FBXO2 Human

F-Box Protein 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

FBXO2 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 319 amino acids (1-296 a.a.) and having a molecular mass of 35.7kDa.
FBXO2 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
F-Box Protein 2 (FBXO2) is a member of the F-box protein family. These proteins constitute one of the four subunits of the ubiquitin protein ligase complex known as SCFs (SKP1-cullin-F-box), which participates in phosphorylation-dependent ubiquitination. F-box proteins are categorized into three classes, with FBXO2 belonging to the Fbxs class. This class is characterized by containing diverse protein-protein interaction modules or lacking recognizable motifs. FBXO2 exhibits high similarity to the rat neural F Box 42 kDa protein, which is predominantly found in the nervous system and contributes to maintaining neurons in a postmitotic state.
Description
Recombinant human FBXO2, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 319 amino acids (specifically, amino acids 1 to 296). It possesses a molecular weight of 35.7 kDa. For purification purposes, FBXO2 is fused to a 23 amino acid His-tag at its N-terminus and subsequently purified using proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless solution.
Formulation
The FBXO2 protein solution is provided at a concentration of 0.25 mg/ml and is prepared in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 30% glycerol, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. It is essential to minimize freeze-thaw cycles.
Purity
The purity of FBXO2 is determined to be greater than 80.0% by SDS-PAGE analysis.
Synonyms
F-box only protein 2, FBXO2, F-Box Protein 2, FBX2, FBG1, Fbs1, NFB42, OCP1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMDGDGDP ESVGQPEEAS PEEQPEEASA EEERPEDQQE EEAAAAAAYL DELPEPLLLR VLAALPAAEL VQACRLVCLR WKELVDGAPL WLLKCQQEGL VPEGGVEEER DHWQQFYFLS KRRRNLLRNP CGEEDLEGWC DVEHGGDGWR VEELPGDSGV EFTHDESVKK YFASSFEWCR KAQVIDLQAE GYWEELLDTT QPAIVVKDWY SGRSDAGCLY ELTVKLLSEH ENVLAEFSSG QVAVPQDSDG GGWMEISHTF TDYGPGVRFV RFEHGGQDSV YWKGWFGARV TNSSVWVEP.

Q&A

What is the molecular structure and function of FBXO2?

FBXO2 is a specialized F-box protein that functions as a substrate-recognizing component within the SCF E3 ubiquitin ligase complex. Its structure includes:

  • An F-box domain that binds to Skp1 within the SCF complex

  • A C-terminal FBA (F-Box Associated) domain responsible for substrate recognition

  • Critical residues Tyr-278 (Y278) and Trp-279 (W279) in the FBA domain that specifically recognize N-glycosylated proteins

Unlike most F-box proteins, FBXO2 uniquely targets N-glycosylated proteins for ubiquitination, marking them for proteasomal degradation. This specificity makes FBXO2 particularly important in quality control of glycoproteins in various cellular compartments .

How does FBXO2 expression vary across human tissues and cell types?

FBXO2 shows variable expression patterns across tissues with particularly notable expression in:

  • Brain tissue: Where it regulates NMDA receptor subunits

  • Liver tissue: Where it influences glucose metabolism pathways

  • Cancer tissues: Often showing dysregulated expression compared to adjacent non-tumor tissues

Interestingly, while transcriptomic data may show upregulation of FBXO2 mRNA in some cancers (such as HCC), protein levels can be downregulated due to post-transcriptional mechanisms, specifically through proteasomal degradation mediated by other ubiquitin ligases like SKP2 . This discrepancy between mRNA and protein levels highlights the importance of examining both transcript and protein expression when studying FBXO2 function.

What are the validated substrates of FBXO2 and how can new substrates be identified?

Validated FBXO2 substrates include:

  • GluN1 and GluN2A (NMDAR subunits) in neurons

  • Hsp47 (Heat shock protein 47) in hepatocellular carcinoma cells

Methods for identifying new FBXO2 substrates:

  • Affinity purification-mass spectrometry (AP-MS): Perform immunoprecipitation with tagged FBXO2 followed by mass spectrometry analysis to identify interacting proteins. This approach successfully identified Hsp47 as an FBXO2 substrate in HCC cells .

  • Protein stability assays: Compare protein half-lives in FBXO2-deficient versus FBXO2-expressing cells using cycloheximide chase experiments. True substrates should show extended half-lives in FBXO2-knockout conditions.

  • Ubiquitination assays: Perform in vitro and in vivo ubiquitination assays with candidate substrates, focusing on K48-linked ubiquitination as the primary signal for proteasomal degradation. For example, FBXO2 promotes K48-linked ubiquitination of Hsp47 .

  • Glycosylation dependency tests: Since FBXO2 specifically recognizes N-glycosylated proteins, potential substrates should be analyzed for N-glycosylation sites and tested with glycosylation inhibitors to confirm dependency.

How is FBXO2 itself regulated at the post-translational level?

FBXO2 undergoes several post-translational modifications that regulate its stability and function:

  • Ubiquitination by SKP2:

    • FBXO2 is targeted for proteasomal degradation by another F-box protein, SKP2

    • This creates a regulatory network where one F-box protein controls another

  • Phosphorylation by DNA-PKcs:

    • DNA-dependent protein kinase catalytic subunit (DNA-PKcs) phosphorylates FBXO2

    • This phosphorylation is required for SKP2-mediated ubiquitination

    • Inhibition of DNA-PKcs prevents SKP2-FBXO2 interaction and stabilizes FBXO2

  • Protein half-life regulation:

    • FBXO2 has a relatively short half-life that can be extended by proteasome inhibitors

    • This allows for rapid changes in FBXO2 levels in response to cellular needs

This multi-layered regulation explains the discrepancy between FBXO2 mRNA and protein levels observed in some cancers and provides potential intervention points for therapeutic strategies .

What is the role of FBXO2 in hepatocellular carcinoma progression?

FBXO2 functions as a tumor suppressor in hepatocellular carcinoma (HCC) through the following mechanisms:

  • Inhibition of epithelial-mesenchymal transition (EMT):

    • FBXO2 targets Hsp47 (a collagen-specific chaperone) for degradation

    • Hsp47 normally promotes EMT and metastasis

    • FBXO2 depletion stabilizes Hsp47, enhancing EMT marker expression and promoting metastasis

  • Clinical correlations:

    • Low FBXO2 expression correlates with more advanced tumor AJCC and Edmonds stages

    • Patients with FBXO2ᶫᵒʷ tumors have significantly worse median survival

    • FBXO2 expression shows inverse correlation with SKP2 and Hsp47 levels in patient samples

  • Regulatory mechanism in HCC:

    • FBXO2 is frequently downregulated at the protein level in HCC despite mRNA upregulation

    • This occurs through SKP2-mediated ubiquitination and proteasomal degradation

    • The SKP2-FBXO2-Hsp47 axis represents a potential therapeutic target in HCC

How does FBXO2 modulate neuronal function and what are the implications for neurological disorders?

FBXO2 plays a critical role in regulating NMDA receptors in the central nervous system:

  • Regulation of NMDA receptor subunits:

    • FBXO2 facilitates the degradation of specific NMDAR subunits (GluN1 and GluN2A)

    • Loss of FBXO2 results in increased surface localization of these subunits

    • Interestingly, not all NMDAR subunits are affected; GluN2B levels remain unchanged in FBXO2 knockout mice

  • Synaptic changes in FBXO2 knockout mice:

    • Increased synaptic markers PSD-95 and Vglut1

    • Increased axo-dendritic shaft synapses

    • These changes occur without alterations in dendritic spine density or significant neurophysiological differences

  • Implications for neurological disorders:

    • NMDAR dysregulation is linked to cognitive deficits and glutamate-induced excitotoxicity

    • FBXO2's selective regulation of specific NMDAR subunits suggests it may play a role in fine-tuning excitatory neurotransmission

    • This makes FBXO2 a potential therapeutic target in conditions with NMDAR dysfunction such as epilepsy, neurodegenerative diseases, and certain psychiatric disorders

What evidence links FBXO2 to metabolic disorders and glucose homeostasis?

Recent research has revealed a connection between FBXO2 and metabolic regulation:

  • Role in hepatic glucose metabolism:

    • FBXO2 downregulation after sleeve gastrectomy (SG) is associated with improved glucose homeostasis

    • This improvement operates through activation of the PI3K-AKT pathway

    • Hepatic-specific overexpression of FBXO2 can partially reverse the glucose homeostasis benefits of sleeve gastrectomy

  • Connection to type 2 diabetes and NAFLD:

    • FBXO2 appears to be involved in the mechanism by which bariatric surgery relieves type 2 diabetes

    • Its regulation may connect dietary factors and metabolic disease progression

    • FBXO2 represents a potential non-invasive therapeutic target for metabolic disorders

  • Regulatory network:

    • FBXO2 likely targets specific glycosylated proteins in metabolic pathways

    • Its expression is influenced by dietary factors and surgical interventions

    • The exact substrates mediating these metabolic effects require further investigation

What are optimal methods for studying FBXO2 function in cell and animal models?

Cell Culture Models:

  • Gene Manipulation Approaches:

    • CRISPR/Cas9 for complete knockout

    • siRNA/shRNA for transient knockdown (effective in HCC cell lines like Hep3B and HCC-LM3)

    • Overexpression using lentiviral or plasmid vectors with wild-type or mutant FBXO2 constructs

    • Site-directed mutagenesis of key residues (e.g., Y278A, W279A) to study substrate recognition mechanisms

  • Substrate Interaction Analysis:

    • Co-immunoprecipitation assays to detect FBXO2-substrate interactions

    • Cycloheximide chase experiments to measure protein half-lives

    • In vivo ubiquitination assays using K48-specific antibodies

Animal Models:

  • FBXO2 Knockout Mice:

    • Complete knockout models have been generated and characterized, particularly for brain studies

    • Conditional tissue-specific knockouts using Cre-loxP system for organ-specific analysis

  • Viral-Mediated Gene Delivery:

    • AAV8-mediated hepatic expression for liver-specific studies

    • Adenoviral delivery for acute expression in various tissues

  • Disease-Specific Models:

    • HCC xenograft models to study tumor growth and metastasis

    • High-fat diet models combined with FBXO2 manipulation to study metabolic effects

    • Tumor metastasis models with tracking (e.g., luciferase-expressing cells for in vivo imaging)

How can contradictory findings about FBXO2 across different tissue types be reconciled?

Researchers frequently encounter seemingly contradictory findings about FBXO2 across different experimental systems. These can be reconciled through:

  • Context-Dependent Function Analysis:

    • Recognize that FBXO2 may target different substrates in different tissues

    • In HCC, it primarily targets Hsp47, affecting EMT and metastasis

    • In neurons, it regulates NMDAR subunits, affecting synaptic composition

    • In liver metabolism, it influences glucose homeostasis through yet undefined targets

  • Expression Level Verification:

    • Always measure both mRNA and protein levels

    • Transcriptional upregulation doesn't necessarily translate to increased protein due to post-translational regulation

    • Use multiple antibodies targeting different epitopes to confirm specificity

  • Substrate Specificity Analysis:

    • Verify glycosylation status of putative substrates in the tissue of interest

    • Perform glycosidase treatments to confirm glycosylation-dependent interactions

    • Use FBXO2 mutants defective in glycoprotein binding (Y278A, W279A) as controls

  • Regulatory Network Mapping:

    • Determine tissue-specific expression of FBXO2 regulators (e.g., SKP2, DNA-PKcs)

    • Map tissue-specific interactomes using proximity labeling or AP-MS approaches

    • Consider compensatory mechanisms from related F-box proteins

What are the structural determinants of FBXO2's glycoprotein substrate specificity?

FBXO2's unique glycoprotein recognition capacity depends on specific structural features:

  • FBA Domain Structure:

    • The FBA domain forms a specific binding pocket for N-glycans

    • Residues Y278 and W279 are critical for this interaction

    • Mutations in these residues (e.g., W279A) abolish substrate binding and subsequent ubiquitination

  • Substrate Recognition Determinants:

    • Primary sequence context surrounding N-glycosylation sites likely contributes to specificity

    • Tertiary structure and accessibility of glycan moieties affect recognition efficiency

    • Not all N-glycosylated proteins are FBXO2 substrates, suggesting additional determinants

  • Experimental Approaches to Map Specificity:

    • Systematic mutagenesis of FBA domain residues

    • Glycan array screening to determine glycan structure preferences

    • Structural studies (X-ray crystallography, cryo-EM) of FBXO2-substrate complexes

    • Computational modeling of interaction interfaces

  • Substrate Competition Analysis:

    • Study how different glycoprotein substrates compete for FBXO2 binding

    • Determine hierarchy of substrate preference under various cellular conditions

    • Investigate whether FBXO2 post-translational modifications alter substrate preferences

How do the dual roles of FBXO2 in cancer (both tumor-promoting and tumor-suppressive) reconcile across different cancer types?

FBXO2 exhibits context-dependent roles in cancer, functioning as:

  • Tumor Suppressor in HCC:

    • Targets Hsp47 for degradation, inhibiting EMT and metastasis

    • Low expression correlates with worse clinical outcomes

    • Frequently downregulated via SKP2-mediated degradation

  • Reported Oncogenic Roles in Other Cancers:

    • Promotes development of endometrial cancer, gastric cancer, and osteosarcoma through distinct pathways

    • These divergent roles likely reflect tissue-specific substrates

  • Reconciliation Approaches:

    • Substrate Profiling: Comprehensive identification of FBXO2 substrates in different cancer types

    • Regulatory Network Analysis: Mapping of FBXO2 regulators (e.g., SKP2) across cancer types

    • Genetic Background Consideration: Analysis of how mutations in related pathway components alter FBXO2 function

    • Microenvironment Effects: Investigation of how tumor microenvironment signals modulate FBXO2 activity

  • Experimental Design for Clarification:

    • Meta-analysis of FBXO2 expression across cancer types using multi-omics approaches

    • Cancer-specific knockout and overexpression models examining common endpoints

    • Patient-derived xenografts to preserve tumor heterogeneity

    • Analysis of FBXO2 mutations and polymorphisms across cancer databases

What is the potential for developing FBXO2-targeting therapeutics for various diseases?

The diverse roles of FBXO2 present several therapeutic opportunities:

  • Strategies for FBXO2 Modulation:

    • Protein Stabilization: Inhibitors of SKP2 or DNA-PKcs to prevent FBXO2 degradation in cancers where it acts as a tumor suppressor

    • Protein Inhibition: Small molecules targeting the substrate-binding pocket for conditions requiring FBXO2 inhibition

    • Expression Modulation: Transcriptional or epigenetic approaches to alter FBXO2 expression levels

  • Disease-Specific Applications:

    • HCC: Strategies to stabilize FBXO2, enhancing its tumor-suppressive functions

    • Neurological Disorders: Modulation of FBXO2 activity to fine-tune NMDAR signaling

    • Metabolic Disorders: Targeting hepatic FBXO2 to improve glucose homeostasis

  • Delivery Challenges and Solutions:

    • Tissue-specific delivery systems (nanoparticles, AAV vectors)

    • Cell-penetrating peptides fused to FBXO2-modulating domains

    • Small molecule approaches for better bioavailability

  • Biomarker Development:

    • FBXO2 expression levels as prognostic indicators in HCC

    • Ratios of FBXO2 to SKP2 or FBXO2 to Hsp47 as predictive biomarkers

    • Monitoring FBXO2 substrates as pharmacodynamic markers

How does FBXO2 function within the broader ubiquitin-proteasome system during cellular stress responses?

FBXO2's specialized role in glycoprotein quality control suggests important functions during cellular stress:

  • ER Stress Response:

    • N-glycosylation occurs in the ER, and misfolded glycoproteins trigger ER stress

    • FBXO2 may participate in ER-associated degradation (ERAD) of misfolded glycoproteins

    • Research should investigate FBXO2 activity during unfolded protein response activation

  • Oxidative Stress Conditions:

    • Oxidative damage can affect protein folding and glycosylation patterns

    • FBXO2's substrate preference may shift under oxidative stress

    • Comparative proteomic analysis of FBXO2 substrates under normal vs. oxidative stress conditions would be informative

  • Methodological Approaches:

    • Stress-specific interactome analysis using BioID or APEX proximity labeling

    • Quantitative glycoproteomics in FBXO2-deficient vs. wild-type cells under stress

    • Live-cell imaging of FBXO2 localization during stress response phases

  • Integration with Other Quality Control Systems:

    • Investigate crosstalk between FBXO2 and other quality control systems (e.g., autophagy)

    • Determine hierarchy and compensatory mechanisms when FBXO2 is compromised

What is the evolutionary significance of FBXO2's specialized glycoprotein recognition capability?

The unique glycoprotein-targeting ability of FBXO2 raises interesting evolutionary questions:

  • Comparative Genomics Analysis:

    • Trace FBXO2 evolution across species, identifying when glycoprotein recognition emerged

    • Compare FBXO2 orthologs for conservation of key residues (Y278, W279) and domain organization

    • Identify species-specific adaptations in substrate recognition

  • Functional Divergence from Related F-box Proteins:

    • Compare FBXO2 with closely related family members (e.g., FBXO6, FBXO17, FBXO27)

    • Determine whether they have overlapping or complementary substrate preferences

    • Investigate tissue-specific expression patterns of FBXO family members

  • Co-evolution with Glycosylation Machinery:

    • Analyze evolutionary relationships between FBXO2 and N-glycosylation pathway components

    • Compare across species with different glycosylation patterns and complexity

    • Identify potential evolutionary pressures (e.g., pathogen interactions, tissue specialization)

  • Experimental Approaches:

    • Reconstruction of ancestral FBXO2 sequences to test substrate recognition

    • Cross-species complementation studies in FBXO2-knockout models

    • Systematic comparison of substrate preferences across species

What are the critical considerations for experimental design when studying the FBXO2-substrate relationship?

Rigorous experimental approaches are essential for accurately characterizing FBXO2-substrate relationships:

  • Controls for Glycosylation Dependency:

    • Include glycosidase treatments (PNGase F, Endo H) to verify N-glycan dependency

    • Use FBXO2 mutants (Y278A, W279A) defective in glycoprotein binding as negative controls

    • Construct substrate mutants lacking N-glycosylation sites to confirm direct targeting

  • Ubiquitination Assay Design:

    • Use K48-specific ubiquitin antibodies to detect degradation-targeting ubiquitination

    • Include proteasome inhibitors to prevent substrate degradation during analysis

    • Perform in vitro reconstitution with purified components to confirm direct effects

  • Protein Half-life Measurements:

    • Use cycloheximide chase assays with appropriate time points for each substrate

    • Include both FBXO2 overexpression and depletion conditions

    • Control for indirect effects through complementation with catalytically inactive FBXO2

  • Consideration of Indirect Effects:

    • Monitor mRNA levels of putative substrates to rule out transcriptional effects

    • Check for effects on global protein synthesis and degradation pathways

    • Consider potential feedback loops where substrate function affects FBXO2 expression

How can researchers address the challenge of distinguishing direct from indirect effects of FBXO2 manipulation?

This is a critical challenge in E3 ligase research, requiring multiple complementary approaches:

  • Biochemical Validation Pipeline:

    • Step 1: Demonstrate physical interaction (co-IP, proximity labeling)

    • Step 2: Show ubiquitination dependency on FBXO2 (in vitro and in vivo assays)

    • Step 3: Confirm protein stability changes upon FBXO2 manipulation

    • Step 4: Verify glycosylation dependency of the interaction

    • Step 5: Demonstrate reversibility of phenotypes through substrate manipulation

  • Substrate Rescue Experiments:

    • Test whether phenotypes of FBXO2 depletion can be reversed by simultaneous knockdown of putative substrates

    • For example, Hsp47 ablation blocks FBXO2 deletion-induced HCC cell growth in lung

  • Substrate Binding-Deficient Controls:

    • Use FBXO2 mutants that maintain structural integrity but lack substrate binding

    • Compare phenotypes between wild-type and binding-deficient FBXO2 expression

  • Temporal Analysis:

    • Use inducible systems to track the sequence of events after FBXO2 manipulation

    • Early changes are more likely to represent direct effects

    • Late-occurring changes may reflect secondary consequences

Product Science Overview

Introduction

F-Box Protein 2 (FBXO2) is a member of the F-box protein family, which is characterized by the presence of an F-box motif. This motif is approximately 50 amino acids long and is crucial for protein-protein interactions. F-box proteins are integral components of the SCF (SKP1-CUL1-F-box protein) complex, a type of E3 ubiquitin ligase complex that mediates the ubiquitination and subsequent proteasomal degradation of target proteins .

Structure and Function

FBXO2, like other F-box proteins, contains an F-box domain that interacts with SKP1, a core component of the SCF complex. Additionally, FBXO2 has other protein-protein interaction motifs, such as leucine-rich repeats (LRRs), which are involved in substrate recognition . The primary function of FBXO2 is to target specific proteins for ubiquitination, thereby regulating various cellular processes, including cell cycle progression, signal transduction, and transcription .

Biological Significance

FBXO2 plays a significant role in maintaining cellular homeostasis by regulating the degradation of misfolded glycoproteins in the endoplasmic reticulum (ER). This process is crucial for preventing the accumulation of potentially toxic proteins that can lead to cellular stress and disease . In addition, FBXO2 has been implicated in various pathological conditions, including neurodegenerative diseases and cancer .

Preparation Methods

The recombinant production of FBXO2 involves cloning the FBXO2 gene into an appropriate expression vector, followed by transformation into a suitable host cell, such as Escherichia coli or mammalian cells. The expressed protein is then purified using affinity chromatography techniques, such as nickel-nitrilotriacetic acid (Ni-NTA) chromatography, which exploits the histidine tags commonly added to recombinant proteins for purification purposes .

Chemical Reactions and Analysis

FBXO2, as part of the SCF complex, facilitates the transfer of ubiquitin from an E2 ubiquitin-conjugating enzyme to the substrate protein. This ubiquitination process involves a cascade of enzymatic reactions, starting with the activation of ubiquitin by an E1 ubiquitin-activating enzyme, followed by its transfer to an E2 enzyme, and finally to the substrate protein via the E3 ligase activity of the SCF complex . The ubiquitinated proteins are then recognized and degraded by the 26S proteasome, a large protease complex responsible for protein turnover in cells .

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.