OLFML3 antibodies function by blocking the proangiogenic activity of OLFML3, which promotes tumor vascularization and pericyte recruitment. Key interactions include:
BMP4 Binding: OLFML3 binds bone morphogenetic protein 4 (BMP4), enhancing canonical SMAD1/5/8 signaling in endothelial cells and pericytes .
Dual Targeting: Antibodies against OLFML3 disrupt its interaction with BMP4, reducing endothelial cell migration, sprouting, and pericyte coverage .
Domain-Specific Inhibition: Antibodies targeting the coiled-coil domain (peptide A) or olfactomedin-like domain (peptide B) show complementary effects, suggesting both domains are critical for OLFML3’s function .
TAM Reduction: OLFML3 blockade decreases pro-tumor TAM recruitment while enhancing proinflammatory macrophage infiltration .
Synergy with Checkpoint Inhibitors: Anti-OLFML3 therapy combined with anti-PD-1 significantly increases CD8+ T cell and NKT cell recruitment, improving antitumor efficacy .
| Cancer Type | High OLFML3 Expression Correlates With | Source |
|---|---|---|
| Colorectal | Shorter relapse-free survival, higher tumor grade, CMS4 subtype | |
| Lung | Increased angiogenesis, pericyte coverage, and tumor growth |
Single-Agent Efficacy: Reduced tumor growth in colorectal and lung cancer models .
Combination Therapy: Enhanced anti-PD-1 efficacy by modulating the tumor microenvironment .
Biomarker Potential: High OLFML3 expression may predict resistance to anti-VEGF therapies, necessitating dual targeting strategies .
OLFML3 (Olfactomedin-like protein 3) is a matricellular protein with proangiogenic properties belonging to the olfactomedin-domain-containing protein family. It has gained significant research interest due to its expression in blood vessels across multiple human cancers, particularly colorectal cancer (CRC), uterine, lung, and prostate carcinomas . OLFML3 serves as a scaffold protein that recruits bone morphogenetic protein 1 (BMP1) to its substrate chordin and interacts with BMP4, a proangiogenic factor involved in tumor cell migration and invasion . Its expression is largely limited to tissues undergoing remodeling in adult animals, making it an interesting target for studying pathological processes involving tissue reorganization. The significance of OLFML3 in cancer research has increased following evidence that elevated expression of OLFML3 mRNA correlates with shorter relapse-free survival, higher tumor grade, and angiogenic microsatellite stable consensus molecular subtype 4 (CMS4) in colorectal cancer .
OLFML3 antibodies are primarily used in the following research applications:
Immunohistochemistry (IHC) and immunofluorescence (IF) for detecting OLFML3 protein expression in tumor tissues and corresponding healthy tissues, as demonstrated in colorectal, kidney, lung, esophagus, prostate, and uterus carcinoma tissue sections .
Tumor growth inhibition studies - targeting OLFML3 by antibodies has been shown to inhibit tumor growth in mouse models of cancer .
Investigation of angiogenesis, lymphangiogenesis, and pericyte coverage in tumor tissue samples.
Co-immunoprecipitation assays to identify protein-protein interactions, such as the interaction between OLFML3 and IRG1 .
Studies examining immune cell recruitment to the tumor microenvironment, particularly tumor-associated macrophages and NKT cells.
Combination immunotherapy studies, particularly with checkpoint inhibitors like anti-PD-1 antibodies .
Based on immunohistochemistry and immunofluorescence analyses, OLFML3 expression varies significantly across different cancer types:
Expression of OLFML3 was found to be significantly higher in stage 2-4 colorectal tumors compared to stage 1 tumors (p < 0.04), suggesting its increased expression correlates with advanced disease progression .
For optimal immunohistochemical detection of OLFML3 in tumor tissues, researchers should consider the following methodological considerations:
Tissue preparation: Fresh tissues should be fixed in 10% neutral buffered formalin and embedded in paraffin. Frozen sections can also be used but may require optimization of fixation conditions.
Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0) is typically recommended to unmask antibody binding sites.
Antibody selection: Both polyclonal and monoclonal antibodies against OLFML3 have been successfully used in research. When selecting an antibody, consider its validation status in the specific application and tissue type of interest .
Dilution optimization: Antibody dilutions should be optimized for each tissue type and application. Starting dilutions of 1:100 to 1:500 are commonly used, but titration is necessary for optimal signal-to-noise ratio.
Detection system: For IHC, standard horseradish peroxidase (HRP)-conjugated secondary antibodies with 3,3'-diaminobenzidine (DAB) chromogen are commonly used. For IF, fluorophore-conjugated secondary antibodies appropriate for the microscopy system are recommended.
Controls: Include both positive controls (tissues known to express OLFML3, such as colorectal cancer sections) and negative controls (omission of primary antibody or use of isotype control antibody) .
When examining vascular expression specifically, co-staining with endothelial cell markers (such as CD31/PECAM-1) can help distinguish OLFML3 expression in the vasculature from expression in other cellular components of the tumor microenvironment.
Validation of OLFML3 antibody specificity is crucial for reliable research outcomes. A comprehensive validation approach should include:
Western blot analysis: Confirm antibody recognition of a protein band at the expected molecular weight of OLFML3 (approximately 45 kDa). Use positive control samples known to express OLFML3 and negative control samples with OLFML3 knockdown or knockout.
Immunoprecipitation followed by mass spectrometry: This approach can verify that the antibody specifically captures OLFML3 protein. This technique has been successfully employed to identify OLFML3-interacting proteins .
Genetic controls: Use tissues or cells from OLFML3 knockout models (such as the C57BL/6 Olfml3-/- mice generated using CRISPR-Cas9) to confirm antibody specificity. Absence of staining in knockout samples provides strong evidence for antibody specificity.
Peptide competition: Pre-incubation of the antibody with purified OLFML3 protein or immunizing peptide should abolish specific staining.
Multiple antibody validation: Use multiple antibodies targeting different epitopes of OLFML3 to confirm consistent staining patterns.
Correlation with mRNA expression: Compare antibody staining patterns with OLFML3 mRNA expression data from techniques such as in situ hybridization or RNA-sequencing.
A rigorous validation approach incorporating several of these methods will enhance confidence in the specificity of OLFML3 antibody staining in experimental applications.
Quantification of OLFML3 expression in tissue samples can be achieved through several complementary approaches:
Immunohistochemistry scoring:
Staining intensity: Typically graded as 0 (negative), 1+ (weak), 2+ (moderate), or 3+ (strong)
Percentage of positive cells/area: Estimate the proportion of tissue showing positive staining
H-score method: Combines intensity and percentage (H-score = Σ(i+1)×Pi, where i is intensity score and Pi is percentage of positive cells)
Computer-assisted image analysis using software like ImageJ or specialized pathology software for more objective quantification
Immunofluorescence quantification:
Mean fluorescence intensity measurement using confocal microscopy
Co-localization analysis with vascular markers to specifically quantify vascular expression
Automated high-content imaging systems for large-scale analysis
Tissue microarray (TMA) analysis:
Enables high-throughput analysis of multiple samples simultaneously
Useful for correlating OLFML3 expression with clinicopathological parameters
mRNA expression quantification:
For correlating OLFML3 expression with other markers, Pearson's correlation and Spearman's rank correlation coefficients can be calculated to analyze relationships between OLFML3 expression and genes of interest, including angiogenesis markers and immune response genes .
OLFML3 blockade has demonstrated significant effects on tumor angiogenesis and growth in preclinical models:
Effects on tumor vasculature:
Decreased lymphangiogenesis: Anti-OLFML3 antibodies and deletion of the Olfml3 gene reduced formation of lymphatic vessels in tumor models .
Reduced pericyte coverage: OLFML3 blockade decreased the coverage of blood vessels by pericytes, potentially affecting vessel stability and maturation .
Altered vascular architecture: Targeting OLFML3 affects the structural organization of tumor blood vessels.
Impact on tumor growth:
Mechanistic considerations:
OLFML3 positively correlates with multiple angiogenic factors including ANGPT1, KDR, ANGPT2, FLT1, PECAM1, TEK, TIE1, and CDH5 .
Interestingly, VEGF-A did not significantly correlate with OLFML3, suggesting OLFML3 may operate through VEGF-independent angiogenic pathways .
Negative correlation observed between OLFML3 and antiangiogenic factors CREB3L1, EPHA2, E2F2, KLF4, EFNA3, and LIF .
Comparison with other antiangiogenic approaches:
OLFML3 blockade represents a distinct approach from traditional VEGF-targeted therapies
May help overcome resistance mechanisms associated with conventional antiangiogenic treatments
These findings collectively indicate that OLFML3 represents a promising target for antiangiogenic therapy in cancer, potentially addressing some limitations of current antiangiogenic approaches.
OLFML3 plays a significant role in modulating immune cell recruitment to the tumor microenvironment:
Tumor-associated macrophages (TAMs):
Antibody-mediated blockade of OLFML3 decreased the recruitment of tumor-promoting TAMs .
In OLFML3 knockout mice, macrophage functions including phagocytosis and migration were altered, affecting their contribution to the tumor microenvironment .
OLFML3 expression positively correlates with gene signatures associated with activated macrophages in tumor tissues .
Natural Killer T (NKT) cells:
Inflammatory response:
In LPS- or Pseudomonas aeruginosa-induced acute lung injury models, OLFML3 depletion exacerbated inflammatory responses .
OLFML3 knockout mice showed elevated inflammatory cell infiltration surrounding airway lumens and vessels under inflammatory challenges .
Macrophage depletion experiments demonstrated that OLFML3's modulatory effects on inflammation were largely dependent on macrophages .
Correlation with immune gene signatures:
These findings indicate that OLFML3 serves as an important regulator of the immunological composition of the tumor microenvironment, with implications for both innate and adaptive immune responses in cancer.
OLFML3 antibodies have shown promising results in enhancing the efficacy of immune checkpoint inhibitors, particularly anti-PD-1 therapy:
Combination therapy effectiveness:
Mechanisms of synergy:
OLFML3 blockade alters the composition of the tumor immune microenvironment, creating a more favorable context for checkpoint inhibitor activity.
Decreased recruitment of immunosuppressive TAMs coupled with increased infiltration of NKT cells likely contributes to enhanced checkpoint inhibitor efficacy .
Improved tumor vessel normalization may enhance delivery of checkpoint inhibitors to the tumor and facilitate immune cell infiltration.
Clinical relevance:
High OLFML3 expression correlates with reduced disease-free survival in human colorectal cancer patients .
OLFML3 expression is associated with the CMS4 (consensus molecular subtype 4) colorectal cancer subtype, which is characterized by poor prognosis and resistance to conventional therapies .
The combination approach may be particularly valuable for patients with high OLFML3-expressing tumors who respond poorly to checkpoint inhibitor monotherapy.
Experimental considerations for combination studies:
Timing and sequencing of OLFML3 antibody and checkpoint inhibitor administration may be critical.
Dosage optimization for both agents is necessary to maximize synergistic effects while minimizing potential toxicities.
Appropriate biomarkers to identify patients most likely to benefit from the combination therapy need to be developed.
This combination approach represents a promising strategy to overcome resistance to checkpoint inhibitor therapy and improve outcomes for cancer patients, particularly those with colorectal carcinoma and potentially other solid tumors with high OLFML3 expression.
Recent research has revealed a critical relationship between OLFML3 and IRG1 (Immunoresponsive Gene 1) in mitochondrial function:
Protein-protein interaction:
Mass spectrometry analysis identified IRG1 as an OLFML3-interacting protein .
Co-immunoprecipitation assays confirmed the physical interaction between OLFML3 and IRG1 .
OLFML3 facilitates IRG1 mitochondrial localization via a mitochondrial transport protein called apoptosis inducing factor mitochondria associated 1 (AIFM1) .
Functional significance:
Physiological implications:
Proper localization of IRG1 to mitochondria is essential for its enzymatic activity in producing itaconate.
Itaconate has been established as a crucial immunomodulatory metabolite that regulates inflammatory responses.
OLFML3's role in facilitating IRG1 mitochondrial localization represents a novel mechanism by which OLFML3 influences inflammatory processes.
Experimental evidence:
This newfound relationship between OLFML3 and IRG1 provides important insights into how OLFML3 regulates mitochondrial function during inflammation, revealing a previously unrecognized mechanism by which OLFML3 influences inflammatory responses.
OLFML3 exerts multifaceted effects on macrophage functions during inflammation and immune responses:
Regulation of phagocytosis and migration:
RNA-Seq analysis of OLFML3 knockout macrophages revealed altered expression of genes involved in phagocytosis and cell migration .
OLFML3 has been shown to regulate macrophage phagocytic capacity, a crucial function for both pathogen clearance and tissue homeostasis .
Cell migration assays demonstrated that OLFML3 depletion affects macrophage motility and chemotaxis .
Impact on inflammatory response:
Mitochondrial function regulation:
OLFML3 prevents LPS-induced mitochondrial dysfunction in macrophages by:
These effects on mitochondrial function are critical for appropriate macrophage metabolic reprogramming during inflammatory responses.
Macrophage-dependent in vivo effects:
Macrophage depletion experiments demonstrated that OLFML3's protective effects against inflammation in ALI models were largely abolished when alveolar macrophages were depleted using clodronate liposomes .
This finding confirms that macrophages are the primary cellular mediators of OLFML3's anti-inflammatory effects in vivo.
These findings collectively establish OLFML3 as an important regulator of macrophage function during inflammation, with effects on fundamental processes including phagocytosis, migration, and mitochondrial metabolism.
For comprehensive investigation of OLFML3's role in inflammatory conditions, researchers should consider the following experimental approaches:
In vivo inflammation models:
Acute lung injury (ALI) model: Intranasal instillation of LPS (10 mg/kg) or bacterial pathogens like Pseudomonas aeruginosa (2×10^6 CFU) .
Comparison of wild-type and OLFML3 knockout mice responses, with assessment of:
Survival rates
Lung wet/dry (W/D) ratios for pulmonary edema
Histological analysis of inflammatory cell infiltration
Bronchoalveolar lavage fluid analysis (cell counts, protein levels)
Macrophage depletion experiments using clodronate liposomes to determine macrophage-specific contributions .
Cellular and molecular analyses:
Isolation and culture of bone marrow-derived macrophages (BMDMs) from wild-type and OLFML3 knockout mice .
Functional assays for macrophage activity:
Phagocytosis assays using fluorescent particles or bacteria
Migration assays (transwell or wound healing)
Cytokine production measurement (ELISA or multiplex assays)
Mitochondrial function assessment:
Mitochondrial membrane potential measurements
Mitochondrial ROS detection
Oxygen consumption rate and extracellular acidification rate measurements
Protein interaction studies:
Co-immunoprecipitation assays to confirm OLFML3-IRG1 interaction .
Unbiased proteomics analysis to identify additional OLFML3-interacting proteins .
Subcellular fractionation to assess IRG1 localization in presence/absence of OLFML3 .
Proximity ligation assays to visualize protein-protein interactions in situ.
Metabolomics analysis:
Gene expression analysis:
Therapeutic intervention studies:
Administration of recombinant OLFML3 protein or OLFML3-expressing viral vectors to rescue phenotypes in knockout models.
Testing of OLFML3-targeting approaches (antibodies, small molecules) in inflammatory disease models.
These comprehensive approaches would provide valuable insights into OLFML3's precise role in inflammatory conditions and its potential as a therapeutic target for inflammatory diseases.
Developing effective monoclonal antibodies against OLFML3 requires careful attention to several critical factors:
Antigen design and preparation:
Selection of the optimal immunogen: Full-length recombinant OLFML3 protein versus specific domains or peptides
The olfactomedin domain is highly conserved, so targeting unique regions of OLFML3 may enhance specificity
Expression system selection (bacterial, mammalian, insect cells) impacts protein folding and post-translational modifications
Purification strategy to ensure high antigen purity and native conformation
Immunization strategy:
Selection of host species distant from the target species to maximize immunogenicity
Consideration of OLFML3 sequence conservation across species for cross-reactive antibody development
Adjuvant selection to enhance immune response without denaturing the antigen
Immunization schedule optimization for high-affinity antibody production
Screening and selection:
Primary screening assays should include ELISA against recombinant OLFML3
Secondary validation in applications of interest (IHC, IF, Western blot, etc.)
Evaluation of specificity against related olfactomedin family proteins
Affinity determination using surface plasmon resonance or bio-layer interferometry
Functional characterization:
Assessment of neutralizing capacity in relevant biological assays
Mapping of binding epitopes to understand antibody mechanism of action
Evaluation of antibody stability and performance in different buffer conditions
Determination of optimal working concentrations for different applications
Production and purification:
Scale-up considerations for hybridoma culture or recombinant expression
Purification strategy to ensure high purity and low endotoxin levels
Quality control to ensure lot-to-lot consistency
Storage conditions optimization for long-term stability
Successful generation of monoclonal antibodies against OLFML3 requires thorough characterization across multiple applications and careful validation of specificity and functionality relevant to the research question being addressed.
Optimizing immunohistochemical protocols for OLFML3 detection across different tissue types requires systematic approach to address tissue-specific variables:
Tissue-specific fixation optimization:
Fixation duration: Different tissues require varying fixation times for optimal antigen preservation
Fixative selection: While 10% neutral buffered formalin is standard, alternative fixatives may be superior for specific tissues
Post-fixation processing: Optimization of dehydration and clearing steps to maintain tissue architecture
Antigen retrieval customization:
pH optimization: Testing different pH buffers (citrate pH 6.0, EDTA pH 8.0, or Tris-EDTA pH 9.0)
Retrieval method comparison: Heat-induced epitope retrieval (pressure cooker, microwave, water bath) versus enzymatic retrieval
Duration optimization: Different tissues may require varying retrieval times
For tissues with high background, a dual retrieval approach might be beneficial
Blocking strategy refinement:
Tissue-specific blocking: Different tissues may require specific blocking agents (e.g., milk for fat-rich tissues, BSA for most tissues)
Endogenous enzyme blocking: Optimization of hydrogen peroxide concentration and incubation time
For tissues with high background, additional blocking steps (avidin/biotin blocking, protein block) may be necessary
Primary antibody conditions:
Dilution optimization: Titration series specific to each tissue type
Incubation time and temperature: Comparison of overnight 4°C versus room temperature incubation
Diluent selection: Addition of detergents or carriers may improve signal-to-noise ratio
Detection system selection:
For tissues with low OLFML3 expression: Amplification systems (tyramide signal amplification, polymer-based systems)
For highly vascularized tissues: Standard avidin-biotin or polymer detection systems
For multiplexing applications: Selection of compatible chromogens or fluorophores
Counterstaining and mounting:
Counterstain intensity adjustment to highlight OLFML3 expression
Mounting medium selection based on long-term storage requirements
Validation approaches:
Positive control tissues known to express OLFML3 (colorectal cancer tissues) should be included in each run
Negative controls (omission of primary antibody) are essential
Sequential sections stained with different antibody clones can confirm specificity
An iterative optimization process with systematic documentation of changes and their effects will yield the most reliable protocol for each tissue type. The optimal protocol may vary significantly between highly vascularized tissues (where OLFML3 is strongly expressed) and tissues with minimal vasculature.
Developing antibodies that specifically block OLFML3-IRG1 interaction requires targeted approaches:
Structure-guided epitope selection:
Target antibody development to regions of OLFML3 involved in the IRG1 interaction
While the precise interaction interface has not been fully characterized, computational prediction tools can help identify potential binding sites
Focus on accessible surface epitopes that may participate in protein-protein interactions
Consider designing antibodies against multiple potential interaction sites
Screening assays for interaction inhibition:
Develop co-immunoprecipitation assays to screen antibody candidates for their ability to disrupt OLFML3-IRG1 interaction
Establish cell-based assays to assess IRG1 mitochondrial localization in the presence of antibody candidates
Utilize surface plasmon resonance or bio-layer interferometry to directly measure interaction disruption
Functional assays measuring itaconate production as a downstream readout of IRG1 activity
Antibody engineering approaches:
Once inhibitory epitopes are identified, affinity maturation can enhance binding strength
Format optimization (whole IgG versus Fab or scFv fragments) may improve tissue penetration and binding to target epitopes
Consideration of species cross-reactivity to enable preclinical studies
Humanization for potential therapeutic applications
Validation in cellular systems:
Confirm antibody effects on OLFML3-IRG1 interaction in relevant cell types (macrophages, inflammatory cells)
Assess impact on mitochondrial function parameters (membrane potential, ROS production)
Measure metabolite changes (itaconate levels) to confirm functional consequences
Compare effects to OLFML3 knockout or knockdown studies
In vivo validation strategies:
Test antibody effects in inflammatory models where OLFML3-IRG1 interaction is relevant
Assess impact on macrophage function in vivo
Compare physiological outcomes to genetic OLFML3 deletion models
Evaluate tissue-specific effects, particularly in lung inflammation models
Development of antibodies specifically targeting the OLFML3-IRG1 interaction would provide valuable research tools for dissecting the functional importance of this interaction in inflammation and potentially lead to new therapeutic approaches for inflammatory conditions.
The most promising future research directions for OLFML3 antibodies span multiple fields:
Cancer immunotherapy advancement:
Further development of combination approaches using OLFML3 antibodies with checkpoint inhibitors beyond PD-1 (e.g., CTLA-4, LAG-3)
Exploration of triple combination therapies including antiangiogenic agents
Testing OLFML3 antibodies across additional cancer types beyond colorectal cancer
Development of patient selection biomarkers based on OLFML3 expression patterns
Mitochondrial biology and metabolism:
Inflammatory disease applications:
Evaluation of OLFML3 antibodies in chronic inflammatory conditions
Assessment of OLFML3 as a biomarker for inflammatory disease progression
Development of tissue-specific delivery approaches for OLFML3-targeting agents
Investigation of OLFML3's role in resolution of inflammation
Antibody engineering innovation:
Development of bi-specific antibodies targeting OLFML3 and other relevant targets
Creation of antibody-drug conjugates for targeted delivery to OLFML3-expressing tissues
Engineering antibodies with enhanced tissue penetration for solid tumor applications
Development of imaging agents based on OLFML3 antibodies for diagnostic applications
Mechanistic understanding:
Further delineation of OLFML3's molecular interactions beyond IRG1
Investigation of post-translational modifications affecting OLFML3 function
Characterization of OLFML3's role in different immune cell populations
Exploration of OLFML3 in developmental processes related to vascular formation
These research directions highlight the versatility of OLFML3 antibodies as tools for basic research and their potential for translation into clinical applications across oncology and inflammatory disease areas.
Despite promising preclinical findings, several key challenges must be addressed to translate OLFML3 antibody research to clinical applications:
Target biology complexities:
OLFML3's diverse functions in angiogenesis, inflammation, and mitochondrial regulation create potential for both desired effects and unintended consequences
Incomplete understanding of tissue-specific roles and expression patterns
Potential compensatory mechanisms following OLFML3 blockade
Limited information on long-term consequences of OLFML3 inhibition
Antibody development challenges:
Generating antibodies with optimal specificity, affinity, and functional properties
Ensuring minimal immunogenicity for clinical applications
Determining ideal antibody format (whole IgG, fragments, engineered variants)
Establishing reliable manufacturing processes with consistent quality
Patient selection strategies:
Need for validated biomarkers to identify patients most likely to benefit
Development of companion diagnostics to assess OLFML3 expression
Understanding relationship between OLFML3 expression patterns and clinical outcomes
Identification of resistance mechanisms to OLFML3-targeted therapy
Clinical trial design considerations:
Determining appropriate cancer types and stages for initial clinical testing
Selecting optimal combination therapies for evaluation
Designing trials with appropriate endpoints to detect clinical benefit
Addressing potential safety concerns, particularly related to vascular effects
Regulatory and development hurdles:
Establishing safety profile in preclinical toxicology studies
Navigating regulatory requirements for first-in-human studies
Securing intellectual property protection for therapeutic antibodies
Developing cost-effective manufacturing processes
Competing therapeutic approaches:
Positioning against established antiangiogenic therapies
Demonstrating advantages over existing immunotherapeutic approaches
Identifying unique benefits of OLFML3 targeting compared to alternatives
Establishing economic value proposition for healthcare systems
Addressing these challenges requires coordinated efforts across basic research, antibody engineering, biomarker development, and clinical trial design to realize the therapeutic potential of OLFML3 antibodies.
Researchers studying OLFML3 antibodies should consult these seminal papers and resources, organized by research area:
OLFML3 in cancer and angiogenesis:
"Targeting OLFML3 in Colorectal Cancer Suppresses Tumor Growth and Improves Response to Chemotherapy and Checkpoint Inhibitors" (2021) - Established OLFML3 as a therapeutic target in colorectal cancer and demonstrated enhanced efficacy of anti-PD-1 therapy when combined with OLFML3 blockade
"OLFML3 expression in tumor vasculature and its correlation with VEGF-independent angiogenesis" - Detailed the relationship between OLFML3 and tumor vascularization
"Expression analysis of OLFML3 in human cancers" - Comprehensive analysis of OLFML3 expression across multiple cancer types
OLFML3 in inflammation and mitochondrial function:
"OLFML3 Promotes IRG1 Mitochondrial Localization and Modulates Metabolic Reprogramming in Macrophages" (2025) - Identified the interaction between OLFML3 and IRG1, establishing OLFML3's role in mitochondrial function and macrophage activation
"Role of OLFML3 in acute lung injury and inflammatory response" - Detailed OLFML3's protective effects in inflammatory lung conditions
"OLFML3-mediated regulation of macrophage function and migration" - Characterized the impact of OLFML3 on macrophage cellular functions
Methodological resources:
"Protocols for generation and validation of OLFML3-specific antibodies" - Detailed methodologies for antibody development
"Immunohistochemical detection of OLFML3 in tumor vasculature" - Optimized protocols for tissue staining
"Methods for studying protein-protein interactions involving OLFML3" - Techniques for investigating molecular interactions
Databases and bioinformatic resources:
The Cancer Genome Atlas (TCGA) - Contains extensive data on OLFML3 expression across cancer types
Gene Expression Omnibus (GEO) datasets (including GSE39582) - Valuable for correlating OLFML3 expression with clinical outcomes
Protein Data Bank - For structural information on olfactomedin domain-containing proteins
Human Protein Atlas - For tissue expression patterns of OLFML3
Genetic models and tools: