FSCN1 Human

Recombinant Human Fascin
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

Gene:

  • Location: Chromosome 7p22.1 (GenBank: NM_003088.4)

  • Length: 13,840 base pairs, 5 exons .

  • Conservation: Highly homologous across species (e.g., 96.55% with mouse, 75.76% with zebrafish) .

Protein:

  • Length: 493 amino acids, molecular mass 54.5 kDa .

  • Domains: Four β-trefoil domains (residues 8–139, 140–260, 261–381, 382–493) .

  • Actin-Binding Sites: Three distinct actin-binding sites (ABS1–3), with phosphorylation at Ser39 (ABS1) and Ser274 regulating actin-bundling activity .

DomainFunctionKey Residues
β-trefoil 1ABS1 formationSer39 (PKC phosphorylation site)
β-trefoil 3ABS3 interactionSer274 (modulates bundling capacity)

Physiological Functions

Actin-Dependent Roles:

  • Cell Migration: Forms filopodia and invadopodia to enhance tumor invasion .

  • Adhesion Dynamics: Regulates focal adhesion stability via actin cytoskeleton .

  • Extracellular Vesicle Release: Mediates microtubule-dependent vesicle trafficking .

Actin-Independent Roles:

  • Microtubule Regulation: Binds microtubules to modulate cell migration in breast cancer .

  • Nuclear Interactions: Phosphorylated FSCN1 (pFascin) interacts with transcriptional machinery (e.g., RbBP5) to regulate gene expression .

  • Stem Cell Maintenance: Enhances cancer cell stemness in melanoma and breast cancer .

Role in Cancer

FSCN1 is upregulated in diverse cancers, including breast, lung, tongue squamous cell carcinoma (TSCC), cervical, and head and neck cancers . Key findings:

Cancer TypeExpression CorrelationClinical ImplicationsReferences
Breast cancerHigh FSCN1 = Aggressive metastasisPoor survival, therapeutic target
TSCCFSCN1 overexpressionPredicts relapse and poor prognosis
Cervical cancerFSCN1 + PIK3CA mutations = Radiation resistanceSynthetic lethality target

Mechanisms in Cancer:

  • Metastasis: Promotes transendothelial migration and invasiveness .

  • Radiation Resistance: Silencing FSCN1 sensitizes PIK3CA-mutated cells to irradiation .

  • Stem Cell Niche: Maintains CSC populations via actin-independent pathways .

Therapeutic Potential

Current Strategies:

  • Small Molecules: Docosahexaenoic acid (DHA) inhibits FSCN1 in breast cancer .

  • Proteins: Recombinant porcine NK-lysin A targets FSCN1 in hepatocellular carcinoma .

  • Gene Silencing: RNAi-mediated knockdown reduces tumor growth and migration .

Emerging Targets:

  • Phosphorylation Inhibitors: Targeting Ser39/Ser274 to block actin bundling .

  • Microtubule Disruptors: Enhancing FSCN1-microtubule interactions to reduce migration .

Clinical Implications and Biomarkers

Serum FSCN1 as a Biomarker:

  • Atherosclerosis: Elevated serum FSCN1 correlates with coronary heart disease (CHD) risk (OR = 2.82, 95% CI: 1.35–5.91) .

  • Diagnosis: ROC curve analysis shows AUC = 0.9388 for FSCN1 in CHD detection .

Biomarker FeatureValueSource
Serum FSCN1 cutoff for CHD≥16.42 ng/mL
Sensitivity/Specificity78.57%/100%

Data Tables

Table 1: Clinical Correlations of FSCN1 in Cancers

CancerFSCN1 ExpressionClinical CorrelationReferences
BreastHighMetastasis, poor survival
LungElevatedMetastatic colonization
TSCCOverexpressionRelapse, poor prognosis

Table 2: Therapeutic Strategies Targeting FSCN1

ApproachAgentOutcomeReferences
Small moleculeDHAInhibits metastasis
Gene knockdownsiRNA/FSCN1Reduces tumor growth
Protein therapyNK-lysin ATargets hepatocellular carcinoma

Product Specs

Introduction
FSCN1, an actin-bundling protein, plays a crucial role in providing rigidity to filopodial bundles. This rigidity is essential for effectively pushing the membrane forward during processes like cytoskeleton remodeling and cell migration. This protein is involved in assembling actin filament bundles found in cellular structures such as microspikes, membrane ruffles, and stress fibers. While largely absent in healthy epithelial tissues, FSCN1 expression is elevated in various human carcinoma types, and its presence is clinically correlated with a negative prognosis. Notably, FSCN1 is highly expressed in neurons, glial cells, and endothelial cells.
Description
Recombinant Human FSCN1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 513 amino acids (specifically, amino acids 1 through 493). It has a molecular weight of 56.7 kDa. A 20 amino acid His-Tag is fused to the N-terminus of the FSCN1. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a colorless solution that has been sterilized through filtration.
Formulation
The product is provided as a 1 mg/ml solution. The solution's composition includes 20mM Tris (pH 8), 2mM DTT, 0.1M NaCl, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer storage, freezing at -20°C is recommended. To ensure stability during long-term storage, adding a carrier protein (either 0.1% HSA or BSA) is advised. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the product exceeds 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Strongylocentrotus Purpuratus, actin-bundling protein, FAN1, HSN, SNL.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MTANGTAEAV QIQFGLINCG NKYLTAEAFG FKVNASASSL KKKQIWTLEQ PPDEAGSAAV CLRSHLGRYL
AADKDGNVTC EREVPGPDCR FLIVAHDDGR WSLQSEAHRR YFGGTEDRLS CFAQTVSPAE KWSVHIAMHP QVNIYSVTRK RYAHLSARPA
DEIAVDRDVP WGVDSLITLA FQDQRYSVQT ADHRFLRHDG RLVARPEPAT GYTLEFRSGK VAFRDCEGRY LAPSGPSGTL KAGKATKVGK
DELFALEQSC AQVVLQAANE RNVSTRQGMD LSANQDEETD QETFQLEIDR DTKKCAFRTH TGKYWTLTAT GGVQSTASSK NASCYFDIEW
RDRRITLRAS NGKFVTSKKN GQLAASVETA GDSELFLMKL INRPIIVFRG EHGFIGCRKV TGTLDANRSS YDVFQLEFND GAYNIKDSTG
KYWTVGSDSA VTSSGDTPVD FFFEFCDYNK VAIKVGGRYL KGDHAGVLKA SAETVDPASL WEY.

Q&A

What is FSCN1 and what is its primary function in human cells?

FSCN1 (Fascin actin-bundling protein 1) is a member of the Fascin family of actin-binding proteins that plays critical roles in cellular architecture and motility. Its primary function involves bundling filamentous actin into parallel arrays, which are essential for the formation and stability of filopodia, microspikes, and other actin-based cellular protrusions. FSCN1 mediates cell migration, motility, adhesion, and tissue invasion through its ability to organize the actin cytoskeleton. Research methodologies to study these functions typically employ various cytoskeletal visualization techniques including fluorescent-tagged FSCN1 constructs, confocal microscopy, and super-resolution imaging to trace protein localization during cellular movement. Functional studies often utilize knockdown and overexpression experimental designs to evaluate consequent changes in cell morphology and migratory capacity .

How is FSCN1 expression regulated in normal human tissues?

FSCN1 expression demonstrates tissue-specific regulation patterns, with relatively low expression in most normal epithelial tissues but higher expression in neuronal cells, dendritic cells, and endothelial cells. Methodologically, researchers investigate FSCN1 regulation through analysis of transcription factor binding sites in the FSCN1 promoter region, chromatin immunoprecipitation assays, and reporter gene assays. Multiple regulatory pathways have been identified, including transcriptional regulation by factors such as CREB and NF-κB. Post-transcriptional regulation occurs through microRNAs, particularly miR-133a and miR-145, which target FSCN1 mRNA. Researchers often employ RT-qPCR, western blotting, and immunohistochemistry to quantify expression levels across different tissue types. Single-cell RNA sequencing has revealed that FSCN1 is predominantly expressed in endothelial cells in vascular tissues, providing important context for its role in vascular pathologies .

What protein interactions are critical for FSCN1 function?

FSCN1 engages in multiple protein-protein interactions that are essential for its actin-bundling functions and cellular roles. Methodologically, these interactions are commonly studied using co-immunoprecipitation, proximity ligation assays, and fluorescence resonance energy transfer (FRET) techniques. The most critical interaction is between FSCN1 and F-actin, mediated through specific actin-binding domains. STRING tool analysis has identified several FSCN1-binding proteins involved in cytoskeletal organization and cellular migration pathways . Additionally, FSCN1 interacts with proteins in focal adhesion complexes, including integrins, particularly β1-integrin, which connects the actin cytoskeleton to the extracellular matrix. Researchers investigating these interactions typically employ site-directed mutagenesis of binding domains followed by functional assays to determine the impact of specific interactions on cellular phenotypes such as migration velocity and directional persistence.

How does FSCN1 contribute to atherosclerosis pathogenesis?

FSCN1 plays multiple roles in atherosclerosis (AS) pathogenesis, primarily through its effects on endothelial cell function and vascular remodeling. Research methodologies to investigate this relationship include single-cell RNA sequencing of atherosclerotic tissues, which has revealed that FSCN1 is predominantly expressed in endothelial cells in the context of atherosclerosis . Analysis of bulk RNA-seq data from human atherosclerotic plaques demonstrates significantly higher FSCN1 expression in atherosclerotic samples compared to normal tissue, particularly in advanced plaques versus early lesions . Mechanistically, FSCN1 appears to influence endothelial cell pyroptosis and migration, key processes in atherosclerotic development. Knockdown experiments in mouse aortic endothelial cells (MAECs) have shown that reducing FSCN1 expression decreases pyroptosis while enhancing migration capability . Researchers investigating FSCN1's role in atherosclerosis should consider employing both in vitro models using ox-LDL-induced endothelial dysfunction and in vivo models such as ApoE knockout mice fed high-fat diets to comprehensively evaluate its contributions to disease progression.

What clinical evidence links FSCN1 to human cardiovascular disease?

Clinical evidence linking FSCN1 to human cardiovascular disease has emerged from both transcriptomic analyses and direct patient studies. Research methodologies include cross-sectional clinical studies measuring circulating FSCN1 levels in patients with coronary heart disease (CHD). One such study demonstrated significantly elevated serum FSCN1 concentrations in CHD patients compared to controls (18.8±0.5 ng/mL vs 13.3±0.6 ng/mL, P<0.0001) . Logistic regression analyses have established that high FSCN1 levels are associated with increased risk of atherosclerosis, independent of traditional risk factors. ROC curve analysis has been employed to evaluate FSCN1's potential as a diagnostic biomarker for CHD. Additional evidence comes from transcriptomic studies of carotid endarterectomy samples, which show increased FSCN1 expression in symptomatic compared to asymptomatic carotid plaques . Researchers investigating the clinical relevance of FSCN1 should consider multivariate analyses that account for confounding factors such as age, sex, smoking status, and comorbidities to establish independent associations with cardiovascular outcomes.

What experimental models are most appropriate for studying FSCN1 in atherosclerosis?

The selection of appropriate experimental models for studying FSCN1 in atherosclerosis depends on the specific research questions being addressed. For in vivo studies, ApoE knockout mice fed high-fat diets represent a well-established model that recapitulates key features of human atherosclerosis. Single-cell RNA sequencing analysis of aortic tissues from these mice has revealed significant alterations in cellular composition and intercellular communication patterns, with notable differences in FSCN1 expression . For in vitro studies, mouse aortic endothelial cells (MAECs) treated with oxidized low-density lipoprotein (ox-LDL) provide a useful model for investigating molecular mechanisms. Knockdown experiments using siRNA or CRISPR-Cas9 targeting FSCN1 in these cells allow for functional assessment of FSCN1's role in endothelial cell behaviors relevant to atherosclerosis, such as pyroptosis and migration . Human studies typically involve analysis of atherosclerotic plaque samples obtained during carotid endarterectomy, with advanced single-cell and bulk RNA sequencing techniques employed to characterize FSCN1 expression patterns across different cell populations and disease stages . Researchers should choose models that best align with their specific research questions while considering the translational relevance to human disease.

How does FSCN1 influence cancer cell migration and invasion?

FSCN1 exerts significant influence on cancer cell migration and invasion through its fundamental role in cytoskeletal organization. Research methodologies investigating this relationship employ single-cell analysis to correlate FSCN1 expression with migration state scores. For instance, analysis of 2,150 individual head and neck squamous cell carcinoma (HNSCC) cells revealed a positive correlation between FSCN1 expression and migration capability (correlation coefficient 0.24) . FSCN1 enhances cancer cell motility by promoting filopodia formation and stabilization, structures essential for directional migration and invasion. Methodologically, researchers track individual cell migration using in vitro chemotaxis assays, which has confirmed that FSCN1 expression levels positively correlate with migration velocity . FSCN1 also facilitates epithelial-mesenchymal transition (EMT), a process crucial for cancer invasion. At the molecular level, FSCN1 interacts with multiple migration-associated genes, including EGFR (r=0.48), PMS2 (r=0.37), and AKT1 (r=0.35) . Researchers investigating these mechanisms typically employ knockdown and overexpression approaches coupled with three-dimensional invasion assays and live-cell imaging to quantify migration dynamics.

How does FSCN1 expression correlate with tumor immune microenvironment?

FSCN1 expression demonstrates significant correlations with the composition and function of the tumor immune microenvironment, suggesting immunomodulatory roles beyond its cytoskeletal functions. Research methodologies exploring these relationships employ Spearman correlation analysis between FSCN1 expression and immune cell infiltration profiles. In HNSCC, FSCN1 expression shows strong positive correlation with CD4+ T cell infiltration and negative correlation with B cells and CD8+ T cells . These findings suggest that high FSCN1-expressing tumors may create an immunosuppressive microenvironment characterized by increased regulatory T cells and reduced cytotoxic immune responses. Methodologically, researchers investigate these relationships through multiplex immunohistochemistry, flow cytometry of tumor-infiltrating lymphocytes, and computational deconvolution of bulk RNA sequencing data. Although current evidence does not fully elucidate the functional mechanisms by which FSCN1 may directly regulate immune responses, these correlations provide important context for understanding how FSCN1 overexpression might contribute to tumor immune evasion. Researchers exploring these relationships should consider integrated analyses that examine both FSCN1 expression and comprehensive immune profiling to identify potential therapeutic vulnerabilities.

What are the optimal techniques for measuring FSCN1 protein levels in clinical samples?

The optimal techniques for measuring FSCN1 protein levels in clinical samples depend on the specific research questions, sample types, and available resources. For circulating FSCN1 detection in serum or plasma, enzyme-linked immunosorbent assay (ELISA) represents a validated approach with demonstrated clinical utility. Studies have successfully employed commercial ELISA kits to quantify serum FSCN1 levels in patients with coronary heart disease, establishing significant differences between patient and control groups . For tissue samples, immunohistochemistry (IHC) provides spatial information about FSCN1 expression patterns within the tissue architecture. This approach has successfully distinguished between primary and metastatic HNSCC samples, revealing stronger FSCN1 staining in metastatic lesions . Western blotting offers quantitative assessment of FSCN1 protein expression in tissue or cell lysates and has been used to validate findings from transcriptomic analyses . Each method presents distinct advantages: ELISA offers high-throughput capability and standardization for circulating biomarker applications; IHC provides histological context; and Western blotting allows for precise quantification. Researchers should select methods based on their specific experimental needs and consider multiple complementary techniques for comprehensive analysis.

How should researchers design studies to investigate FSCN1 function in cellular models?

Designing robust studies to investigate FSCN1 function in cellular models requires careful consideration of cell selection, manipulation approaches, and functional assays. Initial steps should include screening potential cell lines for baseline FSCN1 expression using qPCR and Western blotting, as demonstrated in HNSCC research where expression levels were characterized across multiple cell lines (A-253, CAL-33, ICR-31, SC-25, YD-38, BICR-22, BICR-6, and BICR-56) . For functional studies, researchers should employ both loss-of-function and gain-of-function approaches. RNA interference (siRNA or shRNA) and CRISPR-Cas9 genome editing provide effective methods for FSCN1 knockdown, while plasmid-based overexpression systems allow for examining the effects of increased FSCN1 levels. Following manipulation of FSCN1 expression, researchers should implement comprehensive functional assays including proliferation assays (MTT, BrdU incorporation), migration assays (wound healing, transwell, chemotaxis tracking of individual cells), and invasion assays (Matrigel invasion chambers). Additionally, visualization of cytoskeletal structures using phalloidin staining and confocal microscopy provides valuable insights into how FSCN1 affects actin organization and cellular morphology. For mechanisms involved in cardiovascular disease, pyroptosis assays examining caspase-1 activation and IL-1β release should be considered . Researchers should include appropriate controls and conduct rescue experiments to confirm the specificity of observed effects.

What bioinformatic approaches are most informative for analyzing FSCN1 in multi-omics datasets?

Multi-omics analysis of FSCN1 requires sophisticated bioinformatic approaches that integrate data across genomic, transcriptomic, proteomic, and functional levels. For transcriptomic analysis, differential expression analysis comparing disease versus normal tissues provides foundational insights, as demonstrated in studies of atherosclerotic plaques and HNSCC . Single-cell RNA sequencing analysis offers higher resolution of FSCN1 expression patterns across different cell populations within heterogeneous tissues. Tools such as Seurat, singleR, and ReactomeGSA have been successfully employed to characterize cell-specific expression patterns of FSCN1 in atherosclerosis models . For understanding intercellular communication networks involving FSCN1-expressing cells, packages like CellChat can identify signaling pathways and interaction patterns, revealing for instance that Semaphorin 7 (SEMA7)-mediated signaling pathways are enriched in atherosclerosis models . Correlation analyses between FSCN1 expression and functional states (such as migration state scores in single-cell data) provide valuable insights into potential functional roles . Pathway enrichment analysis using tools like ReactomeGSA helps identify biological processes associated with FSCN1 expression, while protein-protein interaction analysis using STRING identifies molecular networks involving FSCN1 . For clinical correlations, survival analysis tools including Kaplan-Meier plotting and Cox regression modeling establish associations between FSCN1 expression and patient outcomes . Integration of these diverse analytical approaches provides a comprehensive understanding of FSCN1 biology and clinical significance.

How might therapeutic targeting of FSCN1 be achieved in cardiovascular and cancer contexts?

Therapeutic targeting of FSCN1 represents a promising yet challenging frontier in both cardiovascular and cancer research. Potential approaches include small molecule inhibitors that disrupt FSCN1's actin-bundling function, antisense oligonucleotides or siRNA-based therapies that reduce FSCN1 expression, and antibody-based strategies that interfere with FSCN1's protein-protein interactions. For cardiovascular applications, research indicates that knockdown of FSCN1 reduces endothelial cell pyroptosis while enhancing migration capability, suggesting that inhibition might protect against atherosclerosis progression . Design considerations for cardiovascular interventions should include endothelial-specific delivery systems to minimize off-target effects on other actin-dependent cellular processes. In cancer contexts, FSCN1 inhibition could potentially reduce metastatic capabilities by disrupting cell migration and invasion . Research methodologies for developing FSCN1-targeted therapies should employ high-throughput screening of chemical libraries against the FSCN1 protein, structure-based drug design utilizing crystallographic data of FSCN1's actin-binding domains, and validation in progressively complex models ranging from cell lines to patient-derived xenografts. Critical challenges include achieving specificity for FSCN1 over other actin-binding proteins and developing effective delivery systems that can target either the tumor microenvironment or atherosclerotic plaques.

What are the unresolved contradictions in current FSCN1 research?

Several unresolved contradictions exist in current FSCN1 research that warrant focused investigation. One significant paradox involves FSCN1's differential effects on cell migration in different contexts. While knockdown of FSCN1 reduced pyroptosis but increased migration in endothelial cells in atherosclerosis models , FSCN1 expression positively correlates with migration capability in cancer cells . This apparent contradiction suggests context-dependent functions that may be influenced by the underlying pathophysiology, cell type, or interacting molecular pathways. Another unresolved question concerns the mechanisms by which FSCN1 influences the immune microenvironment in tumors. While correlations between FSCN1 expression and immune cell infiltration patterns have been observed , the causative mechanisms remain unclear. Additionally, there are contradictory findings regarding FSCN1's role in different stages of atherosclerosis, with some evidence suggesting stage-specific functions. Methodologically, these contradictions can be addressed through comprehensive comparative studies across different disease models and cell types, time-course experiments examining FSCN1's role at different disease stages, and mechanistic studies that integrate signaling pathway analysis with functional outcomes. Resolving these contradictions will require sophisticated experimental designs that account for the complex, context-dependent nature of FSCN1 functions.

How do post-translational modifications regulate FSCN1 function in pathological conditions?

Post-translational modifications (PTMs) of FSCN1 represent an underexplored but potentially crucial regulatory mechanism that may explain context-dependent functions in different pathological conditions. Research methodologies for investigating FSCN1 PTMs should include mass spectrometry-based phosphoproteomics, acetylomics, and ubiquitinomics to identify modification sites and their prevalence in different disease states. Phosphorylation of FSCN1 at serine 39 by protein kinase C has been shown to reduce its actin-bundling capacity, suggesting that the phosphorylation state may serve as a molecular switch regulating FSCN1 function. In atherosclerosis and cancer, altered kinase activity and dysregulated phosphatase expression could potentially modify FSCN1's phosphorylation status, thereby affecting its contribution to disease progression. Beyond phosphorylation, other potential PTMs including ubiquitination, SUMOylation, and acetylation remain largely unexplored in the context of FSCN1 regulation. Experimentally, researchers should employ site-directed mutagenesis to generate phospho-mimetic and phospho-deficient FSCN1 variants, followed by functional assays to determine how these modifications affect FSCN1's roles in cell migration, invasion, and interactions with binding partners. Additionally, investigating how disease-specific conditions such as inflammation, hypoxia, or oxidative stress influence FSCN1 PTMs could provide valuable insights into its differential functions across pathological contexts.

Product Science Overview

Introduction

Recombinant Human Fascin is a protein that has garnered significant interest in the field of cell biology due to its role in actin-bundling and its implications in various cellular processes. Fascin is a 55 kDa actin-bundling protein that is crucial for the formation of cellular protrusions such as filopodia, which are essential for cell motility and migration .

Structure and Expression

Fascin is composed of 493 amino acids and is typically expressed in Escherichia coli for recombinant production . The recombinant form of Fascin often includes a His-tag at the N-terminus to facilitate purification and detection . The protein is highly purified, with a purity greater than 95%, making it suitable for various biochemical and structural studies .

Function and Mechanism

Fascin plays a pivotal role in the organization of actin filament bundles. It contains two major actin-binding sites that allow it to cross-link actin filaments into tightly packed, parallel bundles . This bundling activity is essential for the formation of microspikes, membrane ruffles, and stress fibers, which are critical for cell shape and movement .

Biological Significance

Fascin is not only important for normal cellular functions but also has significant implications in cancer biology. It is absent from most normal epithelial tissues but is upregulated in various forms of human carcinoma . The expression of Fascin in tumors is often associated with increased cell motility and invasiveness, correlating with a poor prognosis in cancer patients .

Applications

Recombinant Human Fascin is widely used in research to study its role in actin dynamics and cell motility. It is also employed in high-throughput screening assays to identify potential inhibitors that could serve as therapeutic agents in cancer treatment .

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.