The ORM1 Antibody Pair (Catalog: abx117318) from Abbexa Ltd consists of two monoclonal antibodies optimized for sandwich ELISA assays:
Capture Antibody: Unconjugated, 2 mg/ml concentration.
Biotin-Conjugated Detection Antibody: 0.25 mg/ml concentration .
These antibodies target distinct epitopes on the ORM1 protein, enabling precise quantification in biological samples .
Disease Biomarker Studies: ORM1 is upregulated in inflammatory conditions and cancers like hepatocellular carcinoma (HCC). Its expression correlates with tumor stage, vascular invasion, and sorafenib resistance .
Drug Development: ORM1 influences drug pharmacokinetics by binding synthetic compounds, making this antibody pair critical for pharmacodynamic studies .
Immunoassays: Validated for ELISA, it enables high-throughput screening of ORM1 levels in clinical samples .
Sensitivity: Detects ORM1 at low concentrations due to high-affinity binding.
Cross-Reactivity: Broad species reactivity ensures utility in preclinical models (e.g., mouse, rat) .
Validation: Includes data from GEO and TCGA datasets confirming ORM1’s role in HCC progression .
Cancer Research: ORM1 knockdown suppresses HCC cell growth and enhances sorafenib sensitivity in vitro .
Acute-Phase Response Monitoring: ORM1 levels increase up to 50-fold during inflammation, aiding in disease severity assessment .
While Proteintech’s ORM1/2 antibody (66097-1-Ig) is used for Western blotting and immunohistochemistry , the Abbexa pair is uniquely optimized for ELISA, offering higher specificity for quantitative assays .
ORM1 (Orosomucoid 1) is an acute-phase protein primarily synthesized in the liver and secreted into plasma . It functions as a transport protein in the bloodstream, binding various ligands and synthetic drugs, thereby influencing their distribution and availability in the body . ORM1 has gained significant research attention due to its multiple roles in pathological conditions:
Upregulated in the serum of cancer patients, particularly in breast cancer
Promotes chemotherapy resistance (epirubicin resistance in breast cancer, sorafenib resistance in kidney cancer)
Serves as a biomarker in inflammatory conditions such as rheumatoid arthritis
Modulates immune system activity during acute-phase reactions
These diverse functions make ORM1 an important target for research in oncology, immunology, and drug development. Its expression levels correlate with disease progression and survival outcomes in multiple cancer types, positioning it as both a biomarker and potential therapeutic target .
When investigating ORM1 in cancer contexts, several methodological approaches have proven particularly effective:
qRT-PCR for quantifying ORM1 mRNA expression in tissues and cell lines
Western blotting to assess protein expression levels (recommended dilution range: 1:500-1:2000)
Immunohistochemistry to visualize tissue distribution and cellular localization
Colony formation assays to assess effects on cell proliferation
Transwell and wound healing assays to determine migration and invasion capacity
Research has demonstrated that ORM1 knockdown inhibits cancer cell proliferation by approximately 50% in kidney cancer cell lines, while simultaneously reducing migration and invasion capabilities . Additionally, ORM1 manipulation directly affects chemotherapeutic drug sensitivity, with knockdown enhancing drug effectiveness and overexpression reducing it .
Sandwich ELISA using ORM1 antibody pairs offers a powerful quantitative approach for measuring ORM1 levels in biological samples. The methodology involves:
Coating microplate wells with ORM1 capture antibody in appropriate buffer
Blocking non-specific binding sites with blocking buffer
Sample addition (serum, plasma, cell culture supernatants, or tissue lysates)
Binding of target ORM1 to immobilized capture antibody
Addition of biotin-conjugated detection antibody targeting a different epitope
Introduction of streptavidin-HRP conjugate
Substrate addition and colorimetric/chemiluminescent signal development
Quantification against standard curve using recombinant ORM1
Antibody concentrations and dilution ratios
Incubation times and temperatures
Washing stringency
Sample preparation protocols
Detection system sensitivity
The sandwich ELISA format provides superior specificity compared to single-antibody approaches due to the requirement for two distinct epitope recognitions. This is particularly important for ORM1, which has multiple glycoforms and potential cross-reactivity with related proteins .
Multiple factors can significantly impact the sensitivity and specificity of ORM1 detection:
Epitope location relative to glycosylation sites
Affinity and avidity for target protein
Clone selection (monoclonal vs polyclonal)
Production and purification methods
ORM1 concentration in different biological matrices
Presence of binding proteins or drugs that interact with ORM1
Post-translational modifications, especially glycosylation pattern variations
Sample storage and handling conditions
Buffer composition (optimal: PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3)
Blocking reagent selection
Detection system (colorimetric vs fluorescent vs chemiluminescent)
Capture antibody coating density and orientation
Factor | Impact on Detection |
---|---|
ORM2 (closely related protein) | Potential cross-reactivity requiring validation |
Species differences | ORM1 antibodies may show variable cross-reactivity across species |
Glycoform heterogeneity | Different glycosylation patterns can mask epitopes |
Acute-phase status | Dramatically increased levels during inflammation may require optimization |
Researchers should systematically evaluate these factors when establishing ORM1 detection methods, particularly when working with samples from different species or disease states .
ORM1 has been implicated in drug resistance across multiple cancer types, with several mechanistic pathways identified:
Promotes epirubicin resistance through:
ORM1 expression enhances cell proliferation and migration
Down-regulation of ORM1 inhibits these resistance mechanisms
ORM1 affects sorafenib efficacy:
Inhibition rate of sorafenib is enhanced in ORM1 knockdown cells
Overexpression of ORM1 reduces sorafenib effectiveness
Mechanism involves CALR-mediated regulation of apoptotic pathways
ORM1 knockdown upregulates caspase-3 activity and apoptotic rate
ORM1 attenuates doxorubicin-induced oxidative stress through:
Understanding these mechanisms provides potential approaches for overcoming drug resistance. For example, targeting ORM1 in combination with chemotherapy may enhance treatment efficacy in resistant tumors .
Effective sample preparation is crucial for accurate ORM1 detection:
Collection: Use standardized collection tubes (serum or EDTA/heparin for plasma)
Processing: Separate within 2 hours of collection
Avoid hemolysis which can interfere with detection
Storage: Aliquot to avoid freeze-thaw cycles
Collect supernatants from cells at appropriate confluency
Centrifuge to remove cellular debris (typically 1000g for 10 minutes)
Consider concentrating samples if ORM1 levels are low
Include protease inhibitors to prevent degradation
Flash freeze immediately after collection
Homogenize in appropriate buffer with protease inhibitors
Centrifuge at high speed (>10,000g) to remove insoluble material
Determine protein concentration before analysis
Dilution optimization: Test multiple dilutions to ensure readings within the linear range
Sample matrix effects: Consider matrix-matched standards
Interfering substances: Be aware that ORM1 binds various drugs and compounds that might affect detection
Consistency: Maintain identical processing for all samples within a study
For western blot applications specifically, prepare samples in reducing or non-reducing conditions depending on the epitope recognition requirements of the antibodies. For glycoprotein analysis, consider enzymatic treatments to evaluate the impact of glycosylation on detection .
Thorough validation of antibody pair specificity is essential for reliable ORM1 research:
Recombinant protein controls: Test against purified recombinant ORM1
Epitope mapping: Ensure antibody pairs target distinct, non-overlapping regions
Sequence verification: Confirm antibodies recognize the intended sequence (e.g., amino acids 35-201 of human ORM1, NP_000598.2)
Peptide competition: Pre-incubate with specific peptides to confirm epitope specificity
Knockdown/knockout verification:
Test in ORM1 siRNA-treated samples
Compare signal reduction with knockdown efficiency
Species cross-reactivity assessment:
Systematically test samples from different species
Verify performance matches claimed reactivity profile
Western blot analysis:
Confirm single band at expected molecular weight (~23 kDa for core protein)
Higher apparent molecular weight due to glycosylation (~45 kDa)
Mass spectrometry confirmation:
Immunoprecipitate ORM1 using antibodies
Verify protein identity by peptide mass fingerprinting
Cross-reactivity testing:
Test against related proteins (particularly ORM2)
Evaluate in complex biological matrices
Linearity: Assess across relevant concentration range
Precision: Determine intra- and inter-assay variability
Accuracy: Perform spike-recovery experiments
Sensitivity: Establish lower limit of detection
Rigorous validation ensures that observed results genuinely reflect ORM1 biology rather than technical artifacts or cross-reactivity .
Post-translational modifications (PTMs) of ORM1, particularly glycosylation, have profound implications for research:
ORM1 exhibits N-glycan heterogeneity at Asn-33 with three major glycoforms:
Glycan structures can mask protein epitopes
Different glycoforms may alter antibody binding efficiency
Deglycosylation treatments might expose hidden epitopes
Changes in glycosylation patterns during disease may affect detection consistency
ORM1 is phosphorylated by protein kinase Ypk1
Key phosphorylation sites include Ser51, Ser52, and Ser53
Phosphorylation status affects ORM1 function and may influence antibody binding
Biomarker applications: Different PTM patterns may serve as disease-specific markers
Functional studies: PTMs affect ORM1's binding to drugs and other molecules
Assay development: Consider epitope accessibility relative to PTM sites
Data interpretation: Signal changes may reflect PTM alterations rather than expression changes
Enzymatic treatments to remove specific PTMs
Mass spectrometry to characterize PTM profiles
Antibody panels targeting different epitopes
Combined protein and glycan analysis techniques
Understanding and accounting for PTMs is essential for accurate ORM1 quantification and functional characterization across different experimental conditions and disease states .
ORM1 exhibits distinct expression patterns across various pathological conditions:
Breast Cancer:
Kidney Renal Clear Cell Carcinoma (KIRC):
Protein expression significantly stronger in primary tumor tissues compared to normal
Lower methylation level of ORM1 promoter in tumor tissues
Higher ORM1 expression correlates with worse survival probability
Expressed at higher levels in KIRC cell lines (786-O, A498, and Caki-2) compared to control cells
Rheumatoid Arthritis:
Doxorubicin-Induced Cardiotoxicity:
Disease Context | ORM1 Expression | Clinical Correlation | Mechanism |
---|---|---|---|
Breast Cancer | Upregulated | Poor prognosis | AKT/ERK pathway activation |
KIRC | Upregulated | Worse survival | CALR-mediated anti-apoptotic effects |
Rheumatoid Arthritis | Upregulated | Increased disease activity | Immune cell infiltration |
Doxorubicin Cardiotoxicity | Downregulated after DOX | Increased cardiotoxicity | Reduced Nrf2/HO-1 activity |
These expression patterns highlight ORM1's context-dependent roles and potential value as both a diagnostic biomarker and therapeutic target .
Recent research has revealed significant applications of ORM1 in multiple disease contexts:
In KIRC, patients with high ORM1 expression (n=133) showed significantly worse survival probability compared to those with low expression (n=398)
ORM1 expression correlates with tumor grade and vascular invasion in hepatocellular carcinoma
In breast cancer, ORM1 acts as a predictor of chemotherapy response and disease progression
ORM1 serves as a serum biomarker in rheumatoid arthritis, with levels correlating with disease activity
Integration of ORM1 in multi-biomarker panels improves diagnostic accuracy
Relationship to specific immune cell populations provides mechanistic insights into disease pathogenesis
ORM1 shows potential as a biomarker for cardiotoxicity risk assessment
Therapeutic overexpression of ORM1 demonstrates cardioprotective effects
Monitoring ORM1 levels may help predict susceptibility to chemotherapy-induced cardiac damage
Advanced proteomics approaches using mass spectrometry have identified ORM1 as a key biomarker
Machine learning models incorporating ORM1 with other proteins improve diagnostic accuracy
Protein-protein interaction (PPI) network topological analysis methods reveal ORM1's position in disease networks
Development of clinical assays measuring ORM1 glycoform variations in disease states
Integration of ORM1 assessment in therapeutic decision-making algorithms
Exploration of ORM1-targeting approaches to overcome drug resistance
These advances highlight ORM1's growing importance in precision medicine approaches across multiple disease domains .
When investigating ORM1's role in drug resistance, optimal experimental design requires careful consideration of several factors:
Cell Line Models:
Choose cell lines with varying baseline ORM1 expression
Include resistant and sensitive pairs for comparative studies
Consider patient-derived cell lines to increase clinical relevance
Animal Models:
Develop ORM1 knockout or transgenic models
Use xenograft models with manipulated ORM1 expression
Consider patient-derived xenografts for translational studies
Gene Expression Modulation:
Pharmacological Approaches:
Dose-response studies with relevant chemotherapeutic agents
Time-course experiments to track resistance development
Combination studies with ORM1-targeting compounds
Viability and Proliferation:
CCK-8 assay for cytotoxicity assessment
Colony formation for long-term survival
Real-time cell analysis for temporal dynamics
Mechanism Investigation:
Western blot for AKT/ERK pathway activation
qRT-PCR for MMP-2/9 expression
Caspase-3 activity assays for apoptosis assessment
Oxidative stress markers (4-HNE, ROS measurement)
Translational Readouts:
ELISA for secreted ORM1 quantification
Immunohistochemistry for tissue distribution
Patient sample correlation studies
Include appropriate positive and negative controls
Perform time-course studies to capture dynamics of resistance development
Use multiple drug concentrations to generate complete resistance profiles
Incorporate rescue experiments to confirm ORM1-specific effects
This comprehensive approach enables robust investigation of ORM1's mechanistic contributions to drug resistance and identification of potential intervention strategies .
Developing multiplex assays including ORM1 presents several methodological challenges that must be systematically addressed:
Potential interactions between detection antibodies
Ensuring ORM1 antibodies don't recognize related proteins (particularly ORM2)
Validation of specificity in the presence of multiple targets
Accounting for species cross-reactivity when using non-human samples
ORM1 levels can vary dramatically (acute-phase response can increase levels 2-5 fold)
Other biomarkers may have different concentration ranges
Balancing sensitivity requirements across all targets
Developing appropriate dilution strategies for all analytes
Platform | Advantages | Challenges for ORM1 Inclusion |
---|---|---|
Bead-Based | High multiplexing capacity | Potential bead aggregation with glycoproteins |
Planar Arrays | Spatial separation reduces interference | May require larger sample volumes |
Electrochemiluminescence | High sensitivity | Complex optimization with glycosylated proteins |
Mass Spectrometry | Detailed PTM analysis | Limited throughput, complex sample prep |
Selection of complementary biomarkers based on disease context:
Biological relevance of marker combinations
Statistical considerations for data interpretation
Complex biological matrices may affect antibody binding
Different preanalytical requirements for different targets
Consistency in sample preparation across studies
Validating performance across different sample types
By addressing these challenges systematically, researchers can develop robust multiplex assays that include ORM1 alongside other relevant biomarkers, enabling more comprehensive disease profiling while conserving valuable sample material .
ORM1 engages with multiple signaling pathways across different disease contexts:
AKT/ERK Pathway:
MMP Regulation:
CALR-Mediated Apoptotic Regulation:
Nrf2/HO-1 Pathway:
Rigorous controls and validation are essential for reliable ORM1 research:
Antibody Validation Controls:
Positive control: Recombinant ORM1 protein at known concentrations
Negative control: Samples from ORM1 knockout models or cells
Isotype control: Matched antibody isotype to detect non-specific binding
Peptide competition: Pre-incubation with immunizing peptide should block specific signal
Experimental Controls:
ORM1 knockdown validation: Confirm reduction at both mRNA and protein levels
Overexpression validation: Verify increased expression using multiple detection methods
Rescue experiments: Reintroduction of ORM1 should reverse knockdown effects
Dose-response relationships: Demonstrate concentration-dependent effects
Technical Validation:
Antibody specificity confirmation:
Western blot showing single band at expected molecular weight
Immunoprecipitation followed by mass spectrometry identification
Cross-reactivity testing against related proteins (especially ORM2)
Assay performance validation:
Linearity assessment across relevant concentration range
Intra- and inter-assay precision determination
Spike-recovery experiments to confirm accuracy
Limit of detection and quantification establishment
Biological Validation:
Expression pattern confirmation:
Concordance between mRNA and protein levels
Consistency with published literature
Verification in multiple model systems
Functional validation:
Phenotypic changes following ORM1 manipulation
Pathway activation/inhibition as expected
Reproducibility across different experimental conditions
Validation Parameter | Method | Acceptance Criteria |
---|---|---|
Antibody Specificity | Western blot | Single band at expected MW (~45 kDa glycosylated) |
Cross-reactivity | Testing multiple species | Results consistent with claimed reactivity profile |
Assay Linearity | Standard curve analysis | R² > 0.98 across working range |
Intra-assay Precision | Replicate measurements | CV < 10% |
Inter-assay Precision | Multiple independent assays | CV < 15% |
Functional Impact | Knockdown studies | Consistent with published effects on proliferation, migration and drug resistance |
Thorough validation ensures that experimental findings are reliable, reproducible, and truly reflect ORM1 biology rather than technical artifacts .
Researchers may encounter seemingly contradictory findings regarding ORM1's functions. Several methodological approaches can help reconcile these discrepancies:
Disease-Specific Effects:
Concentration-Dependent Effects:
Low versus high concentrations may activate different pathways
Acute versus chronic exposure may yield different outcomes
Establish complete dose-response relationships to identify biphasic effects
Model System Differences:
Compare in vitro vs. in vivo findings systematically
Account for species differences in ORM1 function
Consider cell type-specific responses to ORM1
Technical Considerations:
Antibody epitope differences may detect distinct ORM1 forms
Glycosylation heterogeneity may affect function and detection
Phosphorylation status alters ORM1 activity
Pathway-Based Integration:
Map contradictory findings to specific signaling nodes
Identify context-dependent regulatory mechanisms
Consider crosstalk between pathways (e.g., AKT/ERK and Nrf2/HO-1)
Meta-Analysis Techniques:
Systematically review published literature with standardized criteria
Account for methodological variables in analysis
Identify patterns across seemingly contradictory results
Direct Comparison Studies:
Side-by-side testing under identical conditions
Examination of multiple endpoints simultaneously
Controlled manipulation of key variables
Time-Course Experiments:
Evaluate temporal dynamics of ORM1 effects
Identify early versus late responses
Map sequence of molecular events
Isoform and Modification-Specific Analysis:
Distinguish effects of different glycoforms
Examine phosphorylation-dependent functions
Consider potential protein interaction partners
By applying these approaches, researchers can develop more nuanced and comprehensive models of ORM1 function that accommodate seemingly contradictory findings and explain its diverse roles across different pathophysiological contexts .