SNRPA1 Human

Small Nuclear Ribonucleoprotein Polypeptide A1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to SNRPA1 Human

SNRPA1 (Small Nuclear Ribonucleoprotein Polypeptide A') is a critical protein encoded by the SNRPA1 gene in humans, functioning as a core component of the U2 small nuclear ribonucleoprotein (snRNP) complex. It plays an essential role in pre-mRNA splicing by binding stem-loop IV of U2 snRNA within the spliceosome, ensuring accurate removal of introns and ligation of exons . Dysregulation of SNRPA1 has been implicated in oncogenesis, immune modulation, and cellular responses to environmental toxins .

Protein Structure and Interactions

  • Sequence: Comprises 255 amino acids with a conserved RNA recognition motif (RRM) .

  • Expression: Produced recombinantly in Escherichia coli with >80% purity, suitable for structural and functional studies .

  • Interactions: Partners with spliceosomal components (e.g., SF3A1, SNRPC) and transcription regulators (e.g., CDC5L) .

Table 1: Key Protein Interaction Partners of SNRPA1

Partner ProteinFunctionInteraction Score
AQRRNA helicase involved in spliceosome assembly0.999
SF3A1Splicing factor for U2 snRNP stability0.999
SNRPCU1 snRNP component for 5' splice-site recognition0.999

Role in Pre-mRNA Splicing

SNRPA1 contributes to the "E" and "A" spliceosomal complexes, facilitating recognition of branch sites and ensuring fidelity in mRNA processing .

Oncogenic Signaling Pathways

  • mTOR Activation: Drives SNRPA1 overexpression in hepatocellular carcinoma (HCC), promoting tumor proliferation and inhibiting apoptosis .

  • MYC and E2F Targets: SNRPA1 upregulation enhances DNA repair and cell cycle progression in prostate cancer (PCa) .

Clear Cell Renal Cell Carcinoma (ccRCC)

  • Immune Modulation: High SNRPA1 expression associates with increased CD8+ T-cell infiltration and PD-L1 levels, yet predicts poor immunotherapy response .

  • 5-Year Survival: SNRPA1-high patients exhibit 53.67% survival vs. 73.79% in SNRPA1-low groups (p < 0.001) .

Breast Cancer Metastasis

  • Alternative Splicing: SNRPA1 drives retention of prometastatic exons (e.g., PLEC exon 31) via RNA structural recognition, enhancing invasiveness .

Chemical and Environmental Modulation

SNRPA1 expression is dynamically regulated by xenobiotics and therapeutics:

Table 2: SNRPA1 Modulators and Effects

CompoundEffect on SNRPA1Model SystemReference
Benzo(a)pyrene↑ mRNA (Mouse)In vitro
Beta-lapachone↑ mRNA, ↓ proteinHuman cells
Bisphenol A↑ mRNA, ↓ proteinHuman cells
Copper↑ ProteinRat hepatocytes

Therapeutic Implications

  • Targeted Inhibition: Silencing SNRPA1 suppresses HCC and ccRCC tumorigenesis in vivo .

  • Biomarker Potential: SNRPA1 expression levels predict resistance to anti-PD-L1 therapy in ccRCC (AUC = 0.823 for 10-year survival) .

Product Specs

Introduction
U2 small nuclear ribonucleoprotein A (SNRPA1) is a 255-amino acid protein that is part of the U2 snRNP complex. This complex, formed with U2 snRNP B (U2B), binds to the U2 snRNA hairpin IV. The specific binding of this snRNP-RNA complex is regulated by the structural arrangement of the U2 snRNP A/U2 snRNP B dimer and subtle variations in a few key residues.
Description
Recombinant human SNRPA1 is produced in E. coli. It is a single, non-glycosylated polypeptide chain consisting of 275 amino acids (1-255 a.a.) with a molecular weight of 30.5kDa. The protein has a 20 amino acid His-tag fused at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile filtered solution.
Formulation
The SNRPA1 protein solution (0.5mg/ml) is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 30% glycerol, 0.2M NaCl, 2mM DTT, and 0.1mM PMSF.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 80% as determined by SDS-PAGE analysis.
Synonyms
U2 small nuclear ribonucleoprotein A', U2 snRNP A', SNRPA1, Lea1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MVKLTAELIE QAAQYTNAVR DRELDLRGYK IPVIENLGAT LDQFDAIDFS DNEIRKLDGF PLLRRLKTLL VNNNRICRIG EGLDQALPCL TELILTNNSL VELGDLDPLA SLKSLTYLSI LRNPVTNKKH YRLYVIYKVP QVRVLDFQKV KLKERQEAEK MFKGKRGAQL AKDIARRSKT FNPGAGLPTD KKKGGPSPGD VEAIKNAIAN ASTLAEVERL KGLLQSGQIP GRERRSGPTD DGEEEMEEDT VTNGS.

Q&A

What is SNRPA1 and what is its fundamental role in RNA processing?

SNRPA1, officially known as U2 small nuclear ribonucleoprotein A', is a protein encoded by the SNRPA1 gene in humans. It functions as a critical component of the U2 small nuclear ribonucleoprotein (snRNP) complex, which is an integral part of the spliceosome machinery. The spliceosome is a multimegadalton ribonucleoprotein complex composed of small nuclear RNAs (snRNAs) and small nuclear ribonucleoproteins (snRNPs) that catalyzes the splicing of pre-mRNA into mature mRNA in eukaryotes .

The spliceosome is responsible for removing introns (non-coding regions) from pre-mRNA molecules and joining exons (coding regions) together to form a mature mRNA molecule containing the instructions to build a protein. This process is essential for gene expression in eukaryotic cells . As a component of U2 snRNP, SNRPA1 participates in the recognition and proper assembly of the spliceosome complex during the early stages of the splicing reaction.

Methodological approach: To study SNRPA1's basic function, researchers typically employ RNA immunoprecipitation techniques combined with mass spectrometry to identify its binding partners within the spliceosome complex. Structural studies using X-ray crystallography or cryo-electron microscopy are also valuable for elucidating SNRPA1's position and interactions within the U2 snRNP.

How is SNRPA1 structurally related to other spliceosomal components?

SNRPA1 is specifically a component of the U2 snRNP complex, which binds to the branch site of introns during splicing. Though our search results provide more detailed information about U1 snRNP structure than U2 snRNP, we can understand the general principles of snRNP organization. The spliceosome consists of five main snRNP particles (U1, U2, U4, U5, and U6), each containing specific snRNAs and associated proteins .

SNRPA1 has been shown to interact with CDC5L, another spliceosomal component . This interaction is part of the complex network of protein-protein and RNA-protein interactions that facilitate proper splicing. Similar to how U1-C protein in U1 snRNP contributes to the recognition of the 5' splice site, SNRPA1 likely contributes to the recognition and binding of U2 snRNP to the branch point sequence in pre-mRNA .

Methodological approach: Co-immunoprecipitation experiments followed by Western blotting can confirm SNRPA1's interaction with CDC5L and potentially identify additional interaction partners. Researchers can also use protein crosslinking followed by mass spectrometry to map the precise interaction surfaces between SNRPA1 and other spliceosomal components.

What evidence links SNRPA1 to hepatocellular carcinoma development?

Functional studies provide compelling evidence for SNRPA1's oncogenic role:

  • Proliferation effects: Knockdown of SNRPA1 inhibits cell proliferation, colony formation, and xenografted tumorigenesis of HCC cells both in vitro and in vivo .

  • Apoptosis regulation: SNRPA1 down-regulation induces apoptosis in HCC cells, indicating its role in cancer cell survival .

  • Pathway analysis: SNRPA1 is stimulated by mTOR activation, suggesting it functions downstream of this critical oncogenic pathway .

Methodological approach: Researchers investigating SNRPA1's role in HCC should consider using siRNA or shRNA knockdown approaches in HCC cell lines, followed by functional assays including cell viability, colony formation, and apoptosis assays. Xenograft models in immunodeficient mice can validate in vitro findings in a more physiologically relevant context.

What molecular mechanisms underlie SNRPA1's oncogenic activity?

Whole-genome microarray analysis has identified substantial gene expression changes associated with SNRPA1 knockdown in HCC cells, with 262 genes being up-regulated and 462 genes down-regulated . This suggests SNRPA1 has widespread effects on the transcriptome, potentially through its role in splicing regulation.

Specific genes affected by SNRPA1 knockdown include:

Gene CategoryGenes Down-regulated After SNRPA1 KnockdownGenes Up-regulated After SNRPA1 Knockdown
Growth FactorsFibroblast growth factor-2 (FGF2)-
Tumor MarkersAlpha-fetoprotein (AFP)-
Signaling Moleculesβ-catenin-
Proliferation MarkersKi-67, Cyclin B1-
Tumor Suppressors-p53, p21
Apoptosis Regulators-Caspase 3

The regulation of these genes provides insight into how SNRPA1 might promote HCC development through multiple mechanisms:

  • Enhancement of cell proliferation via FGF2, Ki-67, and cyclin B1

  • Activation of β-catenin signaling, which is frequently dysregulated in HCC

  • Suppression of tumor suppressors p53 and p21

  • Inhibition of apoptosis by downregulating caspase 3

Methodological approach: Researchers should perform RNA-sequencing after SNRPA1 modulation to identify splicing alterations in addition to gene expression changes. ChIP-seq experiments can determine if SNRPA1 has any direct transcriptional regulatory functions. Western blotting and RT-qPCR should be used to validate key target gene expression changes at protein and mRNA levels.

What are the most effective methods for studying SNRPA1 expression patterns across tissues?

Understanding SNRPA1 expression patterns across different tissues and disease states is crucial for characterizing its role in human biology and pathology. Several complementary approaches can be employed:

  • Immunohistochemistry (IHC): This technique allows visualization of SNRPA1 protein expression in tissue sections. The search results indicate that IHC was likely used to compare SNRPA1 expression between HCC tissues and normal liver tissues .

  • Western blotting: For quantitative assessment of SNRPA1 protein levels across different tissue samples or experimental conditions.

  • RT-qPCR: To measure SNRPA1 mRNA expression levels with high sensitivity.

  • RNA-sequencing data mining: Analysis of publicly available RNA-seq datasets from projects like GTEx, TCGA, or single-cell RNA-seq studies can provide insights into SNRPA1 expression patterns across diverse tissue types and disease states.

  • Transcriptional analysis: Methods to study the transcription of snRNA genes, including SNRPA1, have been developed to distinguish genuinely expressed genes from those prone to false identification due to repetitive sequences and bioinformatic misalignment .

Methodological approach: Researchers should consider combining multiple techniques for a comprehensive assessment. For instance, mining public databases can identify tissues of interest, followed by validation using RT-qPCR and Western blotting on tissue samples, with IHC providing spatial information about SNRPA1 expression within tissues.

How can researchers effectively modulate SNRPA1 function for mechanistic studies?

Modulating SNRPA1 expression or function is essential for investigating its mechanistic roles. Several approaches have proven effective:

  • RNA interference (RNAi): siRNA or shRNA targeting SNRPA1 has been successfully used to knockdown its expression in HCC cells, leading to observable phenotypic changes .

  • CRISPR-Cas9 genome editing: For generating SNRPA1 knockout cell lines or introducing specific mutations to study structure-function relationships.

  • Overexpression systems: Plasmid-based overexpression of wild-type or mutant SNRPA1 can complement loss-of-function studies.

  • Inducible expression systems: Tetracycline-inducible or other inducible systems allow temporal control of SNRPA1 expression.

  • Inhibitor development: Though not mentioned in the search results, developing small molecule inhibitors that disrupt SNRPA1's interaction with other spliceosomal components could provide alternative approaches to modulate its function.

Methodological approach: A comprehensive study would employ both loss-of-function (RNAi or CRISPR knockout) and gain-of-function (overexpression) approaches in relevant cell types. For cancer studies, patient-derived xenograft models following SNRPA1 modulation can provide translational insights.

How does SNRPA1 contribute to alternative splicing regulation in cancer?

While the search results don't directly address SNRPA1's role in alternative splicing regulation, its function as a component of U2 snRNP implies involvement in splice site selection. Given that dysregulation of alternative splicing is a hallmark of cancer, SNRPA1's up-regulation in HCC likely contributes to splicing alterations that promote tumorigenesis.

The spliceosome catalyzes the splicing of pre-mRNA molecules by removing introns and joining exons . U2 snRNP, which contains SNRPA1, plays a crucial role in recognizing the branch site during splicing. Alterations in SNRPA1 expression or function could affect:

  • Branch point recognition efficiency

  • Splice site selection in contexts with competing splice sites

  • Exon inclusion/exclusion rates

  • Recognition of weak splice sites

Methodological approach: To investigate SNRPA1's role in alternative splicing, researchers should perform RNA-seq with focused analysis on alternative splicing events (exon skipping, alternative 5' or 3' splice sites, intron retention) following SNRPA1 modulation. Minigene splicing assays for specific candidate alternative splicing events can provide mechanistic validation. RNA-protein interaction studies (CLIP-seq) can identify direct RNA binding targets of SNRPA1 within the transcriptome.

What is the relationship between mTOR signaling and SNRPA1 expression?

The search results indicate that SNRPA1 is stimulated by mTOR activation . This connection between mTOR signaling and SNRPA1 is particularly significant given mTOR's central role in regulating cell metabolism, growth, proliferation, and survival.

The mechanistic details of how mTOR regulates SNRPA1 expression are not fully elaborated in the search results, but several possibilities exist:

  • Transcriptional regulation: mTOR could promote SNRPA1 gene transcription through downstream transcription factors.

  • Post-transcriptional regulation: mTOR might affect SNRPA1 mRNA stability or translation efficiency.

  • Post-translational regulation: mTOR signaling could modulate SNRPA1 protein stability or activity through phosphorylation.

Methodological approach: Researchers investigating this relationship should treat cells with mTOR inhibitors (e.g., rapamycin, torin) and assess SNRPA1 expression at both mRNA and protein levels. Time-course experiments would help determine whether the regulation is direct or indirect. Phosphoproteomic analysis could identify potential mTOR-dependent phosphorylation sites on SNRPA1. Luciferase reporter assays using the SNRPA1 promoter could determine if mTOR regulates SNRPA1 at the transcriptional level.

How might SNRPA1 serve as a therapeutic target in cancer?

Given SNRPA1's oncogenic properties in HCC, it represents a potential therapeutic target. Several approaches could be explored:

  • Direct targeting: Development of small molecules that specifically inhibit SNRPA1 function or its interaction with critical binding partners.

  • RNA-based therapeutics: siRNAs or antisense oligonucleotides targeting SNRPA1 mRNA could reduce its expression in cancer cells.

  • Indirect targeting: Since SNRPA1 is regulated by mTOR signaling, combining mTOR inhibitors with other cancer therapies might enhance efficacy against tumors with high SNRPA1 expression.

  • Synthetic lethality: Identifying genes that, when inhibited alongside SNRPA1, cause synergistic cancer cell death could provide novel therapeutic combinations.

Methodological approach: High-throughput screening of chemical libraries could identify compounds that specifically inhibit SNRPA1 function. Drug combination screens in cells with SNRPA1 knockdown could reveal synthetic lethal interactions. Patient-derived xenografts or organoids would provide platforms for preclinical evaluation of SNRPA1-targeting strategies.

What unresolved questions remain about SNRPA1's fundamental biology?

Despite the insights provided by current research, several fundamental questions about SNRPA1 remain unanswered:

  • Structural determinants: What specific structural features of SNRPA1 contribute to its function within U2 snRNP and how do they facilitate interactions with other spliceosomal components?

  • Tissue-specific functions: Does SNRPA1 have tissue-specific roles or expression patterns that might explain its particular importance in liver cancer?

  • Splicing specificity: Does SNRPA1 contribute to the recognition of specific types of introns or regulate particular subsets of alternative splicing events?

  • Post-translational modifications: How is SNRPA1 function regulated by post-translational modifications, and do these modifications change in cancer?

  • Evolutionary conservation: How conserved is SNRPA1 structure and function across species, and what can this tell us about its fundamental importance?

Methodological approach: Advances in cryo-electron microscopy could elucidate SNRPA1's position and interactions within the assembled spliceosome at different stages of the splicing reaction. Tissue-specific knockout mouse models would help understand SNRPA1's role in different physiological contexts. Single-molecule imaging techniques could provide insights into the dynamics of SNRPA1 within the spliceosome during the splicing reaction.

Product Science Overview

Introduction

Small Nuclear Ribonucleoprotein Polypeptide A1 (SNRPA1) is a crucial component of the spliceosomal U1 small nuclear ribonucleoprotein (snRNP) complex. This protein plays a significant role in the splicing of pre-mRNA, a critical process in the post-transcriptional modification of RNA in eukaryotic cells .

Structure and Function

SNRPA1 is part of the U1 snRNP, which is essential for recognizing the 5’ splice site of precursor mRNAs. This recognition is the first step in the assembly of the spliceosome, a large RNA-protein complex responsible for removing introns from pre-mRNA . The U1 snRNP binds to the 5’ splice site and facilitates the subsequent binding of U2 snRNP and the U4/U6/U5 tri-snRNP, which are necessary for the splicing process .

The protein encoded by the SNRPA1 gene binds to stem loop II of the U1 snRNA. In its snRNP-free form, it may be involved in the coupled pre-mRNA splicing and polyadenylation process . It has been suggested that SNRPA1 binds preferentially to the 5’-UGCAC-3’ motif on RNAs .

Biological Significance

The proper functioning of SNRPA1 is vital for the accurate splicing of pre-mRNA. Any mutations or malfunctions in this protein can lead to various diseases, including mixed connective tissue disease and lymphangiosarcoma . The protein’s role in splicing and polyadenylation highlights its importance in gene expression regulation and RNA processing.

Recombinant SNRPA1

Recombinant SNRPA1 is produced using recombinant DNA technology, which involves inserting the SNRPA1 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, including studies on RNA splicing, gene expression regulation, and the development of therapeutic interventions for diseases associated with splicing defects.

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 2024 Thebiotek. All Rights Reserved.