EXOSC1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to EXOSC1 Antibody

The EXOSC1 antibody is a specialized tool designed to detect and study the EXOSC1 protein, a core component of the mammalian exosome complex. This complex is primarily involved in RNA processing and degradation, particularly AU-rich element-containing RNAs . EXOSC1 antibodies enable researchers to investigate the protein's expression, localization, and functional roles in cellular processes and diseases such as cancer .

Functional Studies in Cancer

EXOSC1 antibodies have been critical in uncovering the protein’s role in promoting DNA damage and mutations in kidney renal clear cell carcinoma (KIRC). Key findings include:

  • DNA Cleavage Activity: EXOSC1 cleaves single-stranded DNA (ssDNA) at cytosine (C) sites, leading to C>A transversions .

  • Therapeutic Sensitivity: High EXOSC1 expression sensitizes cancer cells to PARP inhibitors, suggesting its potential as a therapeutic target .

  • Prognostic Value: KIRC patients with elevated EXOSC1 levels exhibit poorer survival outcomes .

Technical Validation

The antibody has been tested across diverse biological samples:

ApplicationTested Samples
WBSGC-7901, HeLa, Jurkat, and MCF-7 cell lines; human stomach tissue
IHCHuman lymphoma tissue (optimal antigen retrieval: TE buffer pH 9.0)
IPHeLa cell lysates

Dilution Guidelines

ApplicationDilution Range
Western Blot (WB)1:500 – 1:1000
Immunoprecipitation (IP)0.5–4.0 µg per 1–3 mg lysate
Immunohistochemistry (IHC)1:20 – 1:200

Key Protocols

  • WB: Use RIPA lysates with SDS-PAGE under reducing conditions .

  • IHC: Paraffin-embedded tissues require heat-induced epitope retrieval (HIER) with TE buffer (pH 9.0) .

Role in Genomic Instability

Studies using EXOSC1 antibodies revealed that the protein promotes DNA damage via ssDNA cleavage, independent of its role in the exosome complex . This activity correlates with increased γ-H2AX foci (a DNA damage marker) and mutations in VHL genes in KIRC .

Therapeutic Implications

EXOSC1’s mutagenic properties make it a biomarker for predicting PARP inhibitor sensitivity. Preclinical models show that EXOSC1 knockdown reduces tumor growth and DNA damage .

Validation and Quality Control

The antibody’s specificity is confirmed by:

  • Band Detection: A single 21 kDa band in WB across multiple cell lines .

  • Functional Knockdown: Reduced γ-H2AX foci and mutations upon EXOSC1 silencing .

  • Peer-Reviewed Studies: Cited in research on pontocerebellar hypoplasia and COVID-19 .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please contact your local distributors for specific delivery time information.
Synonyms
3'-5' exoribonuclease CSL4 homolog antibody; CGI 108 antibody; CGI-108 antibody; CSL4 antibody; CSL4 exosomal core protein homolog antibody; CSL4, S. cerevisiae, homolog of antibody; Csl4p antibody; EXOS1_HUMAN antibody; Exosc1 antibody; Exosomal core protein CSL4 antibody; Exosome complex component csl4 antibody; Exosome component 1 antibody; hCsl4p antibody; Homolog of yeast exosomal core protein CSL4 antibody; p13 antibody; RP11-452K12.9 antibody; SKI4 antibody; Ski4p antibody
Target Names
EXOSC1
Uniprot No.

Target Background

Function

EXOSC1 is a non-catalytic component of the RNA exosome complex. It exhibits 3'->5' exoribonuclease activity and plays a crucial role in various cellular RNA processing and degradation events.

In the nucleus, the RNA exosome complex participates in the proper maturation of stable RNA species, including rRNA, snRNA, and snoRNA. It also eliminates RNA processing by-products and non-coding transcripts, such as antisense RNA species and promoter-upstream transcripts (PROMPTs). Additionally, it degrades mRNAs with processing defects, thereby limiting or preventing their export to the cytoplasm.

The RNA exosome may be involved in Ig class switch recombination (CSR) and/or Ig variable region somatic hypermutation (SHM) by targeting AICDA deamination activity to transcribed dsDNA substrates.

In the cytoplasm, the RNA exosome complex is involved in general mRNA turnover, specifically degrading inherently unstable mRNAs containing AU-rich elements (AREs) within their 3' untranslated regions. It also participates in RNA surveillance pathways, preventing the translation of aberrant mRNAs. Furthermore, it appears to be involved in the degradation of histone mRNA.

The catalytically inactive RNA exosome core complex of 9 subunits (Exo-9) is believed to play a vital role in binding and presenting RNA for ribonucleolysis. It serves as a scaffold for the association with catalytic subunits and accessory proteins or complexes.

EXOSC1, as a peripheral component of the Exo-9 complex, stabilizes the hexameric ring of RNase PH-domain subunits through interactions with EXOSC6 and EXOSC8.

Gene References Into Functions
  1. The combined expression status of p16 and p53 in cervical metastases of CUP may provide a simple method for risk stratification. Further validation of these biomarkers in large prospective trials is crucial for designing rational trials aimed at optimizing CUP treatment. PMID: 28144684
  2. Human exosome hCsl4p participates in RNA degradation. PMID: 22068837
  3. The association of hCsl4p with the exosome is mediated by protein-protein interactions with hRrp42p and hRrp46p. PMID: 11812149
Database Links

HGNC: 17286

OMIM: 606493

KEGG: hsa:51013

STRING: 9606.ENSP00000359939

UniGene: Hs.632089

Protein Families
CSL4 family
Subcellular Location
Nucleus, nucleolus. Nucleus. Cytoplasm.

Q&A

What is EXOSC1 and what are its primary cellular functions?

EXOSC1 (Exosome Component 1) is a component of the exosome complex with multiple cellular functions that extend beyond its canonical role in RNA processing. Recent research has demonstrated that EXOSC1 has the capacity to cleave single-stranded DNA, preferentially at C sites, leading to DNA damage and subsequent mutations . It functions as an endogenous source of mutation (ESM) in cancer cells, particularly in kidney renal clear cell carcinoma (KIRC) . Additionally, EXOSC1 plays a crucial role in mediating the biogenesis and release of exosomes, which are small extracellular vesicles involved in intercellular communication through the transport of various biomolecules and signaling molecules . This dual function in both genomic integrity and cellular communication makes EXOSC1 a protein of significant interest in cancer biology and cellular physiology research.

How do EXOSC1 antibodies differ from antibodies targeting other exosome components?

EXOSC1 antibodies specifically target the EXOSC1 protein, which has shown unique nuclease activity compared to other exosome complex components (EXOSC2-EXOSC9). Research indicates that among all exosome complex members, EXOSC1 most notably enhances mutations in experimental models . When designing experiments, researchers should recognize that EXOSC1 antibodies will detect processes related to DNA damage and mutation generation that may not be observed with antibodies targeting other exosome components. For instance, studies show that EXOSC1 significantly increased mutations in bacterial assays, even more markedly than AID, a known ESM (p=4.08 × 10⁻⁵) . This distinctive property makes EXOSC1 antibodies particularly valuable for investigating DNA damage mechanisms and mutation signatures in cancer research, offering insights that antibodies against other exosome components cannot provide.

What experimental applications are EXOSC1 antibodies validated for?

EXOSC1 antibodies have been validated for several experimental applications in research settings. Based on current literature, these antibodies are primarily validated for Western blot applications, enabling the detection and quantification of EXOSC1 protein expression in various cell types and tissue samples . They can be effectively used in immunohistochemistry to visualize the localization and distribution of EXOSC1 in tissue sections, particularly in cancer samples. Additionally, EXOSC1 antibodies have been employed in immunoprecipitation experiments to isolate EXOSC1 and its interacting partners, facilitating the investigation of protein-protein interactions and functional complexes. These antibodies are also useful in chromatin immunoprecipitation (ChIP) assays to study the association of EXOSC1 with specific genomic regions, providing insights into its role in DNA damage and mutation generation at the molecular level.

How can researchers effectively use EXOSC1 antibodies to study its role in DNA damage and mutation?

To effectively study EXOSC1's role in DNA damage and mutation, researchers should implement a multi-technique approach. First, establish experimental systems with modulated EXOSC1 expression through stable cell lines expressing EXOSC1 (EXOSC1-OE) or EXOSC1 knockdown (EXOSC1-KD) using lentiviral vectors as described in published protocols . For DNA damage assessment, combine γ-H2AX immunostaining (which increases approximately sevenfold with EXOSC1 overexpression) with neutral comet tail assays to quantify DNA breaks .

For mutation analysis, employ differential DNA denaturation PCR (3D-PCR), which can detect genomic C/G>A/T mutations through amplification at lower denaturation temperatures. This approach has successfully demonstrated that EXOSC1 overexpression increases lower temperature amplicons (LTAs) of cancer-related genes like VHL, indicating enhanced mutation rates . When analyzing mutation signatures, focus particularly on C>A transversions, as EXOSC1 shows higher correlation with these mutations compared to other substitution types. Researchers should also consider investigating the relationship between EXOSC1 and DNA repair proteins like XRCC1, as knockdown experiments have shown that XRCC1 impairs EXOSC1's mutation-promoting capabilities .

What are the key considerations when interpreting EXOSC1 expression data in cancer studies?

Third, evaluate strand asymmetry in mutation patterns, as EXOSC1 preferentially cleaves C sites in single-stranded DNA, leading to strand-specific mutation profiles. Fourth, consider the impact of EXOSC1 on response to DNA-damage targeting therapeutics; research demonstrates that EXOSC1 sensitizes cancer cells to poly(ADP-ribose) polymerase (PARP) inhibitors like niraparib and olaparib . Finally, assess the relationship between EXOSC1 and DNA repair pathways, particularly those involving XRCC1, which has been shown to influence EXOSC1-promoted mutations . Integrating these considerations will provide a more comprehensive understanding of EXOSC1's role in cancer biology and potential therapeutic implications.

How does EXOSC1's DNA cleavage activity influence experimental design when studying cancer therapeutics?

EXOSC1's DNA cleavage activity significantly impacts experimental design for cancer therapeutic studies, particularly those involving DNA repair inhibitors. When designing such experiments, researchers must first establish appropriate cellular models with controlled EXOSC1 expression levels through stable transfection systems using vectors like pCDH-Flag EXOSC1 for overexpression or shRNA constructs (e.g., pLKO shEXOSC1-1) for knockdown . Colony formation assays should be conducted with serial dilutions of PARP inhibitors (niraparib, olaparib) to assess sensitization effects, as EXOSC1 overexpression has demonstrated enhanced sensitivity to these agents .

For in vivo studies, xenograft models comparing EXOSC1-OE and control tumors treated with PARP inhibitors are essential, with careful monitoring of tumor volume and animal weight throughout the experiment . When evaluating therapeutic responses, researchers should analyze DNA damage markers (γ-H2AX) and mutation signatures (particularly C>A transversions) to correlate with treatment outcomes. Additionally, consider combination therapy approaches that exploit EXOSC1's role as an endogenous source of mutation, potentially enhancing the efficacy of DNA repair inhibitors through synthetic lethality. Finally, incorporate DNA repair pathway analysis, particularly focusing on XRCC1-dependent 'A' rule DNA repair, which has been shown to influence EXOSC1-mediated mutation generation and potentially treatment response .

What controls should be included when using EXOSC1 antibodies in Western blot applications?

When using EXOSC1 antibodies in Western blot applications, researchers should implement a comprehensive set of controls to ensure result validity and reliability. First, include positive controls consisting of cell lines known to express EXOSC1, such as KIRC cell lines (769P, TUHR14TKB) that have been well-characterized in literature . These positive controls serve as reference points for antibody performance and protein detection. Second, incorporate negative controls using EXOSC1 knockdown cells generated through verified shRNA constructs (e.g., pLKO shEXOSC1-1, pLKO shEXOSC1-2) that have demonstrated high knockdown efficiency in previous studies .

Third, include loading controls using housekeeping proteins (β-actin, GAPDH) to normalize protein levels across samples and ensure equal loading. Fourth, employ specificity controls by pre-absorbing the antibody with recombinant EXOSC1 protein to confirm signal specificity. Fifth, use molecular weight markers to verify that the detected band corresponds to the expected size of EXOSC1 (approximately 21-23 kDa). Finally, consider including cross-reactivity controls by testing the antibody against recombinant proteins of other exosome components (EXOSC2-EXOSC9) to ensure it does not detect related proteins. This comprehensive control strategy will enhance the validity and interpretability of Western blot results when studying EXOSC1 expression and function in various experimental contexts.

What is the optimal protocol for detecting EXOSC1-induced DNA damage using immunofluorescence?

For optimal detection of EXOSC1-induced DNA damage using immunofluorescence, researchers should follow this methodological approach based on published protocols. Begin by establishing appropriate cell models with controlled EXOSC1 expression through stable transfection systems (EXOSC1-OE or EXOSC1-KD) using vectors such as pCDH-Flag EXOSC1 and shRNA constructs . Culture cells on glass coverslips in appropriate media (e.g., DMEM with 10% FBS) under standard conditions (37°C, 5% CO₂), ensuring mycoplasma-free status through regular testing .

For γ-H2AX staining, fix cells with 4% paraformaldehyde for 15 minutes at room temperature, followed by permeabilization with 0.3% Triton X-100 for 10 minutes. Block with 3% BSA in PBS for 1 hour, then incubate with anti-phospho-γ-H2AX (Ser139) antibody (1:500 dilution, Millipore cat. number 05-636) overnight at 4°C . After washing with PBS, incubate with fluorophore-conjugated secondary antibody for 1 hour at room temperature, followed by nuclear counterstaining with DAPI. Mount slides using anti-fade mounting medium and image using confocal microscopy.

For quantification, count cells with ≥5 γ-H2AX foci as positive, analyzing at least 100 cells per condition across multiple fields. Previous studies have shown that EXOSC1 overexpression increases γ-H2AX-positive cells approximately sevenfold compared to controls . Include appropriate controls: positive controls (cells treated with H₂O₂ or UV radiation), negative controls (primary antibody omission), and expression controls (Western blot verification of EXOSC1 modulation). This comprehensive protocol ensures reliable detection and quantification of EXOSC1-induced DNA damage in cellular systems.

How can researchers effectively characterize EXOSC1-induced mutation patterns using 3D-PCR?

To effectively characterize EXOSC1-induced mutation patterns using 3D-PCR (differential DNA denaturation PCR), researchers should implement the following optimized protocol based on published methodologies. Begin by establishing cell models with controlled EXOSC1 expression (EXOSC1-OE, EXOSC1-KD, and vector controls) using verified expression vectors and shRNA constructs . Extract genomic DNA using standard methods that minimize DNA damage during isolation (e.g., phenol-chloroform extraction or commercial kits optimized for genomic DNA integrity).

For 3D-PCR amplification, design primers specific to genes of interest, such as VHL in KIRC studies, that are suitable for gradient PCR conditions . Perform PCR across a denaturation temperature gradient (typically 80-94°C) with small increments (0.5-1°C steps), maintaining other parameters constant (annealing temperature, extension time). Visualize amplification products on agarose gels, identifying lower temperature amplicons (LTAs) that indicate sequences with increased A/T content due to C/G>A/T mutations .

Extract and purify LTA bands for direct sequencing or cloning and sequencing to identify specific mutation patterns. When analyzing sequencing data, focus particularly on C>A transversions, which have shown significant correlation with EXOSC1 expression in previous studies . Calculate mutation frequencies for different substitution types (C>A, G>T, etc.) and compare between experimental conditions. Include appropriate controls in all experiments, including DNA repair pathway controls (e.g., XRCC1 knockdown) which have been shown to influence EXOSC1-promoted mutations . This comprehensive approach allows for detailed characterization of the unique mutation signatures induced by EXOSC1 activity.

What strategies can be employed to validate the specificity of EXOSC1 antibodies?

To validate the specificity of EXOSC1 antibodies, researchers should implement a multi-faceted approach that addresses both positive and negative controls. Begin with Western blot analysis using lysates from cells with confirmed EXOSC1 knockdown (via verified shRNA constructs such as pLKO shEXOSC1-1) compared against control cells . A specific antibody will show significantly reduced signal intensity in knockdown samples, proportional to the knockdown efficiency. Next, perform peptide competition assays by pre-incubating the antibody with excess recombinant EXOSC1 protein or immunogenic peptide before application to samples; specific antibodies will show diminished or abolished signal.

Overexpression validation using cells transfected with EXOSC1 expression vectors (e.g., pCDH-Flag EXOSC1) serves as a positive control, with specific antibodies showing enhanced signal intensity proportional to overexpression levels . For antibodies used in immunoprecipitation, perform reciprocal co-IP experiments using different antibodies targeting the same protein, and verify precipitated proteins by mass spectrometry. Cross-reactivity testing against other exosome components (EXOSC2-EXOSC9) is essential, as these share structural similarities with EXOSC1 . Finally, conduct immunohistochemistry or immunofluorescence with appropriate controls (blocking peptides, knockdown tissues) to validate specificity in intact tissues or cells. This comprehensive validation approach ensures that experimental outcomes reflect genuine EXOSC1 biology rather than non-specific antibody interactions.

How should researchers address contradictory results between EXOSC1 expression and mutation patterns?

When confronting contradictory results between EXOSC1 expression and mutation patterns, researchers should systematically investigate several potential explanations through rigorous experimental approaches. First, verify antibody specificity and EXOSC1 detection methods using the comprehensive validation techniques outlined in section 4.1, as non-specific antibody binding can lead to misleading expression data. Second, reassess EXOSC1 expression at both protein and mRNA levels using multiple techniques (Western blot, qRT-PCR, immunohistochemistry) to confirm consistent expression patterns across methodologies.

Third, carefully examine mutation detection methods, as different techniques may have varying sensitivities for detecting specific mutation types. For instance, 3D-PCR is particularly sensitive for C/G>A/T transitions but may miss other mutation types . Consider employing next-generation sequencing for comprehensive mutation profiling. Fourth, evaluate the influence of DNA repair pathways, particularly XRCC1-dependent processes, which have been shown to modulate EXOSC1-promoted mutations . Knockdown studies of relevant repair proteins may reveal context-dependent effects on mutation patterns.

Fifth, investigate potential tissue or cell-type specificities, as EXOSC1's effects may vary across different cellular contexts. Finally, consider the impact of experimental conditions such as cell culture stress, passage number, or in vitro versus in vivo models on both EXOSC1 expression and mutation patterns. A systematic investigation of these factors, combined with appropriate statistical analyses accounting for variability and potential confounding factors, will help reconcile apparent contradictions and develop a more nuanced understanding of EXOSC1's role in mutagenesis.

What are the common technical challenges when using EXOSC1 antibodies in immunoprecipitation studies?

Immunoprecipitation (IP) studies using EXOSC1 antibodies present several technical challenges that researchers should anticipate and address. First, antibody-antigen binding efficiency may be limited by epitope accessibility, as EXOSC1 functions within the exosome complex where protein-protein interactions may mask binding sites. To overcome this, consider using multiple antibodies targeting different EXOSC1 epitopes or employing denaturing conditions followed by renaturation before IP. Second, non-specific binding can generate false positives, particularly problematic with polyclonal antibodies. Implement stringent washing protocols, pre-clear lysates with protein A/G beads, and include appropriate negative controls (IgG from the same species, EXOSC1 knockdown samples) .

Third, co-immunoprecipitation of interacting proteins can complicate analysis when studying EXOSC1 specifically. Use crosslinking approaches with titratable crosslinkers to stabilize direct interactions, and consider tandem affinity purification for higher specificity. Fourth, low abundance of EXOSC1 in certain cell types may limit detection. Scale up starting material and optimize lysis conditions to maximize protein extraction while preserving native interactions. Fifth, post-translational modifications might affect antibody recognition. Employ phosphatase or deubiquitinase treatments of samples before IP if modifications are suspected to interfere with antibody binding.

Finally, validation of IP results requires complementary approaches. Confirm IP findings through reciprocal co-IP, mass spectrometry identification of precipitated proteins, and functional studies correlating protein interactions with biological outcomes such as DNA damage or mutation patterns . Addressing these technical challenges through careful experimental design and validation will enhance the reliability and interpretability of EXOSC1 immunoprecipitation studies.

What are the future research directions for EXOSC1 antibodies in cancer research?

Future research directions for EXOSC1 antibodies in cancer research should focus on several promising avenues that build upon current understanding of EXOSC1's dual roles in DNA damage and exosome biology. First, developing antibodies that can distinguish between different functional states or post-translational modifications of EXOSC1 will enable more nuanced studies of its activity regulation in various cancer contexts. Second, exploring the potential of EXOSC1 as a biomarker for predicting response to PARP inhibitors and other DNA damage-targeting therapies represents a significant clinical opportunity, given its demonstrated role in sensitizing cancer cells to these agents .

Third, investigating the relationship between EXOSC1's DNA cleavage activity and its role in exosome biogenesis may reveal novel insights into cancer cell communication and metastasis. Fourth, characterizing tissue-specific and cancer type-specific functions of EXOSC1 beyond KIRC will broaden our understanding of its relevance across different malignancies. Fifth, developing therapeutic strategies targeting EXOSC1 directly or exploiting its mutation-inducing properties for synthetic lethality approaches could yield novel cancer treatments.

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.