SEPW1 Human

Selenoprotein W 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Characteristics

Gene Structure:

  • Located at 19q13.11, the SEPW1 gene contains a UGA codon that encodes selenocysteine instead of functioning as a stop codon. This requires a SECIS element (selenocysteine insertion sequence) in the 3' untranslated region (UTR) .

  • A retroprocessed pseudogene, SEPW1P, is found on chromosome 1p35-34 .

Protein Features:

  • Molecular weight: ~9.8 kDa .

  • Contains a thioredoxin-like fold with a conserved Cys-X-X-Sec motif, enabling antioxidant activity .

Tissue Distribution and Expression

SEPW1 exhibits tissue-specific expression, with highest levels in skeletal muscle, heart, and brain . The table below summarizes key expression data:

TissueExpression LevelSource
Skeletal muscleHigh
HeartHigh
Brain (neurons)Moderate-High
LiverLow
TestisModerate

Synaptic localization of SEPW1 has been confirmed in mouse models, particularly in hippocampal and cerebellar neurons .

Antioxidant Activity

SEPW1 functions as a glutathione-dependent antioxidant, mitigating oxidative damage by reducing hydrogen peroxide and lipid hydroperoxides . Key findings include:

  • Overexpression in H1299 cells reduces susceptibility to H₂O₂-induced stress .

  • Protects neuronal cells against methylmercury toxicity .

Immune Regulation

  • SEPW1 expression increases in response to selenium supplementation, enhancing immune response to challenges like influenza vaccination .

  • SEPW1 mRNA levels rise post-vaccination, correlating with selenium dose (50–200 µg/day) .

Neurological Functions

  • Localized to synapses and dendritic arbors in the brain, SEPW1 may support synaptic plasticity .

  • Co-immunoprecipitates with Staufen 2, an RNA-binding protein, suggesting localized translation in neurons .

Regulatory Mechanisms

SEPW1 expression is tightly regulated by selenium availability:

  • Dietary selenium deficiency reduces SEPW1 levels in muscle and brain .

  • Supplementation with Se-enriched yeast or onions upregulates SEPW1 mRNA, with optimal effects at 50 µg/day .

Key Regulatory Pathways:

  • p53-dependent cell cycle arrest: SEPW1 silencing activates MKK4-p38γ/JNK2 pathways, inducing G₁ arrest via p53 phosphorylation .

  • Synaptic expression is reduced in Sepp1-knockout mice, linking SEPW1 to selenium transport .

Biomarker Potential

  • SEPW1 mRNA levels correlate with selenium status, making it a candidate biomarker for deficiency .

  • Downregulated in colon tissue during selenium depletion, highlighting its role in gut redox balance .

Therapeutic Targets

  • Neurodegenerative diseases: Synaptic SEPW1 loss in selenium-deficient models suggests a role in disorders like Alzheimer’s .

  • Cancer: SEPW1 modulates oxidative stress responses, influencing tumor progression .

Product Specs

Introduction
Selenoprotein W 1, also called SEPW1, is a selenoprotein containing a selenocysteine (Sec) residue at its active site. This selenocysteine is encoded by the UGA codon, typically a stop codon for translation. However, the 3' UTR of selenoprotein genes possesses a common stem-loop structure known as the sec insertion sequence (SECIS). SECIS enables the recognition of UGA as a Sec codon rather than a stop signal. SEPW1 is most abundantly expressed in skeletal muscle and heart tissues. It plays a role in oxidation-reduction reactions.
Description
Recombinant human SEPW1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 110 amino acids (amino acids 1-87). It has a molecular weight of 11.8 kDa. SEPW1 is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The SEPW1 protein solution has a concentration of 0.25 mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 0.15M NaCl, 20% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Selenoprotein W, 1, SelW, Selenoprotein W.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMALAVRV VYCGACGYKS KYLQLKKKLE DEFPGRLDIC GEGTPQATGF FEVMVAGKLI HSKKKGDGYV DTESKFLKLV AAIKAALAQG.

Q&A

What is SEPW1 and what is its primary function in human biology?

Selenoprotein W (SEPW1) is a small selenium-containing protein (approximately 10 kDa) that belongs to the selenoprotein family, characterized by the incorporation of selenocysteine, the 21st amino acid. SEPW1 is abundantly expressed in human brain and muscle tissues, with particularly high expression in neuronal structures. Its primary function appears to be antioxidant protection, with evidence suggesting it plays a crucial role in cellular redox regulation through a CXXU motif (where C is cysteine and U is selenocysteine) that participates in thiol-disulfide exchange reactions .

The protein demonstrates tissue-specific expression patterns and is involved in multiple cellular processes. Research has established that SEPW1 contributes to cell cycle regulation, as depletion studies show it induces p53- and p21-dependent cell cycle arrest. Its high concentration in neuronal tissues further suggests important functions in brain development and neuronal signaling . The protein's expression is selenium-dependent, making it a potential biomarker for selenium status in human studies.

How is SEPW1 expression measured in human research samples?

Several methodological approaches are employed to quantify SEPW1 expression in human samples, each with specific advantages and limitations. The most common technique is quantitative RT-PCR (RT-qPCR), which provides high sensitivity for measuring SEPW1 mRNA levels in peripheral blood mononuclear cells (PBMCs) and other accessible tissues . This approach requires careful selection of reference genes for normalization to ensure accurate quantification.

For protein-level detection, Western blotting with specific antibodies against human SEPW1 is frequently employed, though this can be challenging due to the small size of the protein. Immunohistochemistry and immunofluorescence techniques allow visualization of SEPW1 distribution in tissue sections, providing valuable spatial information about expression patterns and enabling co-localization studies with other cellular markers .

More advanced approaches include RNA sequencing (RNA-Seq) for comprehensive transcriptomic analysis and selenium-specific detection methods such as inductively coupled plasma mass spectrometry (ICP-MS) to measure total selenium levels. These techniques should be selected based on research questions, available samples, and required sensitivity for detecting changes in expression levels.

What is the relationship between SEPW1 and selenium status in humans?

The expression of SEPW1 depends on the selenium transporter selenoprotein P (Sepp1), with studies showing reduced SEPW1 in synaptic regions of Sepp1 knockout mice . This dependency highlights the importance of considering selenium transport mechanisms when studying SEPW1 expression patterns.

Intervention studies demonstrate that SEPW1 expression responds to selenium supplementation in a dose-dependent manner. In human supplementation trials, SEPW1 mRNA levels increased significantly in subjects receiving 50 μg/day selenium-enriched onions compared to those consuming unenriched onions . Interestingly, higher doses may not always produce proportionally greater increases, as evidenced by lower SEPW1 mRNA levels observed in the 200 μg/day selenium-yeast group at certain time points.

Where is SEPW1 primarily expressed in human brain tissues?

SEPW1 demonstrates widespread expression throughout the brain, with particularly high concentrations in specific neuronal populations. Based on studies in mouse models (which share significant homology with human SEPW1), the protein is abundant in both neuronal cell bodies and their processes . Pyramidal neurons of the cortex and hippocampus express high levels of SEPW1, suggesting its importance in these regions associated with higher cognitive functions and memory formation.

Particularly striking is the high expression observed in Purkinje neurons of the cerebellum and their extensive dendritic arbors . These large neurons are critical for motor coordination and certain forms of learning, indicating potential roles for SEPW1 in these functions. The presence of SEPW1 in neuronal processes and at synapses further suggests involvement in synaptic function and potentially in local protein synthesis.

Analysis of synaptosome fractions has confirmed the presence of SEPW1 at synapses, along with several proteins involved in selenoprotein synthesis . This synaptic localization is particularly interesting in light of evidence that SEPW1 mRNA coimmunoprecipitates with Staufen 2 protein in human neuronal cell lines, suggesting roles in RNA transport and localized translation—processes crucial for synaptic plasticity and function.

What methodological considerations are critical when designing selenium supplementation studies to assess SEPW1 response?

Designing rigorous selenium supplementation studies to assess SEPW1 response requires careful attention to multiple methodological factors. Researchers must consider dose selection, supplementation form, timing, and appropriate controls to obtain reliable results.

The dose-response relationship is a critical consideration, as different selenium doses produce varying effects on SEPW1 expression. Research indicates that 50 μg/day selenium-enriched onions can significantly increase SEPW1 mRNA compared to unenriched controls, while higher doses (200 μg/day) might actually reduce expression in certain contexts . Therefore, studies should include multiple dose groups with appropriate intervals to capture non-linear responses.

The chemical form of selenium significantly impacts bioavailability and subsequent effects on gene expression. Organic forms (selenomethionine, selenocysteine) generally have higher bioavailability than inorganic forms (selenite, selenate). Research protocols should clearly specify the chemical form used and consider testing multiple forms when feasible .

Selenium FormTypical BioavailabilityCommon SourcesConsiderations for SEPW1 Research
SelenomethionineHighSe-enriched yeast, plant sourcesNon-specific incorporation into proteins
SelenocysteineMedium-HighAnimal productsDirect incorporation into selenoproteins
Sodium seleniteMediumSupplementsRapid absorption, potential toxicity at high doses
Se-enriched foodsVariableSe-enriched onions, garlic, etc.Food matrix effects on bioavailability

Timing considerations are essential, as SEPW1 expression changes over the supplementation period are often time-dependent. Study designs should include multiple sampling time points to capture temporal dynamics of expression changes . Baseline measurements are crucial, as individual variation in selenium status will influence response to supplementation.

How should researchers account for individual variability in SEPW1 expression when designing human studies?

Individual variability in SEPW1 expression presents a significant challenge for human studies. Multiple factors contribute to this variability, including genetic polymorphisms, baseline selenium status, age, sex, and environmental factors. Addressing these sources of variation requires strategic approaches to study design and analysis.

Sample size calculation is critical and should account for expected individual variability. Based on experimental design principles, a minimum of three biological replicates is necessary, but human studies typically require much larger sample sizes to achieve adequate statistical power . Power analyses should be conducted using realistic effect size estimates based on preliminary data or published literature.

Genetic factors significantly influence SEPW1 expression and function. Several single nucleotide polymorphisms (SNPs) in selenoprotein genes affect protein synthesis efficiency and function. Researchers should consider genotyping participants for key selenoprotein pathway SNPs and including genetic variation as a covariate in analyses, particularly for smaller studies where randomization may not fully balance genetic factors.

Baseline selenium status varies widely between individuals and populations, affecting the response to supplementation. Measuring baseline selenium using multiple biomarkers (plasma selenium, SELENOP, GPX3 activity) provides a more complete picture of selenium status. Stratified analysis or statistical adjustment for baseline status should be incorporated into analytical plans .

Longitudinal study designs with repeated measures can help distinguish treatment effects from individual variability. Mixed-effects statistical models are particularly useful for analyzing such data, as they can account for within-subject correlation while estimating population-level effects.

What evidence exists for SEPW1's role in neuronal function, and how can researchers effectively study this relationship?

Evidence for SEPW1's role in neuronal function comes from its high expression in neuronal tissues and specific subcellular localization patterns. Research shows widespread distribution in neurons and neuropil of the brain, with particularly high expression in pyramidal neurons of the cortex and hippocampus, as well as Purkinje neurons of the cerebellum . Its presence at synapses and association with RNA transport proteins suggests involvement in synaptic function and potentially in local protein synthesis.

Examining SEPW1's neuronal functions requires specialized methodological approaches. Cell-specific expression analysis using single-cell RNA sequencing can identify neuronal subtypes with particularly high SEPW1 expression. This can be complemented by in situ hybridization combined with immunohistochemistry for spatial mapping of expression patterns . For subcellular localization, synaptosome fractionation techniques have successfully demonstrated SEPW1 presence at synapses, while super-resolution microscopy can provide more precise subcellular localization.

Functional studies require careful manipulation of SEPW1 expression. CRISPR/Cas9-mediated knockout or knockdown in neuronal cultures allows assessment of direct functional consequences. For in vivo studies, conditional knockout mouse models with neuron-specific deletion are preferable to global knockouts, which may have confounding developmental effects. Electrophysiological recordings can then assess functional consequences on neuronal activity and synaptic transmission.

The association between SEPW1 mRNA and Staufen 2 protein suggests roles in RNA transport and localized translation . This can be investigated using ribosome profiling to study translation efficiency and FRAP (Fluorescence Recovery After Photobleaching) to examine local protein synthesis. RNA immunoprecipitation followed by sequencing (RIP-seq) can identify other RNAs that may be co-regulated with SEPW1 in neuronal compartments.

How does SEPW1 interact with the immune system, and what are the best methodological approaches to study this relationship?

While direct evidence for SEPW1's role in immune function is limited in the search results, research on related selenoproteins suggests potential immune regulatory functions. Study findings indicate that selenium supplementation influences selenoprotein expression in response to immune challenges such as influenza vaccination, with SEPS1 (another selenoprotein) showing increased mRNA levels 7 days post-vaccination in a dose-dependent manner related to selenium supplementation . This suggests SEPW1 may have similar immune-responsive properties.

Methodological approaches to study SEPW1-immune interactions should include controlled immune challenges. Vaccination serves as a standardized immune stimulus that allows for time-course analysis of SEPW1 expression following challenge. Researchers should collect samples at multiple time points (pre-vaccination and several post-vaccination time points) to capture dynamic expression changes and correlate these with markers of immune activation .

Cell-type specific analysis is essential, as immune responses involve multiple cell populations with distinct functions. Isolation of specific immune cell populations (T cells, B cells, monocytes) followed by analysis of differential SEPW1 expression can identify which immune cells are most responsive to selenium supplementation. Flow cytometry-based approaches can complement this with protein-level assessment in specific cell subsets.

Functional immune parameters should be measured concurrently with SEPW1 expression. These include cytokine production, lymphocyte proliferation, antibody response quantification, and neutrophil function tests. Correlation between SEPW1 expression changes and these functional parameters can provide insights into the biological significance of expression changes.

What are the common experimental design errors in SEPW1 research, and how can they be avoided?

Several experimental design errors can compromise SEPW1 research validity. Understanding these common pitfalls and implementing appropriate mitigation strategies is essential for producing reliable results.

Research ScenarioMinimum ReplicationPreferred ReplicationJustification
Cell culture experiments3 independent cultures5+ independent culturesControls for culture variability
Animal studies5-6 animals per group8-12 animals per groupAccounts for biological variation
Human observational studies20+ subjects per group50+ subjects per groupAccommodates human heterogeneity
Human intervention studies15+ subjects per arm30+ subjects per armAllows for dropouts and covariates

Failure to account for confounding variables represents another significant challenge. When investigating SEPW1 expression in relation to phenotypes such as disease risk, numerous factors influence outcomes, including age, sex, smoking habits, BMI, and even unrelated factors such as sample collection and processing conditions . Researchers should systematically identify potential confounders, measure them when possible, and account for them in study design (through stratification or randomization) and analysis (through statistical adjustment).

Inadequate characterization of selenium status undermines interpretation of SEPW1 findings. Baseline selenium levels vary widely between individuals and populations, affecting SEPW1 expression and response to interventions. Researchers should measure baseline selenium status using established biomarkers and consider this in analysis and interpretation .

What evidence links SEPW1 to neurodegenerative diseases, and how should researchers investigate these connections?

While the search results don't provide direct evidence linking SEPW1 to neurodegenerative diseases, its biological properties strongly suggest potential involvement. SEPW1's high expression in brain tissues, particularly in neuronal populations affected by neurodegenerative conditions (cortical and hippocampal neurons), combined with its antioxidant functions, positions it as a candidate molecule in neuroprotective pathways . Oxidative stress is a well-established component of Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis (ALS) pathogenesis, suggesting SEPW1 may play protective roles.

Investigating SEPW1's role in neurodegeneration requires specialized methodological approaches. Case-control studies comparing SEPW1 expression in affected versus unaffected brain regions from post-mortem samples can provide initial evidence of dysregulation. These studies should control for selenium status, as differences in selenium availability could confound interpretation of SEPW1 changes. Laser capture microdissection can enable cell-type specific analysis, which is particularly important given the cellular heterogeneity of brain tissue.

Animal models of neurodegeneration offer controlled systems to study SEPW1 function. Conditional SEPW1 knockout or knockdown models can assess whether SEPW1 deficiency exacerbates disease phenotypes. Conversely, SEPW1 overexpression models can test potential protective effects. Time-course studies are essential to determine whether SEPW1 changes precede or follow neurodegeneration.

Human genetic association studies represent another approach, examining whether SEPW1 polymorphisms associate with disease risk or progression. These studies should include adequate sample sizes to detect modest effect sizes typical of complex disorders. Functional characterization of identified variants is essential to establish biological plausibility of associations.

How does SEPW1 function relate to cellular redox regulation, and what methodologies best capture this relationship?

SEPW1's function in cellular redox regulation stems from its selenocysteine-containing active site, which participates in thiol-disulfide exchange reactions similar to thioredoxin . This activity positions SEPW1 as a component of cellular antioxidant defense systems, potentially protecting against oxidative damage to proteins, lipids, and nucleic acids.

A computational study exploring the putative reaction mechanism suggests that SEPW1 regulates the oxidation state of a conserved and solvent-exposed cysteine residue . This mechanism would allow SEPW1 to participate in redox signaling pathways, potentially influencing cellular processes through reversible oxidation/reduction of target proteins.

Investigating SEPW1's redox functions requires specialized methodological approaches. Redox proteomics techniques can identify proteins that undergo oxidation/reduction in response to SEPW1 manipulation. These approaches typically use differential labeling of reduced and oxidized thiols followed by mass spectrometry to identify specific targets and sites of modification. For example, biotin-switch techniques or iodoTMT labeling can detect reversible cysteine modifications on target proteins.

Cellular redox state can be monitored using genetically encoded redox sensors. These fluorescent protein-based sensors (such as roGFP or HyPer) can measure redox changes in specific cellular compartments in real-time. By combining these sensors with SEPW1 manipulation (overexpression or knockdown), researchers can directly assess SEPW1's impact on compartment-specific redox states.

Functional consequences of SEPW1-mediated redox regulation can be assessed through measures of cellular antioxidant capacity, lipid peroxidation, protein carbonylation, and DNA oxidation. These endpoints should be examined under both basal conditions and following oxidative challenge to determine how SEPW1 affects stress resistance.

What role does SEPW1 play in cell cycle regulation, and how can this be effectively studied?

Research indicates that SEPW1 plays a significant role in cell cycle regulation, with depletion studies showing it induces p53- and p21-dependent cell cycle arrest . This suggests SEPW1 may influence cell proliferation pathways relevant to both normal development and disease states such as cancer.

The mechanisms linking SEPW1 to cell cycle control appear to involve the mitogen-activated protein kinase (MAPK) pathway, as SEPW1 depletion affects MAPK signaling . This connection provides a plausible molecular mechanism for how a redox-active selenoprotein could influence cell division processes.

Investigating SEPW1's cell cycle functions requires multiple complementary approaches. Cell synchronization techniques allow examination of SEPW1 expression and localization throughout different cell cycle phases. This can determine whether SEPW1 levels fluctuate during the cell cycle and identify phases where its function may be most critical.

Genetic manipulation approaches (CRISPR/Cas9-mediated knockout, RNAi knockdown, or overexpression) followed by cell cycle analysis provide direct evidence of SEPW1's functional impact. Flow cytometry with propidium iodide staining or BrdU incorporation assays can quantify cell cycle distribution changes. Time-lapse microscopy with cell cycle phase markers enables real-time tracking of individual cells through division cycles.

Molecular pathway analysis should focus on known SEPW1-associated signaling components. Western blotting for phosphorylated and total MAPK pathway proteins can determine how SEPW1 manipulation affects this signaling cascade. Immunoprecipitation followed by mass spectrometry can identify novel SEPW1 interaction partners that might mediate its cell cycle effects.

How do SEPW1 expression patterns differ between healthy and disease states in human tissues?

Case-control studies comparing matched healthy and diseased tissue samples represent a fundamental approach. For neurological conditions, given SEPW1's high expression in brain tissues , comparison of affected brain regions from patients with neurodegenerative diseases versus age-matched controls could reveal disease-associated changes. These studies should control for factors known to affect selenoprotein expression, including selenium status, age, sex, and potentially relevant genetic polymorphisms.

Single-cell RNA sequencing offers particular advantages for detecting cell type-specific changes that might be masked in bulk tissue analysis. This approach is especially valuable for heterogeneous tissues where disease may affect specific cell populations differently. For example, in brain tissue, SEPW1 changes might occur specifically in neurons but not in glial cells, or even in specific neuronal subtypes.

Longitudinal studies tracking SEPW1 expression over disease progression provide valuable insights into whether expression changes are early events that might contribute to pathogenesis or later responses to disease processes. Such studies are more feasible in animal models but may be possible in humans through repeated sampling of accessible tissues (blood, skin biopsies) in progressive conditions.

Meta-analysis of existing transcriptomic datasets represents a cost-effective approach to generate hypotheses about SEPW1 expression in disease. Public repositories contain thousands of datasets comparing healthy and diseased tissues across numerous conditions. Mining these resources can identify diseases with consistent SEPW1 dysregulation worthy of focused investigation.

What novel methodologies are emerging for studying SEPW1 function at the synaptic level?

SEPW1's presence at synapses and association with RNA transport proteins suggests important roles in synaptic function . Investigating these roles requires specialized techniques that can capture the complexity of synaptic biology. Several emerging methodologies offer new opportunities for studying SEPW1 at synapses.

Spatially resolved transcriptomics allows visualization of mRNA distribution with subcellular resolution. Techniques such as MERFISH (Multiplexed Error-Robust Fluorescence In Situ Hybridization) or seqFISH (sequential FISH) can map SEPW1 mRNA localization relative to synaptic markers. These approaches can determine whether SEPW1 transcripts are locally enriched near synapses, supporting the hypothesis of local translation.

Proximity labeling methods provide powerful tools for identifying protein interaction networks in specific subcellular compartments. Techniques like BioID or APEX2 fused to SEPW1 can identify nearby proteins in living cells, potentially revealing synaptic-specific interaction partners. When combined with synaptosome preparation , these approaches can generate synaptic-specific interactomes.

MethodologyApplication to SEPW1 ResearchAdvantagesLimitations
Spatially resolved transcriptomicsMap SEPW1 mRNA at synapsesSubcellular resolution, multiple targetsTechnically challenging, fixed samples
Proximity labeling (BioID/APEX2)Identify synaptic SEPW1 interactorsIn vivo labeling, compartment-specificPotential artifacts from fusion proteins
Super-resolution microscopyVisualize SEPW1 relative to synaptic structuresNanoscale resolutionLimited to fixed samples for some methods
Local translation assaysTest if SEPW1 is locally synthesizedFunctional readoutIndirect measures of translation
Optogenetic approachesManipulate SEPW1 function with spatiotemporal precisionCell-type and subcellular specificityComplex setup, potential artifacts

Super-resolution microscopy techniques (STED, STORM, PALM) enable visualization of protein localization at nanoscale resolution. These approaches can precisely map SEPW1 distribution relative to synaptic structures, determining whether it localizes to specific synaptic compartments (pre-synaptic terminals, post-synaptic densities, or perisynaptic regions).

Local translation assays can test whether SEPW1 is locally synthesized at synapses. Techniques like puromycin proximity ligation assay (Puro-PLA) can visualize newly synthesized SEPW1 at synapses. Alternatively, photoconvertible fluorescent protein tags can track protein mobility and local synthesis through optical pulse-labeling.

How might SEPW1 research contribute to precision medicine approaches for neurological disorders?

SEPW1 research has significant potential to contribute to precision medicine approaches for neurological disorders, though this connection is not directly addressed in the search results. Several pathways link SEPW1 biology to personalized treatment strategies for brain disorders.

Genetic variation in selenoprotein pathways could influence individual responses to selenium supplementation. Polymorphisms affecting SEPW1 expression or function might predict whether patients with certain neurological conditions would benefit from selenium supplementation. Pharmacogenomic studies could identify genetic markers predicting treatment response, enabling targeted supplementation strategies.

SEPW1's high expression in brain regions affected by neurodegenerative diseases suggests potential roles in neuroprotection. If SEPW1 indeed has protective functions, therapies aimed at increasing its expression or activity could represent novel treatment approaches. Different neurological conditions might benefit from different levels of SEPW1 modulation, depending on disease mechanisms and affected brain regions.

Biomarker development represents another promising application. If peripheral SEPW1 expression (in blood cells) correlates with brain expression or neurological status, it could serve as an accessible biomarker for disease risk, progression, or treatment response. Longitudinal studies correlating SEPW1 levels with disease trajectories could establish its utility as a monitoring tool.

Drug development targeting SEPW1 pathways might yield new therapeutic options. High-throughput screening could identify compounds that modulate SEPW1 expression or enhance its functional activity. Given SEPW1's involvement in redox regulation , such compounds might have neuroprotective effects in conditions characterized by oxidative stress.

Selenium nutritional interventions might be personalized based on individual SEPW1 status. Rather than general recommendations, selenium supplementation could be tailored to achieve optimal SEPW1 expression or activity on an individual basis. This approach would require development of reliable methods to assess SEPW1 functional status in accessible tissues.

What interdisciplinary approaches could accelerate understanding of SEPW1's role in human biology?

Advancing our understanding of SEPW1's role in human biology requires interdisciplinary approaches that integrate diverse expertise and methodologies. Several collaborative frameworks offer particular promise for accelerating SEPW1 research.

Integration of genomics with nutritional science represents a powerful approach. Nutritional genomics studies examining how genetic variations influence responses to selenium intake could reveal individual differences in SEPW1 regulation. Such studies require expertise in both nutrition and genomics, including advanced statistical methods for gene-nutrient interaction analysis. Large-scale population studies with detailed dietary assessments and genomic data would be particularly valuable.

Combining neuroscience with selenium biology offers another productive intersection. SEPW1's high expression in brain tissues suggests neurological functions that remain largely unexplored. Collaborative research between neuroscientists and selenium experts could investigate how SEPW1 influences neuronal development, synaptic function, and brain aging. Such studies might employ neuroimaging techniques to correlate brain structure or function with selenium status and SEPW1 genetic variants.

Systems biology approaches can integrate multi-omics data to place SEPW1 within broader cellular networks. Combining transcriptomics, proteomics, metabolomics, and functional assays can reveal how SEPW1 interacts with other cellular components under various conditions. This requires collaboration between wet-lab scientists generating biological data and computational biologists developing integration methods and network models.

Translational research connecting basic SEPW1 findings to clinical applications needs partnerships between laboratory scientists and clinicians. Patient samples can be analyzed for SEPW1 expression patterns and correlated with clinical features, while findings from cellular and animal models can inform human studies. Biorepositories linked to clinical data represent valuable resources for such translational approaches.

Methodological innovation through engineering collaborations could develop new tools for SEPW1 research. Partnerships with bioengineers could create improved sensors for measuring SEPW1 activity in living cells or tissues. Collaborations with synthetic biologists might produce engineered cellular systems with controllable SEPW1 expression for mechanistic studies.

What are the most significant knowledge gaps in SEPW1 research, and how might they be addressed?

Despite advances in understanding SEPW1 biology, significant knowledge gaps remain that require targeted research efforts. The search results highlight several areas where current understanding is limited and where methodological innovations could yield important insights.

The precise molecular mechanisms of SEPW1's antioxidant function remain incompletely characterized. While its role in redox regulation is established , the specific substrates and reaction mechanisms in vivo are not fully elucidated. Advanced redox proteomics approaches could identify physiological targets of SEPW1's thiol-disulfide exchange activity. Structural biology studies, including cryo-electron microscopy or X-ray crystallography of SEPW1 with potential substrates, would provide mechanistic insights into its catalytic functions.

SEPW1's role in human neurological function represents a major knowledge gap. Despite high expression in brain tissues , the functional consequences of SEPW1 deficiency or excess in human neurons remain largely unknown. Human iPSC-derived neuronal models with SEPW1 manipulation could address this gap, particularly when combined with functional readouts such as electrophysiology, calcium imaging, or measures of synaptic protein synthesis.

The relationship between SEPW1 and immune function requires further investigation. While selenium supplementation affects immune responses to vaccination , SEPW1's specific contribution to these effects is unclear. Targeted knockout or knockdown studies in immune cell populations, followed by functional immune assays, could clarify SEPW1's immunomodulatory roles. Single-cell approaches would be particularly valuable for understanding cell type-specific functions.

Clinical relevance of SEPW1 genetic variants represents another significant gap. While selenoprotein polymorphisms affect protein function and disease risk, SEPW1-specific associations are less well characterized. Large-scale genetic association studies with deep phenotyping could identify links between SEPW1 variants and disease susceptibility or treatment response, particularly for neurological or immune-related conditions.

Addressing these knowledge gaps requires interdisciplinary collaboration, methodological innovation, and sustained research funding focused on selenoprotein biology. The complex intersection of nutrition, genetics, and cellular function in SEPW1 biology necessitates integrative approaches that can capture this multifaceted reality.

How can SEPW1 research findings be effectively translated to improve human health outcomes?

Translating SEPW1 research findings into improvements in human health outcomes requires strategic approaches that bridge basic science discoveries with clinical applications. Several pathways offer particular promise for this translation process.

Biomarker development represents a direct application of SEPW1 research. If SEPW1 expression or activity consistently correlates with health outcomes, it could serve as a biomarker for selenium status, disease risk, or treatment response. This requires validation studies in diverse populations to establish reference ranges and clinically meaningful thresholds . Point-of-care testing for SEPW1 status could eventually guide personalized nutrition recommendations or medical interventions.

Nutritional guidelines informed by SEPW1 research could improve population health. Current selenium intake recommendations are based primarily on optimization of glutathione peroxidase activity. If SEPW1 has distinct requirements or more sensitive health impacts, nutrition guidelines might need reconsideration. Different life stages or health conditions might benefit from specific selenium intakes optimized for SEPW1 function rather than general recommendations.

Therapeutic development targeting SEPW1 pathways could yield new treatment options. SEPW1's roles in redox regulation and cell cycle control suggest potential applications in conditions characterized by oxidative stress or dysregulated cell proliferation. Drug discovery efforts could focus on compounds that modulate SEPW1 expression or enhance its functional activity, potentially with tissue-specific delivery to minimize off-target effects.

Clinical trial design for selenium interventions should incorporate SEPW1 measurements. Rather than assuming uniform effects of supplementation, stratification based on baseline SEPW1 status could identify subgroups most likely to benefit . This personalized approach would improve the signal-to-noise ratio in trials and could explain inconsistent results from previous studies that didn't account for individual variation.

Implementation science approaches are needed to translate knowledge into practice. Even well-established relationships between SEPW1, selenium, and health outcomes require effective implementation strategies to impact clinical care or public health. This includes education of healthcare providers, development of clinical decision support tools, and creation of accessible testing methods for use in diverse healthcare settings.

Product Science Overview

Introduction

Selenoprotein W 1 (SEPW1) is a small selenoprotein that plays a crucial role in various biological processes. It is a member of the selenoprotein family, which contains the amino acid selenocysteine. Selenocysteine is known as the 21st amino acid and is unique due to its incorporation of selenium, an essential trace element. SEPW1 is widely distributed in various tissues and has been the subject of extensive research due to its potential implications in health and disease.

Structure and Function

SEPW1 is characterized by its small size, approximately 9.5 kDa, and contains a Cys-X-X-Sec motif, where “Sec” stands for selenocysteine . This motif is critical for the protein’s function. The presence of selenocysteine allows SEPW1 to participate in redox reactions, which are essential for maintaining cellular homeostasis and protecting cells from oxidative damage.

Biological Role

The primary function of SEPW1 is related to its antioxidant properties. It helps in the detoxification of reactive oxygen species (ROS) and maintains redox balance within cells . This function is vital for preventing cellular damage and ensuring proper cellular function. SEPW1 is also involved in various other biological processes, including signal transduction, cell proliferation, and immune response .

Expression and Regulation

SEPW1 expression is regulated at both the transcriptional and post-transcriptional levels. The expression of SEPW1 mRNA and protein is correlated, and it is widely distributed across different tissues . The regulation of SEPW1 is influenced by selenium availability, as selenium is a critical component of selenocysteine. Selenium deficiency can lead to reduced expression of SEPW1 and other selenoproteins, which can have significant health implications .

Health Implications

Research has shown that SEPW1 and other selenoproteins play a crucial role in human health. Selenium deficiency has been linked to various health issues, including immune dysfunction, cardiovascular diseases, and certain types of cancer . SEPW1, due to its antioxidant properties, is believed to protect against oxidative stress-related diseases. Additionally, SEPW1 has been implicated in muscle function and development, making it an essential protein for overall health .

Recombinant SEPW1

Recombinant SEPW1 is produced using biotechnological methods to study its structure, function, and potential therapeutic applications. By expressing SEPW1 in a controlled environment, researchers can investigate its properties in detail and explore its potential use in treating diseases related to oxidative stress and selenium deficiency .

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.