CFL1 Human

Cofilin-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CFL1 Human

Cofilin-1 (CFL1) is a critical actin-binding protein encoded by the CFL1 gene located on human chromosome 11 (NC_000011.10). It belongs to the ADF/cofilin family and plays a central role in regulating actin cytoskeleton dynamics through pH-sensitive depolymerization of filamentous F-actin (f-actin) and inhibition of monomeric G-actin (g-actin) polymerization . CFL1 is ubiquitously expressed across tissues, with cytoplasmic and membranous localization, and is essential for processes such as cell migration, mitosis, and neural development .

Actin Dynamics and Cellular Processes

CFL1 binds to actin filaments in a pH-dependent manner, promoting their disassembly. This activity is regulated by phosphorylation:

  • LIMK1-mediated phosphorylation (at Ser3) inactivates CFL1, stabilizing f-actin .

  • SSH1-mediated dephosphorylation reactivates CFL1, enabling actin remodeling .

CFL1 is critical for:

  1. Cell migration and invasion: Overexpression enhances filopodia formation and metastasis in cancers .

  2. Mitosis and cytokinesis: Ensures proper chromosomal segregation and cell division .

  3. Neural development: Required for neural crest migration and neural tube morphogenesis .

Cancer Progression

CFL1 overexpression correlates with increased malignancy and poor prognosis in multiple cancers:

Cancer TypeExpression PatternFunctional ImpactSource
GliomaUpregulated in radioresistant cellsEnhances migration, reduces radiosensitivity
Hepatocellular Carcinoma (HCC)High expression linked to metastasisPromotes invasion, poor survival outcomes
Gallbladder CancerElevated with Arp3 co-expressionAssociated with tumor progression
Breast/Colon CancerOverexpressed in metastatic cellsDrives cell motility and invasion

Chronic Lymphocytic Leukemia (CLL)

CFL1 mRNA and protein levels are significantly elevated in CLL patients, suggesting its role in prosurvival signaling and leukemic clone maintenance .

Tuberculosis (TB) Infection

CFL1 is upregulated in Mycobacterium tuberculosis-infected macrophages and patient blood, indicating potential as a TB biomarker .

Experimental Insights: CFL1 Modulation in U251 Glioma Cells

A study on U251 glioma cells demonstrated CFL1’s dual role in radiosensitivity and metastasis:

ParameterCFL1-Downregulated CellsCFL1-Overexpressing CellsSource
Cell ViabilityReduced survival post-radiationEnhanced proliferation
Migration/InvasionDecreased wound healing and Transwell invasionIncreased metastatic capacity
Cell Cycle ArrestIncreased G2 phase arrestReduced G2 arrest (lower apoptosis risk)

Mechanistic Insight: CFL1 knockdown increases radiosensitivity by arresting cells in G2 phase, while overexpression promotes survival and metastasis .

Therapeutic and Diagnostic Implications

  1. Cancer Therapy: Targeting CFL1 with siRNA or inhibitors may enhance radiosensitivity in gliomas and reduce metastasis in HCC .

  2. Biomarker Potential: Elevated CFL1 levels in plasma or macrophages could aid TB diagnosis .

  3. Drug Resistance: CFL1’s role in radioresistance suggests its modulation may improve treatment outcomes in refractory tumors .

Product Specs

Introduction
Cofilin belongs to a family of actin-binding proteins that play a crucial role in disassembling actin filaments. This widely distributed intracellular protein regulates actin dynamics by binding to and depolymerizing filamentous F-actin. Additionally, it inhibits the polymerization of monomeric G-actin in a pH-dependent manner. Cofilin is involved in the cellular process of translocating the actin-cofilin complex from the cytoplasm to the nucleus.
Description
Recombinant Human Cofilin1 (amino acids 1-166) is expressed in E.Coli with an N-terminal 20 amino acid His-tag. This non-glycosylated polypeptide chain consists of 186 amino acids, resulting in a molecular weight of 20kDa.
Physical Appearance
The product is a sterile-filtered solution, appearing colorless.
Formulation
This protein solution is formulated in 20mM Tris buffer at pH 8.0 and contains 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the protein at -20°C. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles.
Purity
The purity of this protein is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
CFL-1, CFL1, Cofilin1, Cofilin-1, Cofilin, non-muscle isoform, 18 kDa phosphoprotein, p18.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MASGVAVSDG VIKVFNDMKV RKSSTPEEVK KRKKAVLFCLSEDKKNIILE EGKEILVGDV GQTVDDPYAT FVKMLPDKDC RYALYDATYE TKESKKEDLVFIFWAPESAP LKSKMIYASS KDAIKKKLTG IKHELQANCY EEVKDRCTLA EKLGGSAVIS LEGKPL.

Q&A

What is the primary function of CFL1 in human cells?

CFL1 (Cofilin-1) is an essential actin-binding protein that regulates cytoskeletal dynamics by depolymerizing actin filaments. In healthy cells, CFL1 plays multiple roles in maintaining cellular structure and function through its involvement in actin turnover. It binds to both globular (G-actin) and filamentous actin (F-actin), severing actin filaments and creating new barbed ends that enable rapid actin reorganization . This cytoskeletal regulation is crucial for numerous cellular processes including cell division, migration, and morphogenesis. Additionally, CFL1 has been implicated in signal transduction pathways, particularly those involving the regulation of transcription factors such as NF-κB .

How is CFL1 expression and activity regulated in normal human tissues?

CFL1 activity is primarily regulated through phosphorylation at Serine-3, which inactivates the protein by preventing its binding to actin. This phosphorylation is controlled by kinases including LIM kinase (LIMK) and testicular protein kinase (TESK), while dephosphorylation (activation) is mediated by phosphatases such as slingshot (SSH) and chronophin. Regulation also occurs at the transcriptional level, with various cellular stressors and signaling pathways influencing CFL1 expression. In normal tissues, CFL1 activity is carefully balanced to maintain proper cytoskeletal dynamics . Experimental approaches to measure CFL1 activity typically involve Western blot analysis to detect the ratio of phosphorylated to non-phosphorylated CFL1, with higher proportions of non-phosphorylated CFL1 indicating increased activity .

What experimental methods are most effective for detecting CFL1 levels in clinical samples?

For clinical samples, a combination of techniques provides the most comprehensive assessment of CFL1 status:

  • RT-qPCR for mRNA expression analysis, which has demonstrated high sensitivity in detecting differential CFL1 expression between healthy and malignant tissues

  • Western blot analysis for protein levels and phosphorylation status, helping distinguish between active and inactive forms of CFL1

  • Immunohistochemistry (IHC) for tissue microarray analysis, which allows visualization of CFL1 distribution patterns in tissue samples

For diagnostic purposes in conditions such as lung cancer, plasma CFL1 protein detection has shown promise as a biomarker . When designing such experiments, researchers should include appropriate controls and standardize sample collection procedures to minimize variability. Additionally, combining multiple detection methods strengthens the reliability of findings regarding CFL1 expression patterns.

How does CFL1 contribute to cancer progression and metastasis?

CFL1 plays multifaceted roles in cancer progression through several mechanisms:

  • Enhanced cell migration and invasion: Upregulated CFL1 promotes actin dynamics necessary for cancer cell motility, with knockdown studies showing significantly reduced invasiveness in multiple cancer types including hepatocellular carcinoma (HCC) and lung cancer

  • Proliferation support: CFL1 maintains prosurvival signaling in malignancies such as chronic lymphocytic leukemia (CLL), where silencing CFL1 increases apoptosis and alters cell cycle progression

  • Inflammatory signaling modulation: CFL1 regulates NF-κB nuclear translocation, affecting downstream expression of genes involved in cancer progression including MMP9, BCL2, EZH2, COX2, and VCAM1

Research has demonstrated that high CFL1 expression correlates with poor clinical outcomes, including increased recurrence rates (72.9% vs. 42.9%) and mortality (51.1% vs. 20.8%) in HCC patients . In lung cancer, reciprocal expression patterns between CFL1 and Twist-1 proteins were observed in 67.4% of tissue samples, suggesting coordinated roles in cancer progression .

What is the relationship between CFL1 expression and radiotherapy resistance in human cancers?

CFL1 has been implicated in radioresistance across multiple cancer types, particularly in gliomas. Studies have demonstrated that CFL1 is significantly upregulated in radioresistant astrocytomas , suggesting a protective role against radiation-induced damage. The mechanistic basis appears to involve CFL1's ability to modulate actin cytoskeleton dynamics, potentially allowing enhanced DNA damage repair and cell survival following radiation exposure.

Experimental evidence shows that downregulation of CFL1 in U251 human glioma cells increases their radiosensitivity . This finding suggests a therapeutic approach where CFL1 inhibition could be combined with radiotherapy to improve treatment outcomes for resistant tumors. Researchers investigating radioresistance should consider measuring CFL1 expression and activity before and after radiation exposure to determine correlation with treatment response.

How do genetic variations in CFL1 affect disease susceptibility and progression?

Nonsynonymous single nucleotide polymorphisms (nsSNPs) in the CFL1 gene can significantly impact protein function and disease risk. Computational analysis has identified 46 nsSNPs in CFL1, with 20 classified as potentially deleterious or disease-causing . Two variants in particular—L84P (rs199716082) and L99A (rs267603119)—were identified as significant mutations associated with disease development .

These genetic variations can impact:

  • Neurological disorders: CFL1 mutations have been linked to neural tube closure defects, lissencephaly, epilepsy, schizophrenia, and neurodegenerative conditions like Alzheimer's disease

  • Cancer susceptibility: Altered CFL1 function can influence cytoskeletal dynamics critical for tumor cell behavior

  • Developmental abnormalities: Impaired neural crest cell migration due to CFL1 mutations can lead to developmental defects

Researchers studying disease associations should consider genotyping these high-impact SNPs in their patient cohorts to identify potential correlations with disease phenotypes or treatment responses.

What are the most effective approaches for manipulating CFL1 expression in experimental models?

Several approaches have proven effective for modulating CFL1 expression in research settings:

  • RNA interference (RNAi): siRNA targeting CFL1 has been successfully used to examine functional consequences of CFL1 knockdown in multiple cancer cell lines, revealing effects on migration, chemotaxis, apoptosis, and cell cycle progression

  • Inducible expression systems: Tetracycline-inducible systems allow controlled overexpression of CFL1, as demonstrated in H1299 lung cancer cells (HCOXP), enabling temporal studies of CFL1 effects

  • CRISPR-Cas9 gene editing: For permanent modification of CFL1 expression, this approach provides more stable alterations than transient knockdown methods

When designing CFL1 manipulation experiments, researchers should:

  • Include appropriate controls (scrambled siRNA, empty vector)

  • Verify knockdown/overexpression efficiency by both mRNA and protein analysis

  • Carefully time experiments based on the biological process under investigation

  • Consider compensatory mechanisms that may emerge with chronic CFL1 modulation

What methodological approaches best assess CFL1's impact on cell migration and invasion?

To comprehensively evaluate CFL1's effects on cell motility, researchers should employ multiple complementary techniques:

  • Transwell migration/invasion assays: This method has effectively demonstrated reduced chemotaxis and migration in CFL1-silenced CLL cells compared to healthy volunteer cells

  • Wound healing (scratch) assays: For measuring collective cell migration in monolayer cultures

  • 3D invasion models: Using matrices such as Matrigel to better recapitulate the in vivo invasion process

  • Live-cell imaging: To track cell movement dynamics and cytoskeletal changes in real-time

  • Actin dynamics assays: Fluorescence recovery after photobleaching (FRAP) or barbed end assays to directly measure CFL1's effect on actin turnover

Data analysis should quantify multiple parameters including migration distance, velocity, directionality, and invasion depth. Researchers should also correlate these findings with CFL1 activity status (phosphorylated vs. non-phosphorylated) to establish mechanistic links between CFL1 function and observed phenotypes.

How can researchers effectively study the interaction between CFL1 and NF-κB signaling pathways?

To investigate the regulatory relationship between CFL1 and NF-κB signaling, researchers should implement a multi-faceted experimental approach:

  • Subcellular fractionation and Western blot analysis to measure p65 subunit distribution between cytoplasm and nucleus following CFL1 manipulation, as demonstrated in HCC studies where CFL1 knockdown elevated cytoplasmic p65 while reducing nuclear levels

  • Immunofluorescence microscopy to visualize NF-κB translocation in response to stimuli (e.g., TNF-α) under varied CFL1 expression conditions

  • Reporter gene assays using NF-κB response elements to quantify transcriptional activity

  • Chromatin immunoprecipitation (ChIP) to assess NF-κB binding to target gene promoters

  • Co-immunoprecipitation experiments to detect physical interactions between CFL1 and components of the NF-κB pathway

  • qRT-PCR and Western blot analyses of NF-κB target genes (MMP9, BCL2, EZH2, COX2, VCAM1) to confirm downstream effects

This integrated approach would provide comprehensive insights into how CFL1 regulates NF-κB-mediated inflammatory and survival pathways in various pathological contexts.

How reliable is CFL1 as a prognostic biomarker across different cancer types?

CFL1's utility as a prognostic biomarker varies by cancer type but shows consistent patterns in certain malignancies:

  • Hepatocellular carcinoma (HCC): High CFL1 expression correlates significantly with unfavorable clinical features including tumor encapsulation and microvascular invasion, resulting in higher recurrence (72.9% vs. 42.9%) and mortality rates (51.1% vs. 20.8%)

  • Lung cancer: CFL1 expression patterns, particularly when analyzed in relation to other markers like Twist-1, have demonstrated prognostic value in non-small cell lung cancer (NSCLC)

  • Leukemia: Elevated CFL1 activity in CLL correlates with disease aggressiveness

When evaluating CFL1 as a biomarker, researchers should:

  • Use standardized scoring systems for immunohistochemistry

  • Incorporate multivariate analysis to control for confounding factors

  • Consider CFL1 in conjunction with established prognostic markers

  • Validate findings across independent patient cohorts

The most robust approach combines CFL1 assessment with other molecular and clinical parameters to develop integrated prognostic models with superior predictive power.

What strategies show promise for targeting CFL1 therapeutically in human diseases?

Several approaches for therapeutic targeting of CFL1 have shown experimental promise:

  • Small molecule inhibitors: Compounds targeting CFL1 directly or its regulatory kinases (like LIMKs) have demonstrated anti-cancer effects in preclinical models

  • Peptide-based approaches: Designed peptides that interfere with CFL1-actin binding

  • Combination therapies: CFL1 inhibition appears to sensitize cancer cells to radiotherapy in glioma models, suggesting potential for combination approaches

  • Genetic approaches: siRNA or antisense oligonucleotides targeting CFL1 have shown efficacy in reducing cancer cell viability and motility in experimental models

When designing CFL1-targeting strategies, researchers should consider:

  • Selective targeting to minimize off-target effects

  • Tissue-specific delivery approaches

  • Potential compensatory mechanisms (e.g., upregulation of related actin-binding proteins)

  • Biomarkers for patient stratification

These therapeutic approaches remain largely preclinical, highlighting the need for further translational research to advance CFL1-targeted interventions toward clinical application.

How does CFL1 interact with the tumor microenvironment to promote cancer progression?

CFL1's role extends beyond cancer cell-intrinsic functions to include interactions with the tumor microenvironment through several mechanisms:

  • Extracellular matrix (ECM) remodeling: CFL1 contributes to ECM degradation through regulation of matrix metalloproteinases, particularly MMP9 as observed in HCC studies

  • Inflammatory signaling: By modulating NF-κB pathway activity, CFL1 influences the expression of inflammatory mediators that shape the immune landscape within tumors

  • Angiogenesis: Through effects on endothelial cell migration and VEGF signaling

Future research should explore CFL1's role in:

  • Cancer-associated fibroblast interactions

  • Immune cell infiltration and function

  • Extracellular vesicle content and function

  • Premetastatic niche formation

Methodologically, co-culture systems, 3D organoids, and in vivo models that preserve tumor-stroma interactions will be critical for dissecting these complex relationships.

What computational approaches best predict the functional impact of CFL1 mutations?

Advanced computational methods for assessing CFL1 mutations include:

  • Integrated multi-algorithm approaches combining sequence-based predictions with structural analysis, as demonstrated in studies identifying L84P and L99A variants as particularly damaging

  • Molecular dynamics simulations to predict how mutations affect protein stability, flexibility, and interactions

  • Machine learning algorithms trained on known mutation effects to predict novel variant impacts

  • Systems biology approaches integrating CFL1 into broader pathway and network analyses

Researchers should employ multiple complementary computational tools rather than relying on a single prediction algorithm, as this approach identified 20 high-impact nsSNPs from an initial set of 46 variants in CFL1 . Validation of computational predictions through functional assays remains essential for confirming the biological significance of identified mutations.

Product Science Overview

Structure and Function

Cofilin-1 is a small protein with a molecular weight of approximately 21 kDa . It binds to both monomeric (G-actin) and filamentous (F-actin) actin in a pH-dependent manner. At lower pH levels, cofilin-1 binds to F-actin, while at higher pH levels, it severs actin filaments and binds to G-actin . This dynamic binding and severing activity is essential for the reorganization of the actin cytoskeleton in response to various cellular signals .

Regulation

The activity of cofilin-1 is tightly regulated by several mechanisms, including phosphorylation, pH changes, and interactions with phosphoinositides . Phosphorylation of cofilin-1 at serine 3 by LIM kinase (LIMK) inhibits its actin-binding activity, while dephosphorylation by slingshot phosphatase (SSH) restores its activity . This regulation ensures that cofilin-1 activity is precisely controlled in response to cellular needs.

Recombinant Production

Human recombinant cofilin-1 is produced using bacterial expression systems. The recombinant protein is typically purified using ion exchange chromatography to achieve high purity levels (approximately 95%) . The recombinant form is often supplied as a lyophilized powder, which can be reconstituted in a suitable buffer for experimental use .

Biological Activity

The biological activity of recombinant cofilin-1 is assessed by its ability to bind and sever F-actin in a pH-dependent manner. Standard assays involve monitoring the actin-binding and severing activity of cofilin-1 using SDS-PAGE analysis of F-actin/cofilin spin-down assays performed at different pH levels . These assays ensure that the recombinant protein retains its functional properties.

Clinical Significance

Cofilin-1 is implicated in various physiological and pathological processes. It plays a vital role in cancer cell migration and invasion, particularly in colorectal cancer and esophageal squamous cell carcinoma . The differential expression of cofilin-1 and its regulators, such as LIMK1 and SSH1, is associated with tumor progression and metastasis . Understanding the role of cofilin-1 in these processes can provide insights into potential therapeutic targets for 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.