SUPV3L1 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

SUPV3L1 is a mitochondrial ATP-dependent RNA helicase critical for RNA surveillance and degradation of non-coding mitochondrial transcripts . The HRP-conjugated antibody enables chemiluminescent or chromogenic detection of this protein in experimental assays.

Key Assay Performance

ApplicationDilution RangeValidated Samples
ELISA1:1,000–1:5,000Human serum, cell lysates
Western Blot (WB)1:2,000–1:10,000*HEK-293, PC-3, and HeLa cells (70–80 kDa band observed)
Immunohistochemistry1:250–1:1,000*Human stomach cancer tissue (TE/citrate buffer retrieval)

*Non-conjugated parent antibody data; HRP conjugate optimized for ELISA .

Research Findings

  • Mitochondrial RNA Surveillance: SUPV3L1 is essential for degrading non-coding mitochondrial RNAs, as demonstrated in HeLa cell studies using IP and WB .

  • Neurodegenerative Disease Link: Mutations in SUPV3L1 correlate with RNA processing defects in mitochondrial encephalopathies .

  • Apoptosis Regulation: SUPV3L1 knockdown increases susceptibility to apoptosis, underscoring its role in cell survival .

Comparative Analysis of Available Formats

ConjugateProduct CodeApplications
Unconjugated12826-1-AP WB, IHC, IF/ICC, IP
HRPCSB-PA814232LB01HUELISA
FITCCSB-PA814232LC01HUFluorescence microscopy

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Product dispatch occurs within 1-3 business days of order receipt. Delivery times may vary depending on shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
6330443E10Rik antibody; ATP dependent RNA helicase SUPV3L1; mitochondrial antibody; ATP-dependent RNA helicase SUPV3L1 antibody; EC 3.6.1. antibody; MGC109049 antibody; mitochondrial antibody; OTTHUMP00000019719 antibody; S. Cerevisiae suppressor of var 1 3 like 1 antibody; suppressor of var1 (S.cerevisiae) 3 like 1 antibody; Suppressor of var1 3 like 1 (S. cerevisiae) antibody; Suppressor of var1 3 like 1 (S. cerevisiae)(SUV3) antibody; Suppressor of var1 3 like 1 antibody; Suppressor of var1 3 like protein 1 antibody; Suppressor of var1 3-like protein 1 antibody; SUPV 3 L 1 antibody; SUPV3L1 antibody; SUV 3 antibody; SUV 3 L 1 antibody; SUV3 antibody; SUV3 like protein 1 antibody; SUV3-like protein 1 antibody; SUV3_HUMAN antibody; SUV3L1 antibody
Target Names
SUPV3L1
Uniprot No.

Target Background

Function

This antibody targets SUPV3L1, a key helicase in mitochondrial RNA metabolism. It is a component of the mitochondrial degradosome (mtEXO) complex, responsible for the ATP-dependent degradation of 3' overhang double-stranded RNA in a 3'-to-5' direction. SUPV3L1 is involved in the degradation of non-coding mitochondrial transcripts (mt-ncRNA) and tRNA-like molecules. Functioning as an ATPase and ATP-dependent multisubstrate helicase, it unwinds double-stranded (ds) DNA and RNA, and RNA/DNA heteroduplexes in the 5'-to-3' direction. It plays a crucial role in mitochondrial RNA surveillance, regulating the stability of mature mRNAs, removing aberrant mRNAs, and rapidly degrading non-coding processing intermediates. Furthermore, SUPV3L1 is implicated in recombination, chromatin maintenance, and may offer cellular protection against apoptosis. It is also associated with mitochondrial DNA.

Gene References Into Functions

The following research highlights the diverse roles of SUPV3L1:

  1. Regulation of the cell cycle by the nucleolar-associated human SUV3 protein. PMID: 28291845
  2. Influence of inorganic cofactor availability on hSuv3 DNA unwinding activity. PMID: 25446650
  3. Modulation of mt-mRNA poly(A) tail length by SUV3 bridging PNPase and mtPAP, depending on mitochondrial matrix Pi level. PMID: 24770417
  4. Essential role of PNPase-hSuv3 interaction in efficient mitochondrial RNA degradation. PMID: 23221631
  5. hSuv3p's role in regulating the stability of mature mRNAs and removing non-coding processing intermediates. PMID: 19864255
  6. Mitochondrial matrix localization of hSuv3p helicase and its preferential unwinding of dsDNA. PMID: 12466530
  7. hSUV3 as an ATP-dependent multiple-substrate helicase, unwinding dsRNA, dsDNA, and RNA/DNA heteroduplexes. PMID: 15096047
  8. hSUV3 as a housekeeping gene with an enhancer region and regulatory elements in its basal promoter. PMID: 15919122
  9. Interaction between Suv3 protein and HBXIP, a survivin cofactor in apoptosis suppression. PMID: 16176273
  10. Down-regulation of hSUV3 leading to cell cycle perturbations, AIF- and caspase-dependent apoptosis, and p53 induction. PMID: 17352692
  11. Interaction of human SUV3 protein with human WRN and BLM helicases, and the effect of SUV3 gene silencing on homologous recombination. PMID: 17961633
  12. Essential role of SUV3 in maintaining mitochondrial function through mitochondrial RNA regulation. PMID: 18678873
  13. The hSUV3-hPNPase complex as an integral entity for efficient structured RNA degradation in mammalian mitochondria. PMID: 19509288
Database Links

HGNC: 11471

OMIM: 605122

KEGG: hsa:6832

STRING: 9606.ENSP00000352678

UniGene: Hs.106469

Protein Families
Helicase family
Subcellular Location
Nucleus. Mitochondrion matrix. Mitochondrion matrix, mitochondrion nucleoid.
Tissue Specificity
Broadly expressed.

Q&A

What is SUPV3L1 and why is it important for mitochondrial function?

SUPV3L1 (Suppressor of var1, 3-like protein 1) is a crucial ATP-dependent RNA helicase that plays a major role in mitochondrial RNA metabolism. It functions as a component of the mitochondrial degradosome (mtEXO) complex that degrades double-stranded RNA with 3'-to-5' directionality in an ATP-dependent manner.

SUPV3L1 is essential for several mitochondrial processes, including:

  • Regulation of mature mRNA stability

  • Removal of aberrantly formed mRNAs

  • Degradation of non-coding processing intermediates

  • Protection of cells from apoptosis

  • Association with and maintenance of mitochondrial DNA

This protein's critical role in RNA surveillance makes it a significant target for research into mitochondrial dysfunction, aging-related diseases, and metabolic disorders .

What applications are suitable for HRP-conjugated SUPV3L1 antibodies?

HRP-conjugated SUPV3L1 antibodies are particularly valuable for applications where direct enzyme detection eliminates the need for secondary antibodies. Based on the available data, these antibodies are validated for:

  • ELISA (primary application)

  • Western blot (WB) with directly visualized results

  • Immunohistochemistry (IHC) applications requiring enhanced sensitivity

  • Immunocytochemistry with reduced background

The HRP conjugation provides enhanced detection sensitivity while simplifying experimental workflows by eliminating secondary antibody incubation steps .

What dilution ranges are recommended for different applications of SUPV3L1 antibodies?

The optimal dilution ranges vary by application method and specific antibody formulation. For research applications using SUPV3L1 antibodies, the following dilutions are recommended:

ApplicationRecommended Dilution RangeSecondary Antibody Recommendation
Western Blot1:1000 - 1:4000HRP-conjugated secondary at 1:50,000-100,000 (if using non-conjugated primary)
ELISA1:312500Not required for HRP-conjugated antibody
Immunohistochemistry1:50 - 1:200Not required for HRP-conjugated antibody
Immunofluorescence1:50 - 1:200Not required for HRP-conjugated antibody

These dilution ranges should be optimized for each specific experimental setup and sample type .

How can I optimize signal-to-noise ratio when using HRP-conjugated SUPV3L1 antibody for western blot detection?

Optimizing signal-to-noise ratio for HRP-conjugated SUPV3L1 antibody requires a multifaceted approach:

  • Blocking optimization: Test different blocking agents (5% BSA vs. 5% non-fat milk). Data suggests that for SUPV3L1 detection, 3% non-fat dry milk in TBST provides optimal blocking with minimal background .

  • Antibody titration: Perform a dilution series experiment (e.g., 1:1000, 1:2000, and 1:4000) to identify the minimal concentration that provides sufficient signal. The higher dilution range (1:4000) often yields cleaner results for SUPV3L1 when using HRP-conjugated antibodies .

  • Incubation conditions:

    • Primary antibody: Overnight at 4°C with gentle agitation improves specific binding

    • Washing: 5-6 washes with TBST for 5-10 minutes each significantly reduces background

  • Substrate selection: For SUPV3L1 HRP-conjugated antibody, ECL substrates with enhanced sensitivity but controlled signal development time prevent overexposure and maintain signal specificity .

  • Exposure time optimization: Short exposures (1-5 seconds) often provide the best balance between detecting SUPV3L1 signal while minimizing non-specific background .

What challenges might arise when using SUPV3L1 antibodies for immunofluorescence, and how can they be addressed?

Several challenges are common when using SUPV3L1 antibodies for immunofluorescence applications:

  • Mitochondrial localization difficulties: Since SUPV3L1 localizes to mitochondria, distinguishing specific signal from mitochondrial autofluorescence can be challenging. Solution: Use appropriate negative controls and mitochondrial co-staining with established markers (e.g., MitoTracker) to confirm specificity .

  • Fixation-dependent epitope masking: The conformation of SUPV3L1 can be affected by different fixation methods. Solution: Compare multiple fixation protocols (4% paraformaldehyde, methanol, or acetone fixation) to determine optimal epitope preservation .

  • Signal intensity variability: Expression levels vary across cell types. Solution: Validate antibody concentration for each cell line being studied. NIH-3T3 cells have been successfully stained at 1:100 dilution as a reference point .

  • Non-specific nuclear staining: Some SUPV3L1 antibodies may show nuclear staining. Solution: Verify specificity using siRNA knockdown controls and multiple antibodies targeting different epitopes of SUPV3L1 .

  • HRP signal amplification issues: For direct immunofluorescence with HRP-conjugated antibodies, tyramide signal amplification must be carefully controlled. Solution: Optimize hydrogen peroxide concentration and reaction time to prevent excessive signal amplification and background .

How should researchers interpret conflicting data between mitochondrial and non-mitochondrial SUPV3L1 localization?

When faced with conflicting localization data for SUPV3L1 (primarily mitochondrial vs. additional non-mitochondrial pools), researchers should employ a systematic analytical approach:

  • Antibody epitope considerations: The specific region of SUPV3L1 targeted by the antibody may affect localization results. For example, antibodies targeting the 577-786 amino acid region (C-terminal) versus those targeting other regions may give different results .

  • Cell type-specific expression patterns: SUPV3L1 may have different subcellular distributions in different cell types. Compare localizations across multiple validated cell lines.

  • Isoform recognition: Verify whether the antibody recognizes all known SUPV3L1 isoforms. Some antibodies may preferentially detect specific splice variants with altered localization patterns .

  • Validation with orthogonal techniques: Combine immunofluorescence with subcellular fractionation and western blotting to quantitatively assess protein distribution. Enhanced validation using RNAseq data can confirm expression patterns .

  • Functional validation with CRISPR/Cas9: Generate tagged endogenous SUPV3L1 to avoid overexpression artifacts and track true physiological localization.

Remember that dual localization could represent legitimate biological phenomena rather than technical artifacts, as SUPV3L1 has been implicated in both mitochondrial functions and other cellular processes .

What controls should be included when validating SUPV3L1 antibody specificity?

A comprehensive validation strategy for SUPV3L1 antibodies should include multiple control types:

  • Positive controls:

    • Cell lines with confirmed SUPV3L1 expression (validated through RNAseq data)

    • Recombinant SUPV3L1 protein standards

    • Western blots should show a band at approximately 87-88 kDa

  • Negative controls:

    • SUPV3L1 knockout or knockdown samples (siRNA/shRNA-treated cells)

    • Competing peptide blocking experiment using the immunogenic peptide (aa 577-786 for many available antibodies)

    • Primary antibody omission control

  • Orthogonal validation:

    • Multiple antibodies targeting different epitopes of SUPV3L1

    • Correlation with RNAseq expression data across tissues

    • Recombinant expression systems

  • Application-specific controls:

    • For IHC/IF: Include secondary-only controls to assess non-specific binding

    • For WB: Include molecular weight markers and loading controls

    • For HRP-conjugated antibodies: Include enzyme activity controls to confirm conjugate functionality

How can researchers effectively troubleshoot weak or non-specific signals when using SUPV3L1 HRP-conjugated antibodies?

When encountering weak or non-specific signals with SUPV3L1 HRP-conjugated antibodies, consider this systematic troubleshooting approach:

For weak signals:

  • Antibody concentration: Decrease dilution to 1:500 for western blot, but be cautious of increased background.

  • Protein loading: Increase total protein loaded (up to 50 μg) to detect low-abundance SUPV3L1.

  • HRP activity loss: Verify conjugate stability; avoid repeated freeze-thaw cycles of the antibody.

  • Detection system sensitivity: Use high-sensitivity ECL substrates designed for HRP detection.

  • Protein extraction efficiency: Test alternative lysis buffers optimized for mitochondrial protein extraction.

For non-specific signals:

  • Blocking optimization: Increase blocking agent concentration and time.

  • Sample preparation: Ensure proper denaturation of proteins without aggregation.

  • Wash protocol intensification: Increase number and duration of washes.

  • Antibody specificity verification: Perform peptide competition assays with the immunogenic sequence.

  • Alternative antibody selection: Consider antibodies targeting different epitopes of SUPV3L1 .

A systematic testing grid varying multiple parameters simultaneously can efficiently identify optimal conditions for specific SUPV3L1 detection.

What is the significance of the immunogen sequence when selecting a SUPV3L1 antibody for specific applications?

The immunogen sequence is a critical factor that directly impacts antibody performance across different applications:

  • Epitope accessibility in different applications:

    • For SUPV3L1, antibodies raised against the C-terminal region (aa 577-786) have demonstrated good performance in Western blot, IF, and IHC applications .

    • Antibodies targeting the sequence "IQHIPLSLRVRYVFCTAPINKKQPFVCSSLLQFARQYSRNEPLTFAWLRRYIKWPLLPPKNIKDLMDLEAVH" have shown specificity in immunohistochemistry applications .

  • Structural considerations:

    • Epitopes located in highly conserved functional domains may provide better cross-species reactivity but potentially increase cross-reactivity with related helicases.

    • The ATP-binding domain of SUPV3L1 is highly conserved, making it less ideal as a specific immunogen target.

  • Post-translational modifications:

    • Antibodies raised against regions containing phosphorylation or other modification sites may show differential reactivity depending on the protein's modification state.

    • Unmodified SUPV3L1 is targeted by most available antibodies .

  • Application-specific performance:

    • For detecting denatured SUPV3L1 (Western blot), linear epitopes from any region may be suitable.

    • For applications requiring native conformation detection (IP, IF), surface-exposed epitopes are preferable.

When selecting an antibody, researchers should match the immunogen sequence with their specific experimental needs and the structural context of the protein in their application .

How does the performance of HRP-conjugated SUPV3L1 antibodies compare to unconjugated versions for multiplex detection strategies?

HRP-conjugated versus unconjugated SUPV3L1 antibodies present different advantages and limitations in multiplex detection scenarios:

Performance Comparison:

ParameterHRP-ConjugatedUnconjugated
Signal amplificationDirect enzymatic amplification; no secondary requiredRequires secondary antibody for detection
Multiplexing capacityLimited due to single detection channelSuperior for fluorescent multiplexing with different species primaries
Cross-reactivityReduced secondary antibody cross-reactivityPotential cross-reactivity between secondary antibodies
Signal-to-noise ratioHigher background potential from direct enzymatic activityOften cleaner with optimized secondary antibody dilutions
Detection flexibilityFixed to colorimetric/chemiluminescent detectionCompatible with fluorescent, colorimetric or chemiluminescent detection
Workflow complexitySimplified (no secondary antibody step)Additional incubation and washing steps
Cost considerationsHigher initial cost; fewer reagents neededLower primary cost; additional secondary antibody required

For multiplex detection strategies specifically:

  • HRP-conjugated antibodies are ideal for sequential multiplex IHC using tyramide signal amplification and antibody stripping.

  • Unconjugated antibodies offer greater flexibility for simultaneous multiple protein detection when paired with spectrally distinct fluorescent secondary antibodies.

  • For co-localization studies of SUPV3L1 with other mitochondrial markers, unconjugated antibodies raised in different host species provide superior multiplexing capabilities .

What specialized techniques can be applied to enhance detection sensitivity for low-abundance SUPV3L1 in primary tissue samples?

Detecting low-abundance SUPV3L1 in primary tissues requires specialized enhancement techniques:

  • Tyramide Signal Amplification (TSA):

    • HRP-conjugated SUPV3L1 antibodies are particularly suitable for TSA

    • This technique can increase sensitivity 10-100 fold

    • Optimal protocol: Primary antibody at 1:200-1:500, followed by 10-minute TSA reaction

  • Proximity Ligation Assay (PLA):

    • For detecting SUPV3L1 interactions with other mitochondrial RNA processing components

    • Provides single-molecule resolution sensitivity

    • Particularly useful for detecting low-abundance complexes

  • Tissue preprocessing techniques:

    • Heat-mediated antigen retrieval in citrate buffer (pH 6.0) optimizes SUPV3L1 epitope accessibility

    • Prolonged primary antibody incubation (48-72 hours at 4°C) at higher concentration (1:25-1:50)

    • Use of signal enhancing additives (0.1% Triton X-100, 1% BSA)

  • Mitochondrial enrichment prior to analysis:

    • Tissue fractionation to concentrate mitochondria before immunostaining

    • Density gradient separation to purify mitochondrial fractions

    • Digitonin-based permeabilization to preferentially access mitochondrial compartments

  • Signal capture optimization:

    • Extended exposure times with high-sensitivity cameras

    • Deconvolution microscopy to enhance signal discrimination

    • Spectral unmixing to separate SUPV3L1 signal from tissue autofluorescence

These techniques can be combined as needed depending on tissue type and specific experimental requirements.

How can researchers effectively employ SUPV3L1 antibodies in studies investigating mitochondrial dysfunction in neurodegenerative diseases?

SUPV3L1 antibodies offer valuable insights into mitochondrial RNA metabolism dysfunction in neurodegenerative contexts when applied with these specialized approaches:

  • Comparative expression analysis:

    • Quantify SUPV3L1 levels in affected vs. unaffected brain regions using calibrated Western blot

    • Map expression patterns across different neuronal populations using IHC with HRP-conjugated antibodies

    • Recommended dilution: 1:100 for paraffin-embedded tissue sections with 20-minute DAB development

  • Co-localization studies with disease markers:

    • Double immunofluorescence labeling with SUPV3L1 and disease-specific proteins (tau, α-synuclein, etc.)

    • Assess mitochondrial localization changes using SUPV3L1 antibody (1:50) and mitochondrial markers

    • Confocal z-stack imaging to precisely analyze spatial relationships

  • Functional correlation techniques:

    • Combine SUPV3L1 immunostaining with mitochondrial functional indicators (JC-1, MitoSOX)

    • Correlate SUPV3L1 levels with mitochondrial membrane potential in individual neurons

    • Assess relationship between SUPV3L1 expression and mitochondrial RNA granule integrity

  • Dynamic analysis in disease models:

    • Track SUPV3L1 expression changes throughout disease progression in model systems

    • Employ HRP-conjugated antibodies for high-throughput IHC analysis of multiple tissue samples

    • Correlate with biochemical markers of disease advancement

  • Intervention assessment:

    • Use SUPV3L1 antibodies to evaluate the efficacy of mitochondrial-targeted therapies

    • Quantify changes in SUPV3L1 distribution and function following experimental treatments

    • Recommended protocol: Western blot analysis with 1:1000 antibody dilution after mitochondrial isolation

This approach provides multidimensional insights into the relationship between mitochondrial RNA metabolism and neurodegenerative pathology.

How should researchers interpret variations in SUPV3L1 band patterns between different tissue types in Western blot analyses?

Variations in SUPV3L1 band patterns across different tissues require careful interpretation:

  • Expected band patterns:

    • Primary SUPV3L1 band: 87-88 kDa (full-length protein)

    • Potential additional bands: 55-60 kDa (cleavage product) and 100-110 kDa (post-translationally modified forms)

  • Tissue-specific variation analysis:

    • Multiple isoforms: Different tissues may express tissue-specific splice variants of SUPV3L1

    • Post-translational modifications: Phosphorylation states vary between tissues (particularly brain vs. liver)

    • Proteolytic processing: Mitochondrial import can generate tissue-specific cleavage patterns

  • Verification approaches:

    • Peptide mapping using different domain-specific antibodies

    • Mass spectrometry validation of bands

    • Correlation with tissue-specific transcript data from RNAseq

  • Common confounding factors:

    • Sample preparation differences (protease inhibitor cocktail composition)

    • Tissue-specific interfering proteins

    • Variations in mitochondrial enrichment between tissue preparations

  • Quantification considerations:

    • Normalize to tissue-specific mitochondrial markers rather than total protein

    • Consider the ratio of full-length to processed forms as a potential biological indicator

    • Account for tissue-specific background when comparing expression levels

Western blot analysis of NIH-3T3, HeLa, and HEK293 cells shows consistent detection of the main SUPV3L1 band at ~88 kDa, providing reliable positive controls for comparing tissue samples .

What methodological approaches can be used to study SUPV3L1's role in mitochondrial RNA granule formation?

Investigating SUPV3L1's role in mitochondrial RNA granule formation requires specialized methodological approaches:

  • High-resolution co-localization analysis:

    • Super-resolution microscopy (STED, STORM) using HRP-conjugated antibodies with fluorescent tyramide substrates

    • Triple immunolabeling for SUPV3L1 (1:50 dilution), RNA granule markers (GRSF1), and mitochondrial markers

    • 3D reconstruction of confocal z-stacks to analyze spatial distribution of granules

  • Live-cell imaging strategies:

    • CRISPR/Cas9 knock-in of fluorescent tags to endogenous SUPV3L1

    • Optogenetic manipulation of SUPV3L1 activity combined with RNA granule tracking

    • Correlative light-electron microscopy for ultrastructural analysis of immunolabeled granules

  • Biochemical isolation and characterization:

    • Density gradient fractionation of mitochondrial RNA granules

    • Immunoprecipitation using SUPV3L1 antibodies to isolate granule components

    • Mass spectrometry analysis of SUPV3L1-associated proteins in different cell states

  • Functional perturbation experiments:

    • SUPV3L1 depletion followed by RNA FISH to assess granule integrity

    • Structure-function analysis using mutant SUPV3L1 constructs

    • RNA-protein crosslinking immunoprecipitation to identify direct SUPV3L1 RNA targets

  • Stress response and dynamic analysis:

    • Tracking SUPV3L1 redistribution during mitochondrial stress responses

    • Pulse-chase labeling of newly synthesized RNAs to assess SUPV3L1's role in RNA processing

    • Quantitative analysis of granule size, number, and composition under different conditions

These approaches provide complementary insights into SUPV3L1's dynamic role in RNA granule biology.

How can researchers differentiate between specific and non-specific binding when using novel SUPV3L1 antibodies for chromatin immunoprecipitation studies?

Differentiating specific from non-specific binding in chromatin immunoprecipitation (ChIP) studies with SUPV3L1 antibodies requires a comprehensive validation strategy:

  • Sequential ChIP validation approach:

    • Perform antibody titration series (1:50, 1:100, 1:200, 1:500)

    • Compare enrichment patterns across multiple genomic regions (mitochondrial DNA vs. nuclear DNA)

    • Conduct parallel ChIP with different antibodies targeting distinct SUPV3L1 epitopes

  • Critical controls for SUPV3L1 ChIP specificity:

    • SUPV3L1 knockout/knockdown controls to establish background signal

    • ChIP with non-specific IgG from the same species (rabbit for most SUPV3L1 antibodies)

    • Pre-clearing optimization to reduce non-specific binding

    • Peptide competition assays with the immunizing peptide (aa 577-786)

  • Quantitative assessment metrics:

    • Signal-to-noise ratio calculation for each potential binding site

    • Enrichment over input calculation with statistical significance testing

    • Comparative analysis of enrichment patterns between replicate experiments

  • Validation of putative binding regions:

    • Orthogonal techniques (EMSA, DNA footprinting) to confirm direct binding

    • Reporter assays to assess functional significance of binding

    • Analysis of evolutionary conservation of binding sites

  • Technical optimizations for SUPV3L1 ChIP:

    • Crosslinking time optimization (1-2% formaldehyde for 5-15 minutes)

    • Sonication conditions tuned for optimal chromatin fragmentation

    • Buffer composition adjustments to reduce non-specific binding

    • Two-step crosslinking for improved protein-DNA capture

These approaches collectively provide a robust framework for distinguishing authentic SUPV3L1 chromatin interactions from technical artifacts.

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.