SNUPN Human

Snurportin 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Role in Muscular Dystrophy

Recessive mutations in SNUPN have been identified as a cause of a novel subtype of muscular dystrophy characterized by progressive weakness, elevated creatine kinase (CK) levels, and respiratory failure. Key findings:

Pathogenic Variants and Mechanisms

Nine hypomorphic SNUPN variants were identified in 18 patients from 15 families, predominantly clustered in the last coding exon . These mutations disrupt oligomerization, leading to cytoplasmic aggregation and defective nuclear import of snRNPs .

VariantGenomic PositionProtein EffectClinical Impact
m1c.926T>G (p.Ile309Ser)Disrupted oligomerizationSevere muscle weakness, respiratory failure
m6c.902_903delAT (p.Tyr301Cysfs*29)Premature terminationEarly-onset muscle weakness, death by age 15
m9c.164G>A (p.Arg55Gln)Impaired m3G-cap bindingMilder phenotype, neuromuscular symptoms

Molecular and Cellular Consequences

Mutant SNUPN fails to transport U1 snRNPs, causing:

  • Spliceosomal dysfunction: Defective maturation of Cajal bodies and aberrant splicing of sarcolemmal genes (e.g., dystrophin, lamin A/C) .

  • Cytoskeletal disruption: Loss of muscle cell structural integrity due to misregulated actin and myosin expression .

  • ECM dysregulation: Altered extracellular matrix components, exacerbating muscle degeneration .

Role in Spinocerebellar Ataxia (SCA)

SNUPN variants have also been linked to spinocerebellar ataxia (SCA), a neurodegenerative disorder affecting Purkinje cells in the cerebellum. Key mechanisms:

  • Impaired nuclear transport: Mutant SNUPN disrupts U1 snRNP nuclear import, leading to defective splicing of genes critical for Purkinje cell development .

  • Dendritic malformation: Aberrant splicing of Shh (sonic hedgehog) and Rora (RAR-related orphan receptor α) genes impairs dendrite formation and granule cell proliferation .

Mouse Model Insights

Knock-in mice with Snupn variants recapitulate human SCA phenotypes:

PhenotypeMechanismOutcome
Defective U1 snRNP transportImpaired nuclear-cytosol shuttlingPurkinje cell dendrite atrophy
Aberrant splicingMisregulation of Shh and RoraHypoplasia of cerebellar lobes
Granule cell precursorsPremature migration and reduced proliferationCerebellar atrophy

Tissue-Specific Expression and Clinical Relevance

SNUPN exhibits nuclear membranous expression across most tissues, including skeletal muscle, brain, and cerebellum . Its broad distribution aligns with the systemic impact of SNUPN mutations.

TissueExpression PatternFunctional Implication
Skeletal MuscleHigh nuclear expressionCritical for sarcolemmal gene splicing
CerebellumPurkinje cell localizationEssential for dendrite development
LungModerate expressionLinked to respiratory failure in MD

Therapeutic Implications

Current research focuses on leveraging SMA (spinal muscular atrophy) therapies, as SNUPN interacts with SMN1 (the SMA disease gene) . Zebrafish models are being used to explore rescue strategies, such as enhancing snRNP nuclear transport or stabilizing mutant SNUPN oligomers .

Product Specs

Introduction
SNUPN, a member of the Snurportin family, functions as a nuclear import adaptor protein. It resides in both the cytoplasm and nucleus and possesses an N-terminal IBB domain along with a trimethylguanosine (m3G)-cap binding domain. SNUPN exhibits specific binding to the terminal 2,2,7-m3G-cap situated at the 5' end of U snRNPs. Its role involves facilitating the transport of U snRNPs into the nucleus through interaction with Importin β.
Description
Recombinant Human SNUPN, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 380 amino acids (specifically, residues 1-360) and exhibits a molecular weight of 43.3 kDa. A 20 amino acid His-Tag is fused to the N-terminus of SNUPN. Purification is achieved through proprietary chromatographic methods.
Physical Appearance
The product appears as a sterile, colorless solution after filtration.
Formulation
The provided solution of Human SNUPN contains 20mM Tris (pH 8), 2mM DTT, 0.1M NaCl, and 10% glycerol.
Stability
For optimal storage, refrigerate the product at 4°C if the entire vial's contents will be utilized within 2-4 weeks. For extended storage, freeze the product at -20°C. Adding a carrier protein (either 0.1% HSA or BSA) is recommended for long-term storage. It's essential to avoid subjecting the product to multiple freeze-thaw cycles.
Purity
SDS-PAGE analysis indicates a purity exceeding 90.0%.
Synonyms
KPNBL, RNUT1, Snurportin1, SPN1, RNA U transporter 1.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MEELSQALAS SFSVSQDLNS TAAPHPRLSQ YKSKYSSLEQ SERRRRLLEL QKSKRLDYVN HARRLAEDDW TGMESEEENK KDDEEMDIDT VKKLPKHYAN QLMLSEWLID VPSDLGQEWI VVVCPVGKRA LIVASRGSTS AYTKSGYCVN RFSSLLPGGN RRNSTAKDYT ILDCIYNEVN QTYYVLDVMC WRGHPFYDCQ TDFRFYWMHS KLPEEEGLGE KTKLNPFKFV GLKNFPCTPE SLCDVLSMDF PFEVDGLLFY HKQTHYSPGS TPLVGWLRPY MVSDVLGVAV PAGPLTTKPD YAGHQLQQIM EHKKSQKEGM KEKLTHKASE NGHYELEHLS
TPKLKGSSHS PDHPGCLMEN.

Q&A

What is the SNUPN gene and what is its primary function in human cells?

SNUPN (snurportin 1) is a human protein-coding gene with Entrez Gene ID 10073. It functions as an adapter protein specifically involved in the nuclear import of spliceosomal small nuclear ribonucleoproteins (snRNPs), particularly U1, U2, U4, and U5. The protein encoded by this gene interacts specifically with the 5'-2,2,7-terminal trimethylguanosine (m3G) cap structure of U snRNAs and serves as an snRNP-specific nuclear import receptor . This interaction, combined with the Sm core domain, forms a complex nuclear localization signal essential for proper nuclear import of these splicing components. The cellular process of snRNP assembly and trafficking is critical for pre-mRNA splicing and thus gene expression regulation throughout the human body.

What experimental methods are used to determine snurportin-1 localization and trafficking?

Researchers employ several methodological approaches to study snurportin-1 localization and trafficking:

  • Fluorescence microscopy techniques:

    • Immunofluorescence with antibodies against endogenous snurportin-1

    • Expression of fluorescently-tagged wild-type and mutant proteins (GFP, mCherry)

    • Fluorescence recovery after photobleaching (FRAP) to measure shuttling kinetics

  • Biochemical fractionation:

    • Subcellular fractionation to isolate nuclear and cytoplasmic components

    • Western blotting to quantify protein distribution between compartments

  • Live-cell imaging:

    • Real-time visualization of protein movement between compartments

    • Photoactivatable or photoconvertible fluorescent protein tags to track specific populations

  • Electron microscopy:

    • Immunogold labeling for high-resolution localization

    • Visualization of protein complexes at nuclear pores

These methods have revealed that mutations in snurportin-1, like those identified in spinocerebellar ataxia patients, impair the nuclear-cytosol shuttling mechanism, leading to defective nuclear transport of U1 snRNPs in cerebellar Purkinje cells .

What pathogenic variants have been identified in the SNUPN gene?

Several pathogenic variants in SNUPN have been identified in families affected by spinocerebellar ataxia:

Variant TypeNucleotide ChangeProtein ChangePredicted ImpactDetection Method
Missensec.611G>Ap.R204QDisease-causingWhole exome sequencing
Missensec.163C>Tp.R55WDisease-causingWhole exome sequencing
Frameshiftc.927dupTTruncation after M310Loss of C-terminal domainWhole exome sequencing

These variants were found in compound heterozygous states in affected individuals (c.611G>A/c.927dupT in two patients and c.163C>T/c.927dupT in another patient). Both missense mutations were predicted to be "disease causing" by computational prediction tools such as Polyphen2 and Mutation Taster . These findings establish SNUPN as a causative gene for a form of spinocerebellar ataxia with a likely autosomal recessive inheritance pattern.

How can computational tools be utilized to predict the functional impact of novel SNUPN variants?

Computational prediction of novel SNUPN variant effects involves a multi-tiered approach:

  • Sequence-based prediction:

    • Tools like PolyPhen-2 evaluate amino acid conservation and physicochemical properties

    • Mutation Taster assesses evolutionary conservation and protein domain disruption

    • SIFT (Sorting Intolerant From Tolerant) analyzes amino acid conservation in homologous sequences

  • Structural analysis:

    • Homology modeling to predict effects on protein structure

    • Molecular dynamics simulations to assess stability changes

    • Protein-protein interaction interface analysis

  • Splicing effect prediction:

    • Tools that assess potential impacts on splice site recognition or creation

    • Exonic splicing enhancer/silencer disruption analysis

  • Population frequency assessment:

    • Evaluation of variant frequency in population databases (gnomAD, 1000 Genomes)

    • Analysis of allele frequencies across different populations

These computational approaches provide an initial assessment of variant pathogenicity, which should be followed by functional validation. For SNUPN variants, researchers found that predictions using tools like PolyPhen correlated well with experimental evidence of pathogenicity in cases of spinocerebellar ataxia .

What challenges exist in interpreting SNUPN variant pathogenicity?

Researchers face several methodological challenges when determining SNUPN variant pathogenicity:

  • Functional redundancy assessment:

    • Determining whether other nuclear import pathways can compensate for SNUPN dysfunction

    • Evaluating tissue-specific dependencies on SNUPN function

  • Tissue-specific expression patterns:

    • SNUPN expression varies across tissues, complicating interpretation of systemic effects

    • Specific evaluation of expression in cerebellar Purkinje cells is crucial for ataxia-related variants

  • Compound heterozygosity analysis:

    • Determining the combined effect of two different variants in trans

    • Assessing potential synergistic or compensatory effects

  • Separating causative variants from benign polymorphisms:

    • Many rare variants may be identified without clear pathogenicity

    • Need for careful control selection and statistical analysis

  • Limited case numbers:

    • SNUPN-related disorders appear to be rare, making statistical associations challenging

    • Family studies and segregation analysis become particularly important

To address these challenges, integration of computational prediction, in vitro functional studies, animal models, and careful clinical phenotyping is essential for accurate interpretation of SNUPN variant pathogenicity .

How do pathogenic SNUPN variants disrupt normal cellular processes?

Pathogenic SNUPN variants disrupt normal cellular processes through a cascade of molecular events:

  • Impaired m3G-cap recognition:

    • Mutations in the cap-binding domain reduce affinity for the m3G-cap structure

    • This prevents efficient binding to U snRNPs in the cytoplasm

  • Defective nuclear-cytoplasmic shuttling:

    • Mutant snurportin-1 shows altered localization patterns

    • Nuclear import and export dynamics are disrupted

  • Reduced U1 snRNP nuclear import:

    • Decreased nuclear levels of U1 snRNPs, particularly in Purkinje cells

    • Imbalanced snRNP composition in the nucleus

  • Aberrant pre-mRNA splicing:

    • Widespread splicing defects affecting multiple genes

    • Both exon skipping and intron retention events increase

    • Genes essential for neuronal development are particularly affected

  • Protein expression changes:

    • Altered protein isoform ratios due to splicing changes

    • Decreased expression of proteins critical for neuronal function

This molecular cascade is particularly detrimental in cerebellar Purkinje cells, where it leads to defective dendrite formation and subsequently affects cerebellar development through altered interactions with granule cells and interneurons .

What experimental evidence links SNUPN dysfunction to splicing defects?

Multiple lines of experimental evidence connect SNUPN dysfunction to widespread splicing defects:

  • In vitro studies:

    • RNA-seq analysis of cells expressing mutant snurportin-1 reveals global alterations in splicing patterns

    • Direct measurement of U1 snRNP nuclear import efficiency shows significant reduction with mutant proteins

  • Animal model findings:

    • Snupn-variant knock-in mice show cerebellar developmental abnormalities

    • RNA-seq of cerebellar tissue from these mice demonstrates aberrant splicing of genes essential for Purkinje cell development

    • RT-PCR validation confirms specific splicing defects in development-related genes

  • Patient-derived samples:

    • Analysis of available patient tissues shows similar splicing pattern disruption

    • Correlation between severity of splicing defects and clinical phenotypes

  • Mechanistic studies:

    • Direct visualization of U1 snRNP localization shows cytoplasmic accumulation

    • Reduced nuclear U1 snRNP levels lead to inefficient 5' splice site recognition

These experimental approaches have established a clear connection between snurportin-1 dysfunction and aberrant splicing, particularly affecting genes involved in cerebellar Purkinje cell development and function .

How does neuronal cell type specificity arise in SNUPN-related disorders?

The selective vulnerability of cerebellar Purkinje cells in SNUPN-related disorders can be explained by several factors:

  • Differential expression patterns:

    • Cerebellar Purkinje cells may have higher expression or greater dependency on SNUPN

    • Alternative nuclear import pathways may be less active in these neurons

  • Complex morphological demands:

    • Purkinje cells possess the most elaborate dendritic arbors in the brain

    • This requires precise spatiotemporal regulation of gene expression and splicing

    • SNUPN dysfunction particularly impacts genes involved in dendrite formation

  • Developmental timing factors:

    • Critical periods of Purkinje cell development coincide with high dependence on proper splicing

    • Early developmental defects have cascading effects on cerebellar circuit formation

  • Secondary impacts on cerebellar architecture:

    • Purkinje cell abnormalities lead to hypoplasia and premature migration of granule cell precursors

    • Altered Sonic Hedgehog (Shh) signaling from Purkinje cells affects proliferation of granule cell precursors

    • Interneuron positioning and differentiation are secondarily affected

  • Cerebellar network effects:

    • Disrupted Purkinje cell morphology leads to abnormal synaptic connectivity

    • Altered input-output relationships in cerebellar circuits manifest as ataxia

These factors create a unique vulnerability of cerebellar Purkinje cells to SNUPN dysfunction, explaining the predominantly cerebellar phenotype in patients with SNUPN mutations .

What animal models have been developed to study SNUPN function and pathology?

Several animal models have been established to investigate SNUPN function and disease mechanisms:

  • Knock-in mouse models:

    • Mice carrying the same mutations identified in human patients (R204Q, R55W, C-terminal truncation)

    • Generated using CRISPR-Cas9 genome editing technology

    • Reproduce the key phenotypic features of human disease

  • Conditional knockout models:

    • Cell type-specific deletion of SNUPN, particularly in cerebellar Purkinje cells

    • Temporal control of gene inactivation to study developmental versus adult requirements

  • Reporter mouse lines:

    • Fluorescent protein tagging of endogenous SNUPN to monitor expression and localization

    • Combined with cell type-specific markers to study tissue distribution

  • Zebrafish models:

    • Morpholino knockdown of SNUPN orthologs

    • Transgenic lines expressing mutant forms of snurportin-1

    • Useful for high-throughput screening approaches

These models provide valuable tools for studying the molecular, cellular, and behavioral consequences of SNUPN dysfunction. The knock-in mouse models have been particularly informative, recapitulating the human disease phenotypes including motor coordination deficits and cerebellar structural abnormalities .

What biochemical approaches are optimal for studying snurportin-1 interactions?

Researchers employ several sophisticated biochemical techniques to characterize snurportin-1 interactions:

  • Pull-down assays and co-immunoprecipitation:

    • GST-tagged or His-tagged recombinant snurportin-1 to identify binding partners

    • Co-IP from cellular extracts to detect native protein complexes

    • Mutant protein comparison to identify interaction-deficient variants

  • Binding affinity measurements:

    • Surface plasmon resonance (SPR) to determine kinetic parameters of m3G-cap binding

    • Isothermal titration calorimetry (ITC) for thermodynamic binding parameters

    • Fluorescence anisotropy for solution-phase binding measurements

  • Protein-RNA interactions:

    • Electrophoretic mobility shift assays (EMSA) with labeled m3G-capped RNAs

    • UV crosslinking to detect direct protein-RNA contacts

    • CLIP-seq (cross-linking immunoprecipitation with sequencing) for genome-wide binding analysis

  • Structural analysis:

    • X-ray crystallography of snurportin-1 alone or in complex with binding partners

    • Nuclear magnetic resonance (NMR) spectroscopy for dynamic studies

    • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

  • Subcellular fractionation approaches:

    • Isolation of nuclear and cytoplasmic fractions to track protein distribution

    • Density gradient centrifugation to separate snRNP-containing complexes

These biochemical approaches have revealed that mutations in snurportin-1 can disrupt specific protein-protein or protein-RNA interactions, providing mechanistic insight into how these mutations lead to disease .

How can high-throughput screening approaches identify potential therapeutic targets for SNUPN-related disorders?

High-throughput screening methodologies offer several avenues for therapeutic target identification:

  • Small molecule screens:

    • Cell-based assays measuring nuclear import of fluorescently-tagged U1 snRNPs

    • Compounds that rescue nuclear import defects in cells expressing mutant snurportin-1

    • Secondary splicing reporter assays to confirm functional rescue

  • Genetic modifier screens:

    • CRISPR-Cas9 genome-wide or targeted screens in cellular models

    • Identification of genes that, when modulated, can compensate for SNUPN dysfunction

    • Validation in animal models of the disease

  • RNA-based screening approaches:

    • Antisense oligonucleotides targeting specific splice junctions

    • Small interfering RNAs against potential modifier genes

    • Screening for molecules that stabilize U1 snRNP interactions with pre-mRNA

  • Protein-targeting approaches:

    • Screens for stabilizers of mutant snurportin-1 folding

    • Identification of small molecules that enhance residual m3G-cap binding activity

    • Peptide-based approaches to enhance nuclear import

  • Alternative pathway modulation:

    • Screens for molecules that upregulate alternative nuclear import pathways

    • Compounds that modify downstream effects of splicing defects

These screening approaches can identify potential entry points for therapeutic intervention, which could then be developed into targeted treatments for SNUPN-related spinocerebellar ataxia .

How do SNUPN variants affect global splicing networks in Purkinje cells?

SNUPN variants exert complex effects on global splicing networks in Purkinje cells:

  • Differential impact on splice site recognition:

    • Reduced nuclear U1 snRNP primarily affects 5' splice site recognition

    • Weak 5' splice sites are particularly vulnerable to reduced U1 snRNP levels

    • Exons with suboptimal flanking splice sites show increased skipping events

  • Alternative splicing pattern shifts:

    • Altered ratios of protein isoforms due to exon inclusion/exclusion changes

    • Intron retention increases, particularly in genes with high expression levels

    • Activation of cryptic splice sites in genes essential for Purkinje cell development

  • Temporal dysregulation:

    • Developmental stage-specific splicing programs show disrupted transitions

    • Critical developmental switch points fail to occur at appropriate times

    • This affects timing of dendrite elaboration and synapse formation

  • Compensatory mechanisms:

    • Potential upregulation of other splicing factors to partially compensate

    • Changes in RNA binding protein expression to adapt to reduced U1 snRNP

  • Downstream effects on other RNA processing steps:

    • Altered polyadenylation site usage due to interconnections with splicing machinery

    • Changes in mRNA export efficiency for transcripts with retained introns

    • Increased nonsense-mediated decay for improperly spliced transcripts

These widespread splicing alterations particularly affect genes involved in dendrite morphogenesis, synaptic function, and cell-cell signaling, explaining the specific Purkinje cell defects observed in SNUPN-related disorders .

What are the temporal aspects of SNUPN function during cerebellar development?

The temporal regulation of SNUPN function during cerebellar development reveals critical windows of vulnerability:

  • Expression dynamics:

    • SNUPN expression patterns change during cerebellar development

    • Peak expression coincides with periods of intensive dendritogenesis in Purkinje cells

    • Correlation between expression levels and critical periods of circuit formation

  • Developmental transitions:

    • Early embryonic expression supports initial Purkinje cell migration and positioning

    • Postnatal upregulation corresponds with extensive dendrite elaboration

    • Continued expression maintains proper splicing homeostasis in mature neurons

  • Stage-specific requirements:

    • Different developmental stages show varying sensitivity to SNUPN dysfunction

    • Early disruption affects cell fate determination and positioning

    • Later disruption primarily affects dendrite morphogenesis and refinement

  • Interaction with developmental signaling:

    • SNUPN-dependent splicing affects Sonic Hedgehog (Shh) signaling from Purkinje cells

    • This secondarily impacts granule cell proliferation and interneuron migration

    • Creates a temporal cascade of developmental defects

  • Critical windows for intervention:

    • Early intervention may prevent cascading developmental defects

    • Later intervention may be less effective once cellular patterning is established

Understanding these temporal aspects is crucial for developing potential therapeutic interventions and identifying the optimal treatment windows for SNUPN-related disorders .

What research approaches can resolve contradictory findings about SNUPN function?

Several methodological approaches can help resolve contradictions in SNUPN research:

  • Tissue and cell-type specific analysis:

    • Single-cell transcriptomics to isolate Purkinje cell-specific effects

    • Conditional knockout/knockin models with temporal control

    • Region-specific analysis within the cerebellum to identify microenvironmental factors

  • Integrative multi-omics approaches:

    • Combined analysis of transcriptomics, proteomics, and metabolomics data

    • Integration of epigenetic profiling with splicing analysis

    • Systems biology modeling to understand network-level effects

  • Improved disease models:

    • Patient-derived induced pluripotent stem cells (iPSCs) differentiated to cerebellar organoids

    • Comparison of multiple animal models across species

    • Models that incorporate genetic background variation

  • Advanced imaging techniques:

    • Super-resolution microscopy for detailed subcellular localization

    • Long-term in vivo imaging to track developmental processes

    • Multiplexed imaging of multiple molecular markers simultaneously

  • Rigorous statistical approaches:

    • Meta-analysis of published data to identify consistent patterns

    • Power analysis to ensure adequate sample sizes

    • Bayesian approaches to integrate prior knowledge with new data

  • Collaborative validation studies:

    • Multi-laboratory replication of key findings

    • Standardized protocols and reagents

    • Data sharing and transparent reporting of negative results

These approaches can help reconcile apparently contradictory findings by accounting for context-dependent effects, methodological differences, and biological variability in SNUPN function and its role in cerebellar development .

Product Science Overview

Structure and Function

Snurportin 1 contains two key domains:

  1. N-terminal Importin Beta Binding (IBB) Domain: This domain is essential for the interaction with Importin beta, a protein that facilitates the transport of cargo proteins into the nucleus.
  2. Trimethylguanosine (m3G)-Cap Binding Domain: This domain specifically binds to the 2,2,7-trimethylguanosine cap at the 5’ end of U snRNPs .

The primary function of Snurportin 1 is to recognize and bind the m3G-cap structure of U snRNPs and mediate their transport into the nucleus through its association with Importin beta . This process is vital for the proper functioning of the spliceosome, a complex responsible for pre-mRNA splicing.

Recombinant Snurportin 1

Recombinant Snurportin 1 is produced using various expression systems, such as Escherichia coli (E. coli) and HEK-293 cells. The recombinant protein typically includes a tag, such as a His-tag or Myc-DYKDDDDK tag, to facilitate purification and detection .

  • E. coli Expression System: In this system, the recombinant Snurportin 1 is expressed as a single, non-glycosylated polypeptide chain containing 380 amino acids (1-360 a.a.) with a molecular mass of approximately 43.3 kDa. The protein is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques .
  • HEK-293 Cells Expression System: In this system, the recombinant Snurportin 1 is expressed with a Myc-DYKDDDDK tag. The protein is produced with end-sequenced ORF clone and has a purity of over 80% as determined by SDS-PAGE and Coomassie blue staining .
Applications

Recombinant Snurportin 1 is used in various research applications, including:

  • Antibody Production: As a native antigen for optimized antibody production.
  • Positive Controls: In ELISA and other antibody assays.
  • Protein-Protein Interaction Studies: To study the interactions between Snurportin 1 and other proteins involved in nuclear import and spliceosome assembly .
Storage and Handling

Recombinant Snurportin 1 should be stored at -80°C for long-term storage. It is recommended to thaw the protein on ice, aliquot it into individual single-use tubes, and then re-freeze immediately. Only 2-3 freeze-thaw cycles are recommended to maintain protein integrity .

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.