SCGB1A1-deficient (Scgb1a1–/–) mice exhibit strain-specific phenotypes:
C57Bl6 strain: Develop glomerulopathy resembling Goodpasture’s syndrome due to dysregulated IgA-fibronectin interactions .
129 strain: Show no glomerulopathy but display heightened susceptibility to hyperoxia-induced lung injury, increased IL-13/IL-6 production, and altered club cell morphology .
Scgb1a1-iCre mice: Engineered to express Cre recombinase under Scgb1a1 promoters without disrupting endogenous protein expression. Used for lineage tracing in pulmonary epithelial studies .
Alveolar Macrophage Regulation:
Asthma and COPD:
Strain | Phenotype | Key Pathways Affected |
---|---|---|
C57Bl6 | Glomerulopathy, IgA dysregulation | Fibronectin binding, immune complex clearance |
129 | Hyperoxia susceptibility, IL-13/IL-6 surge | Oxidative stress response, cytokine signaling |
Parameter | Specification |
---|---|
Purity | >98% (SDS-PAGE, RP-HPLC) |
Activity | ED₅₀ <5.0 µg/mL (A549 cell adhesion assay) |
Storage | Lyophilized; stable at -18°C (reconstituted: 4°C ≤7 days) |
SCGB1A1, also known as Club Cell Secretory Protein (CCSP) or uteroglobin, is primarily expressed in non-ciliated bronchiolar club cells in the lungs. In mice, SCGB1A1 is detected predominantly in bronchial and alveolar cells as demonstrated by lineage tracing studies using Scgb1a1-Cre/Rosa26-tdTomato mice . While the lung shows the strongest expression, some Scgb1a1-driven recombination has been detected in the spleen, suggesting limited expression in extrapulmonary tissues . Notably, SCGB1A1 is a component of pulmonary surfactant and has documented anti-inflammatory properties.
Multiple complementary methods can be employed to confirm SCGB1A1 expression:
Immunohistochemistry/Immunofluorescence: Fixed tissues can be stained using anti-SCGB1A1 antibodies, as demonstrated in studies where paraformaldehyde-fixed lung sections were stained with anti-SCGB1A1 antibodies (R&D Systems) and visualized with fluorophore-conjugated secondary antibodies (Alexa 488 or Alexa 594) .
RT-PCR/qPCR: RNA isolation followed by reverse transcription and PCR amplification using Scgb1a1-specific primers.
Reporter mouse models: Utilizing Scgb1a1-IRES-Cre mice crossed with reporter lines like Rosa26-tdTomato allows visualization of cells expressing Scgb1a1 through fluorescent protein expression .
Western blotting: Protein extraction from lung tissues followed by immunoblotting with anti-SCGB1A1 antibodies.
For accurate interpretation, researchers should include appropriate positive controls (wild-type lung tissue) and negative controls (tissues known not to express SCGB1A1 or samples from Scgb1a1 knockout mice).
Several mouse models exist for SCGB1A1 research:
Scgb1a1 knockout (KO) mice: Complete germline deletion of the Scgb1a1 gene (Scgb1a1−/−), useful for studying the physiological role of SCGB1A1 .
Scgb1a1-IRES-Cre mice: These mice express Cre recombinase from the endogenous Scgb1a1 locus (C57BL/6Smoc-Scgb1a1em1(IRES-Cre)Smoc), enabling conditional gene manipulation in SCGB1A1-expressing cells .
Inducible Scgb1a1-CreER mice: Allow for temporally controlled recombination specifically in club cells.
Reporter lines: When crossed with Cre-driver lines, mice such as Rosa26-tdTomato can be used to visualize and track SCGB1A1-expressing cells during development or in disease states .
Each model offers distinct advantages depending on the research question being addressed, from studies of complete SCGB1A1 deficiency to lineage tracing of club cells during development and disease.
SCGB1A1 deficiency significantly exacerbates asthma phenotypes in mouse models. When challenged with ovalbumin (OVA), Scgb1a1 knockout mice exhibited:
More severe airway inflammation with increased inflammatory cell infiltration
Downregulation of FoxA2, a transcriptional regulator also reduced in asthma conditions
These findings confirm the protective, anti-inflammatory role of SCGB1A1 in asthma pathogenesis. The research suggests that diminished SCGB1A1 levels, as observed in human asthma patients, may not merely be a consequence of the disease but actively contribute to worsening respiratory symptoms and inflammation . This provides a mechanistic link between the clinical observation of reduced SCGB1A1 levels in human asthma patients and the functional consequences of this reduction.
SCGB1A1 significantly influences alveolar macrophage (AM) development and inflammatory responses:
Developmental impact: Transcriptomic analysis revealed that AMs from Scgb1a1-sufficient mice showed upregulation of 37 biological pathways during development from weaning (4 weeks) to early adulthood (12 weeks), with 30 pathways directly involved in antigen presentation, anti-viral immunity, and inflammation .
Inflammatory regulation: Under Scgb1a1 deficiency, these immune-related pathways were significantly downregulated compared to age-matched Scgb1a1-sufficient mice .
Early inflammation: AMs from Scgb1a1-deficient mice demonstrated premature activation of inflammatory pathways compared to wild-type counterparts .
Cytokine modulation: In vitro experiments demonstrated that SCGB1A1 protein supplementation significantly reduced AM responses to microbial stimuli, blunting the release of pro-inflammatory cytokines and chemokines including IL-1β, IL-6, IL-8, MIP-1α, TNF-α, and MCP-1 after TLR stimulation .
These findings suggest SCGB1A1 plays a crucial role in shaping AM-mediated inflammation and immune responses, particularly in preventing excessive inflammatory responses to stimuli. This represents a novel mechanism by which club cells may influence lung immunity through secreted SCGB1A1.
During respiratory infections and inflammatory conditions, SCGB1A1 expression is significantly altered:
Viral infections: Human rhinovirus infection reduces expression of both SCGB1A1 and its transcriptional regulator FOXA2 in epithelial cells .
Th2 inflammation: Pro-allergic Th2 cytokines (IL-4 and IL-13) repress epithelial expression of SCGB1A1 and FOXA2, linking allergic inflammation to reduced SCGB1A1 levels .
Asthma: Analysis of multiple gene expression datasets revealed that SCGB1A1 mRNA is significantly reduced (by approximately 53%) in epithelial cells from asthma patients compared to healthy controls, while the mucin gene MUC5AC is elevated approximately twofold .
Age-related changes: In Scgb1a1-sufficient mice, AM activation of immune pathways increases from weaning to early adulthood, but this development is impaired in Scgb1a1-deficient mice .
These expression changes suggest SCGB1A1 functions in a complex regulatory network that responds to environmental challenges. The reduction in SCGB1A1 during inflammatory conditions may contribute to disease exacerbation by removing a key anti-inflammatory mediator.
For robust experimental asthma induction in SCGB1A1 mouse models, researchers should consider the following protocol elements:
Ovalbumin (OVA) sensitization and challenge:
Alternative models:
Readout parameters:
Airway hyperreactivity using whole-body plethysmography or forced oscillation techniques
Bronchoalveolar lavage (BAL) cell differential counts
Histopathological analysis of lung sections with H&E, PAS, and Masson's trichrome staining
Immunostaining for SCGB1A1 and MUC5AC to assess club cell integrity and mucus production
Controls:
This comprehensive approach allows for detailed characterization of how SCGB1A1 deficiency influences asthma pathogenesis and provides a platform for testing potential therapeutic interventions.
Effective lineage tracing with Scgb1a1-IRES-Cre mice requires careful experimental design:
Cross with appropriate reporter lines:
Validation of Cre activity:
Confirm recombination efficiency through direct fluorescence microscopy
Analyze tissue-specific recombination patterns (in lung, Scgb1a1-Cre drives recombination in bronchial and alveolar cells)
Test for off-target recombination in other tissues (some recombination has been detected in the spleen with Scgb1a1-Cre)
Temporal studies:
Perform analyses at multiple timepoints to track cell fate
For developmental studies, examine embryonic, neonatal, and adult stages
For injury models, establish baseline recombination before injury and follow recovery
Technical considerations:
Use co-staining with cell-specific markers to confirm identity of lineage-traced cells
Employ confocal microscopy with Z-stack imaging for three-dimensional analysis
Consider flow cytometry for quantitative assessment of lineage-traced populations
Controls:
Include Cre-negative littermates
Use alternative Cre driver lines to validate findings (e.g., other club cell-targeting Cre lines)
This approach allows researchers to trace the fate of Scgb1a1-expressing cells during development, homeostasis, and disease conditions, providing insights into club cell dynamics and potential contribution to epithelial regeneration.
Isolating and maintaining primary club cells requires specialized techniques:
Isolation procedures:
Bronchioalveolar lavage: For collecting superficial club cells (limited yield)
Enzymatic digestion: Using dispase/collagenase treatment of lung tissue
Flow cytometry: Sorting based on Scgb1a1-driven fluorescent reporter expression
Magnetic bead separation: Using antibodies against club cell surface markers
Culture conditions:
Media composition: DMEM/F12 supplemented with insulin, transferrin, selenium, hydrocortisone, EGF, bovine pituitary extract, and retinoic acid
Substratum: Growth on collagen-coated surfaces or air-liquid interface cultures
3D organoid culture: Mixed with Matrigel to form bronchospheres
Co-culture systems: With fibroblasts to better mimic the in vivo niche
Verification of cell identity:
Experimental applications:
These methods enable the study of primary SCGB1A1-expressing cells in controlled environments, allowing detailed investigation of their responses to various stimuli and potential therapeutic interventions.
SCGB1A1 mouse models offer valuable tools for studying airway regeneration:
Lineage tracing approaches:
Injury models compatible with SCGB1A1 studies:
Naphthalene injury: Targets club cells through cytochrome P450-2F2 metabolism
Influenza infection: Causes widespread epithelial damage
Bleomycin: Induces fibrotic responses following epithelial injury
Ozone exposure: Models oxidative damage to the airway epithelium
Analytical approaches:
Single-cell RNA sequencing to characterize regenerative cell populations
Spatial transcriptomics to map regeneration zones
Time-course analysis of SCGB1A1 re-expression during repair
Co-localization studies of SCGB1A1 with proliferation markers (Ki67, BrdU)
Mechanistic investigations:
Conditional knockout of regeneration-associated genes in SCGB1A1+ cells
Selective ablation of SCGB1A1+ cells to assess their contribution to repair
Exogenous SCGB1A1 administration to determine if it accelerates regeneration
These approaches can reveal the contribution of club cells to epithelial maintenance and regeneration, which has implications for understanding chronic lung diseases characterized by impaired repair, such as COPD and pulmonary fibrosis.
SCGB1A1 mouse models have revealed critical immunomodulatory functions of club cells:
Alveolar macrophage programming:
SCGB1A1 influences the developmental trajectory of alveolar macrophages (AMs)
Transcriptomic analysis showed that Scgb1a1 sufficiency is associated with upregulation of 37 biological pathways in AMs during development from weaning to adulthood, with 30 pathways directly involved in antigen presentation, anti-viral immunity, and inflammation
Under Scgb1a1 deficiency, these immune-related pathways were significantly downregulated compared to wild-type mice
Cytokine regulation:
SCGB1A1 protein supplementation significantly reduced AM responses to microbial stimuli in vitro
SCGB1A1 blunted the release of multiple pro-inflammatory cytokines and chemokines including IL-1β, IL-6, IL-8, MIP-1α, TNF-α, and MCP-1 after TLR stimulation
This suggests SCGB1A1 acts as a natural brake on inflammatory responses in the lung
Response to pathogen-associated molecular patterns:
Age-dependent immune regulation:
These findings collectively establish SCGB1A1 as a critical mediator in club cell-macrophage crosstalk and highlight the importance of epithelial-derived factors in shaping lung immune responses.
Effective investigation of SCGB1A1 protein function requires specialized methodological approaches:
Protein sources and preparation:
Cellular models:
Functional assays:
Inflammation studies:
Gene regulation studies:
Molecular interaction studies:
Co-immunoprecipitation to identify SCGB1A1 binding partners
Surface plasmon resonance to measure binding kinetics
Confocal microscopy to track cellular uptake and localization
Controls and validation:
Heat-inactivated SCGB1A1 protein as negative control
Dose-response curves to establish optimal concentrations
Comparison with other secretoglobin family members
Knockdown/knockout validation in relevant cell types
These methodological approaches provide a comprehensive framework for investigating SCGB1A1's functions in inflammation regulation, cellular protection, and immune modulation in controlled experimental systems.
Multiple lines of evidence support SCGB1A1's therapeutic potential:
Anti-inflammatory effects:
Protection in asthma models:
Mitigation of cytokine responses:
Clinical correlations:
Emerging applications:
These findings collectively suggest that recombinant SCGB1A1 administration or strategies to enhance endogenous SCGB1A1 expression could represent novel therapeutic approaches for inflammatory lung diseases and potentially other conditions.
Translating findings from SCGB1A1 mouse models to human disease reveals important parallels and considerations:
Expression patterns and reductions in disease:
Genetic aspects:
Regulatory mechanisms:
Functional outcomes:
Translational challenges:
Species differences in club cell distribution and abundance must be considered
Mouse models may not fully recapitulate the chronic nature of human respiratory diseases
Human genetic diversity creates variability not captured in inbred mouse strains
These translational insights provide a framework for developing SCGB1A1-based therapeutic strategies that could be effective in human disease, while highlighting important considerations for proper interpretation of mouse model findings.
Multiple strategic approaches can target or enhance SCGB1A1 expression:
Direct protein delivery:
Recombinant SCGB1A1 protein administration via aerosolization
Lipid nanoparticle encapsulation for improved stability and delivery
PEGylation to increase half-life in the respiratory tract
Development of SCGB1A1 analogues with enhanced stability or function
Transcriptional enhancement:
Gene therapy approaches:
AAV-mediated delivery of SCGB1A1 expression cassettes
CRISPR activation systems targeting the endogenous SCGB1A1 promoter
Club cell-targeted delivery using Scgb1a1 promoter-driven constructs
Epigenetic modulation:
Histone deacetylase inhibitors to promote chromatin accessibility at the SCGB1A1 locus
DNA methyltransferase inhibitors if SCGB1A1 is silenced by methylation in disease states
Pathway-based approaches:
Combination therapies:
SCGB1A1 enhancement combined with anti-inflammatory corticosteroids
SCGB1A1 with bronchodilators for asthma and COPD applications
Sequential therapy targeting acute inflammation followed by SCGB1A1-based tissue repair
These approaches offer multiple avenues for therapeutic development, with selection depending on the specific disease context, delivery challenges, and desired functional outcomes.
Researchers commonly encounter several challenges when working with SCGB1A1 mouse models:
Phenotypic variability:
Challenge: Inconsistent phenotypes between studies or mouse colonies
Solution: Maintain consistent genetic background through backcrossing; report complete strain information; use littermate controls; standardize housing conditions and microbiome status
Off-target effects in Cre models:
Developmental compensation:
Challenge: Germline Scgb1a1 knockout mice may develop compensatory mechanisms
Solution: Use inducible knockout systems; compare acute vs. chronic deletion effects; examine expression of other secretoglobin family members
Technical detection issues:
Disease model variability:
Translational limitations:
Challenge: Differences between mouse and human club cell biology
Solution: Validate key findings in human samples or cell systems; acknowledge species differences in result interpretation; consider using humanized mouse models when appropriate
Addressing these challenges requires careful experimental design, proper controls, and transparent reporting of methodological details to ensure reproducibility and translatability of findings.
When faced with contradictory findings in SCGB1A1 mouse studies, researchers should employ a systematic approach to reconciliation:
Methodological differences assessment:
Mouse strain variations: Different genetic backgrounds can significantly influence phenotypes
Model specifics: Compare knockout strategies (germline vs. conditional), Cre driver lines, and reporter systems
Age considerations: SCGB1A1 functions change with development—compare if studies used mice of different ages
Sex differences: Determine if contradictions could be explained by male vs. female differences
Experimental context evaluation:
Disease induction protocols: Variations in allergen dose, route, adjuvant, and challenge schedule
Environmental factors: Housing conditions, microbiome composition, and pathogen status
Timing of analysis: Acute vs. chronic responses may differ substantially
Molecular mechanism investigation:
Reconciliation strategies:
Direct replication studies: Using standardized protocols across laboratories
Meta-analysis approaches: Systematically analyzing multiple datasets, as done for human SCGB1A1 expression in asthma
Combinatorial experiments: Simultaneously testing multiple variables to identify interaction effects
Single-cell approaches: Determining if contradictions reflect differences in specific cell populations
Translational relevance assessment:
Compare mouse findings with human data when available
Consider if contradictions reflect the heterogeneity also seen in human disease
This structured approach can help researchers navigate apparent contradictions and extract meaningful biological insights from seemingly discrepant results in the SCGB1A1 literature.
Maintaining high-quality SCGB1A1 mouse lines requires rigorous quality control measures:
Genotyping protocols:
Regular PCR-based genotyping using primers specific to wild-type and modified Scgb1a1 alleles
For Scgb1a1-Cre lines, additional primers to detect Cre recombinase
Periodic verification of genotyping accuracy through alternative methods
Include positive and negative controls with every genotyping batch
Expression verification:
Periodic confirmation of SCGB1A1 protein expression (or absence) through immunohistochemistry of lung sections
For Cre driver lines, validation of recombination patterns using reporter crosses
qPCR assessment of Scgb1a1 transcript levels in lung tissue
Western blot analysis of SCGB1A1 protein in BAL fluid
Colony management:
Maintain detailed pedigree records to monitor inbreeding
Regular backcrossing to foundational strains (typically every 10 generations)
Cryopreservation of embryos from early generations as backup
Periodic health monitoring for pathogens that could affect lung phenotypes
Experimental consistency:
Standardize age for experiments based on developmental SCGB1A1 expression patterns
Consider sex as a biological variable in all experiments
Maintain consistent environmental conditions (temperature, humidity, light cycles)
Control for housing density and cage position effects
Phenotypic drift monitoring:
Regular benchmark testing of key phenotypes (e.g., baseline lung function, club cell numbers)
Periodic comparison to historical data to detect subtle phenotypic drift
Sentinel monitoring for environmental changes that could affect phenotype
Documentation standards:
Detailed record-keeping of breeding performance and health status
Documentation of any unexpected phenotypes or breeding challenges
Transparency in reporting colony management practices in publications
Uteroglobin is a small, secreted, disulfide-bridged dimeric protein. It is composed of two 75 amino acid polypeptide chains, forming a homodimeric structure . The recombinant form of mouse uteroglobin is typically expressed in E. coli or HEK293 cells, and it is often tagged with a polyhistidine tag for purification purposes .
Uteroglobin has several important biological functions:
Recombinant mouse uteroglobin is used in various research applications, including:
Recombinant mouse uteroglobin is typically lyophilized and stored at -20°C to -80°C for long-term stability. Upon reconstitution, it is most stable at -20°C to -80°C and can be stored for up to one week at 2-8°C . It is important to avoid repeated freeze-thaw cycles to maintain its stability and activity.