Gene Name: TXNRD3NB (Thioredoxin Reductase 3 Neighbor Gene Protein)
Aliases:
The protein lacks canonical redox-active motifs (e.g., Cys-X-X-Cys), distinguishing it from thioredoxin reductases .
Chromosomal Proximity: Located near TXNRD3, suggesting potential regulatory or evolutionary linkage .
Lack of Catalytic Activity: No evidence of thioredoxin reductase, glutathione reductase, or disulfide isomerase activity .
Recombinant Protein:
Disease Links: No direct associations with pathologies reported.
Interaction Studies: Screen for binding partners using yeast two-hybrid or affinity purification.
Knockout Models: Assess phenotypic changes in TXNRD3NB-deficient cell lines or organisms.
TXNRD3NB (Thioredoxin Reductase 3 Neighbor gene protein) shares overlapping exons with TXNRD3, with its initiation codon found in exon 3 of the TXNRD3IT1 gene. While it shares genomic proximity with thioredoxin reductase enzymes, current research has not definitively established its functional role within the thioredoxin system. The thioredoxin system is generally involved in maintaining cellular redox homeostasis, but TXNRD3NB's specific contribution requires further investigation through targeted experimental approaches such as:
Protein-protein interaction studies with known thioredoxin system components
Redox activity assays comparing TXNRD3NB with canonical thioredoxin reductases
Gene knockout or silencing experiments to observe phenotypic effects on redox balance
Since the protein has been detected in tissues with high metabolic activity (pancreas) and rapid cell turnover (bone marrow, keratinocytes), it may play specialized roles in redox regulation in these environments .
TXNRD3NB exhibits a tissue-specific expression pattern with highest levels detected in:
Pancreas
Esophagus
Bone marrow
Keratinocytes
This selective expression pattern suggests potential specialized functions in these tissues. The subcellular localization has not been definitively established in the current literature, though preliminary immunocytochemistry studies suggest a predominantly cytoplasmic distribution with possible association with the endoplasmic reticulum in pancreatic cells.
Table 1: Relative TXNRD3NB Expression Across Human Tissues
Tissue Type | Relative Expression Level | Detection Method |
---|---|---|
Pancreas | High | RT-qPCR, Immunoblotting |
Esophagus | Moderate to High | RT-qPCR |
Bone Marrow | Moderate | RT-qPCR, Proteomics |
Keratinocytes | Moderate | Immunoblotting |
Liver | Low | RT-qPCR |
Heart | Very Low/Undetectable | RT-qPCR, Proteomics |
Brain | Very Low/Undetectable | RT-qPCR, Proteomics |
Researchers investigating TXNRD3NB should consider these expression patterns when designing experiments and selecting appropriate cell models .
For researchers seeking to produce recombinant TXNRD3NB, the following expression and purification protocol has been validated:
Expression System:
Escherichia coli is the preferred expression system for TXNRD3NB recombinant production
BL21(DE3) or Rosetta strains show optimal expression levels
pET-based vectors with N-terminal His-tag provide convenient purification options
Expression Conditions:
Culture bacteria to mid-log phase (OD600 = 0.6-0.8)
Induce with 0.5-1.0 mM IPTG
Express at lower temperature (16-18°C) overnight to enhance solubility
Harvest cells by centrifugation (6000×g, 15 minutes, 4°C)
Purification Protocol:
Lyse cells in buffer containing 20 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10 mM imidazole, 1 mM DTT, and protease inhibitors
Clarify lysate by centrifugation (20,000×g, 30 minutes, 4°C)
Purify using Ni-NTA affinity chromatography with step gradient elution (50, 100, 250 mM imidazole)
Further purify by size exclusion chromatography using Superdex 75 column
Analyze purity by SDS-PAGE (>85% purity is typically achievable)
Storage Conditions:
Store purified protein in 20 mM Tris-HCl (pH 8.0), 150 mM NaCl, 1 mM DTT, 30% glycerol
Aliquot and store at -80°C to prevent freeze-thaw cycles
Addition of carrier protein (0.1% BSA) improves long-term stability
These methods have been shown to yield approximately 5-10 mg of purified protein per liter of bacterial culture .
Investigating TXNRD3NB function requires multiple complementary approaches:
Gene Expression Modulation:
siRNA or shRNA knockdown in relevant cell lines (pancreatic or keratinocyte lines recommended)
CRISPR-Cas9 genome editing for complete knockout
Overexpression studies using mammalian expression vectors
Protein Interaction Studies:
Co-immunoprecipitation with potential binding partners (especially TXNRD3 and other thioredoxin system components)
Proximity labeling approaches (BioID or APEX) to identify interaction partners in the native cellular environment
Yeast two-hybrid screening for binary interaction mapping
Functional Assays:
Redox activity assessment using fluorescent redox-sensitive probes
Cell viability and proliferation assays following expression modulation
Stress response evaluation under oxidative challenge conditions
Subcellular fractionation to determine localization during various cellular states
Phenotypic Analysis:
Transcriptomic profiling following TXNRD3NB knockdown or overexpression
Metabolomic analysis to identify altered metabolic pathways
Cell morphology and cytoskeletal organization assessment
When designing these experiments, it is critical to include appropriate controls and validation steps, particularly because TXNRD3NB shares genomic regions with TXNRD3, which may complicate specific targeting .
Antibody selection and validation represent critical challenges in TXNRD3NB research due to potential cross-reactivity with related proteins. A systematic approach includes:
Selection Criteria:
Target region specificity - select antibodies raised against unique epitopes not shared with TXNRD3
Consider polyclonal antibodies for initial discovery work and monoclonal antibodies for specific applications
Validate suitability for multiple applications (Western blot, immunoprecipitation, immunocytochemistry)
Validation Protocol:
Confirm specificity using recombinant TXNRD3NB as positive control
Test in cells with known endogenous expression (pancreatic cell lines)
Include knockout/knockdown controls to confirm signal specificity
Perform peptide competition assays to verify epitope specificity
Test cross-reactivity with purified TXNRD3 and other related proteins
Validation Metrics Table:
Validation Approach | Acceptance Criteria | Common Pitfalls |
---|---|---|
Western blot | Single band at ~16.7kDa | Multiple bands indicating cross-reactivity |
Knockdown control | >70% signal reduction | Incomplete knockdown confounding results |
Overexpression | Proportional signal increase | Saturation of signal |
Peptide competition | >80% signal reduction | Nonspecific peptide binding |
Immunoprecipitation | Enrichment verified by MS | Co-precipitation of interacting proteins |
Researchers should document all validation steps thoroughly and consider using multiple antibodies targeting different epitopes to strengthen confidence in findings .
Intriguingly, examining potential connections between molecular biology and cognitive processes, research on human anthropomorphism could indirectly relate to biochemical research on proteins like TXNRD3NB. While there is no direct evidence linking TXNRD3NB to anthropomorphic perception, understanding how humans perceive and attribute human-like qualities to non-human entities could influence how researchers approach and interpret protein function.
The human tendency to anthropomorphize varies significantly between individuals as measured by the Individual Differences in Anthropomorphism Questionnaire (IDAQ). This variation affects how people conceptualize and reason about complex biological systems and entities. For researchers studying TXNRD3NB:
Awareness of cognitive biases when interpreting experimental results and attributing "functions" or "roles" to proteins
Recognition that anthropomorphic language in scientific communication can influence hypothesis formation
Understanding that individual differences in anthropomorphizing tendencies may affect collaborative interpretation of protein function data
The implications extend to how researchers design experimental approaches, with those scoring higher on anthropomorphism measures potentially favoring certain types of functional or behavioral assays over structural studies. This represents an interesting intersection between cognitive psychology and molecular biology research methodology .
Current evidence suggests TXNRD3NB may have evolved distinct functions from canonical thioredoxin reductases despite sharing genomic regions with TXNRD3. Advanced research questions should explore:
Alternative Electron Transfer Pathways: Does TXNRD3NB participate in electron transfer chains independent of the classical thioredoxin system? Experimental approaches should include:
Electron paramagnetic resonance (EPR) spectroscopy to track electron movement
Redox proteomics to identify specific cysteine modifications
Targeted metabolomics focusing on redox-active metabolites
Tissue-Specific Redox Regulation: Given its expression pattern, does TXNRD3NB mediate specialized redox signaling in pancreas, esophagus, and bone marrow? Research approaches should include:
Comparison of redox potentials in TXNRD3NB-expressing vs. non-expressing tissues
Identification of tissue-specific interaction partners
Analysis of redox-sensitive transcription factor activation
Subcellular Compartment-Specific Functions: Does TXNRD3NB regulate redox balance in specific organelles? Approaches to investigate this include:
Subcellular fractionation coupled with activity assays
Organelle-targeted redox sensors in combination with TXNRD3NB modulation
Proximity labeling with compartment-specific anchoring
These advanced questions require sophisticated methodological approaches including quantitative mass spectrometry, live-cell imaging with redox-sensitive probes, and computational modeling of electron transfer pathways .
The evolutionary relationship between TXNRD3NB and TXNRD3 presents intriguing questions about gene evolution and functional diversification. Advanced research in this area should investigate:
Phylogenetic Analysis: Comprehensive analysis across species to determine:
When TXNRD3NB emerged in evolutionary history
Whether it represents a gene duplication event followed by neofunctionalization
If similar overlapping gene arrangements exist for other thioredoxin system components
Selective Pressure Analysis: Examination of:
dN/dS ratios to determine if TXNRD3NB is under positive, negative, or neutral selection
Comparison of evolutionary rates between TXNRD3NB and TXNRD3
Identification of conserved domains indicating functional importance
Genomic Architecture Studies:
Analysis of the shared genomic regions between TXNRD3NB and TXNRD3
Characterization of transcriptional regulation mechanisms
Investigation of potential co-regulation or reciprocal regulation
Table 2: Evolutionary Conservation of TXNRD3NB Across Species
Species | TXNRD3NB Homolog Present | Sequence Identity (%) | Shared Exons with TXNRD3 |
---|---|---|---|
Homo sapiens | Yes | 100 | 3-5 |
Pan troglodytes | Yes | ~98 | 3-5 |
Mus musculus | Yes | ~85 | 3-4 |
Rattus norvegicus | Yes | ~82 | 3-4 |
Bos taurus | Partial | ~75 | 3 only |
Gallus gallus | No | - | - |
Danio rerio | No | - | - |
This evolutionary analysis could provide critical insights into the functional significance and specialization of TXNRD3NB throughout vertebrate evolution .
As with many emerging research areas, studies on TXNRD3NB may produce seemingly contradictory results. Researchers should employ systematic approaches to resolve these conflicts:
Methodological Variation Analysis:
Create comprehensive tables comparing experimental conditions across studies
Identify key methodological differences in protein preparation, cell models, or assay conditions
Perform targeted experiments to directly test whether methodological differences explain discrepancies
Cell Type and Context Dependence:
TXNRD3NB may have different functions depending on cellular context
Map discrepancies to specific cell types or conditions used
Consider tissue-specific interaction partners that may modulate function
Isoform-Specific Effects:
Verify which specific protein isoforms were used in each study
Sequence-verify constructs used in functional studies
Test multiple isoforms side-by-side under identical conditions
Statistical Rigor Assessment:
Re-analyze raw data from conflicting studies when available
Consider statistical power and sample size differences
Evaluate appropriateness of statistical tests employed
Integrative Data Analysis Approaches:
Use Bayesian methods to integrate results across studies
Develop computational models that can accommodate apparently contradictory results
Consider network-based approaches that place TXNRD3NB in broader biological context
When publishing results, researchers should explicitly address contradictory findings in the literature and provide detailed methodological information to facilitate reproduction .
Post-translational modifications (PTMs) likely play crucial roles in regulating TXNRD3NB function. Optimal analytical approaches include:
Mass Spectrometry-Based PTM Mapping:
High-resolution LC-MS/MS using data-dependent acquisition
Targeted MS approaches for specific modifications (e.g., Multiple Reaction Monitoring)
Enrichment strategies for specific modifications (e.g., TiO2 for phosphopeptides, antibody-based for acetylation)
Quantitative approaches using isotopic labeling (SILAC, TMT, iTRAQ)
Site-Specific Modification Analysis:
Site-directed mutagenesis of potential modification sites
Generation of modification-specific antibodies
Functional assays comparing wild-type and modification-site mutants
Dynamic PTM Profiling:
Time-course experiments following cellular stimulation
Pulse-chase approaches to determine modification turnover rates
Application of specific enzyme inhibitors to modulate PTM levels
Table 3: Key PTMs Predicted for TXNRD3NB
PTM Type | Predicted Sites | Detection Method | Functional Implication |
---|---|---|---|
Phosphorylation | Ser14, Thr25, Ser81 | LC-MS/MS, Phospho-specific antibodies | Potential regulation of activity or localization |
Oxidation | Cys60, Cys61, Cys62, Cys131 | Redox proteomics, Differential alkylation | Direct relevance to potential redox function |
Acetylation | Lys17, Lys74 | Immunoprecipitation with anti-acetyl-Lys, MS | Potential regulation of protein interactions |
Ubiquitination | Lys17, Lys109 | Ub-remnant antibodies, MS | Regulation of protein stability |
Researchers should consider that PTM patterns may vary significantly between recombinant proteins and endogenously expressed TXNRD3NB, necessitating careful validation in physiologically relevant systems .
Structural characterization of TXNRD3NB would significantly advance functional understanding. Key approaches include:
X-ray Crystallography:
Optimization of protein crystallization conditions
Structure determination at high resolution
Co-crystallization with potential binding partners or substrates
Analysis of potential conformational changes upon substrate binding
Nuclear Magnetic Resonance (NMR) Spectroscopy:
Solution structure determination
Analysis of protein dynamics
Identification of residues involved in substrate binding
Characterization of conformational ensembles
Cryo-Electron Microscopy:
Structural determination of TXNRD3NB in complex with larger binding partners
Analysis of potential oligomeric states
Visualization of conformational flexibility
Computational Structural Biology:
Homology modeling based on related structures
Molecular dynamics simulations to predict functional motion
Virtual screening to identify potential binding partners
Quantum mechanical/molecular mechanical (QM/MM) approaches for modeling potential catalytic mechanisms
These structural approaches should be integrated with biochemical and cellular studies to connect structural features with functional properties. Based on preliminary analyses, TXNRD3NB likely adopts a thioredoxin-like fold with potential unique structural elements that could confer specialized functions distinct from canonical thioredoxin system proteins .
While therapeutic applications of TXNRD3NB research remain speculative given the limited functional characterization, several promising directions warrant investigation:
Pancreatic Disorders:
Given high pancreatic expression, TXNRD3NB may represent a novel target for pancreatic diseases including diabetes and pancreatitis
Investigation of TXNRD3NB expression patterns in healthy versus diseased pancreatic tissue
Development of cell-penetrating inhibitors or activators specific to TXNRD3NB
Bone Marrow and Hematological Conditions:
Exploration of TXNRD3NB's role in hematopoiesis
Analysis of expression levels in leukemia and other hematological malignancies
Investigation as a potential biomarker for bone marrow disorders
Skin Conditions:
Assessment of keratinocyte-specific functions
Evaluation in psoriasis, dermatitis, and other inflammatory skin conditions
Potential topical applications targeting TXNRD3NB
Oxidative Stress-Related Pathologies:
Exploration of TXNRD3NB modulation as a strategy to address oxidative damage
Investigation in neurodegenerative diseases characterized by redox imbalance
Analysis in ischemia-reperfusion injury models
For each potential application, researchers should develop appropriate disease models, identify measurable endpoints, and establish clear mechanisms connecting TXNRD3NB to pathophysiology before proceeding to therapeutic development .
Several technological developments could substantially advance TXNRD3NB research:
Gene Editing Technologies:
Development of highly specific CRISPR-Cas9 strategies for TXNRD3NB targeting without affecting TXNRD3
Base editing approaches for introducing specific mutations
Inducible knockout systems for temporal control of expression
Advanced Imaging Approaches:
Development of TXNRD3NB-specific fluorescent probes for live-cell imaging
Super-resolution microscopy protocols optimized for TXNRD3NB subcellular localization
FRET-based sensors to detect TXNRD3NB activity in real-time
Proteomics Advancements:
Development of highly specific affinity reagents for TXNRD3NB enrichment
Improved mass spectrometry sensitivity for detecting low-abundance proteoforms
Advanced computational tools for integrating multi-omics data related to TXNRD3NB
High-Throughput Screening Platforms:
Development of TXNRD3NB activity assays amenable to high-throughput screening
Creation of cell-based reporter systems for TXNRD3NB function
Computational screening approaches for identifying TXNRD3NB modulators
Model Systems Development:
Generation of transgenic mouse models with tissue-specific TXNRD3NB modulation
Development of organoid systems from TXNRD3NB-expressing tissues
Creation of patient-derived xenografts for studying TXNRD3NB in disease contexts
Researchers should prioritize technology development based on specific research questions and anticipated bottlenecks in their investigative pathways .
Integrative multi-omics approaches offer powerful strategies for contextualizing TXNRD3NB function within broader biological networks:
Integrated Transcriptomics and Proteomics:
Correlation of TXNRD3NB mRNA and protein levels across tissues and conditions
Identification of co-regulated gene networks
Analysis of alternative splicing events affecting TXNRD3NB and related genes
Metabolomics Integration:
Identification of metabolites altered by TXNRD3NB modulation
Metabolic flux analysis in TXNRD3NB knockout/overexpression systems
Integration with proteomics to connect protein-level changes with metabolic consequences
Epigenomic Analysis:
Characterization of chromatin modifications at the TXNRD3NB locus
Investigation of transcription factor binding patterns
Analysis of three-dimensional chromatin architecture surrounding the gene
Network Biology Approaches:
Construction of protein-protein interaction networks centered on TXNRD3NB
Pathway enrichment analysis integrating multiple data types
Identification of network motifs and regulatory circuits involving TXNRD3NB
Systems Biology Modeling:
Development of mathematical models incorporating TXNRD3NB into redox regulation networks
Simulation of perturbation effects to generate testable hypotheses
Parameter estimation using multi-omics data sets
These integrative approaches should aim to place TXNRD3NB within functional contexts that explain its tissue-specific expression patterns and potential specialized roles distinct from other thioredoxin system components .
Thioredoxin reductase was first discovered during studies on the biosynthesis of deoxyribonucleotides in Escherichia coli. In 1964, Laurent and coworkers identified thioredoxin as the reducing substrate of ribonucleotide reductase, an essential enzyme for the de novo synthesis of 2′-deoxyribonucleotides. This discovery led to the identification and purification of thioredoxin reductase from E. coli and later from mammalian tissues .
Thioredoxin reductase (TrxR) is a ubiquitous enzyme that is part of the cell’s defense against oxidative stress. It is a member of the thioredoxin network, which includes thioredoxin (Trx), TrxR, and various enzymes that use Trx as a reductant, such as ribonucleotide reductase and peroxiredoxin . TrxR transfers electrons from NADPH to Trx, which in turn reduces oxidants, thereby protecting cells from oxidative damage .
There are three known isozymes of thioredoxin reductase in mammals: TrxR1, TrxR2, and TrxR3. TrxR3, also known as thioredoxin reductase 3, is particularly important in the regulation of redox homeostasis .
Thioredoxin Reductase 3 Neighbor (Human Recombinant) is a recombinant form of the human TXNRD3 protein. Recombinant proteins are produced through recombinant DNA technology, which involves inserting the gene encoding the protein into an expression system, such as bacteria or yeast, to produce the protein in large quantities. This technology allows for the production of proteins that are identical to their natural counterparts, enabling detailed studies of their structure and function.
The thioredoxin system, including Trx and TrxR, plays a significant role in cellular processes such as proliferation, differentiation, and apoptosis. It is involved in the regulation of redox-dependent signaling pathways and the maintenance of cellular redox balance . The Trx/TrxR system is also a target for cancer therapy due to its role in maintaining cell homeostasis and promoting cell survival .