp derivatives have not been reported. Here we describe the heterologous production and kinetic characterization of HtyE, the 2-oxoglutarate-dependent proline hydroxylase from the echinocandin B biosynthetic cluster in Aspergillus pachycristatus Surprisingly, l-proline hydroxylation with HtyE resulted in an even higher proportion (∼30%) of 3-Hyp than that with GloF. This suggests that the selectivity for methylated 3-Hyp in echinocandin B biosynthesis is due solely to a substrate-specific adenylation domain of the nonribosomal peptide synthetase. Moreover, we observed that one product of HtyE catalysis, 3-hydroxy-4-methyl-l-proline, is slowly further oxidized at the methyl group, giving 3-hydroxy-4-hydroxymethyl-l-proline, upon prolonged incubation with HtyE. This dihydroxylated amino acid has been reported as a building block of cryptocandin, an echinocandin produced by CryptosporiopsisIMPORTANCE Secondary metabolites from bacteria and fungi are often produced by sets of biosynthetic enzymes encoded in distinct gene clusters. Usually, each enzyme catalyzes one biosynthetic step, but multiple reactions are also possible. Pneumocandins A0 and B0 are produced by the fungus Glarea lozoyensis They belong to the echinocandin family, a group of nonribosomal cyclic lipopeptides that exhibit a strong antifungal activity. Chemical derivatives are important drugs for the treatment of systemic fungal infections. We have recently shown that in the biosynthesis of pneumocandins A0 and B0, three hydroxyproline building blocks are provided by one proline hydroxylase. Here we demonstrate that the proline hydroxylase from echinocandin B biosynthesis in Aspergillus pachycristatus produces the same hydroxyprolines, with an increased proportion of trans-3-hydroxyproline. However, echinocandin B biosynthesis does not require trans-3-hydroxyproline; its formation remains cryptic. While one can only speculate on the evolutionary background of this unexpected finding, proline hydroxylation in G. lozoyensis and A. pachycristatus provides an unusual insight into peptide antibiotic biosynthesis-namely, the complex interplay between the selectivity of a hydroxylase and the substrate specificity of a nonribosomal peptide synthetase.


Assuntos
Ascomicetos/metabolismo , Aspergillus/metabolismo , Equinocandinas/biossíntese , Proteínas Fúngicas/biossíntese , Hidroxiprolina/metabolismo , Antifúngicos/metabolismo
7.
Z Naturforsch C J Biosci ; 72(1-2): 1-20, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27705900

RESUMO

Echinocandins are an important group of cyclic non-ribosomal peptides with strong antifungal activity produced by filamentous fungi from Aspergillaceae and Leotiomycetes. Their structure is characterized by numerous hydroxylated non-proteinogenic amino acids. Biosynthetic clusters discovered in the last years contain up to six oxygenases, all of which are involved in amino acid modifications. Especially, variations in the oxidation pattern induced by these enzymes account for a remarkable structural diversity among the echinocandins. This review provides an overview of the current knowledge of echinocandin biosynthesis with a special focus on diversity-inducing oxidation steps. The emergence of metabolic diversity is further discussed on the basis of a comprehensive overview of the structurally characterized echinocandins, their producer strains and biosynthetic clusters. For the pneumocandins, echinocandins produced by Glarea lozoyensis, the formation of metabolic diversity in a single organism is analyzed. It is compared to two common models for the evolution of secondary metabolism: the 'target-based' approach and the 'diversity-based' model. Whereas the early phase of pneumocandin biosynthesis supports the target-based model, the diversity-inducing late steps and most oxidation reactions best fit the diversity-based approach. Moreover, two types of diversity-inducing steps can be distinguished. Although incomplete hydroxylation is a common phenomenon in echinocandin production and secondary metabolite biosynthesis in general, the incorporation of diverse hydroxyprolines at position 6 is apparently a unique feature of pneumocandin biosynthesis, which stands in stark contrast to the strict selectivity found in echinocandin biosynthesis by Aspergillaceae. The example of echinocandin biosynthesis shows that the existing models for the evolution of secondary metabolism can be well applied to parts of the pathway; however, thus far, there is no comprehensive theory that could explain the entire biosynthesis.


Assuntos
Antifúngicos/metabolismo , Equinocandinas/biossíntese , Fungos/metabolismo , Aminoácidos/metabolismo , Ascomicetos/metabolismo , Evolução Biológica , Proteínas Fúngicas/metabolismo , Hidroxilação , Microbiologia Industrial/métodos , Redes e Vias Metabólicas , Modelos Biológicos , Estrutura Molecular , Oxirredução , Oxigenases/metabolismo , Metabolismo Secundário
8.
BMC Genomics ; 17: 570, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27502607

RESUMO

BACKGROUND: Echinocandins are nonribosomal lipopeptides produced by ascommycete fungi. Due to their strong inhibitory effect on fungal cell wall biosynthesis and lack of human toxicity, they have been developed to an important class of antifungal drugs. Since 2012, the biosynthetic gene clusters of most of the main echinocandin variants have been characterized. Especially the comparison of the clusters allows a deeper insight for the biosynthesis of these complex structures. RESULTS: In the genome of the echinocandin B producer Aspergillus nidulans NRRL 8112 we have identified a gene cluster (Ani) that encodes echinocandin biosynthesis. Sequence analyses showed that Ani is clearly delimited from the genomic context and forms a monophyletic lineage with the other echinocandin gene clusters. Importantly, we found that the disjunct genomic location of the echinocandin B gene cluster in A. pachycristatus NRRL 11440 on two separate subclusters, Ecd and Hty, at two loci was likely an artifact of genome misassembly in the absence of a reference sequence. We show that both sequences can be aligned resulting a single cluster with a gene arrangement collinear compared to other clusters of Aspergillus section Nidulantes. The reassembled gene cluster (Ecd/Hty) is identical to a putative gene cluster (AE) that was previously deposited at the NCBI as a sequence from A. delacroxii NRRL 3860. PCR amplification of a part of the gene cluster resulted a sequence that was very similar (97 % identity), but not identical to that of AE. CONCLUSIONS: The Echinocandin B biosynthetic cluster from A. nidulans NRRL 8112 (Ani) is particularly similar to that of A. pachycristatus NRRL 11440 (Ecd/Hty). Ecd/Hty was originally reported as two disjunct sub-clusters Ecd and Hty, but is in fact a continuous sequence with the same gene order as in Ani. According to sequences of PCR products amplified from genomic DNA, the echinocandin B producer A. delacroxii NRRL 3860 is closely related to A. pachycristatus NRRL 11440. A PCR-product from the gene cluster was very similar, but clearly distinct from the sequence published for A. delacroxii NRRL 3860 at the NCBI (No. AB720074). As the NCBI entry is virtually identical with the re-assembled Ecd/Hty cluster, it is likely that it originates from A. pachycristatus NRRL 11440 rather than A. delacroxii NRRL 3860.


Assuntos
Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Equinocandinas/biossíntese , Equinocandinas/genética , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/genética , Família Multigênica , Sequência de Bases , Equinocandinas/química , Equinocandinas/metabolismo , Proteínas Fúngicas/química , Homologia de Sequência
9.
Appl Biochem Biotechnol ; 179(7): 1213-26, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27039401

RESUMO

Echinocandin B belongs to lipopeptide antifungal antibiotic bearing five types of direct precursor amino acids including proline, ornithine, tyrosine, threonine, and leucine. The objective of this study is to screen over-producing mutant in order to improve echinocandin B production; a stable mutant Aspergillus nidulans ZJB12073, which can use fructose as optimal carbon source instead of expensive mannitol, was selected from thousand isolates after several cycles of UV and microwave irradiation in turn. The results showed that mutant strain ZJB12073 exhibited 1.9-fold improvement in echinocandin B production to 1656.3 ± 40.3 mg/L when compared with the parent strain. Furthermore, the effects of precursor amino acids and some chemicals on echinocandin B biosynthesis in A. nidulans were investigated, respectively. Tyrosine, leucine, and biotin were selected as key factors to optimize the medium employing uniform design method. The results showed that the optimized fermentation medium provided another 63.1 % increase to 2701.6 ± 31.7 mg/L in final echinocandin B concentration compared to that of unoptimized medium.


Assuntos
Antifúngicos/química , Aspergillus nidulans/metabolismo , Equinocandinas/biossíntese , Proteínas Fúngicas/biossíntese , Antifúngicos/efeitos da radiação , Aspergillus nidulans/efeitos da radiação , Biotina/farmacologia , Equinocandinas/química , Fermentação , Proteínas Fúngicas/química , Micro-Ondas , Raios Ultravioleta
10.
Sheng Wu Gong Cheng Xue Bao ; 31(7): 1082-8, 2015 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-26647583

RESUMO

Anidulafungin is an effective antifungal medicine, which can inhibit activities of candida in vitro and in vivo. Echinocandin B (ECB) is the key precursor of Anidulafungin, thus the price and market prospect of Anidulafungin is directly due to the fermentation titer of ECB. In this study, Aspergillus nidulans was used for ECB fermentation, and the influence of adding microparticles on ECB fermentation was studied, such as talcum powder, Al2O3, and glass beads. The particle size and concentration were the key factors for mycelium morphology and ECB production, and ECB production could reach 1 262.9 mg/L and 1 344.1 mg/L by adding talcum powder of 20 g/L (d50 = 14.2 µm) and 7 glass beads (6 mm), an increase by 33.2% and 41.7%, respectively. The results indicated that the mycelium morphology of filamentous microorganisms and the product yield of fermentation could be improved by adding microparticles remarkably, and it provide an important method for the fermentative optimization of filamentous microorganisms.


Assuntos
Aspergillus nidulans/metabolismo , Equinocandinas/biossíntese , Proteínas Fúngicas/biossíntese , Microbiologia Industrial/métodos , Anidulafungina , Antifúngicos/metabolismo , Fermentação
11.
Bioprocess Biosyst Eng ; 38(10): 1845-54, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26091897

RESUMO

A combination of microbial strain improvement and statistical optimization is investigated to maximize echinocandin B (ECB) production from Aspergillus nidulans ZJB-0817. A classical sequential mutagenesis was studied first by using physical (ultraviolet irradiation at 254 nm) and chemical mutagens (lithium chloride and sodium nitrite). Mutant strain ULN-59 exhibited 2.1-fold increase in ECB production to 1583.1 ± 40.9 mg/L when compared with the parent strain (750.8 ± 32.0 mg/L). This is the first report where mutagenesis is applied in Aspergillus to improve ECB production. Further, fractional factorial design and central composite design were adopted to optimize the culture medium for increasing ECB production by the mutant ULN-59. Results indicated that four culture media including peptone, K2HPO4, mannitol and L-ornithine had significant effects on ECB production. The optimized medium provided another 1.4-fold increase in final ECB concentration to 2285.6 ± 35.6 mg/L compared to the original medium. The results of this study indicated the combined application of a classical mutation and medium optimization can improve effectively ECB production from A. nidulans and could be a promising tool to improve other secondary metabolites production by fungal strains.


Assuntos
Aspergillus nidulans/fisiologia , Técnicas de Cultura Celular por Lotes/métodos , Meios de Cultura/metabolismo , Equinocandinas/biossíntese , Proteínas Fúngicas/biossíntese , Melhoramento Genético/métodos , Mutagênicos/farmacologia , Aspergillus nidulans/efeitos dos fármacos , Aspergillus nidulans/efeitos da radiação , Equinocandinas/genética , Proteínas Fúngicas/genética , Mutação/efeitos dos fármacos , Mutação/genética , Mutação/efeitos da radiação , Especificidade da Espécie , Raios Ultravioleta
12.
Chembiochem ; 15(16): 2365-9, 2014 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-25270390

RESUMO

Echinocandins are cyclic nonribosomal hexapeptides based mostly on nonproteinogenic amino acids and displaying strong antifungal activity. Despite previous studies on their biosynthesis by fungi, the origin of three amino acids, trans-4- and trans-3-hydroxyproline, as well as trans-3-hydroxy-4-methylproline, is still unknown. Here we describe the identification, overexpression, and characterization of GloF, the first eukaryotic α-ketoglutarate/Fe(II) -dependent proline hydroxylase from the pneumocandin biosynthesis cluster of the fungus Glarea lozoyensis ATCC 74030. In in vitro transformations with L-proline, GloF generates trans-4- and trans-3-hydroxyproline simultaneously in a ratio of 8:1; the latter reaction was previously unknown for proline hydroxylase catalysis. trans-4-Methyl-L-proline is converted into the corresponding trans-3-hydroxyproline. All three hydroxyprolines required for the biosynthesis of the echinocandins pneumocandins A0 and B0 in G. lozoyensis are thus provided by GloF. Sequence analyses revealed that GloF is not related to bacterial proline hydroxylases, and none of the putative proteins with high sequence similarity in the databases has been characterized so far.


Assuntos
Equinocandinas/biossíntese , Proteínas Fúngicas/metabolismo , Prolil Hidroxilases/metabolismo , Sequência de Bases , Equinocandinas/química , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Isomerismo , Cinética , Dados de Sequência Molecular , Família Multigênica , Prolina/química , Prolina/metabolismo , Prolil Hidroxilases/química , Prolil Hidroxilases/genética , Saccharomycetales/enzimologia
13.
BMC Genomics ; 14: 339, 2013 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-23688303

RESUMO

BACKGROUND: The antifungal therapy caspofungin is a semi-synthetic derivative of pneumocandin B0, a lipohexapeptide produced by the fungus Glarea lozoyensis, and was the first member of the echinocandin class approved for human therapy. The nonribosomal peptide synthetase (NRPS)-polyketide synthases (PKS) gene cluster responsible for pneumocandin biosynthesis from G. lozoyensis has not been elucidated to date. In this study, we report the elucidation of the pneumocandin biosynthetic gene cluster by whole genome sequencing of the G. lozoyensis wild-type strain ATCC 20868. RESULTS: The pneumocandin biosynthetic gene cluster contains a NRPS (GLNRPS4) and a PKS (GLPKS4) arranged in tandem, two cytochrome P450 monooxygenases, seven other modifying enzymes, and genes for L-homotyrosine biosynthesis, a component of the peptide core. Thus, the pneumocandin biosynthetic gene cluster is significantly more autonomous and organized than that of the recently characterized echinocandin B gene cluster. Disruption mutants of GLNRPS4 and GLPKS4 no longer produced the pneumocandins (A0 and B0), and the Δglnrps4 and Δglpks4 mutants lost antifungal activity against the human pathogenic fungus Candida albicans. In addition to pneumocandins, the G. lozoyensis genome encodes a rich repertoire of natural product-encoding genes including 24 PKSs, six NRPSs, five PKS-NRPS hybrids, two dimethylallyl tryptophan synthases, and 14 terpene synthases. CONCLUSIONS: Characterization of the gene cluster provides a blueprint for engineering new pneumocandin derivatives with improved pharmacological properties. Whole genome estimation of the secondary metabolite-encoding genes from G. lozoyensis provides yet another example of the huge potential for drug discovery from natural products from the fungal kingdom.


Assuntos
Ascomicetos/genética , Ascomicetos/metabolismo , Equinocandinas/biossíntese , Genômica , Família Multigênica/genética , Ascomicetos/enzimologia , Proteínas Fúngicas/biossíntese , Humanos , Dados de Sequência Molecular , Peptídeo Sintases/genética , Peptídeo Sintases/metabolismo , Policetídeo Sintases/genética , Policetídeo Sintases/metabolismo , Análise de Sequência
14.
Appl Microbiol Biotechnol ; 95(1): 113-22, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22555909

RESUMO

Aspergillus nidulans var. roseus ATCC 58397 is an echinocandin B (ECB) producer ascomycete with great industrial importance. As demonstrated by ECB/caspofungin sensitivity assays, A. nidulans var. roseus does not possess any inherent resistance to echinocandins, and its tolerance to these lipopeptide antimycotics are even lower than those of the non-producer A. nidulans FGSC A4 strain. Under ECB producing conditions or ECB exposures, A. nidulans var. roseus induced its ECB tolerance via up-regulating elements of the chitin biosynthetic machinery and, hence, through changing dynamically the composition of its own cell wall. Importantly, although the specific ß-1,3-glucan synthase activity was elevated, these changes reduced the ß-glucan content of hyphae considerably, but the expression of fksA, encoding the catalytic subunit of ß-1,3-glucan synthase, the putative target of echinocandins in the aspergilli, was not affected. These data suggest that compensatory chitin biosynthesis is the centerpiece of the induced ECB tolerance of A. nidulans var. roseus. It is important to note that the induced tolerance to ECB (although resulted in paradoxical growth at higher ECB concentrations) was accompanied with reduced growth rate and, under certain conditions, even sensitized the fungus to other stress-generating agents like SDS. We hypothesize that although ECB-resistant mutants may arise in vivo in A. nidulans var. roseus cultures, their widespread propagation is severely restricted by the disadvantageous physiological effects of such mutations.


Assuntos
Antifúngicos/farmacologia , Aspergillus nidulans/efeitos dos fármacos , Farmacorresistência Fúngica , Equinocandinas/biossíntese , Equinocandinas/farmacologia , Proteínas Fúngicas/biossíntese , Proteínas Fúngicas/farmacologia , Lipopeptídeos/farmacologia , Antifúngicos/metabolismo , Aspergillus nidulans/crescimento & desenvolvimento , Aspergillus nidulans/metabolismo , Biotecnologia , Quitina/biossíntese , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Lipopeptídeos/metabolismo , Testes de Sensibilidade Microbiana , beta-Glucanas/metabolismo
15.
Folia Microbiol (Praha) ; 56(5): 381-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21858538

RESUMO

Polyphasic characterization of the echinocandin B producer Aspergillus nidulans var. roseus ATCC 58397 strain was carried out to elucidate its taxonomical status. According to its carbon source utilization and secondary metabolite spectrum as well as the partial ß-tubulin, calmodulin, and γ-actin gene sequences, A. nidulans var. roseus belongs to the Emericella rugulosa species. Auxotroph mutants of A. nidulans var. roseus ATCC 58397 and E. rugulosa CBS 171.71 and CBS 133.60 formed stable heterokaryons on minimal medium with several A. nidulans strains, and in the case of A. nidulans var. roseus, even cleistothecia were developed.


Assuntos
Actinas/genética , Aspergillus nidulans/genética , Calmodulina/genética , Equinocandinas/biossíntese , Emericella/genética , Proteínas Fúngicas/biossíntese , Tubulina (Proteína)/genética , Actinas/química , Actinas/metabolismo , Aspergillus nidulans/classificação , Aspergillus nidulans/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Cruzamentos Genéticos , Emericella/classificação , Emericella/metabolismo , Fermentação , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Técnicas de Tipagem Micológica , Filogenia , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Esterigmatocistina/biossíntese , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
16.
Yakugaku Zasshi ; 130(11): 1471-8, 2010 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-21048405

RESUMO

Natural fermentation products have long been studied as attractive targets for drug discovery due to their amazing diverse, complex chemical structures and biological activities. As such, a number of revolutionary drugs developed from natural fermentation products have contributed to global human health. To commercialize a drug derived from natural fermentation products, an effective chemical entity must be identified and thoroughly researched, and an effective manufacturing process to prepare a commercial supply must be developed. To construct such a manufacturing process for tacrolimus and micafungin, the following studies were conducted: first, we focused on controlling the production of the tacrolimus-related compound FR900525, a fermentation by-product of tacrolimus which was critical for quality assurance of the drug substance. FR900525 production was reduced by using a mutant strain which produced more pipecolic acid, the biosynthesis material of tacrolimus, than the original strain. Then, to optimize the fermentation process of FR901379, an intermediate of micafungin, a fed-batch culture was adopted to increase FR901379 productivity. Additionally, FULLZONE(TM) impeller was installed into the scaled-up fermenter, reducing the agitation-induced damage to the mycelium. As a result, the mycelial form changed from filamentous to pellet-shaped, and the air uptake rate during fermentation was drastically improved. Finally, we conducted screening for FR901379 acylase-producing microorganisms, as FR901379 acylase is necessary to manufacture micafungin. We were able to easily discover FR901379 acylase-producing microorganisms in soil samples using our novel, convenient screening method, which involves comparing the difference in antibiotic activity between FR901379 and its deacylated product.


Assuntos
Antifúngicos , Produtos Biológicos , Descoberta de Drogas/métodos , Equinocandinas , Fermentação , Imunossupressores , Lipopeptídeos , Streptomyces/metabolismo , Tacrolimo , Amidoidrolases , Equinocandinas/biossíntese , Lipopeptídeos/biossíntese , Micafungina , Micélio/metabolismo , Peptídeos Cíclicos/biossíntese , Microbiologia do Solo , Tacrolimo/metabolismo
17.
J Biosci Bioeng ; 109(2): 138-44, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20129097

RESUMO

Industrial-scale production of FR901379 (WF11899A), which is a novel echinocandin type of lipopeptide antibiotic produced by mutant strain M-7 from Coleophoma empetri F-11899 (FERM BP-2635), was demonstrated. In order to achieve high-level production in fermentor culture, the medium previously developed was modified, in which three types of organic nitrogen were replaced by ammonium sulfate and corn steep liquor. To eliminate increase in viscosity, carbon source was intermittently fed. The viscosity was reduced from 20,000 cP to less than 10,000 cP. The FULLZONE impeller was introduced in the fermentor culture for sufficient mixing. Mixing time was quite improved and high reproducibility was achieved. Surprisingly, the viscosity of the broth was reduced to 1000 cP in a 4 m(3) scale fermentor. When k(L)a was selected as an index for scale-up and industrial scale production using a 15 m(3) fermentor with the FULLZONE impeller was conducted, FR901379 production was successfully obtained at more than 50 U/mL, almost the same level as with the 0.03 m(3) and 4 m(3) fermentors. In addition, superior reproducibility was obtained, and 500-fold scale-up was successfully achieved.


Assuntos
Antibacterianos/biossíntese , Biotecnologia/métodos , Fermentação , Lipopeptídeos/biossíntese , Peptídeos Cíclicos/biossíntese , Antibacterianos/química , Ascomicetos/crescimento & desenvolvimento , Ascomicetos/metabolismo , Reatores Biológicos/microbiologia , Equinocandinas/biossíntese , Equinocandinas/química , Lipopeptídeos/química , Peptídeos Cíclicos/química , Viscosidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...