• No results found

University of Groningen The diversity of glycogen branching enzymes in microbes Zhang, Xuewen

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen The diversity of glycogen branching enzymes in microbes Zhang, Xuewen"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

The diversity of glycogen branching enzymes in microbes

Zhang, Xuewen

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Zhang, X. (2019). The diversity of glycogen branching enzymes in microbes. University of Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 6

Summary and Prospects

(3)

Glycogen, as the major carbon and energy reserve polymer in microorganisms and animals has been linked to a broad range of physiological processes such as cell differentiation (1), environmental survival (2,3), biofilm formation (4), and virulence (5). Impairments in glycogen synthesis or degradation in humans and animals is in most cases fatal, as for example in Pompe’s disease, Laforin dis-ease, and von Gierke’s disease (6-9). In the human pathogenic bacterium Myco-bacterium tuberculosis, the causative agent of tuberculosis killing more than one million people per year, glycogen metabolism has been connected to infection and virulence (10,11). Glycogen is a polymer of α-D-anhydroglucose, consisting of a linear backbone of α-1,4-linked anhydroglucoses with side chains attached via α-1,6-O-glycosidic linkages. The key enzymes in glycogen synthesis are: phosphotransferase, phosphoglucomutase, glucose-1-phosphate adenylyltrans-ferase, glycogenin (Eukaryotes only), glycogen synthase, and glycogen branch-ing enzyme (GBE).

GBE is a member of the α-amylase family, a large collection of the enzymes act-ing on α-glucans, includact-ing glycogen, amylose, amylopectin, and trehalose. GBEs are classified in the glycoside hydrolase families 13 (GH13) and 57 (GH57). The GH13 is a very large and diverse group of enzymes sharing an (α/β)8-barrel cat-alytic domain and catalyzing more than 20 different reactions (http://www.cazy. org). Based on protein homology, GH13 GBEs are divided into the subfamily GH13_8 (mainly Eukaryotes) and GH13_9 (mainly Prokaryotes). Family GH57 has 6 enzyme specificities; α-amylase (EC 3.2.1.1); α-galactosidase (EC 3.2.1.22); amylopullulanase (EC 3.2.1.41); cyclomaltodextrinase (EC 3.2.1.54); branching enzyme (EC 2.4.1.18); 4-α-glucanotransferase (EC 2.4.1.25), which all share an (α/β)7 catalytic barrel structure.

Although the catalytic domains of the GH13 and GH57 GBEs have a different fold they share the same double displacement catalytic mechanism, with retention of the anomeric configuration. All GBEs initiate their reaction by cleaving the α-1,4-linked linear glucan donor substrate forming a covalent glucosyl-enzyme intermediate. In the second step, the glucosyl moiety is transferred to an acceptor glucan or a water molecule to form new branches or release a free glucan chain, respectively, referred to as branching activity or hydrolytic activity. In contrast to the variety of GH13 GBEs that have been characterized (12-17), only three

(4)

GH57 GBEs have been characterized to date (18-20). This in spite of the fact that over 800 GH57 GBE gene sequences are available (21,22). This discrepancy in the number of GH57 GBEs characterized and the vast number of gene sequences available triggered me to study GH57 GBEs in more detail.

Chapter 1 introduces the current insights in the glycogen metabolism in

prokar-yotic microorganisms, the similarities and differences between family GH13 and GH57 GBEs, and the application of GBEs in starch modification. Emphasis is on the current understanding of the biological function of glycogen, the metabolic pathways involved in the synthesis of glycogen and the structural basis of the hydrolysis/branching specificity of GBEs. The differences in the reaction speci-ficity of the two families of GBEs is far from understood. Most microorganisms have one type of GBE, either a GH13 or a GH57. However, various microorgan-isms, including M. tuberculosis and Petrotoga mobilis, have the genes encoding a GH13 as well as a GH57 GBE in their genome. The simultaneous presence of a GH13 and GH57 GBE in a prokaryotic genome was found to occur widely in Nature (Chapter 1).

In contrast to GH13 GBEs, GH57 GBEs have a relatively high hydrolytic activity (20). The activity of AmyC from Thermotoga maritima, shown to be an α-amyl-ase (23,24), was characterized in detail and it was concluded that it actually is a GBE with a high hydrolytic side activity (Chapter 2). The high hydrolytic activ-ity is explained from the crystal structure, which shows that AmyC has a consid-erably shorter catalytic loop (residues 213-220) than most GH57 GBEs. This loop does not reach the acceptor side of the active site groove. In addition, one of the “gatekeeper” residues important for branching activity (19) is positioned away from the active site, thus being unable to fulfill its role as gatekeeper. Sequence analysis predicts that AmyC-like GH57 enzymes are present in species closely related to T. maritima, such as Kosmotoga pacifica, and that these enzymes are also GBEs with a relatively high hydrolytic activity.

In Chapter 1 it is described that a range of microorganisms contain both a GH13 and a GH57 GBE gene in their genome. So far, none of these GBEs have been characterized in detail. In Chapter 3 the two putative GBEs, one GH13 and one GH57, from P. mobilis are overexpressed and characterized. Both GBEs convert

(5)

amylose in branched maltodextrins with a distinct degree of branching; 13% for GH13 GBE and 8.5% for GH57 GBE. The molecular weight of the GH13 GBE product is 107 Da, while that of the GH57 GBE is 104 Da. The results from chapter 3 and those of studies on the GH13 and GH57 GBEs of M. tuberculosis together suggest that the two types of GBEs have different roles in-vivo. It is proposed that the GH13 GBE is involved in glycogen synthesis, which is supported by experimental data, while the function of GH57 GBE remains unclear. In M. tu-berculosis the GH57 GBE might be involved in lipopolysaccharide synthesis, although experimental proof is lacking. The in-vivo function of the GH57 GBE is thus still a mystery.

GBEs employ a double displacement mechanism with either water (hydrolysis) or an α-glucan (branching) as acceptor. In Chapter 4 two GH13 GBEs and two GH57 GBEs are characterized in detail to understand if and how the two family GBEs differ in activity and specificity. The GH13 GBEs have a substantially higher activity towards amylose than the GH57 GBEs. In addition, the GH57 GBEs displayed a considerable higher hydrolytic activity than GH13 GBEs. With respect to the products formed, the GH13 GBEs branched amylose or amylopec-tin more than GH57 GBEs. Moreover, the GH13 GBEs synthesized branched α-glucans with a narrow molecular weight distribution, while the GH57 GBEs products consisted of two or three molecular weight fractions. Most likely the relatively high hydrolytic activity of the GH57 GBEs is not compatible with the synthesis of branched α-glucans with a narrow molecular weight distribution. The results also indicate that the R. marinus GBE, which is commercially availa-ble as Branchzyme of Novozymes, is currently the best GBE for industrial starch processing; this enzyme only slightly hydrolyses the starch resulting in very little formation of low molecular weight material and makes a highly branched malto-dextrin (10%) with a narrow molecular weight distribution.

The GBE modified starches have several advantages over native starches, such as an increased degree of branching and solubility, smaller molecules, and lower viscosity. The higher proportion of α-1,6-linkages makes the branched maltodex-trins more resistant to degradative enzymes such as α-amylases and amylogluco-sidases, the later hydrolyzing α-1,6-linkages at a lower rate than α-1,4-linkages, and thereby makes the (highly) branched maltodextrins slowly digestible. Some

(6)

highly branched maltodextrins have been used in certain specific applications such as the formulation of peritoneal dialysis solution and sport drinks. Chapter

5 analyzes the digestibility of a series of GBE modified starches with distinct

de-gree of branching from 5 to 14%. The various highly branched maltodextrins are digested at different rates. The products with a higher branch density and shorter internal chain length contain more slowly digestible starch and more resistant starch. These results suggest that the starches treated by branching enzymes have a slowly digestible character, which could be used to control postprandial glucose levels.

In conclusion, the results in this thesis have extended the understanding of the catalytic specificity of GBEs, especially those of GH57. The GH57 GBEs display a relatively higher hydrolytic activity than GH13 GBEs. Some GH57 GBEs, such as AmyC from T. maritima, show up to 29% hydrolytic activity. In the case of AmyC, the high hydrolytic activity is correlated to the length of the “LOOP”, a stretch of 20-25 amino acids near the top of the active site. GH57 GBEs such as that of T. kodakarensis KOD1 have a long loop which almost closes the active site and positions a tyrosine residue into the active site (19,20). This tyrosine is im-portant for the branching activity of the GBE of T. thermophilus (20). In AmyC and related GBEs this loop is much shorter and does not reach the active site(23). Engineering the loop of AmyC into KOD1, and vice versa, and mutate the tyros-ine should tell us more on the role of the loop and the tyrostyros-ine.

So far it is not clear if the high hydrolytic activity observed for AmyC is an “arte-fact” of the way in which the activity is measured in-vitro. GBEs are intracellular enzymes “seeing” growing linear α-glucan chains as substrate in an environment with much less water than in the test tubes. In addition, the concentration of sub-strate in the cell is not known. All these parameters, nature and concentration of substrate, concentration of enzyme, and the amount of water all can influence the activity and specificity of the GBE. Two approaches to get some idea on the natural activity and specificity of AmyC are isolating glycogen from T. maritima and synthesizing glycogen in-vitro by combining glycogen synthase or starch/ potato phosphorylase with the GBE (17,25). Potato/starch phosphorylase, com-monly known as α-glucan phosphorylase (E.C. 2.4.1.1), catalyzes the reversible transfer of the glucose unit of glucose-1-phosphate to the non-reducing end of an

(7)

α,1-4 glucan chain with the release of phosphate (26). The Noble Prize 1947 was awarded to Cori and Cori for their instrumental work on polysaccharide phos-phorylases (27). By combining potato phosphorylase with glucose-1-phosphate, a primer such as maltoheptaose, and a GBE in various concentrations, the in-vitro glycogen synthesis can be mimicked. Van der Vlist et al. (25) have shown that using the Deinococcus geothermalis GBE a branched maltodextrin of relatively high molecular weight can be obtained. The question is whether in such an en-zyme cocktail AmyC still has a relatively high hydrolytic activity. The structure of the in-vitro synthesized branched α-glucan can subsequently be compared to that of glycogen extracted from T. maritima cells. So far, the glycogen structure of T. maritima has not been described in the scientific literature, making it not possible to compare the structure of the branched maltodextrins made from am-ylose with the natural glycogen of T. maritima.

The enzyme cocktail approach can also be used to study the GH13 and GH57 GBEs of P. mobilis and M. tuberculosis and get more insights into the roles of these two enzymes in glycogen synthesis. It is still not understood why certain microorganisms have multiple GBEs and which roles these GBEs play. For M. tuberculosis, it seems that the GH13 GBE is directly involved in glycogen syn-thesis while the GH57 GBE may play a role in the synsyn-thesis of the cell wall li-popolysaccharides (28,29). Gusthart (30) studied the M. tuberculosis GH57 GBE in detail. The protein was successfully produced in E. coli, but no activity could be detected on a range of glucans, including p-nitrophenyl-α-D-glucopyranoside, amylose, amylopectin, maltodextrin, and glycogen. What became clear from this PhD thesis is that the GH57 GBE of M. tuberculosis is not an ordinary GBE. Further research on this and the P. mobilis GH57 GBE should shed some light on the activity and role of these peculiar GBEs.

The GH57 GBE (Rv3031), the glycogen synthase (Rv3032), and the S-adenosyl-methionine-dependent methyltransferase (Rv3030) are located in the same gene cluster of M. tuberculosis, and have been proposed to synthesize lipopolysaccha-ride (28,29). However, although the GH57 GBE has been identified in P. mobilis, the other two genes are absent from the genome sequence (31), which suggests that the GH57 GBE of P. mobilis performs a different role in-vivo. The bacteria with “toga” can survive at deep marine and oil well with high osmotic stress, so

(8)

it is very likely that P. mobilis produces stress-protectant molecules to balance the external pressure. The protectants trehalose and cellobiose produced from α-glucans related genes are present in P. mobilis (32), so it seems likely that the GH57 GBE is in one way or another linked to or even involved in the synthesis of such protectants in P. mobilis. Further genetic research on P. mobilis should give some ideas on this.

New biopolymers have been created by using GBEs as a tool to increase branch-ing frequency of post-harvest starches. The highly branched products made from native starches by the action of GBEs have potential in industrial applications, such as sport drink ingredient, slowly digestible starch, and film coating (33-37). The structural properties of the branched glucans can be further diversified by combining the action of GBEs with other enzymes. For example, the amylomal-tase and GBE have been used together to produce highly branched polymers with more short side chains compared to GBE products (38); β-amylase has been applied to hydrolyse GBE products to obtain extremely branched α-glucans (39); potato phosphorylase in combination with GBE produces branched α-glucans with a high Mw (25). Following these ideas, it is proposed to use sucrose as an ideal substrate to produce highly branched α-glucans and fructose. Sucrose is a cheap, renewable biomaterial which is plentiful available while the fructose as the byproduct has more wide applications than sucrose, such as sweetener or as substrate for E. coli or yeast producing succinic acid and ethanol (40,41).

(9)

References

1. Yeo, M., and Chater, K. (2005) The interplay of glycogen metabolism and differentiation provides an insight into the developmental biology of

Streptomyces coelicolor. Microbiology 151, 855-861

2. Bourassa, L., and Camilli, A. (2009) Glycogen contributes to the environmental persistence and transmission of Vibrio cholerae. Mol Microbiol 72, 124-138 3. Bonafonte, M., Solano, C., Sesma, B., Alvarez, M., Montuenga, L.,

García-Ros, D., and Gamazo, C. (2000) The relationship between glycogen synthesis, biofilm formation and virulence in Salmonella enteritidis. FEMS Microbiol Lett 191, 31-36

4. Dinadayala, P., Sambou, T., Daffe, M., and Lemassu, A. (2008) Comparative structural analyses of the α-glucan and glycogen from Mycobacterium bovis.

Glycobiology 18, 502-508

5. Busuioc, M., Mackiewicz, K., Buttaro, B. A., and Piggot, P. J. (2009) Role of intracellular polysaccharide in persistence of Streptococcus mutans. J Bacteriol 191, 7315-7322

6. Bao, Y., Kishnani, P., Wu, J.-Y., and Chen, Y.-T. (1996) Hepatic and neuromuscular forms of glycogen storage disease type IV caused by mutations in the same glycogen branching enzyme gene. J Clin Invest 97, 941-948 7. Stetten Jr, D., and Stetten, M. R. (1960) Glycogen metabolism. Physiol Rev 40,

505-537

8. Field, R. (1960) Glycogen deposition diseases. The Metabolic Basis of Inherited

Disease, edited by JB Stanbury, JB Wyngaarden and DS Fredrickson. New York: McGraw-Hill Co, 156

9. Bruno, C., Servidei, S., Shanske, S., Karpati, G., Carpenter, S., Mckee, D., Barohn, R. J., Hirano, M., Rifai, Z., and Dimauro, S. (1993) Glycogen branching enzyme deficiency in adult polyglucosan body disease. Ann Neurol 33, 88-93 10. Koliwer-Brandl, H., Syson, K., van de Weerd, R., Chandra, G., Appelmelk,

B., Alber, M., Ioerger, T. R., Jacobs, W. R., Geurtsen, J., Bornemann, S., and Kalscheuer, R. (2016) Metabolic network for the biosynthesis of intra and extracellular α-glucans required for virulence of Mycobacterium tuberculosis.

Plos Pathog 12

11. Sambou, T., Dinadayala, P., Stadthagen, G., Barilone, N., Bordat, Y., Constant, P., Levillain, F., Neyrolles, O., Gicquel, B., Lemassu, A., Daffe, M., and Jackson, M. (2008) Capsular glucan and intracellular glycogen of Mycobacterium

tuberculosis: biosynthesis and impact on the persistence in mice. Mol Microbiol

(10)

12. Fan, Q., Xie, Z., Zhan, J., Chen, H., and Tian, Y. (2016) A glycogen branching enzyme from Thermomonospora curvata: Characterization and its action on maize starch. Starch-Stärke 68, 355-364

13. Roussel, X., Lancelon-Pin, C., Vikso-Nielsen, A., Rolland-Sabate, A., Grimaud, F., Potocki-Veronese, G., Buleon, A., Putaux, J. L., and D'Hulst, C. (2013) Characterization of substrate and product specificity of the purified recombinant glycogen branching enzyme of Rhodothermus obamensis. BBA-Gen Subjects 1830, 2167-2177

14. Garg, S. K., Alam, M. S., Kishan, K. R., and Agrawal, P. (2007) Expression and characterization of α-(1,4)-glucan branching enzyme Rv1326c of Mycobacterium

tuberculosis H37Rv. Protein Expres Purif 51, 198-208

15. Van der Maarel, M. J. E. C., Vos, A., Sanders, P., and Dijkhuizen, L. (2003) Properties of the glucan branching enzyme of the hyperthermophilic bacterium

Aquifex aeolicus. Biocatal Biotransfor 21, 199-207

16. Takata, H., Takaha, T., Kuriki, T., Okada, S., Takagi, M., and Imanaka, T. (1994) Properties and active-center of the thermostable branching enzyme from Bacillus

stearothermophilus. Appl Environ Microb 60, 3096-3104

17. Palomo, M., Kralj, S., van der Maarel, M. J. E. C., and Dijkhuizen, L. (2009) The unique branching patterns of Deinococcus glycogen branching enzymes are determined by their N-terminal domains. Appl Environ Microb 75, 1355-1362 18. Na, S., Park, M., Jo, I., Cha, J., and Ha, N. C. (2017) Structural basis for the

transglycosylase activity of a GH57-type glycogen branching enzyme from

Pyrococcus horikoshii. Biochem Bioph Res Co 484, 850-856

19. Santos, C. R., Tonoli, C. C. C., Trindade, D. M., Betzel, C., Takata, H., Kuriki, T., Kanai, T., Imanaka, T., Arni, R. K., and Murakami, M. T. (2011) Structural basis for branching-enzyme activity of glycoside hydrolase family 57: Structure and stability studies of a novel branching enzyme from the hyperthermophilic archaeon Thermococcus kodakaraensis KOD1. Proteins 79, 547-557

20. Palomo, M., Pijning, T., Booiman, T., Dobruchowska, J. M., van der Vlist, J., Kralj, S., Planas, A., Loos, K., Kamerling, J. P., Dijkstra, B. W., van der Maarel, M. J. E. C., Dijkhuizen, L., and Leemhuis, H. (2011) Thermus thermophilus glycoside hydrolase family 57 branching enzyme crystal structure, mechanism of action, and products formed. J Biol Chem 286, 3520-3530

21. Blesak, K., and Janecek, S. (2013) Two potentially novel amylolytic enzyme specificities in the prokaryotic glycoside hydrolase a-amylase family GH57.

Microbiol-Sgm 159, 2584-2593

(11)

from the glycoside hydrolase family GH57. Extremophiles : life under extreme

conditions 16, 497-506

23. Dickmanns, A., Ballschmiter, M., Liebl, W., and Ficner, R. (2006) Structure of the novel α-amylase AmyC from Thermotoga maritima. Acta Crystallogr D Biol

Crystallogr 62, 262-270

24. Ballschmiter, M., Futterer, O., and Liebl, W. (2006) Identification and characterization of a novel intracellular alkaline α-amylase from the hyperthermophilic bacterium Thermotoga maritima MSB8. Appl Environ

Microb 72, 2206-2211

25. van der Vlist, J., Palomo Reixach, M., van der Maarel, M., Dijkhuizen, L., Schouten, A. J., and Loos, K. J. M. R. C. (2008) Synthesis of branched polyglucans by the tandem action of potato phosphorylase and Deinococcus

geothermalis glycogen branching enzyme. Macromol Rapid Comm 29,

1293-1297

26. Rathore, R., Garg, N., Garg, S., and Kumar, A. J. C. r. i. b. (2009) Starch phosphorylase: role in starch metabolism and biotechnological applications.

Crit Rev Biotechnol 29, 214-224

27. Cori, C. F., and Cori, G. T. (1947) Polysaccharide phosphorylase. Nobel lectures 216

28. Kaur, D., Pham, H., Larrouy-Maumus, G., Riviere, M., Vissa, V., Guerin, M. E., Puzo, G., Brennan, P. J., and Jackson, M. (2009) Initiation of methylglucose lipopolysaccharide biosynthesis in Mycobacteria. Plos One 4

29. Stadthagen, G., Sambou, T., Guerin, M., Barilone, N., Boudou, F., Korduláková, J., Charles, P., Alzari, P. M., Lemassu, A., and Daffé, M. J. J. o. B. C. (2007) Genetic basis for the biosynthesis of methylglucose lipopolysaccharides in

Mycobacterium tuberculosis. J Biol Chem 282, 27270-27276

30. Gusthart, J. S. (2018) An investigation into an unusual glycan branching enzyme from Mycobacterium tuberculosis (thesis), University of Southampton

31. Chandra, G., Chater, K. F., and Bornemann, S. (2011) Unexpected and widespread connections between bacterial glycogen and trehalose metabolism.

Microbiol-Sgm 157, 1565-1572

32. Rodionov, D. A., Rodionova, I. A., Li, X. Q., Ravcheev, D. A., Tarasova, Y., Portnoy, V. A., Zengler, K., and Osterman, A. L. (2013) Transcriptional regulation of the carbohydrate utilization network in Thermotoga maritima.

Front Microbiol 4

33. Takata, H., Takaha, T., Nakamura, H., Fujii, K., Okada, S., Takagi, M., Imanaka, T. J. J. o. f., and bioengineering. (1997) Production and some properties of a

(12)

dextrin with a narrow size distribution by the cyclization reaction of branching enzyme. 84, 119-123

34. Van der Maarel, M. J. E. C., Ter Veer, B. C. A., Vrieling-Smit, A., and Delnoye, D. A. P. (2014) Methods and means for coating paper by film coating. Netherlands Patent WO2014/003556Al

35. Takata, H., Akiyama, T., Kajiura, H., Kakutani, R., Furuyashiki, T., Tomioka, E., Kojima, I., and Kuriki, T. (2010) Application of branching enzyme in starch processing. Biocatal Biotransfor 28, 60-63

36. Backer, D., and Saniez, M.-H. (2007) Soluble highly branched glucose polymers and their method of production. United States Patent USOO7211662B2

37. Deremaux, L., Petitjean, C., and Wills, D. (2013) Soluble, highly branched glucose polymers for enteral and parenteral nutrition and for peritoneal dialysis. United States Patent US8445460B2

38. Rashid, A. M., Batey, S. F. D., Syson, K., Koliwer-Brandl, H., Miah, F., Barclay, J. E., Findlay, K. C., Nartowski, K. P., Khimyak, Y. Z., Kalscheuer, R., and Bornemann, S. (2016) Assembly of α-Glucan by GlgE and GlgB in Mycobacteria and Streptomycetes. Biochemistry 55, 3270-3284

39. Lee, B. H., Yan, L., Phillips, R. J., Reuhs, B. L., Jones, K., Rose, D. R., Nichols, B. L., Quezada-Calvillo, R., Yoo, S. H., and Hamaker, B. R. (2013) Enzyme synthesized highly branched maltodextrins have slow glucose generation at the mucosal α-glucosidase level and are slowly digestible in vivo. Plos One 8 40. Berthels, N. J., Otero, R. R. C., Bauer, F. F., Thevelein, J. M., and Pretorius, I.

S. (2004) Discrepancy in glucose and fructose utilisation during fermentation by

Saccharmyces cerevisiae wine yeast strains. Fems Yeast Res 4, 683-689

41. Andersson, C., Hodge, D., Berglund, K. A., and Rova, U. (2007) Effect of different carbon sources on the production of succinic acid using metabolically engineered Escherichia coli. Biotechnol Progr 23, 381-388

(13)

Referenties

GERELATEERDE DOCUMENTEN

Resultaten van de modificatie van amylose suggereren dat de transferase activiteit van GVEs ook gebruikt wordt op complexe substraten zoals zetmeel. Samenvattend heeft dit

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright

Applied Biocatalysis, University of Groningen Mastering your PhD, course, University of Groningen Microbiological Safety Techniques, University of Groningen PhD Welcome Day,

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright

This work described in this thesis was performed in the group of Aquatic Biotechnology and Bioproduct Engineering of the Engineering and Technology Institute Groningen (ENTEG) in

obamensis was six times more active on amylose than on amylopectin and had low activity on glycogen (126), which is because amylopectin and glycogen as GBE products already

Identification of Thermotoga maritima SMB8 GH57 α-amylase AmyC as a glycogen branching enzyme with high hydrolytic activity.. Xuewen Zhang, Hans Leemhuis, Štefan Janeček,

The GH57 GBE, in contrast has a very low branching activity with amylose as substrate and forms branched α-glucans of considerably lower MW (10 4 Da) with a lower degree of