首页 | 本学科首页   官方微博 | 高级检索  
   检索      


Metabolic Engineering of Fungal Strains for Conversion of d-Galacturonate to meso-Galactarate
Authors:Dominik Mojzita  Marilyn Wiebe  Satu Hilditch  Harry Boer  Merja Penttil?  Peter Richard
Institution:VTT Technical Research Centre of Finland, Espoo, Finland
Abstract:d-Galacturonic acid can be obtained by hydrolyzing pectin, which is an abundant and low value raw material. By means of metabolic engineering, we constructed fungal strains for the conversion of d-galacturonate to meso-galactarate (mucate). Galactarate has applications in food, cosmetics, and pharmaceuticals and as a platform chemical. In fungi d-galacturonate is catabolized through a reductive pathway with a d-galacturonate reductase as the first enzyme. Deleting the corresponding gene in the fungi Hypocrea jecorina and Aspergillus niger resulted in strains unable to grow on d-galacturonate. The genes of the pathway for d-galacturonate catabolism were upregulated in the presence of d-galacturonate in A. niger, even when the gene for d-galacturonate reductase was deleted, indicating that d-galacturonate itself is an inducer for the pathway. A bacterial gene coding for a d-galacturonate dehydrogenase catalyzing the NAD-dependent oxidation of d-galacturonate to galactarate was introduced to both strains with disrupted d-galacturonate catabolism. Both strains converted d-galacturonate to galactarate. The resulting H. jecorina strain produced galactarate at high yield. The A. niger strain regained the ability to grow on d-galacturonate when the d-galacturonate dehydrogenase was introduced, suggesting that it has a pathway for galactarate catabolism.d-Galacturonate is the main component of pectin, an abundant and cheap raw material. Sugar beet pulp and citrus peel are both rich in pectin residues. At present, these residues are mainly used as cattle feed. However, since energy-consuming drying and pelletizing of the residues is required to prevent them from rotting, it is not always economical to process the residues, and it is desirable to find alternative uses.Various microbes which live on decaying plant material have the ability to catabolize d-galacturonate using various, completely different pathways (19). Eukaryotic microorganisms use a reductive pathway in which d-galacturonate is first reduced to l-galactonate by an NAD(P)H-dependent reductase (12, 17). In the following steps a dehydratase, aldolase, and reductase convert the l-galactonate to pyruvate and glycerol (9, 11, 14).In Hypocrea jecorina (anamorph Trichoderma reesei) the gar1 gene codes for a strictly NADPH-dependent d-galacturonate reductase. In Aspergillus niger a homologue gene sequence, gar2, exists; however, a different gene, gaaA, is upregulated during growth on d-galacturonate containing medium (16). The gaaA codes for a d-galacturonate reductase with different kinetic properties than the H. jecorina enzyme, having a higher affinity toward d-galacturonate and using either NADH or NADPH as cofactor. It is not known whether gar2 codes for an active protein.Some bacteria, such as Agrobacterium tumefaciens or Pseudomonas syringae, have an oxidative pathway for d-galacturonate catabolism. In this pathway d-galacturonate is first oxidized to meso-galactarate (mucate) by an NAD-utilizing d-galacturonate dehydrogenase. Galactarate is then converted in the following steps to α-ketoglutarate. This route is sometimes called the α-ketoglutarate pathway (20). Galactarate can also be catabolized through the glycerate pathway (20). The products of this pathway are pyruvate and d-glycerate. These pathways have been described in prokaryotes, and it is not certain whether similar pathways also exist in fungi, some of which are able to metabolize galactarate.d-Galacturonate dehydrogenase (EC 1.1.1.203) has been described in Agrobacterium tumefaciens and in Pseudomonas syringae, and the enzymes from these organisms have been purified and characterized (3, 6, 22). Recently, the corresponding genes were also identified (4, 24). Both enzymes are specific for NAD as a cofactor but are not specific for the substrate. They oxidize d-galacturonate and d-glucuronate to meso-galactarate (mucate) and d-glucarate (saccharate), respectively. The reaction product is probably the hexaro-lactone which spontaneously hydrolyzes. The reverse reaction can only be observed at acidic pH where some of the galactarate is in the lactone form (22).We describe here strains of filamentous fungi that have been genetically engineered to produce galactarate by disruption of d-galacturonate reductase and expression of d-galacturonate dehydrogenase (Fig. (Fig.1).1). Galactarate is currently commercially produced from d-galactose by oxidation with nitric acid (1) or from d-galacturonate by electrolytic oxidation (8). Oxidation with nitric acid is expensive and produces toxic wastes. Galactarate is used as a chelator and in skin care products. It was formerly used as a leavening agent in self-rising flour (2) and has potential applications in polymer synthesis (10) and as a platform chemical (for a review, see reference 13).Open in a separate windowFIG. 1.Engineering the d-galacturonic acid pathway in fungi. Deletion of the gene encoding d-galacturonate reductase resulted in strains unable to utilize d-galacturonic acid as a carbon source. The expression of a bacterial udh gene, encoding an NAD-dependent d-galacturonate dehydrogenase, resulted in fungal strains, which were able to oxidize d-galacturonic acid to meso-galactaric acid (mucic acid). d-Galacturonate dehydrogenase forms a galactaro-lactone which spontaneously hydrolyzes.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号