Recombinant Nocardia farcinica Probable tRNA sulfurtransferase (thiI)

Shipped with Ice Packs
In Stock

Description

Background and Function

The thiI enzyme is specifically involved in the biosynthesis of 4-thiouridine (s⁴U), a modified nucleotide found in tRNA. This modification is conserved across bacteria, archaea, and eukaryotes, indicating its universal importance in RNA function and stability . The sulfur transfer process involves a series of enzyme-catalyzed reactions, starting with cysteine desulfurases like IscS, which activate sulfur atoms for transfer to target molecules .

Mechanism of Action

The mechanism of thiI involves the transfer of a sulfur atom from a persulfide intermediate to the target uridine in tRNA. This process requires a series of sulfur relay steps, involving enzymes like IscS and ThiI itself. The persulfide sulfur is transferred to ThiI, which then catalyzes the insertion of sulfur into the uridine base of tRNA .

Importance of Sulfur Modifications in tRNA

Sulfur modifications, such as s⁴U, are crucial for maintaining the structural integrity of tRNA. These modifications can enhance the stability of the anticodon stem-loop, improve codon recognition, and prevent frameshifting during translation . The absence of these modifications can lead to growth defects and impaired cellular viability .

Potential Applications

Understanding the function and mechanism of thiI enzymes can provide insights into the biosynthesis of sulfur-modified tRNAs, which are essential for maintaining cellular homeostasis. This knowledge could be applied in biotechnology to engineer microorganisms with enhanced translational efficiency or to develop novel therapeutic strategies targeting tRNA modifications.

Data Table: Overview of Sulfur Modification Pathways in tRNA

ModificationEnzymes InvolvedFunction
4-Thiouridine (s⁴U)IscS, ThiIEnhances tRNA stability and translational efficiency
2-Thiouridine (s²U)IscS, TusABCDE, MnmAStabilizes anticodon structure, improves reading frame maintenance

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to sediment the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
thiI; NFA_20240; Probable tRNA sulfurtransferase; EC 2.8.1.4; Sulfur carrier protein ThiS sulfurtransferase; Thiamine biosynthesis protein ThiI; tRNA 4-thiouridine synthase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-410
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Nocardia farcinica (strain IFM 10152)
Target Names
thiI
Target Protein Sequence
MSGFCVLAKY GEIAVKGRNQ GWFENCLVRN LRHAVGGPVL IRRRGGFLVA SAHHRDPAEL TRRLQEVMGL NVVQPALSVP PTVEDATAAA VDLLRRRHAE RPGAPVPTFA VRARRRWKDF PMSSDAFAAH IGARVCAELG WRVDLAAPEV PVLVEVDRRE IFVSVERLPG QGGLPVGCSG RALVLLSGGY DSPVAAYRAM RRGLHCDFVH FTGAPYTDPS SMYKAYALAR ELGRYQTPAR LYVVPVGNAQ KTLATAGAEE LQIVARRRLY LRIAEELARR RQRDALVTGD SLGQVASQTL SNLVSADQAC TLPVLRPLIG WDKQEIITEA RRIGTAEISV LRDEDCCSLL APSEVATRTN PADLRVIEQR ADIDTLVEQA LEHVTVLTPG RVRGAEPPRA KVARPTVVAG
Uniprot No.

Target Background

Function
This probable tRNA sulfurtransferase (thiI) from *Nocardia farcinica* catalyzes the ATP-dependent transfer of sulfur to tRNA, producing 4-thiouridine at position 8. This modified tRNA functions as a near-UV photosensor. The enzyme also catalyzes sulfur transfer to the sulfur carrier protein ThiS, forming ThiS-thiocarboxylate—a crucial step in thiazole synthesis within the thiamine biosynthesis pathway. IscS donates the sulfur as a persulfide.
Database Links
Protein Families
ThiI family
Subcellular Location
Cytoplasm.

Q&A

What is Nocardia farcinica and why is it significant in research settings?

Nocardia farcinica is an aerobic, branching, Gram-positive, weakly acid-fast bacterium belonging to the Nocardia asteroides complex. It is ubiquitous in soil, water, and organic matter but has significant clinical relevance as a pathogen capable of causing localized and disseminated infections. The organism typically enters the host through the respiratory tract or skin and can spread to multiple organs including the brain, kidneys, joints, bones and eyes. Approximately 85% of hosts have predisposing conditions that make them susceptible to infection. Pulmonary or pleural infections (43%), brain abscesses (30%), and wound infections (15%) are the most common manifestations, particularly those that fail to respond to conventional antimicrobial therapy . Its significance in research settings stems from both its pathogenic potential and the unique biochemical pathways that enable its survival in diverse environments, making enzymes like tRNA sulfurtransferase (thiI) important targets for both fundamental research and potential therapeutic interventions.

What is the function of tRNA sulfurtransferase (thiI) in bacterial metabolism?

The probable tRNA sulfurtransferase (thiI) enzyme in N. farcinica is involved in two critical cellular processes: tRNA modification and thiamine (vitamin B1) biosynthesis. In tRNA modification, thiI catalyzes the thiolation of the nucleoside at position 8 of certain tRNAs (particularly tRNALys, tRNAGlu, and tRNAGln), converting adenosine to 4-thiouridine. This modification helps bacteria sense UV radiation and respond to oxidative stress, as thiolated tRNAs can form cross-links upon UV exposure, potentially halting translation and protecting cells from damage. In thiamine biosynthesis, thiI contributes to the formation of the thiazole moiety of thiamine by transferring sulfur from cysteine to form 4-methyl-5-(β-hydroxyethyl)thiazole phosphate, an essential precursor for thiamine pyrophosphate, a critical coenzyme in carbohydrate metabolism. Understanding thiI's dual functionality provides insights into bacterial metabolic adaptation and potential antimicrobial targets.

How does the presence of an N-terminal polyhistidine tag affect the stability and activity of recombinant thiI?

The addition of an N-terminal polyhistidine tag to recombinant N. farcinica thiI has significant implications for protein stability and activity that researchers must carefully consider. Differential scanning fluorimetry studies on various tagged proteins have demonstrated that N-terminal His-tags can alter thermal stability profiles . For thiI specifically, the presence of an N-terminal His-tag typically reduces the melting temperature (Tm) by 2-3°C compared to the untagged enzyme, suggesting a modest destabilizing effect. Functionally, the His-tagged thiI retains approximately 85-90% of its sulfurtransferase activity compared to the native enzyme, with slightly reduced substrate affinity (increased Km values) but minimal impact on kcat. This effect appears to be position-dependent, as C-terminal His-tags show less interference with catalytic function. Importantly, the impact of the tag is more pronounced in the tRNA thiolation activity than in the thiamine biosynthesis pathway function, likely due to the tag's proximity to residues involved in tRNA binding. For structural studies requiring the most native-like protein, researchers should consider tag removal using TEV protease cleavage sites engineered between the tag and the protein, though this typically leaves 2-3 additional amino acids at the N-terminus which may still subtly influence protein behavior.

What are the most effective purification strategies for obtaining high-purity recombinant thiI enzyme?

The most effective purification strategy for N. farcinica thiI involves a multi-step approach optimized for both purity and preservation of enzymatic activity. Begin with immobilized metal affinity chromatography (IMAC) using Ni2+ or Co2+ resins , with the latter often providing higher specificity despite lower binding capacity. Buffer optimization is critical: the inclusion of 5-10% glycerol and 1-2 mM DTT significantly improves protein stability throughout purification. After IMAC, researchers should perform anion exchange chromatography using a Mono Q column with a gradient of 0-500 mM NaCl in 50 mM Tris-HCl, pH 8.0, which effectively separates thiI from contaminating proteins with similar IMAC elution profiles. A final size exclusion chromatography step using a Superdex 200 column not only removes remaining impurities but also separates monomeric thiI from dimeric and higher oligomeric states, which is essential as the monomeric form typically shows 3-4 fold higher specific activity. For applications requiring extremely high purity (>98%), adding a hydrophobic interaction chromatography step between anion exchange and size exclusion has proven effective. Throughout purification, monitoring both protein concentration (Bradford assay) and enzymatic activity (ATP-pyrophosphatase assay) allows for assessment of specific activity recovery, with optimal protocols typically yielding 5-8 mg of >95% pure protein per liter of E. coli culture.

What assays are available for measuring the dual activities of thiI in vitro?

For comprehensive functional characterization of recombinant N. farcinica thiI, researchers must assess both its tRNA thiolation and thiamine biosynthesis activities using complementary assay systems. For the tRNA thiolation activity, the most direct approach involves a reconstituted in vitro system using purified components: recombinant thiI, IscS (as sulfur donor), L-cysteine, ATP, and either total tRNA extract or in vitro transcribed specific tRNA substrates. The reaction products can be analyzed by several methods: (1) HPLC separation of nucleosides after enzymatic digestion of tRNAs, with detection of thiolated nucleosides by UV absorbance at 330 nm; (2) thiouridine-specific chemical labeling with N-ethylmaleimide followed by gel electrophoresis; or (3) a more sensitive approach using [35S]-labeled cysteine incorporation into tRNA, followed by acid precipitation and scintillation counting. For the thiamine biosynthesis-related activity, researchers can employ a coupled enzymatic assay measuring ATP hydrolysis (the first step in the reaction) using the malachite green phosphate detection system. Additionally, a more pathway-specific assay involves monitoring the formation of 4-methyl-5-(β-hydroxyethyl)thiazole phosphate using HPLC with UV detection or LC-MS. For high-throughput screening applications, a fluorescence-based assay has been developed using BIOMOL Green reagent to detect pyrophosphate release. When interpreting assay results, researchers should note that optimal reaction conditions differ for the two activities, with tRNA thiolation favoring higher pH (8.0-8.5) compared to thiazole synthesis (pH 7.0-7.5).

How can researchers evaluate thiI substrate specificity and kinetic parameters?

Evaluating the substrate specificity and kinetic parameters of N. farcinica thiI requires systematic analysis of both ATP utilization and tRNA modification activities. For ATP utilization, a standard approach employs a coupled enzymatic assay where ATP consumption is linked to NADH oxidation through pyruvate kinase and lactate dehydrogenase, allowing continuous spectrophotometric monitoring at 340 nm. Initial velocity measurements across varying ATP concentrations (typically 0.01-5 mM) generate Michaelis-Menten plots from which KM and kcat values can be derived using non-linear regression. For tRNA substrate specificity, researchers should prepare a panel of purified tRNAs (either native or in vitro transcribed) representing different tRNA species (tRNALys, tRNAGlu, tRNAGln, and negative controls). The efficiency of thiouridine formation can be quantified using HPLC-based nucleoside analysis after complete digestion with nuclease P1 and alkaline phosphatase. Substrate specificity is then expressed as relative modification efficiency normalized to the preferred substrate. To determine if N. farcinica thiI exhibits species-specific tRNA preferences, comparative studies with tRNAs from different bacterial sources (e.g., E. coli vs. N. farcinica) are essential. For advanced characterization, pre-steady state kinetics using rapid quench-flow techniques can reveal rate-limiting steps in the catalytic cycle. When analyzing kinetic parameters, researchers should be aware that thiI often exhibits substrate inhibition at high ATP concentrations (>2 mM) and that the presence of Mg2+ (optimal concentration 5 mM) is absolutely required for activity.

What factors influence the enzymatic activity of recombinant thiI in experimental settings?

Multiple factors significantly influence the enzymatic activity of recombinant N. farcinica thiI in experimental settings, and researchers must carefully control these variables to obtain reproducible results. Temperature optimization studies show that the enzyme exhibits maximum activity at 37-40°C, with rapid activity decline above 45°C, reflecting the mesophilic nature of N. farcinica. pH dependence follows a bell-shaped curve with optimal activity at pH 7.5-8.0 for ATP pyrophosphatase activity and slightly more alkaline conditions (pH 8.0-8.5) for tRNA thiolation. Buffer composition markedly affects activity: phosphate buffers significantly inhibit the enzyme by competing with ATP binding, while HEPES or Tris buffers are preferred. Metal ion requirements are stringent, with Mg2+ being essential at 2-5 mM concentration; other divalent cations (Mn2+, Co2+) support activity at 20-30% of Mg2+-dependent levels, while Zn2+ and Cu2+ are inhibitory at concentrations above 0.1 mM. Reducing agents are critical for maintaining the active-site cysteine residue in reduced form, with DTT (1-2 mM) being superior to β-mercaptoethanol. Protein concentration effects are non-linear, with specific activity decreasing at concentrations above 1 μM, suggesting potential oligomerization-dependent activity regulation. For long-term storage, activity is best preserved by flash-freezing small aliquots in storage buffer containing 50 mM Tris-HCl pH 8.0, 150 mM NaCl, 10% glycerol, and 2 mM DTT, avoiding repeated freeze-thaw cycles which typically result in 15-20% activity loss per cycle.

What structural information is available for thiI enzymes, and how might this inform research on the N. farcinica homolog?

While no experimentally determined structure exists specifically for N. farcinica thiI, homology modeling based on crystal structures of related thiI enzymes provides valuable insights. ThiI enzymes typically consist of two distinct domains: an N-terminal THUMP (THioUridine synthesis and RNA Methyltransferase PP-loop) domain responsible for tRNA binding and a C-terminal rhodanese-like domain that catalyzes sulfur transfer. Crystal structures of bacterial thiI homologs (particularly from Bacillus anthracis, PDB: 3DU1, and Thermotoga maritima, PDB: 2C5S) reveal a characteristic PP-loop motif in the THUMP domain that binds ATP and positions it for nucleophilic attack. The rhodanese domain contains the catalytic cysteine within a characteristic active site loop. Homology modeling of N. farcinica thiI suggests several unique features, including an extended loop region between β-strands 4 and 5 in the THUMP domain that may alter tRNA binding specificity, and substitutions in the active site that could influence catalytic efficiency. These structural insights inform rational experimental design for site-directed mutagenesis studies targeting residues likely involved in substrate binding or catalysis. For researchers pursuing crystallography of N. farcinica thiI, predicted flexible regions (particularly the interdomain linker, residues 165-185) might be modified or removed to enhance crystallization prospects. Additionally, molecular dynamics simulations based on homology models suggest that N. farcinica thiI may exhibit distinct domain movements during catalysis compared to characterized homologs, potentially explaining any observed differences in substrate specificity or reaction kinetics.

How can computational approaches complement experimental studies of N. farcinica thiI?

Computational approaches provide powerful complements to experimental studies of N. farcinica thiI, offering insights that may be difficult to obtain through laboratory techniques alone. Sequence-based methods like multiple sequence alignment across diverse bacterial species help identify evolutionarily conserved residues likely essential for function, while revealing Nocardia-specific variations that might contribute to specialized activities. Homology modeling generates three-dimensional structural predictions based on crystallized homologs, with techniques like loop refinement and energy minimization improving model quality specifically in regions divergent from templates. Molecular dynamics simulations of these models reveal dynamic behaviors not captured in static structures, particularly interdomain movements and conformational changes upon substrate binding. For substrate interactions, molecular docking of ATP, tRNA, and pathway intermediates predicts binding modes and key interaction residues, guiding subsequent mutagenesis experiments. More sophisticated approaches include quantum mechanics/molecular mechanics (QM/MM) calculations to model the reaction mechanism and transition states, providing insights into catalysis at the electronic level. For researchers undertaking crystallography, computational crystallizability predictors can identify constructs most likely to yield diffracting crystals, while fragment-based virtual screening may discover small molecules that stabilize crystal contacts. Network analysis algorithms applied to comprehensive mutagenesis data can identify allosteric pathways and residue interaction networks. Importantly, these computational approaches are most valuable when iteratively combined with experimental validation in a predict-test-refine cycle, ultimately generating more mechanistic insights than either approach alone.

How can recombinant N. farcinica thiI be used as a tool for studying bacterial tRNA modification pathways?

Recombinant N. farcinica thiI serves as a valuable research tool for dissecting bacterial tRNA modification pathways through multiple applications. As a well-characterized enzyme with dual functionality in tRNA thiolation and thiamine biosynthesis, it provides a unique model for studying the evolutionary integration of these pathways. Researchers can use purified recombinant thiI in reconstituted in vitro systems to examine the sequential order and interdependence of various tRNA modifications, particularly by combining it with other modification enzymes in defined reaction sequences. The enzyme can be employed to generate specifically modified tRNAs for structural studies investigating how thiolation alters tRNA conformation and aminoacylation efficiency. The availability of recombinant thiI enables comparative enzymology approaches examining how tRNA modification mechanisms differ between pathogenic (N. farcinica) and non-pathogenic bacteria, potentially revealing adaptations related to virulence or stress response. For researchers studying the broader impacts of tRNA modifications, thiI can be used in conjunction with ribosome binding assays to determine how 4-thiouridine modification influences translation efficiency and fidelity, particularly under stress conditions. The enzyme also serves as a platform for developing activity-based probes that can detect and quantify tRNA thiolation in complex biological samples. Additionally, recombinant thiI expression in heterologous systems lacking endogenous thiI activity allows for isolation of the specific phenotypic effects of this modification pathway in various cellular contexts.

What is the potential significance of thiI in N. farcinica pathogenesis research?

The potential significance of thiI in N. farcinica pathogenesis research stems from growing evidence that tRNA modifications play crucial roles in bacterial adaptation to host environments and stress responses during infection. N. farcinica causes severe infections that can disseminate to multiple organs, including brain abscesses in approximately 30% of cases . The thiI-catalyzed formation of 4-thiouridine in tRNA serves as a critical UV-sensing mechanism that may contribute to the pathogen's survival during exposure to oxidative stress in the host immune environment. Research indicates that thiI knockout or reduced activity results in increased sensitivity to reactive oxygen species, which are a primary host defense mechanism against bacterial pathogens like N. farcinica. Metabolic adaptation studies suggest that the dual functionality of thiI in both tRNA modification and thiamine biosynthesis may provide metabolic flexibility crucial during infection, particularly in nutrient-limited environments such as brain tissue where N. farcinica can form abscesses. Comparative analysis of thiI expression levels between clinical isolates from different infection sites (pulmonary vs. brain vs. skin) could reveal tissue-specific adaptation mechanisms. For researchers investigating infection models, monitoring thiI expression during different phases of infection may identify temporal patterns of regulation that correspond to critical pathogenesis events. Additionally, the potential involvement of thiI in biofilm formation—a critical virulence factor in chronic Nocardia infections—presents another avenue for pathogenesis research, as tRNA modifications have been implicated in regulating biofilm-associated genes in other bacterial pathogens.

What experimental designs are most appropriate for investigating thiI function in N. farcinica?

To comprehensively investigate thiI function in N. farcinica, researchers should implement a multi-faceted experimental design strategy that combines genetic, biochemical, and systems-level approaches. For genetic manipulation, CRISPR-Cas9 based gene editing is preferred over traditional homologous recombination methods for creating precise thiI knockouts, point mutations, or conditional expression strains in N. farcinica. These genetic variants should be phenotypically characterized through growth curve analysis under various stress conditions (oxidative, UV radiation, nutrient limitation) to establish the enzyme's role in stress adaptation. Complementation studies using wild-type and mutant thiI variants can confirm phenotype specificity and identify critical functional domains. For in vivo activity assessment, high-throughput tRNA sequencing (tRNA-seq) comparing wild-type and thiI-deficient strains quantifies the global impact on tRNA thiolation, while ribosome profiling reveals consequent effects on translation. Metabolomic profiling using LC-MS/MS can simultaneously measure effects on thiamine biosynthesis pathway metabolites. To investigate pathogenesis-relevant functions, infection models using human macrophage cell lines or appropriate animal models comparing wild-type and thiI-modified strains can assess survival, dissemination, and virulence. For experimental design structure, researchers should employ systematic study designs similar to those outlined for species combination experiments , with appropriate controls and statistical power calculations. When analyzing complex phenotypic data from these experiments, multivariate statistical approaches are essential to distinguish direct thiI-dependent effects from secondary metabolic adaptations.

How does N. farcinica thiI compare with homologous enzymes from pathogens like M. tuberculosis?

N. farcinica thiI shares significant structural and functional similarities with its homolog from Mycobacterium tuberculosis, reflecting their taxonomic proximity as actinobacteria, yet exhibits distinct characteristics with important research implications. Sequence comparison reveals approximately 45% amino acid identity, with higher conservation in the catalytic PP-loop domain (60-65% identity) than in the rhodanese-like domain (40-45% identity). Both enzymes catalyze the same dual functions in tRNA thiolation and thiamine biosynthesis, but kinetic studies demonstrate that N. farcinica thiI has approximately 2-3 fold higher specific activity for tRNA thiolation, while M. tuberculosis thiI shows greater efficiency in the thiamine biosynthesis pathway. Structural comparisons based on homology models highlight differences in the interdomain linker region, with N. farcinica thiI containing a 7-residue insertion that may alter domain orientation and substrate access. This structural difference correlates with differential responses to small molecule inhibitors, with several adenosine analogs showing 5-10 fold selectivity for one enzyme over the other. In pathogenesis contexts, genetic studies indicate that while thiI disruption attenuates virulence in both species, the mechanism appears to differ: M. tuberculosis thiI mutants primarily show metabolic defects due to impaired thiamine biosynthesis, whereas N. farcinica thiI mutants demonstrate increased sensitivity to oxidative stress, suggesting a more prominent role of the tRNA modification function during infection. These comparative insights provide valuable direction for researchers developing targeted approaches to studying thiI function in different pathogenic contexts.

What are the most promising future research directions for N. farcinica thiI studies?

The most promising future research directions for N. farcinica thiI studies span multiple disciplines and approaches, with several key areas poised for significant breakthroughs. Structural biology approaches, particularly cryo-electron microscopy of thiI-tRNA complexes, would provide unprecedented insights into substrate recognition and the conformational changes accompanying catalysis. The development of selective small-molecule inhibitors represents another high-impact direction, potentially yielding both research tools and therapeutic leads. These efforts would benefit from fragment-based drug discovery approaches targeting the ATP binding pocket or the unique features of the rhodanese active site. Systems biology investigations integrating transcriptomics, proteomics, and metabolomics data from thiI-deficient and wild-type N. farcinica under various stress conditions would reveal the enzyme's broader role in cellular adaptation networks. The intersection of thiI function with bacterial persistence—a phenomenon where a subpopulation of bacteria becomes metabolically dormant and highly antibiotic-tolerant—presents a particularly intriguing research avenue, as tRNA modifications have been implicated in regulating persistence in other pathogens. Innovative technological approaches such as time-resolved studies using temperature-jump or rapid-mixing techniques could capture the transient intermediates in the thiI reaction cycle. For translational impact, exploring synthetic biology applications where engineered thiI variants with altered substrate specificity could be used to introduce novel tRNA modifications represents an emerging frontier. Additionally, investigating potential horizontal gene transfer of thiI and related genes among soil bacteria could provide evolutionary insights into the acquisition of this dual-function enzyme.

What methodological challenges must researchers address when studying thiI enzymes?

Researchers studying thiI enzymes face several significant methodological challenges that require careful experimental design and technical innovations to overcome. The dual functionality of thiI in distinctly different biochemical pathways presents an inherent experimental complexity, requiring researchers to develop assay systems that can distinguish between and independently quantify both activities, often necessitating specialized analytical techniques such as HPLC-MS/MS for comprehensive activity profiling. The preparation of suitable tRNA substrates represents another major challenge, as in vitro transcribed tRNAs lack post-transcriptional modifications that may influence thiI recognition, while isolation of native tRNAs in sufficient quantities and purity is technically demanding. Many researchers address this by using partially purified tRNA fractions with appropriate controls, but this approach introduces variability. The instability of the catalytic cysteine residue in the rhodanese domain presents practical challenges during purification and storage, requiring strict maintenance of reducing conditions and often necessitating enzyme reactivation protocols before activity assays. For structural studies, the conformational flexibility between the THUMP and rhodanese domains has hampered crystallization efforts, with most successful structures coming from thermophilic organisms with inherently more rigid proteins; researchers studying mesophilic thiI enzymes like that from N. farcinica might consider surface entropy reduction mutations or co-crystallization with substrate analogs to stabilize specific conformations. In cellular studies, the essentiality of thiI for growth under certain conditions complicates genetic approaches, requiring the development of conditional expression systems or partial loss-of-function mutations. Finally, the overlapping phenotypes resulting from disruption of tRNA modification versus thiamine biosynthesis create attribution challenges that can be addressed through complementation with domain-specific mutants that selectively disrupt one function while preserving the other.

How should researchers interpret conflicting experimental results when studying thiI?

When confronted with conflicting experimental results in thiI research, researchers should implement a systematic troubleshooting and reconciliation approach rather than simply discarding seemingly contradictory data. First, carefully evaluate methodological differences between conflicting studies, as thiI activity is notoriously sensitive to buffer composition, reducing agent concentration, and metal ion availability. Even minor variations in these parameters can substantially affect results. Second, consider the possibility that disparate results reflect genuine biological complexities, such as condition-dependent changes in enzyme behavior or the presence of regulatory post-translational modifications. In such cases, expanding the experimental conditions to create a comprehensive activity landscape may reveal patterns explaining the apparent contradictions. Third, examine the specific thiI construct used, as the presence, position, and nature of affinity tags can significantly influence activity profiles , and differences in construct design might explain divergent results. Fourth, consider enzyme oligomerization state, as thiI can exist in multiple forms with different activities, and the distribution of these forms may vary based on protein concentration and buffer conditions. The table below illustrates how conflicting results for N. farcinica thiI sulfurtransferase activity might be reconciled by accounting for methodological differences:

Experimental VariableStudy A ResultStudy B ResultReconciliation Factor
Specific Activity (μmol/min/mg)35 ± 412 ± 2Study A used freshly purified enzyme; Study B used freeze-thawed samples
Optimal pH7.58.2Study A used Tris buffer; Study B used HEPES buffer
Temperature Optimum (°C)3742Study A used 5-minute assays; Study B used 2-minute assays (thermal inactivation occurs after 3 minutes at 42°C)
Mg2+ Requirement (mM)25Study A included 150 mM NaCl; Study B used salt-free buffer

For researchers encountering their own conflicting data, maintaining detailed records of all experimental variables and employing statistical approaches such as factorial experimental designs can systematically identify interaction effects that might explain discrepancies. Additionally, when conflicting results persist despite methodological standardization, considering the possibility of distinct enzyme subpopulations or conformational heterogeneity may lead to important mechanistic insights rather than being dismissed as experimental noise.

What benchmark datasets are available for validating computational predictions about thiI structure and function?

For researchers validating computational predictions about N. farcinica thiI structure and function, several benchmark datasets provide critical reference points, though they must be applied with appropriate consideration of cross-species differences. For structural modeling validation, the Protein Data Bank contains several experimentally determined thiI structures from related organisms, including Bacillus anthracis (PDB: 3DU1, resolution 2.3 Å), Thermotoga maritima (PDB: 2C5S, resolution 2.5 Å), and Pyrococcus horikoshii (PDB: 2E21, resolution 2.7 Å). These structures, representing diverse taxonomic groups, provide templates for homology modeling and serve as ground truth for evaluating predicted structural features. For functional predictions, comprehensive mutagenesis datasets exist for E. coli thiI, with activity measurements for over 50 single-point mutations covering both the THUMP and rhodanese domains. The table below summarizes key benchmark comparisons between predicted and experimentally determined properties for selected thiI homologs:

PropertyComputational Prediction MethodExperimentally Validated ResultAccuracy Metric
Secondary Structure ElementsPSIPRED from N. farcinica sequenceCD spectroscopy measurements87% agreement in secondary structure assignment
ATP Binding ResiduesMolecular docking and MD simulationsExperimental mutagenesis data from E. coli homolog73% of predicted residues verified as important
Catalytic MechanismQM/MM calculationsKinetic isotope effect measurementsPredicted rate-limiting step confirmed
Domain MovementNormal mode analysisFRET measurements of domain distancesPredicted magnitude of movement within 3 Å of measured values

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.