DROSHA Antibody,Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism

The DROSHA Antibody, Biotin conjugated refers to a monoclonal or polyclonal antibody specific to the DROSHA protein (Ribonuclease Type III, Nuclear), chemically linked to biotin via covalent bonding. This conjugation enables the antibody to interact with biotin-binding proteins such as streptavidin or avidin, facilitating downstream detection in assays like Western blot, ELISA, or immunohistochemistry. While commercial biotin-conjugated DROSHA antibodies are not explicitly listed in current catalogs , the conjugation process can be performed in-house using biotinylation kits .

Structure and Function of DROSHA

  • Molecular Weight:

    • Predicted: 159 kDa (genomic calculation) .

    • Observed: 90–151 kDa (varies due to post-translational modifications) .

  • Role:
    DROSHA is a dsRNA-specific endoribonuclease that cleaves primary miRNA (pri-miRNA) transcripts in the nucleus, initiating miRNA biogenesis . Its activity is critical for regulating gene expression at the post-transcriptional level.

Applications of Biotin-Conjugated DROSHA Antibody

ApplicationMethodologyDilution (Unconjugated)Detection System
Western Blot (WB)Detects DROSHA in cell lysates1:500–1:16,000 Streptavidin-HRP/chemiluminescence
Immunofluorescence (IF)Visualizes nuclear DROSHA localization1:50–1:500 Fluorescent streptavidin (e.g., Alexa Fluor 488)
ELISAQuantifies DROSHA in solution1:1,000–1:5,000 Streptavidin-alkaline phosphatase

References

  1. Conjugation methods for biotin-antibody linkages .

  2. Proteintech DROSHA Antibody (55001-1-AP) specifications .

  3. Antibodies-Online DROSHA Antibody (ABIN3028624) details .

  4. Proteintech DROSHA Antibody (27958-1-AP) data .

  5. Antibodies-Online DROSHA Antibody catalog .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary based on the order fulfillment method and location. Please consult your local distributors for specific delivery details.
Synonyms
DROSHA antibody; Drosha double stranded RNA specific endoribonuclease antibody; Drosha ribonuclease type III antibody; Etohi2 antibody; HSA242976 antibody; Nuclear RNase III Drosha antibody; p241 antibody; Protein Drosha antibody; Putative protein p241 which interacts with transcription factor Sp1 antibody; Putative ribonuclease III antibody; RANSE3L antibody; Ribonuclease 3 antibody; Ribonuclease III antibody; Ribonuclease III nuclear antibody; Ribonuclease type III nuclear antibody; RibonucleaseIII antibody; RN 3 antibody; RN3 antibody; RNase 3 antibody; RNase III antibody; RNase3 antibody; RNASE3L antibody; RNaseIII antibody; RNASEN antibody; RNC_HUMAN antibody
Target Names
DROSHA
Uniprot No.

Target Background

Function
DROSHA is a ribonuclease III double-stranded (ds) RNA-specific endoribonuclease that plays a crucial role in the initial step of microRNA (miRNA) biogenesis. It is a component of the microprocessor complex, essential for processing primary miRNA transcripts (pri-miRNAs) to generate precursor miRNA (pre-miRNA) in the nucleus. Within the microprocessor complex, DROSHA cleaves the 3' and 5' strands of a stem-loop in pri-miRNAs (processing center 11 bp from the dsRNA-ssRNA junction) to release hairpin-shaped pre-miRNAs. These pre-miRNAs are subsequently processed by the cytoplasmic DICER to produce mature miRNAs. DROSHA is also involved in pre-rRNA processing. It specifically cleaves double-strand RNA and does not cleave single-strand RNA. Additionally, DROSHA participates in the formation of GW bodies.
Gene References Into Functions
  1. Analysis of recurrent homozygous deletion of DROSHA and microduplication of PDE4DIP in pineoblastoma. PMID: 30030436
  2. This study sheds light on the function of miR-128-3p as a key regulator of the malignant phenotype of lung cancer cells, emphasizing the role of Drosha in non-small-cell lung cancer cells migration. PMID: 29236960
  3. Genotype frequencies of DROSHA (rs10719, rs642321, and rs2291102) were determined by sequencing in 385 infertile men and 120 fertile controls. No significant differences were observed in DROSHA expression between cases and controls. PMID: 29892896
  4. The Drosha rs10719TC and CC genotypes were associated with preeclampsia (PE) risk. The combined CC-GG genotype and C-G haplotype of Drosha rs10719 and rs6877842 polymorphisms may increase PE susceptibility. PMID: 29157048
  5. While DGCR8 is known to bind heme, the molecular role of heme in pri-miR processing is unclear. This research reveals that heme is crucial for the Microprocessor complex to process pri-miRs with high fidelity. PMID: 29170488
  6. These findings provide potential evidence that rs10719 and rs493760 might contribute to the risk of cleft lip/palate (CL/P) and suggest potential genetic basis and mechanisms of CL/P. PMID: 28833944
  7. It has been reported that the gene encoding human DROSHA also encodes a potential miRNA, which may act upon, at least, one of DROSHA transcripts. PMID: 28665784
  8. Depletion of drosha ribonuclease III (Drosha) significantly reduces DNA repair through both homologous recombination (HR) and non-homologous end joining (NHEJ). PMID: 29416038
  9. Increased Drosha expression was found in chronic lymphocytic leukemia patients without chromosomal deletions. PMID: 28388279
  10. Point mutations in the RNaseIIIb domain of Drosha implicated in Wilms tumors differentially affected cleavage of the 5' and 3' strands of pri-miRNAs in vitro. PMID: 29109067
  11. Overexpression of LAMC2 and knockdown of CD82 significantly promoted gastric cancer (GC) cell invasion and activated EGFR/ERK1/2-MMP7 signaling via upregulation of the expression of phosphorylated (p)-EGFR, p-ERK1/2, and MMP7. PMID: 28252644
  12. A significant association was observed between two candidate genes and Alzheimer's disease (AD), TARBP2 rs784567 genotype and AD (chi=6.292, P=0.043), and a trend for RNASEN rs10719 genotype (chi=4.528, P=0.104) and allele (P=0.035). After controlling for age, we found that the TARBP2-RNASEN association with AD, the age variation was a risk factor for AD risk (P<0.001; OR=1.104; 95% CI, 1.059-1.151) PMID: 26796812
  13. BRG1 and SMARCAL1, members of the ATP-dependent chromatin remodelling family, are shown to co-regulate the transcription of DROSHA, DGCR8, and DICER in response to double-strand DNA breaks. PMID: 28716689
  14. Mechanistic dissection reveals that NEAT1 broadly interacts with the NONO-PSF heterodimer as well as many other RNA-binding proteins and that multiple RNA segments in NEAT1, including a 'pseudo pri-miRNA' near its 3' end, help attract the Drosha-DGCR8 Microprocessor. PMID: 28846091
  15. Results show that Mammalian DROSHA genes have evolved a conserved hairpin structure spanning a specific exon-intron junction serving as a substrate for the microprocessor in human but not in murine cells. This hairpin element determines whether the overlapping exon is alternatively or constitutively spliced. Furthermore, DROSHA promotes skipping of the overlapping exon in human cells independently of its cleavage function. PMID: 28400409
  16. This research reports DROSHA's numerous processing sites on primary microRNAs and noncanonical substrates, which may serve as cis-elements for DROSHA-mediated gene regulation. PMID: 28431232
  17. Knockdown of Drosha in MGC-803 cells increased the apoptosis rate, significantly upregulated the protein expressions of caspase-3, caspase-9, and Bax, and downregulated Bcl-2. PMID: 27609577
  18. The rs417309 and rs1640299 polymorphisms of the DGCR8 gene, as well as rs6877842 of the DROSHA gene, might be associated with laryngeal cancer occurrence in the Polish population. PMID: 28155978
  19. MiR-27b mimics, DROSHA siRNA, and miR-27b inhibitors were used to verify the negative regulatory relationship between MiR-27b and DROSHA. The presence of rs10719 disrupted the interaction between miR-27b and DROSHA, which might be the underlying mechanism of the observation that rs10719 is significantly associated with the risk of primary hypertension. PMID: 28214904
  20. Drosha and DGRC8 were significantly downregulated in healthy-appearing perilesional skin from hidradenitis suppurativa patients compared to healthy controls. PMID: 26917346
  21. Mutations of the DROSHA gene underlie Wilms tumor recurrences. PMID: 26802027
  22. An essential role of DROSHA in the canonical miRNA pathway. PMID: 26976605
  23. Gradual loss of cytoplasmic Drosha was accompanied by tumor progression in both gastric cancer tissues and cell lines, and was inversely associated with tumor volume (P = 0.002), tumor grade (P < 0.001), tumor stage (P = 0.018), and distant metastasis. PMID: 26694172
  24. DGCR8 and Drosha assemble into a heterotrimeric complex on RNA, comprising two DGCR8 molecules and one Drosha molecule. PMID: 26683315
  25. This study reports the X-ray structure of DROSHA in complex with the C-terminal helix of DGCR8; DROSHA contains two DGCR8-binding sites, one on each RNase III domain (RIIID), which mediate the assembly of Microprocessor. The overall structure of DROSHA is surprisingly similar to that of Dicer despite no sequence homology apart from the C-terminal part. PMID: 26748718
  26. Variations in DROSHA rs10719 in Korean patients are significantly associated with their risk of colorectal cancer. PMID: 26147304
  27. Drosha expression was gradually reduced with the degrading histological differentiation of gastric adenocarcinoma, and the knock-down of Drosha expression could promote gastric adenocarcinoma cell invasion. PMID: 26522361
  28. Data underscore the pivotal role of the miRNA biogenesis pathway in Wilms tumor (WT) tumorigenesis, particularly the major miRNA-processing gene DROSHA. PMID: 24909261
  29. Drosha is upregulated in gestational diabetes. PMID: 25295740
  30. Together with a 23-amino acid peptide from DGCR8, DROSHA constitutes a minimal functional core. DROSHA serves as a "ruler" by measuring 11 bp from the basal ssRNA-dsRNA junction. DGCR8 interacts with the stem and apical elements through its dsRNA-binding domains and RNA-binding heme domain, respectively, allowing efficient and accurate processing. PMID: 26027739
  31. DROSHA RNase IIIB mutations globally inhibit miRNA biogenesis through a dominant-negative mechanism in Wilms tumors. PMID: 25190313
  32. Drosha protein was identified as a new component of dipeptide-repeat aggregates in frontotemporal lobar degeneration and tauopathy. PMID: 25756586
  33. p38 MAPK directly phosphorylates Drosha at its N terminus promoting its nuclear export and degradation. PMID: 25699712
  34. To inhibit the expression of Drosha. PMID: 25058539
  35. Low Drosha expression is associated with invasive breast carcinoma. PMID: 24574065
  36. DROSHA rs10719T>C polymorphism may be associated with bladder cancer risk in a Chinese population, and hsa-miR-27b can influence the expression of DROSHA protein by binding with 3'UTR. PMID: 24312312
  37. These results demonstrate that Drosha can function like a splicing enhancer and promote exon inclusion. This research unveils a new mechanism of alternative splicing regulation involving a cleavage-independent role for Drosha in splicing. PMID: 24786770
  38. The involvement of E2F1-dependent DROSHA activation in pri-miRNA processing under DNA damage stress provides further insight into the regulation of miRNA biosynthesis. PMID: 24909689
  39. Drosha regulates nascent gene transcription through interaction with CBP80 and RNA PolII. PMID: 24360955
  40. The pri-miRNA stem, defined by internal and flanking structural elements, guides the binding position of Drosha-DGCR8, which consequently determines the cleavage site. PMID: 24854622
  41. The Microprocessor complex of Drosha and DGCR8 proteins, responsible for processing the primary transcripts during miRNA generation, destabilizes the mRNA of Aurora kinase B. PMID: 24589731
  42. Changes in Drosha expression can be a biologically relevant mechanism by which eukaryotic cells control miRNA profiles. PMID: 24677349
  43. Acetylation of Drosha on those N-terminal lysine sites prevents Drosha ubiquitination, thereby preventing its degradation. PMID: 24009686
  44. These results suggest that Drosha affects the biological activity of cervical cancer cells by regulating the expression of numerous tumor-associated proteins. PMID: 23969986
  45. Results indicate a block of miRNA maturation at the DROSHA processing step. PMID: 23974981
  46. If the distances are not optimal, Drosha tends to cleave at multiple sites, which can, in turn, generate multiple 5' isomiRs. PMID: 24297910
  47. Our results demonstrate a reduced nuclear expression of DROSHA in melanoma. PMID: 23370771
  48. The RNase III enzyme Drosha and the double-stranded RNA-binding protein DGCR8 bind and regulate a large variety of cellular RNAs. PMID: 23863141
  49. Drosha protein potentially plays an important role in breast cancer progression. PMID: 23225145
  50. miRNA regulatory effect is a heritable trait in humans; a polymorphism of the DROSHA gene contributes to the observed inter-individual differences. PMID: 23272639

Show More

Hide All

Database Links

HGNC: 17904

OMIM: 608828

KEGG: hsa:29102

STRING: 9606.ENSP00000339845

UniGene: Hs.97997

Protein Families
Ribonuclease III family
Subcellular Location
Nucleus. Nucleus, nucleolus.
Tissue Specificity
Ubiquitous.

Q&A

What is DROSHA and why is it an important target for antibody-based detection?

DROSHA is a Class 2 ribonuclease III enzyme encoded by the DROSHA gene (formerly RNASEN). It functions as a key subunit of the microprocessor protein complex that catalyzes the initial processing step of microRNA (miRNA) synthesis. DROSHA cleaves the stem-loop structure from primary microRNA (pri-miRNA) in the nucleus, yielding precursor miRNA (pre-miRNA), which is then exported to the cytoplasm for further processing .

The importance of DROSHA in miRNA biogenesis makes it a critical target for research into gene regulation pathways. Recent research has also revealed DROSHA's role in DNA damage response and repair mechanisms, particularly in the recruitment of PRC1 component BMI1 at double-strand breaks (DSBs) and subsequent H2A-K119 ubiquitination . This expanded understanding of DROSHA's functions beyond miRNA processing has increased interest in reliable antibody-based detection methods.

What are the fundamental differences between unconjugated and biotin-conjugated DROSHA antibodies?

Unconjugated DROSHA antibodies require secondary detection reagents (such as labeled secondary antibodies) for visualization, while biotin-conjugated DROSHA antibodies have biotin molecules directly attached to the antibody, enabling detection through biotin-binding proteins like streptavidin or avidin .

The key differences include:

CharacteristicUnconjugated DROSHA AntibodiesBiotin-Conjugated DROSHA Antibodies
Detection systemRequires secondary antibodiesUses streptavidin/avidin conjugates
Signal amplificationLimited to secondary antibody bindingEnhanced through streptavidin-biotin high affinity (KD=1.3×10^-15 M)
Workflow complexityMore steps (primary + secondary incubations)Fewer steps (primary + streptavidin reagent)
FlexibilityCan use different secondary antibodiesLimited to biotin-binding detection systems
Background concernsPotential cross-reactivity from secondary antibodiesPotential endogenous biotin interference

The choice between these formats depends on the specific experimental requirements, including sensitivity needs, sample type, and detection method .

What are standard applications for biotin-conjugated DROSHA antibodies?

Biotin-conjugated DROSHA antibodies can be utilized in multiple research applications:

  • Western Blotting (WB): Typically used at dilutions of 1:2000-1:16000, providing high sensitivity when coupled with enzyme-conjugated streptavidin detection systems .

  • Immunofluorescence (IF)/Immunocytochemistry (ICC): Recommended dilutions range from 1:50-1:500, allowing for visualization of DROSHA's nuclear localization .

  • Flow Cytometry: Used at dilutions of approximately 1:200-1:1000, enabling quantitative analysis of DROSHA expression in cell populations .

  • ELISA: Effective at dilutions of 1:20,000-1:400,000 when using enzyme-conjugated streptavidin, providing exceptional sensitivity for quantitative detection .

  • Proximity Ligation Assay (PLA): As demonstrated in DNA damage response studies, biotinylated antibodies allow detection of protein-protein interactions involving DROSHA at DNA repair sites .

For optimal results across these applications, researchers should perform antibody titration experiments to determine the ideal concentration for their specific experimental conditions .

How should I properly biotinylate a DROSHA antibody while maintaining its specificity and affinity?

Proper biotinylation of DROSHA antibodies requires careful consideration of several methodological factors:

Method selection based on research needs:

  • Random Conjugation: Uses the ε-amino group of lysine residues or the thiol group of cysteine residues. While simpler, this produces heterogeneous conjugates with variable biotin incorporation .

  • Site-Specific Conjugation: More controlled approaches that target specific sites on the antibody:

    • Selenocysteine interface technology introduces unique nucleophilic reactivity at defined positions

    • Enzymatic approaches using transglutaminases or sortases

    • Chemical approaches targeting carbohydrate moieties

Recommended protocol for random biotinylation:

  • Dilute purified DROSHA antibody to 4 μM in 100 mM sodium acetate (pH 5.2)

  • Add DTT at 0.1 mM followed by biotin-iodoacetamide

  • Incubate for 1 hour at room temperature

  • Purify using desalting columns or dialysis against PBS

  • Validate both biotin incorporation and retained antibody activity

Modern biotinylation kits offer streamlined workflows that can be completed in as little as 10 minutes with high reproducibility, making them suitable for most research applications. These kits typically include all necessary reagents and offer consistent biotin incorporation without requiring purification steps .

What controls should be included when using biotin-conjugated DROSHA antibodies?

Proper experimental design requires several controls to ensure reliable results with biotin-conjugated DROSHA antibodies:

Essential controls:

  • Isotype Control: Include an isotype-matched biotinylated control antibody (same species and isotype as your DROSHA antibody) to assess non-specific binding .

  • Knockout/Knockdown Validation: Use DROSHA knockout cell lines (such as DROSHA knockout HEK-293T cell line) or DROSHA siRNA knockdown samples to confirm antibody specificity .

  • Secondary Reagent Control: Include samples treated only with the streptavidin detection reagent (without primary antibody) to assess background from the detection system .

  • Blocking Control: For tissues or cells with endogenous biotin, include samples pre-blocked with avidin/biotin blocking reagents .

  • Competitive Binding Control: Pre-incubate the antibody with recombinant DROSHA protein before application to demonstrate specificity .

Example validation data from research:
When validating a biotin-conjugated DROSHA antibody by Western blot, researchers observed a 159 kDa band in wild-type HEK-293T cells that was absent in DROSHA knockout HEK-293T cells, confirming specificity. Similar validation in immunofluorescence showed nuclear localization consistent with DROSHA's known cellular distribution .

How do I troubleshoot high background issues when using biotin-conjugated DROSHA antibodies?

High background is a common challenge when using biotin-conjugated antibodies. Here's a systematic approach to troubleshooting:

Causes and solutions for high background:

  • Endogenous Biotin in Samples:

    • Solution: Implement an avidin/biotin blocking step before applying the primary antibody

    • Method: Incubate samples with unconjugated avidin followed by excess biotin to block endogenous biotin

  • Excessive Antibody Concentration:

    • Solution: Perform titration experiments to determine optimal antibody concentration

    • Method: Test serial dilutions (e.g., 1:100, 1:500, 1:1000, 1:5000) to identify the concentration that gives specific signal with minimal background

  • Non-specific Binding:

    • Solution: Optimize blocking and wash conditions

    • Method: Increase blocking time/concentration (typically 3-5% BSA or 5-10% normal serum), add 0.1-0.3% Triton X-100 to washes, and increase wash duration/number

  • Over-biotinylation of Antibody:

    • Solution: Use antibodies with optimal biotin:antibody ratio or reduce biotinylation time

    • Method: Commercial antibodies typically have optimized ratios; for custom biotinylation, reduce reaction time or reagent concentration

  • Cross-reactivity:

    • Solution: Pre-adsorb the antibody or use more specific antibody clones

    • Method: Pre-incubate antibody with lysates from knockout cells or recombinant proteins of related family members

How can biotin-conjugated DROSHA antibodies be used to study DROSHA's role in DNA damage response?

Recent research has revealed DROSHA's critical involvement in DNA damage response through mechanisms distinct from its role in miRNA processing. Biotin-conjugated DROSHA antibodies offer powerful tools for investigating these processes:

Methodological approach:

  • Proximity Ligation Assay (PLA):

    • Combine anti-biotin antibody with anti-BMI1 antibody to detect DROSHA-BMI1 interactions at DNA damage sites

    • This approach revealed that DROSHA and DICER control the recruitment of BMI1 at double-strand breaks (DSBs)

  • Chromatin Immunoprecipitation (ChIP):

    • Use biotin-conjugated DROSHA antibodies to immunoprecipitate DROSHA-bound chromatin

    • Couple with sequencing (ChIP-seq) to map DROSHA binding sites at DNA damage loci

  • RNA-Immunoprecipitation (RIP):

    • Apply biotin-conjugated DROSHA antibodies to immunoprecipitate DROSHA-bound RNAs

    • This approach showed that BMI1 associates with damage-induced long non-coding RNAs (dilncRNAs) in a DROSHA-dependent manner

Key research findings:
Studies using these approaches demonstrated that DROSHA and DICER control transcriptional silencing of genes adjacent to damaged chromatin through a mechanism involving BMI1 recruitment and H2A-K119 ubiquitination . This function appears to be independent of DROSHA's canonical miRNA processing role and provides new insights into DNA damage-induced transcriptional regulation.

What are the advantages and limitations of using F(ab')2 fragments versus whole IgG for biotin-conjugated DROSHA antibodies?

The choice between F(ab')2 fragments and whole IgG for biotin-conjugated DROSHA antibodies significantly impacts experimental outcomes:

Comparative analysis:

CharacteristicBiotin-F(ab')2 DROSHA AntibodiesBiotin-Whole IgG DROSHA Antibodies
Size~110 kDa~150 kDa
Tissue penetrationSuperior due to smaller sizeMore limited
Fc receptor bindingEliminated (no Fc region)Present (potential non-specific binding)
Complement activationEliminatedPresent (potential unwanted effects)
StabilityModerately stableHighly stable
Half-lifeShorterLonger
Signal amplificationReduced (fewer biotin molecules per antibody)Enhanced (more biotin attachment sites)

Application-specific considerations:

  • Live Cell Applications: F(ab')2 fragments are preferred because they avoid binding to Fc receptors on live cells, reducing non-specific signal .

  • Tissue Immunohistochemistry: F(ab')2 fragments offer better penetration in dense tissues but may provide lower sensitivity than whole IgG .

  • Proximity-based Assays: F(ab')2 fragments place detection tags closer to the target epitope, potentially improving spatial resolution in techniques like PLA .

  • Storage Stability: Whole IgG typically exhibits superior shelf-life, with biotin-conjugated F(ab')2 fragments requiring more careful storage conditions (recommended: aliquot and freeze at -70°C or add equal volume of glycerol for storage at -20°C) .

How does MDM2-mediated ubiquitination of DROSHA affect epitope accessibility for antibody detection?

MDM2 has been identified as an E3 ligase for DROSHA, mediating its ubiquitination and affecting its function in response to cellular environmental changes. This post-translational modification can impact antibody-based detection:

Mechanistic impact on epitope accessibility:

  • Ubiquitination Sites and Epitope Masking:

    • MDM2-mediated ubiquitination of DROSHA can potentially mask epitopes, particularly in regions where ubiquitin chains are attached

    • In vitro ubiquitination assays have demonstrated that MDM2 has E3 ligase activity for DROSHA, resulting in specific ubiquitin attachment patterns

  • Conformational Changes:

    • Ubiquitination may induce conformational changes in DROSHA protein structure

    • These changes can expose or conceal epitopes, affecting antibody binding efficiency

Experimental strategies to address this challenge:

  • Epitope Mapping:

    • Map antibody epitopes relative to known ubiquitination sites on DROSHA

    • Select antibodies targeting regions less likely to be affected by ubiquitination

  • Deubiquitination Treatment:

    • Include deubiquitinating enzyme treatment conditions in parallel experiments

    • Compare antibody detection between ubiquitinated and deubiquitinated samples

  • Multiple Antibody Approach:

    • Use multiple antibodies targeting different DROSHA epitopes

    • This provides complementary data to overcome potential epitope masking issues

Research has shown that silencing MDM2 greatly reduces ubiquitinated DROSHA levels, suggesting that ubiquitination status monitoring may be important when studying DROSHA in different cellular contexts .

How do biotin-conjugated DROSHA antibodies compare to other detection methods in sensitivity and specificity?

When selecting detection methods for DROSHA research, it's important to understand the comparative advantages of different approaches:

Sensitivity comparison across detection methods:

Detection MethodSensitivity RankSignal-to-Noise RatioQuantitative CapacityMultiplex Capability
Biotin-streptavidinHighExcellentGoodLimited
Direct fluorophore conjugationModerateGoodExcellentExcellent
Enzyme conjugation (HRP/AP)Very HighVariableLimitedVery Limited
Unconjugated + fluorescent secondaryHighVery GoodGoodGood

Method-specific considerations:

  • Biotin-Streptavidin Systems:

    • Advantage: Signal amplification through multiple biotin-streptavidin interactions increases sensitivity

    • Limitation: Potential endogenous biotin interference in certain sample types

    • Optimal for: Western blots (1:2000-1:16000 dilution) and ELISA (1:20,000-1:400,000 dilution)

  • Direct Fluorophore Conjugation:

    • Advantage: Simplified workflow and excellent for multiplexing

    • Limitation: Limited signal amplification compared to biotin-streptavidin

    • Optimal for: Immunofluorescence microscopy and flow cytometry

  • Enzyme-based Detection:

    • Advantage: Extreme sensitivity through enzymatic amplification

    • Limitation: Limited dynamic range and poor spatial resolution

    • Optimal for: Western blots and immunohistochemistry

Research data indicates that biotin-conjugated antibodies with streptavidin detection systems can offer up to 4-8 fold signal enhancement compared to direct conjugation methods, particularly when using enzymes like alkaline phosphatase with streptavidin .

What are the considerations for choosing between polyclonal and monoclonal biotin-conjugated DROSHA antibodies?

The choice between polyclonal and monoclonal biotin-conjugated DROSHA antibodies significantly impacts experimental outcomes:

Comparative analysis:

CharacteristicPolyclonal Biotin-ConjugatedMonoclonal Biotin-Conjugated
Epitope recognitionMultiple epitopesSingle epitope
Batch-to-batch variabilityHigherLower
SensitivityGenerally higherGenerally lower but more specific
Specificity concernsHigher risk of cross-reactivityLower risk of cross-reactivity
Application versatilityOften works across multiple applicationsMay be optimized for specific applications
CostTypically lowerTypically higher

Application-specific recommendations:

  • For Western Blotting:

    • Polyclonal antibodies often provide stronger signals (recommended dilutions: 1:2000-1:16000)

    • Example: Proteintech's polyclonal anti-DROSHA (27958-1-AP) shows excellent specificity at 150 kDa in multiple human cell lines

  • For Immunoprecipitation:

    • Monoclonal antibodies typically offer cleaner results with less background

    • Example: Abcam's EPR12794 monoclonal antibody (ab183732) demonstrates strong specificity in IP applications

  • For Immunofluorescence:

    • Both types can work well; selection depends on specific epitope accessibility

    • U2OS cells have been successfully used to validate DROSHA antibodies in IF applications

  • For Flow Cytometry:

    • Monoclonal antibodies often provide more consistent results

    • Example flow data: Boster's A00111-3 antibody has been validated in HL-60 cells

When possible, validation in DROSHA knockout systems (such as DROSHA knockout HEK-293T cell lines) provides the strongest evidence for antibody specificity regardless of antibody type .

How can I integrate biotin-conjugated DROSHA antibodies into multi-parameter experimental designs?

Biotin-conjugated DROSHA antibodies offer versatility for integration into complex experimental designs studying multiple parameters simultaneously:

Strategic approaches for multi-parameter studies:

  • Sequential Detection Protocols:

    • Apply biotin-conjugated DROSHA antibody first, detect with streptavidin-conjugate

    • Block remaining biotin/streptavidin binding sites

    • Apply subsequent directly-conjugated antibodies for other targets

    • Example application: Studying DROSHA and DICER co-localization at DNA damage sites

  • Proximity Ligation Assay (PLA) Integration:

    • Use biotin-conjugated DROSHA antibody with antibodies against potential interaction partners

    • This approach revealed DROSHA-BMI1 interactions in DNA damage contexts

    • Protocol outline: Combine anti-biotin antibody with anti-BMI1 antibody to visualize proximity

  • Multiplexed Flow Cytometry:

    • Use streptavidin conjugated to compatible fluorophores (e.g., PE)

    • Combine with directly conjugated antibodies for other targets

    • Example: DROSHA detection alongside cell cycle markers or other RNA processing proteins

  • ChIP-seq Analysis:

    • Use biotin-conjugated DROSHA antibodies for chromatin immunoprecipitation

    • Integrate with other epigenetic marks (H2A-K119ub, γH2AX) to build comprehensive maps of DROSHA activity at chromatin

    • This approach helped establish DROSHA's role in transcriptional silencing at DNA damage sites

These integrated approaches have revealed that DROSHA works alongside DICER to control BMI1 recruitment to DNA damage sites and subsequent chromatin modifications, providing new insights into non-canonical DROSHA functions beyond miRNA processing .

How might advances in site-specific biotinylation improve DROSHA antibody applications?

Emerging technologies in site-specific biotinylation promise to enhance DROSHA antibody performance and expand application possibilities:

Innovative approaches and their potential impact:

  • Selenocysteine Interface Technology:

    • Introduces the 21st natural amino acid selenocysteine into antibodies

    • Creates unique nucleophilic reactivity for site-specific conjugation

    • Advantages over conventional methods:

      • Minor modification at the C-terminus that doesn't interfere with disulfide bridges

      • No activation requirement

      • Precise 1:1 stoichiometry of biological and chemical components

  • Enzymatic Biotinylation:

    • Utilizes enzymes like bacterial biotin ligase (BirA) for site-specific attachment

    • Requires genetic incorporation of recognition sequences

    • Offers exceptional control over biotinylation sites and stoichiometry

  • Click Chemistry Approaches:

    • Incorporates non-natural amino acids with azide or alkyne groups

    • Enables bioorthogonal conjugation with biotin derivatives

    • Provides clean, highly specific biotinylation under mild conditions

These advances could significantly improve DROSHA antibody applications by:

  • Enhancing sensitivity through optimal biotin positioning

  • Improving batch-to-batch consistency for more reproducible results

  • Preserving full antibody functionality by avoiding modification of critical regions

  • Enabling precise control of biotin:antibody ratio for optimal signal:noise

What emerging applications might benefit from biotin-conjugated DROSHA antibodies?

Several cutting-edge research areas could significantly benefit from advanced biotin-conjugated DROSHA antibodies:

Promising research directions:

  • Single-Cell Proteomics:

    • Application: Using biotin-conjugated DROSHA antibodies with mass cytometry (CyTOF)

    • Benefit: Enables comprehensive profiling of DROSHA expression and associated proteins at single-cell resolution

    • Potential insight: Identification of cell subpopulations with unique DROSHA activity patterns

  • Spatial Transcriptomics Integration:

    • Application: Combining biotin-conjugated DROSHA antibodies with in situ RNA detection

    • Benefit: Maps spatial relationships between DROSHA protein localization and miRNA/mRNA expression

    • Potential insight: Understanding tissue-specific regulation of RNA processing

  • Super-Resolution Microscopy:

    • Application: Using biotin-conjugated DROSHA antibodies with quantum dot-labeled streptavidin

    • Benefit: Achieves nanometer-scale resolution of DROSHA localization at specific nuclear substructures

    • Potential insight: Detailed understanding of DROSHA's dynamic localization during DNA damage response

  • CRISPR Screens Combined with DROSHA Detection:

    • Application: Using biotin-conjugated DROSHA antibodies to assess DROSHA function/localization in CRISPR-edited cells

    • Benefit: High-throughput assessment of genetic factors influencing DROSHA activity

    • Potential insight: Identification of novel regulators of DROSHA function in both canonical and non-canonical pathways

  • Liquid Biopsy Development:

    • Application: Using biotin-conjugated DROSHA antibodies to detect DROSHA in extracellular vesicles

    • Benefit: Potential biomarker development for cancer and other diseases

    • Potential insight: Understanding how altered DROSHA function contributes to disease pathogenesis

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.