SYF2 Human

SYF2 RNA splicing factor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to SYF2 Human

SYF2 (SYF2 Homolog, RNA Splicing Factor) is a nuclear protein encoded by the SYF2 gene, also known as p29, CBPIN, or NTC31. It functions as a critical regulator in pre-mRNA splicing, cell cycle progression, and DNA damage repair . SYF2 interacts with spliceosome components (e.g., Prp19 complex) and cell cycle regulators (e.g., cyclin D-type binding proteins), positioning it as a multifunctional player in cellular homeostasis and disease pathogenesis . Its dysregulation is implicated in cancers and neurodegenerative disorders, notably amyotrophic lateral sclerosis (ALS) .

Gene and Protein Structure

  • Gene location: Human SYF2 is located on chromosome 1p36.31 .

  • Exon-intron organization: Varies across species; human SYF2 contains 6–9 exons, with conserved splicing-related motifs .

  • Protein domains: Includes tetratricopeptide repeats (TPRs) for protein-protein interactions and a conserved C-terminal domain critical for spliceosome assembly .

Interactions

  • Cyclin-D-type binding protein 1: Mediates G1/S phase transition .

  • Prp19 complex (Prp19C): Facilitates splicing and transcription elongation .

  • TDP-43: Linked to ALS pathology via spliceosome regulation .

Cell Cycle Regulation

SYF2 drives G1/S phase transition by interacting with cyclin D1-CDK4/6, promoting cell proliferation. Knockdown induces G1/S arrest, inhibiting cancer growth .

Pre-mRNA Splicing

As part of the NineTeen Complex, SYF2 ensures spliceosome integrity and regulates alternative splicing events (e.g., STMN2 in ALS) .

DNA Replication and Repair

SYF2 modulates DNA replication checkpoints via splicing-dependent regulation of ECT2 isoforms, impacting S-phase progression .

Cancer

SYF2 is overexpressed in multiple malignancies, correlating with poor prognosis:

Cancer TypeKey FindingsReferences
Breast CancerHigh SYF2 levels linked to tumor grade, Ki-67 expression, and poor survival
Ovarian CancerSYF2 promotes proliferation; knockdown inhibits cell growth
Hepatocellular CarcinomaSYF2 drives cisplatin resistance and metastasis

Neurodegeneration

  • ALS: SYF2 suppression reduces TDP-43 cytoplasmic aggregation, restores nuclear TDP-43, and rescues motor neuron degeneration in C9ORF72 and sporadic ALS models .

  • Therapeutic potential: SYF2-targeting antisense oligonucleotides (ASOs) improve motor function in mice .

Tissue-Specific Expression

  • High expression: Human spleen, lung, reproductive organs; mouse cerebellum and thymus .

  • Developmental stages: Enriched in human fetal/juvenile stages and mouse embryonic/postpartum periods .

Disease-Associated Expression

ConditionSYF2 Expression Pattern
Breast CancerUpregulated in tumor lumen, HER2+ subtypes
ALSElevated in degenerating motor neurons

Cancer Therapeutics

  • SYF2 knockdown inhibits proliferation in breast, ovarian, and liver cancer cells .

  • Correlates with chemoresistance, suggesting utility as a biomarker .

Neurodegenerative Therapies

  • SYF2 suppression: Reduces TDP-43 pathology and extends survival in ALS models .

  • Drug development: AcuraStem is advancing SYF2-targeted ASOs to preclinical trials .

Research Gaps and Future Directions

  • Mechanistic insights: SYF2’s dual roles in splicing and cell cycle regulation require further elucidation.

  • Therapeutic optimization: Dose-dependent effects of SYF2 suppression in ALS need validation in human trials .

  • Cross-species variability: Human-mouse expression disparities warrant deeper analysis for translational relevance .

Product Specs

Introduction
SYF2, also known as Pre-mRNA-splicing factor SYF2, is a protein that may play a role in pre-mRNA splicing. This protein interacts with cyclin D-type binding-protein 1, a protein thought to regulate the cell cycle at the G1/S transition. SYF2 is highly expressed in the heart, skeletal muscle, and kidney, with lower expression levels in other tissues.
Description
Recombinant SYF2 protein, produced in E.coli, is a single, non-glycosylated polypeptide chain comprising 173 amino acids (94-243). It has a molecular weight of 20.5 kDa. The protein is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless, and clear solution.
Formulation
The SYF2 solution is provided at a concentration of 0.25 mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 50% glycerol, 0.2M NaCl, 5mM DTT, and 2mM EDTA.
Stability
For short-term storage (2-4 weeks), keep the SYF2 vial refrigerated at 4°C. For long-term storage, freeze the vial at -20°C. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
The purity of the SYF2 protein is greater than 90.0%, as determined by SDS-PAGE analysis.
Synonyms
Pre-mRNA-splicing factor SYF2, CCNDBP1-interactor, p29, SYF2, CBPIN, GCIPIP, NTC31, fSAP29.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSDYEKVKL LEISAEDAER WERKKKRKNP DLGFSDYAAA QLRQYHRLTK QIKPDMETYE RLREKHGEEF FPTSNSLLHG THVPSTEEID RMVIDLEKQI EKRDKYSRRR PYNDDADIDY INERNAKFNK KAERFYGKYT AEIKQNLERG TAV.

Q&A

What is SYF2 and what is its primary function in human cells?

SYF2 is a pre-mRNA splicing factor that belongs to the NineTeen Complex, a collection of proteins recruited to the spliceosome to regulate conformational changes required for the two steps of splicing . It plays critical roles in:

  • Pre-mRNA processing and splicing regulation

  • Cell cycle progression through interaction with cyclin-D-type binding protein 1

  • Potential modulation of TDP-43 localization and activity in neurons

Methodologically, researchers typically study SYF2 function through gene knockdown experiments using antisense oligonucleotides (ASOs) or RNA interference approaches, followed by functional assays examining splicing patterns, cell cycle progression, or in the context of neurodegeneration, motor neuron survival.

How is SYF2 expression regulated during human development?

Human expression profiling data indicates significant enrichment of SYF2 during both fetal and juvenile developmental stages . This developmental expression pattern suggests SYF2 may play important roles in:

  • Embryonic and early postnatal neuronal development

  • Cellular differentiation processes

  • Tissue-specific splicing regulation during development

To properly investigate developmental expression patterns, researchers should employ techniques such as RT-qPCR across developmental timepoints, RNA-seq for transcriptome-wide analysis, and immunohistochemistry on developmental tissue samples. Time-course experiments examining SYF2 expression alongside developmental markers provide valuable insights into its developmental regulation.

What cellular pathways are directly influenced by SYF2 activity?

Based on current research, SYF2 influences several key cellular pathways:

  • Spliceosome assembly and pre-mRNA processing

  • TDP-43 localization and aggregation in motor neurons

  • Cell cycle progression, particularly affecting proliferation rates

  • Potentially exocytosis and cellular waste clearance mechanisms similar to PIKFYVE inhibition effects

To investigate these pathways methodologically, researchers should employ pathway analysis following SYF2 manipulation, combining transcriptomic approaches with functional assays specific to each pathway. For splicing analysis, RNA-seq with junction analysis and splicing-sensitive RT-PCR are recommended.

What experimental approaches best assess SYF2's role in TDP-43 pathology in ALS models?

Several sophisticated experimental approaches can effectively evaluate SYF2's impact on TDP-43 pathology:

  • Induced Motor Neuron (iMN) Models: Patient-derived induced pluripotent stem cells (iPSCs) differentiated into motor neurons provide an excellent platform for studying SYF2 suppression effects on TDP-43 localization . These models should include both C9ORF72 and sporadic ALS patient lines.

  • Quantitative Nuclear/Cytoplasmic TDP-43 Ratio Analysis: Following SYF2 suppression, researchers should perform high-resolution imaging with nuclear/cytoplasmic fractionation to quantify changes in TDP-43 localization .

  • Cryptic Exon Inclusion Analysis: Measuring cryptic exon inclusion in TDP-43 substrates such as STMN2 via RT-PCR and RNA-seq provides valuable functional readouts of TDP-43 activity .

  • In vivo TDP-43 Transgenic Models: TDP-43-overexpressing mice treated with SYF2 ASOs should be assessed for motor function, neuromuscular junction integrity, and neurodegeneration markers .

Methodologically, it's critical to employ multiple model systems and readouts simultaneously, as TDP-43 pathology manifests through several mechanisms. Longitudinal studies tracking individual neurons over time provide particularly valuable insights into the progressive nature of ALS pathology.

How can researchers distinguish between direct and indirect effects of SYF2 suppression on neuronal survival?

Distinguishing direct from indirect effects requires sophisticated experimental design:

  • Temporal Analysis: Implement time-course experiments following SYF2 suppression, tracking immediate transcriptional changes (0-24h) versus later phenotypic outcomes (days-weeks) .

  • Mechanistic Dissection: Perform rescue experiments by expressing SYF2 mutants lacking specific domains or interaction capabilities to identify which functions are essential for neuroprotection.

  • Single-Cell Multi-Omics: Combine single-cell RNA-seq with proteomics following SYF2 manipulation to connect transcriptional changes with protein-level alterations.

  • Pathway Inhibition Studies: Systematically block potential downstream pathways (TDP-43 regulation, splicing alterations) to determine which are necessary for SYF2-mediated neuroprotection.

When interpreting results, researchers should be cautious about potential confounding effects from compensatory mechanisms activated following chronic SYF2 suppression. Short-term inducible systems can help mitigate these concerns.

What are the optimal parameters for ASO-mediated SYF2 suppression in preclinical models?

For effective ASO-mediated SYF2 suppression, researchers should consider:

  • Dose Optimization: Multiple SYF2 ASOs demonstrate improved C9ORF72 and sporadic ALS iMN survival in a dose-dependent manner, with increasing efficacy up to 9 μM in vitro .

  • ASO Chemistry Selection: Compare phosphorothioate, morpholino, and 2'-O-methoxyethyl (MOE) ASO chemistries for optimal CNS tissue penetration and minimal off-target effects.

  • Delivery Route Considerations: For in vivo models, compare intrathecal, intraventricular, and intranasal delivery routes, assessing CNS distribution and target engagement.

  • Temporal Treatment Regimens: Determine whether pre-symptomatic treatment, treatment at symptom onset, or late-stage intervention provides therapeutic benefit.

Researchers should monitor SYF2 suppression efficiency through RT-qPCR and western blotting, while concurrently measuring intended functional outcomes such as motor neuron survival and TDP-43 localization.

How does SYF2's role in cancer biology relate to its function in neurodegenerative conditions?

This complex relationship requires careful comparative analysis:

  • Opposing Functional Requirements: SYF2 overexpression promotes cell proliferation in cancer contexts , whereas SYF2 suppression is neuroprotective in ALS models . This apparent contradiction may reflect tissue-specific requirements.

  • Cell-Type Specific Analysis: Compare SYF2 binding partners and splicing targets between proliferating cancer cells and post-mitotic neurons using techniques like RNA immunoprecipitation sequencing (RIP-seq) and crosslinking immunoprecipitation (CLIP-seq).

  • TDP-43 Interaction Context: Investigate whether SYF2's interaction with TDP-43 differs between cancer and neuronal contexts, potentially explaining divergent functional outcomes.

  • Splicing Program Differences: Analyze differential exon usage following SYF2 manipulation in both cancer and neuronal models to identify context-specific splicing targets.

This research area highlights the importance of cell-type context in interpreting gene function and suggests potential therapeutic windows where SYF2 modulation might benefit neurodegeneration without promoting oncogenic processes.

What cellular assays best measure functional consequences of SYF2 manipulation?

To comprehensively assess SYF2 function, researchers should employ these methodological approaches:

  • Neuronal Survival Tracking: Longitudinal single-cell survival tracking using techniques like automated microscopy with fluorescent reporters is essential for quantifying neuroprotective effects .

  • Splicing Analysis: RT-PCR and RNA-seq focusing on specific splicing events, including intron retention, exon skipping, and cryptic exon inclusion, particularly in TDP-43 targets like STMN2 .

  • TDP-43 Localization Quantification: High-content imaging with nuclear/cytoplasmic segmentation to quantify TDP-43 distribution following SYF2 manipulation .

  • Motor Function Assessment: For in vivo studies, comprehensive motor function testing including rotarod performance, grip strength, and gait analysis provides functional correlates to cellular changes .

  • Neuromuscular Junction Integrity: Fluorescent labeling of pre- and post-synaptic markers at neuromuscular junctions can reveal preservation of connectivity following SYF2 manipulation .

These assays should be performed across multiple timepoints to capture both early molecular changes and later functional outcomes.

How should researchers control for potential off-target effects when manipulating SYF2 expression?

Rigorous control strategies include:

  • Multiple Independent ASO Sequences: Utilize at least 3-4 distinct ASO sequences targeting different regions of SYF2 mRNA to confirm consistent phenotypes .

  • Rescue Experiments: Re-express ASO-resistant SYF2 variants to verify phenotype reversal, confirming specificity of observed effects.

  • Transcriptome-Wide Off-Target Analysis: Perform RNA-seq following SYF2 ASO treatment to identify potential off-target transcripts and validate their expression changes.

  • Dose-Response Relationships: Establish clear dose-response relationships between SYF2 suppression levels and observed phenotypes.

  • Comparison with CRISPR Knockout: Where feasible, compare ASO results with CRISPR-Cas9 mediated SYF2 knockout to confirm consistent mechanisms.

When reporting results, researchers should transparently document all validation steps performed and acknowledge any potential limitations in specificity.

How do we interpret the differential efficacy of SYF2 suppression across various ALS subtypes?

The available data reveals important patterns requiring careful interpretation:

ALS SubtypeResponse to SYF2 SuppressionProposed Mechanism
C9ORF72 ALSSignificant improvementTDP-43 stabilization and nuclear localization
Sporadic ALSSignificant improvementTDP-43 stabilization and nuclear localization
TARDBP ALSSignificant improvementDirect modulation of mutant TDP-43
FUS ALSMinimal effectIndependent pathological mechanism
SOD1 ALSMinimal effectTDP-43-independent pathology
MAPT FTDModerate improvementPotential tau-TDP-43 interaction

When interpreting these results, researchers should consider:

  • The strong correlation between efficacy and TDP-43 involvement suggests SYF2 suppression specifically targets TDP-43-dependent mechanisms.

  • The lack of effect in FUS and SOD1 ALS models supports mechanistic specificity rather than general neuroprotection.

  • For translational applications, patient stratification based on likely TDP-43 involvement may maximize therapeutic impact.

Research approaches should include comparative transcriptomics across responsive and non-responsive ALS subtypes following SYF2 manipulation to identify critical mechanistic differences.

What explains the apparent contradictory roles of SYF2 in cancer progression versus neurodegeneration?

This intriguing paradox requires nuanced interpretation:

  • Cell Cycle Status Differences: SYF2's promotion of proliferation in cancer cells versus its detrimental effects in post-mitotic neurons may reflect fundamentally different requirements in dividing versus non-dividing cells.

  • TDP-43 Context-Dependence: SYF2's interaction with TDP-43 may be particularly relevant in neurons where TDP-43 homeostasis is critical, but less important in cancer cells driven by other pathways.

  • Tissue-Specific Splicing Programs: The downstream targets of SYF2-mediated splicing likely differ substantially between cancer cells and neurons, explaining divergent functional outcomes.

  • Developmental Switch Hypothesis: SYF2's enrichment in developmental stages suggests it may normally be downregulated in mature neurons, where its persistent expression becomes detrimental.

Research approaches to resolve this contradiction should include comparative interactome analysis between cancer and neuronal contexts, and identification of tissue-specific splicing events regulated by SYF2.

What are the most promising approaches to translate SYF2 modulation into clinical applications for ALS?

Based on current evidence, several translational approaches merit investigation:

  • ASO Optimization: Develop CNS-optimized ASO chemistries targeting SYF2 with enhanced stability, cellular uptake, and blood-brain barrier penetration .

  • Patient Stratification Biomarkers: Identify biomarkers predicting responsiveness to SYF2 suppression, focusing on TDP-43 pathology indicators.

  • Combinatorial Approaches: Explore potential synergies between SYF2 suppression and other therapeutic strategies such as PIKFYVE inhibition .

  • Small Molecule Alternatives: Screen for small molecules that modulate SYF2 activity or its interaction with TDP-43 as alternatives to ASO-based approaches.

  • Viral Vector Delivery: Evaluate AAV-mediated RNA interference targeting SYF2 for long-term expression in motor neurons.

Researchers should prioritize safety studies examining potential adverse effects of chronic SYF2 suppression, particularly given its roles in splicing and cell cycle regulation.

What novel technologies could advance our understanding of SYF2's role in human disease?

Emerging technologies with significant potential include:

  • Spatial Transcriptomics: Map SYF2-dependent splicing changes with spatial resolution in brain tissue to identify regional vulnerabilities.

  • CRISPR Base Editing: Create precise SYF2 domain mutations to dissect functional regions critical for neurodegeneration versus normal splicing.

  • Patient-Derived Brain Organoids: Generate 3D organoid models from ALS patient cells to study SYF2 in more complex cellular environments.

  • In Vivo CLIP-seq: Apply crosslinking immunoprecipitation sequencing in animal models to identify direct RNA targets of SYF2 in the CNS.

  • Single-Cell Multi-Omics: Combine transcriptomic, proteomic, and epigenetic profiling at single-cell resolution following SYF2 manipulation.

These approaches would provide unprecedented insight into SYF2's mechanism and potentially identify additional therapeutic targets in its pathway.

Product Science Overview

Introduction

The SYF2 RNA splicing factor, also known as p29, CBPIN, or NTC31, is a nuclear protein encoded by the SYF2 gene. This protein plays a crucial role in the regulation of pre-mRNA splicing and cell cycle progression. The human recombinant form of SYF2 is produced through recombinant DNA technology, allowing for its use in various research and therapeutic applications.

Gene and Protein Structure

The SYF2 gene is located on chromosome 1 and is a protein-coding gene. It encodes a nuclear protein that interacts with cyclin D-type binding-protein 1 (GICP), which is involved in cell cycle regulation at the G1/S transition . The gene undergoes alternative splicing, resulting in different isoforms of the SYF2 protein .

Function and Mechanism

SYF2 is a component of the spliceosome, a complex responsible for the removal of introns from pre-mRNA transcripts. It is involved in the processing of capped intron-containing pre-mRNA, a critical step in the maturation of mRNA molecules . The protein’s RNA binding capability is essential for its function in the spliceosome .

Role in Cell Cycle Regulation

SYF2 interacts with cyclin D-type binding-protein 1, which is thought to be a cell cycle regulator at the G1/S transition . This interaction suggests that SYF2 may play a role in coordinating cell cycle progression with RNA splicing, ensuring that cells progress through the cell cycle in a regulated manner.

Clinical Significance

Research has shown that SYF2 is involved in cancer progression. Its role in cell cycle regulation and RNA splicing makes it a potential target for cancer therapy . Understanding the function and regulation of SYF2 can provide insights into the mechanisms underlying cancer development and progression.

Applications of Human Recombinant SYF2

The human recombinant form of SYF2 is used in various research applications, including studies on RNA splicing, cell cycle regulation, and cancer biology. Recombinant SYF2 can be used to investigate the protein’s function, interactions, and potential as a therapeutic target.

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.