TFF1 Human, His

Trefoil Factor-1 Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Production and Purification

TFF1 Human, His is synthesized via bacterial expression systems:

  • Host Strain: E. coli

  • Expression Vector: Recombinant plasmid with T7 promoter (common in E. coli systems)

  • Purification Steps:

    1. Cell Lysis: Sonication or enzymatic disruption

    2. Nickel Affinity Chromatography: His tag binds to nickel columns

    3. Chromatographic Polishing: Ion-exchange or size-exclusion chromatography

Purity: >95% as confirmed by SDS-PAGE .

Biological Functions and Research Findings

TFF1 Human, His retains critical biological activities of native TFF1:

Mucosal Protection

  • Mucin Binding: Interacts with MUC6 and MUC5AC, stabilizing the gastric mucus layer .

  • Antioxidant Activity: Scavenges reactive oxygen/nitrogen species in the gastric mucosa .

Tumor Suppression

  • Gastric Cancer: TFF1-deficient mice develop antro-pyloric hyperplasia and adenomas; TFF1 inhibits epithelial-mesenchymal transition (EMT) by upregulating E-cadherin and downregulating Snail, Twist, and ZEB transcription factors .

  • Breast Cancer: TFF1 overexpression reduces tumor incidence and metastasis in murine models .

Epithelial Repair

  • Cell Migration: Enhances restitution (rapid epithelial repair) via motogenic and anti-apoptotic effects .

  • Helicobacter pylori Interaction: Binds to H. pylori lipopolysaccharides, modulating bacterial adhesion .

Research Applications

TFF1 Human, His is utilized in:

ApplicationDetailsSource
Mucin Interaction StudiesAssessing binding to MUC5AC/MUC6 via ELISA or surface plasmon resonance
EMT ModelingInhibiting TGF-β/Smad signaling in gastric cancer cell lines (e.g., AGS)
Therapeutic DevelopmentTesting anti-inflammatory effects in colitis or gastritis models

Comparative Analysis of TFF1 Forms

FormMolecular WeightKey FeaturesBiological Relevance
TFF1 Human, His (Monomer)7.9 kDaHis tag, monomeric, free Cys VII thiolPurification, structural studies
TFF1 Homodimer13.2 kDaDisulfide-linked dimer, mucin cross-linkingMucus stabilization
TFF1-FCGBP Heterodimer~25 kDaDisulfide-linked with FCGBP, innate immunityMicrobial defense

Product Specs

Introduction
Trefoil Factor peptides (TFF1, TFF2, and TFF3) are stable secretory proteins found in the gastrointestinal tract, particularly the gastric mucosa. They play crucial roles in protecting and repairing the intestinal mucosa. TFF1 is particularly important for the healthy development of the antral and pyloric gastric mucosa and acts as a tumor suppressor gene specific to the stomach. It helps create a protective mucous gel layer over the gastrointestinal mucosa, acting as a barrier against harmful substances. This protection from damage, stabilization of the mucus layer, and influence on epithelial healing are key functions of TFF1. Elevated levels of TFF1 are frequently observed in tumor cells. Furthermore, TFF1 interacts with the cell membrane of MCF-7 cells. In cases of inflammatory bowel disease, serum levels of both TFF1 and TFF2 are often elevated.
Description
Recombinant Human TFF-1, produced in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 70 amino acids (residues 25-84), including a 10 amino acid His tag, resulting in a total molecular mass of 7.9 kDa. The purification of Recombinant Human TFF-1 is achieved using proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder after filtration.
Formulation
The TFF1 His Tag protein solution, with a concentration of 0.5 mg/ml, was filtered through a 0.4 μm filter. This solution, containing 20mM Tris pH-7.5 and 20mM NaCl, was then lyophilized to obtain the final product.
Solubility
To create a working stock solution, add deionized water to the lyophilized pellet, aiming for a concentration of approximately 0.5 mg/ml. Ensure complete dissolution of the pellet. This product is not sterile. Prior to use in cell culture, filter the solution through a sterile filter of appropriate pore size.
Stability
Lyophilized TFF1 His Tag remains stable at room temperature for up to 3 weeks. However, for extended storage, it is recommended to store the lyophilized product desiccated at temperatures below -18°C. Once reconstituted, TFF1 His Tag can be stored at 4°C for 2-7 days. For long-term storage after reconstitution, store below -18°C. Adding a carrier protein, such as 0.1% HSA or BSA, is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeding 95.0% as determined by SDS-PAGE analysis.
Synonyms
TFF-1, TFF1, pS2, BCEI, HPS, HP1.A, pNR-2, D21S21, pS2 protein, Trefoil factor 1, Breast cancer estrogen-inducible protein.
Source
Escherichia Coli.
Amino Acid Sequence
MKHHHHHHAS EAQTETCTVA PRERQNCGFP GVTPSQCANK GCCFDDTVRG VPWCFYPNTI DVPPEEECEF.

Q&A

What is the molecular structure of human TFF1 and how does His-tagging affect it?

Human TFF1 is a small secreted protein comprising 60 amino acids with a characteristic trefoil domain extending from residues 6-47. The domain is stabilized by three intramolecular disulfide bonds formed between Cys7-Cys33, Cys17-Cys32, and Cys27-Cys44 . A unique feature of TFF1 is the presence of a C-terminal 7th cysteine residue (Cys58) located outside the TFF domain, flanked by four glutamic acid residues (E55EEC58E59) .

When producing recombinant TFF1 with a histidine tag (TFF1-His), researchers should consider tag positioning carefully. N-terminal His-tags are generally preferred as the C-terminal region contains the unpaired Cys58 that is functionally significant. The addition of a His-tag facilitates protein purification through immobilized metal affinity chromatography (IMAC) but may potentially influence protein folding or activity, necessitating validation studies comparing tagged and untagged versions.

How stable is the TFF1 domain under reducing conditions and what implications does this have for experimental handling?

The TFF1 domain exhibits remarkable stability against reducing conditions. Experimental evidence shows that full reduction of TFF1's disulfide bonds only occurs with a large excess of reducing agent (150-fold TCEP), with no partially reduced intermediates observed . This unusual stability is supported by molecular dynamics simulations revealing that the domain substantially retains its compactness and solvent exposure even when one or two disulfide bonds are removed .

For researchers, this has significant implications:

  • Standard reducing buffers may be insufficient for complete denaturation during SDS-PAGE analysis

  • Experimental protocols requiring reduction should use higher concentrations of reducing agents than typically employed for other proteins

  • The protein maintains structural integrity in mildly reducing environments, which may be relevant for studies in different cellular compartments

What are the optimal expression systems for producing functional human TFF1-His protein?

The selection of an expression system for TFF1-His production should consider the protein's disulfide bonding requirements and post-translational modifications.

Recommended expression systems comparison:

Expression SystemAdvantagesLimitationsRecommended for
Mammalian cells (e.g., HEK293, CHO)Native-like folding and disulfide formation; appropriate post-translational modificationsLower yields; higher cost; longer production timeFunctional studies requiring authentic structure
E. coli with oxidizing cytoplasm strains (e.g., SHuffle, Origami)Higher yields; cost-effective; rapid productionMay require refolding; lacks glycosylationStructural studies; applications where glycosylation is not critical
Insect cells (Baculovirus)Proper folding; moderate yields; some post-translational modificationsMore complex than bacterial systems; intermediate costBalance between yield and authentic structure
Yeast (P. pastoris)Good secretion; proper folding; moderate costPotential hyperglycosylationApplications tolerant of different glycosylation patterns

When using bacterial systems, co-expression with disulfide isomerases or directing TFF1-His to the periplasmic space can improve correct disulfide bond formation. For mammalian systems, optimizing secretion signal sequences can enhance yields.

What purification strategy ensures high purity TFF1-His while preserving its functional properties?

A multi-step purification strategy is recommended for TFF1-His:

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-NTA resins (pH 7.4-8.0, 20-50 mM imidazole in binding buffer to reduce non-specific binding)

  • Intermediate purification: Ion-exchange chromatography (IEX) - given TFF1's acidic nature, anion exchange chromatography at pH 8.0 is effective for separating different forms (monomers, dimers)

  • Polishing step: Size exclusion chromatography (SEC) on a high-resolution column to separate monomeric, dimeric, and heterodimeric forms

  • Quality control: Verification of purity by SDS-PAGE under non-reducing and reducing conditions to confirm the presence of different oligomeric states

This approach allows separation of different TFF1 forms, which is crucial since human TFF1 exists in multiple states including monomers with free thiol groups, homodimers, and heterodimers with partners like GKN2 .

How can researchers effectively distinguish between different oligomeric forms of TFF1?

Distinguishing between TFF1 oligomeric forms requires a combination of techniques:

  • Non-reducing vs. reducing SDS-PAGE: The different forms (monomer, homodimer, heterodimers) will show distinct migration patterns under non-reducing conditions but convert to monomers under reducing conditions

  • Size exclusion chromatography: Use high-resolution columns (e.g., Sephacryl S-500) to separate forms based on molecular size

  • Anion-exchange chromatography: Different TFF1 forms show distinct elution patterns, with the TFF1-GKN2 heterodimer and TFF1-FCGBP complexes eluting at different positions than monomers

  • Mass spectrometry: To precisely identify the components in each oligomeric form, particularly for novel heterodimers

  • Thiol-specific labeling: PEG-maleimide can be used to detect the presence of free thiol groups, characteristic of monomeric TFF1 with unpaired Cys58

Research indicates that human gastric TFF1 exists predominantly in monomeric forms with free thiols, which contradicts the expectation that secretory proteins typically have all cysteines engaged in disulfide bonds .

What methodological approaches can verify the tumor suppressor function of TFF1 in experimental models?

To investigate TFF1's tumor suppressor function, researchers can employ multiple complementary approaches:

  • In vitro cellular assays:

    • Gain-of-function: Expressing TFF1 in TFF1-negative cell lines (e.g., MCF10A, MDA-MB-231) to assess changes in migration, invasion, and anchorage-independent growth

    • Loss-of-function: Knockdown TFF1 in TFF1-positive cell lines (e.g., MCF7, ZR75.1) using siRNA or CRISPR-Cas9 to evaluate oncogenic potential

  • In vivo xenograft models:

    • Implant TFF1-expressing vs. control cells in nude mice to assess tumor formation and growth rates

    • TFF1-knockdown cells show enhanced tumorigenicity compared to parental cells

  • Transgenic mouse models:

    • TFF1-deficient (TFF1-KO) mice exhibit higher incidence of mammary tumors and larger tumor sizes compared to wild-type mice when exposed to chemical carcinogens

    • Similar enhanced tumor development is observed in TFF1-KO ovary and lung tissues

  • Molecular mechanism studies:

    • Analyze signaling pathway activation/inhibition (e.g., MAPK, PI3K/AKT)

    • Investigate TFF1 binding partners using co-immunoprecipitation followed by mass spectrometry

    • Examine effects on cell cycle progression and apoptosis markers

Research consistently shows that TFF1 reduces tumor development rather than exhibiting oncogenic properties, aligning with clinical observations that patients with TFF1-positive breast primary tumors have better outcomes .

How does the unique free cysteine residue (Cys58) in TFF1 contribute to its antioxidant properties, and how can this be experimentally validated?

The free cysteine residue (Cys58) in TFF1, flanked by acidic residues (PPEEEC58EF), is thought to function as a scavenger for extracellular reactive oxygen/nitrogen species (ROS/RNS), potentially protecting the gastric mucosa from oxidative damage .

Experimental approaches to validate this function:

  • In vitro ROS/RNS scavenging assays:

    • Compare wild-type TFF1 vs. C58S mutant in H₂O₂ or peroxynitrite neutralization assays

    • Measure thiol oxidation rates using fluorescent probes like dibromobimane

  • Cellular protection assays:

    • Expose gastric epithelial cells to oxidative stressors with/without TFF1 supplementation

    • Assess cell viability, membrane integrity, and intracellular ROS levels

  • Redox state analysis:

    • Use mass spectrometry to characterize post-translational modifications of Cys58 in proteins isolated from gastric mucosa

    • Identify S-nitrosylation, S-glutathionylation, or other oxidative modifications

  • Structure-function studies:

    • Investigate how copper ions, which bind to glutamic acid residues flanking Cys58, might catalyze S-nitrosylation reactions

    • Perform site-directed mutagenesis of flanking acidic residues to assess their contribution to the reactivity of Cys58

The unique position of Cys58 outside the TFF domain and its flanking by acidic residues makes it particularly suited for redox reactions, especially in the acidic environment of the stomach where nitrogen oxide chemistry is active .

What are the molecular mechanisms behind TFF1's selective binding to MUC6 but not MUC5AC, and how does this inform gastric mucosal protection studies?

Despite TFF1 being synthesized in surface mucous cells that produce MUC5AC, binding studies show that TFF1 preferentially interacts with MUC6, which is secreted by mucous neck and antral gland cells . This unexpected finding has significant implications for understanding gastric mucosal protection.

Methodological approaches to investigate this interaction:

  • Binding specificity analysis:

    • Overlay assays using ¹²⁵I-labeled TFF1 homodimer with gastric extracts fractionated by anion-exchange chromatography or SEC

    • Compare binding patterns with lectins specific for MUC5AC vs. MUC6 (e.g., GSA-II for MUC6)

  • Glycan interaction studies:

    • Characterize the glycan structures on MUC6 that mediate TFF1 binding

    • Employ glycosidase treatments to identify critical sugar moieties

    • Utilize glycan arrays to define TFF1's carbohydrate recognition profile

  • Structural biology approaches:

    • X-ray crystallography or cryo-EM studies of TFF1-MUC6 complexes

    • NMR analysis of labeled TFF1 interacting with MUC6-derived glycopeptides

  • Functional analyses:

    • Co-localization studies in gastric tissue sections

    • Investigate how TFF1-MUC6 interaction influences mucus rheological properties

    • Assess barrier function in models with disrupted TFF1-MUC6 binding

Research indicates that TFF1's lectin-like activity may explain its binding to MUC6, similar to how TFF1 interacts with Helicobacter pylori through core oligosaccharide binding . This interaction may contribute to TFF1's role as a tumor suppressor by influencing mucosal organization and integrity.

What strategies can overcome the challenges in detecting and quantifying different TFF1 forms in biological samples?

Accurate detection and quantification of TFF1 forms in biological samples present several challenges due to the protein's complex oligomerization behavior and redox sensitivity.

Recommended analytical approaches:

  • Sample preparation optimization:

    • Use TCA/acetone precipitation followed by dissolution in 1% SDS to recover all TFF1 forms, especially the TFF1-GKN2 heterodimer which may be difficult to detect in standard extractions

    • Include protease inhibitors and alkylating agents (e.g., iodoacetamide) immediately upon sample collection to prevent artificial disulfide shuffling

  • Immunodetection considerations:

    • Develop antibodies against different epitopes to ensure detection of all forms

    • Validate antibodies against recombinant standards of each TFF1 form

    • Use non-reducing conditions when necessary to preserve oligomeric states

  • Mass spectrometry approaches:

    • Targeted multiple reaction monitoring (MRM) assays for specific TFF1 forms

    • Top-down proteomics to characterize intact proteins with post-translational modifications

    • Native MS to preserve non-covalent interactions

  • Standardization of quantification:

    • Develop recombinant standards for each TFF1 form

    • Create isotopically labeled internal standards for absolute quantification

    • Establish correction factors for differential extraction efficiency of various forms

Research shows that significant amounts of TFF1 may exist in hard-to-solubilize forms or in complexes with other proteins, requiring specialized extraction methods for complete recovery .

How can researchers effectively distinguish between experimental artifacts and true biological heterogeneity when studying TFF1?

Distinguishing genuine biological heterogeneity from artifacts is critical when studying a protein like TFF1 that exists in multiple forms sensitive to redox conditions.

Methodological safeguards:

  • Control for extraction artifacts:

    • Compare multiple extraction methods in parallel

    • Include spike-in controls of recombinant TFF1 forms

    • Process samples with minimal delay and under controlled temperature/pH conditions

  • Redox state preservation:

    • Use oxygen-free buffers when appropriate

    • Add thiol-blocking agents at the time of sample collection

    • Compare results with and without reducing agents to identify artifactual disulfide formation

  • Validation across techniques:

    • Confirm observations using orthogonal methods (e.g., western blotting, mass spectrometry, functional assays)

    • Compare in vitro observations with in vivo imaging when possible

  • Biological replicates and controls:

    • Include appropriate tissue/cell controls where TFF1 is absent

    • Analyze samples from multiple individuals/animals

    • Use TFF1-knockout models as negative controls

  • Data interpretation frameworks:

    • Establish clear criteria for distinguishing physiological heterogeneity from artifacts

    • Consider statistical approaches for pattern recognition across multiple samples

    • Document all sample handling conditions meticulously

Research indicates that TFF1 heterogeneity is genuine biological phenomenon, with forms like the TFF1-GKN2 heterodimer being reproducibly detected across multiple studies and species when appropriate extraction methods are used .

Product Science Overview

Introduction

Trefoil Factor-1 (TFF1), also known as pS2, is a small peptide that belongs to the trefoil factor family (TFF) of proteins. These proteins are characterized by the presence of one or more trefoil domains, which are compact, stable structures that play a crucial role in mucosal protection and repair. TFF1 is predominantly expressed in the epithelial cells of the stomach and is involved in maintaining the integrity of the gastrointestinal mucosa.

Biological Properties and Functions

TFF1 is a highly conserved protein that mediates mucosal repair by stimulating cell migration, inhibiting apoptosis, and promoting the barrier function of mucus . It forms a homodimer via a disulfide linkage, which is essential for its wound healing activity . TFF1 is also known to stabilize the mucous gel layer that covers the gastrointestinal mucosa, providing a physical barrier against various noxious agents .

Recombinant Expression

The recombinant form of TFF1 (Human Recombinant, His Tag) is produced using genetic engineering techniques. The gene encoding TFF1 is synthesized and expressed in host cells such as Escherichia coli or Brevibacillus choshinensis . The recombinant protein is then purified using affinity chromatography, typically involving a His tag, which allows for efficient purification.

Applications

Recombinant TFF1 has significant potential in therapeutic applications, particularly in the treatment of gastric damage and wound healing . Studies have shown that recombinant TFF1 produced by Brevibacillus choshinensis exhibits better wound healing capabilities compared to that produced by Escherichia coli . This enhanced activity is attributed to the glycosylation of the recombinant protein, which may contribute to its improved functionality .

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.