Journal articles on the topic 'Yeast cell factory'

To see the other types of publications on this topic, follow the link: Yeast cell factory.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Yeast cell factory.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Spier, R. E. "Yeast as a Cell Factory." Enzyme and Microbial Technology 26, no. 9-10 (June 2000): 639. http://dx.doi.org/10.1016/s0141-0229(00)00223-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Nielsen, Jens. "Yeast Systems Biology: Model Organism and Cell Factory." Biotechnology Journal 14, no. 9 (May 20, 2019): 1800421. http://dx.doi.org/10.1002/biot.201800421.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

dos Santos, Sandra C., and Isabel Sá-Correia. "Yeast toxicogenomics: lessons from a eukaryotic cell model and cell factory." Current Opinion in Biotechnology 33 (June 2015): 183–91. http://dx.doi.org/10.1016/j.copbio.2015.03.001.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

van Dijk, Ralf, Klaas Nico Faber, Jan A. K. W. Kiel, Marten Veenhuis, and Ida van der Klei. "The methylotrophic yeast Hansenula polymorpha: a versatile cell factory." Enzyme and Microbial Technology 26, no. 9-10 (June 2000): 793–800. http://dx.doi.org/10.1016/s0141-0229(00)00173-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Eliasson Lantz, Anna, Songsak Wattanachaisaereekul, Michael Lynge Nielsen, and Jens Nielsen. "Towards a yeast cell factory platform for polyketide production." Journal of Biotechnology 131, no. 2 (September 2007): S199. http://dx.doi.org/10.1016/j.jbiotec.2007.07.355.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Kampranis, Sotirios C., and Antonios M. Makris. "DEVELOPING A YEAST CELL FACTORY FOR THE PRODUCTION OF TERPENOIDS." Computational and Structural Biotechnology Journal 3, no. 4 (October 2012): e201210006. http://dx.doi.org/10.5936/csbj.201210006.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Rustiaty, Banon. "OPTIMALISASI SEL Saccharomyces cerevisiae UNTUK MENINGKATKAN PRODUKTIVITAS DAN EFISIENSI INDUSTRI ETANOL [Optimization of Saccharomyces cerevisiae Cell to Increase Productivity and Efficiency of Ethanol Industry]." Jurnal Teknologi & Industri Hasil Pertanian 23, no. 2 (September 18, 2018): 97. http://dx.doi.org/10.23960/jtihp.v23i2.97-102.

Full text
Abstract:
The development of bioethanol as fuel substitution is believed to overcome the potency of the world energy crisis including Indonesia. The bioethanol development can be done by increasing the production capacity of the existing bioethanol factory plant by improving yeast culture for enhancing the performance of the fermentation process. This study was aimed at obtaining a method of optimizing the ability of Saccharomyces cerevisiae fermentation that can be applied by the alcohol industry in Indonesia for increasing factory productivity, thereby reducing the cost of producing alcohol. In this study, the adaptation of Saccharomyces cerevisiae Watei and Saccharomyces cerevisiae Hakken I were adopted in environment condition with high ethanol content up to 13%. The results showed that the yeast was able to grow in environments with high ethanol content with higher specific growth rate and larger cell size than those within the original yeast. This condition showed that adapted strains can overcome stress caused by high ethanol. These results promise the good performance yeasts with ability in growing and performing metabolic activities in high alcohol-containing environment conditions
APA, Harvard, Vancouver, ISO, and other styles
8

Park, Jongbeom, In Jung Kim, and Soo Rin Kim. "Nonconventional Yeasts Engineered Using the CRISPR-Cas System as Emerging Microbial Cell Factories." Fermentation 8, no. 11 (November 19, 2022): 656. http://dx.doi.org/10.3390/fermentation8110656.

Full text
Abstract:
Because the petroleum-based chemical synthesis of industrial products causes serious environmental and societal issues, biotechnological production using microorganisms is an alternative approach to achieve a more sustainable economy. In particular, the yeast Saccharomyces cerevisiae is widely used as a microbial cell factory to produce biofuels and valuable biomaterials. However, product profiles are often restricted due to the Crabtree-positive nature of S. cerevisiae, and ethanol production from lignocellulose is possibly enhanced by developing alternative stress-resistant microbial platforms. With desirable metabolic pathways and regulation in addition to strong resistance to diverse stress factors, nonconventional yeasts (NCY) may be considered an alternative microbial platform for industrial uses. Irrespective of their high industrial value, the lack of genetic information and useful gene editing tools makes it challenging to develop metabolic engineering-guided scaled-up applications using yeasts. The recently developed clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein (Cas) system is a powerful gene editing tool for NCYs. This review describes the current status of and recent advances in promising NCYs in terms of industrial and biotechnological applications, highlighting CRISPR-Cas9 system-based metabolic engineering strategies. This will serve as a basis for the development of novel yeast applications.
APA, Harvard, Vancouver, ISO, and other styles
9

Porro, Danilo, and Paola Branduardi. "Yeast cell factory: fishing for the best one or engineering it?" Microbial Cell Factories 8, no. 1 (2009): 51. http://dx.doi.org/10.1186/1475-2859-8-51.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Prado, Angelica Rodriguez, Kanchana Kildegaard, Mingji Li, Irina Borodina, and Jens Nielsen. "Development of a yeast cell factory for production of aromatic products." New Biotechnology 31 (July 2014): S130. http://dx.doi.org/10.1016/j.nbt.2014.05.1934.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Saner, Nazan, Jens Karschau, Toyoaki Natsume, Marek Gierliński, Renata Retkute, Michelle Hawkins, Conrad A. Nieduszynski, J. Julian Blow, Alessandro P. S. de Moura, and Tomoyuki U. Tanaka. "Stochastic association of neighboring replicons creates replication factories in budding yeast." Journal of Cell Biology 202, no. 7 (September 23, 2013): 1001–12. http://dx.doi.org/10.1083/jcb.201306143.

Full text
Abstract:
Inside the nucleus, DNA replication is organized at discrete sites called replication factories, consisting of DNA polymerases and other replication proteins. Replication factories play important roles in coordinating replication and in responding to replication stress. However, it remains unknown how replicons are organized for processing at each replication factory. Here we address this question using budding yeast. We analyze how individual replicons dynamically organized a replication factory using live-cell imaging and investigate how replication factories were structured using super-resolution microscopy. Surprisingly, we show that the grouping of replicons within factories is highly variable from cell to cell. Once associated, however, replicons stay together relatively stably to maintain replication factories. We derive a coherent genome-wide mathematical model showing how neighboring replicons became associated stochastically to form replication factories, which was validated by independent microscopy-based analyses. This study not only reveals the fundamental principles promoting replication factory organization in budding yeast, but also provides insight into general mechanisms by which chromosomes organize sub-nuclear structures.
APA, Harvard, Vancouver, ISO, and other styles
12

Chen, Yun, Laurent Daviet, Michel Schalk, Verena Siewers, and Jens Nielsen. "Establishing a platform cell factory through engineering of yeast acetyl-CoA metabolism." Metabolic Engineering 15 (January 2013): 48–54. http://dx.doi.org/10.1016/j.ymben.2012.11.002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
13

Levisson, Mark, Carla Araya-Cloutier, Wouter J. C. de Bruijn, Menno van der Heide, José Manuel Salvador López, Jean-Marc Daran, Jean-Paul Vincken, and Jules Beekwilder. "Toward Developing a Yeast Cell Factory for the Production of Prenylated Flavonoids." Journal of Agricultural and Food Chemistry 67, no. 49 (April 24, 2019): 13478–86. http://dx.doi.org/10.1021/acs.jafc.9b01367.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Nielsen, J. "Yeast as a platform cell factory for production of fuels and chemicals." Journal of Biotechnology 150 (November 2010): 78. http://dx.doi.org/10.1016/j.jbiotec.2010.08.202.

Full text
APA, Harvard, Vancouver, ISO, and other styles
15

Wang, Shaoyu. "Leveraging budding yeast Saccharomyces cerevisiae for discovering aging modulation substances for functional food." Functional Foods in Health and Disease 9, no. 5 (May 30, 2019): 297. http://dx.doi.org/10.31989/ffhd.v9i5.575.

Full text
Abstract:
Background: Discovery of bioactive substances contained in functional food and the mechanism of their aging modulation are imperative steps in developing better, potent and safer functional food for promoting health and compression of morbidity in the aging population. Budding yeast (Saccharomyces cerevisiae) is invaluable model organism for aging modulation and bioactive compounds discovery. In this paper we have conceptualised a framework for achieving such aim. This framework consists of four components: discovering targets for aging modulation, discovering and validating caloric restriction mimetics, acting as cellular systems for screening natural products or compounds for aging modulation and being a biological factory for producing bioactive compounds according to the roles the yeast systems play. It have been argued that the component of being a biological factory for producing bioactive compounds has much underexplored which also present an opportunity for new active substance discovery and validation for health promotion in functional food industry.Keywords: Aging modulation, budding yeast, functional food, bioactive substances, cell factory
APA, Harvard, Vancouver, ISO, and other styles
16

Karim, Ahasanul, Natela Gerliani, and Mohammed Aïder. "Kluyveromyces marxianus: An emerging yeast cell factory for applications in food and biotechnology." International Journal of Food Microbiology 333 (November 2020): 108818. http://dx.doi.org/10.1016/j.ijfoodmicro.2020.108818.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Höhne, Matthias, and Johannes Kabisch. "Brewing Painkillers: A Yeast Cell Factory for the Production of Opioids from Sugar." Angewandte Chemie International Edition 55, no. 4 (January 6, 2016): 1248–50. http://dx.doi.org/10.1002/anie.201510333.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

Do, Diem T. Hoang, Chrispian W. Theron, and Patrick Fickers. "Organic Wastes as Feedstocks for Non-Conventional Yeast-Based Bioprocesses." Microorganisms 7, no. 8 (July 31, 2019): 229. http://dx.doi.org/10.3390/microorganisms7080229.

Full text
Abstract:
Non-conventional yeasts are efficient cell factories for the synthesis of value-added compounds such as recombinant proteins, intracellular metabolites, and/or metabolic by-products. Most bioprocess, however, are still designed to use pure, ideal sugars, especially glucose. In the quest for the development of more sustainable processes amid concerns over the future availability of resources for the ever-growing global population, the utilization of organic wastes or industrial by-products as feedstocks to support cell growth is a crucial approach. Indeed, vast amounts of industrial and commercial waste simultaneously represent an environmental burden and an important reservoir for recyclable or reusable material. These alternative feedstocks can provide microbial cell factories with the required metabolic building blocks and energy to synthesize value-added compounds, further representing a potential means of reduction of process costs as well. This review highlights recent strategies in this regard, encompassing knowledge on catabolic pathways and metabolic engineering solutions developed to endow cells with the required metabolic capabilities, and the connection of these to the synthesis of value-added compounds. This review focuses primarily, but not exclusively, on Yarrowia lipolytica as a yeast cell factory, owing to its broad range of naturally metabolizable carbon sources, together with its popularity as a non-conventional yeast.
APA, Harvard, Vancouver, ISO, and other styles
19

Wang, Guokun, Douglas B. Kell, and Irina Borodina. "Harnessing the yeast Saccharomyces cerevisiae for the production of fungal secondary metabolites." Essays in Biochemistry 65, no. 2 (July 2021): 277–91. http://dx.doi.org/10.1042/ebc20200137.

Full text
Abstract:
Abstract Fungal secondary metabolites (FSMs) represent a remarkable array of bioactive compounds, with potential applications as pharmaceuticals, nutraceuticals, and agrochemicals. However, these molecules are typically produced only in limited amounts by their native hosts. The native organisms may also be difficult to cultivate and genetically engineer, and some can produce undesirable toxic side-products. Alternatively, recombinant production of fungal bioactives can be engineered into industrial cell factories, such as aspergilli or yeasts, which are well amenable for large-scale manufacturing in submerged fermentations. In this review, we summarize the development of baker’s yeast Saccharomyces cerevisiae to produce compounds derived from filamentous fungi and mushrooms. These compounds mainly include polyketides, terpenoids, and amino acid derivatives. We also describe how native biosynthetic pathways can be combined or expanded to produce novel derivatives and new-to-nature compounds. We describe some new approaches for cell factory engineering, such as genome-scale engineering, biosensor-based high-throughput screening, and machine learning, and how these tools have been applied for S. cerevisiae strain improvement. Finally, we prospect the challenges and solutions in further development of yeast cell factories to more efficiently produce FSMs.
APA, Harvard, Vancouver, ISO, and other styles
20

De, Sonakshi, Diethard Mattanovich, Pau Ferrer, and Brigitte Gasser. "Established tools and emerging trends for the production of recombinant proteins and metabolites in Pichia pastoris." Essays in Biochemistry 65, no. 2 (July 2021): 293–307. http://dx.doi.org/10.1042/ebc20200138.

Full text
Abstract:
Abstract Besides bakers’ yeast, the methylotrophic yeast Komagataella phaffii (also known as Pichia pastoris) has been developed into the most popular yeast cell factory for the production of heterologous proteins. Strong promoters, stable genetic constructs and a growing collection of freely available strains, tools and protocols have boosted this development equally as thorough genetic and cell biological characterization. This review provides an overview of state-of-the-art tools and techniques for working with P. pastoris, as well as guidelines for the production of recombinant proteins with a focus on small-scale production for biochemical studies and protein characterization. The growing applications of P. pastoris for in vivo biotransformation and metabolic pathway engineering for the production of bulk and specialty chemicals are highlighted as well.
APA, Harvard, Vancouver, ISO, and other styles
21

Buathong, Phawadee, Nassapat Boonvitthya, Gilles Truan, and Warawut Chulalaksananukul. "Whole-Cell Biotransformation of 1,12-Dodecanedioic Acid from Coconut Milk Factory Wastewater by Recombinant CYP52A17SS Expressing Saccharomyces cerevisiae." Processes 8, no. 8 (August 11, 2020): 969. http://dx.doi.org/10.3390/pr8080969.

Full text
Abstract:
Biotransformation of fatty acids from renewable wastewater as feedstock to value-added chemicals is a fascinating commercial opportunity. α,ω-Dicarboxylic acids (DCAs) are building blocks in many industries, such as polymers, cosmetic intermediates, and pharmaceuticals, and can be obtained by chemical synthesis under extreme conditions. However, biological synthesis can replace the traditional chemical synthesis using cytochrome P450 enzymes to oxidize fatty acids to DCAs. Saccharomyces cerevisiae BY(2R)/pYeDP60-CYP52A17SS (BCM), a transgenic strain expressing the galactose-inducible CYP52A17SS cytochrome P450 enzyme, was able to grow in a coconut milk factory wastewater (CCW) medium and produced 12-hydroxydodecanoic acid (HDDA) and 1,12-dodecanedioic acid (DDA). The supplementation of CCW with 10 g/L yeast extract and 20 g/L peptone (YPCCW) markedly increased the yeast growth rate and the yields of 12-HDDA and 1,12-DDA, with the highest levels of approximately 60 and 38 µg/L, respectively, obtained at 30 °C and pH 5. The incubation temperature and medium pH strongly influenced the yeast growth and 1,12-DDA yield, with the highest 1,12-DDA formation at 30 °C and pH 5–5.5. Hence, the S. cerevisiae BCM strain can potentially be used for producing value-added products from CCW.
APA, Harvard, Vancouver, ISO, and other styles
22

Vandermies, Marie, and Patrick Fickers. "Bioreactor-Scale Strategies for the Production of Recombinant Protein in the Yeast Yarrowia lipolytica." Microorganisms 7, no. 2 (January 30, 2019): 40. http://dx.doi.org/10.3390/microorganisms7020040.

Full text
Abstract:
Recombinant protein production represents a multibillion-dollar market. Therefore, it constitutes an important research field both in academia and industry. The use of yeast as a cell factory presents several advantages such as ease of genetic manipulation, growth at high cell density, and the possibility of post-translational modifications. Yarrowia lipolytica is considered as one of the most attractive hosts due to its ability to metabolize raw substrate, to express genes at a high level, and to secrete protein in large amounts. In recent years, several reviews have been dedicated to genetic tools developed for this purpose. Though the construction of efficient cell factories for recombinant protein synthesis is important, the development of an efficient process for recombinant protein production in a bioreactor constitutes an equally vital aspect. Indeed, a sports car cannot drive fast on a gravel road. The aim of this review is to provide a comprehensive snapshot of process tools to consider for recombinant protein production in bioreactor using Y. lipolytica as a cell factory, in order to facilitate the decision-making for future strain and process engineering.
APA, Harvard, Vancouver, ISO, and other styles
23

Zahoor, Farah, Chayaphathra Sooklim, Pattanan Songdech, Orawan Duangpakdee, and Nitnipa Soontorngun. "Selection of Potential Yeast Probiotics and a Cell Factory for Xylitol or Acid Production from Honeybee Samples." Metabolites 11, no. 5 (May 13, 2021): 312. http://dx.doi.org/10.3390/metabo11050312.

Full text
Abstract:
Excessive use of antibiotics has detrimental consequences, including antibiotic resistance and gut microbiome destruction. Probiotic-rich diets help to restore good microbes, keeping the body healthy and preventing the onset of chronic diseases. Honey contains not only prebiotic oligosaccharides but, like yogurt and fermented foods, is an innovative natural source for probiotic discovery. Here, a collection of three honeybee samples was screened for yeast strains, aiming to characterize their potential in vitro probiotic properties and the ability to produce valuable metabolites. Ninety-four isolates out of one-hundred and four were able to grow at temperatures of 30 °C and 37 °C, while twelve isolates could grow at 42 °C. Fifty-eight and four isolates displayed the ability to grow under stimulated gastrointestinal condition, at pH 2.0–2.5, 0.3% (w/v) bile salt, and 37 °C. Twenty-four isolates showed high autoaggregation of 80–100% and could utilize various sugars, including galactose and xylose. The cell count of these isolates (7–9 log cfu/mL) was recorded and stable during 6 months of storage. Genomic characterization based on the internal transcribed spacer region (ITS) also identified four isolates of Saccharomyces cerevisiae displayed good ability to produce antimicrobial acids. These results provided the basis for selecting four natural yeast isolates as starter cultures for potential probiotic application in functional foods and animal feed. Additionally, these S. cerevisiae isolates also produced high levels of acids from fermented sugarcane molasses, an abundant agricultural waste product from the sugar industry. Furthermore, one of ten identified isolates of Meyerozyma guilliermondiii displayed an excellent ability to produce a pentose sugar xylitol at a yield of 0.490 g/g of consumed xylose. Potentially, yeast isolates of honeybee samples may offer various biotechnological advantages as probiotics or metabolite producers of multiproduct-based lignocellulosic biorefinery.
APA, Harvard, Vancouver, ISO, and other styles
24

Zhou, Pingping, Wenping Xie, Zhen Yao, Yongqiang Zhu, Lidan Ye, and Hongwei Yu. "Development of a temperature-responsive yeast cell factory using engineered Gal4 as a protein switch." Biotechnology and Bioengineering 115, no. 5 (January 24, 2018): 1321–30. http://dx.doi.org/10.1002/bit.26544.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

Hoehne, Matthias, and Johannes Kabisch. "ChemInform Abstract: Brewing Painkillers: A Yeast Cell Factory for the Production of Opioids from Sugar." ChemInform 47, no. 11 (February 2016): no. http://dx.doi.org/10.1002/chin.201611266.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Liu, Xiaoying, Rolf Sommer Kaas, Peter Ruhdal Jensen, and Mhairi Workman. "Draft Genome Sequence of the Yeast Pachysolen tannophilus CBS 4044/NRRL Y-2460." Eukaryotic Cell 11, no. 6 (May 29, 2012): 827. http://dx.doi.org/10.1128/ec.00114-12.

Full text
Abstract:
ABSTRACT A draft genome sequence of the yeast Pachysolen tannophilus CBS 4044/NRRL Y-2460 is presented. The organism has the potential to be developed as a cell factory for biorefineries due to its ability to utilize waste feedstocks. The sequenced genome size was 12,238,196 bp, consisting of 34 scaffolds. A total of 4,463 genes from 5,346 predicted open reading frames were annotated with function.
APA, Harvard, Vancouver, ISO, and other styles
27

Zhu, Hanyu, Dongmei Liu, Yuanyuan Wang, Danfeng Ren, Liesheng Zheng, Liguo Chen, and Aimin Ma. "Use of the yeast-like cells of Tremella fuciformis as a cell factory to produce a Pleurotus ostreatus hydrophobin." Biotechnology Letters 39, no. 8 (May 3, 2017): 1167–73. http://dx.doi.org/10.1007/s10529-017-2343-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Baptista, Marlene, Joana T. Cunha, and Lucília Domingues. "Establishment of Kluyveromyces marxianus as a Microbial Cell Factory for Lignocellulosic Processes: Production of High Value Furan Derivatives." Journal of Fungi 7, no. 12 (December 7, 2021): 1047. http://dx.doi.org/10.3390/jof7121047.

Full text
Abstract:
The establishment of lignocellulosic biorefineries is dependent on microorganisms being able to cope with the stressful conditions resulting from the release of inhibitory compounds during biomass processing. The yeast Kluyveromyces marxianus has been explored as an alternative microbial factory due to its thermotolerance and ability to natively metabolize xylose. The lignocellulose-derived inhibitors furfural and 5-hydroxymethylfurfural (HMF) are considered promising building-block platforms that can be converted into a wide variety of high-value derivatives. Here, several K. marxianus strains, isolated from cocoa fermentation, were evaluated for xylose consumption and tolerance towards acetic acid, furfural, and HMF. The potential of this yeast to reduce furfural and HMF at high inhibitory loads was disclosed and characterized. Our results associated HMF reduction with NADPH while furfural-reducing activity was higher with NADH. In addition, furans’ inhibitory effect was higher when combined with xylose consumption. The furan derivatives produced by K. marxianus in different conditions were identified. Furthermore, one selected isolate was efficiently used as a whole-cell biocatalyst to convert furfural and HMF into their derivatives, furfuryl alcohol and 2,5-bis(hydroxymethyl)furan (BHMF), with high yields and productivities. These results validate K. marxianus as a promising microbial platform in lignocellulosic biorefineries.
APA, Harvard, Vancouver, ISO, and other styles
29

Müller, Waltraud G., Dietmar Rieder, Tatiana S. Karpova, Sam John, Zlatko Trajanoski, and James G. McNally. "Organization of chromatin and histone modifications at a transcription site." Journal of Cell Biology 177, no. 6 (June 18, 2007): 957–67. http://dx.doi.org/10.1083/jcb.200703157.

Full text
Abstract:
According to the transcription factory model, localized transcription sites composed of immobilized polymerase molecules transcribe chromatin by reeling it through the transcription site and extruding it to form a surrounding domain of recently transcribed decondensed chromatin. Although transcription sites have been identified in various cells, surrounding domains of recently transcribed decondensed chromatin have not. We report evidence that transcription sites associated with a tandem gene array in mouse cells are indeed surrounded by or adjacent to a domain of decondensed chromatin composed of sequences from the gene array. Formation of this decondensed domain requires transcription and topoisomerase IIα activity. The decondensed domain is enriched for the trimethyl H3K36 mark that is associated with recently transcribed chromatin in yeast and several mammalian systems. Consistent with this, chromatin immunoprecipitation demonstrates a comparable enrichment of this mark in transcribed sequences at the tandem gene array. These results provide new support for the pol II factory model, in which an immobilized polymerase molecule extrudes decondensed, transcribed sequences into its surroundings.
APA, Harvard, Vancouver, ISO, and other styles
30

Włodarczyk, Paweł, and Barbara Włodarczyk. "Microbial Fuel Cell with Ni–Co Cathode Powered with Yeast Wastewater." Energies 11, no. 11 (November 17, 2018): 3194. http://dx.doi.org/10.3390/en11113194.

Full text
Abstract:
Wastewater originating from the yeast industry is characterized by high concentration of pollutants that need to be reduced before the sludge can be applied, for instance, for fertilization of croplands. As a result of the special requirements associated with the characteristics of this production, huge amounts of wastewater are generated. A microbial fuel cell (MFC) forms a device that can apply wastewater as a fuel. MFC is capable of performing two functions at the same time: wastewater treatment and electricity production. The function of MFC is the production of electricity during bacterial digestion (wastewater treatment). This paper analyzes the possibility of applying yeast wastewater to play the function of a MFC (with Ni–Co cathode). The study was conducted on industrial wastewater from a sewage treatment plant in a factory that processes yeast sewage. The Ni–Co alloy was prepared by application of electrochemical method on a mesh electrode. The results demonstrated that the use of MFC coupled with a Ni–Co cathode led to a reduction in chemical oxygen demand (COD) by 90% during a period that was similar to the time taken for reduction in COD in a reactor with aeration. The power obtained in the MFC was 6.1 mW, whereas the volume of energy obtained during the operation of the cell (20 days) was 1.27 Wh. Although these values are small, the study found that this process can offer an additional level of wastewater treatment as a huge amount of sewage is generated in the process. This would provide an initial reduction in COD (and save the energy needed to aerate wastewater) as well as offer the means to generate electricity.
APA, Harvard, Vancouver, ISO, and other styles
31

Ruffell, Daniela. "A factory within a yeast cell: innovative approaches for a sustainable future – an interview with Irina Borodina." FEBS Letters 596, no. 6 (February 15, 2022): 699–702. http://dx.doi.org/10.1002/1873-3468.14311.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Wang, Pingping, Jiali Wang, Guoping Zhao, Xing Yan, and Zhihua Zhou. "Systematic optimization of the yeast cell factory for sustainable and high efficiency production of bioactive ginsenoside compound K." Synthetic and Systems Biotechnology 6, no. 2 (June 2021): 69–76. http://dx.doi.org/10.1016/j.synbio.2021.03.002.

Full text
APA, Harvard, Vancouver, ISO, and other styles
33

Morrissey, John P., Maria M. W. Etschmann, Jens Schrader, and Gustavo M. de Billerbeck. "Cell factory applications of the yeast Kluyveromyces marxianus for the biotechnological production of natural flavour and fragrance molecules." Yeast 32, no. 1 (December 11, 2014): 3–16. http://dx.doi.org/10.1002/yea.3054.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Zhao, Le, Yunhao Zhu, Haoyu Jia, Yongguang Han, Xiaoke Zheng, Min Wang, and Weisheng Feng. "From Plant to Yeast—Advances in Biosynthesis of Artemisinin." Molecules 27, no. 20 (October 14, 2022): 6888. http://dx.doi.org/10.3390/molecules27206888.

Full text
Abstract:
Malaria is a life-threatening disease. Artemisinin-based combination therapy (ACT) is the preferred choice for malaria treatment recommended by the World Health Organization. At present, the main source of artemisinin is extracted from Artemisia annua; however, the artemisinin content in A. annua is only 0.1–1%, which cannot meet global demand. Meanwhile, the chemical synthesis of artemisinin has disadvantages such as complicated steps, high cost and low yield. Therefore, the application of the synthetic biology approach to produce artemisinin in vivo has magnificent prospects. In this review, the biosynthesis pathway of artemisinin was summarized. Then we discussed the advances in the heterologous biosynthesis of artemisinin using microorganisms (Escherichia coli and Saccharomyces cerevisiae) as chassis cells. With yeast as the cell factory, the production of artemisinin was transferred from plant to yeast. Through the optimization of the fermentation process, the yield of artemisinic acid reached 25 g/L, thereby producing the semi-synthesis of artemisinin. Moreover, we reviewed the genetic engineering in A. annua to improve the artemisinin content, which included overexpressing artemisinin biosynthesis pathway genes, blocking key genes in competitive pathways, and regulating the expression of transcription factors related to artemisinin biosynthesis. Finally, the research progress of artemisinin production in other plants (Nicotiana, Physcomitrella, etc.) was discussed. The current advances in artemisinin biosynthesis may help lay the foundation for the remarkable up-regulation of artemisinin production in A. annua through gene editing or molecular design breeding in the future.
APA, Harvard, Vancouver, ISO, and other styles
35

Liu, Junfeng, Virginija Cvirkaite-Krupovic, Diana P. Baquero, Yunfeng Yang, Qi Zhang, Yulong Shen, and Mart Krupovic. "Virus-induced cell gigantism and asymmetric cell division in archaea." Proceedings of the National Academy of Sciences 118, no. 15 (March 29, 2021): e2022578118. http://dx.doi.org/10.1073/pnas.2022578118.

Full text
Abstract:
Archaeal viruses represent one of the most mysterious parts of the global virosphere, with many virus groups sharing no evolutionary relationship to viruses of bacteria or eukaryotes. How these viruses interact with their hosts remains largely unexplored. Here we show that nonlytic lemon-shaped virus STSV2 interferes with the cell cycle control of its host, hyperthermophilic and acidophilic archaeon Sulfolobus islandicus, arresting the cell cycle in the S phase. STSV2 infection leads to transcriptional repression of the cell division machinery, which is homologous to the eukaryotic endosomal sorting complexes required for transport (ESCRT) system. The infected cells grow up to 20-fold larger in size, have 8,000-fold larger volume compared to noninfected cells, and accumulate massive amounts of viral and cellular DNA. Whereas noninfected Sulfolobus cells divide symmetrically by binary fission, the STSV2-infected cells undergo asymmetric division, whereby giant cells release normal-sized cells by budding, resembling the division of budding yeast. Reinfection of the normal-sized cells produces a new generation of giant cells. If the CRISPR-Cas system is present, the giant cells acquire virus-derived spacers and terminate the virus spread, whereas in its absence, the cycle continues, suggesting that CRISPR-Cas is the primary defense system in Sulfolobus against STSV2. Collectively, our results show how an archaeal virus manipulates the cell cycle, transforming the cell into a giant virion-producing factory.
APA, Harvard, Vancouver, ISO, and other styles
36

Carneiro, Clara Vida Galrão Corrêa, Luana Assis Serra, Thályta Fraga Pacheco, Letícia Maria Mallmann Ferreira, Lívia Teixeira Duarte Brandão, Mariana Nogueira de Moura Freitas, Débora Trichez, and João Ricardo Moreira de Almeida. "Advances in Komagataella phaffii Engineering for the Production of Renewable Chemicals and Proteins." Fermentation 8, no. 11 (October 24, 2022): 575. http://dx.doi.org/10.3390/fermentation8110575.

Full text
Abstract:
The need for a more sustainable society has prompted the development of bio-based processes to produce fuels, chemicals, and materials in substitution for fossil-based ones. In this context, microorganisms have been employed to convert renewable carbon sources into various products. The methylotrophic yeast Komagataella phaffii has been extensively used in the production of heterologous proteins. More recently, it has been explored as a host organism to produce various chemicals through new metabolic engineering and synthetic biology tools. This review first summarizes Komagataella taxonomy and diversity and then highlights the recent approaches in cell engineering to produce renewable chemicals and proteins. Finally, strategies to optimize and develop new fermentative processes using K. phaffii as a cell factory are presented and discussed. The yeast K. phaffii shows an outstanding performance for renewable chemicals and protein production due to its ability to metabolize different carbon sources and the availability of engineering tools. Indeed, it has been employed in producing alcohols, carboxylic acids, proteins, and other compounds using different carbon sources, including glycerol, glucose, xylose, methanol, and even CO2.
APA, Harvard, Vancouver, ISO, and other styles
37

Dikicioglu, Duygu, Pınar Pir, and Stephen G. Oliver. "Predicting complex phenotype–genotype interactions to enable yeast engineering: Saccharomyces cerevisiae as a model organism and a cell factory." Biotechnology Journal 8, no. 9 (August 23, 2013): 1017–34. http://dx.doi.org/10.1002/biot.201300138.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Cai, Peng, Xingpeng Duan, Xiaoyan Wu, Linhui Gao, Min Ye, and Yongjin J. Zhou. "Recombination machinery engineering facilitates metabolic engineering of the industrial yeast Pichia pastoris." Nucleic Acids Research 49, no. 13 (July 1, 2021): 7791–805. http://dx.doi.org/10.1093/nar/gkab535.

Full text
Abstract:
Abstract The industrial yeast Pichia pastoris has been harnessed extensively for production of proteins, and it is attracting attention as a chassis cell factory for production of chemicals. However, the lack of synthetic biology tools makes it challenging in rewiring P. pastoris metabolism. We here extensively engineered the recombination machinery by establishing a CRISPR-Cas9 based genome editing platform, which improved the homologous recombination (HR) efficiency by more than 54 times, in particular, enhanced the simultaneously assembly of multiple fragments by 13.5 times. We also found that the key HR-relating gene RAD52 of P. pastoris was largely repressed in compared to that of Saccharomyces cerevisiae. This gene editing system enabled efficient seamless gene disruption, genome integration and multiple gene assembly with positive rates of 68–90%. With this efficient genome editing platform, we characterized 46 potential genome integration sites and 18 promoters at different growth conditions. This library of neutral sites and promoters enabled two-factorial regulation of gene expression and metabolic pathways and resulted in a 30-fold range of fatty alcohol production (12.6–380 mg/l). The expanding genetic toolbox will facilitate extensive rewiring of P. pastoris for chemical production, and also shed light on engineering of other non-conventional yeasts.
APA, Harvard, Vancouver, ISO, and other styles
39

Carsanba, Erdem, Manuela Pintado, and Carla Oliveira. "Fermentation Strategies for Production of Pharmaceutical Terpenoids in Engineered Yeast." Pharmaceuticals 14, no. 4 (March 26, 2021): 295. http://dx.doi.org/10.3390/ph14040295.

Full text
Abstract:
Terpenoids, also known as isoprenoids, are a broad and diverse class of plant natural products with significant industrial and pharmaceutical importance. Many of these natural products have antitumor, anti-inflammatory, antibacterial, antiviral, and antimalarial effects, support transdermal absorption, prevent and treat cardiovascular diseases, and have hypoglycemic activities. Production of these compounds are generally carried out through extraction from their natural sources or chemical synthesis. However, these processes are generally unsustainable, produce low yield, and result in wasting of substantial resources, most of them limited. Microbial production of terpenoids provides a sustainable and environment-friendly alternative. In recent years, the yeast Saccharomyces cerevisiae has become a suitable cell factory for industrial terpenoid biosynthesis due to developments in omics studies (genomics, transcriptomics, metabolomics, proteomics), and mathematical modeling. Besides that, fermentation development has a significant importance on achieving high titer, yield, and productivity (TYP) of these compounds. Up to now, there have been many studies and reviews reporting metabolic strategies for terpene biosynthesis. However, fermentation strategies have not been yet comprehensively discussed in the literature. This review summarizes recent studies of recombinant production of pharmaceutically important terpenoids by engineered yeast, S. cerevisiae, with special focus on fermentation strategies to increase TYP in order to meet industrial demands to feed the pharmaceutical market. Factors affecting recombinant terpenoids production are reviewed (strain design and fermentation parameters) and types of fermentation process (batch, fed-batch, and continuous) are discussed.
APA, Harvard, Vancouver, ISO, and other styles
40

Włodarczyk, Barbara, and Paweł P. Włodarczyk. "The Membrane-Less Microbial Fuel Cell (ML-MFC) with Ni-Co and Cu-B Cathode Powered by the Process Wastewater from Yeast Production." Energies 13, no. 15 (August 2, 2020): 3976. http://dx.doi.org/10.3390/en13153976.

Full text
Abstract:
Research related to measurements of electricity production was combined with parallel wastewater parameter reduction in a membrane-less microbial fuel cell (ML-MFC) fed with industry process wastewater (from a yeast factory). Electrodes with Ni–Co and Cu–B catalysts were used as cathodes. A carbon electrode (carbon cloth) was used as a reference due to its widespread use. It was demonstrated that all analyzed electrodes could be employed as cathodes in ML-MFC fed with process wastewater from yeast production. Electricity measurements during ML-MFC operations indicated that power (6.19 mW) and current density (0.38 mA·cm−2) were the highest for Ni–Co electrodes. In addition, during the exploitation of ML-MFC, it was recorded that the chemical oxygen demand (COD) removal per time for all types of electrodes was similar to the duration of COD decrease in the conditions for wastewater aeration. However, the COD reduction curve for aeration took the most favorable course. The concentration of NH4+ in ML-MFC remained virtually constant throughout the measurement period, whereas NO3− levels indicated almost complete removal (with a minimum increase in the last days of cell exploitation).
APA, Harvard, Vancouver, ISO, and other styles
41

Tramontin, Larissa Ribeiro Ramos, Kanchana Rueksomtawin Kildegaard, Suresh Sudarsan, and Irina Borodina. "Enhancement of Astaxanthin Biosynthesis in Oleaginous Yeast Yarrowia lipolytica via Microalgal Pathway." Microorganisms 7, no. 10 (October 19, 2019): 472. http://dx.doi.org/10.3390/microorganisms7100472.

Full text
Abstract:
Astaxanthin is a high-value red pigment and antioxidant used by pharmaceutical, cosmetics, and food industries. The astaxanthin produced chemically is costly and is not approved for human consumption due to the presence of by-products. The astaxanthin production by natural microalgae requires large open areas and specialized equipment, the process takes a long time, and results in low titers. Recombinant microbial cell factories can be engineered to produce astaxanthin by fermentation in standard equipment. In this work, an oleaginous yeast Yarrowia lipolytica was engineered to produce astaxanthin at high titers in submerged fermentation. First, a platform strain was created with an optimised pathway towards β-carotene. The platform strain produced 331 ± 66 mg/L of β-carotene in small-scale cultivation, with the cellular content of 2.25% of dry cell weight. Next, the genes encoding β-ketolase and β-hydroxylase of bacterial (Paracoccus sp. and Pantoea ananatis) and algal (Haematococcus pluvialis) origins were introduced into the platform strain in different copy numbers. The resulting strains were screened for astaxanthin production, and the best strain, containing algal β-ketolase and β-hydroxylase, resulted in astaxanthin titer of 44 ± 1 mg/L. The same strain was cultivated in controlled bioreactors, and a titer of 285 ± 19 mg/L of astaxanthin was obtained after seven days of fermentation on complex medium with glucose. Our study shows the potential of Y. lipolytica as the cell factory for astaxanthin production.
APA, Harvard, Vancouver, ISO, and other styles
42

Cui, Na, and Victor Pozzobon. "Food-Grade Cultivation of Saccharomyces cerevisiae from Potato Waste." AgriEngineering 4, no. 4 (October 17, 2022): 951–68. http://dx.doi.org/10.3390/agriengineering4040061.

Full text
Abstract:
Potato waste is generated in a high amount, stably over the year, by operators capable of recovering it. Currently, it is valorized as feed, bioethanol, or biogas. This work explores another avenue to increase the valorization of this waste: the production of yeast production to serve as fodder or single-cell protein. First, potatoes were deconstructed into fermentable sugars by acid hydrolysis using food-grade techniques. Then, after pH adjustment, Saccharomyces cerevisiae was inoculated, and cell growth was monitored. For optimization purposes, this procedure was led over a large range of temperature (90–120 °C) and operation time (30–120 min), for a 1/2 solid/liquid ratio. Response surfaces methodology allowed to achieve a maximum sugar release (44.4 g/L) for 99 min under 103 °C. Then, a numerical model combining biological performances and factory process planning was used to derive process productivity (the best compromise between sugar release and cell growth). Maximal productivity (82.8 gYeast/w/L in batch mode, 110 gYeast/w/L in fed-batch mode) was achieved for 103 min under 94 °C. Furthermore, the process’s robustness was confirmed by a sensibility analysis. Finally, as the proposed procedure preserves the food-grade quality of the substrate, the produced yeast can be used as food or feed.
APA, Harvard, Vancouver, ISO, and other styles
43

Matos-Perdomo, Emiliano, and Félix Machín. "Nucleolar and Ribosomal DNA Structure under Stress: Yeast Lessons for Aging and Cancer." Cells 8, no. 8 (July 26, 2019): 779. http://dx.doi.org/10.3390/cells8080779.

Full text
Abstract:
Once thought a mere ribosome factory, the nucleolus has been viewed in recent years as an extremely sensitive gauge of diverse cellular stresses. Emerging concepts in nucleolar biology include the nucleolar stress response (NSR), whereby a series of cell insults have a special impact on the nucleolus. These insults include, among others, ultra-violet radiation (UV), nutrient deprivation, hypoxia and thermal stress. While these stresses might influence nucleolar biology directly or indirectly, other perturbances whose origin resides in the nucleolar biology also trigger nucleolar and systemic stress responses. Among the latter, we find mutations in nucleolar and ribosomal proteins, ribosomal RNA (rRNA) processing inhibitors and ribosomal DNA (rDNA) transcription inhibition. The p53 protein also mediates NSR, leading ultimately to cell cycle arrest, apoptosis, senescence or differentiation. Hence, NSR is gaining importance in cancer biology. The nucleolar size and ribosome biogenesis, and how they connect with the Target of Rapamycin (TOR) signalling pathway, are also becoming important in the biology of aging and cancer. Simple model organisms like the budding yeast Saccharomyces cerevisiae, easy to manipulate genetically, are useful in order to study nucleolar and rDNA structure and their relationship with stress. In this review, we summarize the most important findings related to this topic.
APA, Harvard, Vancouver, ISO, and other styles
44

Zhurinsky, Jacob, Silvia Salas-Pino, Ana B. Iglesias-Romero, Antonio Torres-Mendez, Benjamin Knapp, Ignacio Flor-Parra, Jiyong Wang, et al. "Effects of the microtubule nucleator Mto1 on chromosomal movement, DNA repair, and sister chromatid cohesion in fission yeast." Molecular Biology of the Cell 30, no. 21 (October 1, 2019): 2695–708. http://dx.doi.org/10.1091/mbc.e19-05-0301.

Full text
Abstract:
Although the function of microtubules (MTs) in chromosomal segregation during mitosis is well characterized, much less is known about the role of MTs in chromosomal functions during interphase. In the fission yeast Schizosaccharomyces pombe, dynamic cytoplasmic MT bundles move chromosomes in an oscillatory manner during interphase via linkages through the nuclear envelope (NE) at the spindle pole body (SPB) and other sites. Mto1 is a cytoplasmic factor that mediates the nucleation and attachment of cytoplasmic MTs to the nucleus. Here, we test the function of these cytoplasmic MTs and Mto1 on DNA repair and recombination during interphase. We find that mto1Δ cells exhibit defects in DNA repair and homologous recombination (HR) and abnormal DNA repair factory dynamics. In these cells, sister chromatids are not properly paired, and binding of Rad21 cohesin subunit along chromosomal arms is reduced. Our findings suggest a model in which cytoplasmic MTs and Mto1 facilitate efficient DNA repair and HR by promoting dynamic chromosomal organization and cohesion in the nucleus.
APA, Harvard, Vancouver, ISO, and other styles
45

Riyanti, Eny Ida, and Edy Listanto. "INHIBITION OF THE GROWTH OF TOLERANT YEAST Saccharomyces cerevisiae STRAIN I136 BY A MIXTURE OF SYNTHETIC INHIBITORS." Indonesian Journal of Agricultural Science 18, no. 1 (September 14, 2017): 17. http://dx.doi.org/10.21082/ijas.v18n1.2017.p17-24.

Full text
Abstract:
<p>Biomass from lignocellulosic wastes is a potential source for biobased products. However, one of the constraints in utilization of biomass hydrolysate is the presence of inhibitors. Therefore, the use of inhibitor-tolerant microorganisms in the fermentation is required. The study aimed to investigate the effect of a mixture of inhibitors on the growth of Saccharomyces cerevisiae strain I136 grown in medium containing synthetic inhibitors (acetic acid, formic acid, furfural, 5-hydroxymethyl furfural/5-HMF, and levulinic acid) in four different concentrations with a mixture of carbon sources, glucose (50 g.l-1) and xylose (50 g.l-1) at 30oC. The parameters related to growth and fermentation products were observed. Results showed that the strain was able to grow in media containing natural inhibitors (BSL medium) with µmax of 0.020/h. Higher level of synthetic inhibitors prolonged the lag phase, decreased the cell biomass and ethanol production, and specific growth rate. The strain could detoxify furfural and 5-HMF and produced the highest ethanol (Y(p/s) of 0.32 g.g-1) when grown in BSL. Glucose was utilized as its level decreased in a result of increase in cell biomass, in contrast to xylose which was not consumed. The highest cell biomass was produced in YNB with Y (x/s) value of 0.25 g.g-1. The strain produced acetic acid as a dominant side product and could convert furfural into a less toxic compound, hydroxyl furfural. This robust tolerant strain provides basic information on resistance mechanism and would be useful for bio-based cell factory using lignocellulosic materials. </p>
APA, Harvard, Vancouver, ISO, and other styles
46

Çiftçi, T., and I. Öztürk. "Anaerobic Treatment of the High Strength Wastes from the Yeast Industry." Water Science and Technology 28, no. 2 (July 1, 1993): 199–209. http://dx.doi.org/10.2166/wst.1993.0104.

Full text
Abstract:
This paper presents the full-scale anaerobic treatment results from a fermentation plant producing baker's yeast from sugar beet molasses. The process of baker's yeast production generates high strength industrial effluents with a chemical oxygen demand (GOD) of 10 000-30 000 mg/liter. In addition to the sugar containing substances sulphur and nitrogen containing substances are added to the batch processes to promote cell growth and to control pH. This results in rather high concentrations of sulphate 0000-2700 mg/l) and ammonia (400-900 mg/l) in the wastewater. The treatment plant at Pakmaya Izmit Factory has two different processes: anaerobic first-stage treatment and aerobic second stage treatment. The anaerobic first-stage treatment system includes a buffer tank, an acid reactor, two methane reactors, lamella separators, a gas storage tank and gas burning facilities. The anaerobic reactors were constructed as upflow anaerobic sludge blanket reactors (UASBR) with internal sludge recirculation facilities. The anaerobic reactors have been operating in series mode at mesophilic temperature ranges. Long term Organic Loading Rates (OLR) in the acid, the first and the second stage methane reactors have been averaging 9.8, 8.6 and 3 kg COD/m3·d respectively. Average COD removal is 75 percent in the anaerobic pretreatment stage. Average biogas production is 8000 m3/d, corresponding to a biogas conversion yield of 0.6 m3 per kg COD removed and it is equivalent to a netbioenergy recovery of 40 000 kWh/d.
APA, Harvard, Vancouver, ISO, and other styles
47

Koirala, Niranjan, Sareeta Khanal, Sujan Chaudhary, Sagar Gautam, Shiv Nandan Sah, Prince Subba, Najat Marraiki, and Gaber El-Saber Batiha. "Potential surface active agent production using very low grade and cheap substrate by Bacillus subtilis as microbial cell factory." Nepal Journal of Biotechnology 9, no. 2 (December 31, 2021): 21–28. http://dx.doi.org/10.54796/njb.v9i2.41910.

Full text
Abstract:
Bio-surfactants are surface-active molecules which are produced by the wide range of microbes including bacteria, fungi, moulds, and yeast. This study was conducted to identify bio-surfactants by Bacillus subtilis combined with use of cheap substrates and industrial wastes (Mustard cake, Whey and Soya cake) which are found locally in Nepal. Bacillus subtilis, one of the most potential bio-surfactants producer; was isolated from soil sample of hydrocarbon contaminated site. Isolates were grown in a Minimal Salt Media (MSM) with 10% (v/v) mustard oil cake, whey and soya cake separately. The presence and potential of surfactant was determined by the oil spreading technique, emulsification index (%E24) and surface tension measurement. It was revealed that the surface tensions of cell free extract were 54.41, 60.02 and 56.64 mN/m for from mustard cake, whey and soya cake respectively as compared to distilled water (72.09) at 25oC. The emulsification index values was found to be highest in engine oil from the bio-surfactant extracted from mustard cake, soya cake and whey respectively. Similarly, mustard oil showed the lowest value of emulsification index. The highest emulsification activity was shown in mustard oil i.e. 1.13 from the cell free extract from mustard oil and lowest in engine oil i.e., 0.07, by the extract from soya cake medium, when measured in spectrophotometer at 540 nm. In conclusion, strain of Bacillus subtilis was found to be the potential surface active agent producers on the mustard oil cake, which can be useful medium for various environmental, food, medicinal and industrial processes.
APA, Harvard, Vancouver, ISO, and other styles
48

Yang, Shuo, Junfeng Shen, Jiliang Deng, Hongxing Li, Jianzhi Zhao, Hongting Tang, and Xiaoming Bao. "Engineering Cell Polarization Improves Protein Production in Saccharomyces cerevisiae." Microorganisms 10, no. 10 (October 11, 2022): 2005. http://dx.doi.org/10.3390/microorganisms10102005.

Full text
Abstract:
Saccharomyces cerevisiae has been widely used as a microbial cell factory to produce recombinant proteins. Therefore, enhancing the protein production efficiency of yeast cell factories to expand the market demand for protein products is necessary. Recombinant proteins are often retained in the secretory pathway because of the limited protein transport performed by vesicle trafficking. Cell polarization describes the asymmetric organization of the plasma membrane cytoskeleton and organelles and tightly regulates vesicle trafficking for protein transport. Engineering vesicle trafficking has broadly been studied by the overexpression or deletion of key genes involved but not by modifying cell polarization. Here, we used α−amylase as a reporter protein, and its secretion and surface−display were first improved by promoter optimization. To study the effect of engineering cell polarization on protein production, fourteen genes related to cell polarization were overexpressed. BUD1, CDC42, AXL1, and BUD10 overexpression increased the activity of surface−displayed α−amylase, and BUD1, BUD3, BUD4, BUD7, and BUD10 overexpression enhanced secreted α−amylase activity. Furthermore, BUD1 overexpression increased the surface−displayed and secreted α−amylase expression by 56% and 49%, respectively. We also observed that the combinatorial modification and regulation of gene expression improved α-amylase production in a dose−dependent manner. BUD1 and CDC42 co−overexpression increased the α−amylase surface display by 100%, and two genomic copies of BUD1 improved α−amylase secretion by 92%. Furthermore, these modifications were used to improve the surface display and secretion of the recombinant β−glucosidase protein. Our study affords a novel insight for improving the surface display and secretion of recombinant proteins.
APA, Harvard, Vancouver, ISO, and other styles
49

Nurdiani, Dini, Hariyatun Hariyatun, Nuruliawaty Utami, Eko Wahyu Putro, and Wien Kusharyoto. "Enhancement in Human Insulin Precursor Secretion by Pichia pastoris through Modification of Expression Conditions." HAYATI Journal of Biosciences 29, no. 1 (November 30, 2021): 22–30. http://dx.doi.org/10.4308/hjb.29.1.22-30.

Full text
Abstract:
Pichia pastoris is an alternative yeast expression system to produce heterologous proteins. It has excellent characteristics for an industrial cell factory, such as its ability to reach high cell densities, high secretory capacity, and a low level of native proteins. In our previous study, we introduced a synthetic insulin precursor (IP)-encoding gene constructed in a pD902 expression vector into P. pastoris. However, the P. pastoris recombinant strains expressed a little amount of IP protein. Here, we modified the expression conditions, including inoculum density, methanol concentration, methanol induction time, pH, and temperature, to obtain a higher amount of secreted IP than our previous result. Protein analysis for studying the five parameters was conducted by SDS-PAGE, and the protein amount was estimated by ImageJ applying lysozyme as standard. We successfully enhanced the IP expression by modifying expression conditions. The highest increased of up to 100 folds was achieved when methanol concentration for induction was arranged at 3% (v/v), and the initial cell density for methanol induction was set at an optical density at 600 nm (OD600) of approximately 10 compared to the standard procedure, where the expression was set at 0.5% (v/v) methanol induction and initial cell density at OD600 = 1.
APA, Harvard, Vancouver, ISO, and other styles
50

Rippert, Dorthe, Federica Linguardo, Andreea Perpelea, Mathias Klein, and Elke Nevoigt. "Identification of the Aldo-Keto Reductase Responsible for d-Galacturonic Acid Conversion to l-Galactonate in Saccharomyces cerevisiae." Journal of Fungi 7, no. 11 (October 27, 2021): 914. http://dx.doi.org/10.3390/jof7110914.

Full text
Abstract:
d-galacturonic acid (d-GalUA) is the main constituent of pectin, a complex polysaccharide abundant in several agro-industrial by-products such as sugar beet pulp or citrus peel. During several attempts to valorise d-GalUA by engineering the popular cell factory Saccharomyces cerevisiae, it became obvious that d-GalUA is, to a certain degree, converted to l-galactonate (l-GalA) by an endogenous enzymatic activity. The goal of the current work was to clarify the identity of the responsible enzyme(s). A protein homology search identified three NADPH-dependent unspecific aldo-keto reductases in baker’s yeast (encoded by GCY1, YPR1 and GRE3) that show sequence similarities to known d-GalUA reductases from filamentous fungi. Characterization of the respective deletion mutants and an in vitro enzyme assay with a Gcy1 overproducing strain verified that Gcy1 is mainly responsible for the detectable reduction of d-GalUA to l-GalA.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography