DOI QR코드

DOI QR Code

The Actinobacterium Corynebacterium glutamicum, an Industrial Workhorse

  • Lee, Joo-Young (Department of Biotechnology, The Catholic University of Korea) ;
  • Na, Yoon-Ah (Department of Biotechnology, The Catholic University of Korea) ;
  • Kim, Eungsoo (Department of Biological Engineering, Inha University) ;
  • Lee, Heung-Shick (Department of Biotechnology and Bioinformatics, Korea University) ;
  • Kim, Pil (Department of Biotechnology, The Catholic University of Korea)
  • Received : 2016.01.21
  • Accepted : 2016.01.31
  • Published : 2016.05.28

Abstract

Starting as a glutamate producer, Corynebacterium glutamicum has played a variety of roles in the industrial production of amino acids, one of the most important areas of white biotechnology. From shortly after its genome information became available, C. glutamicum has been applied in various production processes for value-added chemicals, fuels, and polymers, as a key organism in industrial biotechnology alongside the surprising progress in systems biology and metabolic engineering. In addition, recent studies have suggested another potential for C. glutamicum as a synthetic biology platform chassis that could move the new era of industrial microbial biotechnology beyond the classical field. Here, we review the recent progress and perspectives in relation to C. glutamicum, which demonstrate it as one of the most promising and valuable workhorses in the field of industrial biotechnology.

Keywords

Introduction

Corynebacterium glutamicum was isolated in the search to identify a natural glutamate producer by researchers of the company Kyowa Hakko in 1956 [70]. C. glutamicum is a gram-positive, non-endotoxin, non-sporulating, and generally recognized as safe (GRAS) host that has been widely used for the industrial production of ʟ-glutamate and ʟ-lysine. The publication of the C. glutamicum ATCC 13032 genome sequence by two independent groups [43,50] in 2003 could be regarded as the beginning of a new era for C. glutamicum in the field of industrial microbiology and biotechnology. The release of the complete genome sequence provided a platform that would allow better understanding and easier engineering of C. glutamicum. Heretofore, the progress in metabolic engineering based on the development and integration of genetic engineering tools, systems biology, and omics-based global analysis techniques brought C. glutamicum forward from among the many other workhorses used in industry [10,149]. During the last decade, more than 100 studies reporting on the development and application of high-throughput technologies such as genomics, transcriptomics, proteomics, and metabolomics have been published. The annual number of both publications and patents increased quickly after the complete genome information was released (Fig. 1A). First, transcriptome analysis using DNA microarray was established in the early 2000s [35,84,89,147]. Later, approaches followed for the analysis of global transcriptional regulatory networks in C. glutamicum [8,124,149], the analysis of industrial producer strains [19,35,67,74,128], and the analysis of fermentation processes [15,55]. In addition to the application of high-throughput sequencing in transcriptome analysis, the development of RNA-seq [107] as an alternative to DNA microarray and capillary DNA sequencing allowed faster, deeper, and more precise insights into understanding the transcriptional regulatory mechanisms [92], function, and occurrence of small RNAs [87]. RNA-seq has also been applied in the analysis of adaptive evolution [78] and lysine producer strains [64]. The development and application of proteomics analysis tools such as 2-D PAGE for high-throughput, high-resolution proteome separation and MALDI-TOF MS provided a proteome reference map with comprehensive coverage of proteins [12,37,115]. Here, we summarize the studies and achievements relating to C. glutamicum, including recent progress in the field of bio-based production of value-added chemicals and recombinant proteins. Selected cases of achievements relating to C. glutamicum are provided in this mini-review.

Fig. 1.A brief history of Corynebacterium glutamicum. (A) The annual number of reports relevant to C. glutamicum. (B) Historical and technical milestones in C. glutamicum (upper), together with milestones in biotechnology (lower). Boxes: technical and historical events in biotechnology (gray); fermentation or metabolic engineering in microbes (black); trends in microbial biotechnology (gray-filled). Remarkable historical events are highlighted in bold.

 

Timeline of C. glutamicum as an Industrial Microbe

After isolation as a glutamate producer, much effort was made to understand C. glutamicum as well as to develop this microbe for industrial purposes. Up to 2015, over 2,700 papers and 1,700 patents have been reported relating to C. glutamicum. Among those reports, we selected and summarized the annual number of the reports in Fig. 1A and historical and technical milestones in Fig. 1B.

History of C. glutamicum Strains (Isolation, Engineering, and Future)

In the 1950s, Abe and colleagues at Kyowa Hakko carried out taxonomical studies on 208 glutamate-producing strains, isolating what was designated as Micrococcus glutamicum, but which later came to be known as C. glutamicum [1,70]. These strains and numerous other strains were assorted to the genera Brevibacterium, Microbacterium, Micrococcus, or Arthrobacter. Later, owing to progress in taxonomy (e.g., 16S rDNA sequence analysis and FT-IR spectral comparison), many of the formerly mis-classified strains were reclassified as C. glutamicum [80,93]. Among the first-isolated strains, ATCC13032, a wild-type strain of C. glutamicum, was mostly studied for examination of its physiological properties, metabolism, development of genetic tools, and engineering of C. glutamicum. Another widely used type strain, C. glutamicum R, was first applied for cultivation under conditions of oxygen deprivation. The R strain was then engineered in order to produce ethanol and organic acids such as succinate, lactate, and acetate, using its anaerobic metabolism under oxygen-deprived conditions. Meanwhile, many industrial C. glutamicum strains (screened or engineered) were also studied. To date, 227 type strains, including ATCC13032, have been deposited at American Type Culture Collection (ATCC; http://www.actt.org). In addition, 50 type strains can be found at StrainInfo (http:// www.straininfo.net/taxa/569). Recently, AR strains that were mutated and metabolically engineered to enhance the production of arginine [103], genome-reduced strains initiated from prophage-free strain MB001 [7,138], and a minicells-generating strain that can be applied in drug delivery systems [77] have been reported. Genome information of the first-published complete genome sequence of C. glutamicum has been made available at CoryneRegNet (http://www.coryneregnet.de) and KEGG (http://www.kegg.jp).

To date, genome assembly and annotation reports of 14 strains (9 complete genomes; 5 draft genomes) can be found in the genome database at NCBI (http://www.ncbi.nlm.nih.gov/genome/genomes/469). Reported genome assembly and annotation in NCBI are shown in Table 1. The small cryptic plasmids such as pBL1 and pAG3 were also found in C. glutamicum and other coryneform bacteria (Table 2).

Table 1.a Symbols: ●, complete genome sequence; ◐, draft genome sequence (scaffold or contig).

Table 2.Native plasmids of C. glutamicum registered in NCBI.

Benefits of C. glutamicum as an Industrial Host

C. glutamicum has many fundamental physiological properties that make it an important industrial workhorse. These properties are listed as follows: (i) GRAS, generally C. glutamicum is recognized as a safe strain for human; (ii) fast growth to high cell densities [32]; (iii) genetically stable owing to the lack of a recombination repair system [90]; (iv) limited restriction-modification system [143]; (v) no autolysis and maintenance of metabolic activity under growth-arrested conditions [44]; (vi) low protease activity favoring recombinant protein production [59]; (vii) plasticity of metabolism and strong secondary metabolism properties [148]; and (viii) broad spectrum of carbon utilization (pentoses, hexoses, and alternative carbon sources); stress-tolerance to carbon sources [59,113]. Taken together, these physiological properties render C. glutamicum accessible to manipulation and cultivation in robust industrial conditions.

Toolbox

Numerous physiological and genetic techniques were developed to understand and manipulate C. glutamicum after genome sequences of several strains were made available [43,50,157], and genetic engineering tools have also been developed that are now available to be used in the manipulation of C. glutamicum. Historical events of toolbox development for C. glutamicum are briefly shown in Fig. 1B.

Genome manipulations. Owing to advancements in recombinant DNA technology, various tools for genome manipulation of C. glutamicum have been developed since the 1990s [36,40,120,139]. Basic tools, such as gene integration, replacement, and disruption for genome engineering of C. glutamicum were developed and established from a conjugation system of E. coli using mobilizable (mob-carrying) vectors [120]. Attempts were made to enhance the transfer efficiency and overcome the restriction-modification system (e.g., dam-, dcm-, mcrBC- mutants) [114]. Single-crossover recombination [143] and double-crossover recombination (markerless method) were developed for gene replacement or disruption methods via events [114]. Cre/loxP-mediated recombination systems to rearrange large genome regions have been reported [129-132]. Recently, Unthan et al. [138] reported that combinatory deletions of irrelevant large genes would decrease the genome size to 2,561 kbp.

Promoters and plasmids. In addition to the research on the screening of native promoters of C. glutamicum, there has been development of promoters using mutation and selection for engineering purposes, particularly in strong and constitutive promoters (e.g., Pfbp, Psod, and Ptuf) [99]. Most inducible promoters and vectors were adopted from the inducible expression systems of E. coli promoters (e.g., Plac, Ptac, and Ptrc) and lac operator-repressor. Even leaderless fully synthetic promoters have been recently reported [153]. Promoters used in engineering of C. glutamicum are summarized in Table 3. Attempts to tightly regulate protein expression induction by IPTG have been made numerous times using different vectors such as pEKEx1, pXMJ19, and pVWEx1 [27,45,105]. Owing to the low IPTG permeability of C. glutamicum, IPTG inducible expression systems tend to display a lower level of expression in C. glutamicum than those in E. coli [104]. However, a recent study reported that an engineered C. glutamicum carrying the DE3 region from E. coli BL21 (DE3) showed tight regulation as well as controllable expression using IPTG with a homogenous population [72].

Table 3.Summary of promoters used in engineering of C. glutamicum.

After small native plasmids were discovered in amino acid-producing corynebacteria in the 1980s (Table 2), plasmid vectors were developed [57,112]. Based on native plasmids, vectors for various purposes (cloning, promoter activity report, and expression) were developed starting from the 1990s [27,36,41,120,139,140]. Plasmid vectors used in the engineering of C. glutamicum are summarized in Table 4. Recent progress in synthetic biology (e.g., commercial gene synthesis, biobrick parts) can make combinatorial assembly of various constructions possible with less effort than before.

Table 4.aTc, anhydrotetracycline; Cm, chloramphenicol; IPTG, isopropyl β-D-1-thiogalactopyranoside ; Km, kanamycin.

 

Bio-Based Chemicals from C. glutamicum

C. glutamicum has had excellent success in the large-scale production of glutamate and lysine accompanied by an increase in knowledge of the metabolism and regulatory networks as well as the development of tools for genetic engineering. As such, C. glutamicum has become the preferred microbe in white biotechnology for the industrial production of value-added chemicals. A summary of the current status of the production of bio-based chemicals with C. glutamicum is shown in Table 5.

Table 5.n.d., not determined. Numerical values in titer, yield, and productivity were adapted or calculated based on values in the references. aMaximum conversion yield.

Amino Acids

Starting with ʟ-glutamate and ʟ-lysine, C. glutamicum was applied to industrial production of various amino acids during the half century after its discovery in the 1950s (Fig. 1B). Amino acids have various characteristics in terms of chemical properties, taste, and nutritional value, and thus have many potential uses, such as feed supplements, food additives, and materials for pharmaceuticals and polymers. Most ʟ-amino acids are produced by microbial fermentation. Within a few years after the first report of ʟ-glutamate fermentation with C. glutamicum [70], the microbe was also used to produce ʟ-lysine (Fig. 1B, [91]). In the early era, strain development for amino acid fermentation depended on random mutation and selection such as analog-resistant mutant screening, which contributed to the development of many commercial and potent producers [39]. Along with the advancement in genetic engineering technology, development and application of DNA technology to C. glutamicum for amino acid production began in the 1980s. From the beginning until the present day, the amino acid ʟ-glutamate has possessed the largest market share of amino acid fermentation (2.5 M tons/year), relying on C. glutamicum for its production. Recently, Zhang et al. [161] reported on a ʟ-glutamate-producing C. glutamicum with the highest titer and productivity of 120 g/l and 5 g/l·h, respectively. The amino acid ʟ-lysine represents the second largest share of the market (1.5 M tons/year). Becker et al. [11] reported on an efficient ʟ-lysine hyper-producer with a titer of 120 g/l, productivity of 4.0 g/l·h, and a yield of 0.55 g/g-carbon developed through synthetic metabolic engineering of C. glutamicum. In the 2000s, production expanded to other amino acids such as ʟ-arginine, ʟ- alanine, and ʟ-methionine, and further to derived valuable chemicals such as γ-aminobutyrate (GABA), which can be applied in functional foods, pharmaceuticals, or polyamide.

Alcohols and Organic Acids

Owing to its leading role as an amino acid producer, studies on C. glutamicum have focused on amino acid biosynthetic pathways. However, fermentation of alcohols such as ethanol and isobutanol under oxygen-deprived conditions has been reported [16,44,49,125,151]. Furthermore, efforts to produce organic acids such as succinate, lactate, 2-ketoglutarate, and 2-ketoisovalerate have proven that C. glutamicum can also produce valuable organic acids. The possibilities for the use of C. glutamicum in industrial applications have also been investigated. For example, there have been reports of ethanol (119 g/l), isobutanol (73 g/l), ʟ-lactate (95.6 g/l), D-lactate (120 g/l), and succinate (156 g/l) production under conditions of oxygen deprivation [16,44,49,125,151].

Polymers

There have been efforts made to produce polymers such as diamines (putrescine, cadavarine) and polyhydroxybutyrate (PHB) with engineered C. glutamicum; for example, recombinant ornithine-producing C. glutamicum has been used to produce diamine 1,4-diaminobutane (putrescine) and 1,5-diaminopentane (cadaverine) derived from ʟ-ornithine by decarboxylation [19,69,117-119]. Recombinant C. glutamicum expressing PhbCAB from Ralstonia eutropha accumulated up to 6.0 g/l polyhydroxyalkanoate (PHA) and 0.99 g/l PHB, respectively [83,86].

 

Heterologous Protein Expression in C. glutamicum

As mentioned above, C. glutamicum has been widely used as an industrial workhorse for the production of amino acids and various bio-based chemicals. Moreover, an increasing number of reports indicate C. glutamicum to have great potential as a platform microbe for heterologous protein expression (Table 6). Owing to the many intrinsic attributes of C. glutamicum, a broad range of different approaches has been used to investigate its potential as a host for the expression of heterologous proteins. There are several successful examples showing that heterologous expression in C. glutamicum enhanced the production of amino acids and other value-added chemicals, or allowed the utilization of various carbon sources via metabolic engineering approaches.

Table 6.Summary of the heterologous expression cases in C. glutamicum.

Table 6.n.q., not quantified. aMaximum conversion yield.

Intrinsic Advantages of C. glutamicum as a Protein Expression Host

C. glutamicum has intrinsic advantages as a microbial cell factory for protein production. A low level of extracellular protease activity and the presence of two native protein secretion mechanisms, the general secretory (Sec-dependent) pathway and the twin-arginine translocation (TAT-dependent) pathway have been known to enhance the secretion of homologous and heterologous proteins. Proteome analyses of C. glutamicum showed that approximately 10% of the proteins coded by wild-type C. glutamicum could be secreted [51]. Many species belonging to Corynebacterium are known to possess low protease activity unlike many other soil bacteria such as those of the Streptomyces and Bacillus genera. It has been identified that C. glutamicum has only one extracellular protease coding gene in its genome, the extracellular trypsin-like protease gene etpr (cg1243, cgR1176) [157]. To date, two well-identified independent secretion pathways have been utilized to enhance the secretion of proteins in C. glutamicum: the Sec-dependent and TAT-dependent pathways. Overexpression of components of the Sec and the TAT pathways, such as Sec(E/Y)DF and TatABC, has been suggested as a method to reduce molecular bottlenecks [63]. Moreover, the signal peptides type I and II have also been investigated to enhance secretion efficiency, such as signal sequences 29, 11, and 8 for Sec, Tat (type I), and lipoprotein (type II), respectively [142].

Heterologous Protein Expression

As shown in Table 6, heterologous expression in C. glutamicum has been reported. Amylases from other soil bacteria such as Bacillus amyloliquefaciens, Geobacillus stearothermophilus, Streptomyces bovis, and Streptomyces griseus were successfully expressed in various C. glutamicum strains [20,126,133,146,154]. Numerous cases have shown that heterologous expression enhanced the production of natural or non-natural compounds in C. glutamicum; for example, Pseudomonas taetrolens ArgR enabled the production of D-amino acids in an engineered C. glutamicum strain [127]. More recently, a synthetic Pseudomonas stutzeri ectoine gene cluster enabled the production of ectoine, a chemical chaperone that has a stabilizing effect on biological molecules in C. glutamicum [9].

 

Concluding Remarks and Outlook

Best known for its use over a period of decades in industry as a glutamate and lysine producer, C. glutamicum has been widely used in various applications and is considered as one of the most preferred microbes used for metabolic engineering. A number of approaches for enhancing secretion efficiency have been identified, such as the use of signal peptides. However, C. glutamicum has some disadvantages when compared with E. coli, such as a much lower transformation efficiency and a low number of available expression systems. To overcome these issues, a remarkable effort should be made to develop expression systems and secretion machinery.

Taken together, we still need to develop the toolbox that will allow us to gain a deeper understanding and increase production speed. However, we can surely expect further progress for C. glutamicum as a microbial value-adding producer workhorse in the field of microbial biotechnology.

References

  1. Abe S, Takayama K-I, Kinoshita S. 1967. Taxonomical studies on glutamic acid-producing bacteria. J. Gen. Appl. Microbiol. 13: 279-301. https://doi.org/10.2323/jgam.13.279
  2. Adham SA, Honrubia P, Diaz M, Fernandez-Abalos JM, Santamaria RI, Gil JA. 2001. Expression of the genes coding for the xylanase Xys1 and the cellulase Cel1 from the straw-decomposing Streptomyces halstedii JM8 cloned into the amino-acid producer Brevibacterium lactofermentum ATCC13869. Arch. Microbiol. 177: 91-97. https://doi.org/10.1007/s00203-001-0365-3
  3. An SJ, Yim SS, Jeong KJ. 2013. Development of a secretion system for the production of heterologous proteins in Corynebacterium glutamicum using the Porin B signal peptide. Protein Express. Purif. 89: 251-257. https://doi.org/10.1016/j.pep.2013.04.003
  4. Asakura Y, Kimura E, Usuda Y, Kawahara Y, Matsui K, Osumi T, Nakamatsu T. 2007. Altered metabolic flux due to deletion of odhA causes ʟ-glutamate overproduction in Corynebacterium glutamicum. Appl. Environ. Microbiol. 73: 1308-1319. https://doi.org/10.1128/AEM.01867-06
  5. Bückle-Vallant V, Krause F, Messerschmidt S, Eikmanns B. 2014. Metabolic engineering of Corynebacterium glutamicum for 2-ketoisocaproate production. Appl. Microbiol. Biotechnol. 98: 297-311. https://doi.org/10.1007/s00253-013-5310-2
  6. Bardonnet N, Blanco C. 1991. Improved vectors for transcriptional signal screening in corynebacteria. FEMS Microbiol. Lett. 68: 97-102. https://doi.org/10.1111/j.1574-6968.1991.tb04576.x
  7. Baumgart M, Unthan S, Rückert C, Sivalingam J, Grünberger A, Kalinowski J, et al. 2013. Construction of a prophage-free variant of Corynebacterium glutamicum ATCC 13032 for use as a platform strain for basic research and industrial biotechnology. Appl. Environ. Microbiol. 79: 6006-6015. https://doi.org/10.1128/AEM.01634-13
  8. Becker J, Klopprogge C, Zelder O, Heinzle E, Wittmann C. 2005. Amplified expression of fructose 1,6-bisphosphatase in Corynebacterium glutamicum increases in vivo flux through the pentose phosphate pathway and lysine production on different carbon sources. Appl. Environ. Microbiol. 71: 8587-8596. https://doi.org/10.1128/AEM.71.12.8587-8596.2005
  9. Becker J, Schafer R, Kohlstedt M, Harder BJ, Borchert NS, Stoveken N, et al. 2013. Systems metabolic engineering of Corynebacterium glutamicum for production of the chemical chaperone ectoine. Microb. Cell Fact. 12: 110. https://doi.org/10.1186/1475-2859-12-110
  10. Becker J, Wittmann C. 2012. Systems and synthetic metabolic engineering for amino acid production – the heartbeat of industrial strain development. Curr. Opin. Biotechnol. 23: 718-726. https://doi.org/10.1016/j.copbio.2011.12.025
  11. Becker J, Zelder O, Häfner S, Schröder H, Wittmann C. 2011. From zero to hero - design-based systems metabolic engineering of Corynebacterium glutamicum for ʟ-lysine production. Metab. Eng. 13: 159-168. https://doi.org/10.1016/j.ymben.2011.01.003
  12. Bendt AK, Burkovski A, Schaffer S, Bott M, Farwick M, Hermann T. 2003. Towards a phosphoproteome map of Corynebacterium glutamicum. Proteomics 3: 1637-1646. https://doi.org/10.1002/pmic.200300494
  13. Billman-Jacobe H, Hodgson AL, Lightowlers M, Wood PR, Radford AJ. 1994. Expression of ovine gamma interferon in Escherichia coli and Corynebacterium glutamicum. Appl. Environ. Microbiol. 60: 1641-1645.
  14. Billman-Jacobe H, Wang L, Kortt A, Stewart D, Radford A. 1995. Expression and secretion of heterologous proteases by Corynebacterium glutamicum. Appl. Environ. Microbiol. 61: 1610-1613.
  15. Blombach B, Buchholz J, Busche T, Kalinowski J, Takors R. 2013. Impact of different CO2/HCO3- levels on metabolism and regulation in Corynebacterium glutamicum. J. Biotechnol. 168: 331-340. https://doi.org/10.1016/j.jbiotec.2013.10.005
  16. Blombach B, Riester T, Wieschalka S, Ziert C, Youn JW, Wendisch VF, Eikmanns BJ. 2011. Corynebacterium glutamicum tailored for efficient isobutanol production. Appl. Environ. Microbiol. 77: 3300-3310. https://doi.org/10.1128/AEM.02972-10
  17. Brabetz W, Liebl W, Schleifer KH. 1991. Studies on the utilization of lactose by Corynebacterium glutamicum, bearing the lactose operon of Escherichia coli. Arch. Microbiol. 155: 607-612. https://doi.org/10.1007/BF00245357
  18. Buchholz J, Schwentner A, Brunnenkan B, Gabris C, Grimm S, Gerstmeir R, et al. 2013. Platform engineering of Corynebacterium glutamicum with reduced pyruvate dehydrogenase complex activity for improved production of ʟ-lysine, ʟ-valine, and 2-ketoisovalerate. Appl. Environ. Microbiol. 79: 5566-5575. https://doi.org/10.1128/AEM.01741-13
  19. Buschke N, Schäfer R, Becker J, Wittmann C. 2013. Metabolic engineering of industrial platform microorganisms for biorefinery applications – optimization of substrate spectrum and process robustness by rational and evolutive strategies. Bioresour. Technol. 135: 544-554. https://doi.org/10.1016/j.biortech.2012.11.047
  20. Cadenas RF, Gil JA, Martin JF. 1992. Expression of Streptomyces genes encoding extracellular enzymes in Brevibacterium lactofermentum: secretion proceeds by removal of the same leader peptide as in Streptomyces lividans. Appl. Microbiol. Biotechnol. 38: 362-369. https://doi.org/10.1007/BF00170087
  21. Choi JW, Yim SS, Lee SH, Kang TJ, Park SJ, Jeong KJ. 2015. Enhanced production of gamma-aminobutyrate (GABA) in recombinant Corynebacterium glutamicum by expressing glutamate decarboxylase active in expanded pH range. Microb. Cell Fact. 14: 21. https://doi.org/10.1186/s12934-015-0205-9
  22. Cramer A, Gerstmeir R, Schaffer S, Bott M, Eikmanns BJ. 2006. Identification of RamA, a novel LuxR-type transcriptional regulator of genes involved in acetate metabolism of Corynebacterium glutamicum. J. Bacteriol. 188: 2554-2567. https://doi.org/10.1128/JB.188.7.2554-2567.2006
  23. Date M, Itaya H, Matsui H, Kikuchi Y. 2006. Secretion of human epidermal growth factor by Corynebacterium glutamicum. Lett. Appl. Microbiol. 42: 66-70. https://doi.org/10.1111/j.1472-765X.2005.01802.x
  24. Date M, Yokoyama K, Umezawa Y, Matsui H, Kikuchi Y. 2003. Production of native-type Streptoverticillium mobaraense transglutaminase in Corynebacterium glutamicum. Appl. Environ. Microbiol. 69: 3011-3014. https://doi.org/10.1128/AEM.69.5.3011-3014.2003
  25. Date M, Yokoyama K, Umezawa Y, Matsui H, Kikuchi Y. 2004. High level expression of Streptomyces mobaraensis transglutaminase in Corynebacterium glutamicum using a chimeric pro-region from Streptomyces cinnamoneus transglutaminase. J. Biotechnol. 110: 219-226. https://doi.org/10.1016/j.jbiotec.2004.02.011
  26. Dusch N, Pühler A, Kalinowski J. 1999. Expression of the Corynebacterium glutamicum panD gene encoding ʟ-aspartate-decarboxylase leads to pantothenate overproduction in Escherichia coli. Appl. Environ. Microbiol. 65: 1530-1539.
  27. Eikmanns BJ, Kleinertz E, Liebl W, Sahm H. 1991. A family of Corynebacterium glutamicum/Escherichia coli shuttle vectors for cloning, controlled gene expression, and promoter probing. Gene 102: 93-98. https://doi.org/10.1016/0378-1119(91)90545-M
  28. Gerstmeir R, Cramer A, Dangel P, Schaffer S, Eikmanns BJ. 2004. RamB, a novel transcriptional regulator of genes involved in acetate metabolism of Corynebacterium glutamicum. J. Bacteriol. 186: 2798-2809. https://doi.org/10.1128/JB.186.9.2798-2809.2004
  29. Hänßler E, Müller T, Palumbo K, Patek M, Brocker M, Krämer R, Burkovski A. 2009. A game with many players: control of gdh transcription in Corynebacterium glutamicum. J. Biotechnol. 142: 114-122. https://doi.org/10.1016/j.jbiotec.2009.04.007
  30. Hüser AT, Chassagnole C, Lindley ND, Merkamm M, Guyonvarch A, Elišáková V, et al. 2005. Rational design of a Corynebacterium glutamicum pantothenate production strain and its characterization by metabolic flux analysis and genome-wide transcriptional profiling. Appl. Environ. Microbiol. 71: 3255-3268. https://doi.org/10.1128/AEM.71.6.3255-3268.2005
  31. Hao N, Mu Jr, Hu N, Xu S, Yan M, Li Y, et al. 2015. Improvement of ʟ-citrulline production in Corynebacterium glutamicum by ornithine acetyltransferase. J. Ind. Microbiol. Biotechnol. 42: 307-313. https://doi.org/10.1007/s10295-014-1561-x
  32. Hartbrich A, Schmitz G, Weuster-Botz D, de Graaf AA, Wandrey C. 1996. Development and application of a membrane cyclone reactor for in vivo NMR spectroscopy with high microbial cell densities. Biotechnol. Bioeng. 51: 624-635.
  33. Hasegawa S, Suda M, Uematsu K, Natsuma Y, Hiraga K, Jojima T, et al. 2013. Engineering of Corynebacterium glutamicum for high-yield ʟ-valine production under oxygen deprivation conditions. Appl. Environ. Microbiol. 79: 1250-1257. https://doi.org/10.1128/AEM.02806-12
  34. Hasegawa S, Uematsu K, Natsuma Y, Suda M, Hiraga K, Jojima T, et al. 2012. Improvement of the redox balance increases ʟ-valine production by Corynebacterium glutamicum under oxygen deprivation conditions. Appl. Environ. Microbiol. 78: 865-875. https://doi.org/10.1128/AEM.07056-11
  35. Hayashi M, Mizoguchi H, Shiraishi N, Obayashi M, Nakagawa S, Imai J-I, et al. 2002. Transcriptome analysis of acetate metabolism in Corynebacterium glutamicum using a newly developed metabolic array. Biosci. Biotechnol. Biochem. 66: 1337-1344. https://doi.org/10.1271/bbb.66.1337
  36. Haynes JA, Britz ML. 1990. The effect of growth conditions of Corynebacterium glutamicum on the transformation frequency obtained by electroporation. J. Gen. Microbiol. 136: 255-263. https://doi.org/10.1099/00221287-136-2-255
  37. Hermann T, Pfefferle W, Baumann C, Busker E, Schaffer S, Bott M, et al. 2001. Proteome analysis of Corynebacterium glutamicum. Electrophoresis 22: 1712-1723. https://doi.org/10.1002/1522-2683(200105)22:9<1712::AID-ELPS1712>3.0.CO;2-G
  38. Holatko J, Elisakova V, Prouza M, Sobotka M, Nesvera J, Patek M. 2009. Metabolic engineering of the ʟ-valine biosynthesis pathway in Corynebacterium glutamicum using promoter activity modulation. J. Biotechnol. 139: 203-210. https://doi.org/10.1016/j.jbiotec.2008.12.005
  39. Ikeda M. 2003. Amino acid production processes. Adv. Biochem. Eng. Biotechnol. 79: 1-35.
  40. Ikeda M, Katsumata R. 1992. Metabolic engineering to produce tyrosine or phenylalanine in a tryptophan-producing Corynebacterium glutamicum strain. Appl. Environ. Microbiol. 58: 781-785.
  41. Ikeda M, Katsumata R. 1998. A novel system with positive selection for the chromosomal integration of replicative plasmid DNA in Corynebacterium glutamicum. Microbiology 144: 1863-1868. https://doi.org/10.1099/00221287-144-7-1863
  42. Ikeda M, Katsumata R. 1999. Hyperproduction of tryptophan by Corynebacterium glutamicum with the modified pentose phosphate pathway. Appl. Environ. Microbiol. 65: 2497-2502.
  43. Ikeda M, Nakagawa S. 2003. The Corynebacterium glutamicum genome: features and impacts on biotechnological processes. Appl. Microbiol. Biotechnol. 62: 99-109. https://doi.org/10.1007/s00253-003-1328-1
  44. Inui M, Murakami S, Okino S, Kawaguchi H, Vertès AA, Yukawa H. 2004. Metabolic analysis of Corynebacterium glutamicum during lactate and succinate productions under oxygen deprivation conditions. J. Mol. Microbiol. Biotechnol. 7: 182-196. https://doi.org/10.1159/000079827
  45. Jakoby M, Ngouoto-Nkili C-E, Burkovski A. 1999. Construction and application of new Corynebacterium glutamicum vectors. Biotechnol. Tech. 13: 437-441. https://doi.org/10.1023/A:1008968419217
  46. Jensen JV, Wendisch VF. 2013. Ornithine cyclodeaminase-based proline production by Corynebacterium glutamicum. Microb. Cell Fact. 12: 63. https://doi.org/10.1186/1475-2859-12-63
  47. Jo JH, Seol HY, Lee YB, Kim MH, Hyun HH, Lee HH. 2012. Disruption of genes for the enhanced biosynthesis of alpha-ketoglutarate in Corynebacterium glutamicum. Can. J. Microbiol. 58: 278-286. https://doi.org/10.1139/w11-132
  48. Jojima T, Fujii M, Mori E, Inui M, Yukawa H. 2010. Engineering of sugar metabolism of Corynebacterium glutamicum for production of amino acid ʟ-alanine under oxygen deprivation. Appl. Microbiol. Biotechnol. 87: 159-165. https://doi.org/10.1007/s00253-010-2493-7
  49. Jojima T, Noburyu R, Sasaki M, Tajima T, Suda M, Yukawa H, Inui M. 2015. Metabolic engineering for improved production of ethanol by Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 99: 1165-1172. https://doi.org/10.1007/s00253-014-6223-4
  50. Kalinowski J, Bathe B, Bartels D, Bischoff N, Bott M, Burkovski A, et al. 2003. The comp lete Corynebacterium glutamicum ATCC 13032 genome sequence and its impact on the production of ʟ-aspartate-derived amino acids and vitamins. J. Biotechnol. 104: 5-25. https://doi.org/10.1016/S0168-1656(03)00154-8
  51. Kalinowski J, Wolters D, Poetsch A. 2008. Proteomics of Corynebacterium glutamicum and other corynebacteria, pp. 55-78. In Burkovski A (ed.). Corynebacteria: Genomics and Molecular Biology. Caister Academic Press, Norfolk, UK.
  52. Kang M-K, Lee J, Um Y, Lee T, Bott M, Park S, Woo H. 2014. Synthetic biology platform of CoryneBrick vectors for gene expression in Corynebacterium glutamicum and its application to xylose utilization. Appl. Microbiol. Biotechnol. 98: 5991-6002. https://doi.org/10.1007/s00253-014-5714-7
  53. Kang MK, Eom JH, Kim Y, Um Y, Woo HM. 2014. Biosynthesis of pinene from glucose using metabolically-engineered Corynebacterium glutamicum. Biotechnol. Lett. 36: 2069-2077. https://doi.org/10.1007/s10529-014-1578-2
  54. Kang MS, Han SS, Kim MY, Kim BY, Huh JP, Kim HS, Lee JH. 2014. High-level expression in Corynebacterium glutamicum of nitrile hydratase from Rhodococcus rhodochrous for acrylamide production. Appl. Microbiol. Biotechnol. 98: 4379-4387. https://doi.org/10.1007/s00253-014-5544-7
  55. Kass F, Hariskos I, Michel A, Brandt HJ, Spann R, Junne S, et al. 2014. Assessment of robustness against dissolved oxygen/substrate oscillations for C. glutamicum DM1933 in two-compartment bioreactor. Bioproc. Biosyst. Eng. 37: 1151-1162. https://doi.org/10.1007/s00449-013-1086-0
  56. Katsumata R MT, Kikuchi Y, Kino K. 1986. Threonine production by the lysine producing strain of Corynebacterium glutamicum with amplified threonine biosynthetic operon, pp. 217-226. In Alacevic M, Hranueli D, Toman Z (eds.). Genetics of Industrial Microorganisms. Ognjen Prica Printing Works.
  57. Katsumata R, Ozaki A, Oka T, Furuya A. 1984. Protoplast transformation of glutamate-producing bacteria with plasmid DNA. J. Bacteriol. 159: 306-311.
  58. Kawaguchi H, Sasaki M, Vertès AA, Inui M, Yukawa H. 2009. Identification and functional analysis of the gene cluster for ʟ-arabinose utilization in Corynebacterium glutamicum. Appl. Environ. Microbiol. 75: 3419-3429. https://doi.org/10.1128/AEM.02912-08
  59. Kawaguchi H, Vertès AA, Okino S, Inui M, Yukawa H. 2006. Engineering of a xylose metabolic pathway in Corynebacterium glutamicum. Appl. Environ. Microbiol. 72: 3418-3428. https://doi.org/10.1128/AEM.72.5.3418-3428.2006
  60. Kelle R, Hermann T, Weuster-Botz D, Eggeling L, Krämer R, Wandrey C. 1996. Glucose-controlled l-isoleucine fed-batch production with recombinant strains of Corynebacterium glutamicum. J. Biotechnol. 50: 123-136. https://doi.org/10.1016/0168-1656(96)01554-4
  61. Kikuchi Y, Date M, Yokoyama K, Umezawa Y, Matsui H. 2003. Secretion of active-form Streptoverticillium mobaraense transglutaminase by Corynebacterium glutamicum: processing of the pro-transglutaminase by a cosecreted subtilisin-like protease from Streptomyces albogriseolus. Appl. Environ. Microbiol. 69: 358-366. https://doi.org/10.1128/AEM.69.1.358-366.2003
  62. Kikuchi Y, Itaya H, Date M, Matsui K, Wu LF. 2008. Production of Chryseobacterium proteolyticum protein-glutaminase using the twin-arginine translocation pathway in Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 78: 67-74. https://doi.org/10.1007/s00253-007-1283-3
  63. Kikuchi Y, Itaya H, Date M, Matsui K, Wu LF. 2009. TatABC overexpression improves Corynebacterium glutamicum Tat-dependent protein secretion. Appl. Environ. Microbiol. 75: 603-607. https://doi.org/10.1128/AEM.01874-08
  64. Kim HI, Nam JY, Cho JY, Lee CS, Park YJ. 2013. Nextgeneration sequencing-based transcriptome analysis of ʟ-lysine-producing Corynebacterium glutamicum ATCC 21300 strain. J. Microbiol. 51: 877-880. https://doi.org/10.1007/s12275-013-3236-0
  65. Kim SY, Lee J, Lee SY. 2015. Metabolic engineering of Corynebacterium glutamicum for the production of ʟ-ornithine. Biotechnol. Bioeng. 112: 416-421. https://doi.org/10.1002/bit.25440
  66. Kind S, Jeong WK, Schröder H, Wittmann C. 2010. Systems-wide metabolic pathway engineering in Corynebacterium glutamicum for bio-based production of diaminopentane. Metab. Eng. 12: 341-351. https://doi.org/10.1016/j.ymben.2010.03.005
  67. Kind S, Jeong WK, Schröder H, Zelder O, Wittmann C. 2010. Identification and elimination of the competing N-acetyldiaminopentane pathway for improved production of diaminopentane by Corynebacterium glutamicum. Appl. Environ. Microbiol. 76: 5175-5180. https://doi.org/10.1128/AEM.00834-10
  68. Kind S, Kreye S, Wittmann C. 2011. Metabolic engineering of cellular transport for overproduction of the platform chemical 1,5-diaminopentane in Corynebacterium glutamicum. Metab. Eng. 13: 617-627. https://doi.org/10.1016/j.ymben.2011.07.006
  69. Kind S, Neubauer S, Becker J, Yamamoto M, Völkert M, Abendroth GV, et al. 2014. From zero to hero – production of bio-based nylon from renewable resources using engineered Corynebacterium glutamicum. Metab. Eng. 25: 113-123. https://doi.org/10.1016/j.ymben.2014.05.007
  70. Kinoshita S, Udaka S, Shimono M. 1957. Studies on the amino acid fermentation. Part 1. Production of ʟ-glutamic acid by various microorganisms. J. Gen. Appl. Microbiol. 3: 193-205. https://doi.org/10.2323/jgam.3.193
  71. Knoppova M, Phensaijai M, Vesely M, Zemanova M, Nesvera J, Patek M. 2007. Plasmid vectors for testing in vivo promoter activities in Corynebacterium glutamicum and Rhodococcus erythropolis. Curr. Microbiol. 55: 234-239. https://doi.org/10.1007/s00284-007-0106-1
  72. Kortmann M, Kuhl V, Klaffl S, Bott M. 2015. A chromosomally encoded T7 RNA polymerase-dependent gene expression system for Corynebacterium glutamicum: construction and comparative evaluation at the single-cell level. Microb. Biotechnol. 8: 253-265. https://doi.org/10.1111/1751-7915.12236
  73. Kotrba P, Inui M, Yukawa H. 2001. The ptsI gene encoding enzyme I of the phosphotransferase system of Corynebacterium glutamicum. Biochem. Biophys. Res. Commun. 289: 1307-1313. https://doi.org/10.1006/bbrc.2001.6116
  74. Kromer JO, Sorgenfrei O, Klopprogge K, Heinzle E, Wittmann C. 2004. In-depth profiling of lysine-producing Corynebacterium glutamicum: by combined analysis of the transcriptome, metabolome, and fluxome. J. Bacteriol. 186: 1769-1784. https://doi.org/10.1128/JB.186.6.1769-1784.2004
  75. Lee BH, Lee SB, Kim HS, Jeong KJ, Park JY, Park KM, Lee JW. 2015. Whole cell bioconversion of ricinoleic acid to 12-ketooleic acid by recombinant Corynebacterium glutamicum-based biocatalyst. J. Microbiol. Biotechnol. 25: 452-458. https://doi.org/10.4014/jmb.1501.01001
  76. Lee J. 2014. Development and characterization of expression vectors for Corynebacterium glutamicum. J. Microbiol. Biotechnol. 24: 70-79. https://doi.org/10.4014/jmb.1310.10032
  77. Lee JY, Choy HE, Lee JH, Kim GJ. 2015. Generation of minicells from an endotoxin-free gram-positive strain Corynebacterium glutamicum. J. Microbiol. Biotechnol. 25: 554-558. https://doi.org/10.4014/jmb.1408.08037
  78. Lee JY, Seo J, Kim ES, Lee HS, Kim P. 2013. Adaptive evolution of Corynebacterium glutamicum resistant to oxidative stress and its global gene expression profiling. Biotechnol. Lett. 35: 709-717. https://doi.org/10.1007/s10529-012-1135-9
  79. Lee SK, Keasling JD. 2006. A Salmonella-based, propionateinducible, expression system for Salmonella enterica. Gene 377: 6-11. https://doi.org/10.1016/j.gene.2006.02.013
  80. Liebl W, Ehrmann M, Ludwig W, KH. S. 1991. Transfer of Brevibacterium divaricatum DSM 20297T, “Brevibacterium flavum” DSM 20411, “Brevibacterium lactofermentum” DSM 20412 and DSM 1412, and Corynebacterium lilium DSM 20137T to Corynebacterium glutamicum and their distinction by rRNA gene restriction patterns. Int. J. Syst. Evol. Microbiol. 41: 255-260.
  81. Liebl W, Sinskey AJ, Schleifer KH. 1992. Expression, secretion, and processing of staphylococcal nuclease by Corynebacterium glutamicum. J. Bacteriol. 174: 1854-1861. https://doi.org/10.1128/jb.174.6.1854-1861.1992
  82. Liu Q, Ouyang S-P, Kim J, Chen G-Q. 2007. The impact of PHB accumulation on ʟ-glutamate production by recombinant Corynebacterium glutamicum. J. Biotechnol. 132: 273-279. https://doi.org/10.1016/j.jbiotec.2007.03.014
  83. Liu Q, Zhang J, Wei X-X, Ouyang S-P, Wu Q, Chen G-Q. 2008. Microbial production of ʟ-glutamate and ʟ-glutamine by recombinant Corynebacterium glutamicum harboring Vitreoscilla hemoglobin gene vgb. Appl. Microbiol. Biotechnol. 77: 1297-1304. https://doi.org/10.1007/s00253-007-1254-8
  84. Loos A, Glanemann C, Willis LB, O’Brien XM, Lessard PA, Gerstmeir R, et al. 2001. Development and validation of Corynebacterium DNA microarrays. Appl. Microbiol. Biotechnol. 67: 2310-2318.
  85. Matsuda Y, Itaya H, Kitahara Y, Theresia NM, Kutukova EA, Yomantas YAV, et al. 2014. Double mutation of cell wall proteins CspB and PBP1a increases secretion of the antibody Fab fragment from Corynebacterium glutamicum. Microb. Cell Fact. 13: 56. https://doi.org/10.1186/1475-2859-13-56
  86. Matsumoto K, Kitagawa K, Jo S-J, Song Y, Taguchi S. 2011. Production of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) in recombinant Corynebacterium glutamicum using propionate as a precursor. J. Biotechnol. 152: 144-146. https://doi.org/10.1016/j.jbiotec.2010.07.031
  87. Mentz A, Neshat A, Pfeifer-Sancar K, Puhler A, Ruckert C, Kalinowski J. 2013. Comprehensive discovery and characterization of small RNAs in Corynebacterium glutamicum ATCC 13032. BMC Genomics 14: 714. https://doi.org/10.1186/1471-2164-14-714
  88. Mizukami T, Hamu A, Ikeda M, Oka T, Katsumata R. 1994. Cloning of the ATP phosphoribosyl transferase gene of Corynebacterium glutamicum and application of the gene to ʟ-histidine production. Biosci. Biotechnol. Biochem. 58: 635-638. https://doi.org/10.1271/bbb.58.635
  89. Muffler A, Bettermann S, Haushalter M, Hörlein A, Neveling U, Schramm M, Sorgenfrei O. 2002. Genome-wide transcription profiling of Corynebacterium glutamicum after heat shock and during growth on acetate and glucose. J. Biotechnol. 98: 255-268. https://doi.org/10.1016/S0168-1656(02)00136-0
  90. Nakamura Y, Nishio Y, Ikeo K, Gojobori T. 2003. The genome stability in Corynebacterium species due to lack of the recombinational repair system. Gene 317: 149-155. https://doi.org/10.1016/S0378-1119(03)00653-X
  91. Nakayama K, Tanaka H, Hagino H, Kinoshita S. 1966. Studies on lysine fermentation: Part V. Concerted feedback inhibition of aspartokinase and the absence of lysine inhibition on aspartic semialdehyde-pyruvate condensation in Micrococcus glutamicus. Agric. Biol. Chem. 30: 611-616.
  92. Neshat A, Mentz A, Ruckert C, Kalinowski J. 2014. Transcriptome sequencing revealed the transcriptional organization at ribosome-mediated attenuation sites in Corynebacterium glutamicum and identified a novel attenuator involved in aromatic amino acid biosynthesis. J. Biotechnol. 190: 55-63. https://doi.org/10.1016/j.jbiotec.2014.05.033
  93. Oberreuter H, Charzinski J, Scherer S. 2002. Intraspecific diversity of Brevibacterium linens, Corynebacterium glutamicum and Rhodococcus erythropolis based on partial 16S rDNA sequence analysis and Fourier-transform infrared (FT-IR) spectroscopy. Microbiology 148: 1523-1532. https://doi.org/10.1099/00221287-148-5-1523
  94. Oh Y, Choi J, Kim E, Song B, Jeong K, Park K, et al. 2015. Construction of synthetic promoter-based expression cassettes for the production of cadaverine in recombinant Corynebacterium glutamicum. Appl. Biochem. Biotechnol. 176: 1-11. https://doi.org/10.1007/s12010-015-1555-9
  95. Okibe N, Suzuki N, Inui M, Yukawa H. 2010. Isolation, evaluation and use of two strong, carbon source-inducible promoters from Corynebacterium glutamicum. Lett. Appl. Microbiol. 50: 173-180. https://doi.org/10.1111/j.1472-765X.2009.02776.x
  96. Okino S, Inui M, Yukawa H. 2005. Production of organic acids by Corynebacterium glutamicum under oxygen deprivation. Appl. Microbiol. Biotechnol. 68: 475-480. https://doi.org/10.1007/s00253-005-1900-y
  97. Okino S, Suda M, Fujikura K, Inui M, Yukawa H. 2008. Production of D-lactic acid by Corynebacterium glutamicum under oxygen deprivation. Appl. Microbiol. Biotechnol. 78: 449-454. https://doi.org/10.1007/s00253-007-1336-7
  98. Otten A, Brocker M, Bott M. 2015. Metabolic engineering of Corynebacterium glutamicum for the production of itaconate. Metab. Eng. 30: 156-165. https://doi.org/10.1016/j.ymben.2015.06.003
  99. Pátek M, Holátko J, Busche T, Kalinowski J, Nešvera J. 2013. Corynebacterium glutamicum promoters: a practical approach. Microb. Biotechnol. 6: 103-117. https://doi.org/10.1111/1751-7915.12019
  100. Paradis FW, Warren RA, Kilburn DG, Miller RC Jr. 1987. The expression of Cellulomonas fimi cellulase genes in Brevibacterium lactofermentum. Gene 61: 199-206. https://doi.org/10.1016/0378-1119(87)90114-4
  101. Park S, Lee S, Park I, Choi J, Jeong W, Kim Y, Lee H. 2004. Isolation and characterization of transcriptional elements from Corynebacterium glutamicum. J. Microbiol. Biotechnol. 14: 789-795.
  102. Park SD, Lee JY, Sim SY, Kim Y, Lee HS. 2007. Characteristics of methionine production by an engineered Corynebacterium glutamicum strain. Metab. Eng. 9: 327-336. https://doi.org/10.1016/j.ymben.2007.05.001
  103. Park SH, Kim HU, Kim TY, Park JS, Kim S-S, Lee SY. 2014. Metabolic engineering of Corynebacterium glutamicum for ʟ-arginine production. Nat. Commun. 5: 4618.
  104. Patek M, Nesvera J, Guyonvarch A, Reyes O, Leblon G. 2003. Promoters of Corynebacterium glutamicum. J. Biotechnol. 104: 311-323. https://doi.org/10.1016/S0168-1656(03)00155-X
  105. Peters-Wendisch PG, Schiel B, Wendisch VF, Katsoulidis E, Mockel B, Sahm H, Eikmanns BJ. 2001. Pyruvate carboxylase is a major bottleneck for glutamate and lysine production by Corynebacterium glutamicum. J. Mol. Microbiol. Biotechnol. 3: 295-300.
  106. Peyret JL, Bayan N, Joliff G, Gulik-Krzywicki T, Mathieu L, Schechter E, Leblon G. 1993. Characterization of the cspB gene encoding PS2, an ordered surface-layer protein in Corynebacterium glutamicum. Mol. Microbiol. 9: 97-109. https://doi.org/10.1111/j.1365-2958.1993.tb01672.x
  107. Pfeifer-Sancar K, Mentz A, Rückert C, Kalinowski J. 2013. Comprehensive analysis of the Corynebacterium glutamicum transcriptome using an improved RNAseq technique. BMC Genomics 14: 888. https://doi.org/10.1186/1471-2164-14-888
  108. Plassmeier JK, Busche T, Molck S, Persicke M, Puhler A, Ruckert C, Kalinowski J. 2013. A propionate-inducible expression system based on the Corynebacterium glutamicum prpD2 promoter and PrpR activator and its application for the redirection of amino acid biosynthesis pathways. J. Biotechnol. 163: 225-232. https://doi.org/10.1016/j.jbiotec.2012.08.009
  109. Ravasi P, Peiru S, Gramajo H, Menzella HG. 2012. Design and testing of a synthetic biology framework for genetic engineering of Corynebacterium glutamicum. Microb. Cell Fact. 11: 147. https://doi.org/10.1186/1475-2859-11-147
  110. Rytter J, Helmark S, Chen J, Lezyk M, Solem C, Jensen P. 2014. Synthetic promoter libraries for Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 98: 2617-2623. https://doi.org/10.1007/s00253-013-5481-x
  111. Salim K, Haedens V, Content J, Leblon G, Huygen K. 1997. Heterologous expression of the Mycobacterium tuberculosis gene encoding antigen 85A in Corynebacterium glutamicum. Appl. Environ. Microbiol. 63: 4392-4400.
  112. Santamaria R, Gil J, Mesas JM, Martin JF. 1984. Characterization of an endogenous plasmid and development of cloning vectors and a transformation system in Brevibacterium lactofermentum. J. Gen Microbiol. 130: 2237-2246.
  113. Sasaki M, Jojima T, Inui M, Yukawa H. 2008. Simultaneous utilization of D-cellobiose, D-glucose, and D-xylose by recombinant Corynebacterium glutamicum under oxygen-deprived conditions. Appl. Microbiol. Biotechnol. 81: 691-699. https://doi.org/10.1007/s00253-008-1703-z
  114. Schäfer A, Schwarzer A, Kalinowski J, Pühler A. 1994. Cloning and characterization of a DNA region encoding a stress-sensitive restriction system from Corynebacterium glutamicum ATCC 13032 and analysis of its role in intergeneric conjugation with Escherichia coli. J. Bacteriol. 176: 7309-7319. https://doi.org/10.1128/jb.176.23.7309-7319.1994
  115. Schaffer S, Weil B, Nguyen VD, Dongmann G, Gunther K, Nickolaus M, et al. 2001. A high-resolution reference map for cytoplasmic and membrane-associated proteins of Corynebacterium glutamicum. Electrophoresis 22: 4404-4422. https://doi.org/10.1002/1522-2683(200112)22:20<4404::AID-ELPS4404>3.0.CO;2-2
  116. Scheele S, Oertel D, Bongaerts J, Evers S, Hellmuth H, Maurer K-H, et al. 2013. Secretory production of an FAD cofactor-containing cytosolic enzyme (sorbitol–xylitol oxidase from Streptomyces coelicolor) using the twin-arginine translocation (Tat) pathway of Corynebacterium glutamicum. Microb. Biotechnol. 6: 202-206. https://doi.org/10.1111/1751-7915.12005
  117. Schneider J, Eberhardt D, Wendisch VF. 2012. Improving putrescine production by Corynebacterium glutamicum by fine-tuning ornithine transcarbamoylase activity using a plasmid addiction system. Appl. Microbiol. Biotechnol. 95: 169-178. https://doi.org/10.1007/s00253-012-3956-9
  118. Schneider J, Niermann K, Wendisch VF. 2011. Production of the amino acids ʟ-glutamate, ʟ-lysine, ʟ-ornithine and ʟ-arginine from arabinose by recombinant Corynebacterium glutamicum. J. Biotechnol. 154: 191-198. https://doi.org/10.1016/j.jbiotec.2010.07.009
  119. Schneider J, Wendisch V. 2010. Putrescine production by engineered Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 88: 859-868. https://doi.org/10.1007/s00253-010-2778-x
  120. Schwarzer A, Puhler A. 1991. Manipulation of Corynebacterium glutamicum by gene disruption and replacement. Biotechnology (NY) 9: 84-87. https://doi.org/10.1038/nbt0191-84
  121. Shi F, Jiang J, Li Y, Li Y, Xie Y. 2013. Enhancement of γ-aminobutyric acid production in recombinant Corynebacterium glutamicum by co-expressing two glutamate decarboxylase genes from Lactobacillus brevis. J. Ind. Microbiol. Biotechnol. 40: 1285-1296. https://doi.org/10.1007/s10295-013-1316-0
  122. Shi F, Li Y. 2011. Synthesis of γ-aminobutyric acid by expressing Lactobacillus brevis-derived glutamate decarboxylase in the Corynebacterium glutamicum strain ATCC 13032. Biotechnol. Lett. 33: 2469-2474. https://doi.org/10.1007/s10529-011-0723-4
  123. Siebert D, Wendisch VF. 2015. Metabolic pathway engineering for production of 1,2-propanediol and ʟ-propanol by Corynebacterium glutamicum. Biotechnol. Biofuels 8: 91. https://doi.org/10.1186/s13068-015-0269-0
  124. Silberbach M, Schäfer M, Hüser AT, Kalinowski J, Pühler A, Krämer R, Burkovski A. 2005. Adaptation of Corynebacterium glutamicum to ammonium limitation: a global analysis using transcriptome and proteome techniques. Appl. Environ. Microbiol. 71: 2391-2402. https://doi.org/10.1128/AEM.71.5.2391-2402.2005
  125. Smith KM, Cho KM, Liao JC. 2010. Engineering Corynebacterium glutamicum for isobutanol production. Appl. Microbiol. Biotechnol. 87: 1045-1055. https://doi.org/10.1007/s00253-010-2522-6
  126. Smith MD, Flickinger JL, Lineberger DW, Schmidt B. 1986. Protoplast transformation in coryneform bacteria and introduction of an alpha-amylase gene from Bacillus amyloliquefaciens into Brevibacterium lactofermentum. Appl. Environ. Microbiol. 51: 634-639.
  127. Stäbler N, Oikawa T, Bott M, Eggeling L. 2011. Corynebacterium glutamicum as a host for synthesis and export of D-amino acids. J. Bacteriol. 193: 1702-1709. https://doi.org/10.1128/JB.01295-10
  128. Stansen C, Uy D, Delaunay S, Eggeling L, Goergen JL, Wendisch VF. 2005. Characterization of a Corynebacterium glutamicum lactate utilization operon induced during temperature-triggered glutamate production. Appl. Environ. Microbiol. 71: 5920-5928. https://doi.org/10.1128/AEM.71.10.5920-5928.2005
  129. Suzuki N, Nonaka H, Tsuge Y, Inui M, Yukawa H. 2005. New multiple-deletion method for the Corynebacterium glutamicum genome, using a mutant lox sequence. Appl. Environ. Microbiol. 71: 8472-8480. https://doi.org/10.1128/AEM.71.12.8472-8480.2005
  130. Suzuki N, Nonaka H, Tsuge Y, Okayama S, Inui M, Yukawa H. 2005. Multiple large segment deletion method for Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 69: 151-161. https://doi.org/10.1007/s00253-005-1976-4
  131. Suzuki N, Okayama S, Nonaka H, Tsuge Y, Inui M, Yukawa H. 2005. Large-scale engineering of the Corynebacterium glutamicum genome. Appl. Environ. Microbiol. 71: 3369-3372. https://doi.org/10.1128/AEM.71.6.3369-3372.2005
  132. Suzuki N, Tsuge Y, Inui M, Yukawa H. 2005. Cre/loxP-mediated deletion system for large genome rearrangements in Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 67: 225-233. https://doi.org/10.1007/s00253-004-1772-6
  133. Suzuki N, Watanabe K, Okibe N, Tsuchida Y, Inui M, Yukawa H. 2009. Identification of new secreted proteins and secretion of heterologous amylase by C. glutamicum. Appl. Microbiol. Biotechnol. 82: 491-500. https://doi.org/10.1007/s00253-008-1786-6
  134. Takeno S, Takasaki M, Urabayashi A, Mimura A, Muramatsu T, Mitsuhashi S, Ikeda M. 2013. Development of fatty acid-producing Corynebacterium glutamicum strains. Appl. Environ. Microbiol. 79: 6776-6783. https://doi.org/10.1128/AEM.02003-13
  135. Tateno T, Fukuda H, Kondo A. 2007. Direct production of ʟ-lysine from raw corn starch by Corynebacterium glutamicum secreting Streptococcus bovis alpha-amylase using cspB promoter and signal sequence. Appl. Microbiol. Biotechnol. 77: 533-541. https://doi.org/10.1007/s00253-007-1191-6
  136. Tsuchiya M, Morinaga Y. 1988. Genetic control systems of Escherichia coli can confer inducible expression of cloned genes in coryneform bacteria. Nat. Biotechnol. 6: 428-430. https://doi.org/10.1038/nbt0488-428
  137. Udaka S. 1960. Screening method for microorganisms accumulating metabolites and its use in the isolation of Micrococcus glutamicus. J. Bacteriol. 79: 754-755.
  138. Unthan S, Baumgart M, Radek A, Herbst M, Siebert D, Brühl N, et al. 2015. Chassis organism from Corynebacterium glutamicum – a top-down approach to identify and delete irrelevant gene clusters. Biotechnol. J. 10: 290-301. https://doi.org/10.1002/biot.201400041
  139. van der Rest ME, Lange C, Molenaar D. 1999. A heat shock following electroporation induces highly efficient transformation of Corynebacterium glutamicum with xenogeneic plasmid DNA. Appl. Microbiol. Biotechnol. 52: 541-545. https://doi.org/10.1007/s002530051557
  140. Vasicová P, Abrhámová Z, Nesvera J, Pátek M, Sahm H, Eikmanns B. 1998. Integrative and autonomously replicating vectors for analysis of promoters in Corynebacterium glutamicum. Biotechnol. Tech. 12: 743-746. https://doi.org/10.1023/A:1008827609914
  141. Vasicova P, Patek M, Nesvera J, Sahm H, Eikmanns B. 1999. Analysis of the Corynebacterium glutamicum dapA promoter. J. Bacteriol. 181: 6188-6191.
  142. Vertès AA. 2013. Protein secretion systems of Corynebacterium glutamicum, pp. 351-389. In Yukawa H, Inui M (eds.). Corynebacterium glutamicum. Microbiobiology Monographs 23. Springer, Berlin, Germany.
  143. Vertès AA, Inui M, Kobayashi M, Kurusu Y, Yukawa H. 1993. Presence of mrr- and mcr-like restriction systems in coryneform bacteria. Res. Microbiol. 144: 181-185. https://doi.org/10.1016/0923-2508(93)90043-2
  144. Vogt M, Haas S, Klaffl S, Polen T, Eggeling L, van Ooyen J, Bott M. 2014. Pushing product formation to its limit: metabolic engineering of Corynebacterium glutamicum for ʟ-leucine overproduction. Metab. Eng. 22: 40-52. https://doi.org/10.1016/j.ymben.2013.12.001
  145. Wada M, Awano N, Haisa K, Takagi H, Nakamori S. 2002. Purification, characterization and identification of cysteine desulfhydrase of Corynebacterium glutamicum, and its relationship to cysteine production. FEMS Microbiol. Lett. 217: 103-107. https://doi.org/10.1111/j.1574-6968.2002.tb11462.x
  146. Watanabe K, Tsuchida Y, Okibe N, Teramoto H, Suzuki N, Inui M, Yukawa H. 2009. Scanning the Corynebacterium glutamicum R genome for high-efficiency secretion signal sequences. Microbiology 155: 741-750. https://doi.org/10.1099/mic.0.024075-0
  147. Wendisch VF. 2003. Genome-wide expression analysis in Corynebacterium glutamicum using DNA microarrays. J. Biotechnol. 104: 273-285. https://doi.org/10.1016/S0168-1656(03)00147-0
  148. Wendisch VF, Bott M, Eikmanns BJ. 2006. Metabolic engineering of Escherichia coli and Corynebacterium glutamicum for biotechnological production of organic acids and amino acids. Curr. Opin. Microbiol. 9: 268-274. https://doi.org/10.1016/j.mib.2006.03.001
  149. Wendisch VF, Bott M, Kalinowski J, Oldiges M, Wiechert W. 2006. Emerging Corynebacterium glutamicum systems biology. J. Biotechnol. 124: 74-92. https://doi.org/10.1016/j.jbiotec.2005.12.002
  150. Wieschalka S, Blombach B, Bott M, Eikmanns BJ. 2013. Bio-based production of organic acids with Corynebacterium glutamicum. Microb. Biotechnol. 6: 87-102. https://doi.org/10.1111/1751-7915.12013
  151. Yamamoto S, Suda M, Niimi S, Inui M, Yukawa H. 2013. Strain optimization for efficient isobutanol production using Corynebacterium glutamicum under oxygen deprivation. Biotechnol. Bioeng. 110: 2938-2948. https://doi.org/10.1002/bit.24961
  152. Yim S, An S, Choi J, Ryu A, Jeong K. 2014. High-level secretory production of recombinant single-chain variable fragment (scFv) in Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 98: 273-284. https://doi.org/10.1007/s00253-013-5315-x
  153. Yim SS, An SJ, Kang M, Lee J, Jeong KJ. 2013. Isolation of fully synthetic promoters for high-level gene expression in Corynebacterium glutamicum. Biotechnol. Bioeng. 110: 2959-2969. https://doi.org/10.1002/bit.24954
  154. Yim SS, Choi JW, Lee RJ, Lee YJ, Lee SH, Kim SY, Jeong KJ. 2016. Development of a new platform for secretory production of recombinant proteins in Corynebacterium glutamicum. Biotechnol. Bioeng. 113: 163-172. https://doi.org/10.1002/bit.25692
  155. Youn JW, Jolkver E, Kramer R, Marin K, Wendisch VF. 2008. Identification and characterization of the dicarboxylate uptake system DccT in Corynebacterium glutamicum. J. Bacteriol. 190: 6458-6466. https://doi.org/10.1128/JB.00780-08
  156. Youn JW, Jolkver E, Kramer R, Marin K, Wendisch VF. 2009. Characterization of the dicarboxylate transporter DctA in Corynebacterium glutamicum. J. Bacteriol. 191: 5480-5488. https://doi.org/10.1128/JB.00640-09
  157. Yukawa H, Omumasaba CA, Nonaka H, Kós P, Okai N, Suzuki N, et al. 2007. Comparative analysis of the Corynebacterium glutamicum group and complete genome sequence of strain R. Microbiology 153: 1042-1058. https://doi.org/10.1099/mic.0.2006/003657-0
  158. Zahoor A, Otten A, Wendisch VF. 2014. Metabolic engineering of Corynebacterium glutamicum for glycolate production. J. Biotechnol. 192: 366-375. https://doi.org/10.1016/j.jbiotec.2013.12.020
  159. Zhang B, Zhou N, Liu Y-M, Liu C, Lou C-B, Jiang C-Y, Liu S-J. 2015. Ribosome binding site libraries and pathway modules for shikimic acid synthesis with Corynebacterium glutamicum. Microb. Cell Fact. 14: 71. https://doi.org/10.1186/s12934-015-0254-0
  160. Zhang C, Zhang J, Kang Z, Du G, Chen J. 2015. Rational engineering of multiple module pathways for the production of ʟ-phenylalanine in Corynebacterium glutamicum. J. Ind. Microbiol. Biotechnol. 42: 787-797. https://doi.org/10.1007/s10295-015-1593-x
  161. Zhang D, Guan D, Liang J, Guo C, Xie X, Zhang C, et al. 2014. Reducing lactate secretion by ldhA deletion in ʟ-glutamate-producing strain Corynebacterium glutamicum GDK-9. Braz. J. Microbiol. 45: 1477-1483. https://doi.org/10.1590/S1517-83822014000400044
  162. Zhang Y, Shang X, Lai S, Zhang G, Liang Y, Wen T. 2012. Development and application of an arabinose-inducible expression system by facilitating inducer uptake in Corynebacterium glutamicum. Appl. Environ. Microbiol. 78: 5831-5838. https://doi.org/10.1128/AEM.01147-12
  163. Zhu Q, Zhang X, Luo Y, Guo W, Xu G, Shi J, Xu Z. 2015. ʟ-Serine overproduction with minimization of by-product synthesis by engineered Corynebacterium glutamicum. Appl. Microbiol. Biotechnol. 99: 1665-1673. https://doi.org/10.1007/s00253-014-6243-0

Cited by

  1. Insight of Genus Corynebacterium : Ascertaining the Role of Pathogenic and Non-pathogenic Species vol.8, pp.None, 2017, https://doi.org/10.3389/fmicb.2017.01937
  2. Generic Protocol for Optimization of Heterologous Protein Production Using Automated Microbioreactor Technology vol.130, pp.None, 2016, https://doi.org/10.3791/56234
  3. Heme Derived from Corynebacterium glutamicum: A Potential Iron Additive for Swine and an Electron Carrier Additive for Lactic Acid Bacterial Culture vol.27, pp.3, 2016, https://doi.org/10.4014/jmb.1611.11010
  4. The underlying mechanisms of genetic innovation and speciation in the family Corynebacteriaceae: A phylogenomics approach vol.107, pp.None, 2016, https://doi.org/10.1016/j.ympev.2016.11.009
  5. The whcD gene of Corynebacterium glutamicum plays roles in cell division and envelope formation vol.163, pp.2, 2017, https://doi.org/10.1099/mic.0.000399
  6. A Novel Two-Component System, GluR-GluK, Involved in Glutamate Sensing and Uptake in Streptomyces coelicolor vol.199, pp.18, 2016, https://doi.org/10.1128/jb.00097-17
  7. Construction of Recombinant Pdu Metabolosome Shells for Small Molecule Production in Corynebacterium glutamicum vol.6, pp.11, 2016, https://doi.org/10.1021/acssynbio.7b00167
  8. CRISPR-Cpf1 assisted genome editing of Corynebacterium glutamicum vol.8, pp.1, 2017, https://doi.org/10.1038/ncomms15179
  9. A new genome-scale metabolic model of Corynebacterium glutamicum and its application vol.10, pp.None, 2016, https://doi.org/10.1186/s13068-017-0856-3
  10. Recent advances in metabolic engineering of Corynebacterium glutamicum for bioproduction of value-added aromatic chemicals and natural products vol.102, pp.20, 2016, https://doi.org/10.1007/s00253-018-9289-6
  11. Plasmid copy number mutation in repA gene encoding RepA replication initiator of cryptic plasmid pHM1519 in Corynebacterium glutamicum vol.82, pp.12, 2016, https://doi.org/10.1080/09168451.2018.1508986
  12. New Insights on Steroid Biotechnology vol.9, pp.None, 2018, https://doi.org/10.3389/fmicb.2018.00958
  13. Our microbes not only produce antibiotics, they also overproduce amino acids vol.71, pp.1, 2018, https://doi.org/10.1038/ja.2017.142
  14. Recent advances in metabolic engineering ofCorynebacterium glutamicumas a potential platform microorganism for biorefinery : Corynebacterium glutamicum strains as industrial microorganisms for biorefi vol.12, pp.5, 2016, https://doi.org/10.1002/bbb.1895
  15. Comparative Genomic and Genetic Functional Analysis of Industrial L-Leucine- and L-Valine-Producing Corynebacterium glutamicum Strains vol.28, pp.11, 2016, https://doi.org/10.4014/jmb.1805.05013
  16. A RecET-assisted CRISPR–Cas9 genome editing in Corynebacterium glutamicum vol.17, pp.None, 2018, https://doi.org/10.1186/s12934-018-0910-2
  17. Production of the compatible solute α- D -glucosylglycerol by metabolically engineered Corynebacterium glutamicum vol.17, pp.None, 2016, https://doi.org/10.1186/s12934-018-0939-2
  18. Mutations in Peptidoglycan Synthesis Gene ponA Improve Electrotransformation Efficiency of Corynebacterium glutamicum ATCC 13869 vol.84, pp.24, 2016, https://doi.org/10.1128/aem.02225-18
  19. Reconstruction of tricarboxylic acid cycle in Corynebacterium glutamicum with a genome‐scale metabolic network model for trans‐4‐hydroxyproline production vol.116, pp.1, 2016, https://doi.org/10.1002/bit.26818
  20. Identifying the Growth Modulon of Corynebacterium glutamicum vol.10, pp.None, 2016, https://doi.org/10.3389/fmicb.2019.00974
  21. Impaired oxidative stress and sulfur assimilation contribute to acid tolerance of Corynebacterium glutamicum vol.103, pp.4, 2016, https://doi.org/10.1007/s00253-018-09585-y
  22. Isolation of Novel Exo-type β-Agarase from Gilvimarinus chinensis and High-level Secretory Production in Corynebacterium glutamicum vol.24, pp.1, 2019, https://doi.org/10.1007/s12257-018-0362-x
  23. Recent advances in the applications of promoter engineering for the optimization of metabolite biosynthesis vol.35, pp.2, 2019, https://doi.org/10.1007/s11274-019-2606-0
  24. Glutamate interactions with obesity, insulin resistance, cognition and gut microbiota composition vol.56, pp.5, 2016, https://doi.org/10.1007/s00592-019-01313-w
  25. Metabolic engineering to improve 1,5‐diaminopentane production from cellobiose using β‐glucosidase‐secreting Corynebacterium glutamicum vol.116, pp.10, 2016, https://doi.org/10.1002/bit.27082
  26. Investigation of Potential Brain Microbiome in Alzheimer’s Disease: Implications of Study Bias vol.75, pp.2, 2016, https://doi.org/10.3233/jad-191328
  27. Microbial production of 4‐amino‐1‐butanol, a four‐carbon amino alcohol vol.117, pp.9, 2020, https://doi.org/10.1002/bit.27438
  28. Recent Advances in Microbial Production of cis,cis -Muconic Acid vol.10, pp.9, 2016, https://doi.org/10.3390/biom10091238
  29. CRISPR base editing and prime editing: DSB and template-free editing systems for bacteria and plants vol.5, pp.4, 2016, https://doi.org/10.1016/j.synbio.2020.08.003
  30. Single-Base Genome Editing in Corynebacterium glutamicum with the Help of Negative Selection by Target-Mismatched CRISPR/Cpf1 vol.30, pp.10, 2016, https://doi.org/10.4014/jmb.2006.06036
  31. Improved pEKEx2-derived expression vectors for tightly controlled production of recombinant proteins in Corynebacterium glutamicum vol.112, pp.None, 2016, https://doi.org/10.1016/j.plasmid.2020.102540
  32. GEDpm-cg: Genome Editing Automated Design Platform for Point Mutation Construction in Corynebacterium glutamicum vol.9, pp.None, 2016, https://doi.org/10.3389/fbioe.2021.768289
  33. Biosensor-Based Optimization of Cutinase Secretion by Corynebacterium glutamicum vol.12, pp.None, 2016, https://doi.org/10.3389/fmicb.2021.750150
  34. The Metano Modeling Toolbox MMTB: An Intuitive, Web-Based Toolbox Introduced by Two Use Cases vol.11, pp.2, 2016, https://doi.org/10.3390/metabo11020113
  35. In Silico Prediction and Analysis of Unusual Lantibiotic Resistance Operons in the Genus Corynebacterium vol.9, pp.3, 2016, https://doi.org/10.3390/microorganisms9030646
  36. Diosmetin ameliorate type 2 diabetic mellitus by up-regulating Corynebacterium glutamicum to regulate IRS/PI3K/AKT-mediated glucose metabolism disorder in KK-Ay mice vol.87, pp.None, 2016, https://doi.org/10.1016/j.phymed.2021.153582
  37. Improving the Microbial Production of Amino Acids: From Conventional Approaches to Recent Trends vol.26, pp.5, 2016, https://doi.org/10.1007/s12257-020-0390-1
  38. Tyrosinase-based production of l-DOPA by Corynebacterium glutamicum vol.105, pp.24, 2021, https://doi.org/10.1007/s00253-021-11681-5
  39. Nutritional and functional values of lysed Corynebacterium glutamicum cell mass for intestinal health and growth of nursery pigs vol.99, pp.12, 2021, https://doi.org/10.1093/jas/skab331
  40. Synthetic Biology Toolkits and Metabolic Engineering Applied in Corynebacterium glutamicum for Biomanufacturing vol.10, pp.12, 2016, https://doi.org/10.1021/acssynbio.1c00355
  41. Effect of Clp protease from Corynebacterium glutamicum on heterologous protein expression vol.189, pp.None, 2016, https://doi.org/10.1016/j.pep.2021.105928