PSORS1C1 Antibody

Shipped with Ice Packs
In Stock

Description

Gene and Protein Overview

AttributeDetails
Gene NamePSORS1C1 (C6orf16, SEEK1)
Chromosomal Location6p21.1
Protein NamePSORS1C1 (Protein SEEK1)
Calculated MW~17 kDa
Observed MW~16 kDa (varies due to post-translational modifications or splicing)
ImmunogenSynthetic peptide spanning amino acids 40–100 (human PSORS1C1)

The antibody binds to a conserved region within the PSORS1C1 protein, enabling detection in human samples. Notably, the observed molecular weight discrepancy (16 kDa vs. calculated 17 kDa) may reflect alternative splicing or phosphorylation .

Primary Techniques

ApplicationDetails
Western BlotDetection of PSORS1C1 expression in cell lines (e.g., A-431, LO2)
ELISALimited reports; primarily validated for Western blot

Key Features:

  • Host: Rabbit-derived IgG

  • Reactivity: Human samples

  • Dilution: 1:500–1:2000 (WB)

Disease Associations

DiseaseFindings
Systemic SclerosisPSORS1C1 variants (e.g., rs3130573) linked to SSc susceptibility (OR = 1.25)
PsoriasisGenetic susceptibility locus near MHC class I region
Rheumatoid ArthritisPSORS1C1 SNPs associated with reduced IL-17 production and bone erosion risk

Mechanistic Insights:

  • Autoimmune Regulation: PSORS1C1 modulates inflammatory cytokines (e.g., IL-17, IL-1β) in synovial fibroblasts .

  • Fibrosis Pathway: Reduced TNIP1 expression in SSc patients correlates with enhanced collagen synthesis, suggesting PSORS1C1’s role in inflammation-fibrosis crosstalk .

Product Comparison

VendorProduct CodeHostReactivityObserved MWVerified Samples
ElabscienceE-AB-91932RabbitHuman16 kDaVarious cell lines
Assay GenieCAB17841RabbitHuman16 kDaA-431, LO2, BxPC-3
ThermofisherPA5-69835RabbitHumanN/AN/A
Antibodies-onlineABIN1623683N/AChimpanzeeN/AELISA

Notes:

  • Purification: Affinity-purified via peptide immunogen .

  • Storage: -20°C; avoid freeze-thaw cycles .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributor for specific delivery timelines.
Synonyms
PSORS1C1 antibody; C6orf16 antibody; SEEK1 antibody; Psoriasis susceptibility 1 candidate gene 1 protein antibody; Protein SEEK1 antibody
Target Names
PSORS1C1
Uniprot No.

Q&A

What is PSORS1C1 and why is it significant in research?

PSORS1C1 (also known as SEEK1) is a gene located on chromosome 6 near the major histocompatibility complex (MHC) class I region that confers susceptibility to autoimmune conditions, particularly psoriasis and systemic sclerosis . Genome-wide association studies have identified PSORS1C1 variants (such as rs3130573) as significant risk factors for these disorders, with strong genetic associations demonstrated in multiple populations . The gene's proximity to the HLA complex and associations with HLA-DQB1 make it particularly interesting for immunological research . Understanding PSORS1C1 expression and function provides insights into autoimmune pathogenesis, potentially identifying new therapeutic targets for difficult-to-treat skin disorders. Its involvement in allopurinol-induced severe cutaneous adverse reactions further highlights its significance in pharmacogenomics and personalized medicine approaches .

What types of PSORS1C1 antibodies are available for research?

The primary type of PSORS1C1 antibody available for research is polyclonal antibody derived from rabbit hosts that recognize specific peptide sequences within the human PSORS1C1 protein . Commercial antibodies typically target synthetic peptides corresponding to sequences within amino acids 40-100 of human PSORS1C1 (NP_054787.2) . These antibodies are suitable for Western blotting and ELISA applications, with recommended dilutions ranging from 1:500 to 1:2000 for Western blot procedures . When selecting an antibody, researchers should verify the specific epitope recognized, host species, and validated applications to ensure compatibility with their experimental design. For PSORS1C1, rabbit-derived polyclonal antibodies offer good specificity and sensitivity for detecting the approximately 16-17kDa protein in various human cell and tissue samples .

What cell lines or tissues serve as positive controls for PSORS1C1 antibody validation?

Several cell lines have been identified as reliable positive controls for PSORS1C1 antibody validation, including A-431 (epidermoid carcinoma), LO2 (hepatocyte), BxPC-3 (pancreatic adenocarcinoma), and K-562 (chronic myelogenous leukemia) cells . These cell lines consistently express detectable levels of PSORS1C1 and can be used to confirm antibody specificity in Western blotting experiments. For tissue-based applications, skin biopsies, particularly from patients with psoriasis or systemic sclerosis, often show appreciable levels of PSORS1C1 expression . Fibroblast cultures prepared from lesional skin biopsy specimens also serve as excellent positive controls, as demonstrated in studies of systemic sclerosis patients compared to healthy controls . When validating a new PSORS1C1 antibody or establishing protocols, these positive controls should be included alongside experimental samples to ensure the antibody is functioning as expected.

What is the expected molecular weight of PSORS1C1 protein in Western blot applications?

The calculated molecular weight of PSORS1C1 protein is approximately 17kDa, but the observed molecular weight in laboratory applications such as Western blotting is typically around 16kDa . This slight difference between calculated and observed weights is not uncommon in protein detection and may result from post-translational modifications or protein folding characteristics. When performing Western blots with PSORS1C1 antibodies, researchers should look for protein bands near the 16kDa mark, using appropriate percentage gels (12-15% SDS-PAGE) to achieve optimal separation of this relatively small protein . Including molecular weight markers and positive control lysates from validated cell lines helps confirm the proper identification of PSORS1C1 bands and distinguish them from non-specific binding or cross-reactivity with other proteins.

What are the primary applications for PSORS1C1 antibodies in research?

PSORS1C1 antibodies are primarily used for Western blotting and ELISA applications in research settings . In Western blotting, these antibodies help identify and quantify PSORS1C1 protein expression in various cell types and tissues, with recommended dilutions typically ranging from 1:500 to 1:2000 . They can be used to study differential expression in disease states compared to controls, as well as to monitor changes following experimental treatments. In immunohistochemistry applications, PSORS1C1 antibodies have been used on paraffin-embedded skin sections to visualize protein localization, as demonstrated in studies comparing systemic sclerosis patients with healthy controls . For RNA expression studies, PSORS1C1 antibodies complement RT-qPCR techniques, allowing researchers to correlate mRNA levels with protein expression . This multi-methodological approach provides comprehensive insights into PSORS1C1's role in disease pathogenesis and potential therapeutic responses.

How can PSORS1C1 antibodies be optimized for immunohistochemical studies of skin biopsies?

Optimizing PSORS1C1 antibodies for immunohistochemistry on skin biopsies requires careful protocol development. Begin with antigen retrieval optimization, testing both heat-induced epitope retrieval methods with citrate buffer (pH 6.0) and Tris-EDTA buffer (pH 9.0) to determine which provides optimal staining with minimal background. For PSORS1C1 detection in paraffin-embedded skin sections, previous studies have successfully employed mouse anti-human PSORS1C1 antibodies on specimens from both systemic sclerosis patients and controls . Tissue fixation parameters are critical—standardized fixation in 10% neutral buffered formalin helps preserve antigen integrity while maintaining tissue morphology. Blocking with 5-10% normal serum from the same species as the secondary antibody reduces non-specific binding. When developing the protocol, include both positive controls (known PSORS1C1-expressing tissues) and negative controls (primary antibody omission and isotype controls). For visualization, both chromogenic (DAB) and fluorescent detection methods can be employed, with the latter allowing for co-localization studies with other markers of interest such as immune cell markers or other autoimmune-associated proteins.

What are the critical considerations when performing Western blots with PSORS1C1 antibodies?

When performing Western blots with PSORS1C1 antibodies, several critical factors must be considered for optimal results. Due to PSORS1C1's relatively low molecular weight (~16kDa), researchers should use appropriate percentage gels (12-15% SDS-PAGE) to achieve optimal separation . Protein extraction methods significantly impact results—for skin tissues or fibroblasts, extraction buffers supplemented with protease inhibitors are effective at isolating PSORS1C1 while preserving protein integrity . Efficient protein transfer is crucial—semi-dry transfer systems with PVDF membranes often work well for PSORS1C1, but transfer time and voltage should be optimized for this small protein. Blocking with 5% non-fat milk or BSA in TBST for 1-2 hours at room temperature is recommended before antibody incubation. For primary antibody incubation, dilutions typically range from 1:500 to 1:2000 in blocking buffer . Including positive control cell lysates (such as A-431 or K-562) alongside experimental samples helps confirm antibody specificity and proper technique . Multiple washing steps with TBST are essential to reduce background and improve signal-to-noise ratio in the final results.

How can PSORS1C1 gene expression and protein levels be correlated in experimental studies?

Correlating PSORS1C1 gene expression with protein levels requires a multi-methodological approach. For RNA extraction, specialized kits effectively isolate RNA from samples such as plasma or skin biopsies . RT-qPCR can be performed using PSORS1C1-specific primers (e.g., forward: 5′-CTGACCGACTTTGCCACATGGA-3′, reverse: 5′-GTGGGAAGAGGGAACCAGGATA-3′) with appropriate reference genes like GAPDH . Relative expression should be calculated using the 2^(-ΔΔCq) method following MIQE guidelines for quantitative PCR experiments . In parallel, proteins extracted from the same samples can be analyzed by Western blotting using anti-PSORS1C1 antibodies. For accurate quantification, researchers should use digital imaging systems and normalize PSORS1C1 band intensity to loading controls such as β-actin or GAPDH. This approach has been successfully employed in studies of psoriasis, where PSORS1C1 expression was measured at both RNA and protein levels to establish disease-associated patterns . The correlation analysis between mRNA and protein data should account for potential time lags between transcription and translation as well as differences in the stability of mRNA versus protein in experimental systems.

What experimental approaches can assess PSORS1C1 methylation status in relation to protein expression?

Investigating the relationship between PSORS1C1 methylation and protein expression requires integrating epigenetic and proteomic techniques. Bisulfite conversion of DNA followed by pyrosequencing of specific CpG sites in the PSORS1C1 gene, particularly focusing on sites like cg24926791, provides methylation profiles associated with disease conditions . Studies of allopurinol-induced severe cutaneous adverse reactions have successfully designed primers to capture multiple CpG sites (array site, CpG1, and CpG2 regions) in the PSORS1C1 gene . In parallel, PSORS1C1 protein expression can be assessed using Western blotting or ELISA with specific antibodies to determine how methylation patterns correlate with protein levels. Research has demonstrated that PSORS1C1 hypomethylation (methylation levels <90%) is associated with a 30.22-fold increased risk (95% CI = 4.73–192.96) of developing allopurinol-induced cutaneous reactions . This approach reveals important epigenetic mechanisms potentially underlying PSORS1C1's role in autoimmune and drug hypersensitivity conditions, offering insights beyond genetic association studies.

How do PSORS1C1 antibodies contribute to understanding the genetic basis of psoriasis and systemic sclerosis?

PSORS1C1 antibodies provide critical tools for translating genetic associations into functional understanding of disease mechanisms. While genome-wide association studies have identified PSORS1C1 variants as risk factors for both psoriasis and systemic sclerosis (with odds ratios showing significant association), antibody-based techniques help elucidate how these genetic associations manifest at the protein level . Immunohistochemistry using PSORS1C1 antibodies enables researchers to compare protein expression patterns in lesional versus non-lesional skin from patients with these conditions, revealing tissue-specific alterations associated with disease states . Western blotting can quantitatively assess whether specific PSORS1C1 genetic variants correlate with altered protein expression levels. By combining genotyping data for single nucleotide polymorphisms (SNPs) in the PSORS1C1 locus (such as rs3130573 and rs1062470) with protein quantification in the same patient cohorts, researchers can establish genotype-phenotype correlations . This multi-level approach connecting genetic variation to protein expression and downstream effects provides mechanistic insights into how PSORS1C1 contributes to autoimmune skin disease pathogenesis.

What are common issues with PSORS1C1 antibody specificity and how can they be resolved?

Specificity issues with PSORS1C1 antibodies may manifest as multiple unexpected bands on Western blots or non-specific staining in immunohistochemistry. To resolve these issues, first verify antibody quality through proper controls: positive control samples (A-431, LO2, BxPC-3, or K-562 cell lysates) should show clear bands at the expected molecular weight (~16kDa) . For Western blotting, optimize blocking conditions by testing different blocking agents (5% non-fat milk, 5% BSA, or commercial blocking buffers) and extending blocking time to reduce non-specific binding. Increase washing duration and frequency between antibody incubations (at least 3-5 washes for 5-10 minutes each with TBST). For persistent non-specific bands, titrate the antibody concentration further, using more stringent washing buffers, or pre-absorb the antibody with non-relevant proteins. For immunohistochemistry applications, additional controls should include tissue known to be negative for PSORS1C1, isotype control antibodies, and peptide competition assays where the antibody is pre-incubated with excess target peptide. If problems persist, consider validating results with a different PSORS1C1 antibody that recognizes a distinct epitope to confirm specific binding patterns.

How can researchers address challenges in detecting low levels of PSORS1C1 expression?

Detecting low levels of PSORS1C1 expression requires enhanced sensitivity techniques and careful optimization. For Western blotting, increase protein loading (50-100 μg total protein per lane) and use high-sensitivity chemiluminescent substrates or fluorescent detection systems. When performing RT-qPCR for gene expression analysis, optimize RNA extraction protocols to maximize yield and purity, as demonstrated in studies using the Qiagen miRNeasy mini kit for plasma samples from psoriatic patients . Digital droplet PCR (ddPCR) offers superior sensitivity over conventional qPCR for detecting minimal PSORS1C1 expression. For protein detection in complex samples like plasma, consider employing immunoprecipitation before Western blotting to concentrate PSORS1C1 protein. In cell-based experiments, treating with relevant cytokines (TNFα, IL-1β, IL-6) may upregulate PSORS1C1 expression to detectable levels, as these inflammatory mediators often impact gene expression in autoimmune conditions . This approach has been successfully used in fibroblast cultures from systemic sclerosis patients, where cytokine treatment altered expression patterns of target proteins . Finally, ensure all reagents are fresh and properly stored to maintain optimal activity throughout the detection protocol.

How should researchers approach contradictory data between PSORS1C1 gene expression and protein detection?

Contradictory results between PSORS1C1 gene expression and protein detection require systematic investigation of potential biological and technical explanations. First, verify all technical aspects of both assays: for RT-qPCR, confirm primer specificity through sequencing of amplicons and melt curve analysis; for protein detection, validate antibody specificity with positive and negative controls . Consider temporal dynamics—protein levels often lag behind mRNA changes, so time-course experiments may reveal delayed correlation. Post-transcriptional regulation mechanisms, including microRNA-mediated suppression, can cause discrepancies between mRNA and protein levels. Epigenetic regulation should also be considered—the methylation status of PSORS1C1 significantly impacts gene expression and can be assessed through bisulfite sequencing or methylation-specific PCR . Studies have demonstrated that PSORS1C1 hypomethylation is associated with altered expression patterns in disease states, which may explain apparent contradictions between genetic, transcript, and protein data . Analyze potential splice variants of PSORS1C1 using isoform-specific primers and validate at the protein level with antibodies recognizing different epitopes. By systematically addressing these factors, researchers can resolve apparent contradictions and potentially uncover novel regulatory mechanisms affecting PSORS1C1 expression.

What experimental controls are essential when using PSORS1C1 antibodies in various applications?

When using PSORS1C1 antibodies, several essential controls ensure reliable and interpretable results. For Western blotting, positive control lysates from validated cell lines (A-431, LO2, BxPC-3, or K-562) should be included alongside experimental samples . Loading controls (β-actin, GAPDH, or total protein stains) are crucial for normalization and quantitative comparisons. For immunohistochemistry, positive tissue controls (confirmed PSORS1C1-expressing samples) verify staining protocols, while negative controls including primary antibody omission, isotype controls, and non-expressing tissues detect non-specific binding. When studying PSORS1C1 in disease contexts, including both affected and unaffected tissues from the same patient (such as lesional and non-lesional skin in psoriasis) provides internal controls for disease-specific changes . For gene expression studies, no-template controls and no-reverse transcriptase controls are essential to detect contamination, while reference genes must be validated for stability across experimental conditions . When investigating PSORS1C1 methylation, unmethylated and fully methylated DNA standards should be included in bisulfite conversion and subsequent analyses . These comprehensive controls help distinguish specific PSORS1C1 signals from technical artifacts, enabling confident interpretation of experimental results.

How can PSORS1C1 expression analysis contribute to understanding autoimmune disease mechanisms?

PSORS1C1 expression analysis provides valuable insights into autoimmune disease mechanisms through several research approaches. Genome-wide association studies have established PSORS1C1 as a susceptibility gene for psoriasis and systemic sclerosis, with specific variants showing significant disease associations . Protein-level investigation using PSORS1C1 antibodies reveals how these genetic associations translate to functional effects in target tissues. Comparative expression studies between lesional and non-lesional skin samples from patients with autoimmune conditions demonstrate tissue-specific alterations associated with disease activity . Cell culture models using fibroblasts from patients with systemic sclerosis have shown differential PSORS1C1 expression compared to healthy controls, providing insights into disease mechanisms at the cellular level . Treatment of these cellular models with inflammatory cytokines (TNFα, IL1β, IL6) further elucidates how PSORS1C1 responds to inflammatory stimuli central to autoimmune pathogenesis . By correlating PSORS1C1 expression patterns with clinical parameters such as disease duration, severity, and treatment response, researchers can identify potential biomarkers and therapeutic targets for these challenging conditions.

What is the relationship between PSORS1C1 methylation status and its protein expression in disease states?

The relationship between PSORS1C1 methylation and protein expression represents a critical epigenetic mechanism in disease pathophysiology. Studies of allopurinol-induced severe cutaneous adverse reactions have demonstrated significant hypomethylation of PSORS1C1 during disease onset, with patients showing methylation levels below 90% having a 30.22-fold increased risk (95% CI = 4.73–192.96) of developing these reactions . This hypomethylation appears to affect protein expression, as confirmed by analyses with PSORS1C1 antibodies. The relationship is complex and potentially bidirectional—altered methylation may drive changes in protein expression, while inflammatory disease states may themselves induce epigenetic modifications . Methodologically, researchers can investigate this relationship by combining bisulfite pyrosequencing of specific CpG sites (including the functionally significant cg24926791 site) with protein quantification via Western blotting or ELISA using PSORS1C1 antibodies . While some limitations exist in interpreting results from heterogeneous cell populations in whole blood samples, these approaches have successfully identified disease-associated epigenetic patterns that provide insights beyond genetic association studies .

How can PSORS1C1 antibodies be used to evaluate potential therapeutic interventions for autoimmune skin disorders?

PSORS1C1 antibodies offer valuable tools for evaluating therapeutic interventions in autoimmune skin disorders through multiple experimental approaches. In preclinical studies, researchers can treat cell culture models (such as primary dermal fibroblasts from patients and controls) with potential therapeutic compounds, then use Western blotting with PSORS1C1 antibodies to assess changes in protein expression . This approach has been employed with recombinant TNIP1 treatment in the presence or absence of proinflammatory cytokines (TNFα, IL-1β, IL-6), revealing effects on collagen production and related pathways . For ex vivo studies using skin explants from patients, immunohistochemistry with PSORS1C1 antibodies before and after treatment can visualize changes in protein localization and expression. In clinical research, comparative analysis of PSORS1C1 expression in skin biopsies before and after therapeutic intervention can help identify molecular mechanisms underlying clinical improvement. By establishing PSORS1C1 as a molecular marker of disease activity or treatment response, these antibody-based approaches facilitate development and refinement of targeted therapies for autoimmune skin conditions that remain challenging to treat effectively.

What methodological approaches can best investigate PSORS1C1 interactions with other risk genes in autoimmune disorders?

Investigating PSORS1C1 interactions with other risk genes in autoimmune disorders requires integrated methodological approaches. Genetic association studies have identified several genes with significant associations to conditions where PSORS1C1 plays a role, including HLA-DQB1, TNIP1, and STAT4 in systemic sclerosis . These genes collectively contribute to a substantial proportion of disease risk, with population attributable risk (PAR) estimates of 24% for HLA-DQB1, 4% for TNIP1, and a combined PAR of 47.4% across identified risk genes . To understand functional interactions, co-immunoprecipitation experiments using PSORS1C1 antibodies can identify direct protein-protein interactions between these risk factors. Gene expression correlation analysis between PSORS1C1 and other risk genes using RT-qPCR techniques can reveal coordinated expression patterns suggestive of shared regulatory mechanisms . Chromosome conformation capture methods can detect physical interactions between PSORS1C1 and other risk gene loci within the nuclear space, particularly valuable given PSORS1C1's proximity to the MHC region. CRISPR-based approaches manipulating PSORS1C1 expression while monitoring effects on other risk genes can establish directional relationships. These multi-level approaches help elucidate the complex genetic architecture of autoimmune disorders and may identify central pathways for therapeutic targeting.

How can PSORS1C1 antibodies help discriminate between different clinical subtypes of autoimmune skin disorders?

PSORS1C1 antibodies can help discriminate between clinical subtypes of autoimmune skin disorders through comparative expression profiling across disease variants. In systemic sclerosis, studies have employed PSORS1C1 antibodies to examine protein expression differences between limited cutaneous and diffuse cutaneous disease subtypes . Immunohistochemistry on skin biopsies using these antibodies can reveal distinct tissue distribution patterns that correlate with clinical phenotypes . Quantitative analysis of PSORS1C1 protein levels via Western blotting may identify subtype-specific expression signatures that complement clinical classification. In studies of allopurinol-induced cutaneous adverse reactions, PSORS1C1 methylation and subsequent protein expression patterns showed differences between reaction subtypes including DRESS (Drug Rash with Eosinophilia and Systemic Symptoms) versus SJS/TEN (Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis) . These molecular distinctions support the concept that DRESS and SJS/TEN represent parts of the same spectrum but with different severity profiles . By correlating PSORS1C1 expression patterns with detailed clinical phenotyping, researchers can develop molecular classification systems that may predict disease course, treatment response, and prognosis more accurately than clinical assessment alone.

What emerging technologies might enhance PSORS1C1 antibody applications in autoimmune disease research?

Several emerging technologies promise to enhance PSORS1C1 antibody applications in autoimmune disease research. Single-cell proteomics techniques will allow researchers to examine PSORS1C1 expression at unprecedented resolution, revealing cell-type-specific patterns within heterogeneous skin samples. Mass cytometry (CyTOF) incorporating PSORS1C1 antibodies could simultaneously measure dozens of cellular markers alongside PSORS1C1, providing comprehensive phenotyping of immune and skin cells in disease states. Spatial transcriptomics combined with in situ protein detection using PSORS1C1 antibodies will map both gene and protein expression within tissue architecture, preserving crucial spatial information lost in homogenized samples . CRISPR-based technologies for precise genetic modification of PSORS1C1 will enable functional studies correlating genetic variants with protein expression and disease phenotypes. Advanced imaging techniques including super-resolution microscopy using fluorescently-labeled PSORS1C1 antibodies will reveal subcellular localization patterns previously undetectable with conventional microscopy. Organ-on-chip and 3D skin model technologies incorporating cells with varying PSORS1C1 genotypes will provide dynamic disease models for therapeutic testing. These technological advances will collectively deepen our understanding of PSORS1C1's role in autoimmune pathogenesis and accelerate development of targeted interventions.

How might epigenetic editing of PSORS1C1 advance understanding of its role in disease mechanisms?

Epigenetic editing of PSORS1C1 represents a promising frontier for understanding its role in disease mechanisms. Given the established significance of PSORS1C1 methylation in conditions like allopurinol-induced cutaneous adverse reactions, targeted epigenetic manipulation could provide causal insights into how methylation changes affect protein expression and cellular function . CRISPR-based epigenetic editing systems, including catalytically dead Cas9 (dCas9) fused with DNA methyltransferases (for inducing hypermethylation) or TET enzymes (for demethylation), could target specific CpG sites in the PSORS1C1 gene, particularly the functionally significant cg24926791 site . Following epigenetic manipulation, researchers could use PSORS1C1 antibodies to assess resulting protein expression changes via Western blotting or immunocytochemistry. This approach would help determine whether the hypomethylation observed in disease states is causally linked to altered PSORS1C1 expression or represents a consequence of disease processes. By creating isogenic cell lines differing only in PSORS1C1 methylation status, researchers could isolate epigenetic effects from genetic background variation. These models would be valuable for testing how environmental factors known to influence epigenetic patterns might alter PSORS1C1 expression and contribute to disease susceptibility or progression through epigenetic mechanisms.

What potential exists for developing PSORS1C1-targeted therapeutics for autoimmune skin disorders?

The potential for developing PSORS1C1-targeted therapeutics for autoimmune skin disorders rests on several promising research directions. Studies using recombinant proteins that interact with PSORS1C1 pathways have shown effects on collagen production in fibroblast cultures from systemic sclerosis patients, suggesting modulation of PSORS1C1 signaling could affect disease processes . Antisense oligonucleotides or siRNA approaches targeting PSORS1C1 could provide temporary modulation of expression to assess therapeutic potential before developing more permanent interventions. Small molecule screens using cell-based assays with PSORS1C1 antibody readouts could identify compounds that normalize aberrant expression patterns in disease states. Epigenetic drugs targeting the methylation status of PSORS1C1 might be particularly relevant given the established relationship between PSORS1C1 hypomethylation and certain cutaneous disorders . Monoclonal antibodies targeting PSORS1C1 or its interaction partners could disrupt pathological signaling cascades. For personalized approaches, patient-derived cells could be tested with candidate therapeutics and assessed for PSORS1C1 expression changes as predictive biomarkers of potential clinical response. While still in early conceptual stages, these approaches highlight how fundamental research using PSORS1C1 antibodies could translate into novel therapeutic strategies for difficult-to-treat autoimmune skin conditions.

How can multi-omics approaches incorporating PSORS1C1 antibody data advance precision medicine for autoimmune disorders?

Multi-omics approaches incorporating PSORS1C1 antibody data hold significant promise for advancing precision medicine in autoimmune disorders. By integrating genomic data (PSORS1C1 genotyping), epigenomic information (methylation profiles), transcriptomic data (RNA-seq or RT-qPCR), and proteomic measurements (antibody-based PSORS1C1 quantification), researchers can develop comprehensive molecular signatures of disease subtypes . These integrated profiles enable patient stratification beyond clinical phenotyping alone. For example, combining PSORS1C1 genetic variants, methylation status at key CpG sites like cg24926791, and protein expression patterns measured with specific antibodies could identify distinct molecular subgroups within clinically similar presentations of psoriasis or systemic sclerosis . These molecular subgroups may respond differently to therapies, allowing for more personalized treatment selection. Longitudinal studies incorporating repeated sampling and multi-omics analysis can track molecular changes during disease progression and treatment response, potentially identifying early markers of treatment failure or disease flares. Machine learning algorithms applied to these multi-dimensional datasets can discover complex patterns beyond traditional statistical approaches, potentially revealing new disease mechanisms and therapeutic targets involving PSORS1C1 and its interaction network.

What standardization efforts are needed to improve reproducibility in PSORS1C1 antibody-based research?

Improving reproducibility in PSORS1C1 antibody-based research requires concerted standardization efforts across several domains. First, antibody validation standards should be established, including minimum requirements for demonstrating specificity (such as positive and negative control cell lines, knockout validation, and peptide competition assays) . Detailed reporting of antibody characteristics including catalog numbers, lot numbers, host species, epitope information, and validation data should become standard practice in publications. For gene expression studies, adherence to MIQE guidelines ensures proper RT-qPCR methodology and reporting, which has been successfully implemented in PSORS1C1 research . Sample preparation protocols for various applications (Western blotting, immunohistochemistry, immunoprecipitation) should be standardized and thoroughly documented, including buffer compositions, incubation times, and equipment parameters. Quantification methods for Western blots and immunohistochemistry should employ digital image analysis with defined algorithms rather than subjective assessment. For studies involving PSORS1C1 methylation analysis, standardized bisulfite conversion protocols and pyrosequencing approaches would improve cross-study comparisons . Establishment of reference materials and calibrators for absolute quantification would enable direct comparison of results across laboratories. Finally, creation of open repositories for protocols, validation data, and raw results would facilitate method sharing and collaborative improvement of techniques for PSORS1C1 detection and quantification.

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 2025 TheBiotek. All Rights Reserved.