The antibody targets an internal region of DROSHA (amino acids 1–100), a domain critical for its ribonuclease activity. The immunogen sequence was synthesized as a peptide to ensure high specificity .
Validated applications and suggested dilutions are as follows:
| Application | Dilution Range | Notes |
|---|---|---|
| ELISA | 1:500–1:2000 | Optimal dilution depends on sample type |
| Immunohistochemistry | 1:10–1:50 | Requires antigen retrieval optimization |
Specificity: The antibody demonstrates high specificity for human DROSHA, with no cross-reactivity reported against related ribonucleases .
Functional Role: DROSHA is essential for pri-miRNA processing in the nucleus, and this antibody has been used to study its localization and expression dynamics in cancer models .
Fluorescence Performance: FITC conjugation allows direct visualization in fluorescence microscopy, reducing reliance on secondary antibodies.
Sensitivity: FITC conjugation enhances detection sensitivity in low-abundance samples.
Versatility: Suitable for multiplex assays when combined with other fluorophores.
DROSHA is a ribonuclease III double-stranded (ds) RNA-specific endoribonuclease essential for microRNA (miRNA) biogenesis. As a component of the microprocessor complex, DROSHA cleaves primary miRNA transcripts (pri-miRNAs) to release precursor miRNA (pre-miRNA) in the nucleus . FITC (fluorescein isothiocyanate) conjugation allows direct visualization of DROSHA in cellular compartments through fluorescence microscopy without requiring secondary antibody steps.
For detection methodology, FITC-conjugated antibodies emit green fluorescence (excitation ~495nm, emission ~519nm) when excited with appropriate wavelengths, enabling spatial localization of DROSHA within subcellular structures. This approach is particularly valuable for studying DROSHA's role in miRNA processing and its interactions with chromatin at DNA damage sites .
Optimal sample preparation for DROSHA antibody-FITC staining requires:
Fixation protocol:
4% paraformaldehyde for 15 minutes at room temperature preserves cellular architecture while maintaining antibody accessibility to nuclear DROSHA
Avoid over-fixation which can mask epitopes and reduce signal intensity
Permeabilization method:
Use 0.1-0.5% Triton X-100 for 5-10 minutes to allow antibody access to nuclear DROSHA
For delicate samples, 0.05% saponin may provide gentler permeabilization
Blocking solutions:
2% human IgG (20 mg/ml) has proven more effective than FCS, BSA, or animal sera in reducing background fluorescence, particularly important with ionic fluorochromes like FITC
Alternatively, 2% BSA with 0.1% Tween-20 can be effective for standard applications
Antibody dilution optimization:
Recommended dilutions range from 1:50-1:500 for immunofluorescence applications, with exact optimization needed for each experimental system
Titrate antibody in preliminary experiments to determine optimal signal-to-noise ratio
Nonspecific binding is a significant challenge when using FITC-conjugated antibodies, particularly in samples with positively charged proteins. Research demonstrates several effective strategies:
Blocking optimization:
Human IgG at 2% concentration (20 mg/ml) significantly reduces background fluorescence from ionic fluorochromes like FITC
This concentration was superior to conventional blocking agents such as FCS, BSA, goat, horse, or normal human sera at 1-10% concentrations
Alternative fluorophore selection:
Consider using neutral fluorochromes like BODIPY FL for challenging samples, as they require less stringent blocking conditions (2% BSA is sufficient) compared to ionic FITC conjugates
BODIPY FL-conjugated antibodies demonstrate significantly lower nonspecific binding to positively charged proteins
Pre-absorption protocol:
Incubate antibodies with tissue/cell lysates from species not being studied
This step removes cross-reactive antibodies that contribute to background signal
Buffer optimization:
Adding 0.1-0.5% non-ionic detergents (Tween-20 or Triton X-100) to washing buffers
Including 150-300mM NaCl in buffers to disrupt weak ionic interactions
These methodological adjustments can dramatically improve signal specificity when detecting DROSHA in complex biological samples .
A comprehensive control strategy for DROSHA antibody-FITC experiments should include:
Positive controls:
HeLa nuclear extract serves as a reliable positive control for DROSHA antibody validation
HEK-293T, HEK-293, and HeLa cells have confirmed DROSHA expression detectable by Western blot
U2OS cells are validated for immunofluorescence applications with DROSHA antibodies
Negative controls:
Isotype control using non-specific IgG from the same species as the primary antibody
Secondary antibody-only control to assess non-specific binding
DROSHA knockdown samples using validated siRNAs to confirm antibody specificity
Specificity validation:
Rescue experiments using siRNA-resistant over-expression plasmids can confirm antibody specificity
Different siRNAs targeting DROSHA should produce consistent phenotypes to rule out off-target effects
Autofluorescence controls:
Unstained samples to assess natural autofluorescence of the specimen
Samples stained with only blocking reagents to evaluate background contribution
Implementing these controls ensures reliable interpretation of DROSHA localization and expression data in research applications.
DROSHA mutations significantly impact antibody epitope recognition through multiple mechanisms that researchers must consider when designing experiments:
Mutation-specific effects on epitope accessibility:
The P100L mutation in the Pro-rich domain and R279L in the Arg/Ser-rich domain of DROSHA affect protein conformation, potentially masking epitopes recognized by certain antibodies
These mutations disrupt interactions with cofactors including p68 (DDX5) and phosphorylated Smad1/5/8, which may alter epitope exposure in native conditions
Methodological considerations for mutant detection:
When studying known DROSHA mutants, researchers should select antibodies targeting epitopes distant from mutation sites
For example, antibodies targeting the C-terminal region (residues 1300 to C-terminus) like ab245398 may better detect N-terminal mutations
The D30F3 rabbit monoclonal antibody recognizes residues surrounding His549, making it suitable for detecting mutations in other domains
Validation strategies for mutant DROSHA:
When working with DROSHA mutants, Western blot validation should precede immunofluorescence studies to confirm antibody recognition
For suspected novel mutations, researchers should employ multiple antibodies targeting different epitopes to ensure detection
Expression vectors with epitope tags (FLAG, HA) provide alternative detection methods independent of conformational changes
Experimental interpretation considerations:
Reduced signal may indicate either decreased expression or epitope masking in mutant DROSHA
Researchers should distinguish these possibilities using mRNA quantification alongside protein detection
Research demonstrates DROSHA mutants P100L and R279L maintain similar expression levels despite compromised enzymatic activity
These considerations are crucial for accurate interpretation of results when studying DROSHA mutations implicated in vascular abnormalities and other pathological conditions .
Distinguishing specific DROSHA-FITC signals from autofluorescence requires sophisticated methodological approaches:
Spectral unmixing techniques:
Acquire spectral profiles of FITC and known autofluorescent components separately
Use computational algorithms to mathematically separate overlapping signals based on their spectral signatures
This approach is particularly valuable for tissues with lipofuscin or collagen autofluorescence
Alternative blocking protocols:
For tissues with extensive autofluorescence, treat with 0.1% Sudan Black B in 70% ethanol for 20 minutes after antibody incubation
Copper sulfate treatment (10mM CuSO₄ in 50mM ammonium acetate buffer, pH 5.0) effectively quenches autofluorescence while preserving FITC signal
Signal amplification strategies:
Tyramide signal amplification can enhance specific FITC signal 10-100 fold above background
Biotin-streptavidin systems coupled with FITC provide another amplification approach for low-abundance DROSHA detection
Note that FITC-avidin conjugates have been utilized for specialized immunofluorescence assays and can provide highly targeted detection
Imaging and analytical approaches:
Time-gated detection exploits the longer fluorescence lifetime of FITC compared to autofluorescence
Photobleaching analysis can differentiate between FITC (which bleaches at a characteristic rate) and autofluorescence
Implementing a 488nm narrow bandpass excitation filter with a 520-540nm emission filter maximizes FITC signal while minimizing autofluorescence collection
These approaches collectively enhance signal specificity when detecting DROSHA in tissues with challenging autofluorescence profiles.
Hypoxia significantly alters DROSHA detection through several mechanisms that researchers must account for in experimental design:
Transcriptional repression mechanisms:
Hypoxia induces HIF1α-dependent downregulation of DROSHA through transcriptional repression
ETS1 and ELK1 transcription factors bind to the DROSHA promoter under hypoxic conditions, recruiting repressive HDAC1 and ARID4B complexes
This mechanism leads to epigenetic silencing including increased DNA methylation at CpG islands near the DROSHA promoter
Detection optimization strategies:
Under hypoxic conditions, standard antibody dilutions (1:50-1:500) may yield diminished signals due to reduced DROSHA expression
Researchers should use antibody concentrations at the higher end of recommended ranges (e.g., 1:50 for ICC) when examining hypoxic samples
Signal amplification methods become essential for detecting reduced DROSHA levels in hypoxia
Experimental design considerations:
Include parallel normoxic controls processed with identical antibody concentrations
Establish time course experiments to determine the kinetics of DROSHA downregulation (significant changes typically observed after 24 hours of hypoxia)
Use positive controls targeting proteins known to be upregulated in hypoxia (e.g., CA9, VEGF) to confirm hypoxic response
Validation approaches:
Complement protein detection with mRNA quantification to confirm transcriptional repression
Consider ChIP analysis of the DROSHA promoter to verify ETS1/ELK1 binding and epigenetic modifications
Rescue experiments with siRNAs against ETS1 and ELK1 can restore DROSHA levels and confirm detection specificity
These methodological adaptations are essential when studying DROSHA in hypoxic environments, particularly in cancer research where hypoxia-mediated DROSHA downregulation promotes tumor progression .
Quantitative analysis of DROSHA using FITC-conjugated antibodies requires rigorous methodological approaches:
Image acquisition parameters:
Establish standardized exposure settings using calibration standards
Capture images below pixel saturation to ensure linear relationship between fluorescence and protein quantity
Include reference samples in each imaging session to permit normalization across experiments
Subcellular quantification approaches:
Nuclear-cytoplasmic ratio analysis for DROSHA requires precise nuclear segmentation using DNA counterstains
For chromatin-associated DROSHA, implement co-localization analysis with DNA repair factors (e.g., 53BP1, γH2AX) using Pearson's or Manders' coefficients
Research shows DROSHA drives DNA:RNA hybrid formation around DNA break sites, requiring specialized quantification approaches
Fluorescence intensity quantification strategies:
Standardization and normalization:
Include fluorescent beads of known intensity in each experiment for calibration
Normalize DROSHA signal to nuclear area or total protein content measured by complementary methods
For comparative studies, use relative rather than absolute values and include appropriate statistical analysis
Advanced analytical approaches:
Fluorescence correlation spectroscopy (FCS) for molecular dynamics of DROSHA complexes
Fluorescence recovery after photobleaching (FRAP) to assess DROSHA mobility at chromatin sites
Automated high-content imaging systems for population-level quantification across thousands of cells
These quantitative approaches enable rigorous analysis of DROSHA localization and expression patterns in diverse experimental contexts.
Multi-color imaging with DROSHA antibody-FITC requires careful technical consideration:
Fluorophore selection strategy:
FITC (excitation ~495nm, emission ~519nm) pairs effectively with red fluorophores like Texas Red or Cy3 for nuclear co-factors
For triple labeling, combine with far-red fluorophores (Cy5, Alexa Fluor 647) and blue/UV dyes for nuclear counterstaining
Consider using BODIPY FL-conjugated antibodies instead of FITC when working with highly charged protein targets to reduce nonspecific binding
Sequential staining protocol:
For multiple primary antibodies from the same species (e.g., rabbit anti-DROSHA and rabbit anti-Dicer):
Apply first primary antibody at lower concentration
Detect with fluorophore-conjugated Fab fragments
Block with excess unconjugated Fab fragments
Apply second primary antibody
Detect with differently labeled secondary antibody
Co-localization validation techniques:
Technical controls: single-labeled samples to establish bleed-through parameters
Biological controls: co-staining for known DROSHA interactors like DGCR8 (positive control) and cytoplasmic markers (negative control)
Research shows DROSHA interacts with DNA damage factors, making co-localization with 53BP1 and γH2AX valuable for functional studies
Image acquisition considerations:
Use sequential scanning rather than simultaneous acquisition to minimize crosstalk
Match pinhole sizes across channels for confocal microscopy
Employ chromatic aberration correction using multi-color beads
Advanced co-localization analysis:
Implement pixel intensity correlation analysis using Pearson's or Manders' coefficients
Use object-based co-localization when studying discrete nuclear structures
Distance-based analysis can quantify spatial relationships between DROSHA and other nuclear factors
These methodological considerations ensure accurate multi-color imaging for studying DROSHA's interactions with chromatin factors and other nuclear proteins.
Chromatin immunoprecipitation (ChIP) with DROSHA antibody-FITC conjugates requires specialized methodology:
Antibody selection criteria:
For ChIP applications, select antibodies validated specifically for this purpose, such as the D30F3 Rabbit mAb which has confirmed ChIP reactivity with human and mouse samples
The recommended dilution for ChIP with this antibody is 1:25, significantly more concentrated than for Western blotting (1:1000)
For optimal results, use 20 μl of antibody and 10 μg of chromatin (approximately 4 × 10^6 cells) per IP
FITC modification considerations:
FITC conjugation may interfere with antigen recognition in the chromatin context
Test both conjugated and unconjugated versions of the same antibody clone
If using FITC-conjugated antibodies directly, implement additional controls to ensure epitope recognition remains intact
ChIP protocol modifications:
For DROSHA ChIP, sonication conditions should be optimized to generate chromatin fragments of 200-500 bp
Extended incubation times (overnight at 4°C) improve recovery of DROSHA-associated chromatin
Research demonstrates DROSHA binds to chromatin at DNA break sites, requiring specific experimental design to capture these transient interactions
Validation approaches:
Perform sequential ChIP (ChIP-reChIP) to confirm co-occupancy with known DROSHA-associated factors
Include input DNA, IgG controls, and positive controls (antibodies against histone marks or transcription factors)
For studying DNA:RNA hybrids formed by DROSHA around DNA break sites, combine with DRIP (DNA:RNA Immunoprecipitation) techniques
Data analysis considerations:
Define appropriate positive and negative genomic regions for qPCR validation
For ChIP-seq applications, use specialized peak calling algorithms appropriate for transcription factors
Compare DROSHA binding profiles with known co-factors (DGCR8) and DNA damage markers (γH2AX)
These methodological considerations ensure successful ChIP experiments when studying DROSHA's chromatin associations and DNA damage response functions.
Rigorous validation of DROSHA antibodies requires comprehensive methodological approaches:
siRNA knockdown validation protocol:
Implement multiple siRNAs targeting different regions of DROSHA mRNA
Confirm knockdown efficiency at both mRNA (qRT-PCR) and protein (western blot) levels
Research has demonstrated successful DROSHA knockdown using siRNA approaches in multiple cell types including A549, U2OS, and HeLa cells
For immunofluorescence validation, include quantification of signal reduction following knockdown
Rescue experiment methodology:
Express siRNA-resistant DROSHA constructs in knockdown cells
Confirm restoration of antibody signal with wild-type DROSHA expression
Research demonstrates successful rescue experiments using siRNA-resistant over-expression plasmids
Include mutant DROSHA variants (e.g., catalytically inactive) to distinguish between structural epitope recognition and functional readouts
CRISPR/Cas9 knockout validation:
Generate complete DROSHA knockout cell lines where feasible
For essential genes, implement conditional or inducible knockout systems
Compare antibody signals between wild-type and knockout samples across multiple techniques (WB, IF, ChIP)
Antibody cross-reactivity assessment:
Test antibody against related RNase III family members (Dicer)
Evaluate species specificity using cells from different organisms
Current DROSHA antibodies show confirmed reactivity with human samples, with some also validated for mouse reactivity
These validation approaches ensure accurate interpretation of experimental results and should be implemented before conducting extensive studies with DROSHA antibodies.
Systematic troubleshooting of weak DROSHA-FITC signals requires methodical investigation:
Epitope masking and retrieval strategies:
Implement heat-induced epitope retrieval (10mM citrate buffer, pH 6.0, 95°C for 20 minutes)
Test multiple fixation protocols (4% PFA, methanol, or combination fixation)
Optimize permeabilization (varying Triton X-100 concentrations from 0.1-0.5%)
Extended antibody incubation (overnight at 4°C) may improve signal detection
Signal amplification methods comparison:
| Amplification Method | Signal Enhancement | Implementation Complexity |
|---|---|---|
| Tyramide Signal Amplification | 10-100× | Moderate |
| Biotin-Streptavidin System | 3-8× | Low |
| Multiple secondary antibodies | 2-3× | Very low |
| Antibody-DNA conjugates | 10-1000× | High |
Antibody selection considerations:
Test multiple DROSHA antibodies targeting different epitopes
Research confirms differential reactivity between antibodies - validated options include ab12286 (ICC, WB) , 27958-1-AP (WB, IF/ICC) , and D30F3 Rabbit mAb (WB, ChIP)
Consider antibody format (polyclonal vs. monoclonal) and host species
Protocol optimization for specific cell types:
Adjust blocking reagents based on cell type (2% human IgG superior for cells with charged proteins)
Test neutral fluorophores like BODIPY FL for challenging samples where FITC causes high background
Modify antibody concentration based on DROSHA expression levels (hypoxic conditions may require higher concentrations)
Biological considerations:
Verify DROSHA expression in your specific cell type or tissue
Consider developmental, stress-induced, or disease-related changes in DROSHA expression
Research shows hypoxia significantly downregulates DROSHA expression, potentially resulting in weak signals
Implementing these systematic troubleshooting approaches will help resolve weak or absent DROSHA-FITC signals in challenging experimental contexts.