METHODS FOR THE IMPROVED FORMATION OF ACARBOSE

Abstract
The present invention relates to Actinomycetales strains for the improved formation of acarbose. Provided are Actinomycetales strains which are engineered to overexpress dTDP-D-glucose-4,6-dehydratase (AcbB) and/or uridyltransferase (GtaB). Also provided are Actinomycetales strains which are engineered to have a reduced or absent expression of the small carbohydrate binding protein (Cgt) and/or a reduced or absent expression of genes which are essential for carotenoid synthesis. Also provided are tools, methods and means to generate these strains.
Description
FIELD OF THE DISCLOSURE

The present invention relates to Actinomycetales strains for the improved formation of acarbose. Provided are Actinomycetales strains which are engineered to overexpress dTDP-D-glucose-4,6-dehydratase (AcbB) and/or uridyltransferase (GtaB). Also provided are Actinomycetales strains which are engineered to have a reduced or absent expression of the small carbohydrate binding protein (Cgt) and/or a reduced or absent expression of genes which are essential for carotenoid synthesis. Also provided are tools, methods and means to generate these strains.


BACKGROUND

Acarbose


The therapeutic agent acarviosyl-maltose (acarbose) is used since 1990 in the medical treatment of diabetes mellitus (Wehmeier and Piepersberg 2004; Wehmeier 2004). It shall support the rigorous diet plan of patients and prevent sugar peaks when consuming high carb meals. After oral application acarbose inhibits intestinal α-glucosidases, which leads to a retarded release of monosaccharides from starch- and sucrose-containing diets. By this, acarbose helps to control the rate of absorption of monosaccharides into the blood system and leads to a decreased postprandial blood and serum sugar level, which are assumed to be crucial in the context of cardiovascular disease mortality.


INDUSTRIAL APPLICABILITY

Acarbose is known and marketed in Europe and China as Glucobay (Bayer AG), in North America as Precose (Bayer Pharmaceuticals), and in Canada as Prandase (Bayer AG). Being an important and highly demanded drug for the treatment of diabetes mellitus, there is a need to provide acarbose in high yields and high quality. As the incidence of type II diabetes is continuously increasing worldwide, optimization of product yields and quality is an issue of current concern.


Acarbose Production Strains


Acarbose is naturally produced by different Actinomycetales, like Streptomyces coelicoflavus ZG0656 (Geng et al. 2009), Streptomyces glaucescens GLA.O (Rockser and Wehmeier 2009; Ortseifen et al. 2015) and Actinoplanes sp. SE50/110 (reviewed by Wehmeier and Piepersberg 2004), of which the latter is the wild type of industrial producer strains (Ortseifen 2016; Mahmud et al. 1999). The genus Actinoplanes was first introduced by Couch (1950) as member of the family Micromonosporaceae, order Actinomycetales, phylum Actinobacteria. Actinoplanes sp. SE50/110 (ATCC 31044, CBS 674.73) is a slow-growing natural derivate of SE50 (ATCC 31042, CBS 961.70) (Frommer et al. 1973). SE50 was isolated in 1970 during a screening program by the Bayer AG from a soil sample close to a coffee plantation in Kenia (Frommer et al. 1972). SE50/110 produces approximately 1 g·L1 acarbose, when maltose is provided in the medium (Wendler et al. 2014). Further production strains have been engineered, as described for example in (EP2601209B1) and (CN103298828B).


For Actinoplanes sp. SE50/110 it was shown, that the biosynthesis of acarviosyl-sugars depends on the supply of carbon sources in the culturing medium (Wendler et al. 2014). Growing on glucose, acarviosyl-glucose was formed as the major compound, whereas mainly acarviosyl-maltose was formed, when growing on maltose (Wendler et al. 2014), and acarviosyl-maltotriose, when growing on maltotriose (Ortseifen 2016).


Due to its medical and industrial relevance as wild type of industrial acarbose producer strains, Actinoplanes sp. SE50/110 was extensively studied in the last years: The complete genome (Schwientek et al. 2012), transcriptome (Schwientek et al. 2013) and proteome (Wendler et al. 2015b; Wendler et al. 2015a; Wendler et al. 2013) were analyzed comprehensively. This led to a refined genome sequence and an improved annotation in 2017 (GenBank: LT827010.1) (Wolf et al. 2017b). Also, an intergeneric conjugation system (Gren et al. 2016) as well as advanced genome editing tools by use of CRISPR/Cas9 (Wolf et al. 2016) were established, allowing targeted genetic engineering. Still, a reliable expression system enabling medium to strong gene expression is missing for Actinoplanes sp. SE50/110. Since a suitable system for the medium strong overexpression of singular genes did not exist before, different strategies were tested and evaluated according to the current invention, which led to the development of a novel expression system called pSETT4.


Acarbose Biosynthesis


The biosynthetic pathway of acarbose is based on monofunctional enzymes catalyzing single steps (FIG. 1) (Wehmeier and Piepersberg 2009). According to the model of Zhang et al. (2002), the biosynthesis proceeds via the intermediate valienone-7P. In the refinement by Wehmeier (2003), the steps reduction and dehydration were changed leading to valiolol-7P as intermediate.


As the order of steps is unknown, they are shown in brackets. The first step of acarbose biosynthesis, a cyclic reaction by AcbC to form 2-epi-5-epi-valiolone from sedo-heptulose-7P, is missing in this illustration. Although becoming a focus of research in the last decades, still, the acarbose biosynthetic pathway is not completely unraveled yet. Only the first three steps of biosynthesis were experimentally confirmed. AcbC (ACSP50_3607), the first enzyme of the acarbose biosynthesis, catalyzes a cyclic reaction to generate 2-epi-5-epi-valiolone from sedo heptulose 7P7P (Stratmann et al., 1999). Phosphorylation to 2-epi-5-epi-valiolone-7P is catalyzed by the kinase AcbM (ACSP50_3603) in presence of ATP (Zhang et al. 2002) and epimerization to 5-epi-valiolone-7P by the cofactor independent epimerase AcbO (ACSP50_3606) (Zhang et al. 2002; Zhang et al. 2003).


The remaining steps of the model base on protein homologies and functional predictions (Zhang et al. (2002), Wehmeier (2003), Wehmeier and Piepersberg (2004), Wehmeier and Piepersberg (2009) and Wendler et al. (2013): NADH-dependent (polyol)dehydrogenase/reductase AcbL (ACSP50_3604) and the cyclitol dehydrogenase/oxidoreductase AcbN (ACSP50_3605) have been suggested to catalyze reduction and 5,6 dehydration to 1-epi-valienol-7P. Phosphorylation to 1,7-diphospho-1-epi-valienol is assumed to be catalyzed by the 1-epi-valienol-7-phosphate-1-kinase AcbU (ACSP50_3595) and/or hydrolase AcbJ (ACSP50_3600). Nucleotidylation to NDP-1-epi-valienol-7P is possibly catalyzed by a GlgC-related NDP-polyol synthase AcbR (ACSP50_3597) (1-epi-valienol-1,7-bisphosphate-1-adenylyltransferase), and transfer of the activated intermediate to an activated amino sugar seems to be mediated by the glycosyltransferases Acbl (ACSP50_3599) and/or AcbS (ACSP50_3596) to generate acarviosine-7P.


The activated amino sugar is supposed to be synthesized from D-glucose-1-phosphate in three steps (Wehmeier and Piepersberg 2004; Wehmeier and Piepersberg 2009; Zhang et al. 2002), which are: (i) nucleotidylation by the dTDP-glucose-synthase AcbA (ACSP50_3609) to dTDP-D-glucose, (ii) dehydration by dTDP-D-glucose-4,6-dehydratase AcbB (ACSP50_3608) to dTDP-4-keto-6-deoxy-D-glucose, and (iii) amination by a GabT-like aminotransferase AcbV (ACSP50_3594) to dTDP-4-amino-4,6-dideoxy-D-glucose (Diaz-Guardamino Uribe 2000; Zhang et al. 2019).


Glucose-1P is a branching metabolite displaying an important role in different pathways, like the glycogen metabolism, the galactose metabolism and—after conversion to glucose-6P—the glycolysis (Frey 1996; Purves 2006). UDP-glucose-1P uridyltransferase GtaB catalyzes the conversion of glucose-1P and UDP-glucose into each other.


Last, maltose is transferred in a one-step reaction (Hemker et al. 2001), potentially by AcbS. However Acbl or AcbJ have also been proposed to catalyze the transfer reaction (Wehmeier and Piepersberg 2004; Wendler et al. 2013). Another candidate for this reaction might be the amylomaltase AcbQ (ACSP50_3601).


In Actinoplanes sp. SE50/110, the biosynthesis genes are organized in the acarbose biosynthesis gene cluster (acb gene cluster), which was first identified in 1999 by Stratmann et al. and subsequently sequenced (GenBank: Y18523.4) (Stratmann et al. 1999; Thomas 2001). The cluster contains 22 genes (FIG. 2).


Beside of the already mentioned biosynthetic genes (acbCMOLNUJRSIVBA), the cluster encodes functions in extracellular starch degradation (AcbEZ, ACSP50_3610 and ACSP50_3590), transglycosylation (AcbD, ACSP50_3611) and in export of acarbose (AcbWXY, ACSP50_3591-3). Besides, an acarbose-7-kinase (AcbK, ACSP50_3602) and an intracellular amylomaltase (AcbQ) are encoded, which were assigned to a function within the carbophore (Wendler et al. 2015b; Schwientek et al. 2012; Wehmeier and Piepersberg 2009). The function of AcbP (ACSP50_3598), annotated as NTP-pyrophosphohydrolase, is unknown.



Actinoplanes Proteins with Possible Metabolic Relevance


The singular CBM-20 domain protein Cgt is one of the most strongly expressed genes in Actinoplanes sp. SE50/110 and in derived acarbose producer strains (Ortseifen 2016; Wendler et al. 2015a; Schwientek et al. 2013). It is secreted via the Sec-pathway according to SignalP-analysis (Almagro Armenteros et al. 2019) and makes up to 8% of the total secreted proteome of this organisms (data not shown). Cgt contains 149 amino acids and a CBM-20-domain of fold-family 1, functional group A, characterized by a p-sandwich structure (Schwientek et al. 2013; Guillén et al. 2010). Members of this family are described to bind starch (Guillén et al. 2010).


Methods for Gene Deletion in Actinoplanes


The establishment of an intergeneric conjugation system (Gren et al. 2016) and the CRISPR/Cas9 technique (Wolf et al. 2016), allow genome editing in Actinoplanes sp. SE50/110. In addition, according to the current invention the inventors have successfully established a novel deletion system by homologous recombination, which uses an integrase-free vector backbone and CodA for counter selection, like described by Zhao et al. (2017). By this, the genetic toolbox for Actinoplanes sp. SE50/110 could be further extended. As proof of principle the novel deletion system was successfully tested for deletion of the example gene cgt. Homologous recombination (HR) is a common process in Actinobacteria, which can be technically used to create deletion mutants by double crossover. Temperature-sensitive replicons, like the pSG5 replicon, can support and force this process. (Du et al. 2015; Garg and Parry 2010; Myronovskyy et al. 2009; Zhang and Parry 2007). Further methods exist in the art, e.g. CRISPR-Base Editing System for the exchange of single nucleotides, CRISPR-BEST according to Tong et al. 2019, CRISPRi/dCas9 according to Qi et al. 2013, RNA interference etc.


Methods for Gene Overexpression in Actinoplanes



Actinoplanes sp. SE50/110 has been extensively studied in the last decades. Appropriate expression systems are difficult to design, see Schaffert et al. (2019). The whole content of the publication and in particular the description of the expression systems and promoters for the genetic manipulation of Actinoplanes are included herein in their entirety.


Previous studies have shown successful expression of genes by use of pKC1139 in A. teichomyceticus (Horbal et al. 2012). However, the replicative pSG5-based vector pKC1139 (constructed by Bierman et al. (1992)) turned out to be unsuitable for expression of homologous genes in Actinoplanes sp. SE50/110, as unwanted vector integration by homologous recombination occurs, see Schaffert et al. (2019). This seems to be a favored process, putatively due to the high metabolic costs of vector replication. Without being bound by theory, a protein encoded by ACSP50_7170 in SE50/110, predicted as recombinase A (recA) might catalyze the recombination process. Interestingly, no homologue of recA was found in the genome of A. teichomyceticus. Presence of recA in a. sp. SE50/110 and lack in A. teichomyceticus provides a conclusive explanation, why HR-mediated vector-integration has not been reported for A. teichomyceticus before. A pSG5-based replicative expression system may therefore be implemented by deletion of the recombinase gene recA in Actinoplanes sp. SE50/110.


Other replicative Streptomyces-E. coli shuttle plasmids, like pKC1218, which is based on the SCP2*-replicon (Kieser et al. 2000), and pSOK101, which is based on the plJ101-replicon (Zotchev et al. 2000), did not give exconjugants with Actinoplanes sp. SE50/110 (Gren 2017). These replicons are probably unstable or inactive in SE50/110, which is in accordance with findings from the related species A. teichomyceticus (Horbal et al. 2012).


By use of integrative vector systems, a genetic duplication can be achieved by integration of the complete vector carrying an additional gene copy at a distinct genomic location. This process is mediated by phage integrases. Phage integrases catalyze the targeted and unidirectional recombination of two attachment sites: attP, which is localized on the plasmid, and attB, which is localized in the host chromosome (te Poele et al., 2008). After integration, the vector is flanked by the attachment site left (attL) and right (attR), which are derived from attP-attB-recombination (te Poele et al., 2008).


Four different integrative vectors have been described for Actinoplanes sp. SE50/110 (Gren et al. 2016): Two are based on the integration mechanism of the phage φC31 (pSET152 and plJ6902). The vectors pRT801/2 and pSOK804 are based on the integration mechanism of the phage φBT1 and of the VWB-phage. However, doubling of relative transcript amounts by use of the native promoters was not achieved, see Schaffert et al. (2019).


Evaluation of Homologous and Heterologous Promoters for Integrative Vectors


A method to evaluate homologous and heterologous promoters with regard to their strength is provided in Schaffert et al. (2019), which is incorporated herein in its entirety. In brief, the integrative φC31-based vector pSET152 was used for promoter screening in Actinoplanes sp. SE50/110 (Gren et al. 2016). The promoter strengths of 13 homologous and heterologous promoters were analyzed on protein level, and 12 of these were analyzed on transcript level (Table 1, FIG. 3).









TABLE 1







Constructs with the reporter gene gusA tested in a promoter screening experiment.












abbr.
origin of promoter
construct
source/comment















homologous
cgt
promoter of cgt (ACSP50_5024), annotated
pSETcgtPgusA
Schaffert et al. (2019)




as small carbohydrate binding protein



efp
promoter of efp (ACSP50_6465), annotated
pSETefpPgusA
Schaffert et al. (2019)




as the translation elongation factor P



7457
promoter of ACSP50_7457, annotated as
pSET7457PgusA
Schaffert et al. (2019)




hypothetical protein



katE
promoter of katE (ACSP50_3066),
pSETkatEPgusA
Schaffert et al. (2019)




annotated as catalase hydroperoxidase




(HP) II



rpsJ
promoter of rpsJ (ACSP50_0690),
pSETrpsJPgusA
Schaffert et al. (2019)




annotated as 30S ribosomal protein S10


heterologous
tipA
promoter of tipA from S. lividans, annotated
pSETGUS
Myronovskyi et al. (2011)




as HTH-type transcriptional activator (Chiu




etal. 1999)



moeE5
promoter of moeE5 from S. ghananensis, a
pSETPmoeES
Horbal et al. (2013) and




central moenomycin A biosynthetic gene

R. Makitrynskyy, Ivan




encoding for a nucleotide sugar epimerase

Franko National




(Ostash et al. 2009)

University, Lviv, Ukraine



apm
promoter of aac(3)IV from
pSETaac(3)IVPgusA
Schaffert et al. (2019)




pCRISPomyces-2 (Cobb et al. 2015), an




aminoglycoside 3-N-acetyltransferase,




which mediates apramycin resistance



cdaR
promoter of cdaR from S. coelicolor,
pSETPcdaRgusA
Horbal etal. (2013)




encoding for a transcriptional regulator




(proposed as activator of a calcium-




dependent antibiotic CDA (McKenzie and




Nodwell 2007; Ryding et al. 2002))



act
promoter of actII-4 from S. coelicolor,
pSETactPgusA
Schaffert et al. (2019),




annotated as actinorhodin operon activator

using primers and design




protein (Fernández-Moreno et al. 1991)

from Horbal et al. (2013)



ermE*
promoter of ermE* from S. erythraea,
pGUSPErmE
Schaffert et al. (2019),




annotated as 23S rRNA (adenine-N6)-

referring to Siegl et al.




dimethyltransferase mediating erythromycin

(2013); Bibb et al. (1994)




resistance (Thompson et al. 1982)



gapDH
promoter of gapDH from Eggerthella Ienta
pSETgapDHPgusA
Schaffert et al. (2019)




used on pCRISPomyces-2 (Cobb et al.




2015), annotated as type I glyceraldehyde-




3-phosphate dehydrogenase



rpsL
promoter of rpsL from
pSETrpsLPgusA
Schaffert et al. (2019)




Xylanimonas cellulosilytica used on




pCRISPomyces-2 (Cobb et al. 2015),




annotated as ribosomal protein S12



pGUS
no promoter
pGUS
Myronovskyi et al. (2011)









Strategy


For the current invention, the acarviosyl-maltose metabolism was studied by gene deletion and overexpression, leading to a set of associated tools and methods to engineer strains for the improved production of acarbose. In order to improve the acarviose-synthesis, three different strategies were followed: (i) increasing of the gene dose of acb genes to enhance the flux through the acarbose biosynthesis, (ii) deployment of precursors of acarbose biosynthesis and (iii) reducing the metabolic burden (FIG. 4). Approaches of each of these related strategies surprisingly led to an improved acarbose formation: By overexpression of dTDP-D-glucose-4,6-dehydratase AcbB, the final acarbose concentration was significantly increased by approx. 50%. By overexpression of the uridyltransferase GtaB acarbose yields were improved by 8.5%, presumably because the supply of the precursor glucose-1P was improved. By functional deletion of the small carbohydrate binding protein Cgt, the acarbose formation was significantly enhanced by 8-16%, possibly because the metabolic burden is thereby reduced. The enhancement was robust over long time periods and in different cultivation settings.


Furthermore, growth experiments of the wild type and a regulator mutant ΔmerR exposed to and hidden from light were conducted, revealing a negative influence of light-induced stress and carotenoid formation on the acarbose production. Consequently, the acarbose production can furthermore be improved by reducing the carotenoid formation.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Model of the biosynthesis of acarviosyl-maltose in Actinoplanes sp. SE50/110. Shown are the eleven steps of acarbose biosynthesis from 2-epi-5-epi-valiolone (Zhang (2002)).



FIG. 2. The acarbose biosynthesis gene cluster and gene disposition in the genome of Actinoplanes sp. SE50/110 (GenBank: LT827010.1) (Schaffert, et al. 2019).



FIG. 3. Promoter screening on protein and transcript level in Actinoplanes sp. SE50/110 strains, cf. Table 1. Shown are the normalized glucuronidase activities on the left side (absolute values) and the relative transcript amount of gusA gene calculated by RT-qPCR on the right side. For the glucuronidase assay, the slope of absorption curves of indigo was calculated by linear regression and normalized by the cell dry weights. The normalized activities were tested for significant differences compared to pGUS in a two-sided t-test (p-values: P2475: 0.8889, Pefp: 3.048e-07, PcdaR: 8.967e-07, PrpsL: 1.296e-08, PrpsJ: 0.0003677, Pcgt: 2.183e-06, PtipA: 0.0001651, Papm: 0.0001078, PermE*: 0.007406, PkatE: 0.002577, PmoeE5: 0.001809, PgapDH: 0.0005821, Pact: 0.02042). The relative transcription amounts of gusA gene were analyzed in relation to the pGUS-vector (set to 1). For the act-promoter, no RNA could be isolated due to severe growth deficiencies. For the residual promoters, a significant increase in the relative transcript amount was measured (p-values of a two-sided t-test: P2475: 0.0001133, Pefp: 4.871e-05, PcdaR: 0.002509, PrpsL: 9.928e-06, PrpsJ: 1.167e-08, Pcgt: 5.911e-08, PtipA: 7.158e-06, Papm: 4.596e-05, PermE*: 0.0009364, PkatE: 0.0001373, PmoeE5: 0.0002518, PgapDH: 4.207e-06). Significance levels of the calculated p-values are shown by asterisks: *<α=5%, **<α=1%, ***<α=0.1%. Figure published in (Schaffert, et al. 2019).



FIG. 4. Strategies for improved acarbose production. Three different strategies are provided to improve the acarbose production: 1. Increasing the gene dose of acarbose biosynthesis genes, 2. Deployment of precursors of acarbose biosynthesis and 3. Reducing the metabolic burden by gene deletion. Shown are the target genes evaluated in this work. Furthermore, an overexpression system had to be implemented for the overexpression of single genes.



FIG. 5. Chemical structure of acarbose. Acarbose is a cyclitol-containing aminoglycosid composed of a pseudodisaccharide (valienaminyl-4-amino-4,6-dideoxyglucose), called acarviose, and maltose. Both are connected by an α-1,4-glycosidic bond. Figure published in Wolf 2017.



FIG. 6. Vector card of novel cloning system pSETT4 (cf. SEQ ID No. 110, SEQ ID No. 111). A promoter, such as the strong promoter of the gene gapDH from Eggerthella lenta or the tipA promoter is cloned in front of an expression cassette, e.g. the lacZ-cassette. The lacZ-cassette is flanked by a recognition side of a restriction enzyme e.g. BsaI. The restriction site enables exchange of lacZ by the gene of interest by Gibson Assembly, restriction/ligation cloning or Golden Gate cloning. For termination, T4-terminators are introduced before and after the cloning side. Behind the cloning side, two antiparallel oriented T4-terminators shall prevent read-through from both directions. For exchange of the promoter sequence, further restriction sites, e.g. NdeI and KpnI restriction sites were introduced. Furthermore, the vector comprises the integrase gene int and the attachment site attP of the phage φC31, the origin of transfer (ncP) and relaxosome gene traJ, the high-copy-number ColE1/pMB1/pBR322/pUC origin of replication and an resistance gene (here: apramycin resistance gene aac(3)IV (apmR)).



FIG. 7. Scheme of the novel deletion system and the processes during the homologous recombination (first and second crossover). Selection of vector integration is performed by use of either apramycin or kanamycin (first crossover, resistance mediated by Apm® or Kan®). Counterselection is performed by use of 5-flourouracil (second crossover, sensitivity mediated by codA).



FIG. 8. Workflow of novel deletion system using homologous recombination.



FIG. 9. BlastP analysis of the amino acid sequence of Cgt leads to the identification of 17 other proteins consisting of a singular CBM-20 domain. The protein tree was created and visualized on the basis of multiple sequence alignment performed by BlastP (Altschul et al. 1990). The protein tree shows the distance of the 18 singular CBM-20 domain proteins, identified by the NCBI accession number and their hosts. In brackets the sequence identity and positives of BlastP analysis are shown in percentages.



FIG. 10. Growth of the wild type of Actinoplanes sp. SE50/110 in minimal medium supplemented with different carbon sources (in equal C-molar amounts). Shown are the cell dry weights of at least three biological replicates and the standard deviation (nglc=3, nmal=5, ncel=4, nlac=3, nara=5, nstarch=5).



FIG. 11. A. Relative transcript amounts of cgt in Actinoplanes sp. SE50/110 grown on minimal medium supplemented with starch, C-Pur, glucose, galactose, cellobiose, or lactose as carbon source, compared to a culture grown on maltose minimal medium. Testing for differences in a two-sided t-test displayed significant differential gene expression of the cgt gene on the carbon sources glucose (p-value=0.002848), galactose (p-value=0.002945) and lactose (p-value=0.00114) compared to maltose. B. Relative transcript amount of cgt in Actinoplanes sp. SE50/110 grown on maltose minimal medium complemented with 44.40 g·L−1 maltose compared to a culture grown on 72.06 g·L−1 maltose. Testing for differences in a two-sided t-test displayed significant reduced gene expression of cgt in the medium containing reduced amounts of maltose (p-value=0.04141).



FIG. 12. Growth of the wild type and the deletion mutant Δcgt of Actinoplanes sp. SE50/110 in minimal medium complemented with different carbon sources. Shown are the cell dry weights and the standard deviation over time (wild type: nglc=3, nmal=5, ncel=4, nlac=3, nara=5, nstarch=5, Δcgt: nglc=2, nmal=5, ncel=4, nlac=4, nara=5, nstarch=5).



FIG. 13. Final cell dry weights obtained in cultivations of the wild type and the Δcgt mutant in minimal media supplemented with six different carbon sources. The error bars denote standard deviations.



FIG. 14. Growth of Δcgt and the wild type under limited amounts of starch as carbon source. Medium was supplemented with 1 g·L−1, 2 g·L−1, 3 g·L−1, 4 g·L−1 and 5 g·L−1 starch and cultivation was performed in the RoboLector® system of m2p labs. Shown are the backscatter signals in a bar diagram and standard deviation of at least three biological replicates. No restraint on growth was observed for Δcgt. For 1 g·L−1 growth was even found to be significant enhanced (p-value of a two-sided t-test: 0.006141, nwt=3, nΔcgt=4).



FIG. 15. Final cell dry weights of a pH screening experiment in maltose minimal medium. Wild type and Δcgt mutant of Actinoplanes sp. SE50/110 were grown in 1 mL reaction volume in a 48-well FlowerPlates in the RoboLector® system of m2p-labs. In pH ranging from 4 to 7, no significant differences in final cell dry weights were observed (tested by a two-sided t-test, nwt=3, nΔcgt=4).



FIG. 16. Osmolarity tolerance screening in the RoboLector® system of m2p-labs: Final cell dry weights in maltose minimal medium with maltose monohydrate concentrations ranging between 3.6 and 108.1 g·L−1. No significant growth differences were observed (tested by a two-sided t-test, nwt=3, nΔcgt=4).



FIG. 17. Osmolarity tolerance screening in the RoboLector® system of m2p-labs: Final cell dry weights of an osmolarity-screening experiment in maltose minimal medium. The different osmolarities were achieved by addition of inositol in concentrations ranging from 0 mM to 280 mM. No significant growth differences between the wild type and Δcgt were observed (tested by a two-sided t-test, nwt=3, nΔcgt=4).



FIG. 18. Growth and acarbose production of Actinoplanes sp. SE50/110 wild type and Δcgt mutant in the complex medium NBS supplemented with 11.0 g·L−1 maltose-respectively 10.0 g·L−1 glucose-monohydrate. No differential growth was detected. During growth phase, a significant increased acarbose concentration was measured in Δcgt (significance of t-test after 49 h of cultivation: p-value=0.006778, nwt-acb=3, nΔcgt-acb=3, nwt-cdwGlc=4, nΔcgt-cdwGlc=3, nwt-cdwMal=4, nΔcgt-cdwMal=4).



FIG. 19. A. Final yield coefficient of acarbose with reference to the cell dry weight in a bar chart. Error bars was calculated by Gaussian error propagation. B. Cell dry weights and acarbose concentration in the supernatant during cultivation in maltose minimal medium (ncdw=5, nacb=4).



FIG. 20. Relative transcript amounts of the genes acbZ, acbW, acbV, acbA, acbB, acbE and acbD of the mutant Δcgt compared to the wild type of Actinoplanes sp. SE50/110 grown on maltose minimal medium (n=3-6).



FIG. 21. Reconstruction of the carotenogenesis in Actinoplanes sp. SE50/110. Shown are putative homologous genes in Actinoplanes sp. SE50/110 identified by BLASTX analysis against the NCBI database. Reconstruction was performed by help of the Kyoto Encyclopedia of Genes and Genomes (Kanehisa et al. (2014))


A. Methylerythritolphosphate (MEP) pathway for the biosynthesis of the isoprenoid precursors isopentenyl pyrophosphate (IPP) and dimethylallyl pyrophosphate (DMAPP), which is also known as alternative metabolic pathway of the mevalonate pathway.


B-C: Carotenogenesis. B. Formation of Lycopene from isoprenoid precursors. C. Synthesis of the glycosylated carotenoid Sioxanthin in Salinospora tropica CNB-440 (FIG. 1 of Richter et al. (2015)).


D. Genomic organization of the identified genes in Actinoplanes sp. SE50/110. Gene cluster 2b displays homologies to the sioxanthin gene cluster from Salinospora tropica CNB-440 according to analysis by antiSMASH, a rapid genome-wide identification tool for the annotation and analysis of secondary metabolite biosynthesis gene clusters in bacterial and fungal genomes (Weber et al., 2015).



FIG. 22. Growth, acarbose and pigment formation of Actinoplanes sp. SE50/110 exposed to and covered from light. A. Cultivation of the wild type Actinoplanes sp. SE50/110 in maltose minimal medium exposed to or covered from bulb light (22-44 μE, 1 μE=μmolphotons m−2s−1). Shown are the cell dry weights of five biological replicates and the acarbose concentration in the supernatant of three biological replicates. B. Pellets and supernatants at final cultivation time. C. Growth and pigment formation in solid culture on SFM agar plates exposed to or hidden from natural light.



FIG. 23. Position of a gene encoding a MerR-regulator in terpene cluster 1 and its disposition in the genome of Actinoplanes sp. SE50/110 (cf. FIG. 21 and Table E12). The genes of the cluster encode a MerR-like transcriptional regulator (ACSP50_0145), an isopentenyl-diphosphate delta-isomerase (idi, ACSP50_0146), a phytoene dehydrogenase (crtl, ACSP50_0147), a polyprenyl synthetase (crtE, ACSP50_0148), a phytoene synthase (crtB, ACSP50_0149), a deoxyribodipyrimidine photo-lyase (ACSP50_0150) and a pyridine nucleotide-disulfide oxidoreductase (ACSP50_0151).



FIG. 24. Growth, acarbose and pigment formation of Actinoplanes sp. SE50/110 and the deletion mutant ΔmerR exposed to and covered from light. A. Cultivation of the wild type and the deletion mutant ΔmerR of Actinoplanes sp. SE50/110 in maltose minimal medium exposed to or covered from bulb light (22-44 μE, 1 μE=μmolphotons m−2s−1). Shown are the cell dry weights of at least four biological replicates and the acarbose concentration in the supernatant of three biological replicates. B. Pellets and supernatants at final cultivation time. C. Growth and pigment formation on solid media (SFM agar plates). D. Maximal acarbose concentrations (p-values of a two-sided t-test: wt dark vs. wt light: 0.003975, wt dark vs. ΔmerR dark: 0.09711, wt dark vs. ΔmerR light: 0.007043, ΔmerR dark vs. wt light: 0.02081, ΔmerR dark vs. ΔmerR light: 0.0002131). E. Relative transcript amounts of the genes crtE (ACSP50_0148), crtB (ACSP50_0149), crtl (ACSP50_0147), idi (ACSP50_0146) and merR (ACSP50_0145) in the deletion mutant compared to the wild type (set to a value of 1) when cultivated under dark conditions (p-values of a two-sided t-test: crtE: 0.04245, crtB: 0.01017, crtl: 0.07162, idi: 0.004366). Asterisks indicate the significance level: * p-value<α=5%, ** p-value<α=1%, *** p-value<α=0.1%.



FIG. 25. A ratio/intensity plot of differentially transcribed genes in Actinoplanes sp. SE50/110 exposed to light compared to a cultivation grown in the dark. The ratio (log 2(fold-change)) is plotted against the mean average intensity of a microarray experiment. Darker dots represent genes with significant differential transcription levels in the culture exposed to light compared to a culture hidden from light.



FIG. 26. ReadXplorer (Hilker et al. 2016; Hilker et al. 2014) view showing the TSS of putative antisense promoters behind the gene of interest in the pSET152-vector system. TSS were determined by sequencing of a pooled primary transcript library. Shown are the stacked reads exemplary mapped to the integrated vector-mutant of pGUS::Papm:gusA. Two TSS (surrounded by boxes), are localized behind the gene of interest in antisense orientation (A). These TSS can be assigned to sequence motifs (B) on the vector backbone, which are putatively recognized as promoter sequences by the σA/RNA-polymerase complex. Conserved nucleotides of the −10- and −35-hexamer are highlighted. TG-dimers are shown in bold black letters, if present. The distance between the hexamers is shown by s1; the distance between the −10-motif to the TSS is shown by s2.



FIG. 27. Growth and acarbose production of acbB overexpression strains in maltose minimal medium. Shown are two independent cultivations (A and B). The sampling times for RNA-isolation are indicated by t, (“early growth phase”) and t2 (“linear growth phase”).



FIG. 28. Yield coefficient of acbB overexpression mutants in maltose minimal medium. The mutant with acbB transcribed under control of the heterologous tipA-promoter displayed an enhanced yield coefficient (approx. 50%), whereas only minor differences were observed for the construct with gapDH-promoter. Errors were calculated by Gaussian error propagation. All differences were tested for significance by a two-sided t-test (abbreviations assigned in the picture). Asterisks indicate the significance level: * p-value<α=5%, ** p-value<α=1%, *** p-value<α=0.1%.



FIG. 29. Analysis of intracellular metabolites of acbB-overexpression mutants by LC-MS. Shown are the normalized peak areas of the masses m/z=545 [M-H+]. A glucose-1P and galactose-1P (m/z=259 [M-H+]. B. glucose-6P (m/z=259 [M-H+] and C. UDP-glucose (m/z=565 [M-H+].


D. Significant differences compared to the empty vector control were observed for the normalized peak areas of UDP-glucose (p-values of a two-sided t-test: Ptip: 0.01068, Pgap: 0.001356) and of the mass m/z=545 [M-H+] (p-value of a two-sided t-test: Ptip: 0.0412).



FIG. 30. Relative transcript amounts of the genes acbB, acbA and acbV in acbB-overexpression mutants in the initial growth phase. Shown are the means and standard deviations of at least three biological replicates. The differences to the empty vector control (set to a value of 1) were tested by a two-sided t-test (p-values from left to right corresponding to pSETT4gap::acbB, pSETT4tip::acbB, pSETT4::PacbB:acbB, pSET152::PacbB:acbB): acbB: 4.332e-05, 4.561e-06, 0.3511, 0.7082; acbA: 0.3384, 0.0001164, 0.5967, 0.4246; acbV: 0.3033, 0.0423, 0.73, 0.4687). Asterisks indicate the significance level: * p-value<α=5%, ** p-value<α=1%, *** p-value<α=0.1%.



FIG. 31. Relative transcript amounts of the gene acbB in acbB-overexpression mutants in the linear growth phase. Shown are the means and standard deviations of at least three biological replicates. The RT-qPCR indicates significant differences in gene expression compared to the empty vector control (set to a value of 1), which was tested by a two-sided t-test (p-values from left to right corresponding to pSETT4gap::acbB, pSETT4tip::acbB, pSETT4::PacbB:acbB, pSET152::PacbB:acbB): acbB: 0.02217, 0.02771, 0.03895, 0.1582). Asterisks indicate the significance level: * p-value<α=5%, ** p-value<α=1%, *** p-value<α=0.1%.



FIG. 32. Growth and acarbose production of a gtaB overexpression mutant in maltose minimal medium. The sampling time for RNA-isolation is indicated by an arrow.



FIG. 33. Relative transcript amount of gtaB in an overexpression mutant. The RT-qPCR indicates significant increase of gtaB expression compared to the empty vector control (set to a value of 1) (p-value of a two-sided t-test: 0.01295). Asterisk indicates the significance level: * p-value<α=5%, ** p-value<α=1.



FIG. 34. Analysis of the intracellular metabolites of a gtaB-overexpression mutant by LC-MS. Shown are the peak areas of the masses m/z=545 [M-H+] (A), glucose-1P and galactose-1P (m/z=259 [M-H+], B), glucose-6P (m/z=259 [M-H+], C) and UDP-glucose (m/z=565 [M-H+], 0) in an overexpression strain of the gene gtaB. Significant differences compared to the empty vector control were observed for the normalized peak areas of the mass m/z=545 [M-H+] (p-value of a two-sided t-test: 0.01531). All other peak areas are not significantly different according to a two-sided t-test.





BRIEF DESCRIPTION OF THE SEQUENCE IDs

The Sequence Listing associated with this application is filed in electronic format and hereby incorporated by reference into the specification in its entirety.















SEQ





ID





No.
Type
Name
Sequence


















1
DNA
>acbA
GTGCGCGGAATATTGCTGGCCGGGGGAACCGGCTCACGGCTTCGACC




(ACSP50_
GGTGACCTGGGCGGTTTCCAAACAACTGATGCCGGTCTATGACAAACC




3609)
GATGATCTACTATCCGCTGGCCACGCTCGTCAGCTGCGGGATCCGGG





AGATCCTGGTCATCACGACCGAGACCGAGGCCGCCCAGTTCCAGCGG





TTGCTGGGTGACGGCTCGCAGTGGGGCCTGCGTCTGGAGTTCGCCGT





GCAGCAGCGCCCCGGGGGCATCGCCGAGGCCTTCCTCATCGGCGAG





GAGTTCCTGGCCGGTGGGCCGGTGGCGCTCATGCTCGGCGACAACCT





GCTGCACGGGGTGGACTTCCGCCCCTGCGTGCAGCGGGCACGCGAG





ACGGCCGGTGGGCACGTCTTCGGAGTGGCGGTGGCCGACCCGTCGG





CCTACGGGGTGGTCGAGTTCGACGCCGCCGGGCGGGTGCTGTCCATC





GAGGAGAAACCGGTCCGTCCCCGCTCGCCGTACGCGGTTCCCGGCTT





CTACCTCTACGACGCCGATGTGGTCGAGACGGCCCGGTCGCTGCGGC





CCAGCGCCCGCGGGGAGCTGGAGATCACCGAGGTCAACCAGGCCTA





CCTGCGGCGCGGCGCACTCTCGGTGACGCTGCTGGGTCGGGGCGCG





GTCTGGCTCGACACCGGCACCCTGGCCGACTGCATGCGCGCGGTCGA





CTACGTGCGCGCCATCGACGAGGGCCAGGGCATCAAGATCGGCTGTG





TGGAGGAGGCGGCCTGGCGGGCCGGTTTCCTCGACACCGCGCAGCT





GCGTGCCCTCGCCGAGCCGTTGATGAGCAGCGGCTACGGACAGTACC





TGCTGGCTCTGACCGGCGACGGGCTCAGCCGTACCCCGCAGTGGCC





GGCCTTGACCGCCGCCGCCGGGTGA





2
DNA
>acbB
ATGAAAATCTTGGTCACCGGCGGAGCCGGCTTTATCGGGTCCCATTTT




(ACSP50_
GTAACTTCCCTGATCAGTGGCGACATTGCCACACCACAACCCGTGACG




3608)
CAGGTTACGGTCGTCGACAAACTGGGTTACGGAGGCAATCTCAGAAAT





CTCGCCGAAGCGTCGGCGGACCCTCGTTTCAGCTTCGTTCGGGGCGA





CATCTGTGACGAAGGTCTAATCGAGGGGCTGATGGCGCGGCACGACA





CCGTGGCGCACTTCGCCGCCGAGACCCACGTCGACCGCTCGGTGGTC





GCCTCCGGCCCCTTCGTGGCCAGCAACCTGGTCGGCACTCAGGTGCT





ACTGGACGCCGCGCTACGCCACCATATCGGCCGCTTCCTGCATGTTTC





CACCGACGAGGTGTACGGGTCGATCGACACCGGCTCGTGGGCCGAG





GGCCATCCGCTGGCGCCCAACTCGCCGTACGCCGCGAGCAAAGCCG





GGTCCGACCTCCTCGCTCTGGCCTACCACCAGACGCACGGGATGGAC





GTCGTGGTGACCCGCTGCTCGAACAACTACGGGCCCCGGCAATTCCC





GGAGAAAATGATTCCGCTGTTCGTCACCAGGCTGCTCGACGGGCTCG





ACGTACCGGTCTACGGCGACGGCCGCAACATCCGCGACTGGCTCCAC





GTCAGCGACCATTGCCGCGGTCTCGCCCTGGCCCTGGGTGCCGGCC





GGGCAGGCGAGGTCTATCACATCGGCGGTGGGTGGGAGGCGACGAA





TCTCGAATTGACCGAGATCCTCCTCGAGGCGTGCGGCGCCCCGGCTT





CGCGCATATCTTTCGTGACCGATCGCAAAGGTCACGACCGGCGCTATT





CTCTCGACTATTCGAAAATCGCCGGGGAACTCGGTTACCGGCCGCGG





GTCGATTTCACCGACGGCATCGCGGAAACGGTCGCGTGGTATCGCGC





CAACCGTTCCTGGTGGACCTGA





3
DNA
>acbC
GTGAGTGGTGTCGAGACGGTAGGGGTGCACGCGGATGCGCACCGCG




(ACSP50_
ACTCGTGGCAGGTGCGGGCCCAGAAGCAGATCACCTACGAGGTGCGC




3607)
TTCCGGGACGACGTGTTCGGGCTGGACTCCACCGACCTGCTGGAGGC





CGGGGCGGACGGGGCCGGTTCACGGCGGCGGTTCGTGGTGGTGGAC





AGCGCCGTCGACGCCTTGTACGGGTCCCGGATCCGGGAGTACTTCAC





CCATCACGGCATCGATCATTCGATCCTGGTGATGCGGGTGGGCGAGA





CGGTCAAGGACTTCGACACGGCGGGCCGCATCGTCGCCGCGATGGAC





GCCTTCGGACTGGCCCGCCGCCGGGAGCCGATGATCGTCGTCGGTG





GTGGGGTGCTGATGGACGTGGCCGGTCTGGTGGCCAGCCTCTACCGG





CGCGGCACGCCGTTCCTGCGGGTGCCGACGACACTGGTCGGACTGAT





CGACGCGGGTGTCGGCGCGAAGACCGGGGTCAACTTCAACGGCCACA





AGAACCGGCTGGGTACGTACGCCCCGGCTGATCTGACCCTGCTGGAC





CGCCGCTTCCTGGCCACCCTGGACCGGCGCCACCTCAGCAACGGGCT





CGCCGAGATGCTCAAGATCGCGCTGATCAAGGATGCCGAGCTGTTCC





AGCTGCTGGAGCGGCACGGGCGGGTCCTGATCGAGGAACGGTTCCA





GGGCCGTACCGGAACCGGTGACCGGGCCGCCGTCCGGGCCCTGCGC





GCGGCCACCCATGGCATGCTGGAGGAACTCGGCCCCAATCTGTGGGA





GAGCCGGCTGGAACGCAGTGTCGACTACGGGCACACGTTCAGCCCGA





CCATCGAGATGCGCGCGCTGCCGGCTCTGCTGCACGGCGAGGCCGT





GTGTGTGGACATGGCGCTGACCACGGTGCTGGCGTACCGGCGGGGT





CTGCTCGACGTCGCGCAGCGGGACCGGATCTTCGCGGTGATGACCGC





CCTGGGCCTGCCGACCTGGCATCCGCTGCTCACGCCGGAGGTGCTGG





AGGCGGCGTTGCAGGACACCGTCCGGCACCGGGACGGGTGGCAGCG





GCTGCCACTGCCGGTGGGGATCGGGGGTGTCACGTTCGTCAACGACG





TGACGGCCGCCGAGCTGCAGGCCGCCGCGCTGATGCAGCACCGGCT





CGCCGAGGACGCCCTGCTGCTGCGCGCCTAG





4
DNA
>acbS
ATGCACATCATCGAGACGTACTTCGAATGCGGCGGCTTCGACCACCGG




(ACSP50_
TTCATCCAGGGCGGCACCTCGGTCTATCTCTGGCAGCTGTCGCGTGG




3596)
CCTGGCCGACCTGGGACACCGGGTCTCCATCGTCACACCGGCGCACG





GCCGCCTGGACGATCTGCGCCGGCTGCACGAGGTCGAGGACCTGCC





CGGCACCGACGAGTACGAACTGCCGCTGGTGCTCGACCCGCGCGTGT





GGGGCGAACGGTTCCCGGCCCAGATGGACATCGCCCTGCGGACCAC





CGCGCATCGGATCCGGCTGGCGGGCGTGGACCTGTACTTCCTCTCCA





ACGAACTGCTCGATCAGTTGCCGGACCGGTTCTATCCCCCGTACGAGA





GCAAGGGGGTTGATCTGGTCTTCTTCAAGCCGCTCGCCTATCAGGTGG





CGGCCATCCGGTTCATCAGGTCGCACTTCGGTGACCAGCGCGCGATC





GTGCACGCACACGAGCCGTTCTACCACTACCTGATGCCGGCCGCCTT





CGCCGCGGACCCGGCCAAACACGTGGTCAGCACGGTGCAGAGCAACA





TGCCGATCAACAAGTCGGTGTACCGGGCCGAGGTGGCGCGGCTGCTC





GGCTTCCTCGGCGCCCCGAACGCGCTGCCCGCCGACGATCCGGCCG





GCAGCCGTTCGCCGCACACCGTGGCGATGAGCCAGTACCAGCAGCTG





ACCCACCTGCACTACGAATACCCGCCGGACCACGTGCGGGTCTACGA





CCTGGTGGCCGAGCACGCCGACCGGATCGACTTCCTGTCGCCGGGG





CACCGCGACTACTACACCTGCTTCGCCGACACCCCGTTCGCGCAGCT





GTTCGCCACCCTGCCGGTGTCGCGGACGGTACGGCGCAACGCGGAC





AAGACGTTCGTCGGCGGCTGCGCCGTCGGTGACGAGTGGGTGACCG





GCGAGCTGCCCCCGGTCGACCGGGAGAAGGTGCTGGCCGGGCTCGG





CCTGGACCCGGACCTGCCGGCCTTCTACCACAACGCCCGGTACGCGG





TCAACCACAAGGGGCAGGTCGAGCTGATCCGGGCCGTCGACCGGGTG





CTGAGCGGCGGCGTGCGGGCCAGCTTCATCGTGCGCTGCCTCAGCGA





CGCCGGGATCGCCGACCCGCTCTTCCACGAGGTGGTGGCCCGCCAC





CCGGGCCGGGTGAATCTGGAGTGGCACCGGGTGCCGGAGGACCAGC





TGCGGGAGTACGCCCGAGCCGCGGACTTCTGTCTCTTCCCGTCCAAG





TTCGAGATGGACACCTTCCTGATCGCCCAGGGTGAGGCGATGGCTGC





CGGTGCGGTACCGATCGCCACCGCCCAGCTGGGGATGGCGCACTTCG





GTCACGTCGCCGACCCGCTGACCGGGCCGGACGCGGCGACGGCCAC





CGGATTCGCCGTCAACCGCTCGTTCGCCGAGGACGATCCGCTGCTGG





TCCAGGGCCTGACCGAGCAGATCCGCCGGGCCGTCACGCTCTGGAAC





GAGCAGCCCGGCCAGTACCGCCGGTTGTCCGCCAACGCCGTCGCCC





GGGCCCGCGAGTTCACCTGGCGGCGGGCGGCCCAGGCGCACGAGGC





CGCGTTCGCCGGGGTGTGGGCCGGCCGTACCCCCCGCCTGCCGGTC





GGTGACCTGCTGCGGTTCGGCTGGTTCGACGAGCTGCCCGCGGACGC





CTGGACGCTGCACCGCGACGAGATCGCGGAGGTGGCCCTGGCCCAC





GGCGACGCCGACGCCTACCTGCGCTGCCGGCCCGACGACCTCGACG





CCCTGGCGGCACTCTTCGAGCGGGCCTGGGCCCGGGCCGACTTCCC





GGCCTGCGCGCGGACCGTAGAGCTGGCCGAGGAGCACCGGCAGGAG





CGGGTGCCGCAGTGGCGGGCCCGGCTCGCCGGCCGCGGCCGCATC





GACCGCGACGGTCGGCTGCACTACCGTCCGCCGTCCGCCGAACGGG





TCGAACTGGTCTTGCCCGACCTGGCCGAACCCCTGCGCGGAACGGTC





ACCGTGACCGCGATGGCTCCGACCGGCGACACCTTCACCGGACAGCT





GCCGGCCGGAACCCGGCGTGCCGACCTGCTGCTCACCCTCAGTGACG





GGCGCACCGTCTGGGACGAGGTGACGGCATGA





5
DNA
>acbW
ATGCCCGGGTACGCCCGGCATGCCCGGCCGGACGGCACGACCGGCA




(ACSP50_
TGATCGTCGCCGAGCACCTCAGCAAGCACTTCAAGCGCTACCGGCGC




3593)
GAGCCGGGTCTGCGGGGCAGCCTGCGAACCATGTTCTCGGCCCGGTA





CGACGTGGTCCGGGCCGTCGACGACATCAGCTTCGAGGTCCCGTCCG





GTGTCAAGATCGCCTACATCGGGGCGAACGGCGCGGGCAAGTCCACC





ACGATCAAACTCCTGACCGGCATCATGCGCCCGACCACCGGGCGGGT





CCGGGTCGACGGCCTCGACCCGCACCGGCAGCGCACCCGGGTCGCC





GGCCGGATCGGCGTGGTCTTCGGCCAGCGCAGCCAGCTCTGGTGGG





ATCTGCCGGTCCTCGACTCGTTCCGCATCCTGCGGCACGTCTACGAG





GTGCCGCAGGCGGTGTACGACCGGAACATGCGCCTGTTCCGGGACCG





GCTGGACCTCGGCGCCCTCGGCAACACCCCGGTCCGCCAGCTGAGC





CTGGGCCAGCGCATGCGGGCCGAGATCGCCGCCTCGCTGCTGCACG





ACCCGGCCGTGGTCTTCCTCGACGAACCCACCATCGGCCTGGACCTG





GTCCTCAAGCAGGCGGTCCGGGACCTGATCAACCACATCCACGCCGA





ACTGGGCACCACGGTCATGCTGACCAGCCACGACATCGGCGACATCA





CCAGCATCTGCGATCAGGCGCTGGTCGTGGACCGCGGGACGATCGTC





CACCAGGGAACGATGCGGGACCTGCTGCGGTCGGTGGACACCCGGG





CGGTCACCTTCGAGTACGCCGCCGGCAGCGTCTCCGAGGCCGCCGC





GCTGCGCATCATCACCGAAGGACTGCCCGAGGTGGACGCCACTCCGG





CCGAGTCCGGCCGGATCCGGGTCGAGTTCCCGGTGGACCGCTGGTC





GGCCCGGCAGGTGATCGCCTTCCTGCTGGACCGGTTCGACCTGAGCG





ACGTGCTGGTGCCGGACGCCGATCTGGAGACACTGCTGCGCCGCATC





TACGCCGGGTCGCGCCCGGAGCCGGTCACCGCCGGGGACGGCGCAT





GA





6
DNA
>acbX
ATGATCCGCGCCGCGCGCCGGTACGCGCCGTTCGCCCTCGCCGGACT




(ACSP50_
GCACGCCGTCACCCGTTACCGCTCGACCATCGTCCTGAGCGCACTCA




3592)
CGGCGGCTGCGGCCACCTCGTTGCAGGTGTTCCTGTGGCGAGCCGTC





TACGCCGGCGGACCGGCACCGGCCGGCCTCCCGTTCGCACAGCTCA





CCTCGTACATCGTGCTCGCGCAGGTGCTCGGGATGCTGCACACCAAC





CGGATCGACGAGATGATCGCCGGCGAGGTGTACCGCGGGGACATCG





CGGTCTCCCTGGTACGCCCGGCGAACTACGCGCTCAGCTGTCTGGCG





GTGAACCTGCCGACCGCCGCGCTCAGTGCGCTGCTGGCCGGCGCCC





CGGTGCTCGCCGGTTTCGCGATGTTCGCGTCGCTGCCCGCTCCCCCG





CCCGCCAACCTGCTGCTGTTCGCCGTCGCGCTGCTGCTCTCGGTGAT





CCTCGCCTTCGAGATCAACTTCCTGGTGGGTCTCGCCGCCTTCGTCAC





GACCAACACCTGGGGCATCCGTACGATCAAGAACGCGCTCGTCGCCT





TCCTGGCCGGCCAGGTCGTCCCGCTCGCGCTGTTCCCGGACGGCGTG





GCCCGGCTGCTGCGGCTGCTGCCGTTCCAGGGCCTGATCGACAGCCC





GTTGCGGCTGCTGCTCGGCGGCTACTCCGGCGGTTCCGGCGCCGCT





GCCATCCTCGGTGTCCAGGCGCTCTGGGCGGTACTGCTGTACGGCGT





GCTGGCCCTGGCCTGGAACCGGTCGCTGCGCAGGGTGGAGGTGCTC





GGCGGATGA





7
DNA
>acbY
atgaccgtctccacggcgcgccggtacctgcgcctcacggcggtgctgtgcggggcgagcctgcaccg




(ACSP50_
gctcaccgcgtaccggatggacttcctcatcggggcggccagcttcgtcatccggatcgcctgccagatc




3591)
gccctgatcggggtgatcttccagtacgttccggcgctcggcggctggacccgccagcaggcgctgttcc





tgctcgggttctccctgctgccccgcgggctggaccggctcttcaccgaccagctgtggatcctggcctgg





cagctggtgcgcaccggcgacttcttccgctacctgatccggccggtgaacccgttctacgcgctgctgtc





cgaacggttcctctatccggacgggttcggggagctggccaccggcatcgccatcgtggtcaccgcggc





cgggacgatggacctgcacctgaccgtggcacagtggctgctgttgctgcccctggtcctcggcggcgc





cctgatccacaccttcctcaaggcgttcctggcctccctgtcgttctggatgaccagcagcctcaacgtgat





ggtggcggtcaaccagctcagcgagttcaccgcgtacccgctcaacctctaccacccggtgctgcgcgg





ggtgctcacctgggtgctgccgttcgcgttcaccgcctacctaccggtgcgctacctgctcaccggggacg





ccgggccgctgctgtggatgctgccggtcaccacgctcaccgtcctgctggggtacggcaccttccggct





cgggctgcggcgctacgagatgcccggcagctga





8
DNA
>gtaB
ATGACGACGAACGCGCAAGGGTCGGGCAAGCGCGCGGTGAAAGCAG




galU
TGATTCCGGCGGCCGGCCTAGCCACGCGTTTCCTGCCTGCCACCAAA




(ACSP50_
GCCGTTCCGAAAGAGCTGCTGCCGGTCGTCGACCGGCCGGTCCTGCA




7820)
GTACATCGTCGAGGAGGCCGCCGCGGCCGGCATCACCGACGTGCTG





CTGGTGACCGGGCGTGGCAAGACCTCGATGGTCGACCACTTCGACCG





TCGCCCCGACGTGGAGCAGCGGCTGGAGGAGAAGGGCGACACCGAG





CGGCTCGCCGCCGTCCGGCGCACCAGTGAGCTGGCCGACATCTACAC





CTGCCGACAGGGGGAGCCGCTCGGCCTCGGCCATGCCGTCGGGACC





GCCGCCTCGCACGTCGGGGACAACCCGTTCGCGGTGCTGCTCGGGG





ACGAGTTCGTCGAGGAGGGCAGCCCGCTGCTGCCCGACATGCTCGAC





CTGCAGGCCCGCACCGGCGGCATCGTGCTCGCCTTCATCGAGGTCAC





CCCGGAGGAGACGTCGCGCTACGGGATCGCCTCGGTGCGGGAGTCC





GACCTGGGCGAGGGCGTGGTCGAGGTGACCGGCCTGGTGGAGAAGC





CGTCGCCGGAGGAGGCGCCGAGCAACCTTGCCGTGGTGGGGCGGTA





CGTGCTGCCTGGCAGGATCTTCGAGACGATCGCCGGCACCAAGCCGG





GCAGCGGGGGCGAGATCCAGCTGACCGACGCGATGGCGACGCTGCT





GGCCGAGGGCACCCCGGTGCACGGCATCGTCTACCGCGGTGTCCGG





TACGACACCGGCCAGCCGCTGGGCTACCTGCAGACCGTCGTCCAGCT





CGCGGCTCAGCGTCCCGACCTGGGTGCCGAGTTCCGGGCCTGGCTCA





CCGACTTCGTCGGTGGTCAGAAGGGATGA





9
DNA
>cgt
ATGAATCGCACCACCGTTCGGGCCGGCGTGCTGGCCACCGCCCTGAT




(ACSP50_
CAGCGGCGTGCTCGGGGTGGCCGGCCCGGCGCTCGCCGCCCCGGTC




5024)
ACCGACGCGGCGCCGGTCGCCGCCGCCGGCACCGCCGTCGCGCCGA





TCGCCGCGACCTTCAACGTGACCGCCGGGTTCACCAGCTGGGGTCAG





AACGTCTACGTCGTCGGCAGCATCCCGGCGCTCGGCTCCTGGGACGT





CTCCAAGGCGGTGCCGCTGACCACCACGAGCAGCGCCTTCCCGACCT





GGACCGGGAGCGTGGCGCTGCCGGCGAACACGTACACCGAGTTCCA





GTACGTGGTGAAGAACGCCGACGGCAGCGTCGCCCGCTGGGAGAAG





GGTTTCCAGCAGAACCGCACCACGATCACCCCGCCGACCGGCACCTA





CGTCACGCACGACACCTTCGGCGCGTACTGA





10
DNA
>crtl
ATGATGAAACCCCCCACCCCCTGGAGCCGCGGCGTGCGCACTGTTAC




(ACSP50_
CGGACCCACCGATCGTGTCGTGATAGTGGGGGCCGGCCTGGCCGGC




0147)
CTCTCCTGCGCCTTGCACCTGGCCGCAGCCGGGCGGCAGGTCACCGT





CGTCGAGCGGGAGCCGGTGCCGGGCGGCCGCGCCGGGCGCCTCTC





GGTCGGCGGATACGACTTCGACACCGGCCCGACCGTGCTGACCATGC





CGGAACTGATCGCCGAGCCGCTCGCCGCGGTCGGCGAGAATCTCTCC





GACTGGCTGGAGCTGACCCCGCTCGACCCGGCCTACCGGGCGTACTA





CCCGGACGGCTCCACGCTGGACGTCCGCACCGACACCACCCGGATG





GCGGCCGAGATCGCCCAGGTCTGCGGCGCCCGCGAGGCCGACGGCT





ACCTGCGGTTCGTCGACTACACCCGGCGGCTCTGGCAGCTGGAACGG





GACCACTTCATCGACCGGAACCTGGACAGTCCGCTCGACCTGCTCAAC





CTCAACCTGCTGAAGCTGCTCGGGATGGGCGCTTTCGGTCGCCTGCA





GCCGAAGATCAACGAGTTCTTCCGCGATCCGCGGACCCAGCGGATCT





TCTCGTTCCAGGCGATGTACGCCGGTCTCGCCCCGCACGACGCGATG





GCCATCTACGCGGTGATCGCCTACCTCGACTCGGTCGCCGGGGTGTA





CTACCCCAAGGGCGGCATGCACGCCGTCCCCAAGGCGCTGGCCGGC





GCCGCCGAGAAGCACGGGGTCACCTTCCGTTACGACACGACGGTCGA





GCGGGTGCTCACCCAGCACGGCCGGGCGACCGGGGTGGTGACCGTC





GGCGGGGACGTGATCGAGGCGGACACCGTCGTACTCAATCCCGACCT





GCCCATCGCGTACCGCGACCTGCTGCCTGCCCGGAACAGCCGCAACC





TGCGCTTTTCGCCCTCCTGCGTGGTACTCCACATCGGATCGTCACAGC





GGTATTCGAAGATCGCACACCACAACATCCACTTTGGTACGACGTGGC





GCCGCACCTTCGACGAAGTGATCAACCGTGGGCTGCTGATGAGCGAC





CCGTCACTGCTGGTCACCAATCCCACGCACACCGACCCCTCTGCCGC





GCCCGACGGCAAACAGACCTACTACGTGCTGGCGCCCGCCCCGAACC





TCGTCTCCGGTCCGATGAACTGGCGCGGCGGCCTCGCCGAACGGTAT





GCCGACGAGCTGCTGCGTACCCTGGAGCAGCGCGGCTACATCGGCTT





CCGGGACGGGGTCGAGGTCGAACGGATCATCACGCCGGCCGACTGG





GCCGACGACGGGATGGCGGCCGGCACGCCGTTCGCCGCCGCGCACA





CCTTCGCCCAGACCGGCCCGTTCCGGCCGGCGAACCTGCACCCCACG





CTGCCGAACGTGGTCTTCACCGGTTCGGGCACACAACCCGGGGTCGG





CGTGCCGATGGTGCTCATCTCCGGGAAGCTGGCCGCGAGCCGGATCA





CACAGGGAGCCTCATGA





11
DNA
>merR
GTGGCCGGTGAGGCGTTGAGCGCCGAGATCCCCACCTCGCCGGGCA




(ACSP50_
GCTCGGTCGCCTCCTCGCACGACATCCCGGCCACAGCCGGTCCCGGC




0145)
GCCGTCCGGACCGGCCCGGTGGCTGCCGCGCCCGGTGGCCCGAGCG





ATACGCCCCTGACCGACGCGACAGCTGCCGCGTCGGGTGCCGCGGA





CGACGCCTCCCGGGCCCGCCCGGCGACCGCCACGGACGACGCCTCC





CGCACCGGCCCGGCGACCGCCGCGACGGATTCTCCGGACGACGCCG





TCCGGACCGGCGTGGCAGATGCCGCGCCGGCCGGGCGGGCGGGCG





ATGTGGCGTTGAGTGCCGGGGCGGCCGCGCGGCGGCTGGGAGTGGC





GGTCACGACCCTGCGCACCTGGCACCAGCGGTACGGGCTCGGGCCG





AGCCGGCACGAGCCCGGACATCACCGGCGGTACACCGCCGAGGACA





TGGACCGGCTGCAGGTGATGCAGCGGCTCACCACTCAGGGCGTGGC





GCCCGCCGAGGCCGCCGCCTGGGCGCGGTCCAGGCCCCTCACCCCA





CCGGAGCCCGGCGCGGCGCTGTACGACCCCACCGCCGTGGCGTCGC





CACCCACCCCGGCCGCTCCCGGACAGCCCCCGGTCGGCCCCGCCGG





CCGGGGCACCCGCCCGACCCGCGGACCGGCCCCGGCCGCTCGCGG





GCTGACCCGGGCCGCGATGCGGCTCGACGTGCGCGGCATGCGCGAC





ATCCTCTGCAGCACGCTGCACGACCGCGGCGTGATACCCGCCTGGAC





CGAGGTGATGGTCCCGGCTCTGGCCGCGATCGGCGACCGGTACGAG





GCCACTCGGCGTTTCGTCGAGGTCGAACACCTGCTGTCGCGCGCCGT





CACCGAAATCCTCGCCTCGGTCCCACACCCCGCCGGCTCTCCCCGGG





TGCTGCTCGCCGCCGCCGACGAGGAACAGCACACACTGCCCCTGGAG





GCCCTGGCCGCCGCCCTGGCCGAGGGAGGCGTGCCGAGCCGTCTGT





TCGGCGCCCGGGTGCCGTCACAGGCCCTGCTGGACGCCATCGCCCG





CACCGGCCCGGCTGCCGTCGTGCTCTGGTCGCAGCGCCCGGCCACC





GGCATCGTCACCCAGCTGACCCGGGTCCGCGACATCCCGCACCCGCC





GCTGGTCATCGCCGCCGCCGGCCCCGGCTGGCCGCATGACCTGCCTT





CCGGGATCACCCGCCTGACCGGCCTCACCGAGGCCGTCCACCTGCTC





GCCACGGTCTAG





12
PRT
>AcbA
MRGILLAGGTGSRLRPVTWAVSKQLMPVYDKPMIYYPLATLVSCGIREILVI




(ACSP50_
TTETEAAQFQRLLGDGSQWGLRLEFAVQQRPGGIAEAFLIGEEFLAGGPV




3609)
ALMLGDNLLHGVDFRPCVQRARETAGGHVFGVAVADPSAYGVVEFDAAG





RVLSIEEKPVRPRSPYAVPGFYLYDADVVETARSLRPSARGELEITEVNQA





YLRRGALSVTLLGRGAVWLDTGTLADCMRAVDYVRAIDEGQGIKIGCVEE





AAWRAGFLDTAQLRALAEPLMSSGYGQYLLALTGDGLSRTPQWPALTAA





AG





13
PRT
>AcbB
MKILVTGGAGFIGSHFVTSLISGDIATPQPVTQVTVVDKLGYGGNLRNLAEA




(ACSP50_
SADPRFSFVRGDICDEGLIEGLMARHDTVAHFAAETHVDRSVVASGPFVA




3608)
SNLVGTQVLLDAALRHHIGRFLHVSTDEVYGSIDTGSWAEGHPLAPNSPY





AASKAGSDLLALAYHQTHGMDVVVTRCSNNYGPRQFPEKMIPLFVTRLLD





GLDVPVYGDGRNIRDWLHVSDHCRGLALALGAGRAGEVYHIGGGWEATN





LELTEILLEACGAPASRISFVTDRKGHDRRYSLDYSKIAGELGYRPRVDFTD





GIAETVAWYRANRSWWT





14
PRT
>AcbC
MSGVETVGVHADAHRDSWQVRAQKQITYEVRFRDDVFGLDSTDLLEAGA




(ACSP50_
DGAGSRRRFVVVDSAVDALYGSRIREYFTHHGIDHSILVMRVGETVKDFD




3607)
TAGRIVAAMDAFGLARRREPMIVVGGGVLMDVAGLVASLYRRGTPFLRVP





TTLVGLIDAGVGAKTGVNFNGHKNRLGTYAPADLTLLDRRFLATLDRRHLS





NGLAEMLKIALIKDAELFQLLERHGRVLIEERFQGRTGTGDRAAVRALRAA





THGMLEELGPNLWESRLERSVDYGHTFSPTIEMRALPALLHGEAVCVDMA





LTTVLAYRRGLLDVAQRDRIFAVMTALGLPTWHPLLTPEVLEAALQDTVRH





RDGWQRLPLPVGIGGVTFVNDVTAAELQAAALMQHRLAEDALLLRA





15
PRT
>AcbS
MHIIETYFECGGFDHRFIQGGTSVYLWQLSRGLADLGHRVSIVTPAHGRLD




(ACSP50_
DLRRLHEVEDLPGTDEYELPLVLDPRVWGERFPAQMDIALRTTAHRIRLAG




3596)
VDLYFLSNELLDQLPDRFYPPYESKGVDLVFFKPLAYQVAAIRFIRSHFGD





QRAIVHAHEPFYHYLMPAAFAADPAKHVVSTVQSNMPINKSVYRAEVARL





LGFLGAPNALPADDPAGSRSPHTVAMSQYQQLTHLHYEYPPDHVRVYDL





VAEHADRIDFLSPGHRDYYTCFADTPFAQLFATLPVSRTVRRNADKTFVG





GCAVGDEWVTGELPPVDREKVLAGLGLDPDLPAFYHNARYAVNHKGQVE





LIRAVDRVLSGGVRASFIVRCLSDAGIADPLFHEVVARHPGRVNLEWHRVP





EDQLREYARAADFCLFPSKFEMDTFLIAQGEAMAAGAVPIATAQLGMAHF





GHVADPLTGPDAATATGFAVNRSFAEDDPLLVQGLTEQIRRAVTLWNEQP





GQYRRLSANAVARAREFTWRRAAQAHEAAFAGVWAGRTPRLPVGDLLR





FGWFDELPADAWTLHRDEIAEVALAHGDADAYLRCRPDDLDALAALFERA





WARADFPACARTVELAEEHRQERVPQWRARLAGRGRIDRDGRLHYRPP





SAERVELVLPDLAEPLRGTVTVTAMAPTGDTFTGQLPAGTRRADLLLTLSD





GRTVWDEVTA





16
PRT
>AcbW
MPGYARHARPDGTTGMIVAEHLSKHFKRYRREPGLRGSLRTMFSARYDV




(ACSP50_
VRAVDDISFEVPSGVKIAYIGANGAGKSTTIKLLTGIMRPTTGRVRVDGLDP




3593)
HRQRTRVAGRIGVVFGQRSQLWWDLPVLDSFRILRHVYEVPQAVYDRNM





RLFRDRLDLGALGNTPVRQLSLGQRMRAEIAASLLHDPAVVFLDEPTIGLD





LVLKQAVRDLINHIHAELGTTVMLTSHDIGDITSICDQALVVDRGTIVHQGT





MRDLLRSVDTRAVTFEYAAGSVSEAAALRIITEGLPEVDATPAESGRIRVEF





PVDRWSARQVIAFLLDRFDLSDVLVPDADLETLLRRIYAGSRPEPVTAGDG





A





17
PRT
>AcbX
MIRAARRYAPFALAGLHAVTRYRSTIVLSALTAAAATSLQVFLWRAVYAGG




(ACSP50_
PAPAGLPFAQLTSYIVLAQVLGMLHTNRIDEMIAGEVYRGDIAVSLVRPANY




3592)
ALSCLAVNLPTAALSALLAGAPVLAGFAMFASLPAPPPANLLLFAVALLLSVI





LAFEINFLVGLAAFVTTNTWGIRTIKNALVAFLAGQWPLALFPDGVARLLRL





LPFQGLIDSPLRLLLGGYSGGSGAAAILGVQALWAVLLYGVLALAWNRSLR





RVEVLGG





18
PRT
>AcbY
MTVSTARRYLRLTAVLCGASLHRLTAYRMDFLIGAASFVIRIACQIALIGVIF




(ACSP50_
QYVPALGGWTRQQALFLLGFSLLPRGLDRLFTDQLWILAWQLVRTGDFFR




3591)
YLIRPVNPFYALLSERFLYPDGFGELATGIAIVVTAAGTMDLHLTVAQWLLL





LPLVLGGALIHTFLKAFLASLSFWMTSSLNVMVAVNQLSEFTAYPLNLYHP





VLRGVLTWVLPFAFTAYLPVRYLLTGDAGPLLWMLPVTTLTVLLGYGTFRL





GLRRYEMPGS





19
PRT
>GtaB
MTTNAQGSGKRAVKAVIPAAGLATRFLPATKAVPKELLPWDRPVLQYIVE




GalU
EAAAAGITDVLLVTGRGKTSMVDHFDRRPDVEQRLEEKGDTERLAAVRRT




(ACSP50_
SELADIYTCRQGEPLGLGHAVGTAASHVGDNPFAVLLGDEFVEEGSPLLP




7820)
DMLDLQARTGGIVLAFIEVTPEETSRYGIASVRESDLGEGWEVTGLVEKP





SPEEAPSNLAVVGRYVLPGRIFETIAGTKPGSGGEIQLTDAMATLLAEGTP





VHGIVYRGVRYDTGQPLGYLQTWQLAAQRPDLGAEFRAWLTDFVGGQK





G





20
PRT
>Cgt
MNRTTVRAGVLATALISGVLGVAGPALAAPVTDAAPVAAAGTAVAPIAATF




(ACSP50:
NVTAGFTSWGQNVYVVGSIPALGSWDVSKAVPLTTTSSAFPTWTGSVALP




5024)
ANTYTEFQYVVKNADGSVARWEKGFQQNRTTITPPTGTYVTHDTFGAY





21
PRT
>Crtl
MMKPPTPWSRGVRTVTGPTDRVVIVGAGLAGLSCALHLAAAGRQVTVVE




(ACSP50_
REPVPGGRAGRLSVGGYDFDTGPTVLTMPELIAEPLAAVGENLSDWLELT




0147)
PLDPAYRAYYPDGSTLDVRTDTTRMAAEIAQVCGAREADGYLRFVDYTRR





LWQLERDHFIDRNLDSPLDLLNLNLLKLLGMGAFGRLQPKINEFFRDPRTQ





RIFSFQAMYAGLAPHDAMAIYAVIAYLDSVAGVYYPKGGMHAVPKALAGA





AEKHGVTFRYDTTVERVLTQHGRATGWTVGGDVIEADTWLNPDLPIAYR





DLLPARNSRNLRFSPSCVVLHIGSSQRYSKIAHHNIHFGTTWRRTFDEVIN





RGLLMSDPSLLVTNPTHTDPSAAPDGKQTYYVLAPAPNLVSGPMNWRGG





LAERYADELLRTLEQRGYIGFRDGVEVERIITPADWADDGMAAGTPFAAA





HTFAQTGPFRPANLHPTLPNVVFTGSGTQPGVGVPMVLISGKLAASRITQ





GAS





22
PRT
MerR
MAGEALSAEIPTSPGSSVASSHDIPATAGPGAVRTGPVAAAPGGPSDTPL




(ACSP50_
TDATAAASGAADDASRARPATATDDASRTGPATAATDSPDDAVRTGVAD




0145)
AAPAGRAGDVALSAGAAARRLGVAVTTLRTWHQRYGLGPSRHEPGHHR





RYTAEDMDRLQVMQRLTTQGVAPAEAAAWARSRPLTPPEPGAALYDPTA





VASPPTPAAPGQPPVGPAGRGTRPTRGPAPAARGLTRAAMRLDVRGMR





DILCSTLHDRGVIPAWTEVMVPALAAIGDRYEATRRFVEVEHLLSRAVTEIL





ASVPHPAGSPRVLLAAADEEQHTLPLEALAAALAEGGVPSRLFGARVPSQ





ALLDAIARTGPAAVVLWSQRPATGIVTQLTRVRDIPHPPLVIAAAGPGWPH





DLPSGITRLTGLTEAVHLLATV





23
DNA
>dxs
ATGAGCGACTCCCCTTCGACCCCGGCCGGCCTGCTGGCGAGCGTCAC




(ACSP50_
CGGTCCCGGTGCTCTCAAGCGACTGTCCGCGGAGCAGCTGACCCTGC




7096)
TCGCGGCCGAGATCCGTGACTTCCTCGTGGCCAAGGTGTCGAAGACC





GGGGGGCACCTCGGACCGAACCTGGGCGTGGTCGAGATGACCCTCG





CCATGCACCGGGTCTTCGACTCGCCGCGCGACAAGATCCTCTTCGACA





CCGGCCACCAGGCGTACGTGCACAAGATCGTCACCGGCCGGCAGGAC





GGTTTCGACCTGCTCCGCCAGCGGGGTGGCCTGACCGGCTACCCGAG





CCAGGCGGAGAGCGAGCACGACCTCATCGAGAACTCGCACGCCTCCA





CCGCGTTGTCCTACGCCGACGGCCTGGCCAAGGCGTTCGCGCTGCGC





GGCGAGGACCGGCACGTGGTGGCCGTGGTCGGCGACGGCGCGCTCA





CCGGCGGCATGTGCTGGGAGGCGCTCAACAACATCGCCGCCACGAAG





AACAGGCTGGTCATCGTCGTCAACGACAACGGTCGGTCGTACGCGCC





GACGATCGGCGGCCTGGCCGACCACCTCTCCACGCTGCGGCTCAACC





CCGGCTACGAGAAGGTGCTCGACCTGGTCAAGGACGCGCTCGGCTCG





ACCCCGCTGGTCGGAAAGCCGGTCTTCGAGGTGCTGCACGCGGTCAA





GCGCGGGATCAAGGACGCGGTCAGCCCGCAGCCGATGTTCGAGGAC





CTCGGCCTGAAGTACATCGGGCCGGTCGACGGTCACGACCAGCAGGC





GATGGAGTCCGCGCTGCGCCGGGCCAAGGGGTTCAACGCGCCGGTG





ATCGTGCACGCGGTGACCCGCAAGGGCTACGGCTACCGTCCCGCCGA





GCAGGACGAGGCGGACTGCCTGCACGGCCCGGGCGCCTTCGACCCG





CAGACCGGCGCGCTCACCGCCAAGCCGTCGCTCAAGTGGACCAAGGT





CTTCGCCGAGGAGCTGGTGAAGATCGCCGACGAACGCCCCGACGTGG





TGGGCATCACGGCCGCCATGGCCGAGCCGACCGGCATCGCCGCTCTC





GCCAAGAAGTACCCCGACCGGGCGTACGACGTGGGCATCGCCGAGCA





GCACGCCGCGACCAGCGCCGCGGGCCTGGCGATGGGCGGCCTGCAC





CCGGTGGTGGCGGTCTACGCCACCTTCCTGAACCGCGCTTTCGACCA





GGTGCTGCTGGACGTCGCGATGCATCGGCTGCCGGTGACCTTCGTGC





TGGACCGGGCCGGCATCACCGGGCCGGACGGCCCCAGCCACTACGG





CATCTGGGACATGAGTGTCTTCGGCGCCGTCCCCGGCCTGCGCATCG





CCGCCCCGCGGGACGCCGCCACCCTGCGCGAGGAACTGCGCGAGGC





GGTCGCGGTCGACGACGGCCCGACCATCGTGCGGTTCCCGACCGGT





GCGGTCGCCGCGGACACCCCGGCGGTGCGCCGGGTCGGTCAGGTCG





ACGTGCTGCGCGAGGCGGAGAAGAAGGACATCCTGCTGGTCGCGGTC





GGCTCGTTCGTCGGCCTCGGGCTGGACGCCGCCGAGCGGCTCGCCG





AGCAGGGGTACGGCGTGACCGTGGTCGACCCGCGCTGGGTGCGCCC





GGTGCCGATCGAGCTGACCGGCCTGGCCGCCCAGCACCGCCTGGTG





GTGACCCTGGAGGACGGGATCCGCGCCGGTGGTGTCGGTGACGCGG





TGGCCGCCGCGCTGCGCGACGCCGGGGTGCACGTGCCGCTGCGCGA





TTTCGGCGTGCCGGCCGGTTTCCACCCGCACGGCACCCGGGCCGAGA





TCCTCGCCTCGCTGGGTCTGACCGCGCAGGACGTCGCGCGGGACGT





GACCGGCTGGGTGTCCGGCCTGGACGCCGGCACGTCGGTGGCGGCC





CCGGCGATCTGA





24
DNA
>ispG
GTGACCGCGATCAGTCTCGGAATGCCGGCCGTCCCCCCGCCGCCGCT




(ACSP50_
GGCCCCGCGCCGCCAGAGCCGGCAGATCAACGTCGGAGGAGTCCTG




7248)
GTCGGCGGGGGCGCCCCGGTCAGCGTCCAGTCGATGACCACCACCC





TCACCTCCGACGTCAACGCGACCCTGCAGCAGATCGCCGAGCTGACC





GCGGCCGGCTGCCAGATCGTCCGGGTCGCCGTGCCGTCCCAGGACG





ACGTCGAGGCGCTGCCGGCGATCGCCAAGAAGTCGCAGATCCCGGTG





ATCGCCGACATCCACTTCCAGCCCAAGTACGTGTTCGCCGCGATCGAC





GCGGGCTGCGCGGCGGTCCGGGTCAATCCGGGCAACATCCGCCAGT





TCGACGACAAGGTCAAGGAGATCGCCCGGGCCGCGTCCGACGCCGG





CGTGCCGATCCGGATCGGGGTCAACGCCGGCTCGCTCGACAAGCGG





CTTCTCGAGAAATACGGCAAGGCCACCGCCGAGGCGCTGGTGGAGTC





GGCGCTCTGGGAGTGCTCGCTGTTCGAGGAGCACGGTTTCCGGGACA





TCAAGATCTCGGTCAAACACAACGATCCGGTCGTGATGATCCGCGCCT





ACCGTCAGCTCGCCGAGCAGTGCGACTACCCGCTGCACCTGGGCGTG





ACCGAGGCCGGGCCGGCCTTCCAGGGCACGATCAAGTCGGCGGTGG





CGTTCGGCGCGCTGCTCGCCGAGGGGATCGGCGACACCATCCGGGT





CTCGCTGTCCGCGCCGCCGGTCGAGGAGATCAAGGTCGGGCAGCAG





ATCCTGGAGTCGCTCGGCCTGCGCGAACGCGGCCTGGAGATCGTCTC





CTGCCCGTCCTGCGGGCGGGCCCAGGTCGACGTCTACACGCTGGCC





GAGCAGGTGACCGCGGCGCTCGACGGGTTCCCGGTGCCGCTGCGAG





TGGCCGTGATGGGCTGCGTCGTGAACGGGCCCGGGGAGGCTCGCGA





GGCCGACCTCGGGGTCGCCTCCGGCAACGGCAAGGGGCAGATCTTC





GTCAAGGGCAAGGTGATCAAGACGGTGCCGGAGGCGGTGATCGTCGA





GACGCTGGTCGAGGAGGCGCTGCGGCTCGCCGACGAGATGGGCGCG





GAGCTGCCCGACGAGCTGCGCGAGCTGCTGCCCGGTCCCACGGTCA





CCGTGCACTAG





25
DNA
>dxr
ATGCGTGAGCTTGTGCTGCTGGGGTCGACCGGGTCCATCGGCACCCA




(ACSP50_
GGCCATCGATATCGTCCGGCGCAACCCGGAGCTGTTCCGGGTGGTCG




7250)
CGATCGGGGCCGGGGGTGGCAACGTCGCGTTGCTCGCGGCGCAGGC





GCTGGAGCTGGGCGTCGAGGTGGTCGGGGTGGCCCGGGCCTCGGTC





GTGCAGGATCTGCAGCTGGCCTTCTACGCCGAGGCGCAGAAGCGTGG





CTGGTCGTCCGGCGACTTCAAACTGCCGAAGATCGTGGCCGGGCCGG





ACGCGATGACCGAGCTGGCCCGCTGGCCGTGTGACGTCGTTCTCAAC





GGGGTGGTCGGCAGCCTCGGCCTGGCGCCGACCCTGGCCGCTCTGG





AGTCCGGGCGGATCCTTGCGCTGGCCAACAAGGAGTCGCTGGTCGCC





GGCGGCCCGCTGGTCCGGCGGATCGCCAAGGACGGGCAGATCGTCC





CGGTCGACTCGGAGCATTCGGCGCTGGCCCAGTGCCTGCGCGGCGG





GCGGGCCGCGGAGGTGCGCCGGCTGGTGCTGACCGCCAGCGGGGG





AGCCTTCCGCGGGCGGCGGCGCGCGGAGCTGACGAACGTCACCCCC





GAGGAGGCGCTCAAGCACCCGACCTGGGACATGGGGCCGGTCGTCA





CGATCAACTCGGCGACCATGGTGAACAAGGCGCTGGAAGTGATCGAG





GCGCACGAGCTGTTCGGCGTGCCGTACGACGACATCGCGGTGATGGT





GCACCCGCAGTCGGTGCTGCATTCGCTGGTCGAGTTCACCGACGGCT





CGACGCTGGCCCAGGCCAGCCCGCCGGACATGCGGCTGCCGATCGC





GCTGGCGCTGGCCTGGCCGGACCGGGTGCCGGGGGCGGCCGCCGC





GGTGGACTGGACGCTGGCGCACAACTGGGAGCTGCGACCGCTGGAC





GACGAGGCGTTCCCGGCGGTCGAGCTGGCCAAGGCGGCCGGCCGGT





ACGGTCGCTGCCGTCCGGCGATCTTCAACGCCGCCAACGAGGAGTGT





GTGGCCGCTTTCGCCGCCGGTCGGCTACCTTTCTTGGGCATCGTCGA





CACCCTGGAACGGGTGCTCGCGGCGGCCCCGGATTTCGCGGAGCCG





AGTACCGTCGATGACGTGCTGGCCGCAGAATCCTGGGCGCGTGCCCA





GGCACAGCGGACGATCGCGACTGTGGCTGAAGGAGCCTGA





26
DNA
>ispH
GTGTTGCTCGCCAAGCCGCGTGGTTACTGCGCCGGTGTCGACCGCGC




(ACSP50_
CGTGCAGACCGTCGAGGAGGCGCTGAAACTCTACGGCGCCCCGGTCT




7707)
ACGTGCGTAAGCAGATCGTGCACAACAAGCACGTGGTCAGCACGCTG





GAGGCCCGCGGCGCGATCTTCGTCGAGGAGAACTACGAGGTGCCCGA





GGGCGCCACCGTGGTGTTCTCCGCGCACGGCGTCGCCCCCGAGGTG





CACGACCAGGCCCGCGAGCGCCGGCTCAAGGCGATCGACGCGACCT





GCCCGCTGGTCACCAAGGTGCACCACGAGGCGAAACGGTTCGCCGCC





GAGGACTACGACATCCTGCTGATCGGTCACGAGGGGCACGAGGAGGT





CATCGGCACCTCCGGCGAGGCCCCGGCGCACATCCAGCTCGTCGACG





GCCCCGACGACGTGGCGAACGTCGTCGTCCGCGACCCGGCCAAGGT





CGTCTGGCTGTCGCAGACCACGCTGTCGGTGGACGAGACGATGGAGA





CGGTGGCCCGGCTCAAGACCCGGCTGCCGCTGCTGCAGTCGCCGCC





CAGCGACGACATCTGCTACGCCACCTCGAACCGGCAGCACGTGATCA





AGGAGATCGCGCCGGAGTGCGACGTGGTGATCGTGGTCGGCTCGACC





AACTCGTCGAACTCGGTCCGCCTGGTCGAGGTCGCCCTCGGTGCCGG





CGCCCGGGCCGGTCACCTCGTCGACTACGCCGCCGAGATCCAGGAC





GAGTGGCTGGCCGGCGCCACCACGGTCGGTGTCTCCTCCGGCGCCA





GCGTGCCGGACGAGCTGGTGATGGAGGTGCTGGCGCACCTCGCGGA





GCGTGGCTTCGGCGAGGTCACCGAGTTCACCACGGCCGAGGAGCGG





CTCACCTTCTCCCTCCCGCAGGAGCTCCGCAAGGACATGAAGGCCGC





CGAGGCGGCCCGGGCCGCTGCCGCCGGCTGA





27
DNA
>ispE
ATGACCGAGGCGTGGGGTCCGGACGACGACGAGCCGCGCCCGTACA




(ACSP50_
GCGGCCCGGTCAAGGTCCGCGTGCCGGCCAAAATCAACCTGCACCTC




7802)
GCGGTCGGCCCGCTGCGACCCGACGGCTACCACGAGCTGAACACCGT





CTACCACGCCATCTCGCTGTTCGACGAGATCACCGCCCGGCACGGCG





ACACCCTCACCCTCACCATGGAGGGCGAGGGCACCGGCGACCTCGCC





CTCGACGAGACCAACCTGATCATCCGCGCCGCCCGCGCCCTGGCCGC





CCGCGCCCGCGTCCCCGCCTACGCCCGGCTGCACCTGCGCAAGAGC





ATCCCGCTCGCCGGCGGCCTGGCCGGCGGCAGCGCCGACGCCGCCG





CCACCCTGATCGCCTGCGACCTGCTCTGGGGCCTCGGCATGAGCCGC





GACGAGCTCGCCGAGGTCGGCGCCCAACTCGGCTCCGACATCCCCTT





CCTGCTGCACGGCGGCACCGCCCTCGGCACCGGCCACGGCGAGGCG





GTCAGCCCCATCCTGGCCCGCCCCACCACCTGGCACTGGACCGTCGC





CATCGCCGACGGCGGCCTGGCCACCCCCGCCGTCTACCGCGAGCTC





GACACCCTGCGCGCCGGCACCTGGCCACCCACTCCGCTCGGCAGCG





CCGACACCCTGATGGCCGCCCTGCGCCAGCGCAACCCGGAAATCCTC





GGCGCCGCCCTCGGCAACGACCTGCAACCGGCCGCCCTCGCCCTGC





GCCCCCAGCTCGCCGACGTGCTCAAAGCCGGCACCGAGGCCGGCGC





CCTCGCCGGCCTCGTCTCCGGCTCCGGCCCCACCTGCGTCTTCCTCG





CCGCCGACGCCACACACGCCCAGGAGATCGCCGACAGCCTCACCGAA





GCCGGCGTCTGCCGGGCCGCGGTCACCGCCCGCGGACCCCAGCCCG





GCGCGCGGGTAATCTAG





28
DNA
>ispF
GTGATCATTCCGCGGGTGGGTATCGGCACGGACGTGCACGCATTCGA




(ACSP50_
CGCTGACCGGGCCTGCTGGGTGGCCGGGCTGGAGTGGCCGGGGGAG




8046)
CCGGGGCTGGCCGGGCACTCGGACGCGGACGTGGTGGCCCACGCGG





CCTGTGACGCGCTGCTGTCGGCGGCCGGGCTCGGGGATCTGGGGGG





CAACTTCGGGACGAGCCGGCCGGAGTGGGCCGGGGCAGCCGGGGTC





ACGCTGCTCGCCGAGACGGCGCGGCTGGTCCGGGCGGCCGGGTTCG





CGATCGGCAACGTGTCGGTGCAGGTGATCGGGAACCGGCCGAAGATC





GGGAAGCGGCGGGCCGAGGCCGAGAAGGTGCTCTCCGCGGCGGTGG





GGGCGCCGGTCACCGTGTCCGGGACCACATCCGACGGGCTGGGGCT





CACCGGGCGTGGTGAGGGGCTGGCCGGAGTCGCGGTGGCGATGGTC





TACACGGAGAACGCTCTTCCGGCCTGA





29
DNA
>ispD
GTGATCGCCGACCGCGACGTGACCGCGCAGCTCAATGCTCGCGGTGA




(ACSP50_
CGTCGCGGTCGTCGTTCCGGCGGCGGGGGCGGGTCTCCGGCTCGGC




8047)
CCGGGCGGCCCGAAAGCTCTGCGTCTGCTCGACGGCGAGCCGCTGC





TCGTGCACGCGGTCCGGCGGTTGGCCGCGGCCGCGCCGGTCCGCAT





GATCGTGGTGGCCGCTCCGCCCGCCGAGGTCGACGCGGTGTCCGCG





CTCCTCGCCCCGGTGGCCCCGGTCACCGTCGTGCCCGGCGGCGCCG





AACGCCAGGAATCGGTCGCCGCGGCACTCGCGGTCGTTCCGCCGGAC





GTTCCGATCGTTCTGGTCCACGACGCGGCTCGATGCCTCACCCCGCC





CTCGGTTACGGAGCGTGTCGCCGCCGCTGTCCGGGACGGTGCCGAC





GCGGTGATCCCGGTCCTGCCGGTCGTCGACACGATCAAAGAGGTCGC





GGCCGATGCCACCGTTCTCGGCACGGTCGACCGTTCCGTGCTGCGTG





CGGTACAGACTCCGCAAGGCTTCCGCGCCTCGGTGCTGCGCGCCGCT





CACCGGGCCGCCGCCGACTCACACACCGACGACGCCGGTGCCGTCG





AGAAGCTCGGCATCCCGGTCCTGTGCGTCCCGGGCTCCGACCTCGCG





CTCAAGATCACCCGGCCGATCGATCTGGCGCTCGCCACGCACCTCCT





GGCCCTGCCGGACCCGGACGCCCCTACCGCCTGA





30
DNA
>idi
ATGAGCAGCATCGGTCACCTCAACCGTGAAGATCATCTCGTCGAGCTC




(ACSP50_
GTCAACGAGGAGGGGCAGCCGCTCGGGTCGGCCACCGTCTCCGACG




0146)
CCCACCTCTCGCCGGGTGCGCTGCACCGGGCCTTCTCGGTCTTCCTC





ACCGACGATGAGGGCCGGGTGCTGCTCCAGCAGCGGGCCGCGGCCA





AAACCCGCTTCCCGCTCCGCTGGGGCAACACCTGCTGCGGCCACCCC





GCGCCCGGCGAGCCGGTCACGGTCGCCGCGGCGCGGCGTCTCACCG





AGGAATTGGCGGTACGTGACGTCACGCTGACCGAGATCGGCGTGTAC





ACCTACCGCGCGACCGACCCGGTCACCGGCCGGGTGGAGCACGAATA





CGACCACGTGCTGATCGGCGCCCTGCCGGACGGCGTCGTGCCACACC





CCGATCCGGCGGAGATCGCCACGCTGCGCTGGGCCTCGCTGCCCGG





GCTGCGCACCGGGTTGACGGAGTCCCCCGAGCTGTACGCGCCCTGG





CTCCCCGGGGTGTTCGAGATTCTCACGGAGCGGTCGGGTGTCCTTTC





CACGGAGCGGTCGGGTGGCCGGTGA





31
DNA
>crtE
GTGGCCAATGACACCCTCGAGGGAAATCGCCTTGCCGCGATACCCCG




IdsA
GCAGTCCGTCTCTCACACTGGGCTGGTCGGTGCAGTCGAGGGGACGC




(ACSP50_
TCGCCGACTTCCTCGCCTCCCAGATCGCCTCTCTCGACGCCGTCGACC




0148)
CATCGCTCGGTGGCTTCGGCCGCACCGCCCGTGACCTGGTGATGGCC





GGCGGCAAACGGCTGCGGCCGACGTTCGCGTACTGGGGCTGGCGCG





GCGTCGCCGGGCCGGCCGCGGACGCCGAGACGCTGCTGCCCGCGCT





CGGCGCGCTGGAGCTGATGCACACCTTCGCGCTCGTCCACGACGACG





TGATGGACGACTCGTCCACCCGCCGCGGCCGGCCCACCGCCCACCG





GATCTTCGCGGCCCAGCACGGCGGCCGGTTCGGCACGTCGGCCGCG





ATCCTGGTCGGCGACCTCTGCCTGGTCTGGGCCGACCAGCTGTTGGC





CCGCACCCCGGTGCCGGCGGCCACCCTGCTTGCAGTCCGCGCGCATT





ACGACCGGATGCGGATCGAGGCGGTCGCCGGGCAGTATCTGGACGTC





CTCGGTGAGACCGATCCGGCGTCCTGGTCGGTGGAGCGCGCACTGCT





GGTCGCCCGGCACAAGACCGCCAGCTACACCGTGCAGCGGCCGCTC





GACTTCGGCCTGGCCCTGGCCGGGGTCGAGGACGTGGAGGTCGCCG





AGGCGTACCGGACCTACGGCATCGCCGTCGGCGAGGCCTTCCAGCTG





CGCGACGACCTGCTCGGTGTCTACGGCGACCCGGCGGTGACCGGCA





AACCGGTCAGCGACGACCTGCGCACCGGCAAACCGACCGCACTGCTG





ATGCTGGCCCGTCGGATGGCCACCCCCGGCCAGCTGGCCGAGCTGG





AGTCGGCGGAGATCGAGCGCAAGGCGCAGGTCGTCGCCGAGACCGG





CGCCCCGGCCCGGGTCGAGGAGATGATCCGTGCCCGGGTCACCGAA





GGACTGACCGCGCTGGCCTCGGCGCCGATCGACGCCGAGGCCCGTG





CCACCCTGATCGAGCTGGCCACCGTGGCGACGCAGCGCCCGGCATGA





32
DNA
>crtB
ATGGAAACCGATCTGGCCGCCGCCTATGAGCGGTGCCGTGAGCTACA




(ACSP50_
CCGAGAGCACGGACGCACGTACTACCTGGCGACCCGGTTACTACCGG




0149)
CCTGGAAGCGCCGGCATGTGCACGCTCTGTATGGATTCACCCGGTTC





GCCGACGAGATCGTCGACCGCACCGAGGCGCAACCACCCGCCGAGC





GCGCCGCCGAGCTGGCCACCTGGTCCGCCGGATTCCTCGCCGGACT





GCGCGGCGAGCCGGTCGACGACCCGCTGCTCCCGGCCGTGCTGCAC





ACCATCGCGGTCTTCGGGCTCGACCTGGAGGACTTCGCGAAGTTCCT





GCGCAGCATGGAGATGGACCTCACCGTCACCGGCTACCGCACCTACG





ACGACCTGCTCGACTACATGGAGGGCTCGGCCGCCGTGATCGGCACC





ATGATGCTGCCGATCCTGGGCTCCACCGACCCGGCCGCCGCCCGCGA





ACCGGCCCGCCAGCTCGGCTTCGCCTTCCAGCTCACCAACTTCATCCG





GGACGTCGCCGAGGACCTCGCGCGGGACCGGATCTACCTGCCCGAG





GAGCACCTCGCCGAGTTCGGTGTGACCCGCGCCGACCTGGCCGCCG





GCGTCGCCACCCCGGCGATCCGCGCGCTCATCCGGGCCGAGGTGGA





CCGCGCCCGTGAGCACTACGCGGCCGCCGCCCCCGGCATCCCGCTG





CTCGAACGCACCTCGCAGGCCTGCATGCGGACCGCCTTCCAGCTGTA





CGGCGGGATCCTGGACGAGATCGAGGCGGCCGACTACGACGTGTTCG





CCCGGCGGGTCACGGTGCCGAACCGGCGCCGGGCCGCGGTCGCCGT





CCGCAGCCTGCTCACCCGGCCCGGCACCCCGGTCGAACTGGCGGCC





TGA





33
DNA
>ACSP50_
ATGGGCGCCCGCGTCGCGCTGTTCACCCGCGACCTGCGGATCCACGA




0150
CAACCCGCTGCTCAGCGGGCCCGACCCGGTGGTGCCGCTGTTCGTCC





TCGACCCACGGCTGAGCGGCCTCTCGGCCAACCGCAGCCGCTTTCTC





CACCAGAGCCTGGCCGACCTGCGGAACAGTCTCCGCGAGCGTGGCG





CCGACCTGGTGATCCGGGAGGGCGACCCGGTGGCCGAGACCATCGC





GGTCGCCTCCGAGGTGGACGCGTCGACGATCACGGTGGCCGCCGAC





GTGACCGGTTACGCCCAGCGGCGCGAGCGGCGGCTGCGGGACGAGC





GATTCCGGGTGAAGACGGTGCCGAGCGTCACGGTGCTGCCGCCCGGT





ACGGTCCGGCCGGGCGGGGGAGGCGAGTCGTACCGCGTGTTCACGC





CGTACTTCAAAGCCTGGGAGAAAGCTGGGTGGCGCGCACCCTCCGCA





ACGCCGGGGAAGGTCGCGATGCCGGCCGGCATCGCGCCGGGAAGGC





TCCCCGAGATGCCCGCCGGCGACTCACCGGACGCCGTCGCCGGTGG





CGAGACCGAGGGCCGCCGCCGGCTCCAGGCCTGGCAGAAAGAAATG





GCGCGGTACGCCGAGGACCACGACGACATGGCCGCCGACAACACCA





GCCGGCTCAGCGCCTACCTCCGGTTCGGCTGCCTGTCGCCGCTCGAA





CTGGCGCTGGCCGCGAAAGCCGACGACTCTCCCGGCGCCCAGGCCT





ACCTGCGGCAACTGTGCTGGCGGGACTTCTACTACCAGGTCACCGCG





ACCTTCCCGGAGATCTCCACCCGGCCGCTGCGGGAGAAGGCGGACCA





GAACTGGCGATACGACGACGACGCGCTGCGTCACTGGCAGGACGGCC





TGACCGGGGTGCCGATCGTCGACGCCGGCATGCGCCAGCTCCGCGC





GGAGGGCTGGATGCACAACCGGGCCCGGCTGATCACCGCCGCGTTC





CTCACCAAACACCTGGGCATCGACTGGCGGCCCGGGCTGCAATGGTT





CTTCCGCTGGCTGCTCGACGGCGACGTGCCGAACAACTCCGGCAACT





GGCAGTGGACCGCCGGCACCGGCAACGACACCCGGCCCTATCGCAG





GTTCAATCCCATTCGCCAAGCGCAGCGATTCGATGCGCAGGGCGTGTA





CGTTCGGCGCTACGTACCGGAGTTGAAAGACATCGACGGTGTCACGG





TGCATCAGCCGTGGCGACTGCCGGAATCGGTACGCCGCGGGCTCGAC





TATCCCGGACCGTTGGAGTCACATCGGGACGAGGCGGTCTGGCTGCG





CGACTGA





34
DNA
>ACSP50_
ATGTCTGAAGCGCGGCAAGTGGACGTGGTGGTCGTCGGGCTCGGTGT




0151
CGGCGGCGAGGAGGTCGCCGGTCGCCTGGCCGCGGCCGGCCTGAG





CGTGATCGGCGTCGAACACCGACTGGTCGGTGGCGAATGCCCGTACT





GGGGATGCATCCCCACCAAGATCATGGTCCGCGCCGGGAACGCGCTG





GCCGAGGCCCGCCGGATCCCCGGCCTCGCCGGGACGTCCACGGTGC





GGGCCGACTGGGCGCCGGTCGCCAAACGGATCCGCGACGAGGCCAC





CGACGACTGGAACGACAAGGTCGCCGTCGAGCGGTTCACCGGTAAGG





GCGGAACGTTCGTCCGGGGCACGGCCGAACTGACCGGTCCCGGTCA





GGTCCGGGTCGGGGACCAGGAATTCGCCGCTTCGCGCGGCGTGGTC





ATCGCCACCGGCACCGCCGCTGTGGTCCCACCCATCGAGGGCCTGTC





CGGTACGCCGTTCTGGACGAACCGTGAGGCCGTGGAAGCGGCGGCC





CTGCCCGCATCGATGCTGGTGCTCGGCGGCGGGGCGATCGGGTGCG





AGCTGGCCCAGGCGTACGCCCGGTTCGGCGTGCAGGTGACGGTCATC





GAGGGCTCACCCCGGGTGCTGGCCATGGAGGAACCGGAGTCGTCCG





AGGTGGCGGCCGCCGCCCTGACCGCCGACGGGGTCCGGATCGTCAC





CGGGGTGCGCGCGCAGAAGGTCGCCCACGACGACGGGTTCCACGTG





ACCCTCTCCGACGGCAGCGTGCTGGCCGGCGAGAAGCTGCTGGTCGC





GACCGGGCGGGCGGCCCGGCTCGGCGGGCTCGGGCTGGACCGGGT





GGGGCTGGACCCGTCGGCTCGATTCCTGGCCACCGATGACCGGCTGC





GCGCCGGCGAGGGCATCTGGGCGGTGGGGGACGTGACCGGGAACG





GGGCGTTCACCCACATGGCGATGTACGAGGCGGACATCGCGGTGCGG





GACATCCTGGGGCAGGGCGGCCCGGGAGCCGACTACCGGGCGCGGC





CGCGGGTGACCTTCCTCGACCCGGAGATCGGGGCGGTGGGGATGAC





CGAGCAGCAGGCCCGGGACGCCGGCCTCGAGGTGCGGGTGGGGTAC





GTGCCGCTGAACCAGACCTCGCGAGGGTTCATCCACGGGCCGGGGAA





CGAGGGATTCCTCAAACTTGTCGCGGACGGGGAGCGGGGAGTGCTGG





TCGGCGGGACGACCGCCGGGCAGTCCGGTGGCGAGATGATCGGGGC





GGTGGCGGTGGCGGTGCACGCCGAGGTGCCGGTGTCGACGTTGCTC





AGCCAGATCTGGGCGTACCCGACGTTTCATCGGGGGCTGGGGCAGGC





GCTTCAGTCGCTGGCCTGA





35
DNA
>ACSP50_
GTGAGCGAACCCGTCATCACCGAACCGGCTGCCTGGATCAACCTGCC




1631
CGACCTGTCCGAGAGGCTGGACGTGTCGATCAGCAAGGTGCACCAGA





TGATCAGAGACGGCGACCTGCTCGCGGTCCGCCGCGACGGCATCCGC





GTGGTGCCCGCCGAACTGGTGGCCAACGCCACCGTCCTCAAGCATCT





GCCCGGTGTGCTGAACGTGCTCCGCGACGCCGGGTACAACGACGAAG





AGGCCTTCCGGTGGCTCTACGCCGAGGACGCCGAGGTCGGCGGCAG





CGCCGCGATCGCGCTCGGCGGTCAGCAGGCGCGCGAGATCAAGCGC





CGCGCGCAGGCCCTCGGCTTCTGA





36
DNA
>ACSP50_
ATGAGGCATTTGTCGTACGTCGCGGTGCTGGCCGGATGCCTGGCCGG




1632
GGCGCTGTGGCTGGAACCGATCCTGCGGGTCAACGTGCTGCGCCGGT





GGCGTCGGCTGCTGCTGGCCGTGCTGCCGATGGCGGTCGTCTTCACC





CTGTGGGACCTGGCGGCGATCGCGGCCGGCCACTGGCACTTCGACC





CGGCCCAGATCACCGGCGTCTACCTCGGCGGCGGGCTGCCCCTCGA





CGAGGTGCTGTTCTTCCTGGTGGTGCCGGTCTGCGCGATCCTCGGCT





TCGAGGCCGTGCGGGCCGTGCTGCGACGTCCGGCGGGGGACGAGTG





A





37
DNA
>ACSP50_
GTGACCTACACCACCGCTGCGGTGCTCGGCGTGCTGGCCGCCCTCAC




1633
GCTCGACGTGCTGATCCTGCGGACCCGGCTCGTCGGGCGACTGGTGT





TCTGGGCCACGTACCCCATCATCTTCGTCTTTCAGTTGATCTCGAACG





GCATTCTGACCGGGCGCGACATCGTGATGTACGACCCGGCCGCGATC





CTCGGCCCGCGGCTCGTCCACGCCCCGGTCGAGGACCTGCTGTTCGG





TTTCGCCCTGGTGCTCGGCACGCTGTCGCTGTGGGTGGCGCTGGGCC





GGCGCGGCATCCAGCGCACCCCGCGAGCCGGGTCTAGACGGACCGA





CGAGTAG





38
DNA
>crtE
GTGACGAACTCCCCGCTCGACGAGGCCGGTCTGCGGTCGCGTGTCGA




fps2
CAAGGCGCTGACCGTGTTCCTGGCCGGGCAGCGTGACCGGCTGCTG




(ACSP50_
GCGATCGACCCGGCGCTGGCCGAGATGTCCGCCACGGTCTCCGAGTT




1634)
CGTGCTGGGCGGCGGGAAGCGGCTGCGGCCGGCATTCGCCTACTGG





GGTTTCCGCGGGGCCGGCGGCGCCGACTCGGACGCCGTGGTGGCGG





CCGTCGCCGCGCTGGAGCTGGTGCAGGCCAGCGCGCTGATCCACGA





CGATCTGATGGACCGCTCGGACACCCGGCGCGGGGTGCCGTCGGTG





CACCGTCGGTTCGAGAAACTGCACGCCGGCGAGGGCTGGCGGGGCA





GCGCGGCCGGGTTCGGCGACTGCGCCGCGGTGCTGCTCGGCGACCT





GGCCCTGGTCTGGTCGGACGAGCTGCTGCACACCTCGGGGATGGCG





GTGGCCGACGTGCAACGGGCCCGCCCGATCTTCGACGGGATGCGCA





CCGAGGTGACCGTCGGGCAGTACCTGGACGTGCTCACCCAGGCGACC





GGCGACACGTCGCTGGAGCGGGCCGGCAAGGTGGCCGTCTACAAGG





CCGCGAAATACACCGTGGAGCGTCCGCTGCTGCTGGGCGCGGCGCT





GGCCGGAGCGGCCCCCGGGGTGCACGCGGCGTACTCGGCGTTCGGC





CTGCCGCTGGGCGAGGCGTTCCAGCTGCGCGACGACGTGCTGGGCG





TGTTCGGCGACCCGGAGCGGACCGGCAAGCCGGCCGGCGACGACCT





GCGCGAGGGCAAGCGCACCTATCTGGTCGCGGCCGCCTTCGGCGCG





CTGGACGCGGCCGGGCGGGCCGAACTGGACGCCGCGCTCGGCGACC





CCGGCCTGGACGAGGCCGGGGTGGCCCGGCTGCGCACGGTCATCCG





GGACAGCGGTGCGCTGGCCGCGACCGAGGCCCGGATCGACGAGCTG





ATGACCGCGTCGATCGGCGCGCTGGACGCGGCACCGATCGATCAGGA





CGCCCGGGAGGTGCTGCGCCGGCTGGCCGACGCGGCTACTCGTCGG





TCCGTCTAG





39
DNA
>ACS
GTGTCTCTCGGACTTCCCTCCCGGCTGCCCGGCACCCCGTCGATCGG




P50_1
CGACCTGGTCCGCGGCGCGGCGCCGACGTTCTCCTTCGAGTTCTTCC




635
CGCCGAAGACACCGGACGGGGAGCGGCTGCTCTGGCAGGCCATCCG





GGAGCTGGAGTCGCTGCGGCCCAGCTTCGTCTCGATCACCTACGGGG





CCGGCGGCACCACCCGGGAGACCACGGTCGCGGTCACCGAGCGGGT





CGCCACCGAGACCACGCTGCTGCCGCTGGCCCACCTCACCGCGGTCG





ACCACTCAGTGGCCGACCTGCGCAACGTGATCGGCCGGCTGGCCGGC





GCCGGGATCCGCAACGTGCTGGCGCTGCGCGGCGACCCGCCGGGCG





ACCCGATGGGCGAGTGGGTCCGGCACCCGGACGGCGTCGGTTACGC





CGACGAGCTGGTCCGGCTGATCCGCGAGTCCGGCGACTTCAGCGTCG





GGGTGGCCGCCTTCCCGCACAAACACCCCCGGTCGGCCGGCGTCAA





GGACGACACCCGCAACTTCGTCCGCAAGTGCCGGGCCGGTGCCGACT





ACGCGATCACCCAGATGTTCTTCGACGCCGACGAATATCTGCGGCTGC





GCGACCGGGTGGTGGCCGCCGGCTGTCACACCCCGATCGTGGCCGG





CGTGATGCCGGTGACCCGGATGGCCACCATCGCGCGCTCCACCCAGC





TCTCCGGCGCGCCCTTCCCGCCGGCGCTGCTGCGCGACTTCGAGCG





GGTCGCCGGCGACGACGCGGCGGTGCGCGAGCTGGGCATCGAGACG





TGCGCGGCGATGTGCGCCCGGTTGCTGCGGGAGGGTGTGCCGGGCA





TCCACTTCATCACCATGAACCGGTCCACCGCCACCCGCGAGGTCTGG





CAGCGGCTGGCCCCCGCGGAAGTCGCCGCGTCGGCGTGA





40
DNA
>ACSP50_
GTGCAGCTGCAACAACTCCGGTACTTCCTGGCGGTGGTGGAGACCCG




1650
GCATTTCACCCAAGCAGCGGACATTCTGGGCGTCTCGCAACCTACCTT





GAGTAAGCAGATTCACACCCTTGAGATGTCACTCGGAGCCCCGCTGTT





CGAGCGGATGCGCGGTGCGGTGACCCTGACCGTCGCCGGCGAGACA





TTGCTGCCGATGGCCCAGCGGATCGTCGCCGACGCCGACGCGGCCC





GCGACGCCGTGCAGGACATCGTCGGTCTGCGCCGCGGCGAGGTGCG





CCTGGGTGCCACCCCGAGCCTGTGCTCCTCGCTGGTCCCGGCCGTGT





TGCGCACCTTCCGCGCCGACCACCCGGGGGTCAAGCTGCACATCAGT





GAGGGCAGCTCGCACGACCTGACCGCCGGCCTGCTGGCGCACACCC





TGGATCTGGCCCTGATCGTGCAGCCCGAGCACGGCGTCGATCCGGCC





CTGGTGGCCATCGAGCTGCTGCGCGAGAGCCTGGTGGTGGCCTCGGT





CGCGGCCGGCCCGCCGCCCACCGTGGGCCGCCAACTGGAGCTCTCC





GAGCTGCGCCACACCCCGATGGTGATGTTCCGCGAGGGCTACGACAT





CCGTGAGGTCACCCTGCACGCCTGCGAGCGGGCCGGCTTCGCGCCG





AAGTTCGCGGTCGAGGGTGGTGAGATGGACGCGGTGCTCGCCTTCGT





CGAGGCCGGCCTCGGGGTCGCCCTGGTGCCCAGCATGGTGCTCGCC





AACCGGCCGCTGCTGCGGGCCACCCCGCTCGCGCCGCCGGGGATGC





GCCGGACCATCGCGCTCGCCCAGCGCCGTGCCGCGGTGCTGCCGCA





TGCCGCGGCCGCGCTGCGTGAGGTGGTGCTCGACCACATCGGCTCG





GGCCGGCTGCCGTTCGGCGTGCGCGCCCTGGAGAGACCGTCCACTTA





G





41
DNA
>ACSP50_
ATGGGCGAGTTCCACGACCCGCGACTCGTCGAGGTCTACGACGCCGA




1651
ATGTCCCTGGGGCTGGGACGACGACTTCTTCATGGCCGTGCTCGCCG





AACGCTCCGCGCACCGGGTCGCCGACCTGGGGTGCGGCACCGGCCG





GCTGGCCATCGCGATGGCCGCGGCCGGGCACGAGGTGATCGCGATC





GACCCGGCGCCGGCCGCCCTGGCCGCGGCCCGCCGCAAGCCGGGC





GGCACCCGGGTGCGCTGGCTGCAGGGCTCGGCCGAGCGGCTCGCCC





CGCGCTCGCTCGACGCCGCGTTCATGACCGGTCACGTCGCCCAGTCC





TTCGTCGACGACGAGGAATGGGACACCGTGCTCCGCGGGCTGCGCCG





GGCGCTGGTCCCGGAGGGACGGCTGGTCTTCGACAGCCGGGACCCG





GACGACCGGCCGTGGCAGCAGTGGAACCCGCAGGATTCGTGGCGCA





CCGTGGTGCTCGACGACGGGAGGGTGGTGGAGGCGTGGAGCGAGGC





CGAGCAGGTCGGGCTGAACACCGTGCGCGTCACCGGGCGCTACCGG





TTCGCCGACGGAGGGGAACTGGCGAACTCGGCGACCCTGCGTTTCCG





GACCGAGCCGGAGCTGCGCGACTCACTGCGCGAGGCGGGCTTCCGG





GTCGAGCGGATCTACGGCGGCTGGGGGCGCGAGCCGGTGGGTCTGA





GCGGCGACGGCGAGTTCATCGTGATCGCGGTCGCGACGCCCCGGCT





GATGTCCTGA





42
DNA
>ACSP50_
ATGCCCGAGAACGAGTGGCCCGACGACCCCCGCCCGCCCGACCAGG




1652
GCGAGTGGAGCCAGCCGCATCACGAGCCGCCACCCGGCCGTGGCCG





CGCCCTGCTGGCCGCCGCGGTGGTGGTGCTGGTCCTGCTGGCCGCC





GGCGGCATCGCCTGGCGTCTGATGAGCAGCCGCGGCGCTACGCCGG





TGGCGCAGCCCACCGCGCCCGCCCCGACGCCCACCGCGCAGACCGC





GCCACCCTGCCCACAGCCGCGCCTGCGGGTCGCCGCCGCGCCGGAG





ATCGCCCCGGTGATCCAGCAGGCCGCCGCCGCACTCAGCCAGCCCG





GCCAGCGCTGCTCCGAGGTGCTGGTGCAGGCCGCCGAGCCGGGCGC





CGCGCTGACCGGCAAGCCGGACGTCTGGGTGCCGTCCAGCAGCGTG





TGGCTGGCCCTGGCCAAAAGCCGCGGCGACGTCTACACCACGCAGGG





CGCGTCGCTGGCCTGGTCGCCGCTGGTGATCGCCGGGCCGGAGTCG





ATCGCCAGCCTGTTCGCGCCGAACGGGGTCACCTCCTGGTCCGGCCT





GGTCCAGGGCACCATCCAGAAACGGGTGCCGGCGGTCCGGATGCCC





GATCCGACGCTGACCACGACCGGACTGCTCAGCGTCTACGCGGTGGG





CCAGGCCACGGTCAAGGCCAACCCGGACGCCGGGATCGCCCAGTTG





CAGGCGCTCACCCTGCGCAGCCGGCTGGAGAACGCGGCCGCCGACC





CGGCGGAACTGTTCGCGCAGATGGGCAAGCAGACCGACGCGGCCAC





GGCGATCTACCAGGTCGGGGTCTTCCCGACCACCGAGCAGCAGCTGC





TGACCTATCAGAAGAGTCAGCACGACGTCCGGCTGTCCGGCTCGGCG





CCCGCCGACGGCCAGATCGACGCCGACTATCCGTACGCGGTCCGCAA





GGGCGCCCCGGCCGACCTGGTCGAGAGCCTTCGCGAGGCGATCACC





CCGGACGCGCTGACGACGGCCGGATTCCGGGCCACCGCGACCAAGA





ACGCGCTGCGCCTGCCGGCCCCGGCCGTGCTCGCCGGGGCGGCCCG





GCAGTGGTCGGCGTACAAGTCGGTGGCCTTCCAGGTGCTGCTGCTGA





TCGACGCGTCCGGCTCGATGAACGAGAAGATCACCGACCGGGCCGGC





CGCAGCGTCACCAAGGCCGCGCTGCTGCGCGAGTCCGGGACCAGCG





CGGCCCAGCTCTTCGGTGACGACACCAGCCTCGGCCTGTGGTTCTTC





GGCACCCCGACGGCGGACAGCCCGGCGCACACCGAGGAGGTGCCGT





TCGGCCCGGTCATCGCCACCGTCGACGGCAAGAGCCGCCGTGACCTG





CTGGCCGCCAAGATCGGCGAGTACCGGCCGGTGGCGAACGCCGGGA





CCCCGCTCTACCAGAGCGTGCTGGACGGCGTCGCCGAGATGCGCGG





CCGGGCCAAGCCGGACACGGCGACCGTGGTGGTGGTCCTCACCGAC





GGCTCGGACGGCGGCACGAAGTACCGGATGTCCAACGCGGACTTCCT





GAAGAAGCTGACCGCCGGTGCCGACCCCGCCAAGCCGGTGCCGGTG





ATCGCCGTCGGTTACGGCCCGGCCGCGAACGCCACCGCCCTGCAGG





CCATGGCCAAGGCCACCGGTGGCCAGGCGGTCACCGTCAAGAACCCG





GCCGACCTGGCCGCCGGCATCGCCCAGGCCTTCCTCGCCGCACACAC





CCACTAG





43
DNA
>crtD
ATGAGCGACATCGTGGTGGTCGGGGCTGGGGTCGGCGGGCTGGCCG




(ACSP50_
CGGCGATCCGGCTGGCCGAGGCGGGGCATCGGGTCAGCATCCATGA




1653)
GCGGTCCGGCGTGGTCGGCGGCAAGCTGGCGGCATACGAGAGGGAC





GGCTACCGGTTCGACACCGGCCCCAGCCTGCTCACCCTGCCGGACGT





GTTCACCGGCCTCGGTCTGGACCTGCGCCCGGAGCCGCTGGACCCG





GTGGTGCGGCACTTCTTCCCGGACGGCACGGTGCTGGACTCGTCGTC





GGACCACGAGACCTTCCTGGCCCGGATCACCGACGCGCTGGGCGGT





GCCGCGGCGCGCGACTGGGACCGGTTCTGGCGCCGTGCCGAGCGGA





TCTGGCACGCCTCCTGGGAGTCGGTGCTGCGCCGCCCGGTGACCGC





GGCGTCGCTGGCCCGGCTGTCCTGGCGGCTCGGTGACCTGGCCGCG





ATCGCTCCCGGCCGGTCACTGCGGTCGCTGGGCCGCCGCTATCTGCG





CGACCCGCGGCTGCGGATGCTGCTGGACCGCTATGCGACGTATTCGG





GCGCGGATCCGCGGCGGGCGCCGGCGGCGCTGGCCGCGATCCCCTA





CGCCGAGCTGGCGTTCGGCGGGTGGTATCTGCCGGGTGGGCTGGTC





ACCCTCGCGGAGGCGCTGCTCGCCCGATGCGAGAAACTGGGCGTACG





GGTGCATCTGCACTCACCGGTCGCCTCGATCGCCACGACCGGCGCCC





GGGTGTCCGGGGTCCGGCTGGGGGACGGGACCCGCCTCGCGGCGG





ACGTCGTCGTCTCCAACGTGGACGCCGTCACGCTCTACCGGGATCTG





CTGCCCAGTCCGAAACCGCTGGCCCGCCTCGCCGACCGGAGCCTGG





CCGGATTCGTGCTGCTGCTCGCGGTGCGGGGCGAGACTCCGCGGCT





GGCGCACCACAACGTGTTCTTCCCGCGGGACTACGACGCCGAGTTCG





ACGCGGTCTTCGGGGGGCCGGGGCGGCGGGCGCGGCCGGCCGGCG





ACCCGACCGTCTTCGTCACCCGGGCCGCGGATCCGGCGGTGCGCCC





GGCCGGCGACGAGGCGTGGTTCGTGCTGGTCAACGCGGCGCCACAC





GGCACCTCGTGGTCCACCGTGGACTGGCTGCGGGCGGGGCTGGCCG





ACGCGTACCGGGATCGGGTCCTCGAGGTCCTGGCGGGGCGCGGTCT





CGACGTACGCGATCGGCTGATCTTCGCCGAGACCCGGACCCCGGCGG





ATCTGGCGGCGTCGGCCGCAGCGCCGGGCGGAGCGATCTACGGCAC





CGCCGGCGGCCTGGTCCGGCCGGCGAACCGCGCGCCGGTCGACGG





GTTGTTCCTGGTCGGCGGCTCGACGCATCCCGGCGGCGGGCTGCCG





ATGGTCACCCTCTCCGCCGAGATCGTCGCGGGCATGATCGGATCGAA





CTGA





44
DNA
>cruC
ATGATCGTCGCCTGGCTGATCCTGCCGCCGCTGCTGCTGATCACCGC




(ACSP50_
ACACACCGCCGTCAACGCGCTGCTGCTGCGCCGCCCGCGCCGGGCG




1654)
GCGACCAGCACCGAACGGGTCGCCGTCCTGCTCCCGCTGCGCGACG





AGGCCACCCGGGTCACCCCGTGCCTGCGCGCCCTGCTCGCCCAGCG





CGGCGTCGCCGATCTCACCGTGCACGTGCTCGACGACGGCTCCACCG





ACGGCACCGCGGACGTGGTCCGGGCGGTCGCCGGCGACCGGGTCCG





GCTGCACACCGGCACTCCGCCGCCGCCCGGCTGGCTCGGCAAACCG





GCCGCCTGCCAACGGCTCGCCGACCTGGCCGGGGACGTGGACGTGC





TGGTCTTCGTCGACGCCGACGTGGTGCTCGCGCCGGACGCGGTGGC





CGGGGCCGTCGATCTGCTGCGCCGGGCCGGAGCGGACCTGCTCAGC





CCGTACCCGAAGATCGTCGGTGCCGGCCGGCTGGTCCAGCCGCTGCT





GCAGTGGTCCTGGCTGAGTTTCCTGCCACTGCGCGCGATGGAACGCT





CGGCGCGGCCGTCGCTGGCCGCCGCCGGTGGCCAGTGGCTGGTGCT





GGACCGGGCCGGTTACCGGCGAGCCGGTGGCCACGCCGCGGTGCGC





GGCGAGATCCTGGAGGACATCGCGCTGGCCCGCGCGGTCAAACGGG





CCGGCGGGCGGATCGCCCTGGCCGACGGTTCCGGCCTGGCCACCTG





CCGGATGTACGAGTCCTGGGACGAGCTCGCCGACGGATACGCCAAAT





CGCTGTGGGCGTCATTGGGGTCCGCGGCCGGCGCGACCGCCGTCAC





GCTCCTGCTGATTCTGCTGTACGTGGTGCCACCCCTGCTGGCGCCCTT





CGCCCCGCTTCCGGCGGTGCTCGGCTACCTGCTCGGCGTGACCGGC





CGGATGATCGCCGCCAGGGCCACCGGCGGCCGCGTCCTGCCCGGCA





CGCTGGCCCATCCGGTCTCCATCGTCCTGTTCGGCTACCTGATCGCCC





GCTCCTTCCGGCTGCGCCGGGCCGGCCGCCTGGCCTGGCGCGGCCG





CCCGGTGCCCTGA





45
DNA
>cruF
GTGTCTCCCCGTCATCTGCCCTGGGGCCTGCTCGGGGCGCTCGTGCT




(ACSP50_
CGCCCAGATCTGCTATCCGCTCACCGAGGGTGACACCCGGGCCGGGC




1655)
TGACCGTGCTCACCGTGCTGCTCGGCGTCGCGTTCTCGCTGAGCCAC





GCGCTGCTCACCCGGGGCCCCCGGGCGCTCACGGCGCTGCTGTCGA





CCGCCACCCTGGGCGGGTTCGCGGTGGAGGCGATCGGGGTGGCCAC





CGGTTTCCCGTTCGGTTCCTACGAGTACTCCGGGCGTCTCGGTCCGC





GCCTGCTCGGCGTACCGCTGATCATCCCGCTGGCCTGGACCTGGATG





GCCTGGCCGGCCTGGCTCGCCGCGCTGCGGGTGACCCGGCGGCGGC





TCCCCCGGATCCTGGTCGCCGGGGCCGGCCTGGCCGCCTGGGACGT





CTTCCTCGACCCGCAGATGGTCGCCGAGGACTACTGGCGGTGGCGGC





ACCCGGTGCCCGCGCTGCCCGGCGTGCCCGGTGTGCCGCTCGGCAA





CTACCTGGGCTGGCTCGGCTTCGCGCTGCTGCTGATGACCGCGCTGG





CCGCCGTCGCCGGCCGGGCCGCCGACCGGCCGCTGTCCGCCGACCG





GCCGGCGCTCGCCCTGTGGATCTGGACGTACGCCTCGTCGGTGCTCG





CCCACGCCGTCTTCCTGTCGCTGCCGGCGTCCGCGGCGTGGGGCGC





GCTGATCATGGGCGCCGCGGTCCTCCCGCTGCTCGCCCGGCTGCGC





GCACCCGCATGA





46
DNA
>ACSP50_
ATGAGGCTTGTGGCGTGGCAGCCGGACGACCTGCTGCGGCGGCTCG




1656
ACGACGTGGTCGGGGTCTACGGCGAGGCGATGGGCTACCGCCAGGA





GCTGCTGCAGACCCGCCGGGGATACATCGGGTCGCACGTGCGCCGG





CCCGGGTTCCGGGCGGTGGCCACGCTGACCACCGAGGGCCGGCTGA





TGGGCTTCGGATACGGCTACACCTCCGCCGCCGGCCAGTGGTGGCAC





GACCAGGTCCGGTTCGCTCTCGGCGAGGACGACCGCCGGCAGTGGC





TGACCGACTGCTTCGAGGTGGTCGAGCTGCACGTGCGCCCGGCCGCG





CAGGGCCACGGGGTGGGCGCCCGGCAGCTGCGCGCGCTGCTGGCCA





TGGCCAAAGGCCGCACCGTGCTGCTGTCCACTCCGGAGGCCGACGAG





CAGGCGTCCCGCGCCTGGCGGCTGTACCGGCGGTACGGCTTCGCCG





ACGTGCTGCGGCACTTCTACTTCCCGGGTGACGAGCGGGCCTTCGCG





GTCCTCGGCCGCGAGCTGCCGCTGGCCGAGCGTCCGCTCGAGGACG





CACCGGGCATCGCCGGCGCCTGA





47
DNA
>ACSP50_
ATGACGCACGTCGCCCTGCACGTCTGGCGGGTGCCGCGCAGCGCCG




1657
TCGGCTCGGCCATGCTGCGCATGGCCTTCGCGCGGCGCCATCTGGCC





GGTCTGCGGTTCGGCAAGTTCCTCGGCACCGGCACCGGCACCGGCTT





CGGTCCCGGCGACACCGATCTCACCCGGTGGGCGGCGATCACGGTCA





GTGATGCGCCGGTACGTTTCCCCGTCTGGGAGCGGATCGCCGTCAAC





GGCGCCCGGATCGATCTGGAGCCACTGATCAGCCGGGGCACCTGGG





CCGGCCGTACCCCGTTCGAGCCCACCGGCCGCCGCCCGGACGGTCC





GGTGCTCGCGCTCACCCGGGCCCGGCTGCGGCCGGCTCGCGCGCTG





ACCTTCTGGCGGGCGGTCCCGGCGGTGGTGCGCGAGGTGCACCGGG





CGCCCGGGCTGCTCGCCCGGTTCGGCGTCGGCGAGGCGCCGATCGG





CTGGCAGGGCACCGTCACCGTGTGGCGGGACGCGGCGGATCTCGTC





GCGTTCGCGTACCGTCAGCCGGAGCATCGCGCGGCGATCGCCCGGA





CCCCGGCCGACCGCTGGTACGCCGAGGAGTTGTTCGCCCGGTTCGCG





GTGCTCGGGATCAGCGGTGACCGGTCCGTGCTGGGCTGGACCGCCG





ACGAAGGGGAACGGGCGGAAGCATGA





48
DNA
>ACSP50_
ATGACACAGACCATCGTGATCACCGGGGCCAGCTCCGGGGTCGGGCT




1658
GGCCGCCGCCGAGCAGCTCGCCGCCCGCGGTGACGAGGTGGTGCTG





GTCGGCCGCGACCCGGGCCGGCTCGACGCGGCCGTGCAGCGGGTCC





GGGAGGCCGGCGGCGGCCGCGCGCCCCGGCACTTCCGGGCCGACTT





CGAACGGCTCGACGACGTGCGGGAGCTCGCCGCCGGGCTGCTGGCC





GAGCTGCCCCGGATCGACGTGCTGGCCAACAACGCCGGCGGGATCAT





CAAGCGGCCCCGGCAGACGGTGGACGGCCACGAGGCCACCATCCAG





GGCAACCACCTGGCCCCGTTCCTGCTCACCCACCTGCTGCGGGAGCG





GCTGACCGGGGGCCGGGTGGTGAACACCGCCTCGGCGGCACACGTG





CAGGGCCGGCCCGGCACCCGGTTCACCGACGACCCGAAGTCGTACA





GTCCGTGGCGCTCCTACGGGGCGAGCAAGGCGGCCAACATCCTGTTC





GCCGCCGAGGCCGCCCGCCGCTGGCCGGACGTGTGCAGCGTCTCGT





TCCACCCCGGTGTGGTGCGCACCAACTTCGGGGAGGGCCGGCTGATC





CGGCTGTTCTACCGGTACGCGCCCGGCCTGGTCACCCCGGAGGCCG





CCGGCGAGCTGCTGACCTGGCTGTGCACCACCCCGGCCGGGGAGCT





GGAGAACGGCGCCTACTACGTCAAGCGTCAGGTGACCCGGCCGGCC





GCGCACGCCCGCGACCCGCGGCTGGCCGCCGAGTTGTGGGACGCCA





GCCTGACCGCGACCGGCCTCGCCGGATGA





49
DNA
>crtE
GTGATCGACGACTTCCTCAGCGCGCAACGCGACGTGCTGGCCGAGGT




(ACSP50_
CAGCGACGACTGCGCGCCGCTGGAACGCTACGTGGCCGACCTGATGG




3873)
GCGGCGGCAAACGACTCCGGCCGGCGTTCTGCTACTGGGCGTGGCG





GGCGGCCGGCGCCCCCGACGGCCCGGGCATCGTGGCGGCCGCGAC





ATCCCTGGAGTTCCTGCAGGCCGCCGCGCTGATCCACGACGACATCA





TGGACGATTCGGACACCCGTCGCGGCGCCCCGGCGGTGCACCGCAG





ACTGGCGGCCCTGCACTCCGGCGGCCGCTGGGCCGGGGACGCCGAC





CACTTCGGGCTGTCCGCCGCCGTGCTCGCCGGCGACCTGTGCCTGAC





CTGGAGCGACGCGTTGTATTCGGGCAGCGGCCTGCACCCGTCCGCGC





TGGCCCGGGGCCGGCCGGTCTTCGACCGGATGCGCACCCAGCTGAT





GGGCGGCCAGTATCTGGACCTGCTGGACCAGGCGCGGCCGTCCCGG





GGCGGCGTCGACCGGGCGCGCCGGGTGGTGCACTTCAAGAGCGCCA





AGTACACCGTCGAACATCCGCTGCTGCTCGGCGCCCGGCTCGCCGGC





GCGGACGACGATCTGCTCGCCCGGTTGTCCGCGTTCGGTCTGCCGCT





GGGCGAGGCGTTCCAGCTGCGCGACGACCTGCTCGGGGTCTTCGGC





GACGCGGCGCAGACCGGCAAACCCACCGGCGACGACCTGCGCGAGG





GAAAGCGCACCACGCTGGTCATCCTGGCCGCGGACCGCGCCACCGCA





CCCCAGCAGGCCGCCCTCACCGCGCTGCTCGGCGATCGCGGCCTGA





CCGGGGCCGGCGTCGACACCCTCCGGCAGATCATCGTGGACACCGGT





GCCCGGGCCGAGGTCGAGCGGATGATCGAGCAACTGCTGGCGACGA





GTCTCGGCGTGCTCAGCGGCACGCCCGTCGACGAGGCGGCCCGCTC





GGTGCTGCTCGCCCTCGCCGAGGCGGCGACCGCCCGCAGCTCCTGA





50
DNA
>ACSP50_
ATGGTGAGCACAGTGATCGCCTCGGGGCCCACCGGCCTGGGCACCTC




1950
CGCGGCCCGTCTCTTCGGTCGGGTGGACCGGGACGAGCCGGAGCTC





TTCTGCCCGGCGCCGCTGCGCGACGACCGGGCGCTGGGGGAGCGGG





TCAACGACGCCGTGGTCCAGTGGGCCGAGAAGGCCGGCATCTACCCC





GGCCGGCTGGACAAGCTGCGCGGGGCGAACTTCGGCCGCTTCATGAT





GCTCGCCCACCCGGCCACCAGCGATCCCGACCGGCTGCTCGCCGCG





ACGAAGTGTCTGGTCGCCGAGTGGGCGGCGGACGACTACTACGTCGA





CGAGGTGTCCCTGGGCGCGGATCCGATGGTGGTCGGCTCGCGGCTG





GCCAACCTCTACTCGGTGGTCGACCCGGCCTCGCTGACCCCGCGCTA





TCAGGCCGACTTCGAGAAGCATCACCGCCTGCAGCCGATCTCGGTGG





CGTTCCGCACCGCGATGGAACACCTGGCCGAGTACGCCTCGGTCACC





CAACTGGCCCGGTTCCAGCACCAGATGGCGATCCTGTTCGTCGCCTG





GTCGCAGGAGGCCGACTGGCACGCCAACCGGCGCACCCCGCCGGTC





TGGGAGTATCTGGTGCAGCGGCACCTGAACAGCTATCTGCCGCCGAT





GATCCTGGTCGACGTGCTGGCCGGGTACGAGCTGTCGCCGGCCGAGT





TCTTCGATCCGCGGGTCCGCGCGGCGTTCACCACCGCAGGCAACGCC





GCCGTGCTGGTCAACGACCTCTACTCGGGCAGGAACGAGTCCGAGAC





CGATCACAACCTGCCGACCGTGCTGGTGTCCGGGGAGCGGCTCACGC





CGCGGGCCGCGGTCCGGCGCACCGTGGAGATCCACAACGAGTTGAT





GCACACCTTCGTGACCTCGGCCGCGTCGTTGAGCGCGTCCGGCTCGC





CGCAGCTGCGCCGGTTTCTCGCGGACACCTGGGCCTGGCTGGGCGG





AAGTCGCGAGTGGCACGCCACGAGCGGCCGCTACCACTCATCCAACT





GA





51
DNA
>ACSP50_
ATGACGACCACCGCACCGACTCCCGCCCACCTCGCCGGCAACTTCGC




5522
GCCCGTCACCGGGGAGACCACCACGCTCGACCTGCCGGTCACCGGC





GCCGTCCCGGCCGAACTGACCGGGTGGTATCTGCGCAACGGGCCCAA





CCCCCACCACGGGACCTCGGCGCACTGGTTTCTCGGCGACGGCATGG





TGCACGGCGTCCGCCTCGATCACGGCCGGGCCACCTGGTACCGCAAC





CGCTGGGTGCGGACCCGGGTGCTGACCGACGACGCCCGCGCCTACG





GCCCGGACGGCACCCGCGACCTCACCGCCGGCCCGGCGAACACCAA





CGTCGTGCGCCACGGCGGACGACTGCTGGCGCTGGTCGAGTCCGCG





CTTCCGTACGAGATCACCACCGACCTGGAGACCGTCGGCCCCTACGA





CTTCGGCGGCCGCCTGCACACCCCGATGACCGCCCACCCCAAGGTCT





GTCCCACCACCGGGGAGATGCACTTCTTCGGCTACGGCGGACTCGAG





CCGCCCTACCTCACCTACCACCGCGCCGGCGCGGACGGCCGGCTGT





CGCTCAGCCGCCCGATCGACGTCCCCGCGCACACGATGATGCACGAC





TTCAGCCTCACCGCGGCCCACGTGATCTTCATGGACCTGCCGGTGCT





GTTCAGCCTGGACGGGGCGCGGACCGGCGGCATGCCGTACCGGTGG





GACGACACCTACCAGGCGCGCCTGGGCGTGCTGCGGCGCGACGCCC





CGCAGGGGGAGGTCCGCTGGTACACCATCGATCCCGGATACGTCTTC





CACACCCTGAACGCCCACGACGACGGCGACCGGATCGTCATGCACGT





CGTCCGCCACGAGCACGCGTACCGCCCGGGGCAGCCCGCCGCCGCA





CCGGACCTCTGGCGCTGGACCATCGACCAGCGCACCGGCCGGGTCG





CCGAGGAACGGCTGGACGACGAAGCGGTCGAGTTCCCCCGCATCGAC





GATCGGCGCACCGGGCAGCCGGCCCGTTACGGCTTCGCCGTGACCG





ACAACGTTCCCCGCCGGCTCGCCGACGTCAGCGCCGTCATCCGCTAC





GACCTGCACACCGGCTCGACCACCCGGCACCGCCTGCCGACCGGGC





AGGTACCCGGGGAGGCGGTCTTCGTGCCGGCCGGCGGCGCCCCCGC





CGGATCGGCCGACGGCTGGCTGCTGACGTTCGCCTACGACCCGGGG





CGCGACGCCAGCGATCTGATCATCATCGACGCCACCGACCTCGCCGC





CCCGCCGCTGGCCCGGATCCACCTGCCGCACCGGGTGCCGTTCGGC





TTCCACGGCAACTGGCTGCCCGACCACGACCGCGCAGAATAG





52
PRT
>Dxs
MSDSPSTPAGLLASVTGPGALKRLSAEQLTLLAAEIRDFLVAKVSKTGGHL




(ACSP50_
GPNLGVVEMTLAMHRVFDSPRDKILFDTGHQAYVHKIVTGRQDGFDLLRQ




7096)
RGGLTGYPSQAESEHDLIENSHASTALSYADGLAKAFALRGEDRHWAVV





GDGALTGGMCWEALNNIAATKNRLVIWNDNGRSYAPTIGGLADHLSTLRL





NPGYEKVLDLVKDALGSTPLVGKPVFEVLHAVKRGIKDAVSPQPMFEDLG





LKYIGPVDGHDQQAMESALRRAKGFNAPVIVHAVTRKGYGYRPAEQDEA





DCLHGPGAFDPQTGALTAKPSLKWTKVFAEELVKIADERPDVVGITAAMA





EPTGIAALAKKYPDRAYDVGIAEQHAATSAAGLAMGGLHPVVAVYATFLNR





AFDQVLLDVAMHRLPVTFVLDRAGITGPDGPSHYGIWDMSVFGAVPGLRI





AAPRDAATLREELREAVAVDDGPTIVRFPTGAVAADTPAVRRVGQVDVLR





EAEKKDILLVAVGSFVGLGLDAAERLAEQGYGVTVVDPRWVRPVPIELTGL





AAQHRLWTLEDGIRAGGVGDAVAAALRDAGVHVPLRDFGVPAGFHPHG





TRAEILASLGLTAQDVARDVTGWVSGLDAGTSVAAPAI





53
PRT
>lspG
MTAISLGMPAVPPPPLAPRRQSRQINVGGVLVGGGAPVSVQSMTTTLTSD




(ACSP50_
VNATLQQIAELTAAGCQIVRVAVPSQDDVEALPAIAKKSQIPVIADIHFQPKY




7248)
VFAAIDAGCAAVRVNPGNIRQFDDKVKEIARAASDAGVPIRIGVNAGSLDK





RLLEKYGKATAEALVESALWECSLFEEHGFRDIKISVKHNDPVVMIRAYRQ





LAEQCDYPLHLGVTEAGPAFQGTIKSAVAFGALLAEGIGDTIRVSLSAPPVE





EIKVGQQILESLGLRERGLEIVSCPSCGRAQVDVYTLAEQVTAALDGFPVP





LRVAVMGCVVNGPGEAREADLGVASGNGKGQIFVKGKVIKTVPEAVIVET





LVEEALRLADEMGAELPDELRELLPGPTVTVH





54
PRT
>Dxr
MRELVLLGSTGSIGTQAIDIVRRNPELFRVVAIGAGGGNVALLAAQALELGV




(ACSP50_
EVVGVARASVVQDLQLAFYAEAQKRGWSSGDFKLPKIVAGPDAMTELAR




7250)
WPCDVVLNGVVGSLGLAPTLAALESGRILALANKESLVAGGPLVRRIAKDG





QIVPVDSEHSALAQCLRGGRAAEVRRLVLTASGGAFRGRRRAELTNVTPE





EALKHPTWDMGPWTINSATMVNKALEVIEAHELFGVPYDDIAVMVHPQS





VLHSLVEFTDGSTLAQASPPDMRLPIALALAWPDRVPGAAAAVDWTLAHN





WELRPLDDEAFPAVELAKAAGRYGRCRPAIFNAANEECVAAFAAGRLPFL





GIVDTLERVLAAAPDFAEPSTVDDVLAAESWARAQAQRTIATVAEGA





55
PRT
>lspH
MLLAKPRGYCAGVDRAVQTVEEALKLYGAPVYVRKQIVHNKHVVSTLEAR




(ACSP50_
GAIFVEENYEVPEGATWFSAHGVAPEVHDQARERRLKAIDATCPLVTKVH




7707)
HEAKRFAAEDYDILLIGHEGHEEVIGTSGEAPAHIQLVDGPDDVANWVRD





PAKVVWLSQTTLSVDETMETVARLKTRLPLLQSPPSDDICYATSNRQHVIK





EIAPECDVVIVVGSTNSSNSVRLVEVALGAGARAGHLVDYAAEIQDEWLAG





ATTVGVSSGASVPDELVMEVLAHLAERGFGEVTEFTTAEERLTFSLPQEL





RKDMKAAEAARAAAAG





56
PRT
>lspE
MTEAWGPDDDEPRPYSGPVKVRVPAKINLHLAVGPLRPDGYHELNTVYH




(ACSP50_
AISLFDEITARHGDTLTLTMEGEGTGDLALDETNLIIRAARALAARARVPAY




7802)
ARLHLRKSIPLAGGLAGGSADAAATLIACDLLWGLGMSRDELAEVGAQLG





SDIPFLLHGGTALGTGHGEAVSPILARPTTWHWTVAIADGGLATPAVYREL





DTLRAGTWPPTPLGSADTLMAALRQRNPEILGAALGNDLQPAALALRPQL





ADVLKAGTEAGALAGLVSGSGPTCVFLAADATHAQEIADSLTEAGVCRAA





VTARGPQPGARVI





57
PRT
>lspF
MIIPRVGIGTDVHAFDADRACWVAGLEWPGEPGLAGHSDADVVAHAACD




(ACSP50_
ALLSAAGLGDLGGNFGTSRPEWAGAAGVTLLAETARLVRAAGFAIGNVSV




8046)
QVIGNRPKIGKRRAEAEKVLSAAVGAPVTVSGTTSDGLGLTGRGEGLAGV





AVAMVYTENALPA





58
PRT
>lspD
MIADRDVTAQLNARGDVAVVVPAAGAGLRLGPGGPKALRLLDGEPLLVHA




(ACSP50_
VRRLAAAAPVRMIVVAAPPAEVDAVSALLAPVAPVTWPGGAERQESVAA




8047)
ALAVVPPDVPIVLVHDAARCLTPPSVTERVAAAVRDGADAVIPVLPVVDTIK





EVAADATVLGTVDRSVLRAVQTPQGFRASVLRAAHRAAADSHTDDAGAV





EKLGIPVLCVPGSDLALKITRPIDLALATHLLALPDPDAPTA





59
PRT
>ldi
MSSIGHLNREDHLVELVNEEGQPLGSATVSDAHLSPGALHRAFSVFLTDD




(ACSP50_
EGRVLLQQRAAAKTRFPLRWGNTCCGHPAPGEPVTVAAARRLTEELAVR




0146)
DVTLTEIGVYTYRATDPVTGRVEHEYDHVLIGALPDGVVPHPDPAEIATLR





WASLPGLRTGLTESPELYAPWLPGVFEILTERSGVLSTERSGGR





60
PRT
>CrtE
MANDTLEGNRLAAIPRQSVSHTGLVGAVEGTLADFLASQIASLDAVDPSLG




ldsA
GFGRTARDLVMAGGKRLRPTFAYWGWRGVAGPAADAETLLPALGALELM




(ACSP50_
HTFALVHDDVMDDSSTRRGRPTAHRIFAAQHGGRFGTSAAILVGDLCLVW




0148)
ADQLLARTPVPAATLLAVRAHYDRMRIEAVAGQYLDVLGETDPASWSVER





ALLVARHKTASYTVQRPLDFGLALAGVEDVEVAEAYRTYGIAVGEAFQLRD





DLLGVYGDPAVTGKPVSDDLRTGKPTALLMLARRMATPGQLAELESAEIE





RKAQWAETGAPARVEEMIRARVTEGLTALASAPIDAEARATLIELATVATQ





RPA





61
PRT
>CrtB
METDLAAAYERCRELHREHGRTYYLATRLLPAWKRRHVHALYGFTRFAD




(ACSP50_
EIVDRTEAQPPAERAAELATWSAGFLAGLRGEPVDDPLLPAVLHTIAVFGL




0149)
DLEDFAKFLRSMEMDLTVTGYRTYDDLLDYMEGSAAVIGTMMLPILGSTD





PAAAREPARQLGFAFQLTNFIRDVAEDLARDRIYLPEEHLAEFGVTRADLA





AGVATPAIRALIRAEVDRAREHYAAAAPGIPLLERTSQACMRTAFQLYGGIL





DEIEAADYDVFARRVTVPNRRRAAVAVRSLLTRPGTPVELAA





62
PRT
>ACSP50_
MGARVALFTRDLRIHDNPLLSGPDPVVPLFVLDPRLSGLSANRSRFLHQSL




0150
ADLRNSLRERGADLVIREGDPVAETIAVASEVDASTITVAADVTGYAQRRE





RRLRDERFRVKTVPSVTVLPPGTVRPGGGGESYRVFTPYFKAWEKAGWR





APSATPGKVAMPAGIAPGRLPEMPAGDSPDAVAGGETEGRRRLQAWQK





EMARYAEDHDDMAADNTSRLSAYLRFGCLSPLELALAAKADDSPGAQAYL





RQLCWRDFYYQVTATFPEISTRPLREKADQNWRYDDDALRHWQDGLTG





VPIVDAGMRQLRAEGWMHNRARLITAAFLTKHLGIDWRPGLQWFFRWLL





DGDVPNNSGNWQWTAGTGNDTRPYRRFNPIRQAQRFDAQGVYVRRYVP





ELKDIDGVTVHQPWRLPESVRRGLDYPGPLESHRDEAVWLRD





63
PRT
>ACSP50_
MSEARQVDVVVVGLGVGGEEVAGRLAAAGLSVIGVEHRLVGGECPYWG




0151
CIPTKIMVRAGNALAEARRIPGLAGTSTVRADWAPVAKRIRDEATDDWND





KVAVERFTGKGGTFVRGTAELTGPGQVRVGDQEFAASRGVVIATGTAAV





VPPIEGLSGTPFWTNREAVEAAALPASMLVLGGGAIGCELAQAYARFGVQ





VTVIEGSPRVLAMEEPESSEVAAAALTADGVRIVTGVRAQKVAHDDGFHV





TLSDGSVLAGEKLLVATGRAARLGGLGLDRVGLDPSARFLATDDRLRAGE





GIWAVGDVTGNGAFTHMAMYEADIAVRDILGQGGPGADYRARPRVTFLD





PEIGAVGMTEQQARDAGLEVRVGYVPLNQTSRGFIHGPGNEGFLKLVAD





GERGVLVGGTTAGQSGGEMIGAVAVAVHAEVPVSTLLSQIWAYPTFHRGL





GQALQSLA





64
PRT
>ACSP50_
MSEPVITEPAAWINLPDLSERLDVSISKVHQMIRDGDLLAVRRDGIRVVPAE




1631
LVANATVLKHLPGVLNVLRDAGYNDEEAFRWLYAEDAEVGGSAAIALGGQ





QAREIKRRAQALGF





65
PRT
>ACSP50_
MRHLSYVAVLAGCLAGALWLEPILRVNVLRRWRRLLLAVLPMAVVFTLWD




1632
LAAIAAGHWHFDPAQITGVYLGGGLPLDEVLFFLVVPVCAILGFEAVRAVLR





RPAGDE





66
PRT
>ACSP50_
MTYTTAAVLGVLAALTLDVLILRTRLVGRLVFWATYPIIFVFQLISNGILTGRD




1633
IVMYDPAAILGPRLVHAPVEDLLFGFALVLGTLSLWVALGRRGIQRTPRAG





SRRTDE





67
PRT
>CrtE
MTNSPLDEAGLRSRVDKALTVFLAGQRDRLLAIDPALAEMSATVSEFVLG




fps2
GGKRLRPAFAYWGFRGAGGADSDAVVAAVAALELVQASALIHDDLMDRS




(ACSP50_
DTRRGVPSVHRRFEKLHAGEGWRGSAAGFGDCAAVLLGDLALVWSDELL




1634)
HTSGMAVADVQRARPIFDGMRTEVTVGQYLDVLTQATGDTSLERAGKVA





VYKAAKYTVERPLLLGAALAGAAPGVHAAYSAFGLPLGEAFQLRDDVLGV





FGDPERTGKPAGDDLREGKRTYLVAAAFGALDAAGRAELDAALGDPGLD





EAGVARLRTVIRDSGALAATEARIDELMTASIGALDAAPIDQDAREVLRRLA





DAATRRSV





68
PRT
>ACSP50_
MSLGLPSRLPGTPSIGDLVRGAAPTFSFEFFPPKTPDGERLLWQAIRELES




1635
LRPSFVSITYGAGGTTRETTVAVTERVATETTLLPLAHLTAVDHSVADLRN





VIGRLAGAGIRNVLALRGDPPGDPMGEWVRHPDGVGYADELVRLIRESGD





FSVGVAAFPHKHPRSAGVKDDTRNFVRKCRAGADYAITQMFFDADEYLRL





RDRVVAAGCHTPIVAGVMPVTRMATIARSTQLSGAPFPPALLRDFERVAG





DDAAVRELGIETCAAMCARLLREGVPGIHFITMNRSTATREVWQRLAPAE





VAASA





69
PRT
>ACSP50_
MQLQQLRYFLAWETRHFTQAADILGVSQPTLSKQIHTLEMSLGAPLFERM




1650
RGAVTLTVAGETLLPMAQRIVADADAARDAVQDIVGLRRGEVRLGATPSL





CSSLVPAVLRTFRADHPGVKLHISEGSSHDLTAGLLAHTLDLALIVQPEHG





VDPALVAIELLRESLVVASVAAGPPPTVGRQLELSELRHTPMVMFREGYDI





REVTLHACERAGFAPKFAVEGGEMDAVLAFVEAGLGVALVPSMVLANRPL





LRATPLAPPGMRRTIALAQRRAAVLPHAAAALREVVLDHIGSGRLPFGVRA





LERPST





70
PRT
>ACSP50_
MGEFHDPRLVEVYDAECPWGWDDDFFMAVLAERSAHRVADLGCGTGRL




1651
AIAMAAAGHEVIAIDPAPAALAAARRKPGGTRVRWLQGSAERLAPRSLDA





AFMTGHVAQSFVDDEEWDTVLRGLRRALVPEGRLVFDSRDPDDRPWQQ





WNPQDSWRTVVLDDGRVVEAWSEAEQVGLNTVRVTGRYRFADGGELAN





SATLRFRTEPELRDSLREAGFRVERIYGGWGREPVGLSGDGEFIVIAVATP





RLMS





71
PRT
>ACSP50_
MPENEWPDDPRPPDQGEWSQPHHEPPPGRGRALLAAAVVVLVLLAAGGI




1652
AWRLMSSRGATPVAQPTAPAPTPTAQTAPPCPQPRLRVAAAPEIAPVIQQ





AAAALSQPGQRCSEVLVQAAEPGAALTGKPDVWVPSSSVWLALAKSRGD





VYTTQGASLAWSPLVIAGPESIASLFAPNGVTSWSGLVQGTIQKRVPAVR





MPDPTLTTTGLLSVYAVGQATVKANPDAGIAQLQALTLRSRLENAAADPAE





LFAQMGKQTDAATAIYQVGVFPTTEQQLLTYQKSQHDVRLSGSAPADGQI





DADYPYAVRKGAPADLVESLREAITPDALTTAGFRATATKNALRLPAPAVL





AGAARQWSAYKSVAFQVLLLIDASGSMNEKITDRAGRSVTKAALLRESGT





SAAQLFGDDTSLGLWFFGTPTADSPAHTEEVPFGPVIATVDGKSRRDLLA





AKIGEYRPVANAGTPLYQSVLDGVAEMRGRAKPDTATWVVLTDGSDGG





TKYRMSNADFLKKLTAGADPAKPVPVIAVGYGPAANATALQAMAKATGGQ





AVTVKNPADLAAGIAQAFLAAHTH





72
PRT
>CrtD
MSDIVWGAGVGGLAAAIRLAEAGHRVSIHERSGWGGKLAAYERDGYRF




(ACSP50_
DTGPSLLTLPDVFTGLGLDLRPEPLDPVVRHFFPDGTVLDSSSDHETFLAR




1653)
ITDALGGAAARDWDRFWRRAERIWHASWESVLRRPVTAASLARLSWRLG





DLAAIAPGRSLRSLGRRYLRDPRLRMLLDRYATYSGADPRRAPAALAAIPY





AELAFGGWYLPGGLVTLAEALLARCEKLGVRVHLHSPVASIATTGARVSG





VRLGDGTRLAADVVVSNVDAVTLYRDLLPSPKPLARLADRSLAGFVLLLAV





RGETPRLAHHNVFFPRDYDAEFDAVFGGPGRRARPAGDPTVFVTRAADP





AVRPAGDEAWFVLVNAAPHGTSWSTVDWLRAGLADAYRDRVLEVLAGR





GLDVRDRLIFAETRTPADLAASAAAPGGAIYGTAGGLVRPANRAPVDGLFL





VGGSTHPGGGLPMVTLSAEIVAGMIGSN





73
PRT
>CruC
MIVAWLILPPLLLITAHTAVNALLLRRPRRAATSTERVAVLLPLRDEATRVTP




(ACSP50_
CLRALLAQRGVADLTVHVLDDGSTDGTADVVRAVAGDRVRLHTGTPPPP




1654)
GWLGKPAACQRLADLAGDVDVLVFVDADWLAPDAVAGAVDLLRRAGAD





LLSPYPKIVGAGRLVQPLLQWSWLSFLPLRAMERSARPSLAAAGGQWLVL





DRAGYRRAGGHAAVRGEILEDIALARAVKRAGGRIALADGSGLATCRMYE





SWDELADGYAKSLWASLGSAAGATAVTLLLILLYVVPPLLAPFAPLPAVLG





YLLGVTGRMIAARATGGRVLPGTLAHPVSIVLFGYLIARSFRLRRAGRLAW





RGRPVP





74
PRT
>CruF
MSPRHLPWGLLGALVLAQICYPLTEGDTRAGLTVLTVLLGVAFSLSHALLT




(ACSP50_
RGPRALTALLSTATLGGFAVEAIGVATGFPFGSYEYSGRLGPRLLGVPLIIP




1655)
LAWTWMAWPAWLAALRVTRRRLPRILVAGAGLAAWDVFLDPQMVAEDY





WRWRHPVPALPGVPGVPLGNYLGWLGFALLLMTALAAVAGRAADRPLSA





DRPALALWIWTYASSVLAHAVFLSLPASAAWGALIMGAAVLPLLARLRAPA





75
PRT
>ACSP50_
MRLVAWQPDDLLRRLDDVVGVYGEAMGYRQELLQTRRGYIGSHVRRPG




1656
FRAVATLTTEGRLMGFGYGYTSAAGQWWHDQVRFALGEDDRRQWLTDC





FEVVELHVRPAAQGHGVGARQLRALLAMAKGRTVLLSTPEADEQASRAW





RLYRRYGFADVLRHFYFPGDERAFAVLGRELPLAERPLEDAPGIAGA





76
PRT
>ACSP50_
MTHVALHVWRVPRSAVGSAMLRMAFARRHLAGLRFGKFLGTGTGTGFG




1657
PGDTDLTRWAAITVSDAPVRFPVWERIAVNGARIDLEPLISRGTWAGRTPF





EPTGRRPDGPVLALTRARLRPARALTFWRAVPAVVREVHRAPGLLARFGV





GEAPIGWQGTVTVWRDAADLVAFAYRQPEHRAAIARTPADRWYAEELFA





RFAVLGISGDRSVLGWTADEGERAEA





77
PRT
>ACSP50_
MTQTIVITGASSGVGLAAAEQLAARGDEVVLVGRDPGRLDAAVQRVREAG




1658
GGRAPRHFRADFERLDDVRELAAGLLAELPRIDVLANNAGGIIKRPRQTVD





GHEATIQGNHLAPFLLTHLLRERLTGGRWNTASAAHVQGRPGTRFTDDP





KSYSPWRSYGASKAANILFAAEAARRWPDVCSVSFHPGVVRTNFGEGRLI





RLFYRYAPGLVTPEAAGELLTWLCTTPAGELENGAYYVKRQVTRPAAHAR





DPRLAAELWDASLTATGLAG





78
PRT
>CrtE
MIDDFLSAQRDVLAEVSDDCAPLERYVADLMGGGKRLRPAFCYWAWRAA




(ACSP50_
GAPDGPGIVAAATSLEFLQAAALIHDDIMDDSDTRRGAPAVHRRLAALHSG




3873)
GRWAGDADHFGLSAAVLAGDLCLTWSDALYSGSGLHPSALARGRPVFDR





MRTQLMGGQYLDLLDQARPSRGGVDRARRVVHFKSAKYTVEHPLLLGAR





LAGADDDLLARLSAFGLPLGEAFQLRDDLLGVFGDAAQTGKPTGDDLREG





KRTTLVILAADRATAPQQAALTALLGDRGLTGAGVDTLRQIIVDTGARAEVE





RMIEQLLATSLGVLSGTPVDEAARSVLLALAEAATARSS





79
PRT
>ACSP50_
MVSTVIASGPTGLGTSAARLFGRVDRDEPELFCPAPLRDDRALGERVNDA




1950
WQWAEKAGIYPGRLDKLRGANFGRFMMLAHPATSDPDRLLAATKCLVAE





WAADDYYVDEVSLGADPMVVGSRLANLYSVVDPASLTPRYQADFEKHHR





LQPISVAFRTAMEHLAEYASVTQLARFQHQMAILFVAWSQEADWHANRRT





PPVWEYLVQRHLNSYLPPMILVDVLAGYELSPAEFFDPRVRAAFTTAGNA





AVLVNDLYSGRNESETDHNLPTVLVSGERLTPRAAVRRTVEIHNELMHTFV





TSAASLSASGSPQLRRFLADTWAWLGGSREWHATSGRYHSSN





80
PRT
>ACSP50_
MTTTAPTPAHLAGNFAPVTGETTTLDLPVTGAVPAELTGWYLRNGPNPHH




5522
GTSAHWFLGDGMVHGVRLDHGRATWYRNRWVRTRVLTDDARAYGPDG





TRDLTAGPANTNVVRHGGRLLALVESALPYEITTDLETVGPYDFGGRLHTP





MTAHPKVCPTTGEMHFFGYGGLEPPYLTYHRAGADGRLSLSRPIDVPAHT





MMHDFSLTAAHVIFMDLPVLFSLDGARTGGMPYRWDDTYQARLGVLRRD





APQGEVRWYTIDPGYVFHTLNAHDDGDRIVMHVVRHEHAYRPGQPAAAP





DLWRWTIDQRTGRVAEERLDDEAVEFPRIDDRRTGQPARYGFAVTDNVP





RRLADVSAVIRYDLHTGSTTRHRLPTGQVPGEAVFVPAGGAPAGSADGW





LLTFAYDPGRDASDLIIIDATDLAAPPLARIHLPHRVPFGFHGNWLPDHDRA





E





81
DNA
>tipA
atccctagaacgtccgggcttgcacctcacgtcacgtgaggaggcagcgtggacggcgtggtaccaag




promoter
cttattggcactagtcgagcaacggaggtattccg





82
DNA
>gapDH
gtactggccgatgctgggagaagcgcgctgctgtacggcgcgcaccgggtgcggagcccctcggcga




promoter
gcggtgtgaaacttctgtgaatggcctgttcggttgctttttttatacggctgccagataaggcttgcagcat





ctgggcggctaccgctatgatcggggcgttcctgcaattcttagtgcgagtatctgaaaggggatacgc





83
DNA
>lacZ?
taatgtgagttagctcactcattaggcaccccaggctttacactttatgcttccggctcgtatgttgtgtgga




promoter
attgtgagcggataacaatttcacacaggaaacagctatgacatgattacgaattcgatatcgcgcgcggcc




and gene
gcggatcctctagagtcgacctgcagcccaagcttggcactggccgtcgttttacaacgtcgtgactggg





aaaaccctggcgttacccaacttaatcgccttgcagcacatccccctttcgccagctggcgtaatagcga





agaggcccgcaccgatcgcccttcccaacagttgcgcagcctgaatggcgaatggcgcctgatgcggt





attttctccttacgcatctgtgcggtatttcacaccgcataaattccccaatgtcaagcacttccggaatcggg





agcgcggccgatgcaaagtgccgatcaacataa





84
DNA
>T4
aagctttatgcttgtaaaccgttttgtgaaaaaatttttaaaataaaaaaggggacctctagggtccccaatt




terminator
aattagtaatataatctattaaaggtcattcaaaaggtcatcca





85
DNA
>PhiC
gtggacacgtacgcgggtgcttacgaccgtcagtcgcgcgagcgcgagaattcgagcgcagcaagcc




31
cagcgacacagcgtagcgccaacgaagacaaggcggccgaccttcagcgcgaagtcgagcgcgac




integrase
gggggccggttcaggttcgtcgggcatttcagcgaagcgccgggcacgtcggcgttcgggacggcgga




gene
gcgcccggagttcgaacgcatcctgaacgaatgccgcgccgggcggctcaacatgatcattgtctatga





cgtgtcgcgcttctcgcgcctgaaggtcatggacgcgattccgattgtctcggaattgctcgccctgggcgt





gacgattgtttccactcaggaaggcgtcttccggcagggaaacgtcatggacctgattcacctgattatgc





ggctcgacgcgtcgcacaaagaatcttcgctgaagtcggcgaagattctcgacacgaagaaccttcag





cgcgaattgggcgggtacgtcggcgggaaggcgccttacggcttcgagcttgtttcggagacgaagga





gatcacgcgcaacggccgaatggtcaatgtcgtcatcaacaagcttgcgcactcgaccactccccttac





cggacccttcgagttcgagcccgacgtaatccggtggtggtggcgtgagatcaagacgcacaaacacc





ttcccttcaagccgggcagtcaagccgccattcacccgggcagcatcacggggctttgtaagcgcatgg





acgctgacgccgtgccgacccggggcgagacgattgggaagaagaccgcttcaagcgcctgggacc





cggcaaccgttatgcgaatccttcgggacccgcgtattgcgggcttcgccgctgaggtgatctacaagaa





gaagccggacggcacgccgaccacgaagattgagggttaccgcattcagcgcgacccgatcacgctc





cggccggtcgagcttgattgcggaccgatcatcgagcccgctgagtggtatgagcttcaggcgtggttgg





acggcagggggcgcggcaaggggctttcccgggggcaagccattctgtccgccatggacaagctgta





ctgcgagtgtggcgccgtcatgacttcgaagcgcggggaagaatcgatcaaggactcttaccgctgccg





tcgccggaaggtggtcgacccgtccgcacctgggcagcacgaaggcacgtgcaacgtcagcatggcg





gcactcgacaagttcgttgcggaacgcatcttcaacaagatcaggcacgccgaaggcgacgaagaga





cgttggcgcttctgtgggaagccgcccgacgcttcggcaagctcactgaggcgcctgagaagagcggc





gaacgggcgaaccttgttgcggagcgcgccgacgccctgaacgcccttgaagagctgtacgaagacc





gcgcggcaggcgcgtacgacggacccgttggcaggaagcacttccggaagcaacaggcagcgctg





acgctccggcagcaaggggcggaagagcggcttgccgaacttgaagccgccgaagccccgaagctt





ccccttgaccaatggttccccgaagacgccgacgctgacccgaccggccctaagtcgtggtgggggcg





cgcgtcagtagacgacaagcgcgtgttcgtcgggctcttcgtagacaagatcgttgtcacgaagtcgact





acgggcagggggcagggaacgcccatcgagaagcgcgcttcgatcacgtgggcgaagccgccgac





cgacgacgacgaagacgacgcccaggacggcacggaagacgtagcggcgtag





86
DNA
>PhiC
cccaggtcagaagcggttttcgggagtagtgccccaactggggtaacctttgagttctctcagttgggggc




31
gtagggtcgccgacatgacacaaggggtt




attachment





site






87
DNA
>incP
ccggccagcctcgcagagcaggattcccgttgagcaccgccaggtgcgaataagggacagtgaaga





aggaacacccgctcgcgggtgggcctacttcacctatcctgccc





88
DNA
>traJ
atggctgatgaaaccaagccaaccaggaagggcagcccacctatcaaggtgtactgccttccagacg





aacgaagagcgattgaggaaaaggcggcggcggccggcatgagcctgtcggcctacctgctggccgt





cggccagggctacaaaatcacgggcgtcgtggactatgagcacgtccgcgagctggcccgcatcaat





ggcgacctgggccgcctgggcggcctgctgaaactctggctcaccgacgacccgcgcacggcgcggt





tcggtgatgccacgatcctcgccctgctggcgaagatcgaagagaagcaggacgagcttggcaaggtc





atgatgggcgtggtccgcccgagggcagagccatga





89
DNA
>ColE
ttgagatcctttttttctgcgcgtaatctgctgcttgcaaacaaaaaaaccaccgctaccagcggtggtttgtt




1/pMB
tgccggatcaagagctaccaactctttttccgaaggtaactggcttcagcagagcgcagataccaaatact




1/pBR
gttcttctagtgtagccgtagttaggccaccacttcaagaactctgtagcaccgcctacatacctcgctctgc




322/
taatcctgttaccagtggctgctgccagtggcgataagtcgtgtcttaccgggttggactcaagacgatagt




pUC ori
taccggataaggcgcagcggtcgggctgaacggggggttcgtgcacacagcccagcttggagcgaac





gacctacaccgaactgagatacctacagcgtgagctatgagaaagcgccacgcttcccgaagggaga





aaggcggacaggtatccggtaagcggcagggtcggaacaggagagcgcacgagggagcttccagg





gggaaacgcctggtatctttatagtcctgtcgggtttcgccacctctgacttgagcgtcgatttttgtgatgct





cgtcaggggggcggagcctatggaaa





90
DNA
aac(3)
gtgcaatacgaatggcgaaaagccgagctcatcggtcagcttctcaaccttggggttacccccggcggt




IV
gtgctgctggtccacagctccttccgtagcgtccggcccctcgaagatgggccacttggactgatcgagg





ccctgcgtgctgcgctgggtccgggagggacgctcgtcatgccctcgtggtcaggtctggacgacgagc





cgttcgatcctgccacgtcgcccgttacaccggaccttggagttgtctctgacacattctggcgcctgccaa





atgtaaagcgcagcgcccatccatttgcctttgcggcagcggggccacaggcagagcagatcatctctg





atccattgcccctgccacctcactcgcctgcaagcccggtcgcccgtgtccatgaactcgatgggcaggt





acttctcctcggcgtgggacacgatgccaacacgacgctgcatcttgccgagttgatggcaaaggttccct





atggggtgccgagacactgcaccattcttcaggatggcaagttggtacgcgtcgattatctcgagaatga





ccactgctgtgagcgctttgccttggcggacaggtggctcaaggagaagagccttcagaaggaaggtcc





agtcggtcatgcctttgctcggttgatccgctcccgcgacattgtggcgacagccctgggtcaactgggcc





gagatccgttgatcttcctgcatccgccagaggcgggatgcgaagaatgcgatgccgctcgccagtcga





ttggctga





91
DNA
>cgt
GCCCGGCCCTGTCGAGCTGACGGCTGTCCCGCGGCCTCGTCATCGGT




promoter
GCTGTCGAGCAGGCTGTCGCCTGGTAGGAAGATTGCCATGGTCCAGA





TGGACCCCCTCAGCGCACGTCCCGATGGACGACGTTCCGTCTTGTCG





ACGACTCCGAGCCGCCCGACCCACCGGGCCTGAGCGCGCCCGATCA





CGGCTCCCCGGCCTGACGGGTTCTGCACCTCCGGCGGCTTTCCCGAG





GACGGCGTGGTGGTCGGTGACGGCTGCTGGACCTCCTCCGGTGGGC





AAGCGTTTCGGTGAGGTGGGCAGCCCGGCTGCGGGCACATCGGGGG





CGGAGAGACGCTTAGGTTTATTGCAAGTTCTTTCTTCGGTGGCGCGGC





GTGTCATCAGCAGCCGATTGTGGCATTCTGGTGACGCATTGACGCAGG





TCACAGATTTGTTGGGATAGGCAACGAACAATTCCTAAATCGCCTATTC





GGACAAATAGGCTTGACCTGACGACGCTGTCCCACCACTGTGGATGAC





GCCTACCGCGCAAGTTCTGGAAGTACTTGCAATCAGCGGTGAGGATCA





TCAAAGGGGACTGTC





92
DNA
>efp
TGGAGCACATCTGCCGGTAGACCCGATTCGCCCTCACCAGCGAATCG




promoter
CCGGTAAAGTGGTTCGGTCAACGATTCGAGTCAAGATCAAGGCAGGAC





ATGGCTTCCACCAACGACCT





93
DNA
>rpsJ
ATTGCGGGTTGTCGCCGGTGAGAGCCGGTGACAACCCCCACCGGTGA




promoter
CCCCGATTAGCAATGCTGCGTTCAATCGGGCATACTAGTCAGGTTGCG





TCCGCGCGGGGTGGGTGGCTGGCGTTCGTCAGCCGCCCACCCTCGC





CGGGTGTCCGGGTGTGTTTCCAGCCGCCCGGCGCCCTCAGATCCCCG





CGATCGCGTTCGTCCCCGGCAAGATCGGGGATGGAGGCCGAAAGCTG





AGTGCCCAGCACTCTGTGACGAGGCGCGACACGCCCGACCGCGGGG





GTCGGACAACGCAGGATCAACGGTCCTGCGGGCATGTGGGGGCCACC





GCCTCCGCACGTAGCGGCATCGAGAGAAGGAAACAGAAGCCACC





94
DNA
>katE
ATCTCGGGCTCGGTAGGCATCAGGCACTCGTTTCGTCGGGCTCTCGT




promoter
GACAGTGACCTTGATACTGGAGGGGTACGACAAAACCGGGACCGCCA





CCGACGTCCGGACCGACCCGATCGTCGGCCACGAACAGGGCCGGAT





GGTCGTCGTGACGCGTCCGCGAGACGCCGTCCGGGCCGGGCCGATG





CTCGGCCGGACCGTTTGCCGGGGTTCATGCGGGGTATCCGCCATCCG





ATCACATACCCTTATCGAGGAGTTTGTCCGG





95
DNA
moeE5
AGGGCGCCACCAGCTGGAGCCCCATCCCCGCGGGGACCAGGAGGGC




promoter
GAGCAGCGCCACGGCGGTCCGTTCACCGCGCAGGTAGCGGACAAAC





GTGGAGAGATGCCGCAACGGACTGTCTGCCAACGCGCCCCTCCCCCG





TTCGCCCGGCGGCGAGCGGCCAGCATAAAGTCCTGTGCGCCTCCTTG





TGAATGACGCCTCGTCAACGGCGGCCGGAGCACGCCCTTTCTGCGGG





AAGCCGATAGCGGACGCCGCTCCGGGAGGGGGCGAAGCACACCATT





GCTCGTGATTGACGCATGCTGTTAGACTCCCCACGTCTCTTGGTCCGG





ACATGCGTTTCTCAACGCCGAAAGCCTGGTCAACCGCACTTTCGGCAC





CGCACAGTCCCACGGCGTCCGAGCGGTCGCGCGAGTCGGCCCGGTC





GAGCCAGAGGCAGCCACACGAACGTGCACCGCAATGCACCGCCTTGA





TC





96
DNA
>apm
CTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTGAT




promoter
CCGGCAAACAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGC





AGCAGATTACGCGCAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTT





TTCTACGGGGTCTGACGCTCAGTGGAACGAAAACTCACGTTAAGGGAT





TTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTGGTTC





ATGTGCAGCTCCATCAGCAAAAGGGGATGATAAGTTTATCACCACCGA





CTATTTGCAACAGTGCCGTTGATCGTGCTATGATCGACTGAGC





97
DNA
cdaR
GGGCCCCCCCTCGAGGTCGACGGTATCGATAAGCTTGATTTTTCTGGA




promoter
CAACCTGGACGCCGAGACCTCGGCGGCGACCCTCGCGCTGTTCCTCA





CGGCCCTGTGGGCGCTGGCCGTCATCGCCCGCCCCTACACCTGGTGG





CGCGTGCTGCTGGTGCTGACCATGGCGGTGGGCTTCGCCGTGGTGCT





GGTGGTGCCCTACCTCCAGGAGTTCTTCCAGCTGAAGCTGGTCGGCG





TCACCGCGCCGTGGGCGGCGGTCGCCTGTGCGGCGGTCGCCGGGCT





GGTGCTGGAGTTGGTGTGGGCACGTATGCGGCGTCGTCTCGACGCCG





ACTGAGCCCACCGGGCGGTCGACCCCCGTACCGCCCGGTGAAGAGG





AGGGGACGCCCGGTCCGTGCCGGGCGTCCCCTCCGTCTTTGTGCGCC





CCCCGCCGACCGGAACGGCACGATCCGGCCAAACCTGCGCAGCGGT





GCGGCCGGAGGAGCCGCTTCCGGGCCGTTCGACGGGCGGCCCGCCA





CGGGACCGGAACGAGCCCCGGCATCCGCCGCCACCAGCGGATTTCAC





ATTCCTTACGCAATCGGCGGCGAGAGCGACCGGCAGGTAACCTCGGG





GCTGAATCCAGGCCATCGGGGAATAGCAAACGGCGCACTGACGAAAG





CAAGGGCAGAGACCTGCCGAAAGTTGAGTGTTGGATTCAAAGAAGATC





CGTATTATTCCGACTGCAGGCAGGGGGGAGCCGGCTACGAAGGAAAA





GTTCCGCAGGTCAGATTGGGCCGGGTCGCAGGCAGCGCCGCACCGG





CAACCACGACCGCGACTTTCGTCGACGCACCCCCTCGCACCGCCGCC





CGGCCACCGGTCCGGGCGCACGACCCGAAGGGAAGTGAGGCTCACG





CACGGACCAGCAGCTCCTGACGCAGCGACCCGGACCCGGAGGTGAG





TGACATGACGACGAGGCCCCGACCAGCGGTGAACCCTGCTGACCCGG





CCGTAACGAAGTCTTCATGCCCGTGGCACCCGACGGCTTCGGAGAGT





TTCGGCACGCAGACATCAGCACAACTTGACGCGGGGGTATCAAGAGG





TCATGGATCTTCGGTACC





98
DNA
ermE*
GCGGTTGATCGGCGATCGCAGGTGCACGCGGTCGATCTTGACGGCTG




promoter
GCGAGAGGTGCGGGGAGGATCTGACCGACGCGGTCCACACGTGGCA





CCG





CGATGCTGTTGTGGGCACAATCGTGCCGGTTGGTAGGATCCAGCGAG





CA





99
DNA
>rpsL
TGAGCACGTCCGCGAGCTGGCCCTGCAGGCGGAAGTCAGGTAGACAC




promoter
GACTTCCGCTAGTCCTTGCAAGGTCTGCTGACGTGAGGCGGGGCGGT





CGTTTTTGACCGCCCTGCCTTCGTCATGTAGGCTCGCTCGCTGTGCCT





GGCGTGTCATCAGACGCCCAGGTCCCGGTGCCGTGAGGCCCGGGCC





ATCGAGCCGGTGGTACGTGGCTGCGGTCCCCTTGTGAGGGCTGCGCG





CCGTGTGCTGTCCGGCGCGCACAGCCTTGAATCCACCCGCGGGGGCC





GGCCGGTCTCCGTGAGCTCG AGTAGACGACGGAGACGTA





100
DNA
>ACSP50_
GTGGCGACTCCCACGCAGTCCGAGATCCGCGAGGAAGAGCACGAAGA




1949
GCAGCGGCAGAGCCTGAGCACGGCGGCGGCCCGCAACCTCACGACC





ACCACCAAGACCGCGCCGCAGATGCAGGAGATCACTTCGCGATGGCT





GCTCCGTAAGCTTCCCTGGGTTCAGGTCGCCGGTGGGGCGTATCGGG





TGAACCGGCGGATGACTTATCGGATCGGCGACGGCCGGCTGAGCTTC





ACCAACGTCGGTGCGCAGGTCCGGGTCGTCCCGGCCGAGCTGCGGG





AACTCTCGGTGCTCAGCGAGTTCGACGACGCGGACGTGCTGGCCGCC





ATGGCCGACAAGTTCGTGCAGCAGGAGTACCAGCCCGGTCAGGTGAT





CGTCGAGTTCGGCTCGGTCGCCGACCACGTGTACGTGATCGCGCACG





GCAAGGTGAACAAGGTCGGCGTCGGCAACTACGGCGACCCGGTCAAC





CTGGGGGTGCTCGCCGACGGGGAGGCGTTCGGCGAGAAGTCGCTCA





CCGACGAGGAGCGGATCTGGGACTACACCGCCAAGGCGATGACCGC





GGTGACCCTGCTGGCCATGCCGCGCTCGGCGTTCACCGCGCTGCTCG





GCCAGAGTGACCACCTGCGCACGCACGTCGAGCAGTTCCGGGCCAAG





AACCGCCGGCCGCAGAACAAGCACGGCGAGGCGGAGATCTCGGTGG





CCGCCGGGCACACCGGCGAACCGAAGCTGGACGGCACGTACGTCGA





CTACGAGCTGACGCCGCGCGAATACGAGCTGAGCGTCGCGCAGACCG





TGCTGCGCGTGCACACCCGGGTCGCCGACCTCTACAACGAGCCGATG





AACCAGGTGGAGCAGCAGCTCCGGCTGACCGTCGAGGCGCTGCGCG





AGCGTCAGGAATACGAAATGATCAACAACCGCGAGTTCGGCCTGCTGC





ACAACGCCGACCTGCGGCAGCGCATCCACACCCGGGGCGGCCCGCC





CACCCCGGACGACCTCGACGAGCTGCTCAGCATGCGGCGCGGCACCA





GGATGTTCGTGGCCCACCCGCAGGCGGTCGCCGCGTTCGGCCGGGA





GTGCACCAAGCGGGGCATCTATCCACCGATGCTGGAACAGGACGGCG





GCACCTTCCTGTCCTGGCGCGGGGTCCCGATCCTGCCGTGCGGCAAG





ATCCCGGTGACCGAGACGCACACCACCTCGATCCTGGCGATGCGCAC





CGGGGAGAGCGACCAGGGTGTGGTCGGGCTGCACCAGACCGGGATC





CCGGACGAGTACGAGCCGAGCCTGTCCGTGCGGTTCATGGGGATCAG





CGAGCAGGCGATCATGTCGTACCTGGTGAGCGCGTACTACTCGGCCG





CGGTGCTGGTGCCGGACGCGCTGGGCATCCTGGACCACGTCGAGCT





GTCCCACTGA





101
DNA
>ACSP50_
ATGACAAGTGCTGTTGCTTCGCCACTGCGGACCGACTTCGAGCGCTCG




1951
GTCGCCAGCTACTGGAACACCAACCGGGCCGACCCGGTCAACCTGCG





CCTCGGCGAGGTCGACGGGCTGTACCACCACCACTACGGCGTCGGCG





AGCCCGACCTCAGCGTGCTGGACGGCCCGGCCGACACCCGCGAGCA





GCGGATCATCGCCGAGCTGCACCGGCTGGAGAACGCCCAGGCCGAC





CTGCTGCTCGACCACCTCGGCCCGATCCGGCCGGGCGACGCGCTGCT





CGACGGCGGGTCCGGCCGCGGCGGCACCAGCATCATGGCCAACGCG





CGGTTCGGCTGCCGGGTCGACGGGGTGTCCATCTCGGAATACCAGGT





GGGTTTCGCCAACGAGCAGGCCGCTCAGCGCGGCGTCGCCGACAGG





GTGCGCTTCCACTTCCGCAACATGCTGGACTCCGGATTCGCGACCGG





GTCACGGCAGGCGATCTGGACGAACGAGACGACGATGTACGTCGACC





TGTTCGACCTGTACGCGGAGTTCGCCCGGATGCTCGGCTTCGGCGGC





CGCTACGTGTGCATCACCGGTTGCGCCAACGACGTGACCGGCCGGCG





CTCCAAGGCGGTCAACAGGATCAACGAGCACTACACCTGTGACATCCA





CCCGCGCAGCGACTACTTCAAGGCGCTCGCCGCCCACGATCTCGTGC





CGATCGCCGTCACCGACCTGACCGCGGCCACCATCCCGTACTGGGAG





CTGCGCGCCCGGTCCGAGGTGGCGACCGGGATCGAACAGGCTTTCCT





CACGGCGTACTCAGAAGGCAGTTTCCACTACCTTCTGATCGCCGCCGA





TCGGGTCTGA





102
DNA
>ACSP50_
ATGGCCCTGCCGATCGAGGACTACGCGATCATCGCCGACACCCAGAC




1952
CGCGGCCCTGGTCGGTCGCAACGGATCGATCGACTGGCTCTGCGTGC





CCCGCTTCGACTCCGGCGCGATCTTCGCGGCGCTGCTCGGCGAGGC





GGAGAACGGCCACTGGACCATCGCACCGTCCGGCGAGGTGGTCACCA





CCCGCCGCCGCTACCGGGACGACACGCTGGTGTTGGAGACGGAGTTC





GAGACGGCCGGCGGCGTCGCCCGGTTGATCGACTTCATGCCGCCGC





GCACCGACTCGCCGTCCGTCATCCGGATCGTCGAGGGCGTCCGCGG





GCAGGTGGACTTCGGCATGGAGCTGCGGCTGCGCTTCGACTATGGAC





ACGTCGTGCCATGGGTCTACCGCGAGGGTGGGGCGCTCGTCGCGGT





CGCCGGTCCGGACGCGGCCTGGTTGCGCACCGACGTGCCGACCCGG





GGCGAGAATCTGACCACCAAAGCCGATTTCCGGGTACGGGCGGGGGA





ACGCGCCGCCTTCACCCTGACCTGGCGCCCGTCGCATCTGCCCTCGC





CCGCCCCGCTGGACCCGGCCCACGAGCTCGGCGTGACCGAGGGTTA





CTGGCGCGGCTGGGTGTCCGCCTGCACGTACGAGGGGGAGTGGCGG





GACGCCGTCGTCCGATCGCTGCTCACTCTGAAAGCCCTCACCTACGCA





CCCACCGGCGGCATTGTCGCGGCCGCCACCACCAGCCTCCCGGAGAA





ACTCGGCGGCGTCCGCAACTGGGACTACCGCTTCTGCTGGCTCCGCG





ACGCCACCATCACCCTGCAGTCGCTGCTCTTCTCCGGTTTCCAGAGTG





AGGCGATCGCCTGGCGCAAATGGCTGCTGCGCGCGATCGCCGGCAAC





CCCGCCGAGCTGCAGATCATGTACGGCGTCGCCGGCGAACGCCGCCT





CGACGAGTATCTGGCCGACTGGCTCACCGGCTACGACGGCAACCCGG





TCCGGATCGGCAACGCCGCCGCCGAGCAGTTCCAGTTGGACGTGTAC





GGCGAGGTGATGGACGCCCTGCATCAGGGCCGCCGGGCCGGCCTCA





AAGCCGACGACCCGTCCTGGGGCCTGCAGGTCAAACTGATGGAGTTC





GTCGAGGAGCACTGGCAGGACCCGGACGAGGGCATCTGGGAGGTCC





GCGGCGGCCCCCGCCAGTTCACCCACTCCAAACTGATGGCCTGGGTC





GCCGCCGACCGCGCCGTCAAGGCCGTCGAGGAGTTCGGCCTGGACG





GCCCCGCCGACCGCTGGCGCCGCCTGCGCGACGAGATCCGTCAGGA





CATCCTGGACAAGGGTTACGACCCGGTCCGCAAGACCTTCACCCAGTA





CTACGGCTCCGATGAGCTCGACGCCGCGATGCTGATGGTCCCCCTGG





TCGGCTTCCTCCCCGGGGATGACGAACGCGTCGCCGGCACGGTCGCC





GCCATCGAGCAACACCTGCTGGTCGACGGTTTCGTCCAGCGGTACAC





CCAACATCCGGACGCCGACGTCGACGGCCTTCCCCCGGGCGAGGGC





GCGTTCCTGGCCTGCACGTTCTGGCTGGCCGACAACTACGCGCTGAT





GGGTCGCCACGACGAGGCCCGGGAGACGTTCGCCCGCCTGCTGGCC





CTGCGCAACGACGTGGGTCTGCTCGCCGAGGAGTACGACACCACCAC





CGGCCGCCTGGTCGGCAACTTCCCTCAGGCCTTCAGTCACGTCCCGC





TGATCGACACGGCCCGGACCTTGACCAGCGCGCTGGCGCCGACCGA





GGCCCGGGCCTCGGAGGGCCTCAGGTAG





103
DNA
>ACSP50_
atgcgtacggtgattcgtgggatcgtggtgttggcgctggtggccgggggtggcgccggcatggtggggc




1953
ccgccggagcggcgccggcggtgacgttcaagaactgcactgagctgaacaagaagtacaagcacg





gggtcggcaagcggggcgccgaggacagggtgagcgggtccaccaagccggtcaccaccttctccgt





gaacaacgatctctatgcggcgaacaagaggctggaccgtgacaaggacgggatcgcctgcgagaa





gcggtga





104
PRT
>ACSP50_
MATPTQSEIREEEHEEQRQSLSTAAARNLTTTTKTAPQMQEITSRWLLRKL




1949
PWVQVAGGAYRVNRRMTYRIGDGRLSFTNVGAQVRVVPAELRELSVLSE





FDDADVLAAMADKFVQQEYQPGQVIVEFGSVADHVYVIAHGKVNKVGVG





NYGDPVNLGVLADGEAFGEKSLTDEERIWDYTAKAMTAVTLLAMPRSAFT





ALLGQSDHLRTHVEQFRAKNRRPQNKHGEAEISVAAGHTGEPKLDGTYV





DYELTPREYELSVAQTVLRVHTRVADLYNEPMNQVEQQLRLTVEALRERQ





EYEMINNREFGLLHNADLRQRIHTRGGPPTPDDLDELLSMRRGTRMFVAH





PQAVAAFGRECTKRGIYPPMLEQDGGTFLSWRGVPILPCGKIPVTETHTTS





ILAMRTGESDQGVVGLHQTGIPDEYEPSLSVRFMGISEQAIMSYLVSAYYS





AAVLVPDALGILDHVELSH





105
PRT
>ACSP50_
MTSAVASPLRTDFERSVASYWNTNRADPVNLRLGEVDGLYHHHYGVGEP




1951
DLSVLDGPADTREQRIIAELHRLENAQADLLLDHLGPIRPGDALLDGGSGR





GGTSIMANARFGCRVDGVSISEYQVGFANEQAAQRGVADRVRFHFRNML





DSGFATGSRQAIWTNETTMYVDLFDLYAEFARMLGFGGRYVCITGCANDV





TGRRSKAVNRINEHYTCDIHPRSDYFKALAAHDLVPIAVTDLTAATIPYWEL





RARSEVATGIEQAFLTAYSEGSFHYLLIAADRV





106
PRT
>ACSP50_
MALPIEDYAIIADTQTAALVGRNGSIDWLCVPRFDSGAIFAALLGEAENGH




1952
WTIAPSGEVVTTRRRYRDDTLVLETEFETAGGVARLIDFMPPRTDSPSVIRI





VEGVRGQVDFGMELRLRFDYGHVVPWVYREGGALVAVAGPDAAWLRTD





VPTRGENLTTKADFRVRAGERAAFTLTWRPSHLPSPAPLDPAHELGVTEG





YWRGWVSACTYEGEWRDAVVRSLLTLKALTYAPTGGIVAAATTSLPEKLG





GVRNWDYRFCWLRDATITLQSLLFSGFQSEAIAWRKWLLRAIAGNPAELQI





MYGVAGERRLDEYLADWLTGYDGNPVRIGNAAAEQFQLDVYGEVMDALH





QGRRAGLKADDPSWGLQVKLMEFVEEHWQDPDEGIWEVRGGPRQFTHS





KLMAWVAADRAVKAVEEFGLDGPADRWRRLRDEIRQDILDKGYDPVRKT





FTQYYGSDELDAAMLMVPLVGFLPGDDERVAGTVAAIEQHLLVDGFVQRY





TQHPDADVDGLPPGEGAFLACTFWLADNYALMGRHDEARETFARLLALR





NDVGLLAEEYDTTTGRLVGNFPQAFSHVPLIDTARTLTSALAPTEARASEG





LR





107
PRT
>ACSP50_
MRTVIRGIVVLALVAGGGAGMVGPAGAAPAVTFKNCTELNKKYKHGVGKR




1953
GAEDRVSGSTKPVTTFSVNNDLYAANKRLDRDKDGIACEKR





108
DNA
>anti-
CACTCATTAGGCACCCCAGGCTTTACACTTTATGCTTCCGGCTCGTATG




sense
TTGTGTGG




1 (put.





Anti-





sense





promoters)






109
DNA
>anti-
ACGCGGTCGAACACGCGGTGGTACATGTCCAGCCACGCGCACTGGTA




sense
CTCTTCGGAC




2 (put.





Anti-





sense





promoters)






110
DNA
>pSET
aagcgcggggaagaatcgatcaaggactcttaccgctgccgtcgccggaaggtggtcgacccgtccg




T4gap
cacctgggcagcacgaaggcacgtgcaacgtcagcatggcggcactcgacaagttcgttgcggaacg





catcttcaacaagatcaggcacgccgaaggcgacgaagagacgttggcgcttctgtgggaagccgcc





cgacgcttcggcaagctcactgaggcgcctgagaagagcggcgaacgggcgaaccttgttgcggagc





gcgccgacgccctgaacgcccttgaagagctgtacgaagaccgcgcggcaggcgcgtacgacggac





ccgttggcaggaagcacttccggaagcaacaggcagcgctgacgctccggcagcaaggggcggaa





gagcggcttgccgaacttgaagccgccgaagccccgaagcttccccttgaccaatggttccccgaaga





cgccgacgctgacccgaccggccctaagtcgtggtgggggcgcgcgtcagtagacgacaagcgcgtg





ttcgtcgggctcttcgtagacaagatcgttgtcacgaagtcgactacgggcagggggcagggaacgccc





atcgagaagcgcgcttcgatcacgtgggcgaagccgccgaccgacgacgacgaagacgacgccca





ggacggcacggaagacgtagcggcgtagcgagacacccgggaagcctgatctacgtctgtcgagaa





gtttctgatcgaaaagttcgacagcgtctccgacctgatgcagctctcgcagggcgaagaatctcgtgcttt





cagcttcgatgtaggagggcgtggatatgtcctgcgggtaaatagctgcgccgatggttCTCTGTCG





TCGCTGACGTCTGTAGTCTAGCCTCATTATGATTGTACGCTATTCAGGG





ATTGACTGATACCGGAAGACATCTCAAATGAAGTGGTCAAGCTTTATGC





TTGTAAACCGTTTTGTGAAAAAATTTTTAAAATAAAAAAGGGGACCTCTA





GGGTCCCCAATTAATTAGTAATATAATCTATTAAAGGTCATTCAAAAGGT





CATCCAAGCTTGGCTGTTTTGGCGGATGAGAGAAGATTTTCAGCCTGA





TACAGATTAAATCAGAACGCAGAAGCGGTCTGATAAAACAGAATTTGCC





TGGCGGCAGTAGCGCGGTGGTCCCACCTGACCCCATGCCGAACTCAG





AAGTGAAACGCCGTAGCGCCGATGGTAGTGTGGCCCATGCGAGAGTA





CATATGGTACTGGCCGATGCTGGGAGAAGCGCGCTGCTGTACGGCGC





GCACCGGGTGCGGAGCCCCTCGGCGAGCGGTGTGAAACTTCTGTGAA





TGGCCTGTTCGGTTGCTTTTTTTATACGGCTGCCAGATAAGGCTTGCAG





CATCTGGGCGGCTACCGCTATGATCGGGGCGTTCCTGCAATTCTTAGT





GCGAGTATCTGAAAGGGGATACGCATGGTACCGAGACCTTATGTTGAT





CGGCACTTTGCATCGGCCGCGCTCCCGATTCCGGAAGTGCTTGACATT





GGGGAATTTATGCGGTGTGAAATACCGCACAGATGCGTAAGGAGAAAA





TACCGCATCAGGCGCCATTCGCCATTCAGGCTGCGCAACTGTTGGGAA





GGGCGATCGGTGCGGGCCTCTTCGCTATTACGCCAGCTGGCGAAAGG





GGGATGTGCTGCAAGGCGATTAAGTTGGGTAACGCCAGGGTTTTCCCA





GTCACGACGTTGTAAAACGACGGCCAGTGCCAAGCTTGGGCTGCAGG





TCGACTCTAGAGGATCCGCGGCCGCGCGCGATATCGAATTCGTAATCA





TGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCGCTCACAATTCCACA





CAACATACGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATG





AGTGAGCTAACTCACATTAATTGCGTTGCGCGGTCTCGGCGTTTCGTG





CCGCGTGATTTTCCGCCAAAAACTTTAACGAACGTTCGTTATAATGGTG





TCATGACCTTCACGACGAAGTACTAAAATTGGCCCGAATCATCAGCTAA





GCTTTATGCTTGTAAACCGTTTTGTGAAAAAATTTTTAAAATAAAAAAGG





GGACCTCTAGGGTCCCCAATTAATTAGTAATATAATCTATTAAAGGTCA





TTCAAAAGGTCATCCACCTCACTTCGGTGAATCGAAGCGCGGCATCAG





GGTTACTTTTTGGATACCTGAGACATTCGTCGCTTCCGGGTATGCGCT





CTATGTGACGGTCTTTTGGCGCACAAATGCTCAGCACCATTTAAATTAG





ACCGACTCCAGATCTGTAAGGTCCAACAAAACCCATCGTAGTCCTTAG





ACTTGGCACACTTACACCTGCAGTGGATGACCTTTTGAATGACCTTTAA





TAGATTATATTACTAATTAATTGGGGACCCTAGAGGTCCCCTTTTTTATT





TTAAAAATTTTTTCACAAAACGGTTTACAAGCATAAAGCTTGCCACGCA





GACGACAGCCCACGCTGACCGATCTACCTGAACGGCGACCATCTGTG





TGGTACTGGGGCGGAGAGATAACTACGGTGCCGCTTACCGGgctcactca





aaggcggtaatacggttatccacagaatcaggggataacgcaggaaagaacatgtgagcaaaaggc





cagcaaaaggccaggaaccgtaaaaaggccgcgttgctggcgtttttccataggctccgcccccctgac





gagcatcacaaaaatcgacgctcaagtcagaggtggcgaaacccgacaggactataaagataccag





gcgtttccccctggaagctccctcgtgcgctctcctgttccgaccctgccgcttaccggatacctgtccgcctt





tctcccttcgggaagcgtggcgctttctcatagctcacgctgtaggtatctcagttcggtgtaggtcgttcgct





ccaagctgggctgtgtgcacgaaccccccgttcagcccgaccgctgcgccttatccggtaactatcgtctt





gagtccaacccggtaagacacgacttatcgccactggcagcagccactggtaacaggattagcagag





cgaggtatgtaggcggtgctacagagttcttgaagtggtggcctaactacggctacactagaagaacagt





atttggtatctgcgctctgctgaagccagttaccttcggaaaaagagttggtagctcttgatccggcaaaca





aaccaccgctggtagcggtggtttttttgtttgcaagcagcagattacgcgcagaaaaaaaggatctcaa





gaagatcctttgatcttttctacggggtctgacgctcagtggaacgaaaactcacgttaagggattttggtca





tgagattatcaaaaaggatcttcacctagatccttttggttcatgtgcagctccatcagcaaaaggggatga





taagtttatcaccaccgactatttgcaacagtgccgttgatcgtgctatgatcgactgatgtcatcagcggtg





gagtgcaatgtcgtgcaatacgaatggcgaaaagccgagctcatcggtcagcttctcaaccttggggtta





cccccggcggtgtgctgctggtccacagctccttccgtagcgtccggcccctcgaagatgggccacttgg





actgatcgaggccctgcgtgctgcgctgggtccgggagggacgctcgtcatgccctcgtggtcaggtctg





gacgacgagccgttcgatcctgccacgtcgcccgttacaccggaccttggagttgtctctgacacattctg





gcgcctgccaaatgtaaagcgcagcgcccatccatttgcctttgcggcagcggggccacaggcagagc





agatcatctctgatccattgcccctgccacctcactcgcctgcaagcccggtcgcccgtgtccatgaactc





gatgggcaggtacttctcctcggcgtgggacacgatgccaacacgacgctgcatcttgccgagttgatgg





caaaggttccctatggggtgccgagacactgcaccattcttcaggatggcaagttggtacgcgtcgattat





ctcgagaatgaccactgctgtgagcgctttgccttggcggacaggtggctcaaggagaagagccttcag





aaggaaggtccagtcggtcatgcctttgctcggttgatccgctcccgcgacattgtggcgacagccctgg





gtcaactgggccgagatccgttgatcttcctgcatccgccagaggcgggatgcgaagaatgcgatgccg





ctcgccagtcgattggctgagctcatgagcggagaacgagatgacgttggaggggcaaggtcgcgctg





attgctggggcaacacgtggagcggatcggggattgtctttcttcagctcgctgatgatatgctgacgctca





atgccgtttggcctccgactaacgaaaatcccgcatttggacggctgatccgattggcacggcggacgg





cgaatggcggagcagacgctcgtccgggggcaatgagatatgaaaaagcctgaactcaccgcgacgt





atcgggccctggccagctagctagagtcgacctgcaggtccccggggatcggtcttgccttgctcgtcggt





gatgtacttcaccagctccgcgaagtcgctcttcttgatggagcgcatggggacgtgcttggcaatcacgc





gcaccccccggccgttttagcggctaaaaaagtcatggctctgccctcgggcggaccacgcccatcatg





accttgccaagctcgtcctgcttctcttcgatcttcgccagcagggcgaggatcgtggcatcaccgaaccg





cgccgtgcgcgggtcgtcggtgagccagagtttcagcaggccgcccaggcggcccaggtcgccattga





tgcgggccagctcgcggacgtgctcatagtccacgacgcccgtgattttgtagccctggccgacggcca





gcaggtaggccgacaggctcatgccggccgccgccgccttttcctcaatcgctcttcgttcgtctggaagg





cagtacaccttgataggtgggctgcccttcctggttggcttggtttcatcagccatccgcttgccctcatctg





ttacgccggcggtagccggccagcctcgcagagcaggattcccgttgagcaccgccaggtgcgaataag





ggacagtgaagaaggaacacccgctcgcgggtgggcctacttcacctatcctgcccggctgacgccgtt





ggatacaccaaggaaagtctacacgaaccctttggcaaaatcctgtatatcgtgcgaaaaaggatggat





ataccgaaaaaatcgctataatgaccccgaagcagggttatgcagcggaaaagatccgtcgacctgca





ggcatgcaagctctagcgattccagacgtcccgaaggcgtggcgcggcttccccgtgccggagcaatc





gccctgggtgggttacacgacgcccctctatggcccgtactgacggacacaccgaagccccggcggc





aaccctcagcggatgccccggggcttcacgttttcccaggtcagaagcggttttcgggagtagtgcccca





actggggtaacctttgagttctctcagttgggggcgtagggtcgccgacatgacacaaggggttgtgacc





ggggtggacacgtacgcgggtgcttacgaccgtcagtcgcgcgagcgcgagaattcgagcgcagcaa





gcccagcgacacagcgtagcgccaacgaagacaaggcggccgaccttcagcgcgaagtcgagcgc





gacgggggccggttcaggttcgtcgggcatttcagcgaagcgccgggcacgtcggcgttcgggacggc





ggagcgcccggagttcgaacgcatcctgaacgaatgccgcgccgggcggctcaacatgatcattgtct





atgacgtgtcgcgcttctcgcgcctgaaggtcatggacgcgattccgattgtctcggaattgctcgccctgg





gcgtgacgattgtttccactcaggaaggcgtcttccggcagggaaacgtcatggacctgattcacctgatt





atgcggctcgacgcgtcgcacaaagaatcttcgctgaagtcggcgaagattctcgacacgaagaacctt





cagcgcgaattgggcgggtacgtcggcgggaaggcgccttacggcttcgagcttgtttcggagacgaa





ggagatcacgcgcaacggccgaatggtcaatgtcgtcatcaacaagcttgcgcactcgaccactcccct





taccggacccttcgagttcgagcccgacgtaatccggtggtggtggcgtgagatcaagacgcacaaac





accttcccttcaagccgggcagtcaagccgccattcacccgggcagcatcacggggctttgtaagcgca





tggacgctgacgccgtgccgacccggggcgagacgattgggaagaagaccgcttcaagcgcctggg





acccggcaaccgttatgcgaatccttcgggacccgcgtattgcgggcttcgccgctgaggtgatctacaa





gaagaagccggacggcacgccgaccacgaagattgagggttaccgcattcagcgcgacccgatcac





gctccggccggtcgagcttgattgcggaccgatcatcgagcccgctgagtggtatgagcttcaggcgtgg





ttggacggcagggggcgcggcaaggggctttcccgggggcaagccattctgtccgccatggacaagct





gtactgcgagtgtggcgccgtcatgacttcg





111
DNA
>pSET
aagcgcggggaagaatcgatcaaggactcttaccgctgccgtcgccggaaggtggtcgacccgtccg




T4tip
cacctgggcagcacgaaggcacgtgcaacgtcagcatggcggcactcgacaagttcgttgcggaacg





catcttcaacaagatcaggcacgccgaaggcgacgaagagacgttggcgcttctgtgggaagccgcc





cgacgcttcggcaagctcactgaggcgcctgagaagagcggcgaacgggcgaaccttgttgcggagc





gcgccgacgccctgaacgcccttgaagagctgtacgaagaccgcgcggcaggcgcgtacgacggac





ccgttggcaggaagcacttccggaagcaacaggcagcgctgacgctccggcagcaaggggcggaa





gagcggcttgccgaacttgaagccgccgaagccccgaagcttccccttgaccaatggttccccgaaga





cgccgacgctgacccgaccggccctaagtcgtggtgggggcgcgcgtcagtagacgacaagcgcgtg





ttcgtcgggctcttcgtagacaagatcgttgtcacgaagtcgactacgggcagggggcagggaacgccc





atcgagaagcgcgcttcgatcacgtgggcgaagccgccgaccgacgacgacgaagacgacgccca





ggacggcacggaagacgtagcggcgtagcgagacacccgggaagcctgatctacgtctgtcgagaa





gtttctgatcgaaaagttcgacagcgtctccgacctgatgcagctctcgcagggcgaagaatctcgtgcttt





cagcttcgatgtaggagggcgtggatatgtcctgcgggtaaatagctgcgccgatggttCTCTGTCG





TCGCTGACGTCTGTAGTCTAGCCTCATTATGATTGTACGCTATTCAGGG





ATTGACTGATACCGGAAGACATCTCAAATGAAGTGGTCAAGCTTTATGC





TTGTAAACCGTTTTGTGAAAAAATTTTTAAAATAAAAAAGGGGACCTCTA





GGGTCCCCAATTAATTAGTAATATAATCTATTAAAGGTCATTCAAAAGGT





CATCCAAGCTTGGCTGTTTTGGCGGATGAGAGAAGATTTTCAGCCTGA





TACAGATTAAATCAGAACGCAGAAGCGGTCTGATAAAACAGAATTTGCC





TGGCGGCAGTAGCGCGGTGGTCCCACCTGACCCCATGCCGAACTCAG





AAGTGAAACGCCGTAGCGCCGATGGTAGTGTGGCCCATGCGAGAGTA





CAATCCCTAGAACGTCCGGGCTTGCACCTCACGTCACGTGAGGAGGC





AGCGTGGACGGCGTGGTACCAAGCTTATTGGCACTAGTCGAGCAACG





GAGGTATTCCGATGGTACCGAGACCTTATGTTGATCGGCACTTTGCAT





CGGCCGCGCTCCCGATTCCGGAAGTGCTTGACATTGGGGAATTTATGC





GGTGTGAAATACCGCACAGATGCGTAAGGAGAAAATACCGCATCAGGC





GCCATTCGCCATTCAGGCTGCGCAACTGTTGGGAAGGGCGATCGGTG





CGGGCCTCTTCGCTATTACGCCAGCTGGCGAAAGGGGGATGTGCTGC





AAGGCGATTAAGTTGGGTAACGCCAGGGTTTTCCCAGTCACGACGTTG





TAAAACGACGGCCAGTGCCAAGCTTGGGCTGCAGGTCGACTCTAGAG





GATCCGCGGCCGCGCGCGATATCGAATTCGTAATCATGTCATAGCTGT





TTCCTGTGTGAAATTGTTATCCGCTCACAATTCCACACAACATACGAGC





CGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATGAGTGAGCTAACT





CACATTAATTGCGTTGCGCGGTCTCGGCGTTTCGTGCCGCGTGATTTT





CCGCCAAAAACTTTAACGAACGTTCGTTATAATGGTGTCATGACCTTCA





CGACGAAGTACTAAAATTGGCCCGAATCATCAGCTAAGCTTTATGCTTG





TAAACCGTTTTGTGAAAAAATTTTTAAAATAAAAAAGGGGACCTCTAGG





GTCCCCAATTAATTAGTAATATAATCTATTAAAGGTCATTCAAAAGGTCA





TCCACCTCACTTCGGTGAATCGAAGCGCGGCATCAGGGTTACTTTTTG





GATACCTGAGACATTCGTCGCTTCCGGGTATGCGCTCTATGTGACGGT





CTTTTGGCGCACAAATGCTCAGCACCATTTAAATTAGACCGACTCCAGA





TCTGTAAGGTCCAACAAAACCCATCGTAGTCCTTAGACTTGGCACACTT





ACACCTGCAGTGGATGACCTTTTGAATGACCTTTAATAGATTATATTACT





AATTAATTGGGGACCCTAGAGGTCCCCTTTTTTATTTTAAAAATTTTTTC





ACAAAACGGTTTACAAGCATAAAGCTTGCCACGCAGACGACAGCCCAC





GCTGACCGATCTACCTGAACGGCGACCATCTGTGTGGTACTGGGGCG





GAGAGATAACTACGGTGCCGCTTACCGGgctcactcaaaggcggtaatacggttatc





cacagaatcaggggataacgcaggaaagaacatgtgagcaaaaggccagcaaaaggccaggaac





cgtaaaaaggccgcgttgctggcgtttttccataggctccgcccccctgacgagcatcacaaaaatcgac





gctcaagtcagaggtggcgaaacccgacaggactataaagataccaggcgtttccccctggaagctcc





ctcgtgcgctctcctgttccgaccctgccgcttaccggatacctgtccgcctttctcccttcgggaagcgtgg





cgctttctcatagctcacgctgtaggtatctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgcac





gaaccccccgttcagcccgaccgctgcgccttatccggtaactatcgtcttgagtccaacccggtaagac





acgacttatcgccactggcagcagccactggtaacaggattagcagagcgaggtatgtaggcggtgct





acagagttcttgaagtggtggcctaactacggctacactagaagaacagtatttggtatctgcgctctgctg





aagccagttaccttcggaaaaagagttggtagctcttgatccggcaaacaaaccaccgctggtagcggt





ggtttttttgtttgcaagcagcagattacgcgcagaaaaaaaggatctcaagaagatcctttgatcttttctac





ggggtctgacgctcagtggaacgaaaactcacgttaagggattttggtcatgagattatcaaaaaggatc





ttcacctagatccttttggttcatgtgcagctccatcagcaaaaggggatgataagtttatcaccaccgacta





tttgcaacagtgccgttgatcgtgctatgatcgactgatgtcatcagcggtggagtgcaatgtcgtgcaata





cgaatggcgaaaagccgagctcatcggtcagcttctcaaccttggggttacccccggcggtgtgctgctg





gtccacagctccttccgtagcgtccggcccctcgaagatgggccacttggactgatcgaggccctgcgtg





ctgcgctgggtccgggagggacgctcgtcatgccctcgtggtcaggtctggacgacgagccgttcgatcc





tgccacgtcgcccgttacaccggaccttggagttgtctctgacacattctggcgcctgccaaatgtaaagc





gcagcgcccatccatttgcctttgcggcagcggggccacaggcagagcagatcatctctgatccattgcc





cctgccacctcactcgcctgcaagcccggtcgcccgtgtccatgaactcgatgggcaggtacttctcctcg





gcgtgggacacgatgccaacacgacgctgcatcttgccgagttgatggcaaaggttccctatggggtgc





cgagacactgcaccattcttcaggatggcaagttggtacgcgtcgattatctcgagaatgaccactgctgt





gagcgctttgccttggcggacaggtggctcaaggagaagagccttcagaaggaaggtccagtcggtca





tgcctttgctcggttgatccgctcccgcgacattgtggcgacagccctgggtcaactgggccgagatccgtt





gatcttcctgcatccgccagaggcgggatgcgaagaatgcgatgccgctcgccagtcgattggctgagc





tcatgagcggagaacgagatgacgttggaggggcaaggtcgcgctgattgctggggcaacacgtgga





gcggatcggggattgtctttcttcagctcgctgatgatatgctgacgctcaatgccgtttggcctccgactaa





cgaaaatcccgcatttggacggctgatccgattggcacggcggacggcgaatggcggagcagacgct





cgtccgggggcaatgagatatgaaaaagcctgaactcaccgcgacgtatcgggccctggccagctag





ctagagtcgacctgcaggtccccggggatcggtcttgccttgctcgtcggtgatgtacttcaccagctccgc





gaagtcgctcttcttgatggagcgcatggggacgtgcttggcaatcacgcgcaccccccggccgttttagc





ggctaaaaaagtcatggctctgccctcgggcggaccacgcccatcatgaccttgccaagctcgtcctgct





tctcttcgatcttcgccagcagggcgaggatcgtggcatcaccgaaccgcgccgtgcgcgggtcgtcggt





gagccagagtttcagcaggccgcccaggcggcccaggtcgccattgatgcgggccagctcgcggacg





tgctcatagtccacgacgcccgtgattttgtagccctggccgacggccagcaggtaggccgacaggctc





atgccggccgccgccgccttttcctcaatcgctcttcgttcgtctggaaggcagtacaccttgataggtggg





ctgcccttcctggttggcttggtttcatcagccatccgcttgccctcatctgttacgccggcggtagccggcca





gcctcgcagagcaggattcccgttgagcaccgccaggtgcgaataagggacagtgaagaaggaaca





cccgctcgcgggtgggcctacttcacctatcctgcccggctgacgccgttggatacaccaaggaaagtct





acacgaaccctttggcaaaatcctgtatatcgtgcgaaaaaggatggatataccgaaaaaatcgctata





atgaccccgaagcagggttatgcagcggaaaagatccgtcgacctgcaggcatgcaagctctagcgat





tccagacgtcccgaaggcgtggcgcggcttccccgtgccggagcaatcgccctgggtgggttacacga





cgcccctctatggcccgtactgacggacacaccgaagccccggcggcaaccctcagcggatgccccg





gggcttcacgttttcccaggtcagaagcggttttcgggagtagtgccccaactggggtaacctttgagttctc





tcagttgggggcgtagggtcgccgacatgacacaaggggttgtgaccggggtggacacgtacgcgggt





gcttacgaccgtcagtcgcgcgagcgcgagaattcgagcgcagcaagcccagcgacacagcgtagc





gccaacgaagacaaggcggccgaccttcagcgcgaagtcgagcgcgacgggggccggttcaggttc





gtcgggcatttcagcgaagcgccgggcacgtcggcgttcgggacggcggagcgcccggagttcgaac





gcatcctgaacgaatgccgcgccgggcggctcaacatgatcattgtctatgacgtgtcgcgcttctcgcgc





ctgaaggtcatggacgcgattccgattgtctcggaattgctcgccctgggcgtgacgattgtttccactcag





gaaggcgtcttccggcagggaaacgtcatggacctgattcacctgattatgcggctcgacgcgtcgcac





aaagaatcttcgctgaagtcggcgaagattctcgacacgaagaaccttcagcgcgaattgggcgggta





cgtcggcgggaaggcgccttacggcttcgagcttgtttcggagacgaaggagatcacgcgcaacggcc





gaatggtcaatgtcgtcatcaacaagcttgcgcactcgaccactccccttaccggacccttcgagttcgag





cccgacgtaatccggtggtggtggcgtgagatcaagacgcacaaacaccttcccttcaagccgggcag





tcaagccgccattcacccgggcagcatcacggggctttgtaagcgcatggacgctgacgccgtgccga





cccggggcgagacgattgggaagaagaccgcttcaagcgcctgggacccggcaaccgttatgcgaat





ccttcgggacccgcgtattgcgggcttcgccgctgaggtgatctacaagaagaagccggacggcacgc





cgaccacgaagattgagggttaccgcattcagcgcgacccgatcacgctccggccggtcgagcttgatt





gcggaccgatcatcgagcccgctgagtggtatgagcttcaggcgtggttggacggcagggggcgcggc





aaggggctttcccgggggcaagccattctgtccgccatggacaagctgtactgcgagtgtggcgccgtc





atgacttcg









DETAILED DESCRIPTION
Definitions

Unless otherwise defined, all scientific and technical terms used in the description, Figures and claims have their ordinary meaning as commonly understood by one of ordinary skill in the art. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will prevail. If two or more documents incorporated by reference include conflicting and/or inconsistent disclosure with respect to each other, then the document having the later effective date shall control. The materials, methods, and examples are illustrative only and not intended to be limiting. Unless stated otherwise, the following terms used in this document, including the description and claims, have the definitions given below.


The terms “comprising”, “including”, “containing”, “having” etc. shall be read expansively or open-ended and without limitation. Singular forms such as “a”, “an” or “the” include plural references unless the context clearly indicates otherwise. Unless otherwise indicated, the term “at least” preceding a series of elements is to be understood to refer to every element in the series. The terms “at least one” and “at least one of” include for example, one, two, three, four, five, six, seven, eight, nine, ten or more elements.


It is furthermore understood that slight variations above and below a stated range can be used to achieve substantially the same results as a value within the range. Also, unless indicated otherwise, the disclosure of ranges is intended as a continuous range including every value between the minimum and maximum values.


Where protein or amino acid sequences are provided throughout the application it is also understood by the skilled person that single or multiple amino acids may be exchanged by amino acids with similar properties to achieve substantially the same effect, i.e. an equivalent result.


The skilled person furthermore knows that a defined protein or amino acid sequence may be encoded by various nucleic acid sequences. For a given amino acid sequence as defined herein, each of the countable nucleic acid sequences encoding the specific amino acid sequence shall be deemed to be disclosed herein. Where nucleic acid sequences are provided throughout the application it is furthermore understood that silent mutations may be introduced.


O-{4,6-dideoxy-4[1S-(1,4,6/5)-4,5,6-trihydroxy-3-hydroxymethyl-2-cyclohexen-1-yl]-amino-α-D-gluco-pyranosyl}-(1→4)-O-α-D-glucopyranosyl-(1→4)-D-glucopyranose or “acarbose” is a cyclitol-containing aminoglycoside, composed of a pseudodisaccharide and an α-1,4-glycosidic bound maltose (Wehmeier and Piepersberg 2009). The pseudodisaccharide, named acarviose, is built by an unsaturated C7-aminocyclitol, also referred as valienol or valienamine, which is connected to C4 of a 4,6-didesoxy-D-glucose by a nitrogen bond (cf. FIG. 5) (Wehmeier and Piepersberg 2009). This N-glycosidic bond cannot be hydrolyzed by foreign alpha-1,4-glucoside hydrolases, leading to an almost irreversible inhibitory effect (Wehmeier and Piepersberg 2009; Brayer et al. 2000).


“Overexpression” of a gene product or protein as described herein refers to an increase in expression compared to the wild type or a specified reference strain. Preferably, the reference strain or control is the strain which has not been engineered for the specific overexpression of the respective gene(s) or protein(s). For example, the control does not comprise a vector comprising an expression cassette for the respective gene product or protein. For example, the overexpression of the gene product may be an increase during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


Preferably, overexpression is an increase of the gene product or protein by a factor of at least 1.5 or at least a factor of 2 compared to the control. With regard to transcript amounts and if not defined otherwise herein, strong overexpression refers to a log 2(fold change) >6. With regard to transcript amounts and if not defined otherwise herein, weak overexpression refers to a log 2(fold change)<2. With regard to transcript amounts and if not defined otherwise herein, medium strong overexpression refers to a log 2(fold change) ≥2 and ≤6.


The expression of a gene product or protein as described herein is “absent or reduced” if the respective gene has been deleted or mutated in such a way that its gene product is not expressed at all or in a significantly decreased amount (e.g. less than 0.75 fold or less than 0.5 fold). The expression of a gene product or protein as described herein is also considered absent or reduced if the gene product or protein has lost functionality, e.g. in a transient or permanent way, e.g. by mutation or knockdown. Methods to monitor the amount and or activity of a gene product or protein are known in the art and are also described herein in an exemplary way. In general, suitable methods to obtain an absent or reduced expression of a gene product are methods that alter the genetic sequence or elements of gene expression (e.g. by deletion or point mutations) and/or methods that negatively affect the transcription and translation of a gene or the activity or half-life of the gene product (protein).


If not specified otherwise the symbol “Δ” refers to a “deletion mutant”, i.e. a mutant wherein a specific gene sequence bas been at least partially deleted.


The “early growth phase” is the time, in which the Actinoplanes strain adapts to the medium and in which the cell dry weight is below 3 g·L1. After adaption to the environment, the culture metabolizes the nutrients supplied by the medium and starts to grow. Since Actinoplanes is growing in a spherical mycelium, which can only expand to the outside of the sphere, the cells in the middle are shielded from nutrients and have only limited space for cell division. Therefore, only the cell in the outer layer of the spherical mycelium are dividing. By this, growth of Actinoplanes is linear and not exponential—in contrast to other bacteria, which are growing unicellular. The growth phase is called “linear growth phase” for Actinoplanes ssp. and starts at a cell dry weight of 3 g·L−1. The “stationary phase” is defined as growth phase, in which the cells reach the capacity limits (of space and nutrients) respectively in which growth decreases due to the formation of inhibitory by-products or other chemical and physical factors such as changes in the osmolarity or pH. The stationary phase is the growth phase, in which the number of dying cells equals the number of dividing cells. This phase usually starts at a cell dry weight of 16-18 g·L−1 in maltose minimal medium.


The term “vector”, as used herein, refers to a nucleic acid molecule capable of propagating a nucleic acid molecule to which it is linked.


The term “expression cassette”, as used herein, refers to a nucleic acid molecule comprising at least a gene for expression and a regulatory sequence, such as a promoter.


A “promoter” is a nucleic acid sequence which leads to initiation of transcription of a particular gene.


A “strong promoter” as defined herein is a promoter, which leads to a normalized glucuronidase activity of at least 5·10−4 [L·g−1·min−1] in the glucuronidase assay, and/or which leads to a 350-fold relative transcription (in log 2(fold change)) of the gusA gene compared to the promoterless pGUS control vector. A detailed description of a method for characterizing the strength of a promoter is provided within the examples and in (Schaffert, et al. 2019).


Examples include the promoters of

    • apm: 9.2·10−4 [L·g·1·min−1] and log 2(fold change)=360.78
    • ermE*: 9.7·10−4 [L·g−1·min−1] and log 2(fold change)=291.03
    • katE: 5.1·10−4 [L·g−1·min−1] and log 2(fold change)=342.51
    • moeE5: 9.7·10−4[L·g−1·min−1] and log 2(fold change)=329.32
    • gapDH: 11.5·10−4 [L·g−1·min−1] and log 2(fold change)=931.45, and
    • actP: 22.9·10−4 [L·g−1·min−1].


A “medium strong promoter” is defined as promoter, which leads to a normalized glucuronidase activity of at least 1·10−4 [L·g−1·min−1] was achieved in the glucuronidase assay, and/or which leads to a 10-fold relative transcription (in log 2(fold change)) of the gusA gene compared to the promoterless pGUS control vector. Examples include the promoters of

    • efp: 3.1·10-4 [L·g-1·min-1] and log 2(fold change)=53.08
    • cdaR: 3.1·10-4 [L·g-1·min-1] and log 2(fold change)=86.82
    • rpsL: 3.5·10-4 [L·g-1·-min-1] and log 2(fold change)=98.53
    • rpsJ: 3.7·10-4 [L·g-1·min-1] and log 2(fold change)=123.97
    • cgt: 2.5·10-4 [L·g-1·min-1] and log 2(fold change)=347.29, and
    • tipA: 4.2·10-4 [L·g-1·min-1] and log 2(fold change)=191.


In some cases, the medium strong promoter leads to a normalized glucuronidase activity of at least 1·10−4 [L·g−1·min−1] and maximal 5·10−4 [L·g−1·min−1] in the glucuronidase assay.


A “weak promotor” is defined as promoter, which leads to a normalized glucuronidase activity of below 1·10−4 [L·g−1·min−1], and/or which leads to a relative transcription of below 10-fold compared to the promoterless pGUS control vector.


The term “Cgt” (ACSP50_5024, previously: ACPL_5091) refers to extracellular small carbohydrate binding protein, previously described as cyclomaltodextrin glucanotransferase due the high similarity to the C-terminal domain of cyclodextrin glycosyltransferases, obtained from Actinoplanes sp., e.g. strain ATCC 31044/CBS 674.73/SE50/110. Cgt protein is encoded by the gene cgt. Sequence(s) are described herein (SEQ ID No. 20) or are accessible via UniProt Identifier G8S155 (G8S155_ACTS5). Different isoforms and variants may exist for the different strains and are all comprised by the term. Where a specific mutation can be exchanged without changing the described catalytic properties of the initial sequence, it is clear that the sequence having such a functionally silent mutation is equivalent with regard to the initial sequence. In addition, the protein may furthermore be subject to various modifications, e.g, synthetic or naturally occurring modifications.


The term “AcbB” (ACSP50_3608, previously ACPL_3681) refers to dTDP-D-glucose-4,6-dehydratase obtained from Actinoplanes sp., e.g. strain ATCC 31044/CBS 674.73/SE50/110, which is probably involved in the biosynthesis of the acarviose moiety of acarbose. AcbB protein is encoded by the gene acbB. Sequence(s) are described herein (SEQ ID No. 13) or are accessible via UniProt Identifier Q9ZAE8 (RMLB_ACTS5). Different isoforms and variants may exist for the different strains and are all comprised by the term. Where a specific mutation can be exchanged without changing the described catalytic properties of the initial sequence, it is clear, that the sequence having such a functionally silent mutation is equivalent with regard to the initial sequence. In addition, the protein may furthermore be subject to various modifications, e.g, synthetic or naturally occurring modifications.


The term “GtaB” also “GaIU” (ACSP50_7820, previously ACPL_7811) refers to UTP-glucose-1-phosphate uridylyltransferase obtained from Actinoplanes sp., e.g. strain ATCC 31044/CBS 674.73/SE50/110. GtaB seems to catalyze the conversion of glucose-1P and UDP-glucose into each other and might be involved in the precursor supply for acarbose. GtaB protein is encoded by the gene gtaB. Sequence(s) are described herein (SEQ ID No. 19) or are accessible via UniProt Identifier G8S608 (ACPL_7811). Different isoforms and variants may exist for the different strains and are all comprised by the term. Where a specific mutation can be exchanged without changing the described catalytic properties of the initial sequence, it is clear that the sequence having such a functionally silent mutation is equivalent with regard to the initial sequence. In addition, the protein may furthermore be subject to various modifications, e.g, synthetic or naturally occurring modifications.


As defined herein, a “gene which is essential for carotenoid synthesis” is defined as a gene which is positively required for the synthesis of a carotenoid. Actinoplanes are known to produce a variety of soluble pigments including yellow, orange and pink pigments of the class carotenoids. In Actinoplanes, the set of genes which are essential for carotenoid synthesis include genes from the MEP/DOXP pathway, genes of terpene cluster 1, genes of terpene cluster 2a, genes of terpene cluster 2b and genes of camphene-like monoterpene biosynthesis terpene cluster 3. Genes of the MEP/DOXP pathway comprise

    • i. 1-deoxy-D-xylulose-5-phosphate synthase gene dxs (ACSP50_7096, SEQ ID No. 23),
    • ii. 4-hydroxy-3-methylbut-2-en-1-yl diphosphate synthase gene ispG (ACSP50_7248, SEQ ID No. 24),
    • iii. 1-deoxy-D-xylulose-5-phosphate reductoisomerase gene dxr (ACSP50_7250, SEQ ID No. 25),
    • iv. 4-hydroxy-3-methylbut-2-enyl diphosphate reductase gene ispH (ACSP50_7707, SEQ ID No. 26),
    • v. 4-(cytidine 5′-diphospho)-2-C-methyl-D-erythritol kinase gene ispE (ACSP50_7802, SEQ ID No. 27),
    • vi. 2-C-methyl-D-erythritol 2;4-cyclodiphosphate synthase gene ispF, ACSP50_8046, SEQ ID No. 28), and/or
    • vii. 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase gene ispD (ACSP50_8047, SEQ ID No. 29).


Genes of terpene cluster 1 comprise

    • i. isopentenyl-diphosphate delta-isomerase gene idi (ACSP50_0146, SEQ ID No. 30),
    • ii. zeta-phytoene desaturase gene crtl (ACSP50_0147, SEQ ID No. 10),
    • iii. polyprenyl synthetase gene crtE/ldsA (ACSP50_0148, SEQ ID No. 31),
    • iv. phytoene synthase gene crtB (ACSP50_0149, SEQ ID No. 32),
    • v. deoxyribodipyrimidine photo-lyase gene (ACSP50_0150, SEQ ID No. 33), or
    • vi. pyridine nucleotide-disulfide oxidoreductase gene (ACSP50_0151, SEQ ID No. 34).


Genes of terpene cluster 2a comprise

    • i. transcriptional regulator gene (ACSP50_1631, SEQ ID No. 35),
    • ii. lycopene cyclase gene (ACSP50_1632, SEQ ID No. 36),
    • iii. lycopene cyclase gene (ACSP50_1633, SEQ ID No. 37),
    • iv. polyprenyl synthetase (farnesyl pyrophosphate synthetase 2 gene fps2/crtE (ACSP50_1634, SEQ ID No. 38), and
    • v. methylenetetrahydrofolate reductase (NADPH) gene (ACSP50_1635, SEQ ID No. 39).


Genes of terpene cluster 2b comprise

    • i. LysR-family transcriptional regulator gene (ACSP50_1650, SEQ ID No. 40),
    • ii. methyltransferase type 11 gene (ACSP50_1651, SEQ ID No. 41),
    • iii. CDP-alcoholphosphatidyltransferase pgsA (ACSP50_1652, SEQ ID No. 42),
    • iv. zeta-phytoene desaturase (crtl-family) gene crtD (ACSP50_1653, SEQ ID No. 43),
    • v. glycosyl transferase gene cruC (ACSP50_1654, SEQ ID No. 44),
    • vi. hypothetical protein (put. membrane prot,) gene cruF, (ACSP50_1655, SEQ ID No. 45),
    • vii. GCN5 family acetyltransferase gene (ACSP50_1656, SEQ ID No. 46),
    • viii. monooxygenase gene (ACSP50_1657, SEQ ID No. 47), and
    • ix. short-chain dehydrogenase gene (ACSP50_1658, SEQ ID No. 48).


Another gene which is essential for carotenoid synthesis is polyprenyl synthetase gene crtE (ACSP50_3873, SEQ ID No. 49).


Genes of camphene-like monoterpene biosynthesis terpene cluster 3 comprise

    • i. transcriptional regulator (Crp/Fnr family) gene eshA (ACSP50_1949, SEQ ID No. 104),
    • ii. camphene synthase gene (ACSP50_1950, SEQ ID No. 50),
    • iii. methyltransferase (SAM-dependent) type 11 gene (ACSP50_1951, SEQ ID No. 105),
    • iv. glycosyl-hydrolase gene (ACSP50_1952, SEQ ID No. 106), and
    • v. oxidoreductase/aldo/ketoreductase (ACSP50_1953, SEQ ID No. 107).


EMBODIMENTS

While Actinomycetales strain Actinoplanes sp. SE50/110 was used as a model strain for the current invention, it is clear for the skilled person, that the general mechanisms and findings can be applied for other acarbose producing strains such as those strains which are currently used for the commercial production of acarbose. According to some embodiments, the Actinomycetales strain is a Micromonosporaceae strain. According to some embodiments, the Actinomycetales strain is an Actinoplanes strain. According to some embodiments, the Actinomycetales strain is Actinoplanes SE50 (ATCC 31042, CBS 961.70) (Frommer et al. 1973), Actinoplanes sp. SE50/110 (ATCC 31044, CBS 674.73) or an Actinoplanes strain derived thereof. In some embodiments, the Actinomycetales strain is an Actinoplanes strain which is commercially used for acarbose production. In some embodiments, the Actinomycetales strain is an Actinoplanes strain which is commercially used for Acarbose production, such as SN223-29-47, C445-P47, SN12755-38, SC3687-18-43, SC7177-40-17 or SN19910-37-21 as disclosed e.g. in EP 2601209 B1 and CN103298828 B, or a strain derived thereof.


Improvement of acarbose production refers to an increase in yield of acarbose over a specific time (either in total or relative to cell growth) and/or improvement of the purity of the acarbose, e.g. the decrease of side-products and/or acarbose analogs such as component C. Cultivation of the Actinoplanes strain can occur as known in the art or as described herein. In some embodiments, cultivation of the Actinoplanes strain occurs in maltose minimal medium.


According to a first aspect of the current invention, there is provided a method to engineer an Actinomycetales strain, such as an Actinoplanes strain, for the improved production of acarbose. According to some first embodiments according to the first aspect, the method according to the first aspect comprises engineering the Actinomycetales strain for absent or reduced expression of extracellular small carbohydrate binding protein Cgt (SEQ ID No. 20).


Surprisingly, deletion of carbohydrate binding protein Cgt (SEQ ID No. 20) resulted in an improved production of acarbose. An increase of the final acarbose yield between 8.3 and 16.6% was achieved in three independent shake flask cultivations (cf. example “Δcgt displays improved acarbose formation on maltose minimal medium”, FIG. 18, FIG. 19, Table E10, Table E11).


Furthermore, in comparison with the wildtype, the gene deletion mutant Δcgt displayed no apparent growth phenotype in screening experiments testing for different carbon sources, or under carbon-limited conditions (cf. examples “Analysis of cgt expression during growth on different carbon sources”, “Δcgt on different carbon sources or under carbon-limited conditions”, FIG. 12, FIG. 13, FIG. 14), or pH and osmolyte stress (cf. example “Δcgt has no impact on osmolarity- or pH-tolerance”, FIG. 15, FIG. 16, FIG. 17). The inventors could furthermore show, that deletion of cgt had no negative impact on the expression of acarbose biosynthesis genes (cf. example “Δcgt has no impact on the expression of acarbose biosynthesis genes”, FIG. 20).


Without being bound by theory, Cgt was found to be highly expressed in Actinoplanes sp. SE50/110 according to comprehensive studies of the extracellular proteome (Wendler et al. 2013; Ortseifen 2016) and transcriptome (Schwientek et al. 2013). Its gene product is exported into the extracellular space making up for about 8% of the whole secreted proteome. The inventors have analyzed the distribution of CBM-20 single-domain proteins in the prokaryotic world by BlastP analysis. Interestingly, singular CBM-20 domain-proteins were found in only 17 other species (cf. example “Distribution of single-domain CBM-20 proteins in the eubacterial world”). Most of these are found in species of the order Actinomycetales, for example in all strains of the genus Actinoplanes. Without being bound by theory, by deletion or reduced expression of cgt, energy and resources, such as ATP and amino acids, are relieved.


These resources may then be redirected to the acarbose biosynthesis, which is a growth-associated product.


According to some embodiments according to the first aspect, the method comprises deletion or mutation of the gene encoding extracellular small carbohydrate binding protein Cgt (SEQ ID No. 20). The establishment of an intergeneric conjugation system (Gren et al. 2016) and the CRISPR/Cas9 technique (Wolf et al. 2016), allows genome editing in Actinoplanes sp. SE50/110. In some embodiments according to the first aspect engineering the Actinomycetales strain for absent or reduced expression may occur using CRISPR/Cas9 technique. In some embodiments, engineering the Actinomycetales strain for absent or reduced expression may occur as described by (Wolf et al. 2016). In some embodiments engineering the Actinomycetales strain for absent or reduced expression may occur as described herein, e.g. as described in the example “Deletion of the gene cgt by CRISPR/Cas9 technique” or “Deletion system based on homologous recombination and counterselection with the cytosine deaminase CodA”.


For example, the inventors have successfully established a novel deletion system by homologous recombination, which uses an integrase-free vector backbone and CodA for counter selection, like described by Zhao et al. (2017).


According to some second embodiments according to the first aspect, the method according to the first aspect comprises engineering the Actinomycetales strain for absent or reduced expression of at least one gene which is essential for carotenoid synthesis. In some embodiments, the carotenoid is the orange pigment of Actinoplanes or a derivative thereof. In some different or the same embodiments, the carotenoid is a C40-carotenoid.


Engineering the Actinomycetales strain for absent or reduced expression may occur as described previously for the current aspect. According to some embodiments according to the first aspect, the method comprises deletion or mutation of the gene which is essential for carotenoid synthesis.



Actinoplanes are known to produce a variety of soluble pigments including yellow, orange and pink pigments of the class carotenoids (Parenti and Coronelli 1979). The inventors observed, that strong pigmentation was associated with acarbose production losses. This was confirmed by comparing growth and acarbose yields of cultures exposed to and covered from light (cf. example “Light-dependent carotenoid-formation and oxidative stress reduce acarbose production in Actinoplanes sp. SE50/110”, FIG. 22). While carotenoid formation was induced, acarbose production and growth of Actinoplanes sp. SE50/110 was strongly reduced, when exposed to bulb light (FIG. 22). In total, a loss of 39% of the final acarbose concentration was monitored.


From these findings it is not only plausible that the produced pigments are not essential (e.g. in a technical setup for commercial acarbose production) but also that reducing or depleting the carotenoid synthesis in Actinoplanes can be used to improve the acarbose formation. To this end, the method according to the first aspect comprises reducing or depleting the expression of at least one gene which is essential for carotenoid synthesis.


The inventors could furthermore reconstruct the carotenogenesis in Actinoplanes sp. SE50/110 (cf. example “Analysis of the functional relevance of carotenoid formation”, FIG. 21). The set of genes which are essential for carotenoid synthesis in Actinoplanes include genes from the MEP/DOXP pathway, genes of terpene cluster 1, genes of terpene cluster 2a, genes of terpene cluster 2b, genes of camphene-like monoterpene biosynthesis terpene cluster 3.


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is a gene of the MEP/DOXP pathway, such as

    • i. 1-deoxy-D-xylulose-5-phosphate synthase gene dxs (ACSP50_7096, SEQ ID No. 23),
    • ii. 4-hydroxy-3-methylbut-2-en-1-yl diphosphate synthase gene ispG (ACSP50_7248, SEQ ID No. 24),
    • iii. 1-deoxy-D-xylulose-5-phosphate reductoisomerase gene dxr (ACSP50_7250, SEQ ID No. 25),
    • iv. 4-hydroxy-3-methylbut-2-enyl diphosphate reductase gene ispH (ACSP50_7707, SEQ ID No. 26),
    • v. 4-(cytidine 5′-diphospho)-2-C-methyl-D-erythritol kinase gene ispE (ACSP50_7802, SEQ ID No. 27),
    • vi. 2-C-methyl-D-erythritol 2;4-cyclodiphosphate synthase gene ispF, ACSP50_8046, SEQ ID No. 28), and/or
    • vii. 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase gene ispD (ACSP50_8047, SEQ ID No. 29).


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is a gene of terpene cluster 1, such as

    • i. isopentenyl-diphosphate delta-isomerase gene idi (ACSP50_0146, SEQ ID No. 30),
    • ii. zeta-phytoene desaturase gene crtl (ACSP50_0147, SEQ ID No. 10),
    • iii. polyprenyl synthetase gene crtE/ldsA (ACSP50_0148, SEQ ID No. 31),
    • iv. phytoene synthase gene crtB (ACSP50_0149, SEQ ID No. 32),
    • v. deoxyribodipyrimidine photo-lyase gene (ACSP50_0150, SEQ ID No. 33), or
    • vi. pyridine nucleotide-disulfide oxidoreductase gene (ACSP50_0151, SEQ ID No. 34).


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is zeta-phytoene desaturase gene crtl (ACSP50_0147, SEQ ID No. 10). As discussed before, carotenoid formation is dispensable under laboratory conditions. In order to improve acarbose production, switching off the concurring carotenoid biosynthesis pathway, in particular by deletion of the central gene crtl, can be used for strain development.


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is a gene of terpene cluster 2a, such as

    • i. transcriptional regulator gene (ACSP50_1631, SEQ ID No. 35),
    • ii. lycopene cyclase gene (ACSP50_1632, SEQ ID No. 36),
    • iii. lycopene cyclase gene (ACSP50_1633, SEQ ID No. 37),
    • iv. polyprenyl synthetase (farnesyl pyrophosphate synthetase 2 gene fps2/crtE (ACSP50_1634, SEQ ID No. 38), or
    • v. methylenetetrahydrofolate reductase (NADPH) gene (ACSP50_1635, SEQ ID No. 39),


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is a gene of terpene cluster 2b, such as

    • i. LysR-family transcriptional regulator gene (ACSP50_1650, SEQ ID No. 40),
    • ii. methyltransferase type 11 gene (ACSP50_1651, SEQ ID No. 41),
    • iii. CDP-alcoholphosphatidyltransferase pgsA (ACSP50_1652, SEQ ID No. 42),
    • iv. zeta-phytoene desaturase (crtl-family) gene crtD (ACSP50_1653, SEQ ID No. 43),
    • v. glycosyl transferase gene cruC (ACSP50_1654, SEQ ID No. 44),
    • vi. hypothetical protein (put. membrane prot,) gene cruF, (ACSP50_1655, SEQ ID No. 45),
    • vii. GCN5 family acetyltransferase gene (ACSP50_1656, SEQ ID No. 46),
    • viii. monooxygenase gene (ACSP50_1657, SEQ ID No. 47), or
    • ix. short-chain dehydrogenase gene (ACSP50_1658, SEQ ID No. 48),


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is polyprenyl synthetase gene crtE (ACSP50_3873, SEQ ID No. 49).


According to some embodiments according to the current aspect and embodiments, the at least one gene essential for carotenoid synthesis is a gene of camphene-like monoterpene biosynthesis terpene cluster 3, such as

    • i. transcriptional regulator (Crp/Fnr family) gene eshA (ACSP50_1949, SEQ ID No. 104),
    • ii. camphene synthase gene (ACSP50_1950, SEQ ID No. 50),
    • iii. methyltransferase (SAM-dependent) type 11 gene (ACSP50_1951, SEQ ID No. 105),
    • iv. glycosyl-hydrolase gene (ACSP50_1952, SEQ ID No. 106), or
    • v. oxidoreductase/aldo/ketoreductase (ACSP50_1953, SEQ ID No. 107).


Since carotenoids influence the fluidity of membranes, lack of carotenoids and in particular of the C40-carotenoid can also affect the surface and mycelial structure of Actinoplanes sp. SE50/110. With regard to production break-up of mycelial lumps is advantageous to increase the mycelial surface and the number of biochemically available cells.


According to some further embodiments, the method according to the first aspect comprises engineering the Actinomycetales strain for overexpression of MerR-/HTH-transcriptional regulator gene merR (ACSP50_0145, SEQ ID No. 11). Engineering the Actinomycetales strain for overexpression may occur as described elsewhere herein.


Beside the mentioned genes which are essential for carotenoid synthesis, the inventors surprisingly identified a transcriptional repressor for the carotenoid synthesis among the genes of terpene cluster 1: ACSP50_0145 (SEQ ID No. 11, MerR-/HTH-transcriptional regulator gene merR) cf. example “Deletion of merR in SE50/110 induces carotenoid formation without exposure to light”, FIG. 24. By CRISPR/Cas9 deletion of the corresponding gene in SE50/110, the carotenoid formation was strongly induced without exposure to light (FIGS. 24 B and C).


Consistent with this, the acarbose production was found to be decreased. When illuminated, both wild type and AmerR are strongly pigmented and the final acarbose concentrations were similar for both strains, reaching approx. 0.52 g·L−1 (FIGS. 24 B and D). This corresponds to a reduction of acarbose formation of approx. 38% compared to the wild type under dark conditions (reaching 0.83 g·L−1). This is in accordance to the previous growth experiments of the wild type.


Under dark conditions, AmerR produces approx. 15% less acarbose than the wild type (0.70 g·L−1) (FIG. 24 D). Without being bound by theory, these production losses are assumed to be caused by the waste of resources by carotenoid formation in the deletion mutant (FIG. 24 C). In conclusion, the production losses under light conditions (38-39%) might be assigned to further light-induced stress in both the deletion mutant and the wild type.


According to some third embodiments according to the first aspect the method comprises engineering the Actinomycetales strain for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13).


According to the current invention it was surprisingly found that overexpression of the acb gene encoding the dTDP-D-glucose-4,6-dehydratase AcbB increased the final acarbose concentration significantly by approx. 50%. This was particularly surprising, because other genes of the Acb cluster such as AcbC did not lead to an improved formation of acarbose.


Furthermore, the observed increase was superior compared to the observed increase for overexpression of the complete Acb cluster as described by Zhao et al. (Zhao, Xie, et al. 2017).


According to some embodiments, the strain does not comprise engineering the Actinomycetales strain for overexpression of other genes of the Acb cluster, except for AcbA.


The dTDP-D-glucose-4,6-dehydratase AcbB seems to be involved in the generation of an activated amino sugar from D-glucose-1P which is a feeding pathway of the acarbose biosynthesis (FIG. 1): Without being bound by theory, increased AcbB activity was surprisingly found to also improve the supply of the modified precursor.


Overexpression of AcbB as described herein refers to an increase in expression for AcbB compared to the wild type or a specified reference strain/control. For example, the overexpression of the gene product may be an increase during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


Preferably, as described herein, overexpression of AcbB refers to an increase of AcbB transcript and/or protein by a factor of at least 1.5 or at least a factor of 2 compared to the control. With regard to AcbB transcript amounts, and if not defined otherwise herein, strong overexpression refers to a log 2(fold change) >6. With regard to AcbB transcript amounts and if not defined otherwise herein, medium strong overexpression refers to a log 2(fold change) ≥2 and ≤6.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a factor of a log 2(fold change) of at least 1.5 or at least 2, during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a log 2(fold change) ≥2 and ≤6 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time, such as during the early growth phase and/or during the linear growth phase.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a log 2(fold change) >3 and <5 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a log 2(fold change) >6 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


In overexpression mutants with expression vectors comprising heterologous promoters the relative transcription of acbB decelerated from 4.06- to 3.33-fold (log 2(fold change)) between the two sampling times in pSETT4tip::acbB (medium strong promoter) and from 6.54- to 2.05-fold in in pSETT4gap::acbB (strong promoter) (cf. example “Medium overexpression of acbB leads to improved acarbose formation”).


According to some embodiments, engineering the Actinomycetales strain for overexpression of a gene according to the first aspect may occur by any method known in the art or described herein.


As described within the example “Medium overexpression of acbB leads to improved acarbose formation”, two pSETT4-based overexpression mutants were created, in which acbB is transcribed under control of the medium strong tipA-promoter or the strong gapDH-promoter.


The native promoter was used in both the pSET152- and the pSETT4-vector background as control. In particular the mutant with acbB transcribed under control of the heterologous tipA-promoter displayed enhanced acarbose production compared to the control strains (FIG. 27, FIG. 28). The yield coefficient was increased to 48.6 and 51.9% compared to the empty vector control. By usage of the strong gapDH-promoter, the acarbose yield coefficient was found to be slightly increased (FIG. 28).


According to some embodiments, engineering the Actinomycetales strain for overexpression of a gene according to the first aspect may occur by introducing a vector comprising an expression cassette for AcbB (SEQ ID No. 13) into the Actinomycetales strain. In some embodiments, the expression vector is derived from pSET152. In some embodiments, the expression vector is derived from pSETT4. A vector is derived from another vector, if it comprises at least one, two, three, four elements of the second vector.


According to some embodiments, engineering the Actinomycetales strain for overexpression of a gene according to the first aspect may occur by introducing a vector comprising an expression cassette for AcbB (SEQ ID No. 13) into the Actinomycetales strain. In some of these or other embodiments the expression cassette is under the control of a medium strong promoter, as characterized by a normalized glucuronidase activity of at least 1x·10−4, preferably between 1x·10−4 and 5×10−4 [L·g−1·min−1] in a glucuronidase assay, e.g. as described elsewhere herein. In some embodiments said promoter is selected from efp promoter (SEQ ID No. 92), cdaR promoter (SEQ ID No. 97), rpsL promoter (SEQ ID No. 99), rpsJ promoter (SEQ ID No. 93), cgt promoter (SEQ ID No. 91), or tipA promoter (SEQ ID No. 81). In some embodiments the promoter is the tipA promoter (SEQ ID No. 81). Excellent results for acarbose production were obtained with pSETT4tip::acbB, cf. FIG. 27, FIG. 28.


In some embodiments the expression cassette is under the control of a strong promoter, as characterized by a normalized glucuronidase activity of at least 5×10−5 [L·g−1·min−1] in a glucuronidase assay, e.g. as described elsewhere herein. In some embodiments said promoter is selected from apm promoter (SEQ ID No. 96), ermE* promoter (SEQ ID No. 98), katE promoter (SEQ ID No. 94), moeE5 promoter (SEQ ID No. 95) or gapDH promoter (SEQ ID No. 82).


According to some embodiments, the method according to the first aspect comprises engineering the Actinomycetales strain for medium overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) and optionally AcbA (SEQ ID No. 12). In some embodiments, which are also compatible with all other embodiments described herein if not explicitly stated otherwise, genetic engineering does not result in an increase of transcript and/or protein by a log 2(fold change) ≥2 for Acb genes other than AcbB and AcbA. In some of embodiments which are also compatible with all other embodiments described herein, genetic engineering does not result in an increase of transcript and/or protein by a log 2(fold change) ≥2 for AcbC.


Upon overexpression of AcbB, further genes of the acb gene cluster were not significantly affected, e.g. in the early growth phase, like shown for acbA and acbV (FIG. 30). Only exception is a slightly higher transcription abundance of acbA in pSETT4tip::acbB (log 2(fold change)=1.87).


According to some embodiments, the method according to the first aspect comprises engineering the Actinomycetales strain for overexpression of AcbB (SEQ ID No. 13) and AcbS (ACSP50_3596) and/or Acbl (ACSP50_3599).


By (additional) overexpression of AcbS and/or Acbl, the transfer reaction of the amino sugar to the cyclitol precursor can be strengthened. According to the current model (see FIG. 1), this reaction is catalyzed by AcbS (ACSP50_3596) or Acbl (ACSP50_3599).


According to some embodiments, the method according to the first aspect comprises engineering the Actinomycetales strain for overexpression of AcbB (SEQ ID No. 13) and AcbCUJ (AcbC (ACSP50_3607) and/or AcbU (ACSP50_3595) and/or AcbJ (ACSP50_3600)) and/or AcbSI (AcbS (ACSP50_3596) and/or Acbl (ACSP50_3599)). Without being bound by theory, this combination can plausibly reinforce both acarbose synthesis strands.


According to some fourth embodiments according to the first aspect, the method comprises engineering the Actinomycetales strain for overexpression of UDP-glucose-1P uridyltransferase GtaB (SEQ ID No. 19).


By medium overexpression of gtaB, an increase of 8.5% of the final acarbose concentration was observed, cf. example “Medium overexpression of gtaB leads to improved acarbose formation”, FIG. 32, FIG. 33. Interestingly, the acarbose formation is particularly increased in the late linear to stationary growth phase (FIG. 32). Without being bound by theory, this may result from the improved deployment of the precursor glucose-1P (cf. FIG. 34).


Overexpression of GtaB (SEQ ID No. 19) as described herein refers to an increase in expression for GtaB transcript and/or protein compared to the wild type or a specified reference strain/control. For example, the overexpression of the gene product may be an increase during the early growth phase and/or during the linear growth phase and/or during the stationary phase, and/or an increase during any other time.


Preferably, overexpression is an increase of GtaB transcript and/or protein by a factor of at least 1.5 or at least a factor of 2 compared to the control. With regard to GtaB transcript amounts and if not defined otherwise herein, strong overexpression refers to a log 2(fold change) >6. With regard to GtaB transcript amounts and if not defined otherwise herein, medium strong overexpression refers to a log 2(fold change) ≥2 and ≤6.


According to some embodiments the overexpression of UDP-glucose-1P uridyltransferase GtaB is the increase of the expression of GtaB by a factor of a log 2(fold change) of at least 1.5 or at least 2 during the early growth phase and/or during the linear growth phase and/or during the stationary phase, and/or an increase during any other time.


In one of the overexpression mutants described herein, the relative transcript amount of the gene gtaB is 2.64-fold increased (log 2(fold change)) (FIG. 33).


According to some embodiments the overexpression of UDP-glucose-1P uridyltransferase GtaB is the increase of the expression of GtaB transcript and/or protein by a log 2(fold change) ≥2 and ≤6 during the early growth phase and/or during the linear growth phase and/or during the stationary phase, and/or an increase during any other time. According to some embodiments the overexpression of UDP-glucose-1P uridyltransferase GtaB is the increase of the expression of GtaB by a log 2(fold change) ≥3 and ≤5 during the early growth phase and/or during the linear growth phase and/or during the stationary phase, and/or an increase during any other time.


According to some embodiments the overexpression of UDP-glucose-1P uridyltransferase GtaB is the increase of the expression of GtaB by a log 2(fold change) ≥6 during the early growth phase and/or during the linear growth phase and/or during the stationary phase.


According to some embodiments, engineering the Actinomycetales strain for overexpression of a gene according to the first aspect may occur by introducing a vector comprising an expression cassette for GtaB (SEQ ID No. 19) into the Actinomycetales strain. In some embodiments, the expression vector is derived from pSET152. In some embodiments, the expression vector is derived from pSETT4. A vector is derived from another vector, if it comprises at least one, two, three, four elements of the second vector.


According to some embodiments, engineering the Actinomycetales strain for overexpression of a gene according to the first aspect may occur by introducing a vector comprising an expression cassette for GtaB (SEQ ID No. 19) into the Actinomycetales strain.


In some of these or other embodiments the expression cassette is under the control of a medium strong promoter, as characterized by a normalized glucuronidase activity of between 1x·10−4 and 5×10−5 [L·g−1·min−1] in a glucuronidase assay, e.g. as described elsewhere herein. In some embodiments said promoter is selected from efp promoter (SEQ ID No. 92), cdaR promoter (SEQ ID No. 97), rpsL promoter (SEQ ID No. 99), rpsJ promoter (SEQ ID No. 93), cgt promoter (SEQ ID No. 91), or tipA promoter (SEQ ID No. 81). In some embodiments the promoter is the tipA promoter (SEQ ID No. 81). Good results for acarbose production were obtained for example with pSETT4tip::gtaB, cf. FIG. 32, FIG. 33.


In some embodiments the expression cassette is under the control of a strong promoter, as characterized by a normalized glucuronidase activity of at least 5×10−5 [L·g−1·min−1] in a glucuronidase assay, e.g. as described elsewhere herein. In some embodiments said promoter is selected from apm promoter (SEQ ID No. 96), ermE* promoter (SEQ ID No. 98), katE promoter (SEQ ID No. 94), moeE5 promoter (SEQ ID No. 95) or gapDH promoter (SEQ ID No. 82).


According to some further or the same embodiments of the first aspect, the method comprises engineering the Actinomycetales strain for medium overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) and GtaB (SEQ ID No. 19).


It was surprisingly found that overexpression of GtaB triggers improved acarbose formation. By medium overexpression of acbB (e.g. by use of the tipA-promoter), a positive effect on acarbose production was observed yielding approx. 50% more acarbose in two independent cultivations.


Therefore, the improvement of the acarbose biosynthesis by overexpression of singular acb gene AcbB was achieved. Furthermore, by medium overexpression of gtaB, an increase of 8.5% of the final acarbose concentration was observed. It is plausible, that by a combined overexpression of acbB and gtaB, the flux through the amino sugar biosynthesis is improved leading to a further enhancement of acarbose production.


Without being bound by theory, strong overexpression of AcbB induced only smaller increases of acarbose production compared to medium strong overexpression of AcbB. This may be due to an imbalance in glucose-phosphate-metabolism, occurring upon massive overexpression of AcbB. Overexpression of gtaB might cure this imbalance, and combined overexpression of both, acbB and gtaB therefore plausibly leads to a further increase in acarbose production.


Interestingly, a significant decreased amount of the mass m/z=545 [M-H+] was found in pSETT4tip::gtaB (approx. decrease of 48%), which might correspond to dTDP-4-keto-6-deoxy-D-glucose, the proposed product of AcbB. This may indicate, that the flow through the synthesis strand is more balanced, since the accumulation of this metabolite is reduced in comparison to the empty vector control and AcbB-overexpression mutants (FIG. 34).


According to some embodiments, the method according to the first aspect comprises engineering the Actinomycetales strain

    • (i) for absent or reduced expression of extracellular small carbohydrate binding protein Cgt (SEQ ID No. 20) and/or,
    • (ii) for absent or reduced expression of at least one gene involved in carotenoid synthesis, and/or,
    • (iii) for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13), and/or
    • (iv) for overexpression of UDP-glucose-1P uridyltransferase GtaB (SEQ ID No. 19).


According to some embodiments, the method according to the first aspect further comprises engineering the Actinomycetales strain for absent or reduced expression of treY.


According to some embodiments, the method according to the first aspect further comprises

    • (i) deletion or mutation of the gene encoding extracellular small carbohydrate binding protein Cgt (SEQ ID No. 20) and/or,
    • (ii) deletion or mutation of at least one gene involved in carotenoid synthesis and/or,
    • (iii) introducing a vector comprising an expression cassette for AcbB (SEQ ID No. 13) into the Actinomycetales strain and/or
    • (iv) introducing a vector comprising an expression cassette for GtaB (SEQ ID No. 19) into the Actinomycetales strain.


According to some embodiments, the expression cassette according to (iii) and/or (iv) is under the control of a medium strong promoter, as characterized by a normalized glucuronidase activity of between 1x·10−4 and 5×10−5 [L·g−1·min−1] in a glucuronidase assay.


According to a second aspect there is provided an Actinomycetales strain, such as an Actinoplanes strain, for the production of acarbose. According to some embodiments the Actinomycetales strain is a strain generated by a method according to the first aspect. According to some other embodiments the Actinomycetales strain is genetically engineered for absent or reduced expression of extracellular small carbohydrate binding protein Cgt (SEQ ID No. 20).


According to some embodiments the Actinomycetales strain is a Δcgt mutant. A Δcgt mutant is a variant of an Actinomycetales strain wherein the gene Cgt (SEQ ID No. 20) has been at least partially deleted or inverted.


According to some of these or other embodiments the Actinomycetales strain is genetically engineered for absent or reduced expression of at least one gene which is essential for carotenoid synthesis. According to some embodiments the at least one gene which is essential for carotenoid synthesis has been at least partially deleted or inverted. According to some of these embodiments the at least one gene which is essential for carotenoid synthesis comprises at least one gene selected from any of

    • a. the genes of the MEP/DOXP pathway, such as
      • i. 1-deoxy-D-xylulose-5-phosphate synthase gene dxs (ACSP50_7096, SEQ ID No. 23),
      • ii. 4-hydroxy-3-methylbut-2-en-1-yl diphosphate synthase gene ispG (ACSP50_7248, SEQ ID No. 24),
      • iii. 1-deoxy-D-xylulose-5-phosphate reductoisomerase gene dxr (ACSP50_7250, SEQ ID No. 25),
      • iv. 4-hydroxy-3-methylbut-2-enyl diphosphate reductase gene ispH (ACSP50_7707, SEQ ID No. 26),
      • v. 4-(cytidine 5-diphospho)-2-C-methyl-D-erythritol kinase gene ispE (ACSP50_7802, SEQ ID No. 27),
      • vi. 2-C-methyl-D-erythritol 2;4-cyclodiphosphate synthase gene ispF, ACSP50_8046, SEQ ID No. 28), and/or
      • vii. 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase gene ispD (ACSP50_8047, SEQ ID No. 29),
    • b. the genes of terpene cluster 1, such as
      • i. isopentenyl-diphosphate delta-isomerase gene idi (ACSP50_0146, SEQ ID No. 30),
      • ii. zeta-phytoene desaturase gene crtl (ACSP50_0147, SEQ ID No. 10),
      • iii. polyprenyl synthetase gene crtE/ldsA (ACSP50_0148, SEQ ID No. 31),
      • iv. phytoene synthase gene crtB (ACSP50_0149, SEQ ID No. 32),
      • v. deoxyribodipyrimidine photo-lyase gene (ACSP50_0150, SEQ ID No. 33), or
      • vi. pyridine nucleotide-disulfide oxidoreductase gene (ACSP50_0151, SEQ ID No. 34)
    • c. the genes of terpene cluster 2a, such as
      • i. transcriptional regulator gene (ACSP50_1631, SEQ ID No. 35),
      • ii. lycopene cyclase gene (ACSP50_1632, SEQ ID No. 36),
      • iii. lycopene cyclase gene (ACSP50_1633, SEQ ID No. 37),
      • iv. polyprenyl synthetase (farnesyl pyrophosphate synthetase 2 gene fps2/crtE (ACSP50_1634, SEQ ID No. 38), or
      • v. methylenetetrahydrofolate reductase (NADPH) gene (ACSP50_1635, SEQ ID No. 39),
    • d. the genes of terpene cluster 2b, such as
      • i. LysR-family transcriptional regulator gene (ACSP50_1650, SEQ ID No. 40),
      • ii. methyltransferase type 11 gene (ACSP50_1651, SEQ ID No. 41),
      • iii. CDP-alcoholphosphatidyltransferase pgsA (ACSP50_1652, SEQ ID No. 42),
      • iv. zeta-phytoene desaturase (crtl-family) gene crtD (ACSP50_1653, SEQ ID No. 43),
      • v. glycosyl transferase gene cruC (ACSP50_1654, SEQ ID No. 44),
      • vi. hypothetical protein (put. membrane prot,) gene cruF, (ACSP50_1655, SEQ ID No. 45),
      • vii. GCN5 family acetyltransferase gene (ACSP50_1656, SEQ ID No. 46),
      • viii. monooxygenase gene (ACSP50_1657, SEQ ID No. 47),
      • ix. short-chain dehydrogenase gene (ACSP50_1658, SEQ ID No. 48),
    • e. polyprenyl synthetase gene crtE (ACSP50_3873, SEQ ID No. 49), or
    • f. the genes for camphene-like monoterpene biosynthesis terpene cluster 3, such as
      • i. transcriptional regulator (Crp/Fnr family) gene eshA (ACSP50_1949, SEQ ID No. 104),
      • ii. camphene synthase gene (ACSP50_1950, SEQ ID No. 50),
      • iii. methyltransferase (SAM-dependent) type 11 gene (ACSP50_1951, SEQ ID No. 105),
      • iv. glycosyl-hydrolase gene (ACSP50_1952, SEQ ID No. 106),
      • v. oxidoreductase/aldo/ketoreductase (ACSP50_1953, SEQ ID No. 107).


According to some of these or other embodiments the Actinomycetales strain is genetically engineered for overexpression of MerR-/HTH-transcriptional regulator gene merR (ACSP50_0145, SEQ ID No. 11).


According to some of these or other embodiments the Actinomycetales strain is genetically engineered for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13).


As described elsewhere herein, overexpression of AcbB refers to an increase of AcbB by a factor of at least 1.5 or at least a factor of 2 compared to the control. Preferably, the control is the strain which has not been engineered for the specific overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13). For example, the control does not comprise a vector comprising an expression cassette for AcbB.


For example, the overexpression of the gene product may be an increase during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a factor of a log 2(fold change) of at least 1.5 or at least 2, during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a log 2(fold change) ≥2 and ≤6 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time, such as during the early growth phase and/or during the linear growth phase.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a log 2(fold change) >3 and <5 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) is the increase of the expression of AcbB transcript and/or protein by a log 2(fold change) >6 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the Actinomycetales strain genetically engineered for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) comprises a vector for overexpression of AcbB. According to some of these embodiments, the vector is a vector as described herein, preferably according to an aspect described herein.


According to some embodiments the Actinomycetales strain genetically engineered for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) comprises an expression cassette for AcbB (SEQ ID No. 13) under the control of a medium strong promoter.


According to some embodiments the Actinomycetales strain genetically engineered for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB (SEQ ID No. 13) comprises an expression cassette for AcbB (SEQ ID No. 13) under the control of strong promoter. Preferably, the promoter is not the native promoter of AcbB.


According to some of these or other embodiments the Actinomycetales strain is genetically engineered for overexpression of UDP-glucose-1P uridyltransferase GtaB (SEQ ID No. 19).


Overexpression of GtaB (SEQ ID No. 19) as described elsewhere herein refers to an increase in expression for GtaB compared to the wild type or a specified reference strain/control.


Preferably, the control is the strain which has not been engineered for the specific overexpression of GtaB (SEQ ID No. 19). For example, the control does not comprise a vector comprising an expression cassette for GtaB (SEQ ID No. 19). For example, the overexpression of the gene product may be an increase during the early growth phase and/or during the linear growth phase and/or during the stationary phase, and/or an increase during any other time.


According to some embodiments the overexpression of GtaB is the increase of the expression of GtaB transcript and/or protein by a factor of a log 2(fold change) of at least 1.5, or at least 2, during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of GtaB is the increase of the expression of GtaB transcript and/or protein by a log 2(fold change) ≥2 and ≤6 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time, such as during the early growth phase and/or during the linear growth phase.


According to some embodiments the overexpression of GtaB is the increase of the expression of GtaB transcript and/or protein by a log 2(fold change) >3 and <5 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the overexpression of GtaB is the increase of the expression of GtaB transcript and/or protein by a log 2(fold change) >6 during the early growth phase, during the linear growth phase, during the stationary phase or an increase during any other time.


According to some embodiments the Actinomycetales strain genetically engineered for overexpression of GtaB comprises a vector for overexpression of GtaB. According to some of these embodiments, the vector is a vector as described herein, preferably according to an aspect described herein.


According to some embodiments the Actinomycetales strain genetically engineered for overexpression of GtaB (SEQ ID No. 19) comprises an expression cassette for GtaB (SEQ ID No. 19) under the control of a medium strong promoter.


According to some embodiments the Actinomycetales strain genetically engineered for overexpression of GtaB (SEQ ID No. 19) comprises an expression cassette for GtaB (SEQ ID No. 19) under the control of strong promoter. Preferably, the promoter is not the native promoter of GtaB.


According to a third aspect there is provided an Actinomycetales strain, such as an Actinoplanes strain, for the production of acarbose for use in the production of acarbose.


According to some embodiments there is provided a method for the production of acarbose, wherein the method comprises the use of an Actinomycetales strain according to the second aspect.


For genetic engineering of Actinoplanes, an expression system is required for the overexpression of singular or multiple genes. According to a fourth aspect there is provided an expression vector for Actinoplanes.


According to some embodiments, the vector according to the fourth aspect comprises a medium strong promoter characterized by a normalized glucuronidase activity of at least 1x·10−4 [L·g-1·min-1] in a glucuronidase assay. In some embodiments, the medium strong promoter is selected from efp according to SEQ ID No. 92, cdaR according to SEQ ID No. 97, rpsL according to SEQ ID No. 99, rpsJ according to SEQ ID No. 93, cgt according to SEQ ID No. 91, or tipA according to SEQ ID No. 81.


According to some embodiments, the vector according to the fourth aspect comprises a strong promoter characterized by a normalized glucuronidase activity of at least 5x·10−4 [L·g-1-min-1] in a glucuronidase assay. In some embodiments, the strong promoter is selected from apm according to SEQ ID No. 96, ermE* according to SEQ ID No. 98, katE according to SEQ ID No. 94, moeE5 according to SEQ ID No. 95 or gapDH according to SEQ ID No. 82.


To find further suitable promoters, that allow medium to strong gene expression, a promoter screening can be carried out by use of the screening system of Horbal et al. (2013) and Myronovskyi et al. (2011), which is based on the reporter GusA cloned in a pSET152-vector system, cf. FIG. 3, Table 1.


In some embodiments the vector according to the first aspect comprises an expression cassette.


Preferably the vector comprises an expression cassette for AcbB (SEQ ID No. 13) and/or an expression cassette for GtaB (SEQ ID No. 19) and/or an expression cassette for MerR.


In some embodiments, the expression cassette may furthermore comprise a lacZα-gene under control of the lac-promoter. The lacZα-gene encodes a catalytic domain of a β-galactosidase, that enables quick selection of the integration of a target sequence by blue/white-selection in the cloning strain Escherichia coli DH5αMCR (NC_017638.1) (Grant et al. 1990).


Without being bound by theory, the vector according to the current aspect comprises elements for vector replication, transfer, maintenance and selection. In some embodiments, at least one of these elements is derived from pSET152.


In some embodiments, the vector according to the current aspect comprises parts of the sequence of the pSET152 vector of Bierman et al. (1992).


Preferably, the vector does not comprise putative antisense promoters according to SEQ ID NO 108 and/or SEQ ID No. 109. These antisense promoters were identified by the inventors by sequencing of a 5′-primary transcript library and impair suitability of the vector pSET152. In brief, identification occurred by sequencing of an enriched primary transcript library. The two putative promoters were identified behind the gene of interest in antisense orientation (FIG. 26). These two pseudo-promoters were removed in order to prevent antisense transcription.


Furthermore, a T4-terminator was introduced behind the expression cassette in opposite orientation to prevent further putative antisense reads (cf. e.g. FIG. 6). In some embodiments, the vector comprises at least one T4-terminator (derived from the bacteriophage T4). T4-terminators can block transcription efficiently and prevent read-through from the integrase gene into the gene of interest. In some embodiments, the vector comprises a T4-terminator behind the expression cassette in opposite orientation to prevent further putative antisense reads. For example, the vector may comprise at least one T4-terminator before and/or at least one T4-terminator after the expression cassette. In some embodiments, the vector may comprise three terminators, one before and two after the expression cassette.


In some embodiments the vector comprises the φC31 integrase gene int. In some of these embodiments the φC31 integrase gene int is derived from pSET152. In some embodiments, the vector according to the first aspect furthermore comprises the attachment site attP. The integrase of the φC31 integrase gene int mediates the integration of the vector into the host chromosome at a distinct genomic location by catalyzing the targeted and unidirectional recombination of two attachment sites: attP, localized on the vector, and attB, localized in the host chromosome in the gene ACSP50_6589 (former: ACPL_6602) (te Poele et al. 2008; Gren et al. 2016). Without being bound by theory, after integration, the vector is flanked by the attachment site left (attL) and right (attR), which are derived from attP-attB-recombination (te Poele, Bolhuis und Dijkhuizen 2008).


In some embodiments the vector comprises an origin of transfer such as the origin of transfer (incP) and/or a relaxosome gene such as the relaxosome gene traJ. In some of these embodiments the origin of transfer such as the origin of transfer (incP) and/or the relaxosome gene, such as traJ are derived from pSET152. The origin of transfer and the relaxosome gene enable the transfer of the plasmid from the donor strain (e.g. Escherichia coli ET12567/pUZ8002 (Kieser et al. 2000)).


In some embodiments the vector according to the first aspect comprises an origin of replication such as the high-copy-number ColE1/pMB1/pBR322/pUC origin of replication (ori). In some of these embodiments the origin of replication such as the high-copy-number ColE1/pMB1/pBR322/pUC origin of replication (ori) is derived from pSET152. The origin of replication such as the high-copy-number ColE1/pMB1/pBR322/pUC origin of replication (ori) enables replication of the plasmid in the cloning strain (Escherichia coli DH5αMCR) and donor strain (Escherichia coli ET12567/pUZ8002).


In some embodiments the vector according to the first aspect comprises at least one resistance marker such as a resistance marker mediating apramycin resistance (aac(3)IV, apmR).


Resistance markers mediating apramycin resistance (aac(3)IV, apmR) can be used for selection.


According to some embodiments according to the fourth aspect the expression vector comprises at least one element of pSET152, such as (a) the φC31 integrase gene int according to SEQ ID No. 85, (b) the origin of transfer (incP) according to SEQ ID No. 87, (c) the relaxosome gene traJ according to SEQ ID No. 88, or (d) the high-copy-number ColE1/pMB1/pBR322/pUC according to SEQ ID No. 89, and furthermore does not comprise putative antisense promoters according to SEQ ID NO 108 and SEQ ID No. 109.


According to some embodiments according to the fourth aspect the expression vector comprises (a) the φC31 integrase gene int according to SEQ ID No. 85, and (b) the origin of transfer (incP) according to SEQ ID No. 87, and (c) the relaxosome gene traJ according to SEQ ID No. 88, and (d) an origin of replication, such as the high-copy-number ColE1/pMB1/pBR322/pUC, origin of replication (ori) according to SEQ ID No. 89 and (e) optionally at least one resistance marker, such as a resistance marker mediating apramycin resistance, such as aac(3)IV according to SEQ ID No. 90, apmR, and (f) optionally at least one T4-terminator, and (g) optionally, wherein the vector does not comprise putative antisense promoters according to SEQ ID NO 108 and/or SEQ ID No. 109.


According to some embodiments, the vector comprises the sequence according to SEQ ID No. 110 or SEQ ID No. 111. According to some embodiments, the vector comprises the sequence according to SEQ ID No. 110 or SEQ ID No. 111, or a fragment thereof.


In some embodiments, the vector is excelled by an easy cloning mechanism allowing integration of different promoters. By this, the system can be quickly adapted to further species, e.g. production strains of Acarbose.


EXAMPLES

General Tools and Methods


Strains and Plasmids


All strains used in this work are listed in Table E1. Recombinant strains used or created in this work are listed in Table E2, Table E3 and Table E4 (plasmid-based expression systems in Table E2, deletion and integration constructs cloned and stored in E. coli DH5αMCR in Table E3, deletion and integration mutants of Actinoplanes sp. SE50/110 in Table E4).









TABLE E1







Culture collection of microorganisms.












NCBI reference



strain
strain collection
sequence
reference





Actinoplanes sp. SE50/110
ATCC ®31044, CBS
NZ_LT827010.1
(Wolf etal. 2017b;



674.73

Frommer et al. 1979;





Parenti and Coronelli





1979)



Escherichia coli DH5αMCR

Mcr-deficient
NC_017638.1
(Grant et al. 1990)



derivative of E. coli



DH1



Escherichia coli



(Kieser et al. 2000)


ET12567/pUZ8002


Streptomyces lividans TK23
plasmid-free derivative
NZ_CP009124.1
(Kieser et al. 2000)



of S. lividans 66
(TK24 as




representative




genome)


Streptomyces coelicolor
ATCC ®BAA-471D-5,
NC_003888.3
(Bentley et al. 2002;


A3(2) M145
plasmid-free derivative

Dyson and Schrempf



of S. coelicolor A3(2)

1987)



ATCC ®BAA-471


Streptomyces glaucescens
DSM ®40922
NZ_CP009438.1
(Ortseifen


GLA.O


et al 2015)
















TABLE E2







Replicative and integrative vector systems.














E. coli

Actinoplanes



vector name
promoter and insert
DH5αMCR
sp. SE50/110
source







pSETT4 constructs (backbone created in this work)















pSETT4gap
PgapDH

Ec112

this work


pSETT4tip
PtipA

Ec117

this work


pSETT4::PacbB:acbB
PacbB
acbB (ACSP50_3608)
Ec120
Ac152
this work


pSETT4tip::acbB
PtipA
acbB (ACSP50_3608)
Ec119
Ac153
this work


pSETT4gap::acbB
PgapDH
acbB (ACSP50_3608)
Ec118
Ac154
this work


pSETT4tip::gtaB
PtipA
gtaB (ACSP50_7820)
Ec115
Ac150
this work
















TABLE E3







Vector systems for targeted deletion and integration


based on pCRISPomyces-2 of Cobb et al. (2015).













E. coli




vector name
insert
DH5αMCR
source





pCRISPomyces-2::sp:cgt_flanks
flanks for deletion of the
Ec018
this work



gene cgt (ACSP50_5024)


pCRISPomyces-2::sp1:merR_flanks
flanks for deletion of the
Ec109
this work



gene merR (ACSP50_0145)
















TABLE E4







Deletion and integration mutants obtained in


Actinoplanes ssp. by CRISPR/Cas9- technique.












Actinoplanes



strain
description
sp. SE50/110
source





Actinoplanes sp.
deletion mutant of the gene
Ac064
this work


SE50/110 Δcgt
cgt (ACSP50_5024)


Actinoplanes sp.
deletion mutant of the gene
Ac146
this work


SE50/110 ΔmerR
merR (ACSP50_0145)









Media and Cultivation Conditions


Unless otherwise specified, all chemicals and media components were obtained from Carl Roth GmbH & Co. KG (Karlsruhe, Germany), Sigma-Aldrich (St. Louis, USA), SERVA Electrophoresis GmbH (Heidelberg, Germany) or VWR International (Pennsylvania, USA).


Preparation of Glycerol Stocks of Actinoplanes sp. SE50/110


For preparation of glycerol stocks, Actinoplanes sp. SE50/110 (ATCC 31044) was grown in the complex medium NBS (11 g·L−1 glucose·1H2O, 4 g·L−1 peptone, 4 g·L−1 yeast extract, 1 g·L−1 MgSO4·7H2O, 2 g·L−1 KH2PO4, 4 g·L−1 K2HPO4) and mixed 2:3 with sterile 86% (v/v) glycerol. Glycerol stocks are stored at −80° C.


Growth on Solid Media and Preparation of Spore Solutions


For spore formation, 200-300 μL of a glycerol stock were grown on agar plates of soy flour medium (SFM-agar) (20 g·L−1 soy flour (SOBO® Naturkost (Cologne, Germany)), 20 g·L−1 D-mannitol, 20 g·L−1 Bacto™ agar (Becton-Dickinson, Heidelberg, Germany), 167 μL 10 N NaOH in tap water). Spores could be harvested after 5-7 days of incubation at 28° C. by washing them off in 3 mL ddH2O with a cotton swab, like described by Wolf et al. (2016).


Preparation of Minimal Medium


Maltose minimal medium (72.06 g·L−1 maltose 1H2O, 5 g·L−1 (NH4)2SO4, 0.184 g·L−1 FeCl2·4H2O, 5.7 g·L−1 Na3C6H5O7·2H2O, 1 g·L−1 MgCl2·6H2O, 2 g·L−1 CaCl2·2H2O, trace elements (final concentration: 1 μM CuCl2, 50 μM ZnCl2, 7.5 μM MnCl2 dissolved in 1 M HCl) and phosphate buffer consisting of 5 g·L−1 each K2HPO4 and KH2PO4 in ddH2O) was prepared and filter sterilized following the protocol of Wendler et al. (2013).


For substitution of the carbon source maltose, 79.2 g·L−1 glucose·1H2O, 72.0 g·L−1 C-pur (Cerestar 01908, CerestarGmbH, Krefeld, Germany), 71.9 g·L−1 galactose, 68.4 g·L−1 cellobiose, 71.9 g·L−1 D-arabinose or 72.0 g·L−1 D-lactose were used respectively, instead of maltose-monohydrate. Mixtures of maltose and glucose were prepared in the ratio of 90:10, 80:20 and 50:50 (v/v).


For the starch medium, a 4% (w/v) opalescent solution of “starch soluble” from Acros Organics (part of Thermo Fisher Scientific, Geel, Belgium) was generated. For this, sterile water was preheated to 90° C. in a water bath and the weighed portion of starch added with stirring.


Afterwards, the residual media components were added. To allow comparison to the starch-cultivation, a maltose minimal medium was created with comparable C-molarity (here net weight of 44.4 g·L−1 maltose 1H2O). Media of different pH and osmolarity were created by addition of correcting agents (HCl or NaOH), by varying of the concentration of the carbon-sources maltose respectively by addition of inositol, which is not metabolized according to our study (data not shown).


Furthermore, minimal media with 1 g·L−1, 2 g·L−1, 3 g·L−1, 4 g·L−1 and 5 g·L−1 “starch soluble” from Acros Organics were created for cultivation under limited carbon-source.


The pH and osmolarity of all media were determined by the pH-meter Calimatic of Knick GmbH (Berlin, Germany) and the Osmomat 3000 of Gonotec GmbH (Berlin, Germany) according to the manufacturer's instructions.


Shake Flask Cultivation


Cultivations were performed in 250 mL Corning® Erlenmeyer baffled cell culture flasks at 28° C. and 140 rpm for seven days in the GFL shake-imcubators 3032 or 3033 (Burgwedel, Germany).


For inoculation of 50 mL medium, 1 mL spore solution of an OD=3-5 was used. Cell dry weights were determined like described by Wolf et al. (2017a). The supernatant was stored at −20° C. for later analysis.


Miniaturized cultivation in the BioLector system of m2p-labs GmbH (Baesweiler, Germany)


Comparative growth experiments were performed in a 1 mL reaction volume in a 48-well FlowerPlate covered by a gas-permeable sealing foil (m2p-labs GmbH, Baesweiler, Germany) and incubated for 1 week at 28° C. and 800 rpm in the RoboLector® of m2p-labs. Growth was recorded by the backscatter signal. For determination of final cell dry weights, 800 μL of each well was sampled in a weighed reaction tube (14,000 g, 2 min), washed with deionized water and dried for 1 day at 60-70° C. The supernatant was stored at −20° C. for later analyses.


Recombinant DNA Work


Unless otherwise specified, plasmid construction and assembly was performed by Gibson Assembly (Gibson et al. 2009). Fragments were amplified by PCR (Phusion® High-Fidelity PCR Master Mix with GC Buffer, NEB, Ipswich, Mass., USA) in the Eppendorf thermocycler vapo.protect (Hamburg, Germany) and treated with DpnI (Thermo Fisher Scientific, Waltham, Mass., USA), when necessary. Purification of PCR products and gel extracts was performed by use of the NucleoSpin® Gel and PCR Clean-up kit (Macherey-Nagel, DQren, Germany). Equimolar amounts of the DNA fragments were added to the Gibson Assembly Master Mix in a ratio of 1:4.


The master mix consists of 0.64 μL T5 Exonuclease (10 U·μL−1, NEB, Ipswich, Mass., USA), 20 μL Phusion High-Fidelity DNA Polymerase (2 U·μL−1, Thermo Fisher Scientific, US), 160 μL Taq DNA Ligase (NEB, Ipswich, Mass., USA), 699.36 μL aqua distilled and 320 μL isothermal reaction buffer (25% PEG-8000, 1 mL 1 M Tris-HCl, 100 μL 1 M MgCl2, 100 μL 1 M DTT, 20 μL each 1 mM dNTP, 200 μL NAD). The sample was incubated at 50° C. for at least 1 h and subsequently transferred to Escherichia coli DH5αMCR by chemical transformation according to a protocol of Beyer et al (2015). Selection of E. coli was performed on Luria/Miller broth medium with 15 g·L−1 agar-agar (Carl Roth, GmbH&Co.KG, Karlsruhe, Germany)) and 50 mg·L−1 apramycin-sulfate.


Positive colonies were tested by PCR and gel-electrophoresis as well as by Sanger sequencing by our in-house sequencing core facility.


Construction of Plasmids for the gusA Reporter System


For the construction of plasmids for the gusA reporter system see Schaffert et al. (2019).


Construction of the Novel pSETT4 Expression System


For cloning of the novel pSETT4 expression system, the pSET152 vector of Bierman et al. (1992) was used as template. The vector backbone was linearized by PCR (Table E5).


The cloning cassette, consisting of the gapDH-promoter, a lacZ-gene under control of the lac-promoter and several restriction sites flanked by three T4-terminators, was ordered as string DNA at Integrated DNA Technologies (Iowa, USA). Due to the complex structure, the cassette was ordered in three parts and assembled by GeneSOEing (Horton 1995) by use of the primers in Table E5. Finally, backbone and insert were assembled by Gibson Assembly (Gibson et al. 2009). The novel vector system was named pSETT4gap.


For exchange of the gapDH-promoter by the tipA-promoter, pSETT4gap was digested with NdeI and KpnI and treated with shrimp alkaline phosphatase following the instructions of the supplier.


All enzymes were purchased from Thermo Fisher Scientific (Waltham, Mass., USA). The tipA-promoter was amplified from pSETGUS (Myronovskyi et al. 2011) by use of the primers tipA_GAF and tipA_GAR and assembled with the linearized backbone by Gibson assembly (Gibson et al. 2009). The vector was named pSETT4tip (cf. FIG. 6).









TABLE E5







Gibson Assembly primer for assembly of the novel expression system


pSETT4gap and pSETT4tip.












size



fragment
template
(bp)
primer sequence (5’-3’)





pSET152_lin
pSET152
5114
CTACGGTGCCGCTTACCGGgctcactcaaaggcggtaatac





gg





CAGACGTCAGCGACGACAGAGaaccatcggcgcagctatt





tac





genesoeing_
IDT-order 1 and
1473
CTCTGTCGTCGCTGACGTCTG


for
2




genesoeing_1r


CAGATCTGGAGTCGGTCTAATTT





genesoeing_2f
IDT-order 2 and
 878
AGGGTTTTCCCAGTCACGACG


genesoeing_
3

CCGGTAAGCGGCACCGTAG


rev








tipA_GAF
pSETGUS
 146
GTGGCCCATGCGAGAGTACAATCCCTAGAACGTC





CGGG


tipA_GAR


TCAACATAAGGTCTCGGTACCATCGGAATACCTCC





GTTGCT









Overexpression of Single Genes in the Novel pSETT4 Expression System


For the overexpression of single genes, the insert was amplified by PCR (Table E6). The vector (pSETT4gap or pSETT4tip), was digested with BsaI (NEB, Ipswich, Mass., USA) and assembled with the insert by Gibson Assembly (Gibson et al. 2009). For expression of the acbB gene under control of the native promoter, the vector backbone pSETT4gap was digested with BsaI and NdeI, leading to the linearization of the vector under removal of the promoter. The gene of interest and the native promoter were amplified by use of the primers in Table E6 and assembled with the vector backbone by Gibson Assembly (Gibson et al. 2009).









TABLE E6







Primer for amplification of inserts for Golden Gate cloning and restriction cloning


into the pSETT4gap and pSETT4tip vector system.












size



fragment
template
(bp)
primer sequence (5’-3’)





acbB for
gDNA
1008
GAGTATCTGAAAGGGGATACGCATGAAAATCTTGGTCA


pSETT4gap


CCGGCGGAGC





GGCGGAAAATCACGCGGCACGAATCAGGTCCACCAGG





AACGGTTGGC





acbB for pSETT4tip
gDNA
1006
CGAGCAACGGAGGTATTCCGATGAAAATCTTGGTCACC





GGCGGAGC





GGCGGAAAATCACGCGGCACGAATCAGGTCCACCAGG





AACGGTTGGC





PacbB:acbB for
gDNA
1136
GGCCCATGCGAGAGTACATAGCCAGCCTTTCATGATAT


pSETT4


ATCTC





AATCACGCGGCACGAAACGCACCGGATCCATGTTGTG





TGG





gtaB for pSETT4tip
gDNA
 950
GCAACGGAGGTATTCCGATGACGACGAACGCGCAAGG





G





GGAAAATCACGCGGCACGAAGTCATCCCTTCTGACCA





CCGACG









Construction of pCRISPomyces-2 Deletion and Integration Vectors


For the construction of deletion and integration mutants by CRISPR/Cas9 technique, the plasmid pCRISPomyces-2 (Cobb et al. 2015) was used according to a protocol of Wolf et al. (2016). The spacer and its reverse complement were ordered at metabion GmbH (Steinkirchen, Germany) or Sigma-Aldrich (Taufkirchen, Germany) as oligonucleotides with overlap (Table E7).


The oligonucleotides were annealed to a double-strand and assembled with the plasmid by Golden Gate Assembly (Engler et al. 2008) according to the protocol of Cobb et al. (2015). For repair of the Cas9-induced double-strand break, a DNA template was cloned into the vector backbone by Gibson Assembly (Gibson et al. 2009). As DNA template, flanking sequences up- and downstream of the target gene (each round about 1 kB) were amplified by PCR (Table E8) from genomic DNA.









TABLE E7







Spacer and the reverse complement used in a Golden Gate Assembly with


pCRISPomyes-2.









gene
oligo 1 (5’-3’)
oligo 2 (5’-3’)





cgt (ACSP50_5024)
acgcAGCGTCGCCCGCTGGGAG
aaacTTCTCCCAGCGGGCGACGCT



AA






merR (ACSP50_0145)
acgcGACCGGGGGCTGTCCGG
aaacCTCCCGGACAGCCCCCGGTC



GAG
















TABLE E8







Gibson Assembly primer for pCRISPomyes-2 deletion and integration vectors.













size

size


gene
primers for flank 1 (5′-3′)
(bp)
primers for flank 2 (5′-3′)
(bp)





cgt
tcggttgccgccgggcgttttttatCC
1101
gtatctgagccatatccctcGACCTGC
982


(ACSP50_5024)
GGTACCCTGCTCCTCGT

GTCAATGCGTCAC




C


gcggcctttttacggttcctggcctACCTG







embedded image



ACCCTGCTGAAATGG






embedded image










merR
tcggttgccgccgggcgttttttatCT
1115
gcaggtggacggcctcggtgATCTCG
1129


(ACSP50_0145)
CCGGGCGCCGACCGGC

GCGCTCAACGCCTC




AC


gcggcctttttacggttcctggcctCGGC






gaggcgttgagcgccgagatCAC


AAACAGACCTACTACG




CGAGGCCGTCCACCTGC









Deletion of the gene cgt by CRISPR/Cas9 technique


For the construction of a Δcgt (ΔACSP50_5024) deletion mutant by CRISPR/Cas9 technique (clustered regular interspaced short palindromic repeats/CRISPR-associated endonuclease 9), the plasmid pCRISPomyces-2 was used (Cobb et al. 2015). The spacer sequence was selected according to Wolf et al. (2016) and ordered as oligonucleotides together with its reverse complement at metabion GmbH (Steinkirchen, Germany) (spacer_1: 5′-acgcAGCGTCGCCCGCTGGGAGAA-3′, spacer_2: 5′-aaacTTCTCCCAGCGGGCGACGCT-3′).


The oligonucleotides were annealed to a double-strand and assembled with the plasmid by Golden Gate Assembly (Engler et al. 2008) by use of BsaI (NEB, lpswich, MA, USA) according to the protocol of Cobb et al. (2015) (Cobb et al. 2015). For repair of the Cas9-induced double-strand break, a deoxyribonucleic acid (DNA) template was cloned into the XbaI-linearized vector by Gibson Assembly (Gibson et al. 2009). As DNA template, flanking sequences up- and downstream of the target gene (each round about 1 kB) were amplified by polymerase chain reaction (PCR) with the Phusion® High-Fidelity PCR Master Mix with GC Buffer (NEB, Ipswich, Mass., USA) (Primer sequences: cgt_flank1_fw: 5′-tcggttgccgccgggcgttttttatCCGGTACCCTGCTCCTCGTC-3′, cgt_flank1_rv: 5′-gtgacgcattgacgcaggtcGAGGGATATGGCTCAGATAC-3′, cgt_flank2_fw: 5′-gtatctgagccatatccctcGACCTGCGTCAATGCGTCAC-3′, cgt_flank2_rv: 5′-gcggcctttttacggttcctggcctACCTGACCCTGCTGAAATGG-3′). For Gibson Assembly, the DNA fragments (flank_1: 1101 bp and flank_2: 982 bp) were mixed equimolar added in a ratio of 1:4 to the Gibson Assembly Master Mix consisting of 0.64 μL T5 Exonuclease (10 U/μL, NEB, Ipswich, Mass., USA), 20 μL Phusion High-Fidelity DNA Polymerase (2 U/μL, Thermo Fisher Scientific, US) and 160 μL Taq DNA Ligase (40 U/μL NEB, Ipswich, Mass., USA), 699.36 μL aqua distilled and 320 μL isothermal reaction buffer (25% PEG-8000, 1 mL 1 M Tris-HCl, 100 μL 1 M MgCl2, 100 μL 1 M DTT, 20 μL each 1 mM dNTP, 200 μL NAD). After incubation at 50° C. for at least 1 h, the reaction mix was transferred to Escherichia coli DH5αMCR by chemical transformation according to a protocol of (Beyer et al. 2015). Growth and selection of E. coli was performed by plating them on Luria/Miller broth (LB-media) with 15 g·L−1 agar-agar Kobel (both: Carl Roth, GmbH&Co.KG, Karlsruhe, Germany)) supplemented with 50 mg·L−1 apramycin-sulfate. Plates were incubated for 10-14 h at 37° C. Apramycin-resistant colonies were tested by PCR and gel-electrophoresis first, and second by Sanger sequencing by our in-house sequencing core facility (primer sequences for PCR: for: 5′-GGCGTTCCTGCAATTCTTAG-3′, rev: 5′-TCGCCACCTCTGACTTGAGC-3′, walking primer for sequencing: w1: 5′-CGCTGATCTTCAGCTTCC-3′, w2: 5′-GCCTTCACCTTCCATCTG-3′, w3: 5′-TCGGGAAAGCCGCCGGAG-3′)).


Conjugal transfer to Actinoplanes sp. SE50/110


Competent Actinoplanes sp. SE50/110 cells were prepared from a freshly grown NBS-culture (see above). Cells were washed twice in 10% (w/v) ice-cold sucrose and twice in ice-cold 15% (v/v) glycerol. Finally, the cells were taken up in 15% (v/v) ice-cold glycerol (by addition of round about the four-fold volume of the cell pellet), aliquoted to 100 μL in reaction tubes and snap-frozen in liquid nitrogen. The competent Actinoplanes cells are stored at −80° C.


For conjugation, Escherichia coli ET12567/pUZ8002 (Kieser et al. 2000) was used. After transfer of the desired construct into E. coli ET12567/pUZ8002 according to Beyer et al. (2015) and selection on LB agar plates supplemented with 50 mg·L−1 apramycin-sulfate, 50 mg·L−1 kanamycin-sulfate and 15 mg L−1 chloramphenicol, cells were grown in liquid culture (LB-medium with the same supplements) and harvested at an optical density of 0.4-0.6. The cells were washed twice in ice-cold LB medium and mixed with competent cells of Actinoplanes sp. SE50/110. The cell suspension was plated on SFM agar plates. After 20-24 h of incubation at 28° C., 1 mL 500 mg·L−1 apramycin-sulfate dissolved in ddH2O was distributed on the plate with a sterile swab. First exconjugants of Actinoplanes sp. SE50/110 can be observed after 1 week. Exconjugants were transferred to an SFM agar plate supplemented with 50 mg·L−1 apramycin-sulfate. Repeated streaking is performed for several times to purify Actinoplanes exconjugants from E. coli. To expedite this process, 50 mg·L−1 fosfomycin or trimethoprim can be supplemented to the medium to get rid of the donor strain.


Plasmid Curing to Obtain Marker-Free CRISPR/Cas9 Deletion/Integration Mutants of Actinoplanes sp. SE50/110


Plasmid curing was performed according to the protocol of Wolf et al. (2016) by cultivation in the complex medium NBS at elevated temperatures. Colonies were tested for the presence of the plasmid by parallel streaking on apramycin-containing and apramycin-free SFM plates. Apramycin-sensitive exconjugants were tested for the deletion by PCR (primer sequence data not shown). The PCR fragment was excised from the gel and sequenced by our in-house Sanger sequencing core facility.


Additionally, also genomic DNA of the deletion or integration mutant was sequenced by the Oxford Nanopore technique (Oxford, UK) to exclude off-target effects. For this, genomic DNA of an NBS-grown culture was isolated with the NucleoSpin® Microbial DNA kit (Macherey-Nagel, DQren, Germany). A library was prepared with help of the 1D Genomic DNA by ligation-kit (Oxford Nanopore, Oxford, UK).


Deletion System Based on Homologous Recombination and Counterselection with the Cytosine Deaminase CodA.


Vector integration into genes of the acb gene cluster has occurred by use of the replicative vector pKC1139. Based on this observation, a novel deletion system using homologous recombination was developed and tested by example of the gene cgt (ACSP50_5024).


A vector backbone with origin of transfer (ncP) and relaxosome gene traJ was used to allow conjugation into Actinoplanes sp. SE50/110. In this work, two different antibiotic resistance markers mediating apramycin and kanamycin resistance were tested for selection: aph(3′)II (Kan®, kanamycin) and aac(3)IV (Apm®, apramycin). Furthermore, the high-copy-number ColE1/pMB1/pBR322/pUC origin of replication was integrated to allow replication in the donor strain E. coli. The ori, the oriTncP, tra gene and resistance cassettes were taken from pRT802 respectively pRT801 (Gregory et al. 2003). Since no replicon for replication in Actinoplanes sp. SE50/110 neither an integrase gene with attachment site are contained in the novel deletion system, the vector can only be maintained in Actinoplanes sp. SE50/110 when being integrated into the genome by homologous recombination (FIG. 7). For this, homologous sequences of 2 kB were integrated, which are flanking the gene cgt. After conjugal transfer in Actinoplanes sp. SE50/110, mutants, in which the first crossover has taken place, can be selected by apramycin or kanamycin resistance. To force desintegration of the vector backbone (second crossover), 5-fluorocytosine (5-FC) is added, which is converted into the toxic product 5-fluorouracil (5-FU) by the cytosine deaminase CodA. In this work, codA(s) is used, which is codon-optimized for Streptomyces ssp. (Dubeau et al. 2009). After second crossover either the genotype of the wild type or the genotype of the deletion mutant is present.


The novel deletion system was successfully tested for the gene cgt, which was shown by colony PCR and ONT-sequencing. The proportion of deletion mutants after successful second crossover was between 25% and 32%. The workflow is illustrated in FIG. 8.


Analytical Methods


Acarbose Quantification from the Supernatant by High Performance Liquid Chromatography (HPLC)


Supernatants of maltose-grown cultures of Actinoplanes ssp. were centrifuged (20,000 g, 2 min), mixed 1:5 with methanol by vortexing and centrifuged again to remove the precipitate (20,000 g, 2 min). The samples were transferred to HPLC vials and analyzed in the HPLC system 1100 series of Agilent (G1312A Binary Pump Serial #DE43616357, G1329A ALS autosampler Serial #DE43613/10, G1315A diode-array detector (DAD) Serial #DE72002469). As stationary phase the Hypersil APS-2 column (125×4 mm, 3 μm particle size) of Thermo Fisher Scientific Inc. (Waltham, Mass., USA) was used, heated to 40° C. As mobile phase an isocratic flow of 1 mL·min−1 68% acetonitrile (solvent B) and 32% phosphate buffer (0.62 g·L−1 KH2PO4 and 0.38 g·L−1 Na2HPO4·2H2O) (solvent A) was applied. 40 μL of each sample was injected and separated in a 10 min run. Detection of acarbose was carried out with a DAD detector at 210 nm (Reference 360 nm) and quantified from the peak areas of a calibration curve.


Liquid Chromatography-Mass Spectrometry (LC-MS)


Sample Preparation for Analysis of Intracellular Metabolites


Triplicates of Actinoplanes sp. SE50/110 strains were grown in maltose minimal medium for at least 4 days. 10 mL of the culture were quickly filtrated through filtering paper by a Buchner funnel and washed with 2.63 g·L−1 NaCl solution. Cells were transferred into pre-weighted round bottom screw-cap tubes, snap-frozen in liquid nitrogen and stored at −80° C. Cells were dried overnight in the Centrifugal Evaporator (SpeedVac) of Thermo Fisher Scientific (Waltham, Mass., USA). 4 mg dried cells were transferred into a fresh 2 mL screw-cap tube containing round about 500 μL of a mixture of zirconia/silica micro beads of the sizes 0.1 mm, 0.05 mm and 0.01 mm (Bio Spec Products Inc., Bartlesville, USA). 700 μL 80% MeOH was added to the cells and beads. Cell disruption was carried out in a homogenizer (FastPrep FP120, Thermo Fisher Scientific, Waltham, Mass., USA) for three times 30 s at speed setting 6.5. Samples were cooled for 5 min on ice in between. The cell suspension was centrifuged for 5 min at 13,000 g and 4° C. 500 μL of the supernatant was transferred into HPLC vials, dried under nitrogen flow and taken up in 50 μL distilled water.


Sample Preparation for Analysis of Extracellular Acarviosyl-Metabolites


The sample preparation was conducted according to a protocol described by Ortseifen (2016). Sugars and pseudo-sugars were enriched from 10 mL of the supernatant by solid phase extraction using the Chromabond® Easy columns (Macherey-Nagel, DQren, Germany, REF 730753). The columns were equilibrated with 3 mL methanol, afterwards washed with 3 mL distilled water before loading of the sample. Unspecific bound metabolites were rinsed by 3 mL 95% (v/v) methanol. Elution was conducted in 3 mL methanol.


LC-ESI-MS of Intracellular and Extracellular Metabolites


For LC-MS, the LaChromUltra (Hitachi Europe Ltd., UK) HPLC system coupled to a microTOF-Q hybrid quadrupole/time-of-flight mass spectrometer (Bruker Daltonics, Bremen, Germany) was used, which was equipped with an electrospray ionization (ESI) source.


For the analysis of intracellular metabolites, 2 μL of the sample was separated with the SeQuant® ZIC®-pHILIC 5 μm Polymeric column (150×2.1 mm) (Merck, Darmstadt, Germany). Eluent A (20 mM NH4HCO3, pH 9.3, adjusted with aqueous ammonia solution) and eluent B (acetonitrile) were applied at a flow rate of 0.2 mL·min−1 by use of the following gradient: 0 min B: 90%, 30 min B: 25%, 37.5 min B: 25%, 40.0 min B: 80%.


As standards for the peak identification, 2 μL of 10 μM of UDP-glucose, glucose-1-phosphate, galactose-1-phosphate, glucose-6-phosphate and dTDP-glucose were injected.


For the analysis of extracellular acarviosyl-metabolites, 10 μL of the sample was separated with the Cogent Diamond Hydride™ HPLC column (MicroSolv Technology Corporation; 150 mm×2.1 mm; 3 μL particle size). Eluent A (50% (v/v) acetonitrile, 50% (v/v) H2O und 0.1% (v/v) formic acid) and eluent B (90% (v/v) acetonitrile, 10% (v/v) H2O und 0.1% (v/v) formic acid) were applied at a flow rate of 0.4 mL·min−1 by use of the following gradient: 0 min B: 100%, 8 min B: 0%, 13 min B: 0%, 15.5 min B: 100%, 18 min B: 100%.


The ESI source was operated in the negative ionization mode for analysis of intracellular metabolites and in the positive ionization mode for analysis of extracellular acarviosyl-metabolites. The temperature of the dry gas and the capillary was set to 180° C. The scan range of the MS was set to 200-1,000 m/z (intracellular metabolites) respectively 50-3,000 m/z (extracellular acarviosyl-metabolites)


The peak areas of specific masses were integrated by use of the software Compass™ (Bruker Daltonics, Bremen, Germany). Peaks were normalized on the weighed amount of dried cells (intracellular metabolites) respectively the cell dry weight at sampling time (extracellular acarviosyl-metabolites).


Extraction and Analysis of Carotenoids


Extraction


Cell pellets from Actinoplanes sp. SE50/110 were transferred into a 2 mL screw-cap tube with round about 500 μL of a mixture of zirconia/silica micro beads of the sizes 0.1 mm, 0.05 mm and 0.01 mm (Bio Spec Products Inc., Bartlesville, USA). 1 mL acetone or methanol was added as extracting solvent. Cell disruption was carried out in a homogenizer (FastPrep FP120, Thermo Fisher Scientific, Waltham, Mass., USA) for three times 45 s at speed setting 6.5. Samples were cooled for 5 min on ice in between. The homogenized cell suspension was centrifuged for 20 min at 13,000 g and 4° C. The supernatants were transferred into glass vials. For HPLC-analysis, mixtures of the acetone- and methanol-extracts were created in the ratio of 7:3 and transferred into a novel glass vial.


Thin Layer Chromatography (TLC) and Spectral Analysis


50 μL of the extracted carotenoids were applied in 5 μL-steps onto a silica gel matrix (HPTLC-HL, Cat. 58077, Analtech Inc., Newark, USA) and incubated in a TLC-chamber filled with 100 mL petroleum, 11 mL isopropanol and 50 μL water. The run was carried out in darkness. After drying of the TLC-plate, bands were stripped off with a scalpel and transferred into a novel tube. After addition of 1 mL ethanol, the absorption spectrum was analyzed by use of the Genesys 10S UV-Vis spectrophotometer of Thermo Fisher Scientific (Waltham, Mass., USA).


HPLC Analysis of Carotenoids with Absorbance Scan


Carotenoids were separated by reversed-phase HPLC according to Henke et al. (2017) and Heider et al. (2014) using the Agilent 1200 series HPLC system (Agilent Technologies GmbH&Co. KG, Böblingen, Germany) including diode array detector (DAD) for the UV-Vis spectrum. 20 μL sample volume was applied to a flow of 0.5 mL−min−1. As stationary phase a pre-column (10×4 mm MultoHigh 100 RP18-5) and a main column (ProntoSIL 200-5 C30, 250×4 mm) from CS ChromatographieService GmbH (Langerwehe, Germany) were used, like described before (Heider et al. 2014; Henke et al. 2017).


Following gradient was applied: 0 min A: 100%, 32 min A: 75%, 47 min A: 0%, 70 min A: 0%, 75 min A: 100%, with eluent A consisting of 0.1 M ammonium acetate in deionized water and methanol in the ratio of 15:85 (v/v). Eluent B consists of a mixture of methanol, acetonitrile and acetone in the ratio of 44:43:13 (v/v). Detection of carotenoids was conducted at 470 nm. Additionally, wavelength scans between 360 nm and 700 nm were performed each second during the run.


Assays


Promoter Screening Experiment by Spectrophotometric Measurement of the Glucuronidase Activity


Two different types of glucuronidase assay were carried out: one with protein raw extract and one with entire cells. The protocols described by Horbal et al. (2013) and Siegl et al. (2013) were adapted to Actinoplanes sp. SE50/110. The substrate 5-bromo-4-chloro-3-indolyl-p-D-glucuronide (X-Gluc, AppliChem GmbH, Darmstadt, Germany) was chosen, as the substrate p-nitrophenyl-D-glucuronide turned out to dissociate under our assay conditions.


Growth Conditions and Sample Preparation



Actinoplanes mutants carrying promoter constructs with gusA gene, were cultivated for one week in maltose minimal medium, like described above. The assays were conducted during growth phase. 500 μL of each culture was sampled for an assay with entire cells. 1 mL was sampled for an assay with protein raw extract and transferred to a screw cap tube containing zirconia/silica micro beads (Bio Spec Products Inc., Bartlesville, USA) of the sizes 0.1 mm and 0.05 mm. Cells were disrupted in a homogenizer (FastPrep FP120, Thermo Fisher Scientific, Waltham, Mass., USA) for two times 30 s at speed setting 6.5 and 5 min on ice in between. After centrifugation, the lysate was transferred to a new reaction tube and centrifuged. The supernatant was used for a cell-free assay. Total protein quantification was carried out by a Bradford assay (see above).


Glucuronidase (Gus) Assay


The gus assay was performed in a black microtiter plate (96 well PS F-bottom pCLEAR, black, med. binding, Greiner Bio-One, Kremsmünster, Österreich, REF 655096). 100 μL of each sample (either cell suspension or lysate) was pipetted in three wells, of which one serves as negative control and two as technical replicates. The gus buffer (50 mM phosphate buffer pH 7.0 (5.136 g·L−1 Na2HPO4·2H2O, 3.299 g·L−1 NaH2PO4·2H2O) with 5 mM DTT and 0.1% Triton-X-100) was complemented with 2 mM substrate X-Gluc (stock solution: 0.2 M in DMF). 100 μL was added to 100 μL of the sample. For the negative control, 100 μL gus buffer without substrate was added. Beside of the individual negative control of each sample, also medium and substrate controls were prepared.


The microtiter plate was measured in a pre-warmed Tecan reader Infinite M200 (Ref 30016056, Tecan Group AG, Männedorf, Switzerland) (37° C.) for 3 hours (assay with entire cells), respectively for 2 hours (assay with lysate). The absorption maxima of indigo were measured at 610 and 660 nm. After discounting the absorption value of all controls, the slope of each absorption curve was calculated by linear regression and normalized either on cell dry weight (assay with entire cells) or on whole protein amount (assay with lysate). The normalized slope was used to compare the p-glucuronidase activities in the different mutants.


Screening experiments in the Biolog® OmniLog Phenotypic Microarray System


Pre-screening experiments were performed in the Biolog® OmniLog Identification System (Hayward, Calif., USA) to evaluate respiration on different carbon sources (panel PM1 and PM2). Actinoplanes sp. SE50/110 wild type and the deletion mutant Δcgt were grown on SFM agar plates, as described elsewhere herein. Cells were harvested by use of a sterile swab and diluted in the inoculating fluid IF-0a for PM1 and PM2. The turbidity of the cell suspension was checked to achieve 80% transmittance in the turbidimeter of Biolog®, according to manufacturer's protocol. 2.32 mL of the cell suspension was added to 20 mL IF-0a, 0.24 mL 0.5 M MgCl2, 0.24 mL 0.5 M Na2SO4, 0.24 mL 1.5 M NH4Cl, 0.24 mL 1.0 M Na3PO4, 0.24 mL aqua distilled, 0.24 mL Biolog redox dye mix G, and 0.24 mL metal ion cocktail (5.0 mM each: ZnCl2·7H2O, FeCl2·6H2O, MnCl2·4H2O, CaCl2·2H2O), according to the manufacturer's protocol. The PM panels were inoculated with 100 μL per well of the prepared solution and incubated for 1 week in the OmniLog system (Mode 71000 Serial #406) at 28-30° C. Data evaluation was carried out with the manufacturer's software (Kinetic Analysis, Biolog and Omnilog 2.3, Biolog).


RNA Work


Sampling and RNA Isolation


For transcriptome analysis, 2×1 mL culture were taken during growth phase, separated from the supernatant by centrifugation (10 s) and snap-frozen in liquid nitrogen. Pellets were stored at −80° C. until further processing.


For isolation of ribonucleic acid (RNA), frozen cell pellets were resuspended in 500 μL LB-buffer (NucleoSpin® RNA Plus, Macherey-Nagel, DQren, Germany) and transferred to 2 mL lysing matrix tubes (0.1 mm spherical silica beads, MP Biomedicals, Santa Ana, Calif., USA). Cell disruption was carried out in a homogenizer (FastPrep FP120, Thermo Fisher Scientific, Waltham, Mass., USA) for three times 20 s at speed setting 6.5 and 5 min on ice in between.


Subsequently, the cell suspension was centrifuged for 5 min at 13,000 g and 4° C. The supernatant was used for RNA extraction using the NucleoSpin® RNA Plus kit in combination with rDNase Set (Macherey-Nagel, DQren, Germany) for an on-column DNA digestion. After clean-up and elution according to the manufacturer's protocol, the DNA-digestion was repeated (in solution) and the sample cleaned up again by use of the same kit. With two primer pairs binding to the genomic DNA of Actinoplanes sp. SE50/110 and amplifying small fragments at round about 200-300 nt, the sample was tested for residual DNA. DNA digestion and RNA clean-up was repeated, if necessary. The quantity of RNA was analyzed with the NanoDrop 1000 spectrometer (Peqlab, Erlangen, Germany).


Reverse Transcription Quantitative PCR


Reverse transcription quantitative PCR was carried out according to the protocol of Wolf et al. (2017a) by use of SensiFast SYBR No-Rox One-Step kit (Bioline, London, UK) and 96 well lightcycler plates (Sarstedt, Numbrecht, Germany) in a LightCycler 96 System of Roche (Mannheim, Germany). The relative RNA amount was normalized on total RNA (100 ng) and calculated as 2−ΔCq. ΔCq is the difference of the mean Cq in the mutant strain compared to the control strain. The primers in Table E9 were used for determination of the relative transcription of a gene.









TABLE E9







Primers used in RT-qPCR experiments.













fragment





size


genetic locus
forward primer (5’-3’)
reverse primer (5’-3’)
(bp)





merR (ACSP50_0145)
GAGCGATACGCCCCTGACC
GGTGATGTCCGGGCTCGTG
309





idi (ACSP50_0146)
GCCTTCTCGGTCTTCCTCAC
CGCCAATTCCTCGGTGAGA
168




C






crtl (ACSP50_0147)
CTCTCGGTCGGCGGATAC
GAGCCGTCCGGGTAGTACG
158




C






crtE (ACSP50_0148)
TTCCTCGCCTCCCAGATCG
CGCGAAGGTGTGCATCAG
210





crtB (ACSP50_0149)
CATGTGCACGCTCTGTATG
AAGACCGCGATGGTGTGCA
185



G
G






acbZ (ACSP50_3590)
CGGCAATTCGCTGTTCAGT
TGTGCTTGACGGTGTCCAT
167



G
C






acbY (ACSP50_3591)
TCCGAACGGTTCCTCTATCC
AACTCGCTGAGCTGGTTGA
239




C






acbX (ACSP50_3592)
TCGGGATGCTGCACACCAA
CGACGCGAACATCGCGAAA
191



C
C






acbW (ACSP50_3593)
GGTGTACGACCGGAACATG
GTTCGGCGTGGATGTGGTT
224



C
G






acbV (ACSP50_3594)
GCTTCCACGGCAAGACGAT
GCGCTCACGTTGGGTTTCT
196



G
C






acbS (ACSP50_3596)
GTTGCCGGACCGGTTCTAT
CCCGGTACACCGACTTGTT
248



C
G






acbQ (ACSP50_3601)
TGCTGGCGCAGATCTACTC
AGCCGCAGATACATCGGGT
211



C
C






acbK (ACSP50_3602)
CGAGGTCTACGCCTTCAAC
AGAGGAAGCCGGACACGAA
248



G
C






acbC (ACSP50_3607)
GATCGCGCTGATCAAGGAT
CTGAACGTGTGCCCGTAGT
213



G
C






acbB (ACSP50_3608)
GTCGACAAACTGGGTTACG
GTCCAGTAGCACCTGAGTG
231



G







acbA (ACSP50_3609)
TCATGCTCGGCGACAACCT
GACCGGTTTCTCCTCGATG
173



G
G






acbE (ACSP50_3610)
GCGCGGCATGAAGATCTAC
CGGACGGCTTCTCGAAGAA
218



C
C






acbD (ACSP50_3611)
ACGCCAACTACTGGATGGA
TCGAGCGGTTGGTGTAGAA
231



C
G






cgt (ACSP50_5024)
CACCACGTACTGGAACTC
GCGACCTTCAACGTGAC
192





gtaB/galU
CTCGCCTTCATCGAGGTCA
GGCGATCGTCTCGAAGATC
192


(ACSP50_7820)
C
C






gusA
ACGCGGACATCCGCAACTA
CCCTGGTGCTCCATCACTT
157



C
C









Whole Genome Oligonucleotide Microarray


Whole genome oligonucleotide microarrays were performed according to a protocol of Wolf et al. (2017a), who adapted the hybridization procedure to the high G+C content of Actinoplanes sp. SE50/110.


RNA of triplicates was isolated and equimolar pooled (total amount of 5 μg pooled RNA in 12 μL). For cDNA synthesis, labeling and microarray hybridization the Two-Color Microarray-Based Prokaryote Analysis FairPlay III Labeling kit (Version 1.4, Agilent Technologies, Santa Clara, Calif., USA) was used according to the manufacturer's instructions with practical adjustments described by Wolf et al. (2017a). The Amersham CyDye mono-reactive dye packs (GE Healthcare, Little Chalfont, UK) were utilized for labeling. A custom whole genome oligonucleotide microarray representing the coding sequence of Actinoplanes sp. SE50/110 was used, which was designed by Wolf et al. (2017a) (4×44 K format, 43,803 features representing 8,238 genes and 1,417 control spots, supplier: Agilent Technologies, Santa Clara, Calif., USA). All microarray specific reagents and device including hybridization oven and scanner were used from Agilent Technologies (Santa Clara, Calif., USA). The Agilent Feature Extraction Software Version 10.7.3.1 (Agilent Technologies, Santa Clara, Calif., USA) was used for feature extraction (protocol GE2_107_Sep09). Subsequent data analysis, including LOWESS normalization and statistical analysis were performed by use of the microarray and gene expression (MAGE)-compliant system EMMA 2 (Dondrup et al. 2009). A p-value of 0.05 was used as a cut-off for significance. The M-value cut-offs for a false discovery rate of 0.01 were determined as 1.1 and -1.1 according to previous “yellow experiments” performed by Wolf et al. (2017a).


Analysis of the Functional Relevance of Cgt


Distribution of Single-Domain CBM-20 Proteins in the Eubacterial World


The inventors have analyzed the distribution of CBM-20 single-domain proteins in the prokaryotic world by BlastP analysis.


In brief, the distribution of singular CBM-20-domain proteins was analyzed by BlastP analyses using the NCBI non-redundant protein database (Altschul et al. 2005; Altschul et al. 1990). As CBM-20 domains occur in a variety of different proteins and enzymes, data filtering had to be performed: Of the initial 3,316 BlastP hits, all of eukaryotic origin and all enzymes with function-specific annotation or sizes above 350 amino acids were excluded. The domain structures of the remaining 80 BlastP hits were analyzed (Marchler-Bauer et al. 2017; Marchler-Bauer and Bryant 2004; Marchler-Bauer et al. 2015; Marchler-Bauer et al. 2010). Most of these, 53 proteins in total, contain two CBM-20 domains traversed by a higher domain described as glyco-hydro-77-superfamiliy 4-alpha-glucanotransferase. Ten contain different additional domains: Five of them alpha-amylase inhibitor domains, two CBM-25, respectively, CBM-26 binding domains at the N-terminus, two N-terminal domains of IPT-superfamily with probable regulatory function and one a DUF1393-domain, which was described to occur in several alpha-amylases (information taken from the NCBI database). These candidates were also excluded. Only 18 candidates (including Cgt from Actinoplanes sp. SE50/110) displayed a singular CBM-20 domain. A protein tree was created by Blast tree view 1.17.5 of the NCBI database (NCBI database) on basis of a multiple sequence alignment performed by BlastP (Altschul et al. 1990; Altschul et al. 2005).


Interestingly, singular CBM-20 domain-proteins were found in only 17 other species (FIG. 9). Most of these are found in species of the order Actinomycetales, for example in all strains of the genus Actinoplanes. The majority of the 17 species were originally isolated from soil and environmental samples, namely A. missouriensis (Parenti and Coronelli 1979), A. utahensis (described by Parenti and Coronelli (1979) and first isolated by Couch (1963)), A. teichomyceticus (Wink et al. 2006), Streptomyces sp. 94 (Chu et al. 1996), Streptomyces sp. OK885 (isolated from roots, Tennessee, USA, information taken from GenBank (Benson et al. 2013) of the NCBI (NCBI database)), Streptosporangium roseum (Nolan et al. 2010), Streptosporangium sclerotialus (syn. Chainia antibiotica) (Thirumalachar 1955), Cellulomonas sp. B6 (Piccinni et al. 2016), Paenibacillus sp. P22 (Hanak et al. 2014), and Clostridium sp. DMHC 10, which was isolated from the sludge of a distillery waste treatment plant (Kamalaskar et al. 2010). CBM-20 proteins also occur in Streptomyces sp. D1166, for which the sampling sites has not been reported, and in multi-species of the family Pseudomonadaceae. They belong to genera, which are known to include soil-inhabiting members.


Strains carrying singular CBM-20 proteins without direct connection to the habitats soil or environment occur only occasionally, like in singular isolates of the human pathogens Chlamydia trachomatis (Thomson et al. 2008) and Mycobacterium abscessus (Ryan and Byrd 2018; Moore and Frerichs 1953).


Confirmation of the Starch Binding Function by an In Vitro Assay


CBM-20 domains are described to have a starch binding function, which the inventors wanted to test by an in vitro assay. As the small carbohydrate binding protein Cgt is highly expressed and enriched in the extracellular space due to an N-terminal signal peptide (Wendler et al. 2015a), the protein could be directly concentrated from the supernatant by filtration. A starch binding assay was performed with starch from potato in different concentrations. Both—the starch fraction as well as the supernatant—were analyzed by SDS-PAGE. In all starch fractions (ranging from 1 to 10% (w/v) of starch), a protein band at about 15 kDA was detected, which was clearly identified as Cgt by MALDI-TOF-MS. In contrast, the supernatant fractions were almost completely depleted by Cgt. Residual Cgt in the supernatant was found, indicating, that the added starch was completely saturated by Cgt. In the negative control without starch, most of Cgt remains in the supernatant fraction. Beside Cgt, another small extracellular protein of unknown function, ACSP50_6253, was identified by the starch binding assay (data not shown).


Analysis of Cgt Expression During Growth on Different Carbon Sources


The gene cgt has been reported of being differentially expressed in the presence of different carbon sources, as determined by transcriptome and proteome analysis on glucose and maltose (Schwientek et al. 2013; Wendler et al. 2015a; Ortseifen 2016). The inventors have tested the effects of several carbon sources on the expression of cgt gene by measuring the transcript amounts by reverse transcription quantitative PCR (RT-qPCR). For this purpose, the wild type strain of Actinoplanes sp. SE50/110 was grown on minimal medium supplemented with maltose, glucose, starch, galactose, cellobiose, lactose and C-Pur (Cerestar 01908) (FIG. 10). The latter is a sugar-containing product from the degradation of starch mainly consisting of maltose and maltotriose. All carbon sources were supplemented in equivalent C-molar amounts. The only exception was starch: Due to the low solubility, here, a 4% (w/v) opalescent solution of “starch soluble” from Acros Organics was generated. For comparison, a maltose minimal medium with reduced amount of maltose was prepared (here: 44.40 g·L−1 maltose monohydrate), in which the C-molarity should approximate the one in the starch medium.


For most tested carbon sources, the transcription of the cgt gene was similar or just slightly and insignificantly reduced compared to a maltose grown culture (FIG. 11 A). Differential transcription was observed for galactose to a minor extent (3.4-less transcribed, log2(fold-change)=0.291). A significant reduction of cgt transcript was measured for the carbon sources glucose (142-fold less transcribed, log2(fold-change)=0.007) and lactose (62-fold less transcribed, log2(fold-change)=0.016). When cells were grown on maltose minimal medium with reduced amount of maltose (here: 44.4 g·L−1 instead of 72.06 g·L−1), a 2.9-fold decreased transcription of cgt gene was observed (log2(fold-change)=0.345) (FIG. 11 B).


Analysis of Gene Deletion Mutant ΔCgt


ΔCgt on Different Carbon Sources or Under Carbon-Limited Conditions


The differential transcription profile of cgt in dependence of the carbon source indicated a function within sugar metabolism, like it has been presumed before (Ortseifen 2016). Ortseifen (2016) (Ortseifen 2016) suggested Cgt of being responsible for the retention of carbon as energy source in the context of the carbophore model. Growth of the wild type and the CRISPR/Cas9 deletion mutant Δcgt was tested on different carbon sources in liquid culture.


Before, a pre-screening experiment was performed in the OmniLog Phenotypic Microarray System (Biolog Inc., Hayward, United States of America), which allows fast phenotypic screening by measurement of cellular respiration activity on a total of 190 different carbon sources in multi-well plates. Of these, Actinoplanes displayed respiration on 103 carbon sources.


Except for arabinose and lactose, no differential respiration profile was observed for Δcgt on the remaining 101 carbon sources. In order to validate these results on the level of growth, the carbon sources arabinose and lactose were furthermore tested in a shake flask cultivation. Also, the standard laboratory sugars maltose and glucose, and the complex carbon source starch as well as the disaccharide cellobiose were tested, to imitate natural carbon sources of the habitat soil. No restraint on growth was observed for Δcgt (FIG. 12 and FIG. 13).


Furthermore, growth under carbon-limited conditions (here: 1 g·L−1, 2 g·L−1, 3 g·L−1, 4 g·L−1 and 5 g·L−1 starch) was tested in the RoboLector®-system of m2p-labs. No growth disadvantages for the Δcgt mutant were observed in case of carbon source constraints compared to the wild type (FIG. 14).


Δcgt has No Impact on Osmolarity- or pH-Tolerance Cgt multimers have been proposed to form surface layers through multimerization (Wendler et al. 2015a). This might suggest a potential role in protection against environmental changes, like drought, pH and osmolarity.


A pH screening was performed on solid media as well as in liquid culture in the RoboLector®-system. For screening on solid media, SFM-agar plates of pH ranging from pH 4 to 11 (in steps of 1) were prepared and droplets of a dilution series of spores of the wild type and the deletion mutant Δcgt were applied. Both mutant and wild type were able to grow from pH 5 to 11. No differences in growth or spore formation on agar-plates were observed.


Since an effect of drought tolerance is difficult to assess, the inventors analyzed the colony and spore formation on the surface of the bacterial lawn and found no differences between the wild type and Δcgt.


For pH screening in liquid culture, maltose minimal medium of pH ranging from 4 to 7 was prepared. Higher pH values could not be tested in liquid culture, as medium components tend to precipitate. Both strains grew from pH 4.5 to 7 (FIG. 15). Regarding the final cell dry weights, no differences were observed.


For osmolarity screening, maltose minimal medium was prepared with different concentrations of maltose ranging from 3.6 to 108.1 g·L−1 maltose monohydrate and osmolarity ranging from 323.5 to 681.0 mOsmol·kg−1 (Table E11). No significant growth differences were observed between the wild type and the deletion mutant Δcgt (FIG. 16).


Also, inositol was tested as osmolyte, since it is not consumed by Actinoplanes. Here, osmolarity ranged from 388.5 to 695.0 mOsmol·kg−1, but no growth differences were observed (FIG. 17).


Lower osmolarities between 159-190 mOsmol·kg−1 were tested by use of the complex medium NBS (FIG. 19, Table E10). Again, no significant differences in growth were observed between the wild type strain and the deletion mutant Δcgt.









TABLE E10







Tabular summary of screening experiments. Final cell dry weights, final acarbose concentrations,


pH and osmolarity of different minimal media used in this work for screening of osmolarity and pH.


Different osmolarities in the media used for pH screening are caused by addition of correcting agents.













maltose
glucose

osmolarity

final cell dry weights
final acarbose concentration


x1H2O
x1H2O
inositol
(mOsmol ·

(g · L−1)
(g · L−1)
















(g · L−1)
(g · L−1)
(mM)
kg−1)
pH
wild type
Δcgt
wild type
Δcgt
significance (p-value)










pH Screening















72.06


629.0
4.0
 1.17 ± 0.95
 2.09 ± 0.26
not detectable

















72.06


630.0
4.5
 5.50 ± 0.66
 6.94 ± 1.07
0.27 ± 0.18
0.38 ± 0.05
*0.02216


72.06


606.0
5.0
 9.67 ± 1.28
11.34 ± 0.80
 0.47 ± 0.003
0.65 ± 0.03
*0.00136


72.06


587.0
5.5
11.04 ± 0.71
12.13 ± 0.42
0.66 ± 0.03
0.80 ± 0.02
*0.00124


72.06


569.0
6.0
12.67 ± 0.38
12.97 ± 0.47
0.97 ± 0.17
0.89 ± 0.01



72.06


569.0
6.5
13.79 ± 1.84
12.83 ± 0.76
1.14 ± 0.13
1.06 ± 0.04



72.06


563.0
7.0
13.86 ± 0.57
12.50 ± 1.53
0.94 ± 0.10
0.85 ± 0.06








Osmo-Screening 1















3.6


323.5
6.5
1.75 ± 0.38
 1.97 ± 0.28
not detectable



14.41


361.5
6.4
6.21 ± 0.19
 6.25 ± 0.10
not detectable

















36.03


420.5
6.4
13.29 ± 0.26
12.66 ± 0.41
0.33 ± 0.04
0.39 ± 0.08



57.65


485.0
6.4
14.00 ± 0.45
14.19 ± 0.30
0.49 ± 0.10
0.61 ± 0.08



72.06


531.0
6.4
14.83 ± 0.47
15.17 ± 0.38
0.58 ± 0.05
0.65 ± 0.06



86.47


605.0
6.4
14.79 ± 0.63
16.91 ± 0.77
0.67 ± 0.04
0.69 ± 0.01



108.09


681.0
6.3
16.21 ± 1.20
17.91 ± 0.33
0.56 ± 0.03
0.66 ± 0.02
*0.01432







Osmo-















20

0
388.5
6.4
11.88 ± 0.22
12.25 ± 0.25
not detectable



20

1
406.5
6.4
12.83 ± 0.47
12.75 ± 0.98
not detectable



20

80
469.5
6.4
12.75 ± 0.45
12.06 ± 0.95
not detectable



20

140
550.0
6.4
15.13 ± 0.25
13.16 ± 0.75
not detectable



20

180
580.0
6.4
14.63 ± 0.45
14.03 ± 1.15
not detectable



20

220
610.3
6.4
14.79 ± 0.44
13.13 ± 1.76
not detectable



20

280
695.0
6.3
15.33 ± 0.51
14.00 ± 1.45
not detectable








NBS-















0
10
0
190
6.9
 4.35 ± 0.22
 3.93 + 0.47
acarbose is not main










component








NBS-
















11

0
159
6.9
 4.51 + 0.22
 3.87 ± 0.27
0.38 ± 0.06
0.42 ± 0.04
 *0.006778









Δcgt Displays Improved Acarbose Formation on Maltose Minimal Medium


Although no distinct growth phenotype could be observed under the tested conditions, lack of the highly expressed Cgt protein seems to save metabolic resources of the cell, such as ATP and amino acids. These might be used for cellular growth or other anabolic processes. In the experiments, Δcgt has not displayed significant growth advantages. However, remarkably higher final acarbose concentrations were detected for the deletion mutant Δcgt compared to the wild type (Table E10). For the cultivation in complex medium, this was most striking during the growth phase (FIG. 18).


The improved acarbose-producing phenotype was validated by three independent shake flask cultivations in maltose minimal medium (FIG. 19 and Table Eli). Quantification of acarbose from the supernatant displayed an enhanced acarbose yield coefficient of the deletion mutant compared to the wild type. The differences in the final acarbose yields were significant (tested by a two-sided t-test, p-value=0.04608). Thereby, in Δcgt an increase of 8.3 to 16.6% of final acarbose concentration was reached (cf. Table E11).









TABLE E11







Tabular summary of growth experiments under acarbose producing


conditions. Final acarbose concentrations, final cell dry weights


and the number of replicates n of three independent cultivations


of Δcgt and the wild type in maltose minimal medium.











No.1
No.2
No.3















acarbose
WT
 0.76 (+/− 0.07)
 0.87 (+/− 0.04)
 0.69 (+/− 0.02)


(g · L−1)
n
5
4
3



Δcgt
 0.88 (+/− 0.05)
 0.99 (+/−0.01))
 0.74 (+/− 0.03



n
6
4
3


%

116.6 
114.2 
108.3 


cell dry weight
WT
14.72 (+/− 1.71)
15.49 (+/− 0.65)
15.30 (+/− 1.25)


(g · L−1)
n
7
5
5



Δcgt
16.12 (+/− 0.89)
14.99 (+/− 0.94)
13.85 (+/− 1.46)



n
6
5
5


%

109.5 
 96.8
 90.5









Δcgt has No Impact on the Expression of Acarbose Biosynthesis Genes

Findings that the deletion of the highly expressed gene cgt has no negative impact on growth or viability of the organism under various conditions, but yields into an enhanced acarbose producing phenotype, was surprising. Due to this and to rule out a direct impact on the regulation of acarbose biosynthesis (acb) genes, RT-qPCR of representative acb genes were performed. For this, wild type and Δcgt were grown on maltose-minimal medium and RNA was isolated from samples of the early growth phase. The relative transcript amount of the acarbose biosynthesis cluster genes acbZ, acbW, acbV, acbA, acbB, acbD and acbE were calculated for Δcgt in comparison to the wild type (FIG. 20). The gene acbV is the first of several polycistronically transcribed genes within the main operon of the acarbose biosynthesis gene cluster (Wolf et al. 2017b). The monocistronically transcribed genes acbD and acbE, encoding for proteins of the extracellular acarbose metabolism, have shown to be strongly regulated by the acarbose regulator ΔcrC (Wolf et al. 2017a). The genes acbA, acbB and acbZ are monocistronically transcribed, too, and are annotated as enzymes of the acarbose biosynthesis (acbAB) and its extracellular metabolism (acbZ), respectively. AcbW is the first gene of the acbWXY-operon, putatively encoding for an ABC transporter. For all selected transcripts, no significant change in relative transcript levels was measured in the deletion mutant Δcgt compared to the wild type (FIG. 20).


DISCUSSION

The connection of carbohydrate metabolism and acarbose biosynthesis is of high interest. Recent research has pointed out the importance of carbon utilization in the context of the biosynthesis of acarbose and further acarviosyl metabolites in the wild type (Wendler et al. 2014).


In this context, the starch binding protein Cgt is striking. It is one of the strongest expressed genes in Actinoplanes sp. SE50/110 (Schwientek et al. 2013) making up for about 8% of the whole secreted proteome (unpublished data of the inventors). Its gene product is exported into the extracellular space (Wendler et al. 2013). Excess production and export means high costs for the cell: Only for the translational process, 4 ATP are required per peptide bond (Campbell and Reece 2011; Purves 2006), i.e. not including additional costs for RNA synthesis, amino acid production, protein folding and export. The inventors therefore concluded that Cgt has a significant role in Actinoplanes sp. SE50/110 physiology. Two different functions of Cgt are proposed and analyzed herein: A role within the sugar metabolism and a role as surface protein.


Due to the starch binding domain Ortseifen (2016) (Ortseifen 2016) suggested, that Cgt might be involved in binding and retention of energy sources in the context of the carbophore model (Wehmeier 2003). Evidence was also given here by RT-qPCR, which displayed differential expression of the gene cgt in glucose-, galactose- and lactose-grown cultures compared to cultures grown on maltose, higher maltodextrins and cellobiose. This is in accordance with differential proteome analyses on the carbon sources maltose and glucose (Wendler et al. 2015a; Wendler et al. 2015b). These results indicate a carbon-dependent expression of cgt. It would be exciting to elucidate the regulatory mechanism. However, it remains to be considered that over 900 genes are putatively involved in transcriptional regulation in Actinoplanes sp. SE50/110, of which 697 are annotated as transcriptional regulators according to the annotation of Wolf et al. (2017b) (GenBank: LT827010.1).


A sugar-dependent expression of cgt might indicate a function within the utilization of maltose, higher maltodextrins and—potentially—also cellobiose. However, our studies of the deletion mutant Δcgt have not unveiled phenotypical differences regarding the carbon utilization. This was tested for a total of 105 different carbon sources, of which 103 were analyzed in the OmniLog screening system and six in liquid culture.


As the function of Cgt might be negligible under excess of carbon source but indispensable when growing under conditions with limited carbon source, the inventors have tested growth of the deletion mutant Δcgt and the wild type on minimal medium with low concentrations of starch.


Starch was chosen as carbon source, due to the starch binding activity of Cgt, which was confirmed in a starch binding assay here. Nevertheless, no growth phenotype of the mutant could be observed under limited carbon source conditions.


Another function within the sugar metabolism could consist in binding of insoluble crystalline substrates, which might lead to structural changes, that increases substrate accessibility and enhances the activity of other hydrolyzing enzymes like amylases. Such mechanisms have already been described in the soil bacteria Serratia marcescens for chitinolysis (Vaaje-Kolstad et al. 2005) and Thermobifida fusca for cellulysis (Moser et al. 2008). In the genome of Actinoplanes sp. SE50/110 several genes are encoded with putative α-glyosidic function, of which three, the α-amylases/pullulanases AcbE, AcbZ and PulA, were shown to accumulate in the extracellular space (Wendler et al. 2015a). Additionally, another small extracellular protein of unknown function and starch binding capability (ACSP50_6253) was identified in a starch binding assay. By heterologous expression of extracellular amylases and enzyme assays in presence and absence of both—Cgt and ACSP50_6253—, a supporting function during starch degradation might be detected in future experiments.


Apart from the sugar metabolism, also a function as surface layer protein is conceivable, which is supported by the fact, that Cgt forms multimers (Ortseifen 2016; Wendler et al. 2013). Wendler et al. (2015) (Wendler et al. 2015a) identified two transmembrane domains in the Cgt protein, of which one is involved in translocation by the Sec pathway as part of the leader peptide and the second is assumed to be required for multimerization. Although Cgt is not likely to be physically anchored in the membrane (Wendler et al. 2015a), Cgt proteins may remain as multimers in the mesh of the mycelium, due to the reduced fluid flow. In this context, the starch binding domain might serve also as anchor.


In the role as putative surface protein, the inventors initially assumed a protective function in the context of pH and osmolyte stress or drought. However, the screening experiments showed that the deletion of cgt gene did not lead to significant growth inhibition at different pH in liquid culture.


From the screening experiments on solid media, there was no indication, that Cgt might have a protective function in case of pH or drought.


Hints for a putative function in the context of osmoregulation were given by reverse transcription quantitative PCR of the wild type, grown on different amounts of maltose. Here, the inventors observed a 2.9-fold reduced transcription of the gene cgt, when growing on 44.4 g·L−1 maltose compared to a 72 g·L−1, which might be an effect of osmolarity. The inventors analyzed growth of the deletion mutant Δcgt in several screening experiments in liquid culture with media ranging from 159 to 681 mOsmol·kg−1. Under all tested conditions, no differences in growth and viability were observed for the deletion mutant Δcgt compared to the wild type.


As surprisingly no apparent physiological impact was observed by the deletion of cgt gene neither in utilization of different carbon sources in excess nor in limitation, neither under different pH nor osmolyte conditions, it might be possible that the function of Cgt only becomes apparent in its natural environment and in possible competition with other soil organisms. Interestingly, the inventors found similar independent singular CBM-20 domain proteins in 17 other prokaryotic species, most of which belong to the order Actinomycetales. Although rare, this at least displays a certain distribution and shows, that Cgt is not a strain-specific protein. Most of the species harboring single domain CBM-20 proteins were associated with soil habitats. Together with the fact, that cgt is highly expressed in Actinoplanes sp. SE50/110, this supports the hypothesis that proteins like Cgt fulfill a crucial function in bacteria living within this habitat. A function of Cgt could be tested in future by co-cultivations in direct contact with other microbial competitors.


While it was surprising, that Cgt turned out to be dispensable under the tested laboratory conditions, the inventors observed a positive phenotype regarding the acarbose production. An increase of the acarbose yield between 8.3 and 16.6% was achieved by deletion of cgt. Although the final product yields differ slightly between batch cultivation, the Δcgt mutant always performed significantly better. This was shown over a time period of several month (data not shown) in three independent shake flask and several micro-scale cultivations performed in maltose minimal medium. Thus, the improved production was robust over long time periods and in different cultivation settings.


We assume that this is due to metabolic burden by expression of cgt gene in the wild type, which brings relief of energy and of free resources in Δcgt. These resources are probably redirected to the acarbose biosynthesis, which is a growth-associated product. A direct regulatory effect by deletion of cgt on the expression of the acb genes was not observed.


Analysis of the Functional Relevance of Carotenoid Formation


Light-Dependent Carotenoid-Formation and Oxidative Stress Reduce Acarbose Production in Actinoplanes sp. SE50/110



Actinoplanes are known to produce a variety of soluble pigments including yellow, orange and pink pigments of the class carotenoids (Parenti and Coronelli 1979). The pigment of Actinoplanes sp. SE50/110 is orange. Its formation is intensified when cultivated exposed to light. Since the pigment was found likewise in the supernatant, it seems to be soluble in watery solutions. After cell extraction and separation by thin layer chromatography, spectral analysis display absorption maxima at 450, 475 und 505-510, which was confirmed by an absorbance scan performed during HPLC-separation. Consistent with these findings in silico reconstruction shows, that Actinoplanes sp. SE50/110 has the full genetic equipment to produce a C40-carotenoid with similarity to sioxanthin from Salinospora tropica CNB-440 (Richter et al. 2015; Wolf et al. 2017b) (FIG. 21 and Table E12).









TABLE E12







Reconstruction of the carotinoid synthesis in Actinoplanes sp. SE50/110. Two


terpene synthesis gene cluster were identified by antiSMASH analysis (Blin et al. 2017; Weber


et al. 2015), which could be assigned to the formation of a C40-carotenoid with similarity to the


sioxanthin gene cluster from Salinospora tropica CNB-440 (Richter et al. 2015) (terpene cluster


1-2). Furthermore, a camphene-like monoterpene gene cluster (terpene cluster 3), all genes of


the MEP/DOXP-pathway and a gene coding for the degradation of lycopene were identified by


BLASTP analysis (Altschul et al. 2005) and KEGG (Kanehisa et al. 2014).










locus tag
name
annotation











genes of MEP/DOXP pathway










ACSP50_7096
dxs
1-deoxy-D-xylulose-5-phosphate





synthase



ACSP50_7248
ispG
4-hydroxy-3-methylbut-2-en-1-yl





diphosphate synthase



ACSP50_7250
dxr
1-deoxy-D-xylulose-5-phosphate





reductoisomerase



ACSP50_7707
ispH
4-hydroxy-3-methylbut-2-enyl





diphosphate reductase



ACSP50_7802
ispE
4-(cytidine 5′-diphospho)-2-C-





methyl-D-erythritol kinase



ACSP50_8046
ispF
2-C-methyl-D-erythritol 2;4-





cyclodiphosphate synthase



ACSP50_8047
ispD
2-C-methyl-D-erythritol 4-





phosphate cytidylyltransferase










ACSP50_0145
merR








genes proposed for the biosynthesis of a glycosylated C40-carotenoid with similarity to sioxanthin


terpene cluster 1













homol. genes identity /





in S. tropica positives





CNB-440











ACSP50_0145
merR
MerR-/HTH-transcriptional
merR
55% /




regulator
(STROP_4437)
65%


ACSP50_0146
idi
isopentenyl-diphosphate delta-
idi
62% /




isomerase
(STROP_4438)
66%


ACSP50_0147
crtl
zeta-phytoene desaturase
crtl
76% /





(STROP_4439)
84%


ACSP50_0148
crtE/IdsA
polyprenyl synthetase
crtE
60% /





(STROP_4440)
67%


ACSP50_0149
crtB
phytoene synthase
crtB
73% /





(STROP_4441)
80%


ACSP50_0150

deoxyribodipyrimidine photo-lyase
not found



ACSP50_0151

pyridine nucleotide-disulfide
STROP_4442
58% /




oxidoreductase

70%







terpene cluster 2a











ACSP50_1631

transcriptional regulator
STROP_3252
70% /






83%


ACSP50_1632

lycopene cyclase
not found



ACSP50_1633

lycopene cyclase
not found



ACSP50_1634
fps2/crtE
polyprenyl synthetase (farnesyl
crtE
69% /




pyrophosphate synthetase 2)
(STROP_3251)
78%


ACSP50_1635

methylenetetrahydrofolate
STROP_3250
77% /




reductase (NADPH)

87%







terpene cluster 2b











ACSP50_1650

LysR-family transcriptional
STROP_1711
58% /




regulator

70%


ACSP50_1651

methyltransferase type 11
not found



ACSP50_1652

CDP-
STROP_3594
27% /




alcoholphosphatidyltransferase

39%




pgsA




ACSP50_1653
crtD
zeta-phytoene desaturase (crtl-
crtD
66% /




family)
(STROP_3248)
73%


ACSP50_1654
cruC
glycosyl transferase
cruC
63% /





(STROP_3247)
71%


ACSP50_1655
cruF
hypothetical protein (put.
cruF
64% /




membrane prot.)
(STROP_3246)
73%


ACSP50_1656

GCN5 family acetyltransferase
STROP_3245
75% /






82%


ACSP50_1657

monooxygenase
crtA
65% /





(STROP_3244)
61%


ACSP50_1658

short-chain dehydrogenase
STROP_0722
53% /






64%


ACSP50_3873
crtE-
polyprenyl synthetase
crtE
54% /



hom.

(STROP_3251)
65%







put. genes for cam text missing or illegible when filed











ACSP50_1949
eshA
transcriptional regulator (Crp/Fnr






family)




ACSP50_1950

camphene synthase




ACSP50_1951

methyltransferase (SAM-






dependent) type 11




ACSP50_1952

glycosyl-hydrolase




ACSP50_1953

oxidoreductase/aldo/ketoreductase









degradation of lycopene











ACSP50_5522
ccd
carotenoid oxygenase/carotenoid






cleavage dioxygenase (RPE65






superfamily)






text missing or illegible when filed indicates data missing or illegible when filed







The genes of the C40-carotenoid biosynthesis are organized in three gene cluster: terpene cluster 1, 2a and 2b (cf. FIG. 21 D).


In contrast to S. tropica, homologues of crtY and crtU, encoding a cyclase and a desaturase, could not be identified in Actinoplanes sp. SE50/110 (Wolf et al. 2017b). Instead, two cyclases of the CarR-domain superfamily were found in this work. They are localized in the terpene cluster 2b (FIG. 21). CarR-domain cyclases are common in fungal, archaeal and bacterial genomes (information taken from CDD-search of the NCBI (Marchler-Bauer et al. 2017)). Since the pigment of SE50/110 is orange-colored, a terminal cycling of the red-colored precursor lycopene is highly likely and might be catalyzed by one or both CarR-domain cyclases. Similar to S. tropica, the carotenoid gene cluster of SE50/110 contains a glycosyltransferase CruC (FIG. 21, Table E12). This strongly indicates for a glycosylated carotenoid, which is in accordance with the observation, that the pigment seems to have polar characteristic, since it was found in the supernatant (FIG. 22 B).


Comparative genome analysis by the software platform EDGAR 2.0 (Blom et al. 2016), display similar terpene cluster arrangements in related species of the genus Actinoplanes, whereas a different organization was found in Streptomyces (data not shown). By this, the gene arrangements found in SE50/110 and CNB-440 (Richter et al. 2015; Wolf et al. 2017b) seem to be characteristic for the family Micromonosporaceae.


Besides, genes for the synthesis of the building blocks IPP and DMAPP via the MEP/DOXP-pathway (Table E12), a gene coding for a camphene-like monoterpene synthase (terpene cluster 3, Table E12) as well as a carotenoid cleavage dioxygenase (ACSP50_5522, Table E12) were found in the genome of SE50/110. The latter two might be involved in the formation of odorous substances (Yamada et al. 2015). The inventors observed, that strong pigmentation was associated with production losses. This was confirmed by comparing growth and acarbose yields of cultures exposed to and covered from light (FIG. 22). While carotenoid formation was induced, acarbose production and growth of Actinoplanes sp. SE50/110 was strongly reduced, when exposed to bulb light (36 W, Osram 830U) with an intensity of 22-44 ρE (1ρE=μmolphotons m−2s−1). In total, a loss of 39% of the final acarbose concentration was monitored.


Deletion of merR in SE50/110 Induces Carotenoid Formation without Exposure to Light


Since natural or bulb light was able to induce carotenoid formation (FIG. 22 B, C), this study searched for possible regulatory genes in SE50/110. A MerR-regulator was found within terpene cluster 1 (ACSP50_0145, FIG. 23).


The MerR-family mainly consists of activators, which are able to respond to environmental stimuli, like oxidative stress, heavy metals or antibiotics (Brown et al. 2003). Indeed, several members of the MerR-family have been described as both light-dependent activators or repressors of the carotenoid biosynthesis in non-photosynthetic bacteria, f. e. LitR in the related actinomycete S. coelicolor (Takano et al. 2005; Takano et al. 2006), in the Gram-negative Thermus thermophiles HB27 (Takano et al. 2011) and in the Gram-positive Bacillus megaterium QM B1551 (Takano et al. 2015). Here, cobalamin (vitamin B12) acts as cofactor, which mediates light sensitivity, since it is able to absorb ultraviolet and blue light: By either binding covalently to the regulator or falling off after light excitation, it is able to modulate the conformation and activity of the regulator (van der Horst et al. 2007). The mechanisms of regulation and the binding sites are quite different: Whereas in T. thermophiles and B. megaterium the promoter regions of litR/crtB (Takano et al. 2011) or litR and crtl (Takano et al. 2015) are repressed in the dark and relieved after illumination, LitR in S. coelicolor seems to be an essential light-induced transcriptional activator of the adjacent localized litS, which encodes an ECF sigma factor and directs the transcription of the carotenoid biosynthesis genes (Takano et al. 2005). A gene encoding an ECF sigma factor does not occur within the gene cluster of SE50/110. In the Gram-negative bacterium Myxococcus xanthus a B12-dependent MerR regulator is part of a complex regulatory cascade including eight further regulatory genes (Fontes et al. 2003; Galbis-Martinez et al. 2012). Indeed, no homologues of the regulatory network from M. xanthus were identified in the genome of SE50/110 by BLASTP-analysis (data not shown).


The MerR-family regulator ACSP50_0145 of Actinoplanes sp. SE50/110 contains an N-terminal HTH-motif and a C-terminal B12-binding domain (according to BLASTP-analysis and CDD-search (Marchler-Bauer et al. 2015; Marchler-Bauer et al. 2010; Altschul et al. 2005)). The position of the HTH-domain accounts for a transcriptional repressor (Pérez-Rueda and Collado-Vides 2000).


By CRISPR/Cas9 deletion of the corresponding gene in SE50/110, the carotenoid formation was strongly induced without exposure to light (FIG. 24 B, C). This confirms a function as transcriptional repressor.


Indeed, it has to be noted, that the repressor/operator system is leaky, since the typical orange color is also produced in the wild type without exposure to light. According to this, the transcription of the genes crtEBI and idi (ACSP50_0146-0149) was only doubled in AmerR compared to the wild type under dark conditions (FIG. 24 E). These differences were significant for crtE, crtB and idi. No effects on the transcription of acb genes were observed.


However, in the context of this work, the question was examined, whether pigment formation in AmerR influences the formation of the fine-chemical acarbose. Again, higher carotenoid formation was associated with lower acarbose formation (FIG. 24 A, D). When illuminated, both wild type and AmerR are strongly pigmented and the final acarbose concentrations were similar for both strains, reaching approx. 0.52 g·L−1 (FIG. 24 B, D). This corresponds to a reduction of acarbose formation of approx. 38% compared to the wild type under dark conditions (reaching 0.83 g·L−1). This is in accordance to the previous growth experiments of the wild type as described herein.


Under dark conditions, AmerR produces approximately 15% less acarbose than the wild type (0.70 g·L−1) (FIG. 24 D). It is suggested, that these production losses are assigned to the waste of resources by carotenoid formation in the deletion mutant (FIG. 24 C). In conclusion, the production losses under light conditions (38-39%) might result from further light-induced stress in both the deletion mutant and the wild type.


Comparative transcriptome analyses of the wild type cultivated under dark and light conditions using the microarray technique, display a complex response on transcript level affecting various genes (cf. FIG. 25). Several of the differentially expressed genes indicate a cellular response to combat oxidative stress. Oxidative stress is caused by reactive oxygen species (ROS), which are formed by energy transfer (leading to singlet oxygen) or electron transfer (leading to superoxide, hydrogen peroxides and hydroxyl radicals) (Ziegelhoffer and Donohue 2009). At high concentrations, ROS are toxic and cause protein and membrane oxidation and DNA damage (Ziegelhoffer and Donohue 2009; Gout 2019).


In SE50/110, the tyrosinase MeIC (ACSP50_4950, previously: ACPL_5017), a photo-protector, which is involved in the formation of the brown pigment eumelanin (Wolf et al. 2016), and genes of the riboflavin biosynthesis (ACSP50_6437-40) are stronger transcribed when exposed to light (FIG. 25). Riboflavin is a water-soluble photo-oxidative sensitizer absorbing at 374 and 445 nm (Silva et al. 1999; Kim et al. 1993). It is the precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). These are cofactors of proteins, that are involved in cellular redox metabolism, light-sensing, DNA-repair and further functions (reviewed in Garcia-Angulo (2017)). By this, riboflavin and its derivates are important micro-nutrients, that enable the cells to overcome oxidative stress (Chen et al. 2013).


According to this, also several flavin-dependent oxygenases are stronger transcribed, when exposed to light. One of them is annotated as taurine dioxygenase, which substrate is a degradation product of cysteine. Sulfur-containing amino acids like cysteine belong to the group of low molecular weight thiols (LMW thiols), that are able to catch ROS and function as redox buffers (Gout 2019). Corresponding to this, further genes probably involved in cysteine and methionine metabolism and transport are stronger transcribed in cells exposed to light.


Remarkably, several transcriptional regulator genes and a gene encoding the sigma factor SigE (ACSP50_0558) are stronger transcribed, too (FIG. 25). SigE was associated with oxidative stress-response in the photosynthetic bacterium Rhodococcus sphaeroides (reviewed in Ziegelhoffer and Donohue (2009)) and with envelope stress response in the related species S. coelicolor (Hutchings et al. 2006) and C. glutamicum (Park et al. 2008). It might be possible, that SigE is involved in oxidative stress response in SE50/110.


Interestingly, genes of the carotenoid biosynthesis and of the regulator MerR are not significantly stronger transcribed in the wild type exposed to light compared to the wild type hidden from light. This is noteworthy, since a clear effect of light on carotenoid formation can be observed in the wild type. Since the carotenoid synthesis takes place both in the dark and in the light and the enhancement of relative transcript amounts is quite moderate in the regulator mutant (see above), the effects on transcript level might be inconspicuous. It is assumed, that further regulation of carotenoid synthesis on protein level or metabolome level might exists, f. e. by degradation of carotenoids or terpenoid-precursors by the carotenoid cleavage dioxygenase (ACSP50_5522). However, according to the results obtained from the microarray of the wild type, the crt gene expression does not seem to be a primary target of the global oxidative stress response, similar to findings from Rhodococcus sphaeroides (reviewed in Ziegelhoffer and Donohue (2009)).


Taken all together, illumination triggers oxidative stress response and seems to have an important impact on the distribution of metabolic resources towards growth, carotenoid and acarbose formation. The regulation of carotenoid biosynthesis seems to be decoupled from the global response to oxidative stress, which needs further investigation. With view to direct the metabolic fluxes towards the production of acarbose, it is desirable to gain a better understanding of these processes in future. The sigma factor SigE might be responsible for the oxidative stress response, since it is higher transcribed when exposed to light.


Apart from light stress, this work demonstrates, that a large portion of production losses can be directly assigned to the carotenoid formation. Carotenoids of non-photosynthetic bacteria are assumed to have a function as photo-protectors (Lee and Schmidt-Dannert 2002), since they have shown to protect from photodynamic killing (Mathews and Sistrom 1959). As the influence of light can be excluded by simple structural measures, carotenoid formation is assumed to be dispensable under laboratory conditions. In order to improve acarbose production, switching off the concurring carotenoid biosynthesis pathway, f. e. by deletion of the central gene crtl, can be used for strain development. Since carotenoids influence the fluidity of membranes (Gruszecki and Strzalka 2005), lack of the C40-carotenoid can also affect the surface and mycelial structure of Actinoplanes sp. SE50/110. With regard to production, a break-up of mycelial lumps is advantageous to increase the mycelial surface and the number of biochemically available cells.


Overexpression of acbB and gtaB


Expression vector pSETT4 was tested for the genes acbB and gtaB. Both genes, acbB and gtaB, are probably involved in the amino sugar synthesis, a feeding branch of acarbose biosynthesis: AcbB catalyzes the dehydration of dTDP-D-glucose to dTDP-4-keto-6-deoxy-D-glucose and GtaB is assumed to be involved in the supply of the precursor glucose-1P. Interestingly, both proteins display increased protein amounts in the cytosol of acarbose producer.


pSETT4gap and pSETT4tip Vectors for Overexpression of Single Genes


A novel cloning system was implemented, that allows easy cloning and overexpression of singular genes in Actinoplanes strains such as Actinoplanes sp. SE50/110. For this, the strong promoter of the gene gapDH from Eggerthella lenta was cloned in front of a lacZ-cassette in a pSET152-backbone. The gene lacZ is transcribed under control of the lac-promoter and flanked by the recognition side of the restriction enzyme BsaI, which enables exchange of lacZ by the gene of interest by Gibson Assembly (Gibson et al. 2009), restriction/ligation cloning or Golden Gate cloning (Engler et al. 2008). As strong expression requires strong termination, T4-terminators were introduced before and after the cloning side of the novel expression system. T4-terminators have already been successfully used in the pGUS-cloning system developed by Myronovskyi et al. (2011). Whole track RNAseq analysis of a pGUS-integration mutant performed herein showed, that the T4-terminators block transcription efficiently and prevent read-through from the integrase gene into the gene of interest. Like shown by a pre-experiment, T4-terminators do not have any side effects on the transcription of acb genes, when introduced into Actinoplanes sp. SE50/110 via pSET152-integration.


Besides, by sequencing of an enriched primary transcript library derived from the promoter-screening experiment, two putative promoters were identified behind the gene of interest in antisense orientation (FIG. 26). These two pseudo-promoters were removed in the novel expression system in order to prevent antisense transcription. Furthermore, an additional (third) T4-terminator was introduced behind the cloning side in opposite orientation to prevent further putative antisense reads.


To allow exchange of the promoter sequence, NdeI and KpnI restriction sites were introduced. In this work, the strong gapDH-promoter was exchanged by the medium-strong tipA-promoter from S. lividans. By this, it was shown, that the system can be easily modified, f. e. to adjust it for other species of the order Actinomycetales. The vectors (named pSETT4gap and pSETT4tip) were tested for strong and medium strong overexpression of the genes acbB and gtaB.


Medium Overexpression of acbB Leads to Improved Acarbose Formation


The dTDP-D-glucose-4,6-dehydratase AcbB seems to be involved in the generation of an activated amino sugar from D-glucose-1P—a feeding pathway of the acarbose biosynthesis (FIG. 1): Increased AcbB-activity was found to improve the supply of the modified precursor: In brief, two overexpression mutants were created based on expression vector pSETT4 described elsewhere herein. In these mutants acbB is transcribed under control of the medium strong tipA-promoter or the strong gapDH-promoter. As previously published in (Schaffert, et al. 2019), expression vectors using the native promoter did not lead to a significant overexpression of the genes of the Acb gene cluster. The native promoter was therefore used in both the pSET152- and the pSETT4-vector background as control. Growth and acarbose formation were monitored in two shake flask cultivations in maltose minimal medium (FIG. 27).


The mutant with acbB transcribed under control of the heterologous tipA-promoter displayed enhanced acarbose production compared to the control strains: The yield coefficient was increased to 48.6 and 51.9% compared to the empty vector control in two independent cultivations (FIG. 28). By usage of the strong gapDH-promoter, the acarbose yield coefficient was slightly increased (FIG. 28).


In pSETT4tip::acbB, the normalized peak areas of phosphorylated glucose/galactose and UDP-glucose were similar or even slightly increased compared to the empty vector control (FIG. 29).


Therefore, the supply of activated glucose moieties seems to be guaranteed. In this mutant, increased amounts of the mass m/z=545 [M-H+] were found (FIG. 29, approx. 41%). Without being bound by theory this intermediate accumulates by medium AcbB-overexpression, e.g. using pSETT4tip::acbB.


At beginning growth phase enhanced expression of acbB was observed in the expected range: Strongest overexpression was achieved by use of the gapDH-promoter (log2(fold-change)=6.54) followed by use of the tipA-promoter (log2(fold-change)=4.06) (FIG. 30). Usage of the native promoter does not lead to a significant increase of relative transcript amounts of acbB.


This was tested in both the pSET152- and pSETT4-vector background (FIG. 30). Further genes of the acb gene cluster were not significantly affected, like shown for acbA and acbV (FIG. 30).


Only exception is a slightly higher transcription abundance of acbA in pSETT4tip::acbB (log2(fold-change)=1.87).


Remarkably, the transcription profile in the linear growth phase differs from the early growth phase: Here, only a doubling of transcript amounts was reached by use of the gapDH-promoter (log2(fold-change)=2.05), whereas by use of the tipA-promoter the overexpression of acbB was maintained, but to a lesser extent (log2(fold-change)=3.33) (FIG. 30, FIG. 31).


In overexpression mutants including heterologous promoters the relative transcription of acbB decreases from 4.06- to 3.33-fold (log2(fold-change)) between the two sampling times in pSETT4tip::acbB and from 6.54- to 2.05-fold in in pSETT4gap::acbB. It is assumed, that whereas the transcription of the chromosomal acbB-copy is down-regulated in these mutants, the transcription of the vector copy is maintained by the heterologous promoters. The differences in acbB-transcription at different sampling times furthermore suggest, that the down-regulation of acb gene transcription occurs stronger respectively earlier in pSETT4gap::acbB compared to pSETT4tip::acbB. Overexpression of acbB (pSETT4gap::acbB and pSETT4tip::acbB) seems to decelerate during linear growth phase.


In summary, in particular medium overexpression of acbB by usage of the tipA-promoter seems to be beneficial for acarbose production, whereas strong overexpression by use of the gapDH-promoter seems to have only a smaller effect on acarbose formation. Further improvement in acarbose formation may be achieved by varying of the expression level of acbB, e.g. by using alternative promoters from the promoter screening or by introducing multiple gene copies.


In summary, this work demonstrates, that medium overexpression of AcbB increases the acarbose yields, possibly due to improved amino sugar supply. By medium overexpression of acbB (e.g. by use of the tipA-promoter), a positive effect on acarbose production was observed yielding into round about 50% more acarbose in two independent cultivations. Therefore, the improvement of the acarbose biosynthesis by overexpression of singular acb genes was achieved.


Medium Overexpression of gtaB Leads to Improved Acarbose Formation


GtaB is supposed to catalyze the conversion of UDP-glucose and glucose-1P into each other. It was surprisingly found that overexpression of GtaB triggers acarbose formation. Without being bound by theory this may occur by improved deployment of the precursor glucose-1P. As shown by a shake flask cultivation in maltose minimal medium (FIG. 32), the final yield coefficient for acarbose of overexpression mutants of gtaB introduced into pSETT4tip is increased to 8.56%.


Interestingly, the acarbose formation is particularly increased in the late linear to stationary growth phase. In the overexpression mutant, the relative transcript amount of the gene gtaB is 2.64-fold increased (log2(fold-change)) (FIG. 33).


Since the metabolism of activated sugars is connected or redirected to other metabolic pathways, they are not supposed to accumulate. But—like shown in previous experiments—the supply can be seriously disturbed. Analysis of the intracellular metabolome displays similar amounts of phosphorylated hexoses and/or UDP-glucose (FIG. 34). Therefore, the pool of activated C6-sugars is not significantly affected by overexpression of gtaB.


Interestingly, a significant decreased amount of the mass m/z=545 [M-H+] was found in pSETT4tip::gtaB (approx. decrease of 48%), which might correspond to dTDP-4-keto-6-deoxy-D-glucose, the proposed product of AcbB. This may indicate, that the flow through the synthesis strand is more balanced, since the accumulation of this metabolite is reduced in comparison to the empty vector control and AcbB-overexpression mutants (FIG. 34). Taken together, the introduction of a second gene copy of gtaB has a positive effect on the acarbose production, although the impact of gtaB-overexpression on the distribution of cellular goods remains unclear.


Transfer of this construct to producer strains of Actinoplanes can result in an increase of the beneficial effect, as here the demand for the precursor is higher compared to the wild type. Since strong overexpression of AcbB leads to an imbalance in glucose-phosphate-metabolism combined overexpression of acbB and gtaB would plausibly further improve acarbose production beyond the observed effect for the single overexpressions.


BIBLIOGRAPHY



  • Patent EP2601209B1.

  • Patent CN103298828B.

  • Aboagye, Samuel Yaw, et al. “Isolation of Nontuberculous Mycobacteria from the Environment of Ghanian Communities Where Buruli Ulcer Is Endemic.” Applied and environmental microbiology, 2016: 4320-4329.

  • Aceti, David J., and Wendy C. Champness. “Transcriptional Regulation of Streptomyces coelicolor Pathway-Specific Antibiotic Regulators by the absA and absB Loci.” Journal of bacteriology, 1998: 3100-3106.

  • Adams, Melanie A., and Zongchao Jia. “Structural and biochemical evidence for an enzymatic quinone redox cycle in Escherichia coli: identification of a novel quinol monooxygenase.” Journal of Biological Chemistry, 2005: 8358-8363.

  • Ahmed, E., and S. J. M. Holmström. “Siderophores in environmental research: roles and applications.” Microbial Biotechnology, 2014: 196-208.

  • Albersmeier, Andreas, Katharina Pfeifer-Sancar, Christian Rückert, and Jörn Kalinowski. “Genome-wide determination of transcription start sites reveals new insights into promoter structures in the actinomycete Corynebacterium glutamicum.” Journal of biotechnology, 2017: 99-109.

  • Almagro Armenteros, José Juan, et al. “SignalP 5.0 improves signal peptide predictions using deep neural networks.” Nature biotechnology, 2019.

  • Alonso-Casajús, Nora, et al. “Glycogen phosphorylase, the product of the glgP Gene, catalyzes glycogen breakdown by removing glucose units from the nonreducing ends in Escherichia coli.” Journal of bacteriology, 2006: 5266-5272.

  • Altschul, S. F., et al. “Gapped BLAST and PSI-BLAST: a new generation of protein database search programs.” Nucleic acids research, 1997: 3389-3402.

  • Altschul, Stephen F., et al. “Protein Database Searches Using Compositionally Adjusted Substitution Matrices.” The FEBS journal, 2005: 5101-5109.

  • Altschul, Stephen F., Warren Gish, Webb Miller, Eugene W. Myers, and David J. Lipman. “Basic local alignment search tool.” Journal of molecular biology, 1990: 403-410.

  • Arora, D. K. “Chemotaxis of Actinoplanes missouriensis Zoospores to Fungal Conidia, Chlamydospores and Sclerotia.” Microbiology (Reading, England), 1986: 1657-1663.

  • Avonce, Nelson, Alfredo Mendoza-Vargas, Enrique Morett, and Gabriel Iturriaga. “Insights on the evolution of trehalose biosynthesis.” BMC evolutionary biology, 2006: 109.

  • Azeredo, L. A. I., M. B. Lima, R. R. R. Coelho, and D. M. G. Freire. “A low-cost fermentation medium for thermophilic protease production by Streptomyces sp. 594 using feather meal and corn steep liquor.” Current microbiology, 2006: 335-339.

  • Bailey, T. L., and C. Elkan. “Fitting a mixture model by expectation maximization to discover motifs in biopolymers.” Proceedings. International Conference on Intelligent Systems for Molecular Biology, 1994: 28-36.

  • Bailey, Timothy L., et al. “MEME Suite: tools for motif discovery and searching.” Nucleic acids research, 2009: W202-8.

  • Banerjee, U. C., Y. Chisti, and M. Moo-Young. “Spectrophotometric determination of mycelial biomass.” Biotechnology Techniques, 1993: 313-316.

  • Barral, Patricia, et al. “An olive pollen protein with allergenic activity, Ole e 10, defines a novel family of carbohydrate-binding modules and is potentially implicated in pollen germination.” Biochemical Journal, 2005: 77-84.

  • Bartek, Tobias, et al. “Comparative 13C metabolic flux analysis of pyruvate dehydrogenase complex-deficient, L-valine-producing Corynebacterium glutamicum.” Applied and environmental microbiology, 2011: 6644-6652.

  • Baumgart, Meike, et al. “Construction of a prophage-free variant of Corynebacterium glutamicum ATCC 13032 for use as a platform strain for basic research and industrial biotechnology.” Applied and environmental microbiology, 2013:6006-6015.

  • Bayer A G, annual report, ed. “Bayer Annual Report.” 51368 Leverkusen, Germany, n.d.

  • Becker, Judith, Corinna Klopprogge, Andrea Herold, Oskar Zelder, Christoph J. Bolten, and Christoph Wittmann. “Metabolic flux engineering of L-lysine production in Corynebacterium glutamicum—over expression and modification of G6P dehydrogenase.” Journal of biotechnology, 2007: 99-109.

  • Becker, Judith, Corinna Klopprogge, Oskar Zelder, Elmar Heinzle, and Christoph Wittmann. “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.” Applied and environmental microbiology, 2005: 8587-8596.

  • Benson, Dennis A., et al. “GenBank.” Nucleic acids research, 2013: D36-42.

  • Bentley, S. D., et al. “Complete genome sequence of the model actinomycete Streptomyces coelicolor A3(2).” Nature, 2002:141-147.

  • Beyer, Hannes M., Patrick Gonschorek, Sophia L. Samodelov, Matthias Meier, Wilfried Weber, and Matias D. Zurbriggen. “AQUA Cloning: A Versatile and Simple Enzyme-Free Cloning Approach.” PloS one, 2015: e0137652.

  • Bibb, M. J., J. White, J. M. Ward, and G. R. Janssen. “The mRNA for the 23S rRNA methylase encoded by the ermE gene of Saccharopolyspora erythraea is translated in the absence of a conventional ribosome-binding site.” Molecular microbiology, 1994: 533-545.

  • Bibb, Mervyn J. “Regulation of secondary metabolism in streptomycetes.” Current opinion in microbiology, 2005: 208-215.

  • Bibb, Mervyn J., Gary R. Janssen, and Judy M. Ward. “Cloning and analysis of the promoter region of the erythromycin resistance gene (ermE) of Streptomyces erythraeus.” Gene, 1985: 215-226.

  • Bierman, M., R. Logan, K. O'Brien, E. T. Seno, R. N. Rao, and B. E. Schoner. “Plasmid cloning vectors for the conjugal transfer of DNA from Escherichia coli to Streptomyces spp.” Gene, 1992: 43-49.

  • Bilyk, Bohdan, and Andriy Luzhetskyy. “Unusual site-specific DNA integration into the highly active pseudo-attB of the Streptomyces albus J1074 genome.” Applied microbiology and biotechnology, 2014: 5095-5104.

  • Bischoff, H. “Pharmacology of alpha-glucosidase inhibition.” European journal of clinical investigation, 1994: 3-10.

  • Blin, Kai, et al. “antiSMASH 4.0-improvements in chemistry prediction and gene cluster boundary identification.” Nucleic acids research, 2017: W36-W41.

  • Blom, Jochen, et al. “EDGAR 2.0: an enhanced software platform for comparative gene content analyses.” Nucleic acids research, 2016: W22-8.

  • Boer, H. A., L. J. Comstock, and M. Vasser. “The tac promoter: a functional hybrid derived from the trp and lac promoters.” Proceedings of the National Academy of Sciences, 1983: 21-25.

  • Bolger, Anthony M., Marc Lohse, and Bjoem Usadel. “Trimmomatic: a flexible trimmer for Illumina sequence data.” Bioinformatics, 2014: 2114-2120.

  • Boos, Winfried, and Howard Shuman. “Maltose/Maltodextrin System of Escherichia coli: Transport, Metabolism, and Regulation.” Microbiology and molecular biology reviews: MMBR, 1998: 204-229.

  • Boraston, Alisdair B., David N. Bolam, Harry J. Gilbert, and Gideon J. Davies. “Carbohydrate-binding modules: fine-tuning polysaccharide recognition.” The Biochemical journal, 2004: 769-781.

  • Bornemann, Stephen. “α-Glucan biosynthesis and the GIgE pathway in Mycobacterium tuberculosis.” Biochemical Society transactions, 2016: 68-73.

  • Brandel, Jérémy, Nicolas Humbert, Mourad Elhabiri, Isabelle J. Schalk, Gastan L. A. Mislin, and Anne-Marie Albrecht-Gary. “Pyochelin, a siderophore of Pseudomonas aeruginosa: physicochemical characterization of the iron(III), copper(II) and zinc(II) complexes.” Dalton transactions (Cambridge, England: 2003), 2012: 2820-2834.

  • Brayer, Gary D., et al. “Subsite Mapping of the Human Pancreatic α-Amylase Active Site through Structural, Kinetic, and Mutagenesis Techniques †,‡.” Biochemistry, 2000: 4778-4791.

  • Breyer, Sandra, Katharina Effenberger-Neidnicht, Sebastian Knauer, and Rainer Schobert. “Synthesis, anticancer activity, and iron affinity of the Actinoplanes metabolite 7,8-dihydroxy-1-methylnaphtho2,3-cfuran-4,9-dione.” Bioorganic & medicinal chemistry, 2011: 1264-1267.

  • Brown, Nigel L., Jivko V. Stoyanov, Stephen P. Kidd, and Jon L. Hobman. “The MerR family of transcriptional regulators.” FEMS Microbiology Reviews, 2003: 145-163.

  • Brown-Elliott, B. A., and R. J. Wallace. “Clinical and Taxonomic Status of Pathogenic Nonpigmented or Late-Pigmenting Rapidly Growing Mycobacteria.” Clinical Microbiology Reviews, 2002: 716-746.

  • Brunkhorst, Claudia, and Erwin Schneider. “Characterization of maltose and maltotriose transport in the acarbose-producing bacterium Actinoplanes sp.” Research in microbiology, 2005: 851-857.

  • Buttner, Mark J. “Actinoplanes Swims into the Molecular Age.” Journal of bacteriology, 2017.

  • Campbell, Neil A., and Jane B. Reece. Biologie. [Nachdr.]. München: Pearson Schule, 2012.

  • -. Campbell Biologie. 1. Aufl. Vol. 4900. München: Pearson Studium ein Imprint der Pearson Education, 2011.

  • -. Campbell Biologie. München; [Erscheinungsort nicht ermittelbar]: Pearson Deutschland; Pearson Studium, 2010.

  • Carpinelli, Jorge, Reinhard Kramer, and Eduardo Agosin. “Metabolic engineering of Corynebacterium glutamicum for trehalose overproduction: role of the TreYZtrehalose biosynthetic pathway.” Applied and environmental microbiology, 2006: 1949-1955.

  • Cervantes, Marla, and Francisco J. Murillo. “Role for vitamin B(12) in light induction of gene expression in the bacterium Myxococcus xanthus.” Journal of bacteriology, 2002: 2215-2224.

  • Chandra, Govind, Keith F. Chater, and Stephen Bornemann. “Unexpected and widespread connections between bacterial glycogen and trehalose metabolism.” Microbiology (Reading, England), 2011: 1565-1572.

  • Chapon, C. “Role of the catabolite activator protein in the maltose regulon of Escherichia coli.” Journal of bacteriology, 1982: 722-729.

  • Chapon, C., and A. Kolb. “Action of CAP on the malT promoter in vitro.” Journal of bacteriology, 1983: 1135-1143.

  • Chen, Jun, Jing Shen, Christian Solem, and Peter Ruhdal Jensen. “Oxidative stress at high temperatures in Lactococcus lactis due to an insufficient supply of Riboflavin.” Applied and environmental microbiology, 2013: 6140-6147.

  • Cheng, Zhuo, Haruki Yamamoto, and Carl E. Bauer. “Cobalamin's (Vitamin B12) Surprising Function as a Photoreceptor.” Trends in biochemical sciences, 2016: 647-650.

  • Chiu, M. L., et al. “Broad spectrum thiopeptide recognition specificity of the Streptomyces lividans TipAL protein and its role in regulating gene expression.” Journal of Biological Chemistry, 1999: 20578-20586.

  • Chng, Chinping, Amy M. Lum, Jonathan A. Vroom, and Camilla M. Kao. “A key developmental regulator controls the synthesis of the antibiotic erythromycin in Saccharopolyspora erythraea.” Proceedings of the National Academy of Sciences of the United States of America, 2008: 11346-11351.

  • Choi, Byoung Taek, and Chul Soo Shin. “Reduced formation of byproduct component C in acarbose fermentation by Actinoplanes sp. CKD485-16.” Biotechnology progress, 2003: 1677-1682.

  • Choi, Woo Sik, et al. “Genetic organization of the putative salbostatin biosynthetic gene cluster including the 2-epi-5-epi-valiolone synthase gene in Streptomyces albus ATCC 21838.” Applied microbiology and biotechnology, 2008: 637-645.

  • Chu, Min, et al. “Structure of Sch 68631: A new hepatitis C virus proteinase inhibitor from Streptomyces sp.” Tetrahedron Letters, 1996: 7229-7232.

  • Clark, Karen, Ilene Karsch-Mizrachi, David J. Lipman, James Ostell, and Eric W. Sayers. “GenBank.” Nucleic acids research, 2016: D67-72.

  • Cobb, Ryan E., Yajie Wang, and Huimin Zhao. “High-efficiency multiplex genome editing of Streptomyces species using an engineered CRISPR/Cas system.” ACS synthetic biology, 2015: 723-728.

  • Cobo-Simón, Marta, and Javier Tamames. “Relating genomic characteristics to environmental preferences and ubiquity in different microbial taxa.” BMC genomics, 2017: 499.

  • Cole, Stewart T., and Olivier Raibaud. “The nucleotide sequence of the malT gene encoding the positive regulator of the Escherichia coli maltose regulon.” Gene, 1986: 201-208.

  • Combes, Patricia, Rob Till, Sally Bee, and Margaret C. M. Smith. “The Streptomyces Genome Contains Multiple Pseudo-attB Sites for the φC31-Encoded Site-Specific Recombination System.” Journal of bacteriology, 2002: 5746-5752.

  • Coronelli, C., H. PAGANI, M. R. BARDONE, and G. C. LANCINI. “PURPUROMYCIN, A NEW ANTIBIOTIC ISOLATED FROM ACTINOPLANES IANTHINOGENES N. SP.” The Journal of antibiotics, 1974: 161-168.

  • Couch, J. N. “Actinoplanes, a new genus of the Actinomycetales.” Journal of the Mitchell Society, 1950: 87-92.

  • Couch, John Nathaniel. “Some New Genera and Species of the Actinoplanaceae.” Journal of the Elisha Mitchell Scientific Society, 1963: 53-70.

  • Cross, T. “Aquatic Actinomycetes: A Critical Survey of the Occurrence, Growth and Role of Actinomycetes in Aquatic Habitats.” Journal of Applied Bacteriology, 1981: 397-423.

  • Dardonville, B., and O. Raibaud. “Characterization of malT mutants that constitutively activate the maltose regulon of Escherichia coli.” Journal of bacteriology, 1990: 1846-1852.

  • Dauvillée, David, et al. “Role of the Escherichia coli glgX Gene in Glycogen Metabolism.” Journal of bacteriology, 2005: 1465-1473.

  • Davies, Bryan W., Ryan W. Bogard, Travis S. Young, and John J. Mekalanos. “Coordinated regulation of accessory genetic elements produces cyclic di-nucleotides for V. cholerae virulence.” Cell, 2012: 358-370.

  • den Hengst, Chris D., Ngat T. Tran, Maureen J. Bibb, Govind Chandra, Brenda K. Leskiw, and Mark J. Buttner. “Genes essential for morphological development and antibiotic production in Streptomyces coelicolor are targets of BIdD during vegetative growth.” Molecular microbiology, 2010: 361-379.

  • Deutsche Diabetes Gesellschaft, and Deutsche Diabetes-Hilfe. “Deutscher Gesundheitsbericht.” Mainz, 2018.

  • diabetesDE. “Deutscher Gesundheitsbericht Diabetes 2015.” Edited by diabetesDE—Deutsche Diabetes-Hilfe, DDG—Deutsche Diabetes Gesellschaft. 2015.

  • American Diabetes Association, ed. “Diagnosis and classification of diabetes mellitus.” Vol. 37 Suppl 1. 2014.

  • Diaz-Guardamino Uribe, Paz Marta. Untersuchungen zum Einbaudes Stickstoffes in der Acarviose-Einheit der Acarbose bei Actinoplanes sp. 50/110: die Aminotransferase AcbV. Wuppertal: Bergische Universitat, 2000.

  • Dippel, Renate, Tobias Bergmiller, Alex Bohm, and Winfried Boos. “The maltodextrin system of Escherichia coli: glycogen-derived endogenous induction and osmoregulation.” Journal of bacteriology, 2005: 8332-8339.

  • Dondrup, Michael, et al. “EMMA 2-a MAGE-compliant system for the collaborative analysis and integration of microarray data.” BMC bioinformatics, 2009: 50.

  • Doroghazi, James R., and Daniel H. Buckley. “Widespread homologous recombination within and between Streptomyces species.” The ISME journal, 2010: 1136-1143.

  • Du, Deyao, Lu Wang, Yuqing Tian, Hao Liu, Huarong Tan, and Guoqing Niu. “Genome engineering and direct cloning of antibiotic gene clusters via phage φBT1 integrase-mediated site-specific recombination in Streptomyces.” Scientific reports, 2015: 8740.

  • Dubeau, Marie-Pierre, Mariana Gabriela Ghinet, Pierre-Etienne Jacques, Nancy Clermont, Carole Beaulieu, and Ryszard Brzezinski. “Cytosine deaminase as a negative selection marker for gene disruption and replacement in the genus Streptomyces and other actinobacteria.” Applied and environmental microbiology, 2009: 1211-1214.

  • Dyson, P., and H. Schrempf. “Genetic instability and DNA amplification in Streptomyces lividans 66.” Journal of bacteriology, 1987: 4796-4803.

  • Edgar, Robert C. “MUSCLE: multiple sequence alignment with high accuracy and high throughput.” Nucleic acids research, 2004: 1792-1797.

  • Elliot, M. A., M. J. Bibb, M. J. Buttner, and B. K. Leskiw. “BdD is a direct regulator of key developmental genes in Streptomyces coelicolor A3(2).” Molecular microbiology, 2001: 257-269.

  • Engler, Carola, Romy Kandzia, and Sylvestre Marillonnet. “A one pot, one step, precision cloning method with high throughput capability.” PloS one, 2008: e3647.

  • Federhen, Scott. “The NCBI Taxonomy database.” Nucleic acids research, 2012: D136-43.

  • Fernández-Moreno, M. A., J. L. Caballero, D. A. Hopwood, and F. Malpartida. “The act cluster contains regulatory and antibiotic export genes, direct targets for translational control by the bIdA tRNA gene of Streptomyces.” Cell, 1991: 769-780.

  • Fong, Jiunn C. N., and Fitnat H. Yildiz. “Interplay between cyclic AMP-cyclic AMP receptor protein and cyclic di-GMP signaling in Vibrio cholerae biofilm formation.” Journal of bacteriology, 2008: 6646-6659.

  • Fontes, Marta, Lilian Galbis-Martinez, and Francisco J. Murillo. “A novel regulatory gene for light-induced carotenoid synthesis in the bacterium Myxococcus xanthus.” Molecular microbiology, 2003: 561-571.

  • Frey, P. A. “The Leloir pathway: a mechanistic imperative for three enzymes to change the stereochemical configuration of a single carbon in galactose.” The FASEB Journal, 1996: 461-470.

  • Frommer, W., B. Junge, L. Müller, D. Schmidt, and E. Truscheit. “Neue Enzyminhibitoren aus Mikroorganismen.” Planta medica, 1979: 195-217.

  • Frommer, Werner, Walter Puls, and Delf Schmidt. Process for the production of a saccharase inhibitor. United States of America Patent U.S. Pat. No. 4,019,960. 1977.

  • Frommer, Werner, Walter Puls, Dietmar Schafer, and Delf Schmidt. Inhibitoren für Glykosidhydrolasen aus Actinomyceten. Germany Patent P 20 64 092.0. 1972.

  • Fukami-Kobayashi, Kaoru, Yoshio Tateno, and Ken Nishikawa. “Parallel evolution of ligand specificity between LacI/GaIR family repressors and periplasmic sugar-binding proteins.” Molecular biology and evolution, 2003: 267-277.

  • Galbis-Martinez, Marisa, S. Padmanabhan, Francisco J. Murillo, and Montserrat Ellas-Arnanz. “CarF mediates signaling by singlet oxygen, generated via photoexcited protoporphyrin IX, in Myxococcus xanthus light-induced carotenogenesis.” Journal of bacteriology, 2012: 1427-1436.

  • Gama-Castro, Socorro, et al. “RegulonDB version 9.0: high-level integration of gene regulation, coexpression, motif clustering and beyond.” Nucleic acids research, 2016: D133-43.

  • Garcla-Angulo, Victor Antonio. “Overlapping riboflavin supply pathways in bacteria.” Critical reviews in microbiology, 2017: 196-209.

  • Garg, Ram P., and Ronald J. Parry. “Regulation of valanimycin biosynthesis in Streptomyces viridifaciens: characterization of VIml as a Streptomyces antibiotic regulatory protein (SARP).” Microbiology (Reading, England), 2010:472-483.

  • Geng, P., G. Bai, Q. Shi, L. Zhang, Z. Gao, and Q. Zhang. “Taxonomy of the Streptomyces strain ZG0656 that produces acarviostatin alpha-amylase inhibitors and analysis of their effects on blood glucose levels in mammalian systems.” Journal of applied microbiology, 2009: 525-533.

  • Geng, Peng, Feng Qiu, Yuanyuan Zhu, and Gang Bai. “Four acarviosin-containing oligosaccharides identified from Streptomyces coelicoflavus ZG0656 are potent inhibitors of alpha-amylase.” Carbohydrate research, 2008: 882-892.

  • Giaever, H. M., O. B. Styrvold, I. Kaasen, and A. R. Strom. “Biochemical and genetic characterization of osmoregulatory trehalose synthesis in Escherichia coli.” Journal of bacteriology, 1988: 2841-2849.

  • Gibson, Daniel G., Lei Young, Ray-Yuan Chuang, J. Craig Venter, Clyde A. Hutchison, and Hamilton O. Smith. “Enzymatic assembly of DNA molecules up to several hundred kilobases.” Nature methods, 2009: 343-345.

  • Glaeser, Jens, and Gabriele Klug. “Photo-oxidative stress in Rhodobacter sphaeroides: protective role of carotenoids and expression of selected genes.” Microbiology (Reading, England), 2005: 1927-1938.

  • Goldstein, Sara, and Joseph Rabani. “Mechanism of nitrite formation by nitrate photolysis in aqueous solutions: the role of peroxynitrite, nitrogen dioxide, and hydroxyl radical.” Journal of the American Chemical Society, 2007: 10597-10601.

  • -. “Mechanism of nitrite formation by nitrate photolysis in aqueous solutions: the role of peroxynitrite, nitrogen dioxide, and hydroxyl radical.” Journal of the American Chemical Society, 2007: 10597-10601.

  • Goodfellow, M., and T. Cross. “Classification.” In The biology of the actinomycetes, by Michael Goodfellow, M. Mordarski, & S. T. Williams, 7-164. London usw.: Acad. Pr, 1984.

  • Görke, Boris, and Jörg Stülke. “Carbon catabolite repression in bacteria: many ways to make the most out of nutrients.” Nature reviews. Microbiology, 2008: 613-624.

  • Gout, Ivan. “Coenzyme A: a protective thiol in bacterial antioxidant defence.” Biochemical Society transactions, 2019: 469-476.

  • Gramajo, H. C., E. Takano, and M. J. Bibb. “Stationary-phase production of the antibiotic actinorhodin in Streptomyces coelicolor A3(2) is transcriptionally regulated.” Molecular microbiology, 1993: 837-845.

  • Gramajo, Hugo C., Eriko Takano, and Mervyn J. Bibb. “Stationary-phase production of the antibiotic actinorhodin in Streptomyces coelicolor A3(2) is transcriptionally regulated.” Molecular microbiology, 1993: 837-845.

  • Grant, Charles E., Timothy L. Bailey, and William Stafford Noble. “FIMO: scanning for occurrences of a given motif.” Bioinformatics (Oxford, England), 2011: 1017-1018.

  • Grant, S. G., J. Jessee, F. R. Bloom, and D. Hanahan. “Differential plasmid rescue from transgenic mouse DNAs into Escherichia coli methylation-restriction mutants.” Proceedings of the National Academy of Sciences, 1990: 4645-4649.

  • Gregory, M. A., R. Till, and M. C. M. Smith. “Integration Site for Streptomyces Phage BT1 and Development of Site-Specific Integrating Vectors.” Journal of bacteriology, 2003: 5320-5323.

  • Gren, Tetiana. Development and application of genetic engineering methods for Actinoplanes sp. SE50/110. 2017.

  • Gren, Tetiana, et al. “Genetic engineering in Actinoplanes sp. SE50/110—development of an intergeneric conjugation system for the introduction of actinophage-based integrative vectors.” Journal of biotechnology, 2016: 79-88.

  • Gruszecki, Wiestaw I., and Kazimierz Strzatka. “Carotenoids as modulators of lipid membrane physical properties.” Biochimica et biophysica acta, 2005: 108-115.

  • Guillén, Daniel, Sergio Sanchez, and Romina Rodriguez-Sanoja. “Carbohydrate-binding domains: multiplicity of biological roles.” Applied microbiology and biotechnology, 2010: 1241-1249.

  • Guo, X., et al. “Draft genome sequence of Streptomyces coelicoflavus ZG0656 reveals the putative biosynthetic gene cluster of acarviostatin family α-amylase inhibitors.” Letters in applied microbiology, 2012: 162-169.

  • Ha, Heon-Su, Yong-II Hwang, and Sun-Uk Choi. “Application of conjugation using phiC31 att/int system for Actinoplanes teichomyceticus, a producer of teicoplanin.” Biotechnology letters, 2008: 1233-1238.

  • Hanak, Anne M., et al. “Draft Genome Sequence of the Growth-Promoting Endophyte Paenibacillus sp. P22, Isolated from Populus.” Genome Announcements, 2014.

  • Hansmeier, Nicole, et al. “Classification of hyper-variable Corynebacterium glutamicum surface-layer proteins by sequence analyses and atomic force microscopy.” Journal of biotechnology, 2004: 177-193.

  • Hansmeier, Nicole, Tzu-Chiao Chao, Alfred Pühler, Andreas Tauch, and Jörn Kalinowski. “The cytosolic, cell surface and extracellular proteomes of the biotechnologically important soil bacterium Corynebacterium efficiens YS-314 in comparison to those of Corynebacterium glutamicum ATCC 13032.” Proteomics, 2006: 233-250.

  • Hayakawa, Masayuki, Tomohiko Tamura, and Hideo Nonomura. “Selective isolation of Actinoplanes and Dactylosporangium from soil by using γ-collidine as the chemoattractant.” Journal of Fermentation and Bioengineering, 1991: 426-432.

  • Heider, Sabine A. E., Petra Peters-Wendisch, Roman Netzer, Marit Stafnes, Trygve Brautaset, and Volker F. Wendisch. “Production and glucosylation of C50 and C 40 carotenoids by metabolically engineered Corynebacterium glutamicum.” Applied microbiology and biotechnology, 2014: 1223-1235.

  • Hemker, M., et al. “Identification, cloning, expression, and characterization of the extracellular acarbose-modifying glycosyltransferase, AcbD, from Actinoplanes sp. strain SE50.” Journal of bacteriology, 2001: 4484-4492.

  • Henke, Nadja A., Sabine A. E. Heider, Silvin Hannibal, Volker F. Wendisch, and Petra Peters-Wendisch. “Isoprenoid Pyrophosphate-Dependent Transcriptional Regulation of Carotenogenesis in Corynebacterium glutamicum.” Frontiers in microbiology, 2017: 633.

  • Higgins, Michael L. “Release of Sporangiospores by a Strain of Actinoplanes.” Journal of bacteriology, 1967: 495-498.

  • Hihara, Yukako, Masayuki Muramatsu, Kinu Nakamura, and Kintake Sonoike. “A cyanobacterial gene encoding an ortholog of Pirin is induced under stress conditions.” FEBS letters, 2004: 101-105.

  • Hilker, Rolf, et al. “ReadXplorer 2-detailed read mapping analysis and visualization from one single source.” Bioinformatics (Oxford, England), 2016: 3702-3708.

  • -. “ReadXplorer—visualization and analysis of mapped sequences.” Bioinformatics (Oxford, England), 2014: 2247-2254.

  • Hirosawa, M., M. Hoshida, M. Ishikawa, and T. Toya. “MASCOT: multiple alignment system for protein sequences based on three-way dynamic programming.” Computer applications in the biosciences: CABIOS, 1993: 161-167.

  • Hofnung, Maurice, Maxime Schwartz, and Dolph Hatfield. “Complementation studies in the maltose-A region of the Escherichia coli K12 genetic map.” Journal of molecular biology, 1971: 681-694.

  • Holger Thomas. Acarbose-Metabolismus in Actinoplanes sp. SE50/110. 2001.

  • Hollander, P. “Safety profile of acarbose, an alpha-glucosidase inhibitor.” Drugs, 1992: 47-53.

  • Horbal, Lilia, Nestor Zaburannyy, Bohdan Ostash, Sergiy Shulga, and Victor Fedorenko. “Manipulating the regulatory genes for teicoplanin production in Actinoplanes teichomyceticus.” World journal of microbiology & biotechnology, 2012: 2095-2100.

  • Horbal, Liliya, et al. “Evaluation of heterologous promoters for genetic analysis of Actinoplanes teichomyceticus—Producer of teicoplanin, drug of last defense.” Journal of biotechnology, 2013: 367-372.

  • Horton, R. M. “PCR-mediated recombination and mutagenesis. SOEing together tailor-made genes.” Molecular biotechnology, 1995: 93-99.

  • Hossain, S. A., K. Tanizawa, Y. Kazuta, and T. Fukui. “Overproduction and characterization of recombinant UDP-glucose pyrophosphorylase from Escherichia coli K-12.” Journal of biochemistry, 1994: 965-972.

  • Huang, Jianqiang, et al. “Cross-regulation among disparate antibiotic biosynthetic pathways of Streptomyces coelicolor.” Molecular microbiology, 2005: 1276-1287.

  • Hull, Travis D., et al. “Cyclic Di-GMP phosphodiesterases RmdA and RmdB are involved in regulating colony morphology and development in Streptomyces coelicolor.” Journal of bacteriology, 2012: 4642-4651.

  • Huson, Daniel H., and Celine Scornavacca. “Dendroscope 3: an interactive tool for rooted phylogenetic trees and networks.” Systematic biology, 2012: 1061-1067.

  • Hutchings, Matthew I., Hee-Jeon Hong, Emmanuelle Leibovitz, lain C. Sutcliffe, and Mark J. Buttner. “The sigma(E) cell envelope stress response of Streptomyces coelicolor is influenced by a novel lipoprotein, CseA.” Journal of bacteriology, 2006: 7222-7229.

  • International Diabetes Federation, ed. “IDF Diabetes Atlas.” 2017.

  • Ihaka, Ross, and Robert Gentleman. “R: A Language for Data Analysis and Graphics.” Journal of Computational and Graphical Statistics, 1996: 299.

  • Irla, Marta, Armin Neshat, Trygve Brautaset, Christian Rückert, Jörn Kalinowski, and Volker F. Wendisch. “Transcriptome analysis of thermophilic methylotrophic Bacillus methanolicus MGA3 using RNA-sequencing provides detailed insights into its previously uncharted transcriptional landscape.” BMC genomics, 2015: 73.

  • Iwasa, T., E. Higashide, and M. Shibata. “Studies of validamycins, new antibiotics. 3. Bioassay methods for the determination of validamycin.” The Journal of antibiotics, 1971: 114-118.

  • Iwasa, T., E. Higashide, H. Yamamoto, and M. Shibata. “Studies on validamycins, new antibiotics. II. Production and biological properties of validamycins A and B.” The Journal of antibiotics, 1971: 107-113.

  • Iwasa, T., H. Yamamoto, and M. Shibata. “Studies on validamycins, new antibiotics. I. Streptomyces hygroscopicus var. limoneus nov. var., validamycin-producing organism.” The Journal of antibiotics, 1970: 595-602.

  • Jarling, Martin, Thomas Cauvet, Matthias Grundmeier, Katharina Kuhnert, and Hermann Pape. “Isolation of mak1 from Actinoplanes missouriensis and evidence that Pep2 from Streptomyces coelicolor is a maltokinase.” Journal of basic microbiology, 2004: 360-373.

  • Kaasen, I., P. Falkenberg, O. B. Styrvold, and A. R. Strom. “Molecular cloning and physical mapping of the otsBA genes, which encode the osmoregulatory trehalose pathway of Escherichia coli: evidence that transcription is activated by katF (AppR).” Journal of bacteriology, 1992: 889-898.

  • Kabus, Armin, Tobias Georgi, Volker F. Wendisch, and Michael Bott. “Expression of the Escherichia coli pntAB genes encoding a membrane-bound transhydrogenase in Corynebacterium glutamicum improves L-lysine formation.” Applied microbiology and biotechnology, 2007: 47-53.

  • Kalscheuer, Rainer, et al. “Self-poisoning of Mycobacterium tuberculosis by targeting GIgE in an alpha-glucan pathway.” Nature chemical biology, 2010: 376-384.

  • Kamalaskar, Leena B., P. K. Dhakephalkar, K. K. Meher, and D. R. Ranade. “High biohydrogen yielding Clostridium sp. DMHC-10 isolated from sludge of distillery waste treatment plant.” International Journal of Hydrogen Energy, 2010:10639-10644.

  • Kanehisa, Minoru, Susumu Goto, Yoko Sato, Masayuki Kawashima, Miho Furumichi, and Mao Tanabe. “Data, information, knowledge and principle: back to metabolism in KEGG.” Nucleic acids research, 2014: D199-205.

  • Kawaguchi, Hideo, Miho Sasaki, Alain A. Vertés, Masayuki Inui, and Hideaki Yukawa. “Identification and functional analysis of the gene cluster for L-arabinose utilization in Corynebacterium glutamicum.” Applied and environmental microbiology, 2009: 3419-3429.

  • Kieser, Tobias, Mervyn J. Bibb, Mark J. Buttner, K. F. Chater, and D. A. Hopwood. Practical Streptomyces genetics. Norwich: John Innes Foundation, 2000.

  • Kim, H., L. J. Kirschenbaum, I. Rosenthal, and P. Riesz. “Photosensitized formation of ascorbate radicals by riboflavin: an ESR study.” Photochemistry and photobiology, 1993: 777-784.

  • Klein, Claudio, and Georg E. Schulz. “Structure of cyclodextrin glycosyltransferase refined at 2.0 Å resolution.” Journal of molecular biology, 1991: 737-750.

  • Koepff, Joachim, et al. “Fast and reliable strain characterization of Streptomyces lividans through micro-scale cultivation.” Biotechnology and bioengineering, 2017: 2011-2022.

  • Kolesnikov, Nikolay, et al. “Array Express update—simplifying data submissions.” Nucleic acids research, 2015: D1113-6.

  • Koliwer-Brandl, Hendrik, et al. “Metabolic Network for the Biosynthesis of Intra- and Extracellular α-Glucans Required for Virulence of Mycobacterium tuberculosis.” PLoS pathogens, 2016: el 005768.

  • Komatsu, Mamoru, Takuma Uchiyama, Satoshi Omura, David E. Cane, and Haruo Ikeda. “Genome-minimized Streptomyces host for the heterologous expression of secondary metabolism.” Proceedings of the National Academy of Sciences of the United States of America, 2010: 2646-2651.

  • Kona, R. P., N. Qureshi, and J. S. Pai. “Production of glucose oxidase using Aspergillus niger and corn steep liquor.” Bioresource technology, 2001: 123-126.

  • Kramida, Alexander, and Yuri Ralchenko. “NIST Atomic Spectra Database, NIST Standard Reference Database 78.” 1999.



Krause, Jens P., Tino Polen, Jung-Won Youn, Denise Emer, Bernhard J. Eikmanns, and Volker F. Wendisch. “Regulation of the malic enzyme gene malE by the transcriptional regulator MalR in Corynebacterium glutamicum.” Journal of biotechnology, 2012: 204-215.

  • Kremling, A., J. Geiselmann, D. Ropers, and H. Jong. “Understanding carbon catabolite repression in Escherichia coli using quantitative models.” Trends in microbiology, 2015: 99-109.
  • Krogh, A., B. Larsson, G. Heijne, and E. L. Sonnhammer. “Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes.” Journal of molecular biology, 2001: 567-580.
  • Kumar, A., R. A. Malloch, N. Fujita, D. A. Smillie, A. Ishihama, and R. S. Hayward. “The minus 35-recognition region of Escherichia coli sigma 70 is inessential for initiation of transcription at an “extended minus 10” promoter.” Journal of molecular biology, 1993: 406-418.
  • Kusunoki, S., and T. Ezaki. “Proposal of Mycobacterium peregrinum sp. nov., nom. rev., and elevation of Mycobacterium chelonae subsp. abscessus (Kubica et al.) to species status: Mycobacterium abscessus comb. nov.” International Journal of Systematic Bacteriology, 1992: 240-245.
  • Laios, Konstantinos, Marianna Karamanou, Zenia Saridaki, and George Androutsos. “Aretaeus of Cappadocia and the first description of diabetes.” Hormones (Athens, Greece), 2012: 109-113.
  • Lakhtakia, Ritu. “The History of Diabetes Mellitus.” Sultan Qaboos University Medical Journal, 2013: 368-370.
  • Langmead, Ben, and Steven L. Salzberg. “Fast gapped-read alignment with Bowtie 2.” Nature methods, 2012: 357-359.
  • Lawford, H. G., and J. D. Rousseau. “Corn steep liquor as a cost-effective nutrition adjunct in high-performance Zymomonas ethanol fermentations.” Applied biochemistry and biotechnology, 1997: 287-304.
  • Lee, Jin-Sook, Tran Hai, Hermann Pape, Tae-Jong Kim, and Joo-Won Suh. “Three trehalose synthetic pathways in the acarbose-producing Actinoplanes sp. SN223/29 and evidence for the TreY role in biosynthesis of component C.” Applied microbiology and biotechnology, 2008: 767-778.
  • Lee, P. C., and C. Schmidt-Dannert. “Metabolic engineering towards biotechnological production of carotenoids in microorganisms.” Applied microbiology and biotechnology, 2002: 1-11.
  • Leemhuis, Hans, Udo F. Wehmeier, and Lubbert Dijkhuizen. “Single amino acid mutations interchange the reaction specificities of cyclodextrin glycosyltransferase and the acarbose-modifying enzyme acarviosyl transferase.” Biochemistry, 2004: 13204-13213.
  • Lengeler, Joseph W., G. Drews, and Hans Günter Schlegel. Biology of the prokaryotes. Stuttgart; New York; Malden, Mass.: Thieme; Distributed in the USA by Blackwell Science, 1999.
  • Leprince, Audrey, van Passel, Mark W J, and dos Santos, Vitor A P Martins. “Streamlining genomes: toward the generation of simplified and stabilized microbial systems.” Current opinion in biotechnology, 2012: 651-658.
  • Li, Chunlin, Yi-Jen Hung, Karim Qamruddin, Aziz, Mohamed Fahmy Abdel, Herbert Stein, and Birgit Schmidt. “International noninterventional study of acarbose treatment in patients with type 2 diabetes mellitus.” Diabetes research and clinical practice, 2011: 57-64.
  • Li, Kun-tai, Jia Zhou, Sai-jin Wei, and Xin Cheng. “An optimized industrial fermentation processes for acarbose production by Actinoplanes sp. A56.” Bioresource technology, 2012: 580-583.
  • Lieberman, Michael, Allan D. Marks, and Alisa Peet. Marks' basic medical biochemistry. Fourth edition. Philadelphia: Wolters Kluwer Health/Lippincott Williams & Wilkins, 2013.
  • Liu, Gang, Keith F. Chater, Govind Chandra, Guoqing Niu, and Huarong Tan. “Molecular regulation of antibiotic biosynthesis in streptomyces.” Microbiology and molecular biology reviews: MMBR, 2013: 112-143.
  • Liu, H. W., and J. S. Thorson. “Pathways and mechanisms in the biogenesis of novel deoxysugars by bacteria.” Annual review of microbiology, 1994: 223-256.
  • Lonetto, M. A., K. L. Brown, K. E. Rudd, and M. J. Buttner. “Analysis of the Streptomyces coelicolorsigE gene reveals the existence of a subfamily of eubacterial RNA polymerase sigma factors involved in the regulation of extracytoplasmic functions.” Proceedings of the National Academy of Sciences, 1994: 7573-7577.
  • Love, Michael, Simon Anders, and Wolfgang Huber. “DESeq2.” 2017.
  • Lu, M., and N. Kleckner. “Molecular cloning and characterization of the pgm gene encoding phosphoglucomutase of Escherichia coli.” Journal of bacteriology, 1994: 5847-5851.
  • Luo, Yun, et al. “A hierarchical cascade of second messengers regulates Pseudomonas aeruginosa surface behaviors.” mBio, 2015.
  • Luo, Yunzi, Lu Zhang, Katherine W. Barton, and Huimin Zhao. “Systematic Identification of a Panel of Strong Constitutive Promoters from Streptomyces albus.” ACS synthetic biology, 2015: 1001-1010.
  • Luzhetskii, A. N., B. E. Ostash, and V. A. Fedorenko. “Interspecies conjugation of Escherichia coli-Streptomyces globisporus 1912 using integrative plasmid pSET152 and its derivatives.” Russian Journal of Genetics, 2001: 1123-1129.
  • Luzhetskyy, A., et al. “IncP plasmids are most effective in mediating conjugation between Escherichia coli and streptomycetes.” Russian Journal of Genetics, 2006: 476-481.
  • Mahmud, T., S. Lee, and H. G. Floss. “The biosynthesis of acarbose and validamycin.” Chemical record (New York, N.Y.), 2001: 300-310.
  • Mahmud, Taifo, et al. “Biosynthetic Studies on the α-Glucosidase Inhibitor Acarbose in Actinoplanes sp.: 2-epi-5-epi-Valiolone Is the Direct Precursor of the Valienamine Moiety.” Journal of the American Chemical Society, 1999: 6973-6983.
  • Makitrynskyy, Roman, et al. “Pleiotropic regulatory genes bIdA, adpA and absB are implicated in production of phosphoglycolipid antibiotic moenomycin.” Open biology, 2013: 130121.
  • Makkar, N. S., and T. Cross. “Actinoplanetes in soil and on plant litter from freshwater habitats.” Journal of Applied Bacteriology, 1982: 209-218.
  • Marchler-Bauer, Aron, and Stephen H. Bryant. “CD-Search: protein domain annotations on the fly.” Nucleic acids research, 2004: W327-31.
  • Marchler-Bauer, Aron, et al. “CDD/SPARCLE: functional classification of proteins via subfamily domain architectures.” Nucleic acids research, 2017: D200-D203.
  • -. “CDD: a Conserved Domain Database for the functional annotation of proteins.” Nucleic acids research, 2010: D225-9.
  • -. “CDD: NCBI's conserved domain database.” Nucleic acids research, 2015: D222-6.
  • Marcone, Giorgia Letizia, Elisa Binda, Marcella Reguzzoni, Luciano Gastaldo, Claudia Dalmastri, and Flavia Marinelli. “Classification of Actinoplanes sp. ATCC 33076, an actinomycete that produces the glycolipodepsipeptide antibiotic ramoplanin, as Actinoplanes ramoplaninifer sp. nov.” International journal of systematic and evolutionary microbiology, 2017: 4181-4188.
  • Markowitz, Victor M., et al. “IMG: the Integrated Microbial Genomes database and comparative analysis system.” Nucleic acids research, 2012: D115-22.
  • Mathews, M. M., and W. R. Sistrom. “Function of carotenoid pigments in non-photosynthetic bacteria.” Nature, 1959: 1892-1893.
  • Matsushima, P., M. C. Broughton, J. R. Turner, and R. H. Baltz. “Conjugal transfer of cosmid DNA from Escherichia coli to Saccharopolyspora spinosa: effects of chromosomal insertions on macrolide A83543 production.” Gene, 1994: 39-45.
  • McKenzie, Nancy L., and Justin R. Nodwell. “Phosphorylated AbsA2 negatively regulates antibiotic production in Streptomyces coelicolor through interactions with pathway-specific regulatory gene promoters.” Journal of bacteriology, 2007: 5284-5292.
  • Mertes, G. “Safety and efficacy of acarbose in the treatment of Type 2 diabetes: data from a 5-year surveillance study.” Diabetes research and clinical practice, 2001: 193-204.
  • Meyer, D., C. Schneider-Fresenius, R. Horlacher, R. Peist, and W. Boos. “Molecular characterization of glucokinase from Escherichia coli K-12.” Journal of bacteriology, 1997: 1298-1306.
  • Miah, Farzana, Maureen J. Bibb, J. Elaine Barclay, Kim C. Findlay, and Stephen Bornemann. “Developmental delay in a Streptomyces venezuelae glgE null mutant is associated with the accumulation of α-maltose 1-phosphate.” Microbiology (Reading, England), 2016: 1208-1219.
  • Mistou, Michel-Yves, lain C. Sutcliffe, and Nina M. van Sorge. “Bacterial glycobiology: rhamnose-containing cell wall polysaccharides in Gram-positive bacteria.” FEMS microbiology reviews, 2016: 464-479.
  • Moore, M., and J. B. Frerichs. “An unusual acid-fast infection of the knee with subcutaneous, abscess-like lesions of the gluteal region; report of a case with a study of the organism, Mycobacterium abscessus, n. sp.” The Journal of investigative dermatology, 1953: 133-169.
  • Morgan, Jacob L. W., Joshua T. McNamara, and Jochen Zimmer. “Mechanism of activation of bacterial cellulose synthase by cyclic-di-GMP.” Nature structural & molecular biology, 2014: 489-496.
  • Morimoto, Takuya, et al. “Enhanced recombinant protein productivity by genome reduction in Bacillus subtilis.” DNA research: an international journal for rapid publication of reports on genes and genomes, 2008: 73-81.
  • Moser, Felix, Diana Irwin, Shaolin Chen, and David B. Wilson. “Regulation and characterization of Thermobifida fusca carbohydrate-binding module proteins E7 and E8.” Biotechnology and bioengineering, 2008: 1066-1077.
  • Mouri, Yoshihiro, Kenji Konishi, Azusa Fujita, Takeaki Tezuka, and Yasuo Ohnishi. “Regulation of Sporangium Formation by BIdD in the Rare Actinomycete Actinoplanes missouriensis.” Journal of bacteriology, 2017.
  • Murakami, T., T. G. Holt, and C. J. Thompson. “Thiostrepton-induced gene expression in Streptomyces lividans.” Journal of bacteriology, 1989: 1459-1466.
  • Muth, G., W. Wohlleben, and A. Pühler. “The minimal replicon of the Streptomyces ghanaensis plasmid pSG5 identified by subcloning and Tn5 mutagenesis.” Molecular & general genetics: MGG, 1988: 424-429.
  • Myronovskyi, Maksym, and Andriy Luzhetskyy. “Native and engineered promoters in natural product discovery.” Natural product reports, 2016: 1006-1019.
  • Myronovskyi, Maksym, Elisabeth Welle, Viktor Fedorenko, and Andriy Luzhetskyy. “Beta-glucuronidase as a sensitive and versatile reporter in actinomycetes.” Applied and environmental microbiology, 2011: 5370-5383.
  • Myronovskyy, M., B. Ostash, I. Ostash, and V. Fedorenko. “A gene cloning system for the siomycin producer Streptomyces sioyaensis NRRL-B5408.” Folia microbiologica, 2009: 91-96.
  • NCBI database. National Center for Biotechnology Information (NCBI) [Internet]. Edited by Bethesda (Md.): National Library of Medicine (US), National Center for Biotechnology Information. 1988. https://www.ncbi.nlm.nih.gov/(accessed cited April 2019).
  • Nguyen, J., F. Francou, M. J. Virolle, and M. Guerineau. “Amylase and chitinase genes in Streptomyces lividans are regulated by reg1, a pleiotropic regulatory gene.” Journal of bacteriology, 1997: 6383-6390.
  • Nguyen, Jacqueline. “The regulatory protein Reg1 of Streptomyces lividans binds the promoter region of several genes repressed by glucose.” FEMS Microbiology Letters, 1999: 51-58.
  • Nolan, Matt, et al. “Complete genome sequence of Streptosporangium roseum type strain (NI 9100).” Standards in genomic sciences, 2010: 29-37.
  • Nonomura, Hideo, and Shigemitsu Takagi. “Distribution of Actinoplanetes in Soils of Japan.” Journal of fermentation technology, 1977: 423-428.
  • Ogawa, Seiichiro, Takao Ogawa, Yoshikazu Iwasawa, Tatsushi Toyokuni, Noritaka Chida, and Tetsuo Suami. “Synthetic studies on the validamycins. 10. Total synthesis of DL-validoxylamines A and B.” The Journal of Organic Chemistry, 1984: 2594-2599.
  • Ortseifen, Vera. Genombasierte Modellbildung zur Biosynthese von Acarviostatin-Metaboliten in drei Actinoplanes sp. SE50/110-Stammen. Bielefeld, 2016.
  • Ortseifen, Vera, et al. “Complete genome sequence of the actinobacterium Streptomyces glaucescens GLA.O (DSM 40922) consisting of a linear chromosome and one linear plasmid.” Journal of biotechnology, 2015: 81-83.
  • Ostash, Bohdan, et al. “Complete characterization of the seventeen step moenomycin biosynthetic pathway.” Biochemistry, 2009: 8830-8841.
  • Parenti, F., and C. Coronelli. “Members of the genus Actinoplanes and their antibiotics.” Annual review of microbiology, 1979: 389-411.
  • Parenti, F., H. PAGANI, and G. Beretta. “Lipiarmycin, a new antibiotic from Actinoplanes. I. Description of the producer strain and fermentation studies.” The Journal of antibiotics, 1975: 247-252.
  • Park, Jong-Tae, et al. “Role of maltose enzymes in glycogen synthesis by Escherichia coli.” Journal of bacteriology, 2011:2517-2526.
  • Park, S. M., A. J. Sinskey, and Gregory Stephanopoulos. “Metabolic and physiological studies of Corynebacterium glutamicum mutants.” Biotechnology and Bioengineering, 1997: 864-879.
  • Park, S. Y., H. K. Kim, S. K. Yoo, T. K. Oh, and J. K. Lee. “Characterization of glk, a gene coding for glucose kinase of Corynebacterium glutamicum.” FEMS Microbiology Letters, 2000: 209-215.
  • Park, Soo-Dong, Jung-Won Youn, Young-Joon Kim, Seok-Myung Lee, Younhee Kim, and Heung-Shick Lee. “Corynebacterium glutamicum sigmaE is involved in responses to cell surface stresses and its activity is controlled by the anti-sigma factor CseE.” Microbiology (Reading, England), 2008: 915-923.
  • Pérez-Marin, Mari Cruz, S. Padmanabhan, Marla Carmen Polanco, Francisco José Murillo, and Montserrat Ellas-Arnanz. “Vitamin B12 partners the CarH repressor to downregulate a photoinducible promoter in Myxococcus xanthus.” Molecular microbiology, 2008: 804-819.
  • Pérez-Rueda, E., and J. Collado-Vides. “The repertoire of DNA-binding transcriptional regulators in Escherichia coli K-12.” Nucleic acids research, 2000: 1838-1847.
  • Pérez-Rueda, Ernesto, and Julio Collado-Vides. “The repertoire of DNA-binding transcriptional regulators in Escherichia coli K-12.” Nucleic acids research, 2000: 1838-1847.
  • Perkins, David N., Darryl J. C. Pappin, David M. Creasy, and John S. Cottrell. “Probability-based protein identification by searching sequence databases using mass spectrometry data.” Electrophoresis, 1999: 3551-3567.
  • Pesavento, Christina, and Regine Hengge. “Bacterial nucleotide-based second messengers.” Current opinion in microbiology, 2009: 170-176.
  • Petersen, Thomas Nordahl, Soren Brunak, Gunnar Heijne, and Henrik Nielsen. “SignalP 4.0: discriminating signal peptides from transmembrane regions.” Nature methods, 2011: 785-786.
  • Pfeifer-Sancar, Katharina, Almut Mentz, Christian Rückert, and Jörn Kalinowski. “Comprehensive analysis of the Corynebacterium glutamicum transcriptome using an improved RNAseq technique.” BMC genomics, 2013: 888.
  • Piccinni, Florencia, Yanina Murua, Silvina Ghio, Paola Talia, Maximo Rivarola, and Eleonora Campos. “Draft Genome Sequence of Cellulolytic and Xylanolytic Cellulomonas sp. Strain B6 Isolated from Subtropical Forest Soil.” Genome Announcements, 2016.
  • Piepersberg, W. “Pathway engineering in secondary metabolite-producing actinomycetes.” Critical reviews in biotechnology, 1994: 251-285.
  • Pósfai, György, et al. “Emergent properties of reduced-genome Escherichia coli.” Science (New York, N.Y.), 2006: 1044-1046.
  • Prágai, Zoltán, and Colin R. Harwood. “Regulatory interactions between the Pho and sigma(B)-dependent general stress regulons of Bacillus subtilis.” Microbiology (Reading, England), 2002: 1593-1602.
  • Preiss, Jack. Bacterial Glycogen Inclusions: Enzymology and Regulation of Synthesis. Vol. 1, in Inclusions in Prokaryotes, by Jessup M. Shively, 71-108. Berlin, Heidelberg: Springer-Verlag Berlin Heidelberg, 2006.
  • Purves, William K. Biologie. 7. Aufl. München: Elsevier Spektrum Akad. Verl., 2006.
  • Raibaud, O., D. Vidal-Ingigliardi, and E. Richet. “A complex nucleoprotein structure involved in activation of transcription of two divergent Escherichia coli promoters.” Journal of molecular biology, 1989: 471-485.
  • Ravcheev, Dmitry A., et al. “Comparative genomics and evolution of regulons of the LacI-family transcription factors.” Frontiers in microbiology, 2014: 294.
  • Reese, M. G. “Application of a time-delay neural network to promoter annotation in the Drosophila melanogaster genome.” Computers & chemistry, 2001: 51-56.
  • Richet, E., and O. Raibaud. “Purification and properties of the MalT protein, the transcription activator of the Escherichia coli maltose regulon.” Journal of Biological Chemistry, 1987: 12647-12653.
  • -. “MalT, the regulatory protein of the Escherichia coli maltose system, is an ATP-dependent transcriptional activator.” The EMBO Journal, 1989: 981-987.
  • Richter, Taylor K. S., Chambers C. Hughes, and Bradley S. Moore. “Sioxanthin, a novel glycosylated carotenoid, reveals an unusual subclustered biosynthetic pathway.” Environmental microbiology, 2015: 2158-2171.
  • Rocha, Eduardo P. C. “Is there a role for replication fork asymmetry in the distribution of genes in bacterial genomes?” Trends in microbiology, 2002: 393-395.
  • Rockser, Yvonne, and Udo F. Wehmeier. “The gac-gene cluster for the production of acarbose from Streptomyces glaucescens GLA.O: identification, isolation and characterization.” Journal of biotechnology, 2009: 114-123.
  • Rodriguez-Garcla, Antonio, Patricia Combes, Rosario Pérez-Redondo, Matthew C. A. Smith, and Margaret C. M.
  • Smith. “Natural and synthetic tetracycline-inducible promoters for use in the antibiotic-producing bacteria Streptomyces.” Nucleic acids research, 2005: e87.
  • Rodriguez-Sanoja, Romina, Norma Oviedo, and Sergio Sanchez. “Microbial starch-binding domain.” Current opinion in microbiology, 2005: 260-267.
  • Römling, Ute, Michael Y. Galperin, and Mark Gomelsky. “Cyclic di-GMP: the first 25 years of a universal bacterial second messenger.” Microbiology and molecular biology reviews: MMBR, 2013: 1-52.
  • Rosak, Christoph, and Gabriele Mertes. “Critical evaluation of the role of acarbose in the treatment of diabetes: patient considerations.” Diabetes, metabolic syndrome and obesity: targets and therapy, 2012: 357-367.
  • Ross, P., et al. “Regulation of cellulose synthesis in Acetobacter xylinum by cyclic diguanylic acid.” Nature, 1987: 279-281.
  • Ryan, Keenan, and Thomas F. Byrd. “Mycobacterium abscessus: Shapeshifter of the Mycobacterial World.” Frontiers in microbiology, 2018: 2642.
  • Ryding, N. Jamie, Todd B. Anderson, and Wendy C. Champness. “Regulation of the Streptomyces coelicolor calcium-dependent antibiotic by absA, encoding a cluster-linked two-component system.” Journal of bacteriology, 2002: 794-805.
  • Sauer, Uwe, Fabrizio Canonaco, Sylvia Heri, Annik Perrenoud, and Eliane Fischer. “The soluble and membrane-bound transhydrogenases UdhA and PntAB have divergent functions in NADPH metabolism of Escherichia coli.” The Journal of biological chemistry, 2004: 6613-6619.
  • Schäffer, Christina, Michael Graninger, and Paul Messner. “Prokaryotic glycosylation.” Proteomics, 2001: 248-261.
  • Schaffert, Lena, et al. “Evaluation of vector systems and promoters for overexpression of the acarbose biosynthesis gene acbC in Actinoplanes sp. SE50/110.” Microbial cell factories, 2019: 114.
  • Schlösser, Andreas, Arnim Weber, and Hildgund Schrempf. “Synthesis of the Streptomyces lividans maltodextrin ABC transporter depends on the presence of the regulator MaIR.” FEMS Microbiology Letters, 2001: 77-83.
  • Schneider, D., C. J. Bruton, and K. F. Chater. “Duplicated gene clusters suggest an interplay of glycogen and trehalose metabolism during sequential stages of aerial mycelium development in Streptomyces coelicolor A3(2).” Molecular & general genetics: MGG, 2000: 543-553.
  • Schönert, S., T. Buder, and M. K. Dahl. “Properties of maltose-inducible alpha-glucosidase MalL (sucrase-isomaltase-maltase) in Bacillus subtilis: evidence for its contribution to maltodextrin utilization.” Research in microbiology, 1999: 167-177.
  • Schönert, Stefan. Maltose-und Maltodextrin-Verwertung in Bacillus subtilis. Konstanz, 2004.
  • Schönert, Stefan, et al. “Maltose and maltodextrin utilization by Bacillus subtilis.” Journal of bacteriology, 2006: 3911-3922.
  • Schönert, Stefan, Thomas Buder, and Michael K. Dahl. “Identification and Enzymatic Characterization of the Maltose-Inducible α-Glucosidase MalL (Sucrase-Isomaltase-Maltase) of Bacillus subtilis.” Journal of bacteriology, 1998: 2574-2578.
  • Schumacher, Maria A., Wenjie Zeng, Kim C. Findlay, Mark J. Buttner, Richard G. Brennan, and Natalia Tschowri. “The Streptomyces master regulator BdD binds c-di-GMP sequentially to create a functional BIdD2-(c-di-GMP)4 complex.” Nucleic acids research, 2017: 6923-6933.
  • Schwartz, M. “Expression phenotypique et localisation genetique de mutations affectant le metabolisme du maltose chez Escherichia coli K 12.” Annales de l'Institut Pasteur, 1967: 673-698.
  • Schwientek, Patrick. Genomics and Transcriptomics of the Industrial Acarbose Producer Actinoplanes sp. SE50/110. Bielefeld, 2012.
  • Schwientek, Patrick, et al. “The complete genome sequence of the acarbose producer Actinoplanes sp. SE50/110.” BMC genomics, 2012: 112.
  • -. “Comparative RNA-sequencing of the acarbose producer Actinoplanes sp. SE50/110 cultivated in different growth media.” Journal of biotechnology, 2013: 166-177.
  • -. “Improving the genome annotation of the acarbose producer Actinoplanes sp. SE50/110 by sequencing enriched 5′-ends of primary transcripts.” Journal of biotechnology, 2014: 85-95.
  • Seibold, Gerd. Charakterisierung des Glycogen-und Maltosestoffwechsels von Corynebacterium glutamicum. Ulm, 2008.
  • Seibold, Gerd M., and Bernhard J. Eikmanns. “Inactivation of the phosphoglucomutase gene pgm in Corynebacterium glutamicum affects cell shape and glycogen metabolism.” Bioscience reports, 2013.
  • -. “The glgX gene product of Corynebacterium glutamicum is required for glycogen degradation and for fast adaptation to hyperosmotic stress.” Microbiology (Reading, England), 2007: 2212-2220.
  • Seibold, Gerd M., et al. “The glgB-encoded glycogen branching enzyme is essential for glycogen accumulation in Corynebacterium glutamicum.” Microbiology (Reading, England), 2011: 3243-3251.
  • Seibold, Gerd M., Martin Wurst, and Bernhard J. Eikmanns. “Roles of maltodextrin and glycogen phosphorylases in maltose utilization and glycogen metabolism in Corynebacterium glutamicum.” Microbiology (Reading, England), 2009: 347-358.
  • Seibold, Gerd, Stefan Dempf, Joy Schreiner, and Bernhard J. Eikmanns. “Glycogen formation in Corynebacterium glutamicum and role of ADP-glucose pyrophosphorylase.” Microbiology (Reading, England), 2007: 1275-1285.
  • Sekurova, Olga N., et al. “In vivo analysis of the regulatory genes in the nystatin biosynthetic gene cluster of Streptomyces noursei ATCC 11455 reveals their differential control over antibiotic biosynthesis.” Journal of bacteriology, 2004: 1345-1354.
  • -. “In vivo analysis of the regulatory genes in the nystatin biosynthetic gene cluster of Streptomyces noursei ATCC 11455 reveals their differential control over antibiotic biosynthesis.” Journal of bacteriology, 2004: 1345-1354.
  • Seshasayee, Aswin S. N., Gillian M. Fraser, and Nicholas M. Luscombe. “Comparative genomics of cyclic-di-GMP signalling in bacteria: post-translational regulation and catalytic activity.” Nucleic acids research, 2010: 5970-5981.
  • Shachrai, Irit, Alon Zaslaver, Uri Alon, and Erez Dekel. “Cost of unneeded proteins in E. coli is reduced after several generations in exponential growth.” Molecular cell, 2010: 758-767.
  • Shao, Zengyi, Guodong Rao, Chun Li, Zhanar Abil, Yunzi Luo, and Huimin Zhao. “Refactoring the silent spectinabilin gene cluster using a plug-and-play scaffold.” ACS synthetic biology, 2013: 662-669.
  • Sharma, Priyanka, and Bijender Kumar Bajaj. “Cost-effective-substrates for production of poly-beta-hydroxybutyrate by a newly isolated Bacillus cereus PS-10.” Journal of environmental biology/Academy of Environmental Biology, India, 2015: 1297-1304.
  • Sheeler, Nancy L., Susan V. MacMillan, and Justin R. Nodwell. “Biochemical activities of the absA two-component system of Streptomyces coelicolor.” Journal of bacteriology, 2005: 687-696.
  • Shim, Jae-Hoon, et al. “Role of maltogenic amylase and pullulanase in maltodextrin and glycogen metabolism of Bacillus subtilis 168.” Journal of bacteriology, 2009: 4835-4844.
  • Shirling, E. B., and D. Gottlieb. “Cooperative Description of Type Strains of Streptomyces: V. Additional Descriptions.” International Journal of Systematic Bacteriology, 1972: 265-394.
  • Shoseyov, Oded, Ziv Shani, and Ilan Levy. “Carbohydrate binding modules: biochemical properties and novel applications.” Microbiology and molecular biology reviews: MMBR, 2006: 283-295.
  • Shu, Xiaokun, Nathan C. Shaner, Corinne A. Yarbrough, Roger Y. Tsien, and S. James Remington. “Novel chromophores and buried charges control color in mFruits.” Biochemistry, 2006: 9639-9647.
  • Siegl, Theresa, Bogdan Tokovenko, Maksym Myronovskyi, and Andriy Luzhetskyy. “Design, construction and characterisation of a synthetic promoter library for fine-tuned gene expression in actinomycetes.” Metabolic engineering, 2013: 98-106.
  • Silva, E., A. M. Edwards, and D. Pacheco. “Visible light-induced photooxidation of glucose sensitized by riboflavin.” The Journal of nutritional biochemistry, 1999: 181-185.
  • Smet, K. A., A. Weston, I. N. Brown, D. B. Young, and B. D. Robertson. “Three pathways for trehalose biosynthesis in mycobacteria.” Microbiology (Reading, England), 2000: 199-208.
  • Sohoni, Sujata Vijay, Alessandro Fazio, Christopher T. Workman, Ivan Mijakovic, and Anna Eliasson Lantz. “Synthetic promoter library for modulation of actinorhodin production in Streptomyces coelicolor A3(2).” PloS one, 2014: e99701.
  • Song, Kyung-Mo, et al. “Transglycosylation properties of maltodextrin glucosidase (MaIZ) from Escherichia coli and its application for synthesis of a nigerose-containing oligosaccharide.” Biochemical and biophysical research communications, 2010: 87-92.
  • Soo, Po-Chi, et al. “Pirin Regulates Pyruvate Catabolism by Interacting with the Pyruvate Dehydrogenase E1 Subunit and Modulating Pyruvate Dehydrogenase Activity t.” Journal of bacteriology, 2006: 109-118.
  • Statista, ed. “Statista.” 2016.
  • Stratmann, A., T. Mahmud, S. Lee, J. Distler, H. G. Floss, and W. Piepersberg. “The AcbC Protein from Actinoplanes Species Is a C7-cyclitol Synthase Related to 3-Dehydroquinate Synthases and Is Involved in the Biosynthesis of the-Glucosidase Inhibitor Acarbose.” Journal of Biological Chemistry, 1999: 10889-10896.
  • Sung, Woo Sang, In-Seon Lee, and Dong Gun Lee. “Damage to the cytoplasmic membrane and cell death caused by lycopene in Candida albicans.” Journal of microbiology and biotechnology, 2007: 1797-1804.
  • Takano, Hideaki, et al. “Involvement of CarA/LitR and CRP/FNR family transcriptional regulators in light-induced carotenoid production in Thermus thermophilus.” Journal of bacteriology, 2011: 2451-2459.
  • -. “Role and Function of LitR, an Adenosyl B12-Bound Light-Sensitive Regulator of Bacillus megaterium QM B1551, in Regulation of Carotenoid Production.” Journal of bacteriology, 2015: 2301-2315.
  • Takano, Hideaki, Saemi Obitsu, Teruhiko Beppu, and Kenji Ueda. “Light-induced carotenogenesis in Streptomyces coelicolor A3(2): identification of an extracytoplasmic function sigma factor that directs photodependent transcription of the carotenoid biosynthesis gene cluster.” Journal of bacteriology, 2005: 1825-1832.
  • Takano, Hideaki, Teruhiko Beppu, and Kenji Ueda. “The CarA/LitR-family transcriptional regulator: Its possible role as a photosensor and wide distribution in non-phototrophic bacteria.” Bioscience, biotechnology, and biochemistry, 2006: 2320-2324.
  • Tao, Fei, Ya-Wen He, Dong-Hui Wu, Sanjay Swarup, and Lian-Hui Zhang. “The cyclic nucleotide monophosphate domain of Xanthomonas campestris global regulator CIp defines a new class of cyclic di-GMP effectors.” Journal of bacteriology, 2010: 1020-1029.
  • Tapio, S., F. Yeh, H. A. Shuman, and W. Boos. “The malZ gene of Escherichia coli, a member of the maltose regulon, encodes a maltodextrin glucosidase.” Journal of Biological Chemistry, 1991: 19450-19458.
  • Tauch, A., O. Kirchner, L. Wehmeier, J. Kalinowski, and A. Pühler. “Corynebacterium glutamicum DNA is subjected to methylation-restriction in Escherichia coli.” FEMS Microbiology Letters, 1994: 343-347.
  • Taurino, Carlo, Luca Frattini, Giorgia Letizia Marcone, Luciano Gastaldo, and Flavia Marinelli. “Actinoplanes teichomyceticus ATCC 31121 as a cell factory for producing teicoplanin.” Microbial cell factories, 2011: 82.
  • te Poele, Evelien M, Henk Bolhuis, and Lubbert Dijkhuizen. “Actinomycete integrative and conjugative elements.” Antonie van Leeuwenhoek, 2008: 127-143.
  • Terns, Rebecca M., and Michael P. Terns. “CRISPR-based technologies: prokaryotic defense weapons repurposed.” Trends in genetics: TIG, 2014: 111-118.
  • Thirumalachar, M. J. “Chainia, a New Genus of the Actinomycetales.” Nature, 1955: 934 EP.
  • Thoma, Lina, Bernd Vollmer, and Gunther Muth. “Fluorescence microscopy of Streptomyces conjugation suggests DNA-transfer at the lateral walls and reveals the spreading of the plasmid in the recipient mycelium.” Environmental microbiology, 2016: 598-608.
  • Thomas, Holger. Acarbose-Metabolisums in Actinoplanes sp. SE50/110. PhD thesis, Wuppertal: University Wuppertal, 2001.
  • Thompson, C. J., T. Kieser, J. M. Ward, and D. A. Hopwood. “Physical analysis of antibiotic-resistance genes from Streptomyces and their use in vector construction.” Gene, 1982: 51-62.
  • -. “Physical analysis of antibiotic-resistance genes from Streptomyces and their use in vector construction.” Gene, 1982: 51-62.
  • Thomson, Nicholas R., et al. “Chlamydia trachomatis: genome sequence analysis of lymphogranuloma venereum isolates.” Genome research, 2008: 161-171.
  • Tran, Ngat T., Chris D. den Hengst, Juan Pablo Gomez-Escribano, and Mark J. Buttner. “Identification and characterization of CdgB, a diguanylate cyclase involved in developmental processes in Streptomyces coelicolor.” Journal of bacteriology, 2011: 3100-3108.
  • Truscheit, Ernst, Werner Frommer, Bodo Junge, Lutz Müller, Delf D. Schmidt, and Winfried Wingender. “Chemistry and Biochemistry of Microbiala-Glucosidase Inhibitors.” Angewandte Chemie International Edition in English, 1981: 744-761.
  • Tschowri, Natalia. “Cyclic Dinucleotide-Controlled Regulatory Pathways in Streptomyces Species.” Journal of bacteriology, 2016: 47-54.
  • Tschowri, Natalia, Susan Busse, and Regine Hengge. “The BLUF-EAL protein YcgF acts as a direct anti-repressor in a blue-light response of Escherichia coli.” Genes & development, 2009: 522-534.
  • Tzvetkov, Mladen, Corinna Klopprogge, Oskar Zelder, and Wolfgang Liebl. “Genetic dissection of trehalose biosynthesis in Corynebacterium glutamicum: inactivation of trehalose production leads to impaired growth and an altered cell wall lipid composition.” Microbiology (Reading, England), 2003: 1659-1673.
  • Uguru, Gabriel C., Karen E. Stephens, Jonathan A. Stead, Jane E. Towle, Simon Baumberg, and Kenneth J. McDowall. “Transcriptional activation of the pathway-specific regulator of the actinorhodin biosynthetic genes in Streptomyces coelicolor.” Molecular microbiology, 2005: 131-150.
  • Unthan, Simon, et al. “Chassis organism from Corynebacterium glutamicum—a top-down approach to identify and delete irrelevant gene clusters.” Biotechnology journal, 2015: 290-301.
  • V. Solovyev, A. Salamov. “Automatic Annotation of Microbial Genomes and Metagenomic Sequences.” In Metagenomics and its Applications in Agriculture, Biomedicine and Environmental Studies, by Nova Science Publishers, p. 61-78. 2011.
  • Vaaje-Kolstad, Gustav, Svein J. Horn, Daan M. F. van Aalten, Bjørnar Synstad, and Vincent G. H. Eijsink. “The non-catalytic chitin-binding protein CBP21 from Serratia marcescens is essential for chitin degradation.” Journal of Biological Chemistry, 2005: 28492-28497.
  • van der Horst, Michael A., Jason Key, and Klaas J. Hellingwerf. “Photosensing in chemotrophic, non-phototrophic bacteria: let there be light sensing too.” Trends in microbiology, 2007: 554-562.
  • van Wezel, G. P., J. White, M. J. Bibb, and P. W. Postma. “The maIEFG gene cluster of Streptomyces coelicolor A3(2): characterization, disruption and transcriptional analysis.” Molecular & general genetics: MGG, 1997: 604-608.
  • van Wezel, G. P., J. White, P. Young, P. W. Postma, and M. J. Bibb. “Substrate induction and glucose repression of maltose utilization by Streptomyces coelicolor A3(2) is controlled by malR, a member of the lacI-galR family of regulatory genes.” Molecular microbiology, 1997: 537-549.
  • Vašicová, Pavla, Miroslav Pátek, Jan Nešvera, Hermann Sahm, and Bernhard Eikmanns. “Analysis of the Corynebacterium glutamicum dapA Promoter.” Journal of bacteriology, 1999: 6188-6191.
  • Voeykova, T., L. Emelyanova, V. Tabakov, and N. Mkrtumyan. “Transfer of plasmid pTO1 from Escherichia coli to various representatives of the order Actinomycetales by intergeneric conjugation.” FEMS Microbiology Letters, 1998: 47-52.
  • Wagman, G. H., et al. “New Polyene Antifungal Antibiotic Produced by a Species of Actinoplanes.” Antimicrobial agents and chemotherapy, 1975: 457-461.
  • Walter, Frederik, Sebastian Grenz, Vera Ortseifen, Marcus Persicke, and Jörn Kalinowski. “Corynebacterium glutamicum ggtB encodes a functional γ-glutamyl transpeptidase with γ-glutamyl dipeptide synthetic and hydrolytic activity.” Journal of biotechnology, 2016: 99-109.
  • Wang, J. C. “Helical repeat of DNA in solution.” Proceedings of the National Academy of Sciences, 1979: 200-203.
  • Wang, Weishan, Xiao Li, Juan Wang, Sihai Xiang, Xiaozhou Feng, and Keqian Yang. “An engineered strong promoter for streptomycetes.” Applied and environmental microbiology, 2013: 4484-4492.
  • Wang, Wenfeng, Zhiqi Qiu, Hongming Tan, and Lixiang Cao. “Siderophore production by actinobacteria.” Biometals: an international journal on the role of metal ions in biology, biochemistry, and medicine, 2014: 623-631.
  • Watson, James D., and Francis H. C. Crick. “Molecular Structure of Nucleic Acids: A Structure for Deoxyribose Nucleic Acid.” Nature, 1953: 737-738.
  • Weber, Tilmann, et al. “antiSMASH 3.0-a comprehensive resource for the genome mining of biosynthetic gene clusters.” Nucleic acids research, 2015: W237-43.
  • Wehmeier, U. F., and W. Piepersberg. “Biotechnology and molecular biology of the alpha-glucosidase inhibitor acarbose.” Applied microbiology and biotechnology, 2004: 613-625.
  • Wehmeier, Udo. “Acarbose, ein therapeutisch eingesetzter Wirkstoff: Biosynthese ud Funktion.” BIOspektrum, 2004: 34-36.
  • Wehmeier, Udo F. “The Biosynthesis and Metabolism of Acarbose in Actinoplanes sp. SE 50/110: A Progress Report.” Biocatalysis and Biotransformation, 2003: 279-284.
  • Wehmeier, Udo F., and Wolfgang Piepersberg. “Chapter 19 Enzymology of Aminoglycoside Biosynthesis-Deduction from Gene Clusters.” Methods in Enzymology, 2009: 459-491.
  • Wendler, Sergej, et al. “Carbon source dependent biosynthesis of acarviose metabolites in Actinoplanes sp. SE50/110.” Journal of biotechnology, 2014: 113-120.
  • -. “The cytosolic and extracellular proteomes of Actinoplanes sp. SE50/110 led to the identification of gene products involved in acarbose metabolism.” Journal of biotechnology, 2013: 178-189.
  • -. “Comparative proteome analysis of Actinoplanes sp. SE50/110 grown with maltose or glucose shows minor differences for acarbose biosynthesis proteins but major differences for saccharide transporters.” Journal of proteomics, 2015.
  • -. “Comprehensive proteome analysis of Actinoplanes sp. SE50/110 highlighting the location of proteins encoded by the acarbose and the pyochelin biosynthesis gene cluster.” Journal of proteomics, 2015: 1-16.
  • Wendler, Wolfgang M. F., Elisabeth Kremmer, Reinhold Forster, and Ernst-Ludwig Winnacker. “Identification of Pirin, a Novel Highly Conserved Nuclear Protein.” Journal of Biological Chemistry, 1997: 8482-8489.
  • Willoughbly, L. G. “Observations on some aquatic Actinomycetes of streams and rivers.” Freshwater Biology, 1971: 23-27.
  • Wilson, Wayne A., et al. “Regulation of glycogen metabolism in yeast and bacteria.” FEMS microbiology reviews, 2010: 952-985.
  • Wink, Joachim M., Reiner M. Kroppenstedt, Peter Schumann, Gerhard Seibert, and Erko Stackebrandt. “Actinoplanes liguriensis sp. nov. and Actinoplanes teichomyceticus sp. nov.” International journal of systematic and evolutionary microbiology, 2006: 2125-2130.
  • Wolf, Andreas, Reinhard Kramer, and Susanne Morbach. “Three pathways for trehalose metabolism in Corynebacterium glutamicum ATCC13032 and their significance in response to osmotic stress.” Molecular microbiology, 2003: 1119-1134.
  • Wolf, Timo. Transcriptional regulation of acarbose biosynthesis in Actinoplanes sp. SE50/110 analyzed by next-generation sequencing, transcriptomics and genome editing. 2017.
  • Wolf, Timo, et al. “The MaIR type regulator AcrC is a transcriptional repressor of acarbose biosynthetic genes in Actinoplanes sp. SE50/110.” BMC genomics, 2017: 562.
  • -. “Genome improvement of the acarbose producer Actinoplanes sp. SE50/110 and annotation refinement based on RNA-seq analysis.” Journal of biotechnology, 2017.
  • -. “Targeted genome editing in the rare actinomycete Actinoplanes sp. SE50/110 by using the CRISPR/Cas9 System.” Journal of biotechnology, 2016: 122-128.
  • -. “The transcription of the acarbose biosynthetic gene cluster of Actinoplanes sp. SE50/110 is dependent on the growth phase.” in preparation.


Woo, Han Min, Stephan Noack, Gerd M. Seibold, Sabine Willbold, Bernhard J. Eikmanns, and Michael Bott. “Link between phosphate starvation and glycogen metabolism in Corynebacterium glutamicum, revealed by metabolomics.” Applied and environmental microbiology, 2010: 6910-6919.

  • Wozniak, Rachel A. F., and Matthew K. Waldor. “Integrative and conjugative elements: mosaic mobile genetic elements enabling dynamic lateral gene flow.” Nature reviews. Microbiology, 2010: 552-563.
  • Wu, Gang, Qiang Yan, J. Andrew Jones, Yinjie J. Tang, Stephen S. Fong, and Mattheos A. G. Koffas. “Metabolic Burden: Cornerstones in Synthetic Biology and Metabolic Engineering Applications.” Trends in biotechnology, 2016: 652-664.
  • Wu, Hang, et al. “Capturing the target genes of BdD in Saccharopolyspora erythraea using improved genomic SELEX method.” Applied microbiology and biotechnology, 2015: 2683-2692.
  • Xiang, Wen-Sheng, et al. “Isolation and identification of chlorinated genistein from Actinoplanes sp. HBDN08 with antioxidant and antitumor activities.” Journal of agricultural and food chemistry, 2010: 1933-1938.
  • Yakobson, E. A., and D. G. Guiney. “Conjugal transfer of bacterial chromosomes mediated by the RK2 plasmid transfer origin cloned into transposon Tn5.” Journal of bacteriology, 1984: 451-453.
  • Yamada, Yuuki, et al. “Terpene synthases are widely distributed in bacteria.” Proceedings of the National Academy of Sciences of the United States of America, 2015: 857-862.
  • Yamauchi, Yuto, Takashi Hirasawa, Masato Nishii, Chikara Furusawa, and Hiroshi Shimizu. “Enhanced acetic acid and succinic acid production under microaerobic conditions by Corynebacterium glutamicum harboring Escherichia coli transhydrogenase gene pntAB.” The Journal of general and applied microbiology, 2014: 112-118.
  • Yan, Wenjuan, et al. “The effect of c-di-GMP (3′-5′-cyclic diguanylic acid) on the biofilm formation and adherence of Streptococcus mutans.” Microbiological research, 2010: 87-96.
  • Yanisch-Perron, C., J. Vieira, and J. Messing. “Improved M13 phage cloning vectors and host strains: nucleotide sequences of the M13mp18 and pUC19 vectors.” Gene, 1985: 103-119.
  • Yu, Lei, Ting Lei, Xiaodong Ren, Xiaolin Pei, and Yan Feng. “Response surface optimization of I-(+)-lactic acid production using corn steep liquor as an alternative nitrogen source by Lactobacillus rhamnosus CGMCC 1466.” Biochemical Engineering Journal, 2008: 496-502.
  • Zahoor, Ahmed, Steffen N. Lindner, and Volker F. Wendisch. “Metabolic engineering of Corynebacterium glutamicum aimed at alternative carbon sources and new products.” Computational and structural biotechnology journal, 2012: e201210004.
  • Zhang, Chang-Sheng. Genomic Analysis of Secondary Metabolite Producing Actinomycetes: AcbM is a 2-epi-5-epi-valiolone 7-kinase. Wuppertal, 2002.
  • Zhang, Chang-Sheng, et al. “Biosynthesis of the C(7)-cyclitol moiety of acarbose in Actinoplanes species SE50/110. 7-O-phosphorylation of the initial cyclitol precursor leads to proposal of a new biosynthetic pathway.” The Journal of biological chemistry, 2002: 22853-22862.
  • Zhang, Chang-Sheng, Michael Podeschwa, Hans-Josef Altenbach, Wolfgang Piepersberg, and Udo F. Wehmeier. “The acarbose-biosynthetic enzyme AcbO from Actinoplanes sp. SE 50/110 is a 2-epi-5-epi-valiolone-7-phosphate 2-epimerase.” FEBS letters, 2003: 47-52.
  • Zhang, Chang-Sheng, Michael Podeschwa, Oliver Block, Hans-Josef Altenbach, Wolfgang Piepersberg, and Udo F.
  • Wehmeier. “Identification of a 1-epi-valienol 7-kinase activity in the producer of acarbose, Actinoplanes sp. SE50/110.” FEBS letters, 2003: 53-57.
  • Zhang, Dan, Qinqing Zhao, Ming Jiang, Qianjin Kang, and Linquan Bai. “(translated from chinese:) Biosynthetic pathway of deoxyaminosugar moiety in acarbose from Actinoplanes sp. SE50/110.” Acta Microbiol. Sin., 2019.
  • Zhang, Xiujun, and Ronald J. Parry. “Cloning and characterization of the pyrrolomycin biosynthetic gene clusters from Actinosporangium vitaminophilum ATCC 31673 and Streptomyces sp. strain UC 11065.” Antimicrobial agents and chemotherapy, 2007: 946-957.
  • Zhao, Qinqin, et al. “A severe leakage of intermediates to shunt products in acarbose biosynthesis.” Nature communications, n.d.
  • Zhao, Qinqin, Huixin Xie, Yao Peng, Xinran Wang, and Linquan Bai. “Improving acarbose production and eliminating the by-product component C with an efficient genetic manipulation system of Actinoplanes sp. SE50/110.” Synthetic and Systems Biotechnology, 2017: 302-309.
  • Zhou, Zhihui, et al. “Increasing available NADH supply during succinic acid production by Corynebacterium glutamicum.” Biotechnology progress, 2015: 12-19.
  • Ziegelhoffer, Eva C., and Timothy J. Donohue. “Bacterial responses to photo-oxidative stress.” Nature reviews. Microbiology, 2009: 856-863.
  • Zotchev, S., K. Haugan, O. Sekurova, H. Sletta, T. E. Ellingsen, and S. Valla. “Identification of a gene cluster for antibacterial polyketide-derived antibiotic biosynthesis in the nystatin producer Streptomyces noursei ATCC 11455.” Microbiology (Reading, England), 2000: 611-619.

Claims
  • 1. A method to engineer an Actinomycetales strain, preferably an Actinoplanes strain, for the improved production of acarbose, the method comprising engineering the Actinomycetales strain (i) for absent or reduced expression of extracellular small carbohydrate binding protein Cgt according to SEQ ID No. 20 and/or,(ii) for absent or reduced expression of at least one gene which is essential for carotenoid synthesis, and/or,(iii) for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB according to SEQ ID No. 13, and/or(iv) for overexpression of UDP-glucose-1P uridyltransferase GtaB according to SEQ ID No. 19.
  • 2. The method according to claim 1, said method comprising (i) deletion or mutation of the gene encoding extracellular small carbohydrate binding protein Cgt according to SEQ ID No. 20 and/or,(ii) deletion or mutation of at least one gene which is essential in the carotenoid synthesis and/or,(iii) introducing a vector comprising an expression cassette for AcbB into the Actinomycetales strain and/or(iv) introducing a vector comprising an expression cassette for GtaB into the Actinomycetales strain.
  • 3. The method according to claim 2, wherein the expression cassette according to (iii) and/or (iv) is under the control of a medium strong promoter, as characterized by a normalized glucuronidase activity of at least 1x·10−4 [L·g−1·min−1] in a glucuronidase assay or a strong promoter as characterized by a normalized glucuronidase activity of at least 5x·10−4 [L·g-1·min-1] in a glucuronidase assay.
  • 4. An Actinomycetales strain, preferably an Actinoplanes strain, for the production of acarbose, wherein the Actinomycetales strain is genetically engineered for absent or reduced expression of extracellular small carbohydrate binding protein Cgt according to SEQ ID No. 20.
  • 5. The Actinomycetales strain according to claim 4, wherein the Actinomycetales strain is a cgt deletion mutant.
  • 6. An Actinomycetales strain, preferably an Actinoplanes strain, for the production of acarbose, wherein the Actinomycetales strain is genetically engineered for absent or reduced expression of at least one gene which is essential for carotenoid synthesis.
  • 7. An Actinomycetales strain, preferably an Actinoplanes strain, for the production of acarbose, wherein the Actinomycetales strain is genetically engineered for overexpression of dTDP-D-glucose-4,6-dehydratase AcbB according to SEQ ID No. 13.
  • 8. An Actinomycetales strain, preferably an Actinoplanes strain, for the production of acarbose, wherein the Actinomycetales strain is genetically engineered for overexpression of UDP-glucose-1P uridyltransferase GtaB according to SEQ ID No. 19.
  • 9. An Actinomycetales strain for the production of acarbose according to claim 6 or a method according to any of claims 1, 2 or 3, wherein the at least one gene which is essential for carotenoid synthesis comprises at least one gene selected from any of a. the genes of the MEP/DOXP pathway i. 1-deoxy-D-xylulose-5-phosphate synthase gene dxs, ACSP50_7096 according to SEQ ID No. 23,ii. 4-hydroxy-3-methylbut-2-en-1-yl diphosphate synthase gene ispG, ACSP50_7248 according to SEQ ID No. 24,iii. 1-deoxy-D-xylulose-5-phosphate reductoisomerase gene dxr, ACSP50_7250 according to SEQ ID No. 25,iv. 4-hydroxy-3-methylbut-2-enyl diphosphate reductase gene ispH, ACSP50_7707 according to SEQ ID No. 26,v. 4-(cytidine 5′-diphospho)-2-C-methyl-D-erythritol kinase gene ispE, ACSP50_7802 according to SEQ ID No. 27,vi. 2-C-methyl-D-erythritol 2;4-cyclodiphosphate synthase gene ispF, ACSP50_8046 according to SEQ ID No. 28, and/orvii. 2-C-methyl-D-erythritol 4-phosphate cytidylyltransferase gene ispD, ACSP50_8047 according to SEQ ID No. 29,b. the genes of terpene cluster 1 i. isopentenyl-diphosphate delta-isomerase gene idi, ACSP50_0146 according to SEQ ID No. 30,ii. zeta-phytoene desaturase gene crtl, ACSP50_0147 according to SEQ ID No. 10,iii. polyprenyl synthetase gene crtE/ldsA, ACSP50_0148 according to SEQ ID No. 31,iv. phytoene synthase gene crtB, ACSP50_0149 according to SEQ ID No. 32,v. deoxyribodipyrimidine photo-lyase gene, ACSP50_0150 according to SEQ ID No. 33, orvi. pyridine nucleotide-disulfide oxidoreductase gene, ACSP50_0151 according to SEQ ID No. 34,c. the genes of terpene cluster 2a i. transcriptional regulator gene ACSP50_1631 according to SEQ ID No. 35,ii. lycopene cyclase gene ACSP50_1632 according to SEQ ID No. 36,iii. lycopene cyclase gene ACSP50_1633 according to SEQ ID No. 37,iv. polyprenyl synthetase, farnesyl pyrophosphate synthetase 2 gene fps2/crtE, ACSP50_1634 according to SEQ ID No. 38, orv. methylenetetrahydrofolate reductase (NADPH) gene, ACSP50_1635 according to SEQ ID No. 39,d. the genes of terpene cluster 2b i. LysR-family transcriptional regulator gene, ACSP50_1650 according to SEQ ID No. 40,ii. methyltransferase type 11 gene, ACSP50_1651 according to SEQ ID No. 41,iii. CDP-alcoholphosphatidyltransferase pgsA, ACSP50_1652 according to SEQ ID No. 42,iv. zeta-phytoene desaturase (crtl-family) gene crtD, ACSP50_1653 according to SEQ ID No. 43,v. glycosyl transferase gene cruC, ACSP50_1654 according to SEQ ID No. 44,vi. hypothetical protein (put. membrane prot,) gene cruF, ACSP50_1655 according to SEQ ID No. 45,vii. GCN5 family acetyltransferase gene, ACSP50_1656 according to SEQ ID No. 46,viii. monooxygenase gene, ACSP50_1657 according to SEQ ID No. 47, orix. short-chain dehydrogenase gene, ACSP50_1658 according to SEQ ID No. 48,e. polyprenyl synthetase gene crtE, ACSP50_3873 according to SEQ ID No. 49, orf. the genes for camphene-like monoterpene biosynthesis terpene cluster 3 i. transcriptional regulator (Crp/Fnr family) gene eshA, ACSP50_1949 according to SEQ ID No. 104,ii. camphene synthase gene, ACSP50_1950 according to SEQ ID No. 50,iii. methyltransferase (SAM-dependent) type 11 gene, ACSP50_1951 according to SEQ ID No. 105,iv. glycosyl-hydrolase gene, ACSP50_1952 according to SEQ ID No. 106, orv. oxidoreductase/aldo/ketoreductase, ACSP50_1953 according to SEQ ID No. 107.
  • 10. An expression vector for Actinoplanes comprising an expression cassette for AcbB according to SEQ ID No. 13 and/or an expression cassette for GtaB according to SEQ ID No. 19, and/or an expression cassette for MerR according to SEQ ID No. 22.
  • 11. The expression vector according to claim 10 furthermore comprising a medium strong promoter characterized by a normalized glucuronidase activity of at least 1x·10−4 [L·g−1·min−1] in a glucuronidase assay or a strong promoter characterized by a normalized glucuronidase activity of at least 5x·10−4 [L·g−1·min−1] in a glucuronidase assay.
  • 12. The expression vector according to claim 10 or 11 furthermore comprising a. the φC31 integrase gene int according to SEQ ID No. 85, andb. the origin of transfer (incP) according to SEQ ID No. 87, andc. the relaxosome gene traJ according to SEQ ID No. 88, andd. an origin of replication, preferably the high-copy-number ColE1/pMB1/pBR322/pUC, origin of replication (ori) according to SEQ ID No. 89 ande. optionally at least one resistance marker, preferably a resistance marker mediating apramycin resistance, more preferably aac(3)IV according to SEQ ID No. 90, apmR, andf. optionally at least one T4-terminator,and optionally, wherein the vector does not comprise putative antisense promoters according to SEQ ID NO 108 and/or SEQ ID No. 109.
  • 13. The expression vector according to claim 11 or 12 wherein said strong promoter is selected from the group consisting of a. apm according to SEQ ID No. 96,b. ermE* according to SEQ ID No. 98,c. katE according to SEQ ID No. 94,d. moeE5 according to SEQ ID No. 95, ore. gapDH according to SEQ ID No. 82,
  • 14. An Actinomycetales strain for the production of acarbose according to any of claims 4 to 9, wherein the strain comprises a vector according to any of claims 10 to 13.
  • 15. Use of an Actinomycetales strain according to any previous claim in the production of acarbose.
Priority Claims (1)
Number Date Country Kind
19203693.7 Oct 2019 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2020/077688 10/2/2020 WO