SNRPD2 is a 118-amino-acid protein with a molecular mass of 14,773 Dalton when expressed in Sf9 insect cells or 15.6 kDa in E. coli . Key characteristics include:
SNRPD2 contains a conserved Sm domain critical for binding snRNA and forming the heptameric Sm core complex .
As part of the U1, U2, U4, and U5 snRNPs, SNRPD2 facilitates spliceosome activation and catalytic steps .
Required for both major (U2-dependent) and minor (U12-dependent) spliceosomes .
Depletion of SNRPD2 in hepatocellular carcinoma (HCC) cells disrupts splicing of DNA repair genes (e.g., BRCA1, FANCA), leading to:
Acetylation of SNRPD2 modulates spliceosome activity under DNA damage, linking splicing regulation to HCC progression .
Binds IgG autoantibodies in SLE patient sera, validated via ELISA and Western blot .
Optimal coating concentration: 0.4–0.8 µg/ml for diagnostic assays .
Upregulated in HCC tissues vs. normal liver (p < 0.001), correlating with poor prognosis .
Knockdown reduces HCC cell proliferation by 60–70% in 3D cultures and suppresses tumorigenesis in murine models .
CRISPR/Cas9-mediated SNRPD2 knockout in MYC/p53-driven HCC mice extends median survival from 41.5 days (controls) to >70 days (experimental group) .
Targeted by Salmonella ubiquitin ligase SlrP, which modifies lysine residues (e.g., K53, K55) without altering protein stability .
Splicing Dysregulation in HCC:
Acetylation-Dependent Splicing Control:
Species-Specific Essentiality:
Investigate SNRPD2 acetylation as a therapeutic vulnerability in spliceosome-addicted cancers.
Develop SNRPD2-targeted diagnostics for early HCC detection using autoantibody profiling.
SNRPD2 encodes the SmD2 protein, a core component of the spliceosome Sm proteins. As part of the small nuclear ribonucleoproteins (snRNPs), SmD2 plays an essential role in pre-mRNA splicing processes. Beyond its canonical role in mRNA processing, recent research indicates that SmD2 also participates in coordinated protein production and mitotic processes, suggesting multifunctional involvement in cellular homeostasis . Methodologically, researchers can confirm these functions through RNA-protein interaction studies, co-immunoprecipitation experiments, and functional genomics approaches.
Transcriptomic analyses reveal that SNRPD2 maintains differential expression patterns across human tissues. Non-malignant cell cultures typically demonstrate approximately 3-fold lower SNRPD2 expression compared to cancer cells, particularly melanoma cultures . Research methodologies for investigating this differential expression include RNA-seq, RT-qPCR validation, and tissue-specific expression analysis using resources such as The Human Protein Atlas, which provides comprehensive tissue expression data through its BRAIN, TISSUE, and SINGLE CELL databases .
SmD2 functions within a highly ordered protein network in the spliceosome. Structurally, it forms direct interactions with other Sm proteins (SmB/B', SmD1, SmD3, SmE, SmF, and SmG) to create the heptameric ring that binds to snRNAs . Researchers can investigate these interactions using protein structure modeling, yeast two-hybrid screens, and mass spectrometry-based interactome analysis. The Protein Atlas resource provides valuable structural and interaction data to inform experimental designs targeting these protein-protein interactions .
Pan-cancer analysis using TCGA and GTEx datasets reveals significant SNRPD2 overexpression in almost all solid tumor types compared to matched normal tissues, with the rare exception of chromophobe renal cell carcinoma . This expression pattern is consistent across all seven Sm genes. To investigate this differential expression, researchers should employ integrated bioinformatic approaches combining RNA-seq data analysis with statistical validation across large patient cohorts.
Cancer cells exhibit selective vulnerability to SNRPD2 silencing that normal cells do not share. RNAi screening data indicate that cancer cell lines with higher SNRPD2 expression show increased sensitivity to its knockdown (Pearson correlation r=0.23, p<0.0001) . This suggests cancer cells develop a dependency on elevated SmD2 levels, potentially due to heightened splicing demands or reliance on non-canonical functions. Experimentally, this differential vulnerability can be investigated using paired cancer/normal models subjected to carefully titrated knockdown approaches.
Lentiviral vectors expressing short hairpin RNA (shRNA) targeting SNRPD2 have proven effective for gene silencing. As demonstrated in multiple studies, transduction with LV-shSNRPD2 at approximately 4000 genome copies per cell achieves 50-80% knockdown efficiency across various cell types . For complete gene knockout, CRISPR-Cas9 approaches yield more profound effects, as evidenced by significantly lower dependency scores in CRISPR screens (median Chronos score -2.10) compared to RNAi screens (median score -0.97) . Researchers should consider experimental goals when choosing between these methodologies, as they provide different insights into gene dependency.
Quantitative reverse transcription PCR (RT-qPCR) represents the standard approach for validating SNRPD2 knockdown at the transcript level. Typically, knockdown confirmation should be performed approximately 3 days post-transduction . For protein-level validation, western blotting with SmD2-specific antibodies is recommended. Additionally, functional readouts of splicing activity, such as exon-inclusion assays, can provide insight into the biological consequences of reduced SmD2 levels.
CellTiter-Blue viability assay has been successfully employed to measure cellular responses to SNRPD2 silencing one week post-transduction . This approach allows quantitative assessment of metabolic capacity as a proxy for cell viability. Complementary assays, including Annexin V/PI staining for apoptosis detection and cell cycle analysis by flow cytometry, provide mechanistic insights into how SNRPD2 depletion affects cancer versus non-malignant cells.
Gene set enrichment analysis (GSEA) reveals that SNRPD2 expression positively correlates with several oncogenic pathways, including MYC and E2F target genes, oxidative phosphorylation, G2M checkpoint regulation, DNA repair mechanisms, and mTORC1 signaling . Conversely, UV response and inflammatory signaling pathways exhibit negative correlation with SNRPD2 expression. These pathway associations suggest that SNRPD2 upregulation may be integrated with broader oncogenic programs that drive cancer progression.
Despite both being subtypes of non-small cell lung cancer (NSCLC), LUAD and LUSC demonstrate striking differences in SNRPD2 expression patterns and their clinical implications . While high SNRPD2 expression correlates with poor outcomes in LUAD, it appears to be a protective marker in LUSC. This discrepancy may relate to differential pathway interactions, as LUSC exhibits strong negative correlations between SNRPD2 expression and epithelial-mesenchymal transition (EMT) and interferon response gene sets that are not observed in LUAD . Researchers investigating SNRPD2 in lung cancer should carefully differentiate between these histological subtypes.
While the search results don't explicitly address epigenetic regulation of SNRPD2, related research on spliceosome components suggests potential regulatory mechanisms. Methodologically, researchers can investigate SNRPD2 regulation through chromatin immunoprecipitation (ChIP) studies targeting histone modifications, DNA methylation analysis of the SNRPD2 promoter region, and evaluation of transcription factor binding sites. These approaches would help elucidate the molecular basis for SNRPD2 overexpression in cancer.
Multiple lines of evidence establish SNRPD2 as a promising cancer-selective target. First, silencing SNRPD2 effectively kills established cancer cell lines and primary melanoma cultures while sparing non-malignant cells . This selective toxicity was demonstrated across diverse normal cell types, including endothelial cells, stellate cells, lung fibroblasts, and cancer-associated fibroblasts from NSCLC resections, none of which showed decreased viability following 50-80% SNRPD2 knockdown . The therapeutic window appears to be related to differential dependency rather than expression levels alone.
SNRPD2 expression levels correlate with cancer cell responses to several FDA-approved anti-tumor drugs, particularly those targeting cell cycle processes . This suggests potential synergistic applications where SNRPD2-targeting approaches could enhance conventional chemotherapy efficacy. Researchers can explore these relationships using drug sensitivity databases coupled with expression data to identify optimal combination strategies.
The heightened dependency of cancer cells on SNRPD2 appears to involve both canonical and non-canonical functions. Beyond mRNA splicing, SNRPD2 participates in coordinated protein production and mitotic processes . Analysis of genes with similar essentiality profiles reveals that SmD2 loss creates intersecting lethal stress with loss of gene products involved in multiple mRNA processing steps as well as mitosis regulation . This suggests that cancer cells, with their elevated metabolic and proliferative demands, become particularly vulnerable to perturbations in these SmD2-dependent processes.
SNRPD2 silencing produces distinct alternative splicing switches that demonstrate cancer selectivity. Previous studies with related Sm proteins (SmB/B' and SmD3) in NSCLC revealed cancer-specific splicing alterations in the ADD3 gene, switching from a cancer-associated variant to one predominantly found in non-malignant tissues . Additionally, silencing of any Sm gene induced cytotoxic splicing switches in the PSMB3 gene specifically in cancer cells . Researchers investigating these phenomena should employ RNA-seq with splicing-aware analysis algorithms followed by experimental validation of candidate events using PCR-based approaches.
Analysis of genes with essentiality profiles similar to SNRPD2 uncovers connections to mitotic processes beyond the expected mRNA processing functions . This suggests that SmD2 may either directly participate in mitotic regulation or indirectly influence mitosis through specialized splicing events affecting cell cycle regulators. Experimental approaches to investigate this relationship include cell synchronization experiments, mitotic phenotype analysis following SNRPD2 depletion, and identification of mitotic substrates that undergo SmD2-dependent splicing.
GSEA reveals negative correlations between SNRPD2 expression and inflammatory response gene sets , suggesting potential antagonistic relationships. This interaction may be particularly relevant to understanding differential SNRPD2 roles in cancer subtypes such as LUAD versus LUSC, where interferon response signatures show distinct correlations with SNRPD2 . Methodologically, researchers can explore these interactions through cytokine stimulation experiments, evaluation of SNRPD2 expression and function under inflammatory conditions, and assessment of inflammatory marker changes following SNRPD2 manipulation.
SNRPD2 is essential for the biogenesis of small nuclear ribonucleoproteins and the splicing of pre-mRNA . It is a core component of the spliceosomal U1, U2, U4, and U5 snRNPs, which are the building blocks of the spliceosome . The spliceosome is responsible for removing introns from pre-mRNA, a critical step in the maturation of messenger RNA (mRNA) molecules .
Human recombinant SNRPD2 is produced using recombinant DNA technology, which involves inserting the SNRPD2 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in various research applications to study its function, interactions, and role in disease.