SNURF Human

SNRPN Upstream Reading Frame Human Recombinant
Shipped with Ice Packs
In Stock

Description

Genomic Context and Imprinting

SNURF is encoded by a bicistronic transcript with SNRPN, forming part of the 15q11-q13 imprinted domain. Key features include:

  • Imprinting Control:

    • The SNURF-SNRPN gene cluster is paternally expressed in somatic cells due to an imprinting control region (ICR) at the 5′ end .

    • Maternal expression of UBE3A (antisense to SNURF-SNRPN) is regulated by the IC, which silences the maternal allele .

  • Transcription Complexity:

    • The locus spans >460 kb, with 148 exons, including 11 snoRNAs (e.g., HBII-13, HBII-85) encoded within introns .

    • Alternative splicing generates multiple isoforms, including a truncated monocistronic transcript in some cases .

  • Evolutionary Conservation:

    • High conservation of SNURF across eutherians (93–100% amino acid identity), with substitutions primarily at wobble codon positions .

Expression and Tissue Distribution

SNURF is expressed in diverse tissues, with developmental and cell-specific regulation:

TissueExpressionSource
BrainDetected in fetal brain; imprinting-dependent paternal expression
HeartPresent in adult heart and skeletal muscle
LymphoblastsConsistently detected in cell lines
PancreasExpression in pancreatic cell models (ongoing research)

Key Findings:

  • Developmental Regulation: SNURF is expressed in the preimplantation embryo, with SNRPN upregulated postnatally .

  • Spermatogenesis: SNURF-SNRPN upstream transcripts are expressed in post-meiotic germ cells, regulated by epigenetic factors like MBD1 and EZH2 .

Functional Insights and Research Trends

Despite its conserved nature, SNURF’s precise function remains elusive. Recent studies focus on:

  • Translational Mechanisms:

    • SNURF and SNRPN are cotranslated from a single mRNA, with potential ribosome reinitiation or internal ribosome entry sites (IRES) .

    • Kozak consensus sequences suggest preferential translation of SNURF over SNRPN .

  • Epigenetic Regulation:

    • SNURF-SNRPN upstream transcripts may recruit chromatin-modifying enzymes to establish imprinting marks .

    • PWRN1, a novel upstream exon, is part of the SNURF-SNRPN transcript and imprinted in the brain .

  • Ongoing Research:

    • FPWR-Funded Study (2024): Investigating SNURF-SNRPN regulation in brain, heart, and pancreatic cells to identify downstream targets and stress-response pathways .

    • Protein Interactions: New tools to identify SNURF-binding partners in cellular models .

Clinical Relevance

Deletions or imprinting defects in the SNURF-SNRPN region are linked to:

DisorderMechanismSource
Prader-WilliPaternal deletion or imprinting failure; SNURF loss implicated in neonatal lethality
AngelmanMaternal deletion or UBE3A silencing due to antisense SNURF-SNRPN expression

Diagnostic Insights:

  • PWS Models: SNURF protein is absent in PWS-affected human and mouse tissues, unlike SNRPN .

  • Therapeutic Targets: Modulating SNURF-SNRPN expression or epigenetic regulators may address PWS/AS pathogenesis .

Product Specs

Introduction
SNURF, a highly basic protein found exclusively in the nucleus, is encoded by an evolutionarily conserved open reading frame. This open reading frame is situated on a bicistronic transcript, with a downstream ORF responsible for encoding the small nuclear ribonucleoprotein polypeptide N. The transcript's initial three exons are utilized by the upstream coding region, recognized as an imprinting center. While the complete length of these transcripts remains to be fully elucidated, multiple transcription initiation sites have been pinpointed, and extensive alternative splicing occurs within the 5' untranslated region. Notably, an alternative exon exists, substituting for exon 4 and leading to a truncated, monocistronic transcript. Disruptions such as deletions or alternative splicing arising from translocations in the 5' UTR or coding region of this gene can lead to Prader-Willi syndrome or Angelman syndrome due to disruptions in the parental imprint switching mechanism.
Description
Recombinant human SNURF, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 94 amino acids (1-71) with a molecular weight of 10.8 kDa. The SNURF protein includes a 23 amino acid His-tag fused to its N-terminus.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The SNURF solution is provided at a concentration of 0.25mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.15M NaCl, 1mM DTT, and 40% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product should be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein such as HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the SNURF protein is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
SNRPN Upstream Reading Frame Protein.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMERARDR LHLRRTTEQH VPEVEVQVKR RRTASLSNQE CQLYPRRSQQ QQVPVVDFQA ELRQAFLAET PRGG

Q&A

What is the SNURF-SNRPN gene and why is it significant in human genetics research?

The SNURF-SNRPN gene is a bicistronic imprinted gene located on chromosome 15 that produces two distinct proteins: SNURF (Small Nuclear Ribonucleoprotein Polypeptide N Upstream Reading Frame) and SNRPN (Small Nuclear Ribonucleoprotein Polypeptide N). This gene is particularly significant because it is deleted or inactivated in nearly all patients with Prader-Willi Syndrome (PWS), making it a critical focus for researchers studying genetic disorders . The gene's importance extends beyond its protein-coding capabilities, as it serves as a host for multiple small nucleolar RNAs (snoRNAs), including HBII-13, HBII-85, and HBII-52, as well as newly identified snoRNAs like HBII-436, HBII-437, HBII-438A, and HBII-438B . The SNURF-SNRPN sense/UBE3A antisense transcription unit spans more than 460 kb and contains at least 148 exons, making it one of the most complex gene structures in the human genome.

How does genomic imprinting affect SNURF-SNRPN expression?

Genomic imprinting of the SNURF-SNRPN gene operates through an imprinting center (IC) located at the 5' end of the gene. This IC coordinates the control of two oppositely imprinted gene clusters on human chromosome 15 . The SNURF-SNRPN gene is part of a centromeric gene cluster that is transcribed exclusively from the paternal chromosome, while genes like UBE3A and ATP10C in the telomeric cluster show preferential expression from the maternal chromosome in certain tissues . Unlike other imprinted genes in this region that feature 5' differentially methylated regions (DMRs), maternal-only expression of genes like UBE3A may be regulated indirectly through a paternally expressed antisense transcript that starts at the IC . This complex regulatory mechanism demonstrates how imprinting controls gene expression in a parent-of-origin specific manner, critical for normal development and function.

What cellular and tissue distribution patterns does SNURF show in human development?

SNURF exhibits distinct expression patterns across human tissues, with highest expression levels observed in the brain, heart, and pancreas . This tissue-specific distribution pattern suggests specialized functions in these organs and explains why SNURF-SNRPN deficiencies manifest primarily in neurological, cardiac, and metabolic symptoms in conditions like Prader-Willi Syndrome. During development, SNURF expression follows temporally regulated patterns that correlate with critical developmental windows. Research methods to study this distribution typically involve RNA-sequencing of tissue samples, immunohistochemistry, and in situ hybridization techniques to visualize expression in specific cell types and developmental stages.

What experimental designs are most effective for studying SNURF function in human cellular models?

When investigating SNURF function in human cellular models, researchers should implement true experimental designs featuring both control and experimental groups with appropriate variable manipulation . The most effective approach involves:

  • Cell Model Selection: Using laboratory cells that mimic human brain, heart, and pancreas tissues, which produce the highest levels of SNURF-SNRPN gene products .

  • Variable Definition and Control: Clearly identifying independent variables (IVs) such as SNURF expression levels or mutations, and dependent variables (DVs) such as cellular phenotypes or molecular interactions .

  • Randomization Methods: Implementing either completely randomized designs or randomized block designs to minimize the influence of confounding variables .

  • Between-Subjects vs. Within-Subjects Approaches: Determining whether to use different cell populations for each experimental condition (between-subjects) or the same population under different conditions at different times (within-subjects) .

For SNURF specifically, a combination of gene editing techniques (CRISPR-Cas9), protein interaction studies, and functional readouts provides the most comprehensive understanding. These cellular models should ideally be derived from human induced pluripotent stem cells (iPSCs) differentiated into relevant cell types to ensure physiological relevance of findings.

How should researchers approach protein interaction studies to understand SNURF function?

To effectively study SNURF protein interactions, researchers should employ a systematic methodology combining multiple complementary techniques:

  • Protein Binding Partner Identification: Implement immunoprecipitation followed by mass spectrometry to identify proteins that physically interact with SNURF . This approach should be conducted in relevant cell types, particularly those derived from brain, heart, and pancreatic tissues.

  • Validation of Interactions: Confirm identified interactions using techniques such as co-immunoprecipitation, proximity ligation assays, or fluorescence resonance energy transfer (FRET).

  • Functional Significance Assessment: Employ gene silencing or overexpression of identified interaction partners to determine their functional significance in SNURF-mediated cellular processes.

  • Structural Analysis: Where possible, characterize the structural basis of key interactions using X-ray crystallography or cryo-electron microscopy.

These approaches should be conducted in physiologically relevant cell models, with careful attention to potential artifacts introduced by protein tags or non-physiological expression levels. The goal should be to build a comprehensive protein interaction network that can inform hypotheses about SNURF's cellular functions.

What are the recommended controls when studying SNURF expression in experimental settings?

  • Negative Controls: Include cell lines or tissues where SNURF is known to be absent or expressed at minimal levels.

  • Positive Controls: Incorporate samples from tissues known to express high levels of SNURF (brain, heart, and pancreas) .

  • Technical Controls: Employ housekeeping genes (e.g., GAPDH, β-actin) for normalization in qPCR experiments, and include isotype controls for antibody-based detection methods.

  • Biological Replicates: Use multiple biological samples to account for natural variation in expression levels.

  • Developmental Stage Controls: When studying developmental expression patterns, include samples from multiple developmental timepoints to capture dynamic changes.

  • Treatment Controls: For experiments involving manipulations that might affect SNURF expression, include vehicle-only controls and time-matched untreated controls.

These controls should be systematically incorporated into experimental designs to ensure that observed changes in SNURF expression are genuinely attributable to the variables being tested rather than to technical artifacts or biological variation.

How does the bicistronic nature of SNURF-SNRPN affect experimental approaches to studying either protein individually?

The bicistronic nature of the SNURF-SNRPN gene presents unique challenges for researchers studying either protein individually. This gene produces two distinct proteins from a single mRNA transcript, requiring specialized approaches:

  • Selective Targeting Strategies: When studying SNURF or SNRPN individually, researchers must develop methods that selectively target one protein without disrupting the other. Traditional gene knockout approaches affect both proteins, necessitating more refined techniques .

  • Protein-Specific Antibodies: Developing highly specific antibodies that recognize either SNURF or SNRPN without cross-reactivity is essential for immunodetection studies.

  • Translational Regulation Analysis: Investigating the mechanisms controlling the differential translation of SNURF versus SNRPN from the same mRNA is critical. Research suggests that "a small part of the gene helps control how much of each protein is made and may also regulate the production of each protein in response to cell stress" .

  • Independent Function Assessment: To determine unique functions, researchers can use complementation studies where either SNURF or SNRPN is reintroduced into cells lacking both proteins.

  • Tissue-Specific Expression Analysis: Examining whether the ratio of SNURF to SNRPN varies across different tissues provides insights into their independent regulation and function.

This complex genetic structure necessitates multifaceted experimental approaches that can distinguish between effects attributable to SNURF versus SNRPN, requiring careful experimental design and interpretation.

What are the key considerations when analyzing SNURF's role in the regulation of other genes?

When analyzing SNURF's role in gene regulation, researchers should consider several methodological factors:

  • Comprehensive Proteome Analysis: After SNRPN deletion in laboratory cells, measuring "the types and amounts of proteins expressed from all other genes" provides insights into downstream regulatory effects . This approach requires sophisticated proteomics techniques and careful statistical analysis.

  • Direct vs. Indirect Effects: Distinguishing between direct regulatory effects of SNURF and secondary effects caused by disruption of cellular homeostasis requires time-course experiments and targeted validation studies.

  • Tissue Context Dependency: Considering that SNURF may regulate different gene sets in different tissues, analyses should be conducted in tissue-specific contexts relevant to the research question .

  • Interaction with Epigenetic Mechanisms: Investigating whether SNURF influences gene expression through interaction with epigenetic regulatory mechanisms, such as DNA methylation or histone modifications.

  • Integration with Transcriptomic Data: Combining proteomics with transcriptomics to determine whether SNURF-mediated changes occur at the transcriptional or post-transcriptional level.

These considerations help ensure that the complex regulatory networks influenced by SNURF are accurately characterized, providing a more complete understanding of its functional significance in human biology.

How can researchers effectively study the relationship between SNURF and small nucleolar RNAs (snoRNAs)?

Studying the relationship between SNURF-SNRPN and its hosted snoRNAs requires specialized methodological approaches:

  • Intronic snoRNA Processing Analysis: Investigate how snoRNAs are processed from the introns of the SNURF-SNRPN transcript. Northern blot analysis indicates that "most if not all of these snoRNAs are indeed expressed by processing from these introns" .

  • Expression Correlation Studies: Determine whether SNURF-SNRPN expression levels correlate with snoRNA levels across different tissues and conditions, which helps establish regulatory relationships.

  • Functional Interdependence Assessment: Analyze whether disruption of SNURF-SNRPN expression affects snoRNA function and vice versa, using targeted gene editing approaches.

  • Host Gene Structure Analysis: Map the exact locations of snoRNAs within the SNURF-SNRPN gene structure, as it "serves as the host for the previously identified HBII-13, HBII-85 and HBII-52 snoRNAs as well as for four additional snoRNAs (HBII-436, HBII-437, HBII-438A and HBII-438B)" .

  • Pathological Significance Exploration: Investigate whether loss of these snoRNAs contributes to disease phenotypes, as "a lack of these snoRNAs may be causally involved in [Prader-Willi syndrome]" .

These approaches provide a comprehensive framework for understanding the complex relationship between the SNURF-SNRPN host gene and its embedded snoRNAs, which may have significant implications for understanding PWS pathophysiology.

How should researchers address the challenges of studying SNURF in the context of imprinting disorders?

Studying SNURF in imprinting disorders presents unique challenges requiring specialized methodological approaches:

  • Parent-of-Origin Effect Analysis: Implement experimental designs that can distinguish maternal versus paternal contributions to SNURF expression. This requires careful pedigree analysis and parental genotyping in human studies, or specialized breeding schemes in animal models.

  • Methylation Status Characterization: Analyze the methylation status of the imprinting center (IC) at the 5' end of the SNURF-SNRPN gene, which "controls two oppositely imprinted gene clusters" . This requires techniques such as bisulfite sequencing or methylation-specific PCR.

  • Antisense Transcript Investigation: Study the complex relationship between SNURF-SNRPN sense transcription and UBE3A antisense transcription, as "maternal-only expression of UBE3A may be regulated indirectly through a paternally expressed antisense transcript" .

  • Tissue-Specific Imprinting Analysis: Examine whether imprinting of SNURF-SNRPN varies across different tissues, particularly focusing on brain, heart, and pancreas where expression is highest .

  • Development of Appropriate Cellular Models: Create cellular models that accurately reflect the imprinting status of SNURF-SNRPN, which may require patient-derived cells or specialized genome editing approaches that maintain imprinting marks.

These approaches help researchers navigate the complexities of imprinting when studying SNURF, ensuring that experimental findings accurately reflect the unique regulatory mechanisms governing this gene's expression in imprinting disorders.

What experimental approaches can resolve contradictory findings in SNURF function studies?

When facing contradictory findings in SNURF function studies, researchers should employ systematic approaches to resolve discrepancies:

  • Standardization of Experimental Systems: Implement standardized cell models and experimental conditions across research groups to minimize system-dependent variations. Focus on "laboratory cells that mimic the human brain, heart, and pancreas, which are the tissues that produce the highest levels of PWS-gene products" .

  • Meta-Analysis Approaches: Conduct formal meta-analyses of published data using statistical methods that account for inter-study heterogeneity and potential publication bias.

  • Multi-Omics Integration: Combine data from multiple molecular levels (genomics, transcriptomics, proteomics, metabolomics) to obtain a more comprehensive view of SNURF function.

  • Replication in Independent Laboratories: Establish collaborations for independent replication of key findings, using identical protocols and reagents where possible.

  • Context-Dependent Function Analysis: Systematically investigate whether SNURF functions differ depending on cell type, developmental stage, or physiological conditions, which could explain apparently contradictory findings.

  • Methodological Validation: Compare results obtained using different methodological approaches (e.g., gene silencing versus gene editing) to determine whether contradictions arise from methodological artifacts.

This systematic approach helps reconcile conflicting results and builds a more coherent understanding of SNURF function, advancing the field beyond contradictory individual findings.

How can researchers effectively study the evolutionary conservation of SNURF function across species?

To effectively study evolutionary conservation of SNURF function, researchers should employ a comprehensive comparative approach:

  • Phylogenetic Analysis: Conduct detailed phylogenetic analyses of SNURF sequences across diverse species to identify conserved domains and potential functional motifs. This helps establish the evolutionary history of SNURF and identify key functional regions under selective pressure.

  • Cross-Species Functional Complementation: Perform rescue experiments in which SNURF from different species is expressed in human cells lacking SNURF to determine functional equivalence across evolutionary distances.

  • Comparative Expression Pattern Analysis: Compare the tissue-specific and developmental expression patterns of SNURF across species to identify conserved regulatory mechanisms and expression domains.

  • Conserved Interaction Partner Identification: Identify protein interaction partners that are conserved across species, which likely represent evolutionarily ancient and functionally critical interactions.

  • Imprinting Status Comparison: Compare the imprinting status of SNURF across species to determine when this regulatory mechanism evolved and its relationship to species-specific developmental patterns.

  • Phenotypic Analysis of Orthologs: Compare phenotypes resulting from SNURF ortholog disruption across model organisms to identify conserved versus species-specific functions.

This multifaceted approach provides insights into both the core conserved functions of SNURF that emerged early in evolution and more recent functional adaptations that may be specific to certain evolutionary lineages, particularly in the context of complex developmental processes.

What statistical approaches are most appropriate for analyzing SNURF expression data across different tissues?

When analyzing SNURF expression data across different tissues, researchers should implement robust statistical methodologies tailored to the specific experimental design:

  • Normalization Strategies: Apply appropriate normalization methods to account for technical variations in sample preparation and measurement. For RNA-seq data, methods such as RPKM/FPKM or TPM should be used, while for quantitative PCR, reference gene normalization is essential.

  • Mixed-Effects Models: Implement mixed-effects statistical models that can account for both fixed effects (tissue type, experimental conditions) and random effects (individual variation, batch effects) when analyzing expression data from multiple samples and tissues.

  • Multiple Testing Correction: Apply appropriate multiple testing corrections (e.g., Benjamini-Hochberg procedure) when comparing expression across numerous tissues to control the false discovery rate.

  • Tissue-Specific Reference Ranges: Establish tissue-specific reference ranges for normal SNURF expression, accounting for factors such as developmental stage and physiological state.

  • Multivariate Analysis: Use multivariate statistical methods such as principal component analysis or hierarchical clustering to identify patterns of co-expression between SNURF and other genes across different tissues.

  • Power Analysis: Conduct a priori power analyses to determine the sample size needed to detect statistically significant differences in SNURF expression across tissues, ensuring experiments are adequately powered.

These statistical approaches ensure rigorous analysis of tissue-specific SNURF expression patterns, providing a solid foundation for interpreting biological significance and developing hypotheses about tissue-specific functions.

What are the best practices for validating SNURF antibodies for research applications?

Validating antibodies for SNURF research requires a systematic approach to ensure specificity and reliability:

  • Knockout/Knockdown Controls: Test antibodies in cells where SNURF has been knocked out or knocked down to confirm specificity. The absence of signal in these negative controls provides strong evidence for antibody specificity.

  • Overexpression Systems: Test antibodies in systems overexpressing tagged versions of SNURF to confirm detection sensitivity and correlation between antibody signal and known protein levels.

  • Multiple Antibody Comparison: Validate results using multiple antibodies targeting different epitopes of SNURF, as concordance between different antibodies increases confidence in specificity.

  • Cross-Reactivity Assessment: Test for potential cross-reactivity with related proteins, particularly SNRPN which is produced from the same bicistronic transcript .

  • Application-Specific Validation: Validate antibodies separately for each application (Western blotting, immunohistochemistry, immunoprecipitation, etc.) as performance can vary across applications.

  • Lot-to-Lot Consistency Testing: Assess consistency between different antibody lots to ensure reproducibility of results over time.

  • Mass Spectrometry Confirmation: When possible, confirm the identity of immunoprecipitated proteins using mass spectrometry to verify that the antibody is truly pulling down SNURF.

These validation steps are essential for ensuring the reliability of SNURF detection in research applications, preventing misinterpretation of results due to non-specific antibody binding or other technical artifacts.

How might single-cell technologies advance our understanding of SNURF function in development and disease?

Single-cell technologies offer revolutionary approaches to understanding SNURF's role in complex biological processes:

  • Cell-Type Specific Expression Patterns: Single-cell RNA sequencing (scRNA-seq) can reveal the precise cell types that express SNURF within heterogeneous tissues like brain, heart, and pancreas . This fine-grained resolution may uncover previously unrecognized expression patterns with functional significance.

  • Developmental Trajectory Analysis: Single-cell technologies enable researchers to map SNURF expression changes along developmental trajectories, potentially identifying critical windows where SNURF function is essential for normal development.

  • Disease-Associated Cell Population Identification: In conditions like Prader-Willi Syndrome, single-cell approaches can identify specific cell populations most affected by SNURF deficiency, providing insights into disease mechanisms.

  • Co-Expression Network Construction: Single-cell data allows construction of gene co-expression networks at unprecedented resolution, potentially revealing cell-type-specific SNURF functions and molecular interactions.

  • Spatial Context Integration: Spatial transcriptomics technologies can map SNURF expression within tissue architecture, providing insights into how spatial organization influences function.

  • Chromatin Accessibility Analysis: Single-cell ATAC-seq can reveal how SNURF expression correlates with chromatin accessibility changes, potentially uncovering regulatory mechanisms.

These approaches significantly enhance our ability to understand SNURF's complex roles in development and disease, moving beyond population-level analyses to cell-specific resolution that better captures biological complexity.

What are the methodological considerations for studying SNURF in the context of the human tumor microbiome?

Investigating SNURF in the context of human tumor microbiomes presents unique methodological challenges requiring specialized approaches:

  • Microbial-Host Gene Expression Correlation: Develop computational methods to correlate SNURF expression with specific microbial signatures identified through comprehensive tumor microbiome analysis. Such approaches should build on existing methodologies that have studied "1526 tumors and their adjacent normal tissues across seven cancer types" .

  • Contamination Control: Implement rigorous controls to distinguish true microbial signals from contamination, particularly important when studying low-biomass samples or when using sensitive molecular techniques to detect SNURF expression.

  • Multi-omics Integration: Combine host transcriptomics (including SNURF expression), microbial metagenomics, and metabolomics to build integrated models of microbiome-host interactions potentially mediated by SNURF.

  • Functional Microbial Analysis: Move beyond taxonomic profiling to understand the functional capacities of the microbiome that might influence SNURF expression, using tools like PICRUSt 2 that transform "bacterial species into functions" .

  • Spatial Distribution Analysis: Apply spatial techniques such as correlative light and electron microscopy (CLEM) to understand the spatial relationships between microbial communities and cells expressing SNURF within the tumor microenvironment .

  • Causality Testing: Design experiments that can distinguish correlation from causation in microbiome-SNURF interactions, potentially using gnotobiotic models or targeted microbial manipulations.

These methodological considerations enable researchers to rigorously investigate potential interactions between SNURF expression and the tumor microbiome, an emerging area that may reveal novel insights into cancer biology.

What ethical considerations should researchers address when conducting SNURF research using patient-derived samples?

SNURF research using patient-derived samples requires careful attention to several ethical dimensions:

  • Informed Consent Protocols: Develop comprehensive informed consent procedures that clearly explain the nature of SNURF research, potential findings, and how these might impact participants, particularly for studies involving patients with Prader-Willi Syndrome where SNURF is implicated .

  • Return of Research Results: Establish protocols for determining when and how to return individual research results to participants, especially when findings might have clinical implications.

  • Privacy and Data Protection: Implement robust data protection measures for genomic and other molecular data, recognizing the uniquely identifying nature of such information.

  • Pediatric Research Considerations: Develop special protections for research involving pediatric populations, which is particularly relevant for SNURF studies given its role in developmental disorders.

  • Equity in Sample Collection: Ensure diversity in patient recruitment to capture population-specific variations in SNURF function and expression, avoiding research that exacerbates existing healthcare disparities.

  • Long-term Sample Storage and Use: Establish clear policies regarding biobanking of samples for future SNURF research, including provisions for withdrawal of consent and limitations on commercial use.

These ethical considerations should be addressed proactively through engagement with ethics committees, patient advocacy groups, and other stakeholders to ensure SNURF research respects participant rights while advancing scientific knowledge.

How can researchers effectively translate findings from basic SNURF research into clinical applications?

Translating basic SNURF research into clinical applications requires systematic approaches that bridge the gap between fundamental science and medical practice:

  • Target Validation Pipeline: Establish a systematic pipeline for validating potential therapeutic targets identified through basic SNURF research, progressing from in vitro studies to appropriate animal models before human applications.

  • Biomarker Development: Identify and validate biomarkers related to SNURF function that could serve as diagnostics, prognostic indicators, or measures of treatment response in conditions like Prader-Willi Syndrome.

  • Phenotypic Screening Approaches: Develop phenotypic screening assays based on cellular manifestations of SNURF dysfunction to identify compounds that normalize these phenotypes without requiring detailed mechanistic understanding.

  • Patient Stratification Methods: Develop methods to stratify patients based on SNURF-related molecular profiles, enabling more personalized therapeutic approaches for conditions involving SNURF abnormalities.

  • Academic-Industry Partnerships: Establish collaborative models between academic researchers studying SNURF and industry partners with the resources to develop therapeutic applications.

  • Regulatory Pathway Planning: Early consideration of regulatory requirements for SNURF-targeting therapeutics, including engagement with regulatory agencies about novel therapeutic modalities.

These translational approaches help ensure that insights from fundamental SNURF research can be effectively developed into clinical applications that benefit patients with conditions involving SNURF dysfunction, particularly those with imprinting disorders like Prader-Willi Syndrome.

Product Science Overview

Introduction

The SNRPN Upstream Reading Frame (SNURF) is a protein encoded by the SNURF-SNRPN gene in humans. This gene is located on chromosome 15q11.2 and is known for its role in the regulation of gene expression through imprinting mechanisms . The SNURF-SNRPN gene is bicistronic, meaning it encodes two distinct proteins from a single mRNA transcript: the SNRPN upstream reading frame protein (SNURF) and the small nuclear ribonucleoprotein polypeptide N (SNRPN) .

Gene Structure and Expression

The SNURF-SNRPN gene utilizes multiple transcription initiation sites and undergoes extensive alternative splicing in its 5’ untranslated region . The upstream coding region of this gene includes the first three exons of the transcript, which have been identified as an imprinting center . This region is evolutionarily conserved and plays a crucial role in the regulation of gene expression.

Function and Localization

The SNURF protein is highly basic and localized to the nucleus . Although the exact function of SNURF is not fully understood, it is believed to be involved in RNA splicing and mRNA processing . The SNRPN protein, on the other hand, is a component of the spliceosome complex and is essential for the proper splicing of pre-mRNA .

Clinical Significance

Mutations or deletions in the SNURF-SNRPN gene can lead to several genetic disorders, including Angelman syndrome and Prader-Willi syndrome . These disorders are caused by the failure of the parental imprint switch, which results in the loss of function of the SNURF-SNRPN gene .

Research and Applications

The study of the SNURF-SNRPN gene and its encoded proteins has significant implications for understanding the mechanisms of gene imprinting and the regulation of gene expression. Human recombinant SNURF protein is used in various research applications to study its function and interactions with other proteins .

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.