Thiosulfate sulfurtransferase-like domain-containing 3 (TSTD3) is a member of the sulfurtransferase superfamily, annotated in the human genome as a cytoplasmic protein. Unlike its isoforms TSTD1 and TSTD2, TSTD3 lacks a catalytic cysteine residue critical for enzymatic sulfur transfer activity . This distinction suggests a non-functional or structural role in sulfur metabolism pathways.
TSTD3 is expressed in multiple tissues, though its distribution remains poorly characterized. Recombinant TSTD3 is produced in Escherichia coli for research purposes:
Parameter | Value/Description |
---|---|
Expression Host | E. coli (codon-optimized synthetic DNA) |
Purity | >90% (SDS-PAGE validated) |
Formulation | 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 10% glycerol, 1 mM DTT |
Stability | Stable at -20°C; avoid repeated freeze-thaw cycles |
Recombinant TSTD3 includes a 23-amino acid N-terminal His-tag for purification via nickel affinity chromatography .
TSTD3 is not catalytically active due to the absence of the critical cysteine residue required for sulfur transfer . Comparative kinetic data for TSTD1 and rhodanese highlight TSTD3’s distinct profile:
Enzyme | Substrate Pair | K<sub>m</sub> (Donor) | K<sub>cat</sub> | Efficiency (k<sub>cat</sub>/K<sub>m</sub>) |
---|---|---|---|---|
TSTD1 | Thiosulfate → Thioredoxin | 22 ± 3 µM | 0.116 s⁻¹ | 6.8 × 10³ M⁻¹s⁻¹ |
Rhodanese | Thiosulfate → KCN | 39.5 ± 2.5 µM | 910 s⁻¹ | 23 × 10³ M⁻¹s⁻¹ |
TSTD3 | No Activity Observed | N/A | N/A | N/A |
TSTD3 has been implicated in experimental liver cirrhosis, though evidence remains limited to computational annotations . Copy number variations (CNVs) involving TSTD3 (e.g., deletions in 6q16.1-16.3) are associated with pathogenic phenotypes in genetic databases .
TSTD3’s precise biological role remains undefined. Prioritizing studies on:
Protein interactions: Potential partnerships with active sulfurtransferases (e.g., TSTD1, MST).
Structural biology: High-resolution crystallography to map domain organization.
Disease mechanisms: Functional validation of CNV associations in liver pathology .
Full sequence of recombinant TSTD3 (1–97 aa):
MGSSHHHHHHSSGLVPRGSHMGSMKIEKCGWSEGLTSIKGNCHNFYTAISKDV TYKELKNLLNSKNIMLIDVREIWEILEYQKIPESINVPLDEVGEALQMNPRDFKEKY NEVKPSKSDS
TSTD3 is a 97-amino acid protein involved in sulfur metabolism that plays a critical role in the detoxification of cyanide by converting it into less harmful thiocyanate . As a member of the rhodanese protein family, it contains a characteristic rhodanese-like domain that facilitates sulfurtransferase activity. Researchers studying this protein should consider its role within broader detoxification pathways and potential interactions with other sulfur metabolism enzymes.
For detecting TSTD3 in experimental systems, researchers can employ:
Method | Applications | Advantages | Limitations |
---|---|---|---|
Western blotting | Protein expression | Specific detection | Semi-quantitative |
qRT-PCR | mRNA expression | Highly sensitive | Doesn't confirm protein levels |
Immunohistochemistry | Tissue localization | In situ detection | Antibody specificity concerns |
Mass spectrometry | Protein identification | Definitive identification | Complex sample preparation |
ELISA | Quantitative detection | High throughput | Requires validated antibodies |
When working with recombinant TSTD3, the protein can be analyzed using SDS-PAGE and is suitable for mass spectrometry characterization .
TSTD3 expression patterns across tissues can be analyzed using:
RNA-seq data from tissue atlases to determine baseline expression
Promoter analysis using luciferase reporter assays to identify regulatory elements
ChIP-seq to identify transcription factor binding sites
Epigenetic profiling (DNA methylation, histone modifications) to understand chromatin-level regulation
Expression quantitative trait loci (eQTL) studies can also reveal genetic variants that affect TSTD3 expression levels. These studies involve analyzing genetically distinct populations to detect genomic differences such as SNPs and linking them to gene expression variations through statistical testing .
For efficient production of recombinant TSTD3:
Expression system: Escherichia coli is a proven system for producing active TSTD3
Construct design: Include a His-tag for purification (typically N-terminal)
Expression conditions: Optimize temperature, IPTG concentration, and induction time
Purification protocol: Use immobilized metal affinity chromatography followed by size exclusion chromatography
Quality control: Verify purity by SDS-PAGE (>90% purity is achievable) and confirm identity by mass spectrometry
Activity assessment: Develop a sulfurtransferase activity assay using appropriate substrates
To characterize TSTD3's sulfurtransferase activity:
Spectrophotometric assays:
Monitor thiocyanate formation using colorimetric methods
Track thiosulfate consumption in coupled enzyme systems
Chromatographic methods:
HPLC separation of reaction products
LC-MS/MS for definitive product identification and quantification
Enzyme kinetics:
Determine Km and Vmax for various substrates
Evaluate the effects of pH, temperature, and ionic strength on activity
Assess potential inhibitors and activators
Isotope labeling:
Use 35S-labeled substrates to track sulfur transfer
Apply 13C-labeled cyanide to monitor product formation
Select appropriate cellular models based on research objectives:
Model Type | Applications | Considerations |
---|---|---|
HEK293 cells | Overexpression studies, localization | Easy transfection, moderate endogenous expression |
HepG2 cells | Liver detoxification pathways | Relevant for cyanide metabolism studies |
Primary hepatocytes | Physiological relevance | Limited availability, short lifespan |
CRISPR-edited cell lines | Loss-of-function studies | Time-intensive but highly specific |
Inducible expression systems | Temporal control | Allows dose-dependent expression studies |
For generating TSTD3-deficient experimental models:
CRISPR/Cas9 gene editing:
Design guide RNAs targeting early exons
Screen for indels causing frameshifts
Validate knockout by sequencing, Western blot, and activity assays
RNAi approaches:
Design siRNAs targeting TSTD3 mRNA
Optimize transfection protocols for target cells
Confirm knockdown efficiency by qRT-PCR and Western blotting
Antisense oligonucleotides:
Design ASOs complementary to TSTD3 pre-mRNA
Test for efficiency in relevant cell types
Monitor off-target effects
To identify and characterize TSTD3 protein interactions:
Affinity purification coupled with mass spectrometry (AP-MS)
Yeast two-hybrid screening against human cDNA libraries
Proximity-dependent biotin identification (BioID)
Co-immunoprecipitation followed by Western blotting for suspected partners
Bimolecular fluorescence complementation to confirm interactions in live cells
These approaches can reveal functional connections between TSTD3 and other proteins involved in sulfur metabolism, cyanide detoxification, or other cellular pathways.
To study TSTD3 genetic variation:
Database mining:
Analyze gnomAD, dbSNP, and 1000 Genomes Project data
Focus on variants in coding regions or regulatory elements
Variant classification:
Distinguish between common polymorphisms and rare variants
Use bioinformatic tools to predict functional consequences
Functional characterization:
Express variant forms using site-directed mutagenesis
Compare activity, stability, and localization to wild-type
Clinical correlation:
Investigate associations with disease phenotypes
Consider potential impacts on cyanide sensitivity
When comparing TSTD3 research to other rhodanese proteins:
Size considerations:
TSTD3 is smaller (97 aa) than many rhodanese proteins
May require modified purification protocols
Substrate specificity:
Design assays that distinguish TSTD3 activity from related enzymes
Compare kinetic parameters across the rhodanese family
Structural analysis:
Use comparative modeling to identify unique features
Target distinctive residues for mutagenesis studies
Evolution and conservation:
Phylogenetic analysis to place TSTD3 in evolutionary context
Identify conserved motifs unique to TSTD3 versus other family members
When faced with contradictory results:
Cross-validation approaches:
Use multiple independent methods to measure the same parameter
Compare results across different cell types and experimental conditions
Controls and standards:
Include well-characterized positive and negative controls
Develop standardized assay protocols
Reproducibility assessment:
Perform rigorous statistical analysis
Conduct blind replication studies
Methodological transparency:
Document all experimental variables
Share detailed protocols and raw data
Given TSTD3's role in cyanide detoxification , investigate potential disease associations:
Toxicology considerations:
Variation in cyanide sensitivity between individuals
Potential role in smoke inhalation outcomes
Occupational exposure response differences
Metabolic disorders:
Research approaches:
Compare TSTD3 expression in patient vs. control samples
Analyze TSTD3 SNPs in case-control studies
Develop cellular disease models with modified TSTD3 expression
For selective modulation of TSTD3:
Small molecule screening:
Develop high-throughput assays for inhibitor/activator discovery
Perform structure-based virtual screening
Peptide-based approaches:
Design peptides that mimic interaction surfaces
Develop cell-penetrating peptides for intracellular targeting
Nucleic acid therapeutics:
Design antisense oligonucleotides for specific knockdown
Consider RNA aptamers as potential modulators
Allosteric regulation:
Identify non-catalytic binding sites
Explore the potential for isoform-selective targeting
These approaches provide tools for dissecting TSTD3 function in complex biological systems.
The TSTD3 gene is located on chromosome 6 and encodes a protein that is 120 amino acids long with a molecular mass of approximately 13.7 kDa . The protein is produced in Escherichia coli (E. coli) as a single, non-glycosylated polypeptide chain . The recombinant form of this protein is often fused with a His-tag at the N-terminus to facilitate purification through chromatographic techniques .
The primary function of TSTD3 is to facilitate the transfer of sulfur atoms within the cell. This is crucial for various biochemical pathways, including the detoxification of cyanide and the biosynthesis of iron-sulfur clusters . The active site cysteine residue in the rhodanese domain plays a pivotal role in these sulfur transfer reactions .
Mutations or dysregulation of the TSTD3 gene have been linked to certain diseases. For instance, TSTD3 has been associated with Leber Congenital Amaurosis 19, a genetic disorder that leads to severe vision loss at an early age . Understanding the function and regulation of TSTD3 can provide insights into the pathogenesis of such diseases and potentially lead to the development of targeted therapies.
Recombinant TSTD3 is widely used in laboratory research to study sulfur transfer reactions and their implications in various biological processes. The protein is available in different quantities and purities for research purposes . It is typically stored in a Tris-HCl buffer solution with glycerol and DTT to maintain stability .