Ancient enzymes such as hydrogenase had to evolve to accommodate

Ancient enzymes such as hydrogenase had to evolve to accommodate into an O2-containing environment. From a biotechnological point of view, oxygen tolerance is a relevant Go6983 mw characteristic with obvious interest

[31]. The initial model described for the oxygen-sensitive hydrogenase from Desulfovibrio gigas[32] has been enriched by recent crystal structures of oxygen tolerant hydrogenases from Hydrogenovibrio marinus, R. eutropha, and E. coli, showing that in the case of oxygen-tolerant enzymes, the iron-sulfur cluster proximal to NiFe cofactor corresponds to an unprecedented [4Fe3S] type coordinated with six cysteines [33–35]. This cluster provides redox protection to the NiFe cofactor, by allowing the enzyme to catalyze https://www.selleckchem.com/products/pf-06463922.html reduction of O2 to water “in situ” as well as the oxidation

of hydrogen. An oxidative environment may also require protection during enzyme biosynthesis. From a genetic point of view, a relevant variation lies in the presence of two additional genes, hupF and hupK and their homologues, encoding auxiliary proteins in hydrogenase systems from aerobic bacteria. Using a specific deletion mutant we have shown in this work that HupF is essential for hydrogenase activity in R. leguminosarum, as it has been described in the R. eutropha system [20]. The results obtained here indicate that HupF has a dual role during hydrogenase biosynthesis: it is required for hydrogenase large subunit BAY 11-7082 order processing and also acts as a chaperone to stabilize HupL when hydrogenase is synthesized in the presence of oxygen. Data from experiments on exposure of HupL-containing cells to different oxygen tensions indicate that, in the absence of HupF, unprocessed HupL gradually Avelestat (AZD9668) disappears at high oxygen tensions. Since there is no P fixN -driven expression of hupL at 21% O2[18], the decrease in the level of HupL is likely due to a loss of stability of the protein. Analysis of the C-terminal deletion mutant of HupF suggests that this domain might be relevant for HupL stabilization and might provide additional support for the role of HupF as an oxygen protective chaperone. The C-terminally truncated protein is functionally indistinguishable

from the full-size protein under symbiotic, ultra-low oxygen conditions, whereas the functionality of the truncated protein is increasingly compromised in free-living cells under 1% and 3% O2. Preliminary analysis of the mutant protein indicates that it still binds HupL, although at lower level, whereas it appears as fully competent in HupK binding (data not shown). The results presented in this work indicate the exis-tence of physical interactions between HupF, HupK, and HupL during biosynthesis of the hydrogenase large subunit in R. leguminosarum. This subunit contains cysteine motifs involved in the binding of the NiFe cluster [1]. The identification of similar motifs in HupK-like proteins had led to the hypothesis of a scaffolding role for HupK similar to that of NifE protein in nitrogenase synthesis [36].

Comments are closed.