SNRPD1 (small nuclear ribonucleoprotein D1 polypeptide) is a 13–16 kDa protein encoded by the SNRPD1 gene (Entrez Gene ID: 6632; UniProt ID: P62314). It functions as a charged scaffold to facilitate snRNP assembly and stabilize RNA-protein interactions during spliceosome formation . SNRPD1 is also a known autoantigen in systemic lupus erythematosus (SLE), where antibodies target snRNP complexes .
Recombinant monoclonal antibodies against SNRPD1 are produced using phage display or hybridoma technology, ensuring high specificity and batch-to-batch consistency . Key examples include:
Clone/Product | Host | Applications | Target Species | Citations |
---|---|---|---|---|
24E2 (MA5-50236) | Mouse | WB, IF, IP | Human | Thermo Fisher |
10352-1-AP | Rabbit | WB, IHC, ELISA | Human, Mouse | Proteintech |
ab233115 | Rabbit | WB, IHC-P | Human, Rat, Pig | Abcam |
Western Blot (WB): Detects SNRPD1 at ~13 kDa in human cell lines (e.g., HEK-293, HL-60) .
Immunohistochemistry (IHC): Localizes SNRPD1 in nuclei of paraffin-embedded tissues (liver, kidney, prostate) .
Autoimmunity: SNRPD1 antibodies are biomarkers for SLE, aiding in diagnostic assays .
Cancer: SNRPD1 overexpression correlates with tumor progression; targeted studies use these antibodies to dissect spliceosome dysregulation .
Parameter | Detail |
---|---|
Immunogen | Recombinant full-length SNRPD1 |
Recommended Dilution | WB: 1:500–1:1000; IHC: 1:50–1:200 |
Observed MW | 13 kDa (monomer); 30 kDa (dimer) |
Knockdown Validation: Reduced signal in SNRPD1-silenced Jurkat cells confirms specificity .
Cross-Reactivity: Reacts with human, mouse, rat, and pig homologs .
SNRPD1 is a small nuclear ribonucleoprotein that belongs to the SNRNP core protein family. It plays a crucial role in pre-mRNA splicing as a core component of the spliceosomal U1, U2, U4, and U5 small nuclear ribonucleoproteins (snRNPs), which are the building blocks of the spliceosome . SNRPD1 serves as a component of both the pre-catalytic spliceosome B complex and activated spliceosome C complexes . Additionally, as part of the minor spliceosome, it participates in the splicing of U12-type introns in pre-mRNAs .
At the molecular level, SNRPD1 may function as a charged protein scaffold to promote snRNP assembly or strengthen snRNP-snRNP interactions through non-specific electrostatic contacts with RNA . This scaffolding role is critical for maintaining the structural integrity of the spliceosome during the dynamic process of RNA splicing.
SNRPD1 has a calculated molecular weight of approximately 16 kDa, although it is typically observed at around 13 kDa in Western blot analyses . The protein is also known by several aliases, including:
The human SNRPD1 gene is located on chromosome 18 and is identified by the following database entries:
Based on the technical data from multiple sources, SNRPD1 recombinant monoclonal antibodies have been validated for the following applications:
Application | Recommended Dilution | Validated Species |
---|---|---|
Western Blot (WB) | 1:500-1:2000 | Human, Mouse, Rat |
Immunohistochemistry (IHC) | 1:50-1:200 | Human |
Immunofluorescence (IF) | 1:50-1:100 | Human |
Most SNRPD1 antibodies have been validated for human samples, with certain antibodies also showing cross-reactivity with rat and pig samples due to sequence homology .
While recommended dilutions provide a starting point, optimal antibody concentration should be determined empirically for each experimental system. Begin with the manufacturer's suggested dilution range, then perform a dilution series to identify the concentration that provides:
Strong specific signal with minimal background
Linear detection range for quantitative analyses
Reproducible results across replicates
For Western blotting, a typical approach is to prepare a dilution series (e.g., 1:500, 1:1000, 1:2000) and assess both signal strength and background. For immunohistochemistry and immunofluorescence, a similar titration approach is recommended, starting with 1:50 and progressing to more dilute solutions .
It's important to note that optimal dilutions may vary depending on sample type, fixation method, detection system, and specific experimental conditions .
When selecting an SNRPD1 recombinant monoclonal antibody for research applications, consider these critical factors:
Epitope recognition: Different antibody clones recognize different epitopes on the SNRPD1 protein. For example, some antibodies target full-length recombinant SNRPD1 protein , while others may target specific peptide sequences. The epitope location can affect antibody performance in different applications.
Validated applications: Confirm that the antibody has been validated for your specific application. Some antibodies perform well in Western blot but poorly in IHC, or vice versa .
Cross-reactivity profile: Evaluate whether the antibody cross-reacts with SNRPD1 from other species if you're working with non-human models. Some antibodies show strong cross-reactivity with mouse and rat SNRPD1 due to sequence conservation .
Performance in specific experimental conditions: Some antibodies may perform better under specific conditions (native vs. denatured proteins, different fixation methods, etc.) .
Publication record: Consider antibodies that have been cited in peer-reviewed publications related to your research area, as this provides evidence of successful application .
Recombinant monoclonal antibodies offer several advantages over traditional antibodies for SNRPD1 research:
Superior batch-to-batch consistency: Recombinant antibodies exhibit higher lot-to-lot reproducibility compared to traditional monoclonal antibodies, as they avoid issues of genetic drift and instability associated with hybridoma-based technology . This consistency is crucial for longitudinal studies or when comparing results across different experimental timeframes.
Defined sequence and production: Recombinant antibodies are produced from entirely defined genetic sequences under controlled conditions, eliminating the variability introduced by animal immunization .
Ethical considerations: Production of recombinant antibodies eliminates the need for animal immunization, addressing ethical concerns regarding animal use in research .
Customizability: Once the peptide sequence of an antibody is known, it can be modified to fit specific research needs, such as changing the Fc fragment to another antibody class or species, or modifying the antigen-binding region to increase binding affinity or specificity .
Production efficiency: Recombinant antibodies can be produced within weeks rather than months, as they don't require animal immunization or hybridoma optimization .
Antibody Type | Batch Consistency | Production Time | Customizability | Ethical Considerations |
---|---|---|---|---|
Recombinant Monoclonal | Excellent | Weeks | High | No animal use |
Traditional Monoclonal | Good | Months | Limited | Requires animals |
Polyclonal | Variable | Months | Very limited | Requires animals |
The following protocol is recommended for Western blot analysis of SNRPD1:
Sample preparation:
Prepare cell or tissue lysates in an appropriate lysis buffer containing protease inhibitors
For optimal SNRPD1 detection, include RNase inhibitors in your lysis buffer as SNRPD1 is an RNA-binding protein
Protein separation:
Transfer and blocking:
Transfer proteins to a PVDF or nitrocellulose membrane
Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Antibody incubation:
Dilute primary SNRPD1 antibody at 1:500-1:1000 in blocking buffer
Incubate overnight at 4°C with gentle agitation
Wash 3-5 times with TBST
Incubate with appropriate HRP-conjugated secondary antibody at 1:5000-1:10000 for 1 hour at room temperature
Wash 3-5 times with TBST
Detection:
For optimal SNRPD1 detection in tissue sections by immunohistochemistry, consider the following protocol adjustments:
Antigen retrieval optimization:
Test both heat-induced epitope retrieval methods (citrate buffer pH 6.0 and EDTA buffer pH 9.0) to determine which provides optimal signal-to-noise ratio
Adjust retrieval time (10-30 minutes) based on tissue type and fixation conditions
Antibody concentration:
Begin with the recommended dilution range (1:50-1:200)
Perform a dilution series to identify optimal concentration for your specific tissue type
Higher concentrations may be needed for tissues with low SNRPD1 expression
Incubation conditions:
Test both overnight incubation at 4°C and 1-2 hour incubation at room temperature
For some tissue types, extended incubation times may improve signal intensity
Detection system selection:
Polymer-based detection systems often provide superior sensitivity and lower background compared to traditional ABC methods
For low-abundance targets like SNRPD1, tyramide signal amplification can significantly enhance detection sensitivity
Counterstaining optimization:
Adjust hematoxylin counterstaining time to ensure nuclear details remain visible without obscuring specific SNRPD1 staining
Consider nuclear fast red as an alternative counterstain if hematoxylin interferes with visualization
Controls:
Include known positive control tissues (tonsil or lymphoid tissues often express SNRPD1)
Use isotype controls at the same concentration as the primary antibody
Consider including tissues from SNRPD1 knockdown models as negative controls if available
Generating custom recombinant monoclonal antibodies against SNRPD1 involves several steps:
Sequence determination:
Expression vector construction:
Design DNA geneblocks optimized for expression in human cells using codon optimization tools
For full-length antibodies, add an N-terminal signal peptide sequence to the geneblock
Clone the resulting DNA fragments into an appropriate expression vector (e.g., pEGFP-N1 with GFP removed)
For each full-length antibody, generate both heavy chain (HC) and light chain (LC) plasmids for co-expression
Antibody expression:
Purification:
Antibody modification approaches:
Species specificity swapping: Generate geneblocks corresponding only to the variable regions of the HCs and LCs, then combine them with PCR fragments for the constant regions of the target species (e.g., changing from mouse to rabbit or human specificity)
Generation of antibody fragments: Convert full-length antibodies to scFv (single chain variable fragment), scFvC (single chain variable fragment plus truncated constant region), or Fab (antigen binding fragment) formats for specific applications
Direct labeling: Conjugate purified antibodies or fragments with fluorescent dyes for applications like super-resolution microscopy
SNRPD1 antibodies provide powerful tools for investigating spliceosome assembly and dynamics through several advanced techniques:
Immunoprecipitation-based analyses:
Perform RNA immunoprecipitation (RIP) using SNRPD1 antibodies to identify RNA species associated with SNRPD1-containing snRNPs
Conduct co-immunoprecipitation experiments to map protein-protein interactions within the spliceosome complex
Combine with mass spectrometry for comprehensive characterization of spliceosome composition at different assembly stages
Chromatin immunoprecipitation (ChIP):
Use SNRPD1 antibodies to investigate co-transcriptional splicing events and potential chromatin associations
Combine with sequencing (ChIP-seq) to identify genome-wide distribution of splicing machinery
Live-cell imaging approaches:
Convert SNRPD1 antibody fragments (scFv) into intrabodies for live-cell tracking of spliceosome dynamics
Bead-load fluorescently labeled SNRPD1 antibody fragments to visualize spliceosome distribution in real-time
Perform FRAP (Fluorescence Recovery After Photobleaching) experiments using labeled antibodies to assess spliceosome turnover rates
Super-resolution microscopy:
Proximity ligation assay (PLA):
Combine SNRPD1 antibodies with antibodies against other spliceosome components to visualize and quantify protein-protein interactions in situ
This approach can reveal spatial organization of spliceosome assembly intermediates
To ensure the specificity of SNRPD1 antibody staining, implement these validation strategies:
Genetic approaches:
Perform siRNA knockdown of SNRPD1 (validated siRNA sequence: available in literature) and confirm reduction of signal by Western blot and immunostaining
If feasible, use CRISPR/Cas9 to generate SNRPD1 knockout cell lines as negative controls
Rescue experiments with exogenous SNRPD1 expression can provide additional validation
Peptide competition assays:
Pre-incubate the antibody with excess purified SNRPD1 protein or immunizing peptide
Compare staining patterns with and without competition
Specific signals should be significantly reduced or eliminated after competition
Multiple antibody validation:
Use two or more antibodies raised against different epitopes of SNRPD1
Concordant staining patterns across different antibodies suggest specific detection
This approach is particularly valuable for novel or contentious findings
Correlation with mRNA expression:
Compare antibody staining intensity with SNRPD1 mRNA levels (from RNA-seq or qPCR)
Positive correlation supports antibody specificity
Expected subcellular localization:
Confirm that staining patterns are consistent with known SNRPD1 localization (primarily nuclear with speckled distribution pattern in interphase cells)
Aberrant localization patterns may indicate non-specific binding
SNRPD1 antibodies offer valuable tools for investigating spliceosome dysfunction in various diseases:
Autoimmune disorders:
Cancer research:
Investigate altered splicing patterns in cancer cells using SNRPD1 antibodies
Perform immunohistochemistry on tissue microarrays to correlate SNRPD1 expression with tumor grade, stage, and patient outcomes
Combine with RNA-seq to link SNRPD1 distribution changes with specific splicing alterations
Neurodegenerative disorders:
Several neurodegenerative diseases feature aberrant RNA processing
Use SNRPD1 antibodies to examine spliceosome composition and distribution in affected tissues
Study co-localization with disease-specific protein aggregates (e.g., tau, alpha-synuclein)
Developmental disorders:
Investigate the developmental expression pattern of SNRPD1 in embryonic tissues
Study potential splicing changes during critical developmental windows
Examine SNRPD1 interactions with developmental regulators
Cellular stress response:
Monitor SNRPD1 localization and post-translational modifications during various cellular stresses (oxidative stress, heat shock, etc.)
Investigate potential stress-induced alterations in spliceosome composition or activity
Recent innovative approaches combining SNRPD1 antibodies with other methodologies include:
Proximity-dependent biotinylation (BioID or TurboID):
Generate SNRPD1-BioID fusion proteins to identify proteins that interact with or are in close proximity to SNRPD1 in living cells
Validate interactions using co-immunoprecipitation with SNRPD1 antibodies
This approach can reveal transient interactions within the dynamic spliceosome
CRISPR-Cas9 genomic tagging combined with antibody detection:
Insert small epitope tags into the endogenous SNRPD1 locus
Use highly specific recombinant antibodies against these tags for enhanced detection sensitivity
This strategy preserves endogenous expression levels while improving detection specificity
Single-molecule RNA visualization techniques:
Combine SNRPD1 immunofluorescence with single-molecule FISH (smFISH) to visualize both the spliceosome component and its target pre-mRNAs
This approach can reveal spatial relationships between splicing machinery and specific transcripts
Mass spectrometry-based proteomics:
Use SNRPD1 antibodies for immunoprecipitation followed by mass spectrometry
Apply quantitative proteomics to compare spliceosome composition under different cellular conditions
Identify post-translational modifications on SNRPD1 and associated proteins
Cryo-electron microscopy:
Use SNRPD1 antibodies or antibody fragments to locate SNRPD1 within cryo-EM structures of spliceosome complexes
This can provide structural insights into SNRPD1's role in maintaining spliceosome architecture
These cutting-edge approaches represent the frontier of research utilizing SNRPD1 antibodies to understand fundamental mechanisms of RNA processing and their dysregulation in disease states.
Several emerging technologies hold promise for advancing SNRPD1 antibody development and applications:
AI-driven antibody design:
Machine learning algorithms can predict optimal epitopes for SNRPD1 antibody generation
Computational design of antibody sequences with improved affinity and specificity
In silico screening of antibody candidates before experimental validation
Nanobody and single-domain antibody development:
Generation of camelid-derived nanobodies against SNRPD1 for improved tissue penetration and reduced immunogenicity
These smaller antibody formats may access epitopes unavailable to conventional antibodies
Superior performance in certain applications like super-resolution microscopy
Genetically encoded intrabodies:
Development of SNRPD1-targeting scFv fragments optimized for intracellular expression
These "intrabodies" could be used to track endogenous SNRPD1 in living cells without the need for protein overexpression
Potential for acute disruption of SNRPD1 function through targeted degradation approaches
Multiplexed antibody detection systems:
Development of SNRPD1 antibodies compatible with highly multiplexed imaging techniques (CODEX, MIBI, etc.)
This would allow simultaneous visualization of SNRPD1 along with dozens of other proteins
Critical for understanding SNRPD1's role in complex cellular processes
Antibody engineering for improved tissue penetration:
Modification of SNRPD1 antibodies to enhance blood-brain barrier penetration for neuroscience applications
Development of tissue-targeted antibody variants for improved in vivo imaging
Creation of antibody fragments with enhanced tissue diffusion properties
These technologies represent promising directions for future development that could significantly enhance the utility of SNRPD1 antibodies in both basic and translational research.