SEPSECS Mouse

Selenocysteinyl-tRNA(Sec) synthase Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Genetic Background

SEPSECS Mouse refers to murine models with targeted mutations in the Sepsecs gene, which encodes selenocysteine synthase. This enzyme catalyzes the final step in selenocysteine-tRNA (Sec-tRNA) biosynthesis, essential for incorporating selenium into selenoproteins .

  • Key Genetic Modifications:

    • Sepsecs Y334C: A missense mutation mimicking the human pathogenic variant p.Y334C, causing perinatal lethality due to cardio-respiratory failure .

    • GPX4-Sec46Cys Crossbreed: Rescues perinatal lethality in Sepsecs Y334C/Y334C mice, highlighting GPX4’s role in ferroptosis regulation .

Phenotypic Characteristics

FeatureObservationSource
Perinatal SurvivalHomozygous Sepsecs Y334C mice die within 24 hours of birth due to respiratory collapse.
Brain DevelopmentReduced selenoprotein expression (e.g., GPX4, TXNRD1) in cortical neurons.
Oxidative StressUpregulation of NRF2-regulated antioxidant genes (e.g., Hmox1, Nqo1).
Rescue MechanismCrossbreeding with GPX4-Sec46Cys mice restores viability without normalizing selenoprotein levels.

Molecular Insights

  • Enzyme Dysfunction: The Y334C mutation disrupts SEPSECS tetramer formation, impairing Sec-tRNA synthesis .

  • Selenoprotein Deficiencies: Global reduction in selenoproteins (e.g., GPX4, SELENOP) in brain tissue .

  • Antioxidant Compensation: Persistent NRF2 activation mitigates oxidative damage despite low selenium utilization .

Comparative Analysis with Human Disorders

AspectSEPSECS Mouse ModelHuman Disorders
Primary SymptomsPerinatal death, respiratory failurePontocerebellar hypoplasia type 2D (PCH2D)
Selenoprotein ImpactGPX4 deficiency drives pathologyGPX4 mutations cause Sedaghatian dysplasia
Therapeutic ImplicationsPartial GPX4 activity suffices for survivalPotential for NRF2-targeted therapies

Recombinant SEPSECS Protein

  • Source: Escherichia coli-expressed mouse SEPSECS with N-terminal His-tag .

  • Molecular Weight: 57.7 kDa (527 amino acids) .

  • Functional Role: Converts O-phosphoseryl-tRNA to selenocysteinyl-tRNA using selenophosphate .

Applications in Disease Modeling

  • Neurological Disorders: Mimics progressive cerebellocerebral atrophy (PCCA) via selenoprotein deficiency .

  • Ferroptosis Research: Links GPX4 dysfunction to cardio-respiratory pathology .

  • Therapeutic Testing: Demonstrates that incomplete GPX4 activity can rescue lethal phenotypes, informing treatment strategies .

Limitations and Future Directions

  • Phenotypic Discrepancy: Murine perinatal lethality contrasts with human PCH2D’s postnatal onset .

  • Unanswered Questions: Mechanisms linking selenoprotein loss to specific brain regions remain unclear.

Product Specs

Introduction
SEPSECS is an enzyme responsible for the final step in selenocysteine (sec) synthesis. It converts O-phosphoseryl-tRNA(sec) to selenocysteinyl-tRNA(sec) utilizing selenophosphate as the selenium donor. In addition to its role in sec synthesis, SEPSECS is recognized as a specific biomarker for autoimmune hepatitis.
Description
Produced in E. coli, our SEPSECS is a single, non-glycosylated polypeptide chain consisting of 527 amino acids (with the mature protein encompassing amino acids 1-504). It has a molecular weight of 57.7 kDa. For purification and ease of use, SEPSECS is tagged with a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile solution.
Formulation
The SEPSECS protein is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 7.5), 1mM DTT, 0.2M NaCl, and 50% glycerol.
Purity
Purity is determined to be greater than 90% using SDS-PAGE analysis.
Stability
For optimal storage and product longevity, we recommend the following: If the entire vial will be used within 2-4 weeks, store at 4°C. For extended storage, freeze at -20°C. To further enhance stability during long-term storage, the addition of a carrier protein like 0.1% HSA or BSA is advised. Minimize repeated freeze-thaw cycles to maintain product integrity.
Synonyms

AA986712, D5Ertd135e, SecS, SLA, SLA/LP autoantigen, SLA-p35, Selenocysteinyl-tRNA(Sec) synthase, Selenocysteine synthase, Liver-pancreas antigen, Soluble liver antigen, Sec synthase, UGA suppressor tRNA-associated protein, SepSecS. 

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMNPESFA AGERRVSPAY VRQGCEARRA HEHLIRLLLE QGKCPEDGWD ESTLELFLHE LAVMDSNNFL GNCGVGEREG RVASALVARR HYRFIHGIGR SGDISAVQPK AAGSSLLNKI TNSLVLNVIK LAGVHSVASC FVVPMATGMS LTLCFLTLRH

KRPKAKYIIW PRIDQKSCFK SMVTAGFEPV VIENVLEGDE LRTDLKAVEA KIQELGPEHI LCLHSTTACF APRVPDRLEE LAVICANYDI PHVVNNAYGL QSSKCMHLIQ QGARVGRIDA FVQSLDKNFM VPVGGAIIAG FNEPFIQDIS KMYPGRASAS PSLDVLITLL SLGCSGYRKL

LKERKEMFVY LSTQLKKLAE AHNERLLQTP HNPISLAMTL KTIDGHHDKA VTQLGSMLFT RQVSGARAVP LGNVQTVSGH TFRGFMSHAD NYPCAYLNAA AAIGMKMQDV DLFIKRLDKC LNIVRKEQTR ASVVSGADRN KAEDADIEEM ALKLDDVLGD VGQGPAL.

Q&A

What is SEPSECS and what is its role in selenoprotein biosynthesis?

SEPSECS (O-Phosphoseryl-tRNA:selenocysteinyl-tRNA synthase) catalyzes the final step in selenocysteine (Sec) biosynthesis, converting O-phosphoseryl-tRNA^Sec (Sep-tRNA^Sec) to selenocysteinyl-tRNA^Sec using selenophosphate as the selenium donor. This process is essential because:

  • Selenocysteine is the 21st amino acid and is incorporated into selenoproteins

  • The reaction is tRNA-dependent, with SEPSECS recognizing specific features of tRNA^Sec

  • The process is PLP (pyridoxal phosphate)-dependent

The crystal structure of human tRNA^Sec in complex with SepSecS demonstrates that two tRNA^Sec molecules bind to each SepSecS tetramer through their 13-base pair acceptor-TΨC arm (where Ψ indicates pseudouridine) . This binding induces a conformational change in the enzyme's active site that properly orients the phosphoserine attached to tRNA^Sec for the reaction to occur.

What is the significance of the SEPSECS Y334C mouse model in selenium research?

The SEPSECS Y334C mouse model recapitulates a human pathogenic variant identified in patients with progressive cerebello-cerebral atrophy. This model provides several important insights:

  • Homozygous Y334C/Y334C mice die perinatally with signs of cardio-respiratory failure

  • The phenotype resembles Sedaghatian spondylometaphyseal dysplasia caused by mutations in glutathione peroxidase 4 (GPX4)

  • Protein expression levels of selenoproteins are generally reduced in brain and isolated cortical neurons

  • Transcriptomics analysis reveals upregulation of NRF2-regulated genes, indicating an antioxidant stress response

Importantly, crossbreeding Y334C/Y334C Sepsecs mice with mice harboring a selenium-independent GPX4 (where selenocysteine is replaced by cysteine) rescues perinatal death, demonstrating that GPX4 deficiency is the critical driver of the lethal phenotype .

How does the SEPSECS Y334C mouse model differ from what is observed in human SEPSECS-related disorders?

The SEPSECS Y334C mouse model shows notable differences from human SEPSECS-related disorders:

FeatureSEPSECS Y334C MiceHuman SEPSECS Disorders
SurvivalPerinatal deathLonger survival with progressive deterioration
Primary manifestationCardio-respiratory failureProgressive cerebello-cerebral atrophy
OnsetImmediate postnatalVariable, often infantile or childhood
GPX4 relationshipClear rescue with selenium-independent GPX4Not established in humans

These differences suggest that while the biochemical defect is similar, there may be species-specific requirements for selenoproteins during development or differences in compensatory mechanisms. The mouse model may represent a more severe manifestation of selenoprotein deficiency than typically seen in human patients with SEPSECS mutations.

What are the key techniques for generating SEPSECS mouse models?

The generation of SEPSECS mouse models involves several sophisticated genetic engineering approaches:

  • Site-directed mutagenesis approach (Y334C model):

    • Homologous recombination in embryonic stem cells

    • Introduction of the Y334C mutation in exon 8 of the Sepsecs gene

    • Addition of silent mutations to create restriction sites for genotyping

  • Vector construction process:

    • PCR amplification of homologous arms using appropriate primers

    • Cloning into intermediary vectors (e.g., pGEMT-Easy)

    • Introduction of mutations using site-directed mutagenesis

    • Assembly into final targeting constructs

  • ES cell targeting:

    • Linearization of the targeting construct

    • Electroporation into embryonic stem cells

    • Selection of correctly targeted clones

    • Blastocyst injection and chimera generation

This process requires careful design to ensure the mutation is properly introduced while minimizing disruption to gene regulation and expression patterns.

What methods are most effective for analyzing selenoprotein expression in SEPSECS mouse tissues?

Analysis of selenoprotein expression in SEPSECS mouse tissues requires specialized approaches:

  • Western blotting:

    • Use of selenoprotein-specific antibodies (e.g., anti-GPX4, anti-SELENOP)

    • Careful sample preparation with reducing agents to prevent selenoprotein oxidation

    • Quantification relative to housekeeping proteins

  • Enzymatic activity assays:

    • Glutathione peroxidase activity assays for GPX4 and other GPXs

    • Thioredoxin reductase activity measurements

    • Deiodinase activity assays for thyroid hormone metabolism

  • 75Se metabolic labeling:

    • In vivo or ex vivo labeling with radioactive selenium

    • Autoradiography to visualize the complete selenoproteome

    • Quantitative comparison between genotypes

  • Mass spectrometry approaches:

    • Targeted proteomics for specific selenoproteins

    • Global proteomics to assess compensatory responses

    • Detection of selenocysteine-containing peptides

In SEPSECS Y334C mice, these methods have revealed generally reduced selenoprotein expression across tissues, with variations in the degree of reduction between different selenoproteins .

How can researchers design rescue experiments for SEPSECS-deficient mice?

Designing rescue experiments for SEPSECS-deficient mice requires careful consideration of several factors:

  • Target identification:

    • Determine which selenoprotein deficiency drives the phenotype

    • For SEPSECS Y334C mice, GPX4 was identified as the critical selenoprotein

  • Rescue construct design:

    • For GPX4 rescue, a selenium-independent version (Sec→Cys) was used

    • Consider appropriate promoters (ubiquitous vs. tissue-specific)

    • Include traceable markers (fluorescent proteins, epitope tags)

  • Delivery methods:

    • Transgenic expression through crossbreeding

    • Viral vector delivery (AAV, lentivirus)

    • Potential for mRNA or protein-based approaches

  • Validation of rescue:

    • Survival assessment

    • Molecular marker normalization

    • Histopathological evaluation

    • Assessment of residual phenotypes

The successful rescue of SEPSECS Y334C/Y334C mice with selenium-independent GPX4 provides a valuable blueprint for this approach, demonstrating that specific selenoprotein replacement can overcome the lethal consequences of global selenoprotein deficiency .

What are the analytical challenges in assessing the NRF2 pathway activation in SEPSECS mouse models?

Assessing NRF2 pathway activation in SEPSECS mouse models presents several analytical challenges:

  • Temporal dynamics:

    • Determining whether NRF2 activation is an early or late event

    • Capturing the dynamics of pathway activation across development

  • Tissue heterogeneity:

    • Different cell types may show varying degrees of NRF2 activation

    • Bulk tissue analysis may mask cell-specific responses

  • Specificity of markers:

    • Identifying NRF2-specific target genes versus general stress responses

    • Validating transcriptional changes at the protein level

  • Causality determination:

    • Establishing whether NRF2 activation is protective or maladaptive

    • Determining if NRF2 activation is a direct response to selenoprotein deficiency

  • Methodological approaches:

    • ChIP-seq for direct NRF2 binding assessment

    • RNA-seq for transcriptional profiling

    • Proteomics for validation of protein-level changes

    • Single-cell approaches to address cellular heterogeneity

Transcriptomics analysis in SEPSECS Y334C mice has revealed upregulation of NRF2-regulated genes, suggesting this pathway represents a compensatory response to increased oxidative stress resulting from selenoprotein deficiency .

What are the major phenotypic differences between heterozygous and homozygous SEPSECS Y334C mice?

The phenotypic differences between heterozygous and homozygous SEPSECS Y334C mice are substantial:

FeatureHeterozygous (Y334C/+)Homozygous (Y334C/Y334C)
ViabilityViable and fertilePerinatal death within 24 hours
GrowthNormalN/A due to early lethality
Selenoprotein levelsMildly reducedSignificantly reduced
NRF2 pathwayMinimal activationStrong upregulation
Cardio-respiratory functionNormalSevere dysfunction leading to death
Embryonic developmentNormalNormal until birth

These differences highlight the dosage-sensitive nature of SEPSECS function, where one functional allele is sufficient for survival but two mutant alleles result in lethal selenoprotein deficiency. The normal embryonic development suggests that the critical requirement for selenoproteins, particularly GPX4, becomes acute at the transition to air breathing .

What is the mechanism by which the Y334C mutation affects SEPSECS function?

The Y334C mutation affects SEPSECS function through several potential mechanisms:

  • Catalytic efficiency:

    • Tyrosine 334 may be involved in the active site or substrate positioning

    • Mutation to cysteine likely reduces catalytic efficiency

  • Structural impact:

    • May affect protein folding, stability, or tetramer formation

    • Could disrupt PLP cofactor binding or positioning

  • tRNA interaction:

    • May alter the binding affinity for tRNA^Sec

    • Could disrupt conformational changes induced by tRNA binding

  • Biochemical consequences:

    • Reduced conversion of Sep-tRNA^Sec to Sec-tRNA^Sec

    • Decreased incorporation of selenocysteine into selenoproteins

The result is a general reduction in selenoprotein levels rather than selective deficiencies, suggesting the mutation affects the core catalytic function of SEPSECS. The human SepSecS-tRNA^Sec complex structure reveals that tRNA binding induces conformational changes in the enzyme's active site that properly orient the phosphoserine for the reaction , a process that may be compromised by the Y334C mutation.

How does selenoprotein hierarchy influence the phenotypic manifestations in SEPSECS mouse models?

Selenoprotein hierarchy significantly shapes the phenotypic manifestations in SEPSECS mouse models:

  • Differential sensitivity:

    • Under limited selenocysteine availability, certain selenoproteins maintain near-normal levels while others show dramatic decreases

    • GPX4 appears to be most critically affected in SEPSECS Y334C mice

  • Tissue-specific effects:

    • Heart and lung show particular vulnerability, explaining the cardio-respiratory failure

    • Brain shows reduced selenoprotein expression but does not cause immediate lethality

  • Developmental stage dependency:

    • The hierarchy shifts during development, with GPX4 becoming acutely critical at birth

    • Transition to air breathing likely increases oxidative stress, requiring optimal GPX4 function

  • Rescue implications:

    • Selenium-independent GPX4 expression rescues lethality despite continued deficiency of other selenoproteins

    • This demonstrates that GPX4 deficiency is the critical driver of perinatal death

This hierarchical organization explains why specific phenotypes emerge despite global selenoprotein deficiency and provides insights into potential therapeutic targeting priorities.

What is the relationship between SEPSECS deficiency, GPX4, and ferroptosis?

The relationship between SEPSECS deficiency, GPX4, and ferroptosis represents a key mechanistic link:

  • GPX4 as a ferroptosis regulator:

    • GPX4 reduces lipid hydroperoxides in biological membranes

    • This prevents iron-dependent lipid peroxidation (ferroptosis)

  • SEPSECS deficiency impact:

    • Reduced SEPSECS activity decreases selenocysteine incorporation

    • This leads to decreased GPX4 levels below critical thresholds

  • Tissue vulnerability patterns:

    • Tissues with high metabolic rates and oxygen consumption (heart, brain) are particularly vulnerable

    • This explains the cardio-respiratory failure in SEPSECS Y334C mice

  • Rescue mechanism:

    • Selenium-independent GPX4 (with cysteine replacing selenocysteine) prevents ferroptosis

    • This is sufficient to rescue perinatal death despite continued deficiency of other selenoproteins

  • NRF2 pathway activation:

    • Upregulation of NRF2-regulated genes likely represents a compensatory response to ferroptotic pressure

    • This persists even after GPX4 rescue, suggesting ongoing oxidative stress

This relationship explains why SEPSECS deficiency phenotypically resembles conditions caused by GPX4 mutations and highlights ferroptosis inhibition as a potential therapeutic approach.

How can SEPSECS mouse models inform our understanding of human selenoprotein-related diseases?

SEPSECS mouse models provide valuable insights into human selenoprotein-related diseases:

  • Disease mechanism elucidation:

    • Reveal critical selenoproteins (e.g., GPX4) for specific phenotypes

    • Identify pathways (ferroptosis, NRF2 activation) involved in pathogenesis

  • Phenotypic spectrum interpretation:

    • Explain why different selenoprotein mutations produce distinct clinical presentations

    • Provide context for variable severity and progression in human patients

  • Biomarker identification:

    • NRF2 target genes as potential disease biomarkers

    • Ferroptosis products as indicators of disease activity

  • Cross-disease relationships:

    • Connect SEPSECS deficiency with Sedaghatian spondylometaphyseal dysplasia (GPX4 mutations)

    • Suggest common mechanisms across selenoprotein disorders

  • Species differences awareness:

    • Highlight important differences between mouse and human selenoprotein requirements

    • Caution against direct extrapolation without cross-species validation

The successful rescue of SEPSECS Y334C mice with selenium-independent GPX4 suggests that targeting specific critical selenoproteins might be a viable approach for human selenoprotein disorders, even when the primary defect affects global selenoprotein synthesis .

What approaches can be used to identify the mechanisms behind the differential tissue sensitivity to SEPSECS deficiency?

Identifying mechanisms of differential tissue sensitivity to SEPSECS deficiency requires multi-faceted approaches:

  • Comparative tissue transcriptomics:

    • RNA-seq of affected versus resistant tissues

    • Analysis of selenoprotein mRNA expression patterns

    • Identification of tissue-specific compensatory pathways

  • Tissue-specific proteomics:

    • Quantification of selenoprotein levels across tissues

    • Correlation with phenotypic vulnerability

    • Analysis of post-translational modifications

  • Metabolic profiling:

    • Assessment of redox status in different tissues

    • Measurement of ferroptosis markers (lipid peroxidation products)

    • Evaluation of energy metabolism differences

  • Conditional knockout strategies:

    • Tissue-specific deletion of SEPSECS

    • Determination of tissue-autonomous versus systemic effects

    • Temporal control to identify developmental windows of vulnerability

  • Single-cell analysis:

    • Identification of particularly vulnerable cell types within tissues

    • Cell-specific selenium utilization patterns

    • Cellular heterogeneity in compensatory responses

  • Ex vivo tissue culture:

    • Comparative survival of different tissues under selenium limitation

    • Response to ferroptosis inducers and inhibitors

    • Rescue experiments with selenium-independent selenoprotein analogs

These approaches would help explain why cardio-respiratory tissues show particular vulnerability in SEPSECS Y334C mice while other tissues can tolerate the mutation at least through embryonic development .

What therapeutic strategies might be developed based on insights from SEPSECS mouse models?

Insights from SEPSECS mouse models suggest several promising therapeutic strategies:

  • Selenium-independent selenoprotein analogs:

    • Development of modified selenoproteins with cysteine replacing selenocysteine

    • Potential gene therapy approaches delivering modified GPX4

    • Focus on the most critical selenoproteins for specific phenotypes

  • Ferroptosis inhibitors:

    • Small molecules that prevent ferroptotic cell death

    • Compounds like ferrostatin-1, liproxstatin-1, or derivatives

    • Potential to functionally mimic GPX4 activity

  • NRF2 pathway modulation:

    • Compounds that activate the NRF2 antioxidant response

    • Enhancement of endogenous compensatory mechanisms

    • Combined approach with selenoprotein replacement

  • Selenium supplementation strategies:

    • Optimized forms of selenium with enhanced bioavailability

    • Targeted delivery to most vulnerable tissues

    • Potential to enhance residual SEPSECS activity

  • Gene therapy approaches:

    • AAV-mediated delivery of functional SEPSECS

    • Delivery of selenium-independent GPX4 (as validated in mouse models)

    • CRISPR/Cas9-based correction of specific mutations

  • Cell-based therapies:

    • Transplantation of cells with normal selenoprotein synthesis

    • Ex vivo corrected patient cells

    • Focus on most affected tissues

The successful rescue of SEPSECS Y334C mice with selenium-independent GPX4 provides a particularly promising proof-of-concept for selenoprotein replacement strategies even when the primary defect affects the selenocysteine incorporation machinery .

How might chemical biology approaches be used to further investigate SEPSECS function and develop potential therapeutics?

Chemical biology approaches offer powerful tools for investigating SEPSECS function and developing therapeutics:

  • High-throughput screening:

    • Identification of small molecules that enhance residual SEPSECS activity

    • Screening for compounds that stabilize mutant SEPSECS proteins

    • Discovery of GPX4 mimetics that prevent ferroptosis

  • Structure-based drug design:

    • Using the human SepSecS-tRNA^Sec complex structure

    • Development of compounds that facilitate SEPSECS-tRNA interaction

    • Design of molecular chaperones that stabilize SEPSECS folding

  • Chemical genetics:

    • Development of small-molecule inducible SEPSECS systems

    • Temporal control over selenoprotein synthesis

    • Determination of critical windows for intervention

  • Selenocysteine analogs:

    • Development of novel selenocysteine derivatives with enhanced properties

    • Creation of non-hydrolyzable selenocysteinyl-tRNA analogs

    • Compounds that can bypass the SEPSECS-dependent pathway

  • Targeted protein degradation:

    • Identification of proteins whose degradation might compensate for selenoprotein deficiency

    • Development of PROTACs targeting ferroptosis sensitizers

    • Selective degradation of negative regulators of NRF2

  • Bioorthogonal chemistry:

    • Tools for tracking selenoprotein synthesis in vivo

    • Click chemistry approaches for monitoring spatial and temporal aspects of selenoprotein function

    • Development of selenocysteine tagging systems for proteomics

These approaches could lead to novel research tools for understanding SEPSECS biology and potential therapeutic candidates for SEPSECS-related disorders.

What are the key challenges in developing viable mouse models for studying the neurological aspects of SEPSECS deficiency?

Developing viable mouse models for studying neurological aspects of SEPSECS deficiency presents several challenges:

  • Overcoming perinatal lethality:

    • Conditional knockout approaches (e.g., Cre-loxP systems)

    • Inducible systems to bypass critical developmental windows

    • Hypomorphic alleles with residual activity

  • Brain-specific manipulation:

    • Neuron-specific or glial-specific SEPSECS deletion

    • Regional targeting (e.g., cerebellum-specific) to model specific aspects

    • Spatiotemporal control of SEPSECS inactivation

  • Balancing severity:

    • Creating models with sufficient deficiency to produce neurological phenotypes

    • Avoiding overwhelming systemic phenotypes that preclude neurological assessment

    • Calibrating selenium availability to modulate phenotype severity

  • Recapitulating progressive nature:

    • Developing models that show progressive neurodegeneration

    • Long-term studies to capture late-onset phenotypes

    • Aging effects on selenoprotein requirements

  • Validation approaches:

    • Confirmation of selenoprotein deficiency in neural tissues

    • Correlation with human pathology

    • Functional assessment of neurological phenotypes

A promising approach might be to use the selenium-independent GPX4 rescue strategy to overcome perinatal lethality while still allowing the study of neurological consequences of other selenoprotein deficiencies.

How can researchers address the discrepancy between mouse and human phenotypes in SEPSECS deficiency?

Addressing the discrepancy between mouse and human SEPSECS deficiency phenotypes requires multi-faceted approaches:

  • Humanized mouse models:

    • Introduction of human SEPSECS variants into mice

    • Creation of mice expressing human selenoproteins

    • Chimeric models with human neural cells

  • Systematic mutation analysis:

    • Testing different human SEPSECS mutations in mice

    • Comparing hypomorphic versus null alleles

    • Identifying mutation-specific effects

  • Cross-species comparisons:

    • Detailed comparison of selenoprotein hierarchies between species

    • Analysis of tissue-specific selenoprotein expression patterns

    • Evaluation of compensatory mechanism differences

  • Human cellular models:

    • Patient-derived iPSCs differentiated into relevant cell types

    • CRISPR-engineered human cell lines with SEPSECS mutations

    • Organoids to model tissue-specific effects

  • Developmental timing analysis:

    • Careful staging of selenoprotein requirements across development

    • Identification of species-specific critical windows

    • Temporal manipulation of SEPSECS function

  • Genetic background effects:

    • Testing SEPSECS mutations on different mouse strain backgrounds

    • Creating mouse genetic diversity panels with SEPSECS mutations

    • Identifying genetic modifiers

Understanding these species differences is crucial for translating findings from mouse models to human therapeutics and may reveal important aspects of selenoprotein biology evolution.

What methodological innovations would advance our understanding of selenoprotein hierarchy in SEPSECS mouse models?

Several methodological innovations could significantly advance our understanding of selenoprotein hierarchy:

  • Advanced proteomics approaches:

    • Single-cell proteomics to resolve cellular heterogeneity

    • Absolute quantification of all selenoproteins simultaneously

    • Monitoring selenoprotein turnover rates with pulse-chase techniques

  • Real-time selenoprotein synthesis monitoring:

    • Fluorescent reporters for selenoprotein translation

    • CRISPR activation/repression screens for hierarchy regulators

    • Optogenetic control of SEPSECS activity

  • Tissue-specific selenoproteome manipulation:

    • Selective depletion of individual selenoproteins in SEPSECS backgrounds

    • Combinatorial selenoprotein knockout/rescue experiments

    • Selenoprotein competition assays in limiting conditions

  • Ribosome profiling adaptations:

    • Specialized techniques to capture UGA recoding events

    • tRNA-ribosome interactions during selenocysteine incorporation

    • Translation efficiency determinants for selenoprotein mRNAs

  • Multi-omics integration:

    • Correlation of transcriptome, selenoproteome, and metabolome data

    • Network analysis of selenoprotein hierarchy regulation

    • Mathematical modeling of selenoprotein synthesis under limiting conditions

  • In vivo selenoprotein activity sensors:

    • Genetically encoded sensors for specific selenoprotein activities

    • Real-time monitoring of selenoprotein function in living tissues

    • Correlation of activity with expression levels

These innovations would provide more dynamic and comprehensive views of selenoprotein hierarchy, helping to explain the tissue-specific and developmental consequences of SEPSECS deficiency.

What are the most promising future directions for research on SEPSECS and selenoprotein biology using mouse models?

The most promising future directions for SEPSECS and selenoprotein biology research include:

  • Integrating ferroptosis and selenoprotein biology:

    • Comprehensive analysis of tissue-specific ferroptosis susceptibility

    • Identification of selenoproteins beyond GPX4 involved in ferroptosis regulation

    • Development of targeted ferroptosis modulators for selenoprotein disorders

  • Exploring nutritional and environmental influences:

    • Interaction between dietary selenium and SEPSECS efficiency

    • Environmental stressors that exacerbate selenoprotein deficiency

    • Maternal-fetal selenium transfer in pregnancy

  • Investigating metabolic consequences:

    • Role of selenoproteins in metabolic regulation

    • Connecting selenoprotein deficiency to mitochondrial dysfunction

    • Exploring selenoprotein requirements in different metabolic states

  • Defining critical developmental windows:

    • Temporal requirement mapping for selenoproteins during development

    • Identification of developmental processes most sensitive to selenoprotein deficiency

    • Intervention timing optimization

  • Therapeutic translation:

    • Development of selenium-independent versions of critical selenoproteins

    • Ferroptosis inhibitor optimization for clinical application

    • Gene therapy approaches for selenoprotein disorders

  • Selenoproteins in aging and neurodegeneration:

    • Role of selenoprotein deficiency in age-related neurodegeneration

    • Selenium status, selenoprotein function, and cognitive decline

    • Neuroprotective strategies based on selenoprotein pathway modulation

The successful rescue of SEPSECS Y334C mice with selenium-independent GPX4 provides a foundational proof-of-concept that could be expanded to address various selenoprotein-related disorders, potentially transforming treatment approaches for these conditions.

Product Science Overview

Introduction

Selenocysteinyl-tRNA(Sec) synthase, often abbreviated as SepSecS, is a crucial enzyme involved in the biosynthesis of selenocysteine (Sec), the 21st amino acid. This enzyme is particularly significant in the context of selenoprotein synthesis, where selenocysteine is incorporated into proteins at specific UGA codons, which typically signal stop codons in the genetic code .

Selenocysteine Biosynthesis

Selenocysteine is synthesized on its cognate transfer RNA (tRNA), known as tRNA(Sec). The biosynthesis of selenocysteine involves several steps:

  1. Aminoacylation: Initially, tRNA(Sec) is aminoacylated with serine by seryl-tRNA synthetase (SerRS) to form Ser-tRNA(Sec) .
  2. Phosphoseryl-tRNA(Sec) Formation: The serine attached to tRNA(Sec) is then phosphorylated to form O-phosphoseryl-tRNA(Sec) (Sep-tRNA(Sec)) .
  3. Selenocysteinyl-tRNA(Sec) Formation: Finally, SepSecS catalyzes the conversion of Sep-tRNA(Sec) to selenocysteinyl-tRNA(Sec) (Sec-tRNA(Sec)) . This step is pyridoxal phosphate-dependent and involves the replacement of the phosphate group with selenium .
Structural Insights

The crystal structure of SepSecS in complex with tRNA(Sec), phosphoserine, and thiophosphate has provided significant insights into the enzyme’s mechanism. The structure reveals that two tRNA(Sec) molecules bind to each SepSecS tetramer through their 13-base pair acceptor-TPsiC arm . This binding likely induces a conformational change in the enzyme’s active site, allowing the proper orientation of phosphoserine for the reaction to occur .

Biological Significance

Selenocysteine is an essential component of several selenoproteins, which play critical roles in various biological processes, including antioxidant defense, thyroid hormone metabolism, and redox homeostasis . Defects in the biosynthesis or incorporation of selenocysteine can lead to various pathological conditions .

Recombinant Production

The recombinant production of mouse SepSecS involves cloning the gene encoding the enzyme into an expression vector, followed by expression in a suitable host system, such as E. coli. The recombinant enzyme can then be purified using affinity chromatography techniques. This recombinant enzyme is valuable for studying the detailed mechanisms of selenocysteine biosynthesis and for potential therapeutic applications.

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.