DROSHA Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to DROSHA Antibody

DROSHA Antibody (e.g., #3364 from Cell Signaling Technology) is a monoclonal rabbit antibody designed to recognize endogenous levels of Drosha protein in human and mouse samples. Drosha, a 160 kDa nuclear RNase III enzyme, is essential for cleaving primary miRNA (pri-miRNA) transcripts into precursor miRNA (pre-miRNA) in the nucleus . The antibody is validated for Western blotting (WB) and immunoprecipitation (IP) .

Applications in Research

The antibody has been instrumental in advancing our understanding of Drosha’s roles:

  • Nuclear Localization: Drosha is predominantly nuclear, but truncation of its N-terminal domain (ΔN-Drosha) results in cytoplasmic mislocalization due to loss of nuclear localization signals (NLS) .

  • Interaction Studies: Drosha forms a complex with DGCR8 (Pasha in Drosophila), which stabilizes Drosha and is critical for pri-miRNA processing .

  • Transcriptional Regulation: Beyond miRNA processing, Drosha binds promoter-proximal regions of genes, interacts with RNA Polymerase II (Pol II), and enhances transcription .

Drosha’s Dual Roles

  • miRNA Biogenesis: Drosha processes pri-miRNAs in the nucleus, forming pre-miRNAs for export to the cytoplasm .

  • Non-Canonical Functions: Drosha regulates transcription by binding gene promoters and interacting with Pol II and CBP80, independent of RNA cleavage .

Experimental Validation

  • In HeLa cells, Drosha knockdown reduced mRNA levels of β-actin and other genes by ~75%, confirming its role in transcriptional activation .

Practical Considerations

  • Sample Preparation: Nuclear fractionation is recommended for WB/IP due to Drosha’s nuclear localization .

  • Controls: Use ΔN-Drosha mutants (cytoplasmic) to validate antibody specificity .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary based on shipping method and destination. For specific delivery times, please consult your local distributor.
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 double-stranded (ds) RNA-specific endoribonuclease that plays a critical 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) into precursor miRNA (pre-miRNA) within the nucleus. In this complex, DROSHA cleaves the 3' and 5' strands of a stem-loop structure in pri-miRNAs, specifically at a processing center located 11 bp from the dsRNA-ssRNA junction, releasing hairpin-shaped pre-miRNAs. These pre-miRNAs are further processed by the cytoplasmic DICER to generate mature miRNAs. DROSHA is also involved in pre-rRNA processing. It exhibits specificity for cleaving double-strand RNA, unlike single-strand RNA. Furthermore, DROSHA participates in the formation of GW bodies.
Gene References Into Functions
  1. This study analyzed the recurrent homozygous deletion of DROSHA and microduplication of PDE4DIP in pineoblastoma. PMID: 30030436
  2. This research provides mechanistic insights into the function of miR-128-3p as a key regulator of the malignant phenotype of lung cancer cells, particularly highlighting the role of Drosha in non-small-cell lung cancer cell migration. PMID: 29236960
  3. This study determined the genotype frequency of DROSHA (rs10719, rs642321, and rs2291102) using sequencing methods 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 genotype of CC-GG and the C-G haplotype of Drosha rs10719 and rs6877842 polymorphisms may increase PE susceptibility. PMID: 29157048
  5. Primary microRNA transcripts (pri-miRs) are cleaved by Microprocessor, a complex containing the RNase Drosha and its partner protein, DGCR8. While DGCR8 is known to bind heme, the molecular role of heme in pri-miR processing is not fully understood. This study demonstrates that heme is crucial for Microprocessor to process pri-miRs with high fidelity. PMID: 29170488
  6. These findings provide potential evidence suggesting that rs10719 and rs493760 might contribute to the risk of cleft lip/palate (CL/P), hinting at possible genetic basis and mechanisms for CL/P. PMID: 28833944
  7. It has been reported that the gene encoding human DROSHA also encodes a potential miRNA that may regulate at least one of DROSHA transcripts. PMID: 28665784
  8. Depletion of drosha ribonuclease III (Drosha) significantly reduces DNA repair by 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 the 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 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, it was found that the TARBP2-RNASEN association with AD 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 remodeling family, were found to co-regulate the transcription of DROSHA, DGCR8, and DICER in response to double-strand DNA breaks. PMID: 28716689
  14. Mechanistic dissection revealed that NEAT1 interacts broadly with the NONO-PSF heterodimer, as well as numerous other RNA-binding proteins, and that multiple RNA segments in NEAT1, including a 'pseudo pri-miRNA' near its 3' end, contribute to attracting the Drosha-DGCR8 Microprocessor. PMID: 28846091
  15. These 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. Additionally, DROSHA promotes skipping of the overlapping exon in human cells independently of its cleavage function. PMID: 28400409
  16. This report identifies numerous processing sites on primary microRNAs and noncanonical substrates that may serve as cis-elements for DROSHA-mediated gene regulation. PMID: 28431232
  17. Knockdown of Drosha increased the apoptosis rate of MGC-803 cells, significantly upregulating the protein expressions of caspase-3, caspase-9, and Bax, while downregulating 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 the risk of 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 for the observed significant association between rs10719 and 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 is established. 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 were significantly associated with their risk of colorectal cancer. PMID: 26147304
  27. Drosha expression was reduced gradually with the degrading histological differentiation of gastric adenocarcinoma, and the knock-down of Drosha expression could promote gastric adenocarcinoma cell invasion. PMID: 26522361
  28. These data highlight the pivotal role of the miRNA biogenesis pathway in Wilms 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. This study aimed 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. This research concludes that Drosha can function like a splicing enhancer and promote exon inclusion. These results reveal 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 primary transcripts during the generation of microRNAs, 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. These 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. This study demonstrates 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

Basic Research Questions

Effective sample preparation is crucial for successful DROSHA detection:

For Western Blotting:

  • Nuclear extraction is recommended as DROSHA is predominantly nuclear

  • Use of protease inhibitors is essential to prevent degradation

  • For cell lysis, RIPA buffer with phosphatase inhibitors has shown good results

  • Recommended loading: 20-40 μg of total protein per lane

For Immunoprecipitation:

  • Crosslinking with formaldehyde (1% for 10 minutes) may help preserve protein-protein interactions

  • Sonication conditions should be optimized to maintain protein integrity

  • Pre-clearing lysates with appropriate control IgG is recommended

For RNA Immunoprecipitation (RIP):

  • Tandem RNA immunoprecipitation (RIP 2) has been developed for studying DROSHA interactions with RNA

  • When studying DROSHA-RNA complexes, RNase inhibitors must be included in all buffers

Sample preparation protocols should be optimized based on the specific research question and cell/tissue type being studied.

How can researchers optimize DROSHA immunoprecipitation for studying microRNA processing complexes?

Optimizing DROSHA immunoprecipitation for microRNA processing studies requires careful consideration of several factors:

Critical Protocol Considerations:

  • Buffer composition is crucial - low-salt buffers (150 mM NaCl) work well for maintaining the microprocessor complex

  • Include RNase inhibitors when studying DROSHA-RNA interactions

  • Mild detergents (0.5% NP-40 or 0.1% Triton X-100) help maintain complex integrity

  • Incubation time and temperature affect complex stability (4°C overnight generally yields good results)

Advanced Approach: Tandem RIP Assay
The tandem RNA-immunoprecipitation (RIP 2) assay, based on chromatin re-immunoprecipitation (reChIP), has been successfully used to study DROSHA interactions:

  • First RNA-immunoprecipitation with anti-DROSHA antibodies

  • Elution of precipitates

  • Second RNA-immunoprecipitation using antibodies against suspected interacting partners (e.g., SAFB)

  • Detection of specific RNA transcripts by RT-PCR

This method has successfully demonstrated simultaneous interaction of DROSHA and SAFB on native Nfib mRNA in dental granule neural stem cells .

Validation Strategy:
In vitro processing assays can be used to validate immunoprecipitated DROSHA activity. For example, varying SAFB levels in immunoprecipitated DROSHA complexes showed a direct relationship to Nfib 3' UTR hairpin processing efficiency, confirming functional complex isolation .

What are the considerations when studying non-canonical functions of DROSHA using antibodies?

Research into non-canonical DROSHA functions presents unique challenges requiring specialized experimental approaches:

Non-canonical Functions of DROSHA:

  • Alternative splicing regulation of pre-mRNA exons (e.g., eIF4H gene)

  • Pre-rRNA processing

  • Maintenance of CG methylation

  • Regulation of cellular localization via its nuclear localization signal (NLS)

  • Control of cell fate decisions in stem cells

Experimental Design Considerations:

  • Antibody Selection:

    • Use antibodies targeting different DROSHA domains to distinguish canonical vs. non-canonical functions

    • For studying splicing functions, antibodies against the N-terminal region may be more informative

    • Domain-specific antibodies help identify which regions are necessary for specific functions

  • Controls for Mechanistic Studies:

    • Include catalytically inactive DROSHA mutants (e.g., E1045Q mutation) to distinguish between enzymatic activity and structural roles

    • Utilize DROSHA fragments corresponding to different functional domains:

      • RS-rich region (amino acids 1-390)

      • Central domain (amino acids 390-875)

      • RNase III domains (amino acids 875-1365)

  • Advanced Techniques:

    • Combine RIP with splicing assays to study DROSHA's role in alternative splicing

    • RNA structure analysis (e.g., using mFold) to identify potential DROSHA binding sites

    • In vitro RNA processing assays using immunopurified DROSHA to test substrate specificity

Research has shown that DROSHA can function in splicing enhancement independent of its cleavage activity, highlighting the importance of distinguishing structural from enzymatic roles .

How do researchers interpret conflicting results when DROSHA antibodies show different subcellular localization patterns?

Conflicting DROSHA localization results can stem from several factors that require careful analysis:

Common Sources of Discrepancy:

  • Domain-specific antibodies detect different DROSHA forms:

    • Full-length DROSHA (FL-DROSHA) is predominantly nuclear

    • N-terminal truncated DROSHA (ΔN-DROSHA) shows predominantly cytoplasmic localization due to loss of nuclear localization signal (NLS) in the RS-rich region

  • Cell type-specific localization:

    • Expression patterns vary between stem cells, differentiated cells, and cancer cell lines

    • DROSHA shuttling between nucleus and cytoplasm is regulated differently across cell types

  • Experimental conditions affect localization:

    • Fixation methods may alter epitope accessibility

    • Permeabilization conditions influence antibody penetration

    • Cell cycle stage affects DROSHA distribution

Validation Approaches for Resolving Conflicts:

  • Cell fractionation coupled with western blotting:

    • Compare nuclear versus cytoplasmic fractions using multiple antibodies

    • Example data from one study showed FL-DROSHA predominantly in nuclear fractions while ΔN-DROSHA appeared in cytoplasmic fractions

  • Fluorescent tagging confirmations:

    • Express DROSHA-GFP fusion proteins to confirm antibody staining patterns

    • Use domain-specific constructs to identify localization signals

  • Microscopy technique considerations:

    • Super-resolution microscopy provides more detailed localization information than standard confocal microscopy

    • Live cell imaging with fluorescent tags can reveal dynamic trafficking

When reporting DROSHA localization, researchers should clearly specify the antibody epitope, cell type, and experimental conditions to facilitate accurate interpretation and reproducibility.

What technical challenges exist in optimizing DROSHA antibodies for chromatin immunoprecipitation (ChIP) experiments?

ChIP experiments with DROSHA antibodies present unique challenges due to DROSHA's primary role as an RNA-processing enzyme rather than a direct DNA-binding protein:

Key Technical Challenges:

  • Indirect DNA association:

    • DROSHA primarily interacts with RNA, not DNA

    • ChIP signal may represent indirect association via RNA intermediates or protein partners

    • RNase treatment controls are essential to distinguish RNA-mediated interactions

  • Cross-linking optimization:

    • Standard formaldehyde cross-linking (1%) may be insufficient

    • Dual cross-linking approaches using DSG (disuccinimidyl glutarate) followed by formaldehyde have shown improved results for RNA-binding proteins

    • Over-cross-linking can reduce epitope accessibility

  • Sonication considerations:

    • Chromatin fragmentation must be optimized to maintain protein complex integrity

    • Excessive sonication may disrupt DROSHA-containing complexes

    • Recommended: 10-15 cycles of 30 seconds on/30 seconds off at medium power

  • Antibody selection criteria:

    • Antibodies validated for IP may not work for ChIP

    • Epitope accessibility in cross-linked chromatin differs from solution-based IP

    • Test multiple antibodies targeting different regions of DROSHA

Recommended Controls and Validation Approaches:

  • Sequential ChIP-RIP:

    • Perform ChIP followed by RNA immunoprecipitation to identify RNA intermediates

    • Alternatively, perform RIP followed by DNA analysis to identify associated genomic regions

  • DROSHA knockdown/knockout controls:

    • Generate DROSHA-deficient cells as negative controls

    • Use rescue experiments with wild-type and catalytically inactive DROSHA mutants to distinguish direct binding from enzymatic effects

  • Bioinformatic analysis:

    • Look for enrichment of sequences capable of forming hairpin structures

    • Compare ChIP-seq peaks with known DROSHA RNA targets

DROSHA ChIP experiments should be interpreted with caution and complemented with RNA-based interaction studies for comprehensive understanding of DROSHA genomic associations.

How to design and interpret in vitro RNA processing assays using immunopurified DROSHA?

In vitro RNA processing assays with immunopurified DROSHA provide valuable insights into substrate specificity and processing mechanisms:

Experimental Design Considerations:

  • DROSHA complex immunopurification:

    • Use antibodies targeting DROSHA (C-terminal region often yields better results)

    • Co-immunoprecipitate DGCR8, essential for efficient processing

    • Verify complex integrity by western blot before proceeding

  • RNA substrate preparation:

    • In vitro transcribed RNAs with predicted secondary structures

    • Include both canonical (miRNA precursors) and potential non-canonical substrates

    • For miRNA processing, pri-miRNA fragments of ~150-300 nt work well

    • Label RNA substrates (radioactive or fluorescent) for detection

  • Reaction conditions optimization:

    • Buffer: Typically Tris-HCl (pH 7.5), MgCl₂ (3-5 mM), NaCl (50-100 mM), DTT (1 mM)

    • Incubation time: 30-60 minutes at 37°C

    • Include RNase inhibitors to prevent non-specific degradation

Results Interpretation Framework:

  • Expected outcomes for canonical processing:

    • Pri-miRNA substrates should be cleaved at specific sites

    • Products should be discrete bands of expected size (~65-70 nt pre-miRNAs)

    • Processing efficiency varies between different miRNA substrates

  • Quantitative analysis approaches:

    • Calculate processing efficiency as ratio of product to substrate

    • Compare relative processing efficiencies across different substrates

    • Plot processing kinetics over time to determine reaction rates

  • Example data interpretation:
    One study examining SAFB's impact on DROSHA processing showed:

    • DROSHA complexes from SAFB knockdown cells failed to cleave Nfib 3' UTR hairpin probe

    • Mixing experiments with varying SAFB levels demonstrated dose-dependent processing

    • This approach established direct relationship between SAFB levels and DROSHA processing efficiency

Validation Controls:

  • Include known DROSHA substrates as positive controls

  • Use catalytically inactive DROSHA mutants (E1045Q) as negative controls

  • Perform processing assays with varying protein concentrations to establish dose-dependency

How can researchers use DROSHA antibodies to investigate its role in stem cell fate determination?

DROSHA antibodies are instrumental in exploring the emerging role of DROSHA in stem cell biology and fate determination:

Experimental Approaches:

  • Cell-type specific DROSHA expression profiling:

    • Compare DROSHA levels across stem cells and differentiated lineages

    • Use antibodies targeting different DROSHA domains to identify potential isoforms

    • Correlate expression patterns with differentiation markers

  • DROSHA-dependent RNA regulation in stem cells:

    • Combine DROSHA immunoprecipitation with RNA-seq to identify cell-type specific targets

    • Compare RNA targets between self-renewing and differentiating conditions

    • Use CLIP-seq (cross-linking immunoprecipitation) to map direct DROSHA-RNA interactions

  • Protein interaction networks:

    • Co-immunoprecipitation followed by mass spectrometry to identify stem cell-specific DROSHA partners

    • Validation of interactions using reciprocal co-IP and proximity ligation assays

    • Compare interaction partners between different cell states

Research Findings on DROSHA in Stem Cell Fate:

Studies have revealed that DROSHA regulates hippocampal stem cell fate through several mechanisms:

  • Oligodendrocyte fate regulation:

    • DROSHA, in complex with SAFB protein, targets specific mRNAs like Nfib

    • This regulation controls the balance between neuronal and oligodendrocyte differentiation

    • SAFB binding to DROSHA influences processing of specific RNA substrates in hippocampal stem cells

  • Neural stem cell differentiation control:

    • Dental granule neural stem cells (DG NSCs) predominantly generate neurons and astrocytes but not oligodendrocytes

    • This fate restriction is partially controlled by DROSHA through post-transcriptional repression of NFIB expression

    • DROSHA acts through both canonical miRNA-dependent and direct mRNA targeting mechanisms

  • Methodological approaches used in these studies:

    • Tandem RNA immunoprecipitation (RIP 2) assays demonstrated simultaneous binding of DROSHA and SAFB to the same native Nfib transcripts

    • In vitro processing assays showed SAFB levels directly affect DROSHA processing of specific hairpins

    • Knockdown studies followed by differentiation assays established functional relevance

When investigating DROSHA in stem cell contexts, researchers should combine molecular techniques with functional differentiation assays to establish causal relationships between DROSHA-mediated RNA regulation and cell fate outcomes.

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.