Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
Add more filters










Publication year range
1.
J Biosci Bioeng ; 137(5): 329-334, 2024 May.
Article in English | MEDLINE | ID: mdl-38461105

ABSTRACT

Hyperthermostable endoglucanases of glycoside hydrolase family 12 from the archaeon Pyrococcus furiosus (EGPf) catalyze the hydrolysis of ß-1,4-glucosidic linkages in cellulose and ß-glucan structures that contain ß-1,3- and ß-1,4-mixed linkages. In this study, EGPf was heterologously expressed with Aspergillus niger and the recombinant enzyme was characterized. The successful expression of EGPf resulted as N-glycosylated protein in its secretion into the culture medium. The glycosylation of the recombinant EGPf positively impacted the kinetic characterization of EGPf, thereby enhancing its catalytic efficiency. Moreover, glycosylation significantly boosted the thermostability of EGPf, allowing it to retain over 80% of its activity even after exposure to 100 °C for 5 h, with the optimal temperature being above 120 °C. Glycosylation did not affect the pH stability or salt tolerance of EGPf, although the glycosylated compound exhibited a high tolerance to ionic liquids. EGPf displayed the highest specific activity in the presence of 20% (v/v) 1-butyl-3-methylimidazolium chloride ([Bmim]Cl), reaching approximately 2.4 times greater activity than that in the absence of [Bmim]Cl. The specific activity was comparable to that without the ionic liquid even in the presence of 40% (v/v) [Bmim]Cl. Glycosylated EGPf has potential as an enzyme for saccharifying cellulose under high-temperature conditions or with ionic liquid treatment due to its exceptional thermostability and ionic liquid tolerance. These results underscore the potential of N-glycosylation as an effective strategy to further enhance both the thermostability of highly thermostable archaeal enzymes and the hydrolysis of barley cellulose in the presence of [Bmim]Cl.


Subject(s)
Cellulase , Ionic Liquids , Pyrococcus furiosus , Cellulase/metabolism , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Glycosylation , Cellulose/metabolism , Enzyme Stability
2.
Biochem Biophys Res Commun ; 693: 149377, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38101000

ABSTRACT

In most of the eukaryotes and archaea, isopentenyl pyrophosphate (IPP) and dimethyl allyl pyrophosphate (DMAPP) essential building blocks of all isoprenoids synthesized in the mevalonate pathway. Here, the first enzyme of this pathway, acetoacetyl CoA thiolase (PFC_04095) from an archaea Pyrococcus furiosus is structurally characterized. The crystal structure of PFC_04095 is determined at 2.7 Å resolution, and the crystal structure reveals the absence of catalytic acid/base cysteine in its active site, which is uncommon in thiolases. In place of cysteine, His285 of HDAF motif performs both protonation and abstraction of proton during the reaction. The crystal structure shows that the distance between Cys83 and His335 is 5.4 Å. So, His335 could not abstract a proton from nucleophilic cysteine (Cys83), resulting in the loss of enzymatic activity of PFC_04095. MD simulations of the docked PFC_04095-acetyl CoA complex show substrate binding instability to the active site pocket. Here, we have reported that the stable binding of acetyl CoA to the PFC_04095 pocket requires the involvement of three protein complexes, i.e., thiolase (PFC_04095), DUF35 (PFC_04100), and HMGCS (PFC_04090).


Subject(s)
Acetyl-CoA C-Acetyltransferase , Pyrococcus furiosus , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl Coenzyme A/metabolism , Pyrococcus furiosus/metabolism , Cysteine/metabolism , Protons , Models, Molecular
3.
Sci Rep ; 13(1): 16821, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37798298

ABSTRACT

Amongst the potential contribution of protein or peptide-display systems to study epitopes with relevant immunological features, the RAD display system stands out as a highly stable scaffold protein that allows the presentation of constrained target peptides. Here, we employed the RAD display system to present peptides derived from the SARS-CoV-2 Spike (S) protein as a tool to detect specific serum antibodies and to generate polyclonal antibodies capable of inhibiting SARS-CoV-2 infectivity in vitro. 44 linear S-derived peptides were genetically fused with the RAD scaffold (RAD-SCoV-epitopes) and screened for antigenicity with sera collected from COVID-19-infected patients. In a second step, selected RAD-SCoV-epitopes were used to immunize mice and generate antibodies. Phenotypic screening showed that some of these antibodies were able to recognize replicating viral particles in VERO CCL-81 and most notably seven of the RAD-SCoV-epitopes were able to induce antibodies that inhibited viral infection. Our findings highlight the RAD display system as an useful platform for the immunological characterization of peptides and a potentially valuable strategy for the design of antigens for peptide-based vaccines, for epitope-specific antibody mapping, and for the development of antibodies for diagnostic and therapeutic purposes.


Subject(s)
COVID-19 , Pyrococcus furiosus , Humans , Animals , Mice , Epitopes , Spike Glycoprotein, Coronavirus/metabolism , Pyrococcus furiosus/metabolism , Antibodies, Viral , Viral Envelope Proteins , SARS-CoV-2 , Peptides/chemistry , Antibodies, Neutralizing
4.
Biol Chem ; 404(11-12): 1085-1100, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37709673

ABSTRACT

Posttranscriptional processes in Bacteria include the association of small regulatory RNAs (sRNA) with a target mRNA. The sRNA/mRNA annealing process is often mediated by an RNA chaperone called Hfq. The functional role of bacterial and eukaryotic Lsm proteins is partially understood, whereas knowledge about archaeal Lsm proteins is scarce. Here, we used the genetically tractable archaeal hyperthermophile Pyrococcus furiosus to identify the protein interaction partners of the archaeal Sm-like proteins (PfuSmAP1) using mass spectrometry and performed a transcriptome-wide binding site analysis of PfuSmAP1. Most of the protein interaction partners we found are part of the RNA homoeostasis network in Archaea including ribosomal proteins, the exosome, RNA-modifying enzymes, but also RNA polymerase subunits, and transcription factors. We show that PfuSmAP1 preferentially binds messenger RNAs and antisense RNAs recognizing a gapped poly(U) sequence with high affinity. Furthermore, we found that SmAP1 co-transcriptionally associates with target RNAs. Our study reveals that in contrast to bacterial Hfq, PfuSmAP1 does not affect the transcriptional activity or the pausing behaviour of archaeal RNA polymerases. We propose that PfuSmAP1 recruits antisense RNAs to target mRNAs and thereby executes its putative regulatory function on the posttranscriptional level.


Subject(s)
Archaeal Proteins , Pyrococcus furiosus , RNA, Small Untranslated , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , RNA, Messenger/metabolism , RNA, Archaeal/genetics , RNA, Archaeal/chemistry , RNA, Archaeal/metabolism , Binding Sites , Bacteria/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , RNA, Small Untranslated/metabolism
5.
Appl Environ Microbiol ; 89(6): e0056323, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37289085

ABSTRACT

A genome-scale metabolic model, encompassing a total of 623 genes, 727 reactions, and 865 metabolites, was developed for Pyrococcus furiosus, an archaeon that grows optimally at 100°C by carbohydrate and peptide fermentation. The model uses subsystem-based genome annotation, along with extensive manual curation of 237 gene-reaction associations including those involved in central carbon metabolism, amino acid metabolism, and energy metabolism. The redox and energy balance of P. furiosus was investigated through random sampling of flux distributions in the model during growth on disaccharides. The core energy balance of the model was shown to depend on high acetate production and the coupling of a sodium-dependent ATP synthase and membrane-bound hydrogenase, which generates a sodium gradient in a ferredoxin-dependent manner, aligning with existing understanding of P. furiosus metabolism. The model was utilized to inform genetic engineering designs that favor the production of ethanol over acetate by implementing an NADPH and CO-dependent energy economy. The P. furiosus model is a powerful tool for understanding the relationship between generation of end products and redox/energy balance at a systems-level that will aid in the design of optimal engineering strategies for production of bio-based chemicals and fuels. IMPORTANCE The bio-based production of organic chemicals provides a sustainable alternative to fossil-based production in the face of today's climate challenges. In this work, we present a genome-scale metabolic reconstruction of Pyrococcus furiosus, a well-established platform organism that has been engineered to produce a variety of chemicals and fuels. The metabolic model was used to design optimal engineering strategies to produce ethanol. The redox and energy balance of P. furiosus was examined in detail, which provided useful insights that will guide future engineering designs.


Subject(s)
Pyrococcus furiosus , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Ethanol/metabolism , Fermentation , Genetic Engineering , Acetates/metabolism
6.
Appl Environ Microbiol ; 89(6): e0001223, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37162365

ABSTRACT

Genetic engineering of hyperthermophilic organisms for the production of fuels and other useful chemicals is an emerging biotechnological opportunity. In particular, for volatile organic compounds such as ethanol, fermentation at high temperatures could allow for straightforward separation by direct distillation. Currently, the upper growth temperature limit for native ethanol producers is 72°C in the bacterium Thermoanaerobacter ethanolicus JW200, and the highest temperature for heterologously-engineered bioethanol production was recently demonstrated at 85°C in the archaeon Pyrococcus furiosus. Here, we describe an engineered strain of P. furiosus that synthesizes ethanol at 95°C, utilizing a homologously-expressed native alcohol dehydrogenase, termed AdhF. Ethanol biosynthesis was compared at 75°C and 95°C with various engineered strains. At lower temperatures, the acetaldehyde substrate for AdhF is most likely produced from acetate by aldehyde ferredoxin oxidoreductase (AOR). At higher temperatures, the effect of AOR on ethanol production is negligible, suggesting that acetaldehyde is produced by pyruvate ferredoxin oxidoreductase (POR) via oxidative decarboxylation of pyruvate, a reaction known to occur only at higher temperatures. Heterologous expression of a carbon monoxide dehydrogenase complex in the AdhF overexpression strain enabled it to use CO as a source of energy, leading to increased ethanol production. A genome reconstruction model for P. furiosus was developed to guide metabolic engineering strategies and understand outcomes. This work opens the door to the potential for 'bioreactive distillation' since fermentation can be performed well above the normal boiling point of ethanol. IMPORTANCE Previously, the highest temperature for biological ethanol production was 85°C. Here, we have engineered ethanol production at 95°C by the hyperthermophilic archaeon Pyrococcus furiosus. Using mutant strains, we showed that ethanol production occurs by different pathways at 75°C and 95°C. In addition, by heterologous expression of a carbon monoxide dehydrogenase complex, ethanol production by this organism was driven by the oxidation of carbon monoxide. A genome reconstruction model for P. furiosus was developed to guide metabolic engineering strategies and understand outcomes.


Subject(s)
Pyrococcus furiosus , Fermentation , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Carbon Monoxide/metabolism , Ethanol/metabolism , Metabolic Engineering , Pyruvic Acid/metabolism , Acetaldehyde/metabolism
7.
Molecules ; 28(10)2023 May 11.
Article in English | MEDLINE | ID: mdl-37241758

ABSTRACT

The development of tungsten biochemistry is sketched from the viewpoint of personal participation. Following its identification as a bio-element, a catalogue of genes, enzymes, and reactions was built up. EPR spectroscopic monitoring of redox states was, and remains, a prominent tool in attempts to understand tungstopterin-based catalysis. A paucity of pre-steady-state data remains a hindrance to overcome to this day. Tungstate transport systems have been characterized and found to be very specific for W over Mo. Additional selectivity is presented by the biosynthetic machinery for tungstopterin enzymes. Metallomics analysis of hyperthermophilic archaeon Pyrococcus furiosus indicates a comprehensive inventory of tungsten proteins.


Subject(s)
Aldehyde Oxidoreductases , Pyrococcus furiosus , Aldehyde Oxidoreductases/genetics , Tungsten/chemistry , Oxidation-Reduction , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism
8.
Viruses ; 15(3)2023 02 21.
Article in English | MEDLINE | ID: mdl-36992305

ABSTRACT

Parvovirus B19 (B19V) is pathogenic to humans and causes various human diseases. However, no antiviral agents or vaccines currently exist for the treatment or prevention of B19V infection. Therefore, developing sensitive and specific methods for B19V infection diagnosis is essential for accurate diagnoses. Previously, a Clustered Regularly Interspaced Palindromic Repeats (CRISPR)-Cas12a (cpf1)-based electrochemical biosensor (E-CRISPR) with a picomole sensitivity for B19V detection was established. Herein, we set up a novel nucleic acid detection system based on Pyrococcus furiosus Argonaute (PfAgo)-mediated nucleic acid detection, targeting the nonstructural protein 1 (NS1) region of the B19V viral genome (abbreviated B19-NS1 PAND). Benefiting from independent protospacer adjacent motif (PAM) sequences, PfAgo can recognize their target with guide DNA (gDNA) that is easy to design and synthesize at a low cost. In contrast to E-CRISPR, without preamplification with Polymerase Chain Reaction (PCR), the Minimum Detectable Concentration (MDC) of three guide- or single guide-mediated B19-NS1 PAND was about 4 nM, approximately 6-fold more than E-CRISPR. However, when introducing an amplification step, the MDC can be dramatically decreased to the aM level (54 aM). In addition, the diagnostic results from clinical samples with B19-NS1 PAND revealed 100% consistency with PCR assays and subsequent Sanger sequencing tests, which may assist in molecular testing for clinical diagnosis and epidemiological investigations of B19V.


Subject(s)
Parvoviridae Infections , Parvovirus B19, Human , Pyrococcus furiosus , Humans , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Argonaute Proteins/genetics , DNA/metabolism , Real-Time Polymerase Chain Reaction/methods , DNA, Viral/genetics , DNA, Viral/metabolism
9.
Carbohydr Res ; 522: 108682, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36194965

ABSTRACT

Glycoside hydrolase family 12 endocellulase (GH family12) plays a key role in the degradation of ß-glucan and cellulose. Hyperthermostable GH family 12 endocellulase from the archaeon Pyrococcus furiosus (EGPf) catalyzes the hydrolysis of ß(1 â†’ 4) glucosidic linkages in cellulose and ß-glucan containing ß(1 â†’ 3),(1 â†’ 4) mixed-linkages. Therefore, in the combination with the hyperthermophilic ß-glucosidase from P. furiosus (BGLPf), non-crystalline cellulose and ß-glucan can be degraded to glucose completely by EGPf at high temperature. X-ray crystallography and protein engineering were used to reveal how the ß(1 â†’ 4) and ß(1 â†’ 3) linkages in ß-glucan substrates are recognized by the enzyme. Structural and functional analyses clarified that the active site of EGPf consists of six subsites: the reducing end subsites (+1 and + 2) recognize both ß(1 â†’ 4) and ß(1 â†’ 3) linkages of various substrates in a productive binding mode, and recognition is controlled by Trp121 and Gln208 located at subsite +2. It was also revealed that the deep cleft in subsite -4 can accommodate the torsion angles of substrates consisting of ß(1 â†’ 3),(1 â†’ 4) mixed-linkages due to the changing tilt of the Trp62 side chain. From the structural similarity, it is proposed that the substrate specificity of family 12 endocellulases towards ß(1 â†’ 3),(1 â†’ 4) mixed-linkage substrates are controlled by the subsites (+1, +2, and -4). Furthermore, the function of family 12 endocellulase could be improved by protein engineering method using the information of the analysis.


Subject(s)
Pyrococcus furiosus , beta-Glucans , Pyrococcus furiosus/metabolism , Crystallography, X-Ray , beta-Glucosidase/metabolism , Substrate Specificity , Cellulose/chemistry
10.
J Biol Inorg Chem ; 27(8): 747-758, 2022 12.
Article in English | MEDLINE | ID: mdl-36269456

ABSTRACT

Five tungstopterin-containing oxidoreductases were characterized from the hyperthermophile Pyrococcus furiosus. Each enzyme catalyzes the reversible conversion of one or more aldehydes to the corresponding carboxylic acid, but they have different specificities. The physiological functions of only two of these enzymes are known: one, termed GAPOR, is a glycolytic enzyme that oxidizes glyceraldehyde-3-phosphate, while the other, termed AOR, oxidizes multiple aldehydes generated during peptide fermentation. Two of the enzymes have known structures (AOR and FOR). Herein, we focus on WOR5, the fifth tungstopterin enzyme to be discovered in P. furiosus. Expression of WOR5 was previously shown to be increased during cold shock (growth at 72 â„ƒ), although the physiological substrate is not known. To gain insight into WOR5 function, we sought to determine both its structure and identify its intracellular substrate. Crystallization experiments were performed with a concentrated cytoplasmic extract of P. furiosus grown at 72 â„ƒ and the structure of WOR5 was deduced from the crystals that were obtained. In contrast to a previous report, WOR5 is heterodimeric containing an additional polyferredoxin-like subunit with four [4Fe-4S] clusters. The active site structure of WOR5 is substantially different from that of AOR and FOR and the significant electron density observed adjacent to the tungsten cofactor of WOR5 was modeled as an aliphatic sulfonate. Biochemical assays and product analysis confirmed that WOR5 is an aliphatic sulfonate ferredoxin oxidoreductase (ASOR). A catalytic mechanism for ASOR is proposed based on the structural information and the potential role of ASOR in the cold-shock response is discussed.


Subject(s)
Pyrococcus furiosus , Tungsten , Tungsten/chemistry , Oxidoreductases/metabolism , Aldehyde Oxidoreductases/metabolism , Pyrococcus furiosus/metabolism , Aldehydes/metabolism
11.
Biotechnol Bioeng ; 119(9): 2388-2398, 2022 09.
Article in English | MEDLINE | ID: mdl-35661137

ABSTRACT

Prokaryotic Argonaute proteins (pAgos) play an important role in host defense against invading genetic elements. The functional diversities make pAgos very promising in development of novel nucleic acid manipulation tools and attract increasing attentions. Here, we reported the in vitro characterization of an Argonaute protein from archaeon Thermococcus thioreducens (TtrAgo) and its example of application in hepatitis B virus DNA detection. The results showed that TtrAgo functions as a programmable DNA endonuclease by utilizing both short 5'-phosphorylated and 5'-hydroxylated single-stranded DNA guides, and presents high efficiency and accuracy at optimal temperatures ranging from 75°C to 95°C. In addition, TtrAgo also possesses stepwise cleavage activity like PfAgo (Pyrococcus furiosus) and chopping activity toward double-stranded DNA similar to MjAgo (Methanocaldococcus jannaschii). This study increases our understanding of pAgos and expands the Ago-based DNA detection toolbox.


Subject(s)
Pyrococcus furiosus , Thermococcus , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , DNA/metabolism , Methanocaldococcus/genetics , Pyrococcus furiosus/metabolism , Thermococcus/genetics , Thermococcus/metabolism
12.
Biochem Biophys Res Commun ; 593: 116-121, 2022 02 19.
Article in English | MEDLINE | ID: mdl-35063766

ABSTRACT

Ribosome dimerization is one of the bacterial events that suppresses protein synthesis in the stationary phase. Protein factors responsible for ribosome dimerization in bacteria are well characterized, whereas no information is available for the corresponding factors in archaeal and eukaryotic cells. Here we describe a protein found among the ribosome-associated proteins which dimerizes the 30S ribosomal subunit of the archaeon Pyrococcus furiosus. The ribosome-associated proteins were prepared by high-salt wash of crude ribosomes, and analyzed by nanoflow liquid chromatography-tandem mass spectrometry (nano LC-MS/MS). Of the detected proteins we focused on a protein (PF0560) whose Protein Score was the highest of all of the function-unknown proteins. PF0560 protein had a pronounced effect on the sedimentation pattern of the 30S ribosomal subunit; addition of this protein to isolated 30S subunit reduced the 30S fraction and increased the amount of the 50S fraction. This increase presumably corresponds to the dimer of the 30S subunit. The PF0560-dependent 30S-dimerization, was also observed by gel electrophoretic analysis. This effect was not observed in EDTA-treated 30S subunit, with protein-free 16S rRNA or with bacterial/eukaryotic ribosomal small subunits. Furthermore, PF0560 protein suppressed the formation of functional 70S ribosomes. These results suggest that PF0560 is a novel 30S dimerization factor, which might participate in regulation of archaeal translation.


Subject(s)
Archaeal Proteins/metabolism , Dimerization , Proteome/metabolism , Pyrococcus furiosus/metabolism , RNA, Ribosomal, 16S/chemistry , Ribosomal Proteins/metabolism , Ribosomes/chemistry , Amino Acid Sequence , Archaeal Proteins/genetics , Magnesium/chemistry , Proteome/analysis , Pyrococcus furiosus/genetics , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Ribosomal Proteins/genetics , Sequence Homology
13.
Int J Biochem Cell Biol ; 144: 106171, 2022 03.
Article in English | MEDLINE | ID: mdl-35093572

ABSTRACT

B-family DNA polymerases, which are found in eukaryotes, archaea, viruses, and some bacteria, participate in DNA replication and repair. Starting from the N-terminus of archaeal and bacterial B-family DNA polymerases, three domains include the N-terminal, exonuclease, and polymerase domains. The N-terminal domain of the archaeal B-family DNA polymerase has a conserved deoxyuracil-binding pocket for specially binding the deoxyuracil base on DNA. The exonuclease domain is responsible for removing the mismatched base pair. The polymerase domain is the core functional domain and takes a highly conserved structure composed of fingers, palm and thumb subdomains. Previous studies have demonstrated that the thumb subdomain mainly functions as a DNA-binding element and has coordination with the exonuclease domain and palm subdomain. To further elucidate the possible functions of the thumb subdomain of archaeal B-family DNA polymerases, the thumb subdomain of Pyrococcus furiosus DNA polymerase was mutated, and the effects on three activities were characterized. Our results demonstrate that the thumb subdomain participates in the three activities of archaeal B-family DNA polymerases as a common structural element. Both the N-terminal deoxyuracil-binding pocket and thumb subdomain are critical for deoxyuracil binding. Moreover, the thumb subdomain assists DNA polymerization and hydrolysis reactions, but it does not contribute to the fidelity of DNA polymerization.


Subject(s)
Pyrococcus furiosus , Amino Acid Sequence , DNA/metabolism , DNA Polymerase I/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Exonucleases/chemistry , Exonucleases/metabolism , Hydrolysis , Models, Molecular , Nucleotides , Polymerization , Protein Structure, Tertiary , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Thumb
14.
J Biol Chem ; 296: 100710, 2021.
Article in English | MEDLINE | ID: mdl-33930466

ABSTRACT

Pyrococcus furiosus is a hyperthermophilic anaerobic archaeon whose metabolism depends on whether elemental sulfur is (+S0) or is not (-S0) included in growth medium. Under +S0 conditions, expression of respiratory hydrogenase declines while respiratory membrane-bound sulfane reductase and the putative iron-storage protein IssA increase. Our objective was to investigate the iron content of WT and ΔIssA cells under these growth conditions using Mössbauer spectroscopy. WT-S0 cells contained ∼1 mM Fe, with ∼85% present as two spectroscopically distinct forms of S = 0 [Fe4S4]2+ clusters; the remainder was mainly high-spin FeII. WT+S0 cells contained 5 to 9 mM Fe, with 75 to 90% present as magnetically ordered thioferrate-like (TFL) iron nanoparticles. TFL iron was similar to chemically defined thioferrates; both consisted of FeIII ions coordinated by an S4 environment, and both exhibited strong coupling between particles causing high applied fields to have little spectral effect. At high temperatures with magnetic hyperfine interactions abolished, TFL iron exhibited two doublets overlapping those of [Fe4S4]2+ clusters in -S0 cells. This coincidence arose because of similar coordination environments of TFL iron and cluster iron. The TFL structure was more heterogeneous in the presence of IssA. Presented data suggest that IssA may coordinate insoluble iron sulfides as TFL iron, formed as a byproduct of anaerobic sulfur respiration under high iron conditions, which thereby reduces its toxicity to the cell. This was the first Mössbauer characterization of the ironome of an archaeon, and it illustrates differences relative to the iron content of better-studied bacteria such as Escherichia coli.


Subject(s)
Iron/metabolism , Pyrococcus furiosus/metabolism , Sulfur/metabolism , Hydrogenase/metabolism , Oxidation-Reduction
15.
RNA ; 27(4): 496-512, 2021 04.
Article in English | MEDLINE | ID: mdl-33483369

ABSTRACT

Ribosomal RNA (rRNA) carries extensive 2'-O-methyl marks at functionally important sites. This simple chemical modification is thought to confer stability, promote RNA folding, and contribute to generate a heterogenous ribosome population with a yet-uncharacterized function. 2'-O-methylation occurs both in archaea and eukaryotes and is accomplished by the Box C/D RNP enzyme in an RNA-guided manner. Extensive and partially conflicting structural information exists for the archaeal enzyme, while no structural data is available for the eukaryotic enzyme. The yeast Box C/D RNP consists of a guide RNA, the RNA-primary binding protein Snu13, the two scaffold proteins Nop56 and Nop58, and the enzymatic module Nop1. Here we present the high-resolution structure of the eukaryotic Box C/D methyltransferase Nop1 from Saccharomyces cerevisiae bound to the amino-terminal domain of Nop56. We discuss similarities and differences between the interaction modes of the two proteins in archaea and eukaryotes and demonstrate that eukaryotic Nop56 recruits the methyltransferase to the Box C/D RNP through a protein-protein interface that differs substantially from the archaeal orthologs. This study represents a first achievement in understanding the evolution of the structure and function of these proteins from archaea to eukaryotes.


Subject(s)
Archaeal Proteins/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Nuclear Proteins/chemistry , Pyrococcus furiosus/genetics , Ribonucleoproteins, Small Nucleolar/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Binding Sites , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , Gene Expression , Methylation , Models, Molecular , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Pyrococcus furiosus/metabolism , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Structural Homology, Protein
16.
Proteins ; 89(2): 251-255, 2021 02.
Article in English | MEDLINE | ID: mdl-32875643

ABSTRACT

The Rad50-Mre11 nuclease complex plays a vital role in DNA repair in all domains of life. It recognizes and processes DNA double-strand breaks. Rad50 proteins fold into an extended structure with a 20 to 60 nm long coiled coil connecting a globular ABC ATPase domain with a zinc hook dimerization domain. A published structure of an archaeal Rad50 zinc hook shows coiled coils pointing away from each other. Here we present the crystal structure of an alternate conformation displaying co-aligned coiled coils. Archaeal Rad50 may thus switch between rod-shaped and ring-like conformations as recently proposed for a bacterial homolog.


Subject(s)
Archaeal Proteins/chemistry , DNA Repair , DNA, Archaeal/chemistry , Endodeoxyribonucleases/chemistry , Exodeoxyribonucleases/chemistry , Pyrococcus furiosus/genetics , Zinc/chemistry , Amino Acid Motifs , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Binding Sites , Cations, Divalent , Cloning, Molecular , Crystallography, X-Ray , DNA, Archaeal/genetics , DNA, Archaeal/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Pyrococcus furiosus/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Zinc/metabolism
17.
RNA Biol ; 18(9): 1300-1309, 2021 09.
Article in English | MEDLINE | ID: mdl-33111609

ABSTRACT

H/ACA ribonucleoproteins catalyse the sequence-dependent pseudouridylation of ribosomal and spliceosomal RNAs. Here, we reconstitute site-specifically fluorophore labelled H/ACA complexes and analyse their structural dynamics using single-molecule FRET spectroscopy. Our results show that the guide RNA is distorted into a substrate-binding competent conformation by specific protein interactions. Analysis of the reaction pathway using atomic mutagenesis establishes a new model how individual protein domains contribute to catalysis. Taken together, these results identify and characterize individual roles for all accessory proteins on the assembly and function of H/ACA RNPs.


Subject(s)
Archaeal Proteins/metabolism , Pseudouridine/metabolism , Pyrococcus furiosus/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Nucleolar/metabolism , Ribonucleoproteins, Small Nucleolar/metabolism , Archaeal Proteins/genetics , Base Pairing , Catalysis , Pseudouridine/genetics , Pyrococcus furiosus/genetics , RNA, Guide, Kinetoplastida/genetics , RNA, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/genetics , Spliceosomes
18.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33372143

ABSTRACT

The electron-transferring flavoprotein-menaquinone oxidoreductase ABCX (EtfABCX), also known as FixABCX for its role in nitrogen-fixing organisms, is a member of a family of electron-transferring flavoproteins that catalyze electron bifurcation. EtfABCX enables endergonic reduction of ferredoxin (E°' ∼-450 mV) using NADH (E°' -320 mV) as the electron donor by coupling this reaction to the exergonic reduction of menaquinone (E°' -80 mV). Here we report the 2.9 Å structure of EtfABCX, a membrane-associated flavin-based electron bifurcation (FBEB) complex, from a thermophilic bacterium. EtfABCX forms a superdimer with two membrane-associated EtfCs at the dimer interface that contain two bound menaquinones. The structure reveals that, in contrast to previous predictions, the low-potential electrons bifurcated from EtfAB are most likely directly transferred to ferredoxin, while high-potential electrons reduce the quinone via two [4Fe-4S] clusters in EtfX. Surprisingly, EtfX shares remarkable structural similarity with mammalian [4Fe-4S] cluster-containing ETF ubiquinone oxidoreductase (ETF-QO), suggesting an unexpected evolutionary link between bifurcating and nonbifurcating systems. Based on this structure and spectroscopic studies of a closely related EtfABCX, we propose a detailed mechanism of the catalytic cycle and the accompanying structural changes in this membrane-associated FBEB system.


Subject(s)
Electron-Transferring Flavoproteins/metabolism , Quinone Reductases/metabolism , Quinone Reductases/ultrastructure , Bacterial Proteins/metabolism , Catalysis , Cryoelectron Microscopy/methods , Electron Transport , Electrons , Ferredoxins/metabolism , Flavins/metabolism , Flavoproteins/metabolism , Iron-Sulfur Proteins/metabolism , Models, Molecular , NAD/metabolism , Nitrogen Fixation/physiology , Oxidation-Reduction , Pyrococcus furiosus/metabolism , Quinone Reductases/physiology , Vitamin K 2/metabolism
19.
Appl Environ Microbiol ; 86(21)2020 10 15.
Article in English | MEDLINE | ID: mdl-32859593

ABSTRACT

Arsenate is a notorious toxicant that is known to disrupt multiple biochemical pathways. Many microorganisms have developed mechanisms to detoxify arsenate using the ArsC-type arsenate reductase, and some even use arsenate as a terminal electron acceptor for respiration involving arsenate respiratory reductase (Arr). ArsC-type reductases have been studied extensively, but the phylogenetically unrelated Arr system is less investigated and has not been characterized from Archaea Here, we heterologously expressed the genes encoding Arr from the crenarchaeon Pyrobaculum aerophilum in the euryarchaeon Pyrococcus furiosus, both of which grow optimally near 100°C. Recombinant P. furiosus was grown on molybdenum (Mo)- or tungsten (W)-containing medium, and two types of recombinant Arr enzymes were purified, one containing Mo (Arr-Mo) and one containing W (Arr-W). Purified Arr-Mo had a 140-fold higher specific activity in arsenate [As(V)] reduction than Arr-W, and Arr-Mo also reduced arsenite [As(III)]. The P. furiosus strain expressing Arr-Mo (the Arr strain) was able to use arsenate as a terminal electron acceptor during growth on peptides. In addition, the Arr strain had increased tolerance compared to that of the parent strain to arsenate and also, surprisingly, to arsenite. Compared to the parent, the Arr strain accumulated intracellularly almost an order of magnitude more arsenic when cells were grown in the presence of arsenite. X-ray absorption spectroscopy (XAS) results suggest that the Arr strain of P. furiosus improves its tolerance to arsenite by increasing production of less-toxic arsenate and nontoxic methylated arsenicals compared to that by the parent.IMPORTANCE Arsenate respiratory reductases (Arr) are much less characterized than the detoxifying arsenate reductase system. The heterologous expression and characterization of an Arr from Pyrobaculum aerophilum in Pyrococcus furiosus provides new insights into the function of this enzyme. From in vivo studies, production of Arr not only enabled P. furiosus to use arsenate [As(V)] as a terminal electron acceptor, it also provided the organism with a higher resistance to arsenate and also, surprisingly, to arsenite [As(III)]. In contrast to the tungsten-containing oxidoreductase enzymes natively produced by P. furiosus, recombinant P. aerophilum Arr was much more active with molybdenum than with tungsten. It is also, to our knowledge, the only characterized Arr to be active with both molybdenum and tungsten in the active site.


Subject(s)
Archaeal Proteins/genetics , Arsenate Reductases/genetics , Gene Expression Regulation, Archaeal , Pyrococcus furiosus/genetics , Thermoproteaceae/genetics , Archaeal Proteins/metabolism , Arsenate Reductases/metabolism , Arsenic/metabolism , Microorganisms, Genetically-Modified/enzymology , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/metabolism , Pyrococcus furiosus/enzymology , Pyrococcus furiosus/metabolism
20.
FEBS Lett ; 594(20): 3338-3355, 2020 10.
Article in English | MEDLINE | ID: mdl-32805768

ABSTRACT

Inteins catalyze self-excision from host precursor proteins while concomitantly ligating the flanking substrates (exteins) with a peptide bond. Noncatalytic extein residues near the splice junctions, such as the residues at the -1 and +2 positions, often strongly influence the protein-splicing efficiency. The substrate specificities of inteins have not been studied for many inteins. We developed a convenient mutagenesis platform termed "QuickDrop"-cassette mutagenesis for investigating the influences of 20 amino acid types at the -1 and +2 positions of different inteins. We elucidated 17 different profiles of the 20 amino acid dependencies across different inteins. The substrate specificities will accelerate our understanding of the structure-function relationship at the splicing junctions for broader applications of inteins in biotechnology and molecular biosciences.


Subject(s)
Inteins/genetics , Mutagenesis, Insertional/methods , Amino Acid Sequence , Conserved Sequence , Gene Library , Models, Molecular , Plasmids/genetics , Protein Splicing , Pyrococcus furiosus/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...