Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Microb Cell Fact ; 21(1): 22, 2022 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-35164756

RESUMO

BACKGROUND: Lovastatin is one of the first statins to be extensively used for its cholesterol-lowering ability. It is commercially produced by fermentation. Species belonging to the genus Aspergillus are well-studied fungi that have been widely used for lovastatin production. In the present study, we produced lovastatin from sago processing wastewater (SWW) under submerged fermentation using oleaginous fungal strains, A. terreus KPR12 and A. caespitosus ASEF14. RESULTS: The intra- and extracellular concentrations of lovastatin produced by A. terreus KPR12 and A. caespitosus ASEF14 were lactonized. Because A. caespitosus ASEF14 produced a negligible amount of lovastatin, further kinetics of lovastatin production in SWW was studied using the KPR12 strain for 9 days. Lovastatin concentrations in the intra- and extracellular fractions of the A. terreus KPR12 cultured in a synthetic medium (SM) were 117.93 and 883.28 mg L-1, respectively. However, these concentrations in SWW were 142.23 and 429.98 mg L-1, respectively. The yeast growth inhibition bioassay confirmed the antifungal property of fungal extracts. A. terreus KPR12 showed a higher inhibition zone of 14 mm than the ASEF14 strain. The two-way analysis of variance (ANOVA; p < 0.01) showed significant differences in the localization pattern, fungal strains, growth medium, and their respective interactions. The lovastatin yield coefficient values were 0.153 g g-1 on biomass (YLOV/X) and 0.043 g g-1 on the substrate, starch (YLOV/S). The pollutant level of treated SWW exhibited a reduction in total solids (TS, 59%), total dissolved solids (TDS, 68%), biological oxygen demand (BOD, 79.5%), chemical oxygen demand (COD, 57.1%), phosphate (88%), cyanide (65.4%), and void of nutrients such as nitrate (100%), and ammonia (100%). CONCLUSION: The starch-rich wastewater serves as a suitable medium for A. terreus KPR12 for the production of lovastatin. It simultaneously decontaminates the sago processing wastewater, enabling its reuse for irrigation/recreation.


Assuntos
Aspergillus/metabolismo , Lovastatina/biossíntese , Manihot , Águas Residuárias , Biomassa , Meios de Cultura , Fermentação , Cinética , Lovastatina/análise , Lovastatina/química , Espectroscopia de Infravermelho com Transformada de Fourier , Amido/metabolismo
2.
PLoS Comput Biol ; 17(9): e1009372, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34570757

RESUMO

Secondary metabolites (SMs) are a vast group of compounds with different structures and properties that have been utilized as drugs, food additives, dyes, and as monomers for novel plastics. In many cases, the biosynthesis of SMs is catalysed by enzymes whose corresponding genes are co-localized in the genome in biosynthetic gene clusters (BGCs). Notably, BGCs may contain so-called gap genes, that are not involved in the biosynthesis of the SM. Current genome mining tools can identify BGCs, but they have problems with distinguishing essential genes from gap genes. This can and must be done by expensive, laborious, and time-consuming comparative genomic approaches or transcriptome analyses. In this study, we developed a method that allows semi-automated identification of essential genes in a BGC based on co-evolution analysis. To this end, the protein sequences of a BGC are blasted against a suitable proteome database. For each protein, a phylogenetic tree is created. The trees are compared by treeKO to detect co-evolution. The results of this comparison are visualized in different output formats, which are compared visually. Our results suggest that co-evolution is commonly occurring within BGCs, albeit not all, and that especially those genes that encode for enzymes of the biosynthetic pathway are co-evolutionary linked and can be identified with FunOrder. In light of the growing number of genomic data available, this will contribute to the studies of BGCs in native hosts and facilitate heterologous expression in other organisms with the aim of the discovery of novel SMs.


Assuntos
Vias Biossintéticas/genética , Evolução Molecular , Genes Essenciais , Família Multigênica , Software , Aspergillus/genética , Aspergillus/metabolismo , Biologia Computacional , Bases de Dados de Proteínas , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Fungos/genética , Fungos/metabolismo , Genes Sintéticos , Genoma Fúngico , Genômica , Lovastatina/biossíntese , Lovastatina/genética , Filogenia , Proteoma/genética
3.
J Sci Food Agric ; 101(13): 5652-5659, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33740266

RESUMO

BACKGROUND: Monacolin K, an important secondary metabolite of Monascus, possesses a cholesterol-lowering effect and is widely used in the manufacture of antihypertensive drugs. In the present study, we constructed an extractive fermentation system by adding non-ionic surfactant and acquired a high monacolin K yield. The mechanism was determined by examining both cell morphology and the transcription levels of the related mokA-I genes in the monacolin K biosynthetic gene cluster. RESULTS: The monacolin K yield was effectively increased to 539.59 mg L-1 during extraction, which was an increase of 386.16% compared to that in the control group fermentation. The non-ionic surfactant showed good biocompatibility with Monascus. Electron scanning microscopy revealed alterations in the morphology of Monascus. The loosened mycelial structure and increased number of cell surface wrinkles were found to be related to the increased cell-membrane permeability and extracellular accumulation of monacolin K. Gene expression levels were measured via a quantitative reverse transciptase-polymerase chain reaction. By contrast, in the control group, mokA, mokB, mokC, mokD and mokF showed higher-level and longer-term expression in the extractive fermentation group, whereas mokE and mokG did not present a similar trend. The expression levels of mokH and mokI, encoding a transcription factor and efflux pump, respectively, were also higher than the control levels. CONCLUSION: The addition of a non-ionic surfactant to Monascus fermentation effectively increases the yield of monacolin K by transforming the fungus morphology and promoting the expression of monacolin K biosynthesis genes. © 2021 Society of Chemical Industry.


Assuntos
Proteínas Fúngicas/genética , Lovastatina/biossíntese , Monascus/crescimento & desenvolvimento , Monascus/metabolismo , Vias Biossintéticas , Fermentação , Proteínas Fúngicas/metabolismo , Monascus/genética , Família Multigênica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
J Photochem Photobiol B ; 217: 112164, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33676287

RESUMO

Light or low frequency magnetic field (LF-MF) as one of the cultivation environments affects secondary metabolites (SMs) production of M. purpureus. Phytochrome (Phy) is a hybrid histidine kinase possessing dual properties of photoreceptor and kinase to sense red and far-red light. The interaction effects of LF-MF and light on SMs of M. purpureus was investigated by knocking out the Phy-like gene in M. purpureus (MpPhy) by homologous recombination. A MpPhy-deletion (ΔMpPhy) strain produced less Monascus pigments (MPs) and monacolin K (mon K) than the wild-type (WT) strain and reduced citrinin production by 78.3% on 10th day but didn't affect the biomass. These results indicated that the MpPhy gene is involved in SMs biosynthesis of M. purpureus. MPs production in WT was decreased significantly when the inoculum was exposed to white/blue/green/red light (500 Lux). But it in ΔMpPhy was no significant difference when exposed to white/red light. The colony size of ΔMpPhy was smaller on potato dextrose agar media containing 0.01% SDS. These results indicated that the deletion of MpPhy gene affected the aerial hyphae and increased sensitivity to cell membrane stress but decreased sensitivity to red light. The inoculum of both WT and ΔMpPhy was exposure to the LF-MF (50 Hz). The accumulation of WT secondary metabolites was not changed, while SMs production of ΔMpPhy was significantly enhanced under exposed to 2.0 mT LF-MF. This indicated that the decrease of SMs caused by the deletion of MpPhy gene was restored by LF-MF. It revealed that there is a crosstalk between magnetoreception and photosensitivity.


Assuntos
Luz , Monascus/metabolismo , Fitocromo/genética , Metabolismo Secundário/efeitos da radiação , Biomassa , Citrinina/biossíntese , Meios de Cultura/química , Lovastatina/biossíntese , Monascus/citologia , Monascus/crescimento & desenvolvimento , Mutagênese , Fitocromo/metabolismo , Pigmentos Biológicos/metabolismo
5.
Nat Commun ; 12(1): 867, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33558520

RESUMO

Statins are effective cholesterol-lowering drugs. Lovastatin, one of the precursors of statins, is formed from dihydromonacolin L (DML), which is synthesized by lovastatin nonaketide synthase (LovB), with the assistance of a separate trans-acting enoyl reductase (LovC). A full DML synthesis comprises 8 polyketide synthetic cycles with about 35 steps. The assembling of the LovB-LovC complex, and the structural basis for the iterative and yet permutative functions of the megasynthase have remained a mystery. Here, we present the cryo-EM structures of the LovB-LovC complex at 3.60 Å and the core LovB at 2.91 Å resolution. The domain organization of LovB is an X-shaped face-to-face dimer containing eight connected domains. The binding of LovC laterally to the malonyl-acetyl transferase domain allows the completion of a L-shaped catalytic chamber consisting of six active domains. This architecture and the structural details of the megasynthase provide the basis for the processing of the intermediates by the individual catalytic domains. The detailed architectural model provides structural insights that may enable the re-engineering of the megasynthase for the generation of new statins.


Assuntos
Lovastatina/biossíntese , Lovastatina/química , Biocatálise , Modelos Moleculares , Naftalenos/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Policetídeo Sintases/química , Policetídeo Sintases/metabolismo , Policetídeo Sintases/ultraestrutura , Domínios Proteicos , Especificidade por Substrato
6.
Prep Biochem Biotechnol ; 51(2): 164-172, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32795118

RESUMO

Lovastatin is a drug in the statin class which acts as a natural inhibitor of 3-hydroxy-3-methylglutaryl, a coenzyme reductase reported as being a potential therapeutic agent for several diseases: Alzheimer's, multiple sclerosis, osteoporosis and due to its anti-cancer properties. Aspergillus terreus is known for producing a cholesterol reducing drug. This study sets out to evaluate the production of lovastatin by Brazilian wild strains of A. terreus isolated from a biological sample and natural sources. Carbon and nitrogen sources and the best physicochemical conditions using factorial design were also evaluated. The 37 fungal were grown to produce lovastatin by submerged fermentation. A. terreus URM5579 strain was the best lovastatin producer with a level of 13.96 mg/L. Soluble starch and soybean flour were found to be the most suitable substrates for producing lovastatin (41.23 mg/L) and biomass (6.1 mg/mL). The most favorable production conditions were found in run 16 with 60 g/L soluble starch, 15 g/L soybean flour, pH 7.5, 200 rpm and maintaining the solution at 32 °C for 7 days, which led to producing 100.86 mg/L of lovastatin and 17.68 mg/mL of biomass. Using natural strains and economically viable substrates helps to optimize the production of lovastatin and promote its use.


Assuntos
Aspergillus/metabolismo , Biotecnologia/métodos , Lovastatina/biossíntese , Biomassa , Brasil , Carbono , Colesterol/química , Cromatografia Líquida de Alta Pressão , Fermentação , Concentração de Íons de Hidrogênio , Nitrogênio , Espectrofotometria Ultravioleta , Amido/química , Temperatura , Fatores de Tempo
7.
Molecules ; 25(19)2020 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-32987627

RESUMO

The aim of this project was to improve the Aspergillus terreus strain and pretreatment of sugarcane bagasse as carrier substrate for bulk production of lovastatin, a cholesterol-lowering drug, in solid state fermentation. Sugarcane bagasse was treated with alkali (1-3% NaOH) for the conversion of complex polysaccharides into simple sugars for better utilization of carrier substrate by microorganism for maximum lovastatin production. Ethidium bromide (time of exposure 30-180 min) was used to induce mutation in Aspergillus terreus and the best mutant was selected on the basis of inhibition zone appeared on petri plates. Fermented lovastatin was quantified by high-performance liquid chromatography. The fermented lovastatin, produced by parent and mutant Aspergillus terreus strain, was checked on body weight, blood glucose and serum cholesterol, ALT, AST, HDL-C, LDL-C, TG and TC levels of rats for their cholesterol lowering capacity. Our results indicate that selected strain along with 2% NaOH treated sugar cane bagasse was best suitable for bulk production of lovastatin by fermentation and fermented lovastatin effectively lower the cholesterol level of rats.


Assuntos
Anticolesterolemiantes , Aspergillus , Colesterol/sangue , Lovastatina , Animais , Anticolesterolemiantes/isolamento & purificação , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/farmacologia , Aspergillus/genética , Aspergillus/crescimento & desenvolvimento , Celulose/química , Avaliação Pré-Clínica de Medicamentos , Lovastatina/biossíntese , Lovastatina/isolamento & purificação , Lovastatina/farmacocinética , Lovastatina/farmacologia , Masculino , Ratos , Saccharum/química
8.
Food Funct ; 11(7): 5738-5748, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32555902

RESUMO

Lovastatin/monacolin K (MK) is used as a lipid lowering drug, due to its effective hypercholesterolemic properties, comparable to synthetic statins. Lovastatin's biosynthetic pathway and gene cluster composition have been studied in depth in Aspergillus terreus. Evidence shows that the MK biosynthetic pathway and gene cluster in Monascus sp. are similar to those of lovastatin in A. terreus. Currently, research efforts have been focusing on the metabolic regulation of MK/lovastatin synthesis, and the evidence shows that a combination of extracellular and intracellular factors is essential for proper MK/lovastatin metabolism. Here, we comprehensively review the research progress on MK/lovastatin biosynthetic pathways, its synthetic precursors and inducing substances and metabolic regulation, with a view to providing reference for future research on fungal metabolism regulation and metabolic engineering for MK/lovastatin production.


Assuntos
Regulação Fúngica da Expressão Gênica , Lovastatina/biossíntese , Lovastatina/genética , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Aspergillus/enzimologia , Aspergillus/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Monascus/enzimologia , Monascus/genética , Regiões Promotoras Genéticas
9.
Fungal Genet Biol ; 138: 103367, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32198121

RESUMO

Filamentous fungi are well known for producing secondary metabolites applied in various industrial segments. Among these, lovastatin and itaconic acid, produced by Aspergillus terreus, have applications in the pharmaceutical and chemical industries. Lovastatin is primarily used for the control of hypercholesterolemia, while itaconic acid is a building block for the production of synthetic fibers, coating adhesives, among others. In this study, for the first time, 35 strains of Aspergillus sp. from four Brazilian culture collections were evaluated for lovastatin and itaconic acid production and compared to a reference strain, ATCC 20542. From an initial screening, the strains ATCC 20542, URM 224, URM1876, URM 5061, URM 5254, URM 5256, URM 5650, and URM 5961 were selected for genomic comparison. Among tested strains, the locus corresponding to the lovastatin genomic cluster was assembled, showing that all genes essential for lovastatin biosynthesis were present in producing URM 5961 and URM 5650 strains, with 100% and 98.5% similarity to ATCC 20542, respectively. However, in the no producing URM 1876, URM 224, URM 5254, URM 5061, and URM 5256 strains, this cluster was either fragmented or missing. Among the 35 strains evaluated for itaconic acid production in this study, only three strains had titers above 0.5 g/L, 16 strains had production below 0.5 g/L, and the remaining 18 strains had no production, with the highest production of itaconic acid observed in the URM 5254 strain with 2.2 g/L. The essential genes for itaconic acid production, mttA, cadA msfA were also mapped, where all three genes linked to itaconic acid production were found in a single contig in the assembly of each strain. In contrast to lovastatin loci, there is no correlation between the level of itaconic acid production and genetic polymorphisms in the genes associated with its biosynthesis.


Assuntos
Aspergillus , Lovastatina , Succinatos , Aspergillus/genética , Aspergillus/metabolismo , Biodiversidade , Brasil , Genes Fúngicos , Variação Genética , Genoma Fúngico , Lovastatina/biossíntese , Lovastatina/genética , Filogenia , Succinatos/metabolismo
10.
Appl Microbiol Biotechnol ; 104(7): 3049-3060, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32043189

RESUMO

Monascus is a filamentous fungus that produces several secondary metabolites. Here, we investigated the effects of the global regulator LaeA on the synthesis of pigments and monacolin K in Monascus purpureus with spectrophotometer and HPLC methods. The LaeA gene was isolated from M. purpureus M1 to create an overexpression construct. An LaeA-overexpressing strain L3 was with 48.6% higher monacolin K production than the M1 strain. The L3 strain also produced higher Monascus pigments than the M1 strain. SEM showed that LaeA overexpression resulted in altered mycelial morphology. Compared with the M1 strain, the L3 strain expressed higher levels of monacolin K synthesis-related genes mokA, mokB, mokE, and mokH. Overall, these results suggest that LaeA plays a role in regulating the production of secondary metabolites and mycelial growth in Monascus. This study provides important insights into the mechanisms underlying the effects of the LaeA gene on the secondary metabolites of M. purpureus.


Assuntos
Proteínas Fúngicas/genética , Genes Fúngicos , Monascus/metabolismo , Metabolismo Secundário , Fatores de Transcrição/genética , Proteínas Fúngicas/metabolismo , Expressão Gênica , Regulação Fúngica da Expressão Gênica , Lovastatina/biossíntese , Monascus/genética , Monascus/crescimento & desenvolvimento , Micélio/genética , Micélio/crescimento & desenvolvimento , Micélio/metabolismo , Pigmentos Biológicos/biossíntese , Metabolismo Secundário/genética , Fatores de Transcrição/metabolismo
11.
J Microbiol Methods ; 164: 105672, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31326443

RESUMO

PURPOSE: This study aimed to assess the effect of nitrogen, salt and pre-culture conditions on the production of lovastatin in A. terreus ATCC 20542. METHODS: Different combinations of nitrogen sources, salts and pre-culture combinations were applied in the fermentation media and lovastatin yield was analysed chromatographically. RESULT: The exclusion of MnSO4 ·5H2O, CuSO4·5H2O and FeCl3·6H2O were shown to significantly improve lovastatin production (282%), while KH2PO4, MgSO4·7H2O, and NaCl and ZnSO4·7H2O were indispensable for good lovastatin production. Simple nitrogen source (ammonia) was unfavourable for morphology, growth and lovastatin production. In contrast, yeast extract (complex nitrogen source) produced the highest lovastatin yield (25.52 mg/L), while powdered soybean favoured the production of co-metabolites ((+)-geodin and sulochrin). Intermediate lactose: yeast extract (5:4) ratio produced the optimal lovastatin yield (12.33 mg/L) during pre-culture, while high (5:2) or low (5:6) lactose to yeast extract ratio produced significantly lower lovastatin yield (7.98 mg/L and 9.12 mg/L, respectively). High spore concentration, up to 107 spores/L was shown to be beneficial for lovastatin, but not for co-metabolite production, while higher spore age was shown to be beneficial for all of its metabolites. CONCLUSION: The findings from these investigations could be used for future cultivation of A. terreus in the production of desired metabolites.


Assuntos
Aspergillus/metabolismo , Meios de Cultura/química , Lovastatina/biossíntese , Técnicas Microbiológicas/métodos , Compostos de Amônio , Benzoatos , Benzofuranos , Biomassa , Técnicas de Cultura de Células/métodos , Fermentação , Lactose/metabolismo , Nitrogênio , Esporos/crescimento & desenvolvimento
12.
Curr Pharm Biotechnol ; 20(15): 1244-1259, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31333127

RESUMO

Statins are drugs used for people with abnormal lipid levels (hyperlipidemia) and are among the best-selling medications in the United States. Thus, the aspects related to the production of these drugs are of extreme importance for the pharmaceutical industry. Herein, we provide a non-exhaustive review of fungal species used to produce statin and highlighted the major factors affecting the efficacy of this process. The current biotechnological approaches and the advances of a metabolic engineer to improve statins production are also emphasized. The biotechnological production of the main statins (lovastatin, pravastatin and simvastatin) uses different species of filamentous fungi, for example Aspergillus terreus. The statins production is influenced by different types of nutrients available in the medium such as the carbon and nitrogen sources, and several researches have focused their efforts to find the optimal cultivation conditions. Enzymes belonging to Lov class, play essential roles in statin production and have been targeted to genetic manipulations in order to improve the efficiency for Lovastatin and Simvastatin production. For instance, Escherichia coli strains expressing the LovD have been successfully used for lovastatin production. Other examples include the use of iRNA targeting LovF of A. terreus. Therefore, fungi are important allies in the fight against hyperlipidemias. Although many studies have been conducted, investigations on bioprocess optimization (using both native or genetic- modified strains) still necessary.


Assuntos
Biotecnologia/métodos , Fungos/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/biossíntese , Engenharia Metabólica/métodos , Pravastatina/biossíntese , Animais , Fermentação , Fungos/genética , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Hiperlipidemias/tratamento farmacológico , Lovastatina/farmacologia , Lovastatina/toxicidade , Pravastatina/farmacologia , Pravastatina/toxicidade
13.
Appl Microbiol Biotechnol ; 103(13): 5301-5310, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31049618

RESUMO

Monascus purpureus is a traditional Chinese microbe that can be used as a medicinal herb and is edible. To improve the yield of monacolin K, we optimized the medium of M. purpureus with high-yield monacolin K strains. When high-yield strains C8, D8, E3, and I1 were grown in glutamic medium instead of the original medium, monacolin K production was increased. Among these strains, C8 exhibited the highest monacolin K production in glutamic acid medium, with levels increased 4.80-fold. RT-qPCR demonstrated that glutamic acid enhanced the expression of mokC and mokG. Observation of Monascus mycelium morphology using SEM showed that mycelia exhibited more folds, swelling, curves, and fractures. Thus, glutamic acid may promote the growth of the mycelium and appeared to increase the permeability of the cell membrane. This lays a foundation for research on the regulatory effect of glutamic acid and provides a theoretical basis for the industrialization and commercialization of Monascus.


Assuntos
Ácido Glutâmico/farmacologia , Lovastatina/biossíntese , Monascus/efeitos dos fármacos , Monascus/metabolismo , Meios de Cultura/química , Fermentação , Proteínas Fúngicas/genética , Expressão Gênica , Microbiologia Industrial , Microscopia Eletrônica de Varredura , Micélio/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real
14.
Appl Microbiol Biotechnol ; 103(14): 5593-5605, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31098686

RESUMO

The goal of the study was to compare the production of secondary metabolites by Aspergillus terreus ATCC 20542 under the conditions of submerged mono- and co-cultivation. The suggested experimental scheme encompassed a diverse set of co-culture initiation strategies differing mostly with respect to the development stage of tested fungal strains at the moment of their confrontation. Three species of filamentous fungi exhibiting distinct patterns of morphological evolution under submerged conditions, namely Penicillium rubens, Chaetomium globosum, and Mucor racemosus, were selected as the co-cultivation partners of A. terreus. The choice of the co-cultivated species and the approach of co-culture triggering noticeably influenced the levels of lovastatin (mevinolinic acid), (+)-geodin, asterric acid, and butyrolactone I in the broth. Even though the evaluated co-cultures did not lead to the increased titers of lovastatin relative to standard monocultures, the biosynthesis of the remaining three metabolites was either enhanced or inhibited depending on the experimental variant. The production of butyrolactone I turned out to be particularly affected by the presence of C. globosum. Interestingly, in the A. terreus/C. globosum co-cultures, the decrease of lovastatin concentration was recorded. According to the most probable scenario, lovastatin was in this case converted to monacolin J acid, a polyketide molecule that may be applied as a substrate for the synthesis of statin drugs. The study revealed that the spores of two distinct fungal species, namely A. terreus and C. globosum, co-agglomerate under submerged conditions to form pellets. Finally, the biosynthetic performance of co-cultures involving four fungal species was evaluated.


Assuntos
Aspergillus/metabolismo , Reatores Biológicos , Lovastatina/biossíntese , Metabolismo Secundário , Biomassa , Técnicas de Cocultura , Cinética , Técnicas Microbiológicas , Naftalenos/metabolismo , Penicillium/metabolismo , Esporos/fisiologia
15.
Bioprocess Biosyst Eng ; 42(7): 1205-1213, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30949844

RESUMO

In bubble column bioreactors, the hydrodynamic behavior like mixing time, bubble size and morphology of filamentous fungi are influenced by the construction of spargers. Sparger pore size is an important factor influencing formation of bubbles. In this study for the first time, a 5-L bubble column bioreactor with different porous spargers was used to investigate the effect of mean air bubble diameter (at 0.36, 0.18 and 0.09 cm) on fungal growth, broth viscosity, fungal pellet morphology and lovastatin production by the filamentous fungus Aspergillus terreus. All cultivations were carried out at air flow rate equal to 0.5 Lair L-1 min-1. The viscosity of the broth was influenced by both biomass concentration and size of the fungal pellets. The highest values of viscosity were observed at bubbles of 0.09 cm diameter after 192 h of cultivation. The largest fluffy pellets and the highest yield of lovastatin (443 mg/L) were obtained at air bubbles diameter of 0.18 cm. Lovastatin yield on biomass growth in this condition was, respectively, 1.7-fold and 3.5-fold higher than in the cultivations performed with air bubbles of 0.36 and 0.09 cm diameters. These laboratory scale experiment indicates that air bubble diameter has the impact on lovastatin production and A. terreus culture conditions.


Assuntos
Aspergillus/crescimento & desenvolvimento , Biomassa , Reatores Biológicos , Lovastatina/biossíntese , Ar , Cinética , Porosidade
16.
N Biotechnol ; 52: 19-24, 2019 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-30995533

RESUMO

Lovastatin is widely prescribed to reduce elevated levels of cholesterol and prevent heart-related diseases. Cultivation of Aspergillus terreus (ATCC 20542) with carbohydrates or low-value feedstocks such as glycerol produces lovastatin as a secondary metabolite and (+)-geodin as a by-product. An A. terreus mutant strain was developed (gedCΔ) with a disrupted (+)-geodin biosynthesis pathway. The gedCΔ mutant was created by inserting the antibiotic marker hygromycin B (hyg) within the gedC gene that encodes emodin anthrone polyketide synthase (PKS), a primary gene responsible for initiating (+)-geodin biosynthesis. The effects of emodin anthrone PKS gene disruption on (+)-geodin and lovastatin biosynthesis and the production of the precursors acetyl-CoA and malonyl-CoA were investigated with cultures based on glycerol alone and in combination with lactose. The gedCΔ strain showed improved lovastatin production, particularly when cultivated on the glycerol-lactose mixture, increasing lovastatin production by 80% (113 mg/L) while simultaneously inhibiting (+)-geodin biosynthesis compared to the wild-type strain. This study thus shows that suppression of the (+)-geodin pathway increases lovastatin yield and demonstrates a practical approach of manipulating carbon flux by modulating enzyme activity.


Assuntos
Aspergillus/metabolismo , Benzofuranos/metabolismo , Vias Biossintéticas , Lovastatina/biossíntese , Acetilcoenzima A/biossíntese , Glicerol/metabolismo , Lactose/metabolismo , Malonil Coenzima A/biossíntese
17.
Pak J Pharm Sci ; 32(1(Supplementary)): 363-370, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30829216

RESUMO

During intrinsic cholesterol formation 3-hydroxy-3-methylgutaryl coenzyme A reductase (HMGCR) converts HMGCoA to mevalonate, in biosynthetic cascade of cholesterol. Statins, competitive inhibitors of HMGCR, now-a-days commonly used to lower the blood-cholesterol level in the hyper-cholesterolemic patients. Lovastatin, one of the most potent natural statins, was produced from wild-type indigenous isolate Aspergillus terreus PU-PCSIR-1, through solid state fermentation (SSF). This study was carried out to investigate different parameters influencing lovastatin production such as pH, carbon source, nitrogen source and media components etc. Each parameter was investigated separately to optimize lovastatin production. Maximum yield of 2860mg/Kg of total lovastatin, comprising 1700 and 1160mg/Kg of hydroxy and lactone forms respectively, was achieved after incubating for 14 days, pH 5.5 and at 28°C. The integrity of biotechnologically-produced lovastatin was analyzed using high performance liquid chromatography (HPLC). Lovastatin was purified by preparative HPLC, and was characterized by FT-IR and LC-MS analyses. The study revealed that A. terreus PU-PCSIR-1 has been proved to be a potent strain for the production of lovastatin that has great pharmaceutical and commercial applications.


Assuntos
Aspergillus/metabolismo , Biotecnologia/métodos , Lovastatina/biossíntese , Carbono/metabolismo , Cromatografia Líquida de Alta Pressão , Fibras na Dieta/metabolismo , Fermentação , Glicerol/metabolismo , Concentração de Íons de Hidrogênio , Lovastatina/análise , Nitrogênio/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
18.
Biomed Res Int ; 2019: 5832496, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30733961

RESUMO

Aluminum oxide nanoparticles were supplemented to Aspergillus terreus ATCC 20542 precultures and the outcomes of the process were evaluated relative to the results of microparticle-enhanced and standard cultivations. The selected morphological parameters of fungal pellets (projected area, elongation, convexity, and shape factor) were monitored throughout the experiment, together with biomass, lactose, and lovastatin concentration. The qualitative and quantitative chemical analysis was performed with the use of liquid chromatography coupled with high resolution mass spectrometry. The results of the study indicated that the application of nanoparticles was indeed associated with morphological consequences, most notably the decreased pellet size. However, it turned out that the term "nanoparticle-enhanced cultivation" could not be used in the context of lovastatin production, as no marked increase of product titer was observed in nanoparticle-influenced variants relative to standard and microparticle-enhanced cultivation. In addition, the concentration of biomass in the nanoparticle-influenced runs was relatively low. Comparative analysis of total ion chromatograms revealed the presence of a molecule of unknown structure that could be detected solely in broths from standard and microparticle-containing cultures. This study represents the first evaluation of nanoparticles as the tools of morphological engineering aimed at enhanced lovastatin biosynthesis in A. terreus cultures.


Assuntos
Óxido de Alumínio/farmacologia , Aspergillus/citologia , Aspergillus/crescimento & desenvolvimento , Lovastatina/biossíntese , Nanopartículas/química , Aspergillus/efeitos dos fármacos , Biomassa , Lactose/metabolismo , Fatores de Tempo
19.
J Agric Food Chem ; 67(9): 2563-2569, 2019 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-30734557

RESUMO

Monascus purpureus is an important food and drug microbial resource through the production of a variety of secondary metabolites, including monacolin K, a well-recognized cholesterol-lowering agent. However, the high production costs and naturally low contents of monacolin K have restricted its large-scale production. Thus, in this study we sought to improve the production of monacolin K in M. purpureus through overexpression of four genes ( mokC, mokD, mokE, and mokI). Four overexpression strains were successfully constructed by protoplast electric shock conversion, which resulted in a 234.3%, 220.8%, 89.5%, and 10% increase in the yield of monacolin K, respectively. The overexpression strains showed clear changes to the mycelium surface with obvious folds and the spores with depressions, whereas the pBC5 mycelium had a fuller structure with a flatter surface. Further investigation of these strains can provide the theoretical basis and technical support for the development of functional Monascus varieties.


Assuntos
Benzopiranos/metabolismo , Lovastatina/biossíntese , Lovastatina/genética , Monascus/genética , Pigmentos Biológicos/metabolismo , Policetídeos/metabolismo , Expressão Gênica , Engenharia Genética/métodos , Monascus/metabolismo
20.
ACS Synth Biol ; 7(12): 2783-2789, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30403849

RESUMO

For many secondary metabolites, heterologous synthesis is the definitive step to determine their required biosynthetic genes. Using a multivector expression system in Saccharomyces cerevisiae, we reconstituted not only two natural statins from two fungal species, i.e., lovastatin from Aspergillus terreus and FR901512 from Xylaria grammica, but also new statin structures by mixing their genes. Combinatorial gene exchange experiments revealed the functional promiscuity of two polyketide synthases in A. terreus, lovB, and lovF; they could synthesize FR901512 with Xylaria genes. Key structure determinants of statins are essential accessory genes that are irreplaceable across species.


Assuntos
Aspergillus/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Xylariales/genética , Cromatografia Líquida de Alta Pressão , Inibidores de Hidroximetilglutaril-CoA Redutases/análise , Lovastatina/análise , Lovastatina/biossíntese , Espectrometria de Massas , Plasmídeos/genética , Plasmídeos/metabolismo , Policetídeo Sintases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...