FHIT Human, GST

Fragile Histidine Triad Human Recombinant, GST Tag
Shipped with Ice Packs
In Stock

Description

Biochemical Function and Mechanism

FHIT Human GST exhibits tumor suppressor activity through multiple pathways:

Primary Enzymatic Activity

  • Substrate: Ap₃A → AMP + ADP

  • Active Site: Requires conserved histidine residues (e.g., H96) for hydrolysis

  • Role: The FHIT-Ap₃A complex may act as a signaling molecule, inducing apoptosis in cancer cells

Interactions and Pathways

PartnerInteraction TypeFunctional Impact
Gαq ProteinsDirect binding (activated state)Modulates GPCR signaling, enhancing Fhit’s growth-inhibitory effects
Ferredoxin Reductase (Fdxr)Mitochondrial bindingIncreases reactive oxygen species (ROS) production and Ca²⁺ uptake, sensitizing cells to apoptosis
Src KinasePhosphorylation at Y114Regulates Fhit stability and degradation

Clinical Relevance in Cancer

FHIT inactivation is observed in >60% of human tumors, including lung, breast, colon, and gastric cancers .

Mechanisms of Inactivation

MechanismExamples
Chromosomal DeletionsHomozygous or hemizygous deletions at 3p14.2 (FRA3B fragile site)
Transcriptional AberrationsAberrant splicing or truncated transcripts
Proteasomal DegradationInduced by Src-mediated phosphorylation

Therapeutic Potential

  • Re-expression in Tumors: Restoring FHIT in deficient cells induces apoptosis and suppresses tumorigenicity

  • Synergy with Tumor Suppressors: Cooperates with VHL and HER2/neu to inhibit cancer progression

Research Applications

FHIT Human GST is widely used in:

Experimental Techniques

ApplicationMethodPurpose
Co-immunoprecipitationGST pull-down assaysIdentify protein interactions (e.g., Gαq, Fdxr)
Enzymatic AssaysAp₃A hydrolysis quantificationMeasure catalytic activity
Cellular StudiesStable overexpression in cancer linesAssess apoptosis, proliferation, and drug sensitivity

Key Findings from Functional Studies

StudyObservation
Zanesi et al. (1997)Hydrolase activity is not required for tumor suppression
Li et al. (2013)GST-tagged Fhit binds activated Gαq proteins via α2-β4 region
Duolink AssayConfirms endogenous Fhit-Fdxr interaction in mitochondria

Table 1: FHIT Gene and Protein Variants

Transcript IDLength (bp)Protein Length (aa)UniProt Match
ENST00000492590.63116147P49789
ENST00000468189.51634147P49789
ENST00000488467.560178E9PBZ0

Table 2: Specific Activity of FHIT in Transfected Cell Lines

Cell LineSpecific Activity (nmol/min/mg)
293/CMV0.62
293/FHIT clNK324.2
293/FHIT H96N cl30.62

Source: Zanesi et al. (1997)

Product Specs

Introduction
The FHIT gene, a member of the histidine triad family, encodes a diadenosine 5',5'''-P1,P3-triphosphate hydrolase that plays a role in purine metabolism. This gene encompasses the fragile site FRA3B on chromosome 3, a region susceptible to carcinogen-induced damage that can lead to translocations and the production of abnormal transcripts. FHIT acts as a tumor suppressor, with reduced or absent expression observed in various cancer types. It is suggested that FHIT may also exhibit tumor suppressor activity in normal cells. Alterations and deletions affecting the FHIT gene are strongly associated with the development and progression of human tumors, particularly in the lung, cervix, breast, colon, stomach, and pancreas. The loss of FHIT expression is more prevalent in cancers of individuals carrying familial mutations in DNA repair genes like BRCA1, BRCA2, and MSH2. In vitro studies have demonstrated that FHIT functions as a hydrolase, cleaving diadenosine triphosphate (Ap3A) into ADP and AMP. The FHIT-Ap3A enzyme-substrate complex is thought to be crucial for its tumor suppressor function. Restoring FHIT expression in FHIT-deficient cancer cells triggers apoptosis through the intrinsic caspase pathway, both in vitro and in tumor xenografts.
Description
This product consists of full-length recombinant human FHIT protein expressed in E.coli, exhibiting a molecular weight of 43 kDa on SDS-PAGE. The FHIT protein is fused to a GST-Tag and purified using proprietary chromatographic techniques.
Physical Appearance
Clear solution that has been sterilized by filtration.
Formulation
The FHIT protein is supplied in a buffer consisting of 50mM Tris-Acetate (pH 7.5), 1mM EDTA, and 20% Glycerol.
Stability
For long-term storage, it is recommended to store the vial at a temperature between -20°C and -80°C. Under these conditions, the protein remains stable for a period of 12 months. It is important to avoid repeated freeze-thaw cycles to maintain protein integrity.
Applications
- Enzyme-Linked Immunosorbent Assay (ELISA) - Inhibition Assays - Western Blotting
Synonyms
Bis(5'-adenosyl)-triphosphatase, EC 3.6.1.29, Diadenosine 5',5'''-P1,P3-triphosphate hydrolase, Dinucleosidetriphosphatase, AP3A hydrolase, AP3Aase, Fragile histidine triad protein, FHIT, FRA3B.
Source
Escherichia Coli.

Q&A

What is the genomic structure of the FHIT gene and how does it relate to its fragility?

The FHIT gene spans approximately 1.6 Mb of genomic DNA and comprises ten exons. It encompasses the FRA3B common fragile site on chromosome 3p14.2, which represents one of the most susceptible regions to breakage in the human genome . This extensive genomic structure partially explains the gene's vulnerability to carcinogen-induced damage, resulting in various structural alterations including deletions, translocations, and aberrant transcripts. The gene encodes a relatively small 1.1 kb mRNA that is expressed at low levels in most tissues . The disparity between the expansive genomic region and the compact transcript highlights why FHIT is frequently targeted by genomic rearrangements in multiple human cancers.

Methodologically, researchers investigating FHIT's genomic structure should employ long-range PCR techniques, chromosome walking, and next-generation sequencing approaches to accurately map deletion breakpoints and structural variations.

How is FHIT's catalytic function related to its tumor suppressor activity?

The tumor suppressor signal appears to be the FHIT-Ap3A enzyme-substrate complex rather than the hydrolysis products themselves . This finding represents a significant methodological insight for researchers, suggesting that experiments should focus on FHIT protein-substrate interactions rather than solely on hydrolase activity measurements when investigating tumor suppression mechanisms.

What is the molecular mechanism through which FHIT acts as a tumor suppressor?

FHIT exerts its tumor suppressive function through direct interaction with the β-catenin signaling pathway. FHIT associates with the lymphoid enhancer-binding factor 1/T cell factor/β-catenin complex by directly binding to the C-terminal domain of β-catenin, a major player in the canonical Wnt pathway that is deregulated in numerous human cancers . This interaction results in the repression of transcription of target genes such as cyclin D1, axin2, MMP-14, and survivin .

Through chromatin immunoprecipitation (ChIP) experiments, researchers have confirmed that FHIT and β-catenin form complexes at the promoters of these target genes. Two-step ChIP assays have verified that FHIT and β-catenin are associated in a complex at these promoters . Methodologically, researchers investigating FHIT's tumor suppressor function should employ a combination of co-immunoprecipitation, ChIP, and reporter gene assays to fully characterize these molecular interactions.

How does FHIT expression correlate with cancer progression and prognosis?

Loss of FHIT expression has been established as an early event during multistep carcinogenesis and correlates with tumor progression and worse prognosis in multiple cancer types . FHIT expression is frequently lost in cancers of individuals with familial mutations causing deficiencies in DNA repair genes, including BRCA1, BRCA2, and MSH2 .

Erroneous FHIT transcripts have been detected in approximately half of all esophageal carcinomas, stomach carcinomas, and other cancer types . Researchers have observed that structural alterations tend to result from deletions in both FHIT alleles, leading to loss of exons and subsequent absence of full-length FHIT transcript and protein .

For methodological approaches, researchers should employ immunohistochemistry, quantitative RT-PCR, and Western blotting in both primary tumor samples and adjacent normal tissues to accurately assess FHIT expression levels and correlate these with clinical outcomes and histopathological parameters.

What are the optimal experimental approaches for reconstituting FHIT expression in cancer cells?

When designing experiments to reconstitute FHIT expression in FHIT-deficient cancer cells, researchers should consider multiple methodological approaches:

  • Transfection systems: Transient transfection with wild-type FHIT has been successfully used in cancer cell lines such as SW480 and MCF-7 . For stable expression, lentiviral or retroviral systems provide more consistent results.

  • Expression verification: Following transfection, FHIT expression should be confirmed at both mRNA (real-time RT-PCR) and protein levels (Western blotting) .

  • Functional validation: After reconstituting FHIT expression, researchers should assess:

    • Changes in target gene expression (cyclin D1, axin2, MMP-14, survivin)

    • Effects on cell growth, apoptosis, and cell cycle progression

    • Impact on anchorage-independent growth using soft agar assays

    • Alterations in tumorigenicity in nude mice xenograft models

  • Controls: Experiments should include both negative controls (empty vector) and FHIT mutants (e.g., hydrolase-dead mutants) to distinguish between enzymatic and non-enzymatic functions .

This methodological framework enables comprehensive evaluation of FHIT's tumor-suppressive effects in various cancer models.

What techniques are most effective for studying FHIT-protein interactions?

To investigate FHIT's interactions with other proteins, particularly β-catenin, researchers should employ a multi-technique approach:

  • Co-immunoprecipitation (Co-IP): This technique allows detection of endogenous protein-protein interactions. Studies have successfully used Co-IP to demonstrate that Fhit associates with the lymphoid enhancer-binding factor 1/T cell factor/β-catenin complex .

  • Chromatin Immunoprecipitation (ChIP): ChIP experiments have confirmed that FHIT and β-catenin are recruited to the promoters of target genes such as axin2, cyclin D1, MMP14, and survivin .

  • Two-step ChIP assays: These have been used to prove that FHIT and β-catenin are associated in a complex at target gene promoters .

  • GST pull-down assays: For in vitro validation of direct interactions, GST-tagged FHIT can be used to pull down potential binding partners.

  • Protein domain mapping: To identify the specific domains involved in protein-protein interactions, researchers should generate deletion mutants and perform interaction studies.

These methodological approaches provide complementary information about FHIT's interactome and functional protein complexes.

How does FHIT interact with multiple signaling pathways beyond the Wnt/β-catenin pathway?

FHIT's tumor suppressor activity extends beyond the Wnt/β-catenin pathway, interacting with several other key signaling networks:

  • NF-κB Pathway: FHIT has been reported to inhibit the NF-κB signaling pathway, suggesting a role in modulating inflammatory responses and cell survival .

  • Ras/Rho GTPase Signaling: Research indicates that FHIT targets multiple components of Ras/Rho GTPase signaling, potentially affecting cell motility, morphology, and invasion capabilities .

  • EGFR-Src Axis: FHIT is a target of Src kinase, and Src-mediated phosphorylation of FHIT in response to EGF receptor stimulation induces proteasomal degradation of FHIT . This suggests FHIT's involvement in growth factor signaling regulation.

  • Akt-Survivin Pathway: FHIT has been shown to modulate the Akt-survivin pathway through tyrosine phosphorylation-dependent mechanisms, contributing to its proapoptotic activity .

Methodologically, researchers investigating these pathway interactions should employ phosphorylation-specific antibodies, kinase inhibitors, and pathway-specific reporter assays to delineate the precise mechanisms of FHIT's involvement in each signaling network.

What is the relationship between FHIT and apoptotic mechanisms in cancer cells?

FHIT exhibits proapoptotic activity that contributes to its tumor suppressor function. In cancer-derived cells and tumor xenografts, restoring FHIT expression in FHIT-deficient cancer cells triggers apoptosis via the intrinsic caspase pathway .

The molecular mechanisms underlying this process involve:

  • Survivin regulation: FHIT contributes to reduced expression of survivin at the mRNA level, as demonstrated in SW480 and MCF-7 cells . Since survivin is an inhibitor of apoptosis, its downregulation promotes apoptotic cell death.

  • Akt pathway modulation: FHIT's proapoptotic activity appears to be mediated by tyrosine phosphorylation-dependent modulation of the Akt-survivin pathway .

  • β-catenin target gene repression: By repressing β-catenin-mediated transcription of survival genes, FHIT may shift the balance toward apoptosis.

Researchers investigating FHIT's role in apoptosis should employ flow cytometry with Annexin V/PI staining, TUNEL assays, and measurement of caspase activation to quantify apoptotic responses. Western blotting for key apoptotic proteins (including survivin, caspases, and Bcl-2 family members) provides additional mechanistic insights.

How might FHIT's evolutionary conservation as a human accelerated region inform its functional studies?

FHIT is recognized as a human accelerated region that may have played an important role in distinguishing humans from apes . This evolutionary aspect raises intriguing questions about FHIT's functions beyond tumor suppression.

For researchers exploring this dimension, methodological approaches should include:

  • Comparative genomics: Analyzing FHIT sequence and structure across primate species can identify human-specific changes.

  • Functional conservation studies: Testing whether FHIT proteins from different species can complement human FHIT deficiency in cancer cells.

  • Tissue-specific expression analysis: Examining whether FHIT expression patterns differ in human versus non-human primate tissues, particularly in brain and other organs that underwent significant evolution in humans.

  • Interactome comparison: Identifying species-specific interaction partners using proteomics approaches.

These evolutionary insights may reveal novel functions of FHIT related to human-specific traits or disease susceptibilities.

What are the implications of FHIT's involvement in DNA damage response for targeted cancer therapies?

FHIT's expression is more commonly lost in cancers of individuals with familial mutations causing deficiencies in DNA repair genes including BRCA1, BRCA2, and MSH2 . This association suggests potential involvement in DNA damage response pathways.

Researchers exploring this connection should consider these methodological approaches:

  • Synthetic lethality screening: Testing whether FHIT-deficient cells show increased sensitivity to specific DNA-damaging agents or PARP inhibitors.

  • DNA damage response assays: Measuring γ-H2AX foci formation, comet assays, and chromosome aberrations in FHIT-proficient versus deficient cells after exposure to various genotoxic agents.

  • Protein-protein interaction studies: Investigating whether FHIT directly interacts with components of DNA repair machinery.

  • Therapeutic combination testing: Evaluating whether FHIT status influences response to therapies targeting DNA repair pathways.

This research direction could identify new therapeutic vulnerabilities in FHIT-deficient tumors and inform personalized treatment strategies based on FHIT status.

Product Science Overview

Introduction

The Fragile Histidine Triad (FHIT) protein is a member of the histidine triad gene family. The FHIT gene encodes a diadenosine 5’,5’‘’-P1,P3-triphosphate hydrolase, an enzyme involved in purine metabolism . This gene includes the common fragile site FRA3B on chromosome 3, where carcinogen-induced damage can lead to translocations and abnormal transcripts of this gene .

Structure and Expression

The FHIT protein is expressed as a human recombinant protein with a Glutathione S-transferase (GST) tag. This recombinant protein is produced in Escherichia coli and shows a 43 kDa band on SDS-PAGE . The GST tag aids in the purification and detection of the protein. The FHIT protein is a single, non-glycosylated polypeptide chain containing 155 amino acids and has a molecular mass of 17.9 kDa .

Function

The primary function of the FHIT protein is to act as a hydrolase that cleaves diadenosine triphosphate (Ap3A) to ADP and AMP . This enzyme-substrate complex is believed to be the tumor suppressor signal . The FHIT protein plays a crucial role in purine metabolism and is involved in the regulation of various cellular processes.

Role in Cancer

The FHIT gene is a tumor suppressor gene, and its expression is often reduced or lost in numerous types of cancer, including lung, cervix, breast, colon, stomach, and pancreas . Alterations and deletions of the FHIT gene are strongly linked to the genesis and establishment of these tumors . The restoration of FHIT expression in FHIT-deficient cancer cells leads to apoptosis, involving the intrinsic caspase pathway, in cancer-derived cells and in tumor xenografts .

Applications

The recombinant FHIT protein with a GST tag is used in various laboratory research applications, including ELISA, inhibition assays, and Western blotting . It is essential for studying the protein’s function, interactions, and role in cancer biology.

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.