TXNRD1 Human 161-649 a.a.

Thioredoxin Reductase 1 161-649 a.a. Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of TXNRD1 Human 161-649 a.a.

TXNRD1 (Thioredoxin Reductase 1) Human 161-649 a.a. is a recombinant protein corresponding to residues 161–649 of the human thioredoxin reductase 1 enzyme. This isoform is a critical component of the thioredoxin system, which regulates redox homeostasis and protects against oxidative stress . The protein is produced in E. coli and widely used in biochemical and biomedical research due to its enzymatic activity and relevance to diseases such as cancer, HIV, and autoimmune disorders .

Key Features:

  • Amino Acid Sequence: Residues 161–649 (isoform 1, UniProt ID Q16881-1), excluding the N-terminal propeptide (residues 151–152) .

  • Molecular Weight: ~55.7–60 kDa (observed via SDS-PAGE) .

  • Domains:

    • Glutaredoxin domain (aa 56–156) .

    • Pyridine nucleotide-disulphide oxidoreductase dimerization domain (aa 520–632) .

    • Catalytic selenocysteine at position 648 .

  • Post-Translational Modifications: Phosphotyrosines at positions 161, 163, and 281 .

Expression and Purification

The recombinant protein is expressed in E. coli and purified via nickel-affinity chromatography . Key steps include:

  1. Cloning: A DNA sequence encoding residues 161–649 is inserted into an expression vector .

  2. Solubility: Maintained in Tris-HCl (pH 8.0) or PBS with glycerol to prevent aggregation .

  3. Validation: Confirmed by western blot and enzymatic activity assays .

Biological Functions

TXNRD1 catalyzes the NADPH-dependent reduction of thioredoxin, supporting cellular antioxidant defenses and redox signaling . Specific roles include:

  • Redox Regulation: Maintains reduced thioredoxin for DNA synthesis and apoptosis regulation .

  • Disease Links:

    • Enhances transcriptional activity of estrogen receptors (isoform 4) .

    • Mediates cell death in interferon-β and retinoic acid therapies (isoform 5) .

    • Implicated in HIV latency and lymphoma progression .

Table 2: Functional Implications of TXNRD1 Isoforms

IsoformKey FunctionsReference
1Glutaredoxin activity, actin/tubulin polymerization, redox regulation
4Enhances estrogen receptor transcriptional activity
5Mediates interferon-β/retinoic acid-induced cell death

Research Applications

  • Enzymatic Assays: Used to study inhibition by gold-based compounds (e.g., auranofin) in cancer therapy .

  • Disease Models:

    • HIV latency studies show altered redox/iron metabolism .

    • Linked to oxidative stress in neurodegenerative diseases .

  • Structural Studies: Crystallography and mutagenesis to explore catalytic mechanisms .

Technical Considerations

  • Activity Assays: Requires NADPH as a cofactor; activity measured via DTNB reduction .

  • Storage: Lyophilized or in glycerol-containing buffers to retain stability .

  • Limitations: Sensitivity to freeze-thaw cycles; requires reconstitution with carrier proteins for long-term storage .

Product Specs

Introduction
TXNRD1, a member of the selenium-dependent pyridine nucleotide-disulfide oxidoreductase family, plays a crucial role in cellular redox balance and antioxidant defense. This enzyme, characterized by its conserved Cys-Val-Asn-Val-Gly-Cys catalytic site, catalyzes the reduction of thioredoxins and other substrates, contributing to selenium metabolism and protection against oxidative stress. Notably, TXNRD1 inhibition has emerged as a promising therapeutic strategy for various diseases, including cancer, AIDS, autoimmune disorders, bacterial infections, and parasitic diseases.
Description
Recombinant human TXNRD1, spanning amino acids 161 to 649, is expressed in E. coli with an N-terminal 21-amino acid His tag. This non-glycosylated polypeptide chain consists of 510 amino acids, resulting in a molecular weight of 55.9 kDa. The protein undergoes purification using proprietary chromatographic techniques to ensure high purity.
Physical Appearance
A clear, sterile-filtered solution without any visible color.
Formulation
The TXNRD1 solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution can be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To further enhance stability during long-term storage, consider adding a carrier protein like HSA or BSA (0.1%). Repeated freezing and thawing should be avoided to preserve protein integrity.
Purity
SDS-PAGE analysis confirms a purity exceeding 90.0%.
Synonyms
Thioredoxin reductase 1 cytoplasmic, TR, Gene associated with retinoic and interferon-induced mortality 12 protein, GRIM-12, Gene associated with retinoic and IFN-induced mortality 12 protein, KM-102-derived reductase-like factor, Thioredoxin reductase TR1, TXNRD1, GRIM12, KDRF, TR1, TXNR, TRXR1, MGC9145.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MYDYDLIIIG GGSGGLAAAK EAAQYGKKVM VLDFVTPTPL GTRWGLGGTC VNVGCIPKKL MHQAALLGQA LQDSRNYGWK VEETVKHDWD RMIEAVQNHI GSLNWGYRVA LREKKVVYEN AYGQFIGPHR IKATNNKGKE KIYSAERFLI ATGERPRYLG IPGDKEYCIS SDDLFSLPYC PGKTLVVGAS YVALECAGFL AGIGLDVTVM VRSILLRGFD QDMANKIGEH MEEHGIKFIR QFVPIKVEQI EAGTPGRLRV VAQSTNSEEI IEGEYNTVML AIGRDACTRK IGLETVGVKI NEKTGKIPVT DEEQTNVPYI YAIGDILEDK VELTPVAIQA GRLLAQRLYA GSTVKCDYEN VPTTVFTPLE YGACGLSEEK AVEKFGEENI EVYHSYFWPL EWTIPSRDNN KCYAKIICNT KDNERVVGFH VLGPNAGEVT QGFAAALKCG LTKKQLDSTI GIHPVCAEVF TTLSVTKRSG ASILQAGCCG.

Q&A

What is the functional role of TXNRD1 in cellular redox homeostasis?

TXNRD1 serves as a critical antioxidant enzyme in the defense against oxidative stress by regulating the dithiol/disulfide balance of interacting proteins . It catalyzes the NADPH-dependent reduction of thioredoxin (TXN), which in turn reduces disulfide bonds in various proteins. This system protects cells from reactive oxygen species (ROS) damage and maintains essential cellular proteins in their reduced, functional states.

The thioredoxin system (including TXNRD1) affects multiple cellular processes beyond simple redox regulation, including DNA synthesis, transcriptional control, and cell signaling. The importance of TXNRD1 is highlighted in knockout studies where homozygous Txn1 knockout mice are embryonically lethal, indicating its essential role in development .

What is the significance of the 161-649 amino acid region in human TXNRD1?

The 161-649 amino acid region of human TXNRD1 encompasses several functionally critical domains:

  • NADPH binding domain: Essential for cofactor binding that provides electrons for the reduction reaction

  • FAD binding domain: Required for electron transfer from NADPH to the active site

  • Interface residues for homodimerization: TXNRD1 functions as a homodimer

  • C-terminal active site containing the essential selenocysteine residue (Sec)

Mutations within this region can significantly impair enzyme activity, as demonstrated in the F54L mutation in rats that showed approximately one-third of the normal insulin-reducing activity . Specifically, the C-terminal region contains the conserved -Gly-Cys-Sec-Gly motif that forms the active site where thioredoxin reduction occurs.

How do mutations in TXNRD1 affect neurological function?

Mutations in TXNRD1 can have profound effects on neurological function. Research with the Txn1-F54L rat model demonstrated that:

  • Rats developed running seizures at approximately five weeks of age

  • Vacuolar degeneration occurred in the midbrain, primarily in the thalamus and inferior colliculus

  • Neuronal and oligodendrocyte cell death was observed in affected regions

  • Mitochondria showed morphological changes in neurons of mutant rats

  • Progression followed a specific timeline: degeneration began at three weeks, seizures at five weeks, and spontaneous repair started at seven weeks

This age-dependent pathology suggests TXNRD1 plays a critical developmental role in the midbrain. The temporal nature of these changes indicates complex interactions between TXNRD1 function, oxidative stress, and developmental processes in the central nervous system.

What are the optimal conditions for measuring TXNRD1 activity using the insulin-coupled TXN1 reduction assay?

The insulin-coupled TXN1 reduction assay is a standard method for measuring TXNRD1 activity. Optimal conditions include:

ParameterRecommended ConditionNotes
TXNRD1 concentration0.2 μMPre-reduced with NADPH
NADPH concentration100 μMFor pre-reduction and in master mix
TXN1 concentration5-10 μMIn master mix
Insulin concentration~160 μMIn master mix
Buffer50 mM Tris-HCl, 1 mM EDTA, pH 7.5TE buffer
TemperatureRoom temperature (20-25°C)Maintain constant
Monitoring wavelength340 nmFor NADPH oxidation
Measurement duration30 minutes10-second intervals
DMSO concentration<2%When testing inhibitors

The procedure involves first reducing 0.2 μM TXNRD1 with 100 μM NADPH for 10 minutes, then adding 30 μL of this mixture to each well of a 96-well plate . Add 170 μL of TXN1-insulin master mix and monitor absorbance at 340 nm to track NADPH oxidation . The slope of absorbance change directly correlates with TXNRD1 activity.

How can the DTNB reduction assay be optimized for high-throughput screening of TXNRD1 inhibitors?

The DTNB (5,5'-dithiobis-2-nitrobenzoic acid) reduction assay can be adapted for high-throughput screening with these optimizations:

  • Assay miniaturization:

    • Adapt to 384-well or 1536-well formats

    • Reduce reaction volumes to 20-50 μL

    • Use automated liquid handling systems

  • Reaction components optimization:

    • TXNRD1: 10-20 nM (pre-reduced with NADPH)

    • DTNB: 2-5 mM (optimized to avoid inhibition at high concentrations)

    • NADPH: 100-200 μM (ensure excess)

    • Buffer: 50 mM Tris-HCl, 1 mM EDTA, pH 7.5

  • Detection parameters:

    • Wavelength: 412 nm for TNB anion formation

    • Reading frequency: Every 5 seconds for 5 minutes

    • Detection mode: Kinetic rather than endpoint

  • Quality control measures:

    • Z' factor calculation (aim for >0.5)

    • Include known inhibitors as positive controls

    • Use DMSO-only controls as negative controls

    • Include NADPH-only controls to account for background

For the screening procedure, incubate pre-reduced TXNRD1 (reduced with NADPH for 10 minutes) with test compounds for 60 minutes, then add DTNB master mix and monitor absorbance at 412 nm . The rate of TNB formation directly correlates with residual TXNRD1 activity.

What considerations should be made when designing experimental controls for TXNRD1 knockdown studies?

When designing TXNRD1 knockdown experiments, include these essential controls:

  • Knockdown validation controls:

    • qRT-PCR to confirm mRNA reduction

    • Western blot to verify protein reduction

    • DTNB or insulin reduction assays to confirm functional impact

    • Time course analysis to determine optimal assessment timepoint

  • Technical controls:

    • Multiple siRNA/shRNA sequences targeting different regions of TXNRD1

    • Non-targeting siRNA/shRNA with similar GC content

    • Empty vector controls for shRNA experiments

    • Wild-type cells (no treatment)

  • Functional controls:

    • Rescue experiments using siRNA-resistant TXNRD1 constructs

    • Pharmacological TXNRD1 inhibition as a comparison

    • Assessment of related enzymes (TXNRD2, glutathione system) to identify compensation

  • Pathway analysis controls:

    • Assessment of mTOR signaling components and MYC targets (shown to be affected by TXNRD1)

    • Oxidative stress markers (ROS levels, protein carbonylation)

    • Cell type-specific effects may vary - research shows TXNRD1 knockdown exacerbates proliferation, migration, and apoptosis resistance in pulmonary artery smooth muscle cells

Knockdown efficiency should be at least 70-80% at the protein level to observe reliable phenotypic effects. Document changes in cellular redox state as TXNRD1 reduction will affect the cellular thiol/disulfide balance.

How does TXNRD1 function as a biomarker and prognostic factor in cancer?

TXNRD1 has emerged as a significant biomarker for multiple cancer types:

  • Hepatocellular Carcinoma (HCC):

    • TXNRD1 is overexpressed in 57 of 120 (47.5%) clinical HCC samples

    • Expression increases with advancing clinical stage

    • Positively correlates with N classification (p = 4.4e-4) and M classification (p = 0.037)

    • Kaplan-Meier analysis shows patients with high TXNRD1 expression have significantly shorter survival

    • Multivariate analysis confirms TXNRD1 as an independent prognostic factor

  • Pulmonary Arterial Hypertension:

    • Serum TXNRD1 levels are lower in idiopathic pulmonary arterial hypertension (IPAH) patients

    • Shows predictive efficiency with AUC of 0.795

    • Negatively correlates with mean pulmonary arterial pressure (mPAP) and pulmonary vascular resistance (PVR)

  • Other Cancers:

    • Oral squamous cell carcinomas

    • Lung cancer

    • Breast cancer

The relationship between TXNRD1 expression and patient outcomes makes it valuable for risk stratification and treatment planning in oncology.

What approaches are used to develop selective TXNRD1 inhibitors?

Development of selective TXNRD1 inhibitors involves several strategic approaches:

  • Structure-based targeting:

    • Focus on unique structural features distinguishing TXNRD1 from related enzymes

    • Target the C-terminal selenocysteine-containing redox center

    • Exploit differences between human TXNRD1 and pathogen thioredoxin reductases for antimicrobial applications

  • Reversible vs. irreversible inhibition strategies:

    • Reversible inhibitors: Typically compete with substrates (NADPH, thioredoxin)

    • Irreversible inhibitors: Form covalent bonds with the selenocysteine residue

    • Time-dependent inhibition analysis for covalent inhibitors

  • Assessment methodology:

    • Enzyme-based activity determination using insulin-coupled TXN1 or DTNB reduction assays

    • Differential scanning fluorimetry to evaluate thermal stability changes upon inhibitor binding

    • LC-MS/MS analysis to characterize molecular interactions and binding sites

  • Selectivity screening:

    • Counter-screening against related enzymes (TXNRD2, glutathione reductase)

    • Cell-based assays to confirm target engagement

    • Assessment of effects on cellular redox balance

For irreversible inhibitors, the protocol includes reducing TXNRD1 with NADPH, incubating with inhibitors, and measuring residual activity through established assays . Successful inhibitors typically achieve IC50 values in the nanomolar to low micromolar range with at least 10-fold selectivity over related enzymes.

How does TXNRD1 interact with the mTOR signaling pathway and MYC targets?

TXNRD1 has significant interactions with critical cellular signaling networks:

  • mTOR signaling pathway:

    • GSEA analysis shows enrichment between TXNRD1 and mTOR pathway components

    • TXNRD1 may influence mTOR through redox regulation of upstream components like PTEN

    • Cellular redox state affects amino acid sensing and energy status, which regulate mTOR

    • Inhibition of TXNRD1 can modulate mTOR activity through altered ROS levels

  • MYC targets:

    • TXNRD1 is itself a transcriptional target of MYC in many cell types

    • TXNRD1 supports MYC-driven metabolic reprogramming by maintaining redox balance

    • The thioredoxin system is required for the high proliferation rates associated with MYC activation

    • GSEA analysis confirms enrichment of MYC target genes with TXNRD1 expression

  • Unfolded protein response (UPR):

    • TXNRD1 helps maintain proper protein folding by regulating thiol/disulfide status

    • Inhibition leads to accumulation of misfolded proteins, triggering UPR

    • TXNRD1 protects against ER stress through thioredoxin-dependent mechanisms

These interactions position TXNRD1 at a critical junction of cellular growth, proliferation, and stress response pathways, explaining its importance in both normal physiology and disease states like cancer and pulmonary hypertension.

How can liquid chromatography-tandem mass spectrometry (LC-MS/MS) be utilized to analyze TXNRD1-inhibitor interactions?

LC-MS/MS offers powerful approaches for characterizing TXNRD1-inhibitor interactions:

  • Binding site identification:

    • Digest inhibitor-treated TXNRD1 with proteases (trypsin, chymotrypsin)

    • Identify peptides with mass shifts corresponding to inhibitor modification

    • MS/MS fragmentation pinpoints exact amino acids modified

    • Special attention to the selenocysteine-containing C-terminal active site peptide

  • Characterization workflow:

    • Pre-reduce TXNRD1 with NADPH to activate the enzyme

    • Incubate with inhibitors under controlled conditions

    • Perform proteolytic digestion

    • LC separation of peptides

    • MS/MS analysis with high-resolution instruments (Orbitrap, Q-TOF)

  • Quantitative assessment:

    • Selected/Multiple Reaction Monitoring (SRM/MRM) for specific modified peptides

    • Ratio of modified to unmodified peptides indicates modification extent

    • Time-course experiments reveal kinetics of covalent bond formation

  • Special considerations for TXNRD1:

    • Selenium-containing peptides have distinctive isotope patterns

    • C-terminal peptide may require special extraction/enrichment

    • Fragmentations patterns differ from typical peptides

    • Standard peptide analysis parameters may need optimization

This approach provides molecular-level insights into how inhibitors engage with TXNRD1, supporting rational drug design and mechanism-of-action studies. The technique is particularly valuable for irreversible inhibitors where understanding the exact site and nature of covalent modification is crucial .

What are the advantages and limitations of differential scanning fluorimetry (DSF) for studying TXNRD1 inhibitors?

DSF (thermal shift assay) offers several advantages and has specific limitations for TXNRD1 inhibitor research:

Advantages:

  • High-throughput capability in 96/384-well formats

  • Small sample requirements (micrograms of protein)

  • Label-free interaction analysis

  • Detection of both reversible and irreversible inhibitors

  • Complementary data to activity assays

  • Accessible instrumentation (real-time PCR machines)

  • Provides insights into binding mechanisms through thermal stability changes

Limitations:

  • Indirect measurement of binding (thermal stability ≠ inhibition)

  • Potential false negatives if binding doesn't significantly affect stability

  • Potential false positives from non-specific effects on protein stability

  • Limited structural information compared to crystallography/NMR

  • Challenges with multi-domain proteins like TXNRD1

  • Buffer constraints may limit testing conditions

  • Complicated interpretation with covalent/irreversible inhibitors

Implementation for TXNRD1:

  • Prepare TXNRD1 at 2-5 μM concentration in buffer

  • Add SYPRO Orange or similar fluorescent dye

  • Test compounds at multiple concentrations (typically 10-100 μM)

  • Include DMSO-only controls

  • Heat samples from 25-95°C with 0.5-1°C increments

  • Monitor fluorescence increase as protein unfolds

  • Calculate melting temperature (Tm) shift compared to controls

For TXNRD1 inhibitor studies, DSF serves best as a screening tool within a multi-method approach, complementing activity assays and structural studies .

What are the most promising research areas for TXNRD1 inhibitors in disease treatment?

Several high-priority research areas for TXNRD1 inhibitors show therapeutic promise:

  • Cancer therapy:

    • TXNRD1 overexpression in multiple cancers makes it an attractive target

    • Selective inhibitors may exploit cancer cells' increased dependence on thioredoxin system

    • Combination with existing chemotherapeutics to overcome resistance

    • Exploration of tumor microenvironment effects through TXNRD1 modulation

  • Neurodegenerative disorders:

    • TXNRD1 mutations are linked to epilepsy and neurodegenerative phenotypes

    • Neuroprotective strategies through targeted TXNRD1 modulation

    • Age-dependent effects suggest critical developmental windows for intervention

    • Mitochondrial protection in vulnerable neurons

  • Pulmonary hypertension:

    • TXNRD1 is downregulated in IPAH patients

    • Therapeutic strategies to restore TXNRD1 function

    • Counteracting proliferative disorder and apoptosis resistance in PASMCs

  • Biomarker development:

    • Prognostic stratification in hepatocellular carcinoma

    • Early detection of IPAH through serum TXNRD1 measurements

    • Therapy response prediction based on TXNRD1 levels

  • Multi-target approaches:

    • Combined inhibition of TXNRD1 and complementary redox systems

    • Targeting TXNRD1-mTOR-MYC interconnections

    • Modulation of unfolded protein response in disease states

These directions require further mechanistic studies, improved inhibitor selectivity, and robust in vivo validation before clinical translation.

What gaps exist in our understanding of TXNRD1 structure-function relationships?

Despite significant advances, several knowledge gaps remain in TXNRD1 research:

  • Region-specific functions:

    • Precise roles of different domains within the 161-649 amino acid region

    • Structure-function relationships of specific motifs and residues

    • Dynamic changes during catalytic cycles

  • Tissue-specific regulation:

    • Mechanisms behind tissue-specific expression patterns

    • Differential responses to oxidative stress across tissues

    • Why midbrain regions show particular vulnerability to TXNRD1 dysfunction

  • Post-translational modifications:

    • Comprehensive mapping of physiological modifications

    • Impact on activity, localization, and protein interactions

    • Regulatory mechanisms through PTMs

  • Developmental dynamics:

    • Age-dependent functions as observed in epilepsy models

    • Role in developmental processes versus homeostatic maintenance

    • Compensatory mechanisms during development

  • Protein-protein interactions:

    • Complete interactome beyond thioredoxin

    • Structural basis of specific protein interactions

    • Regulatory protein complexes in different cellular compartments

  • Isoform-specific functions:

    • Functional distinctions between splice variants

    • Subcellular localization patterns of different isoforms

    • Potential specialized roles in specific cellular contexts

Addressing these gaps requires integrated approaches combining structural biology, proteomics, developmental biology, and advanced imaging techniques to fully understand TXNRD1's multifaceted roles in health and disease.

Product Science Overview

Structure and Production

The human recombinant form of TXNRD1, specifically the 161-649 amino acid (a.a.) segment, is produced in Escherichia coli and is fused with a 21 amino acid His tag at the N-terminus . This recombinant protein is a single, non-glycosylated polypeptide chain containing 510 amino acids and has a molecular mass of approximately 55.9 kDa . The protein is purified using proprietary chromatographic techniques to ensure high purity and functionality .

Function and Mechanism

TXNRD1 plays a pivotal role in reducing thioredoxins and other substrates, which is essential for various cellular processes, including DNA synthesis, repair, and defense against oxidative stress . The enzyme catalyzes the reduction of thioredoxin by transferring electrons from NADPH to the active site disulfide of thioredoxin, thereby maintaining it in a reduced state .

Biological Significance

The thioredoxin system, including TXNRD1, is vital for cellular redox homeostasis and protection against oxidative damage. It is involved in numerous physiological processes such as cell growth, apoptosis, and immune response . Dysregulation of TXNRD1 activity has been implicated in various diseases, including cancer, autoimmune disorders, and neurodegenerative diseases .

Applications in Research and Medicine

Due to its critical role in cellular redox regulation, TXNRD1 is a target for therapeutic interventions. Inhibitors of TXNRD1 are being explored as potential treatments for cancer, AIDS, and other autoimmune diseases . Additionally, recombinant TXNRD1 is used in laboratory research to study its function and to develop new therapeutic strategies .

Storage and Stability

The recombinant TXNRD1 protein is typically stored at 4°C for short-term use and at -20°C for long-term storage. It is recommended to add a carrier protein, such as 0.1% HSA or BSA, to prevent degradation during storage . The protein should be handled carefully to avoid multiple freeze-thaw cycles, which can affect its stability and activity .

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 2024 Thebiotek. All Rights Reserved.