FUCA2 Human

Fucosidase Alpha-L- 2 Plasma Human Recombinant
Shipped with Ice Packs
In Stock

Description

FUCA2 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 462 amino acids (29-467 a.a) and having a molecular mass of 53.3kDa.FUCA2 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Alpha-L-fucosidase 2, found in plasma (FUCA2), constitutes 10-20% of total cellular fucosidase activity. This protein, a member of the glycosyl hydrolase 29 family, facilitates the hydrolysis of alpha-1,6-linked fucose attached to the reducing-end N-acetylglucosamine within glycoprotein carbohydrate moieties. Notably, FUCA2 plays a crucial role in the adhesion of Helicobacter pylori to human gastric cancer cells.
Description
Recombinant human FUCA2, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 462 amino acids (residues 29-467). With a molecular weight of 53.3kDa, it includes a 23 amino acid His-tag fused at the N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution.
Formulation
The FUCA2 protein solution is provided at a concentration of 1mg/ml in a buffer containing 20mM phosphate (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept 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. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the product exceeds 85.0%, as determined by SDS-PAGE analysis.
Synonyms
Fucosidase, alpha-L- 2, plasma, dJ20N2.5, RP1-20N2.5, Plasma alpha-L-fucosidase, Alpha-L-fucoside fucohydrolase 2, Alpha-L-fucosidase 2, PSEC0151, UNQ227/PRO260.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHSATRFD PTWESLDARQ LPAWFDQAKF GIFIHWGVFS VPSFGSEWFW WYWQKEKIPK YVEFMKDNYP PSFKYEDFGP LFTAKFFNAN QWADIFQASG AKYIVLTSKH HEGFTLWGSE YSWNWNAIDE GPKRDIVKEL EVAIRNRTDL RFGLYYSLFE WFHPLFLEDE SSSFHKRQFP VSKTLPELYE LVNNYQPEVL WSDGDGGAPD QYWNSTGFLA WLYNESPVRG TVVTNDRWGA GSICKHGGFY TCSDRYNPGH LLPHKWENCM TIDKLSWGYR REAGISDYLT IEELVKQLVE TVSCGGNLLM NIGPTLDGTI SVVFEERLRQ MGSWLKVNGE AIYETHTWRS QNDTVTPDVW YTSKPKEKLV YAIFLKWPTS GQLFLGHPKA ILGATEVKLL GHGQPLNWIS LEQNGIMVEL PQLTIHQMPC KWGWALALTN VI.

Q&A

What is FUCA2 and what is its basic cellular function?

FUCA2 (alpha-L-fucosidase 2) is a protein-coding gene in humans that encodes a plasma alpha-L-fucosidase enzyme. This enzyme represents approximately 10-20% of the total cellular fucosidase activity. As a member of the glycosyl hydrolase 29 family, FUCA2 primarily catalyzes the hydrolysis of alpha-1,6-linked fucose joined to the reducing-end N-acetylglucosamine of carbohydrate moieties in glycoproteins. This enzymatic activity plays an essential role in glycoprotein processing and modification within cellular systems. Notably, research has identified FUCA2 as essential for Helicobacter pylori adhesion to human gastric cancer cells, suggesting its involvement in host-pathogen interactions .

How is FUCA2 structurally different from other fucosidases?

FUCA2 differs from other fucosidases, particularly FUCA1, in its structural properties and cellular distribution. While both enzymes belong to the glycosyl hydrolase family 29, FUCA2 is primarily a plasma enzyme with specific activity toward alpha-1,6-linked fucose residues. Structurally, FUCA2's active site contains specific amino acid residues that determine its substrate specificity, distinguishing it from other fucosidases. The enzyme's three-dimensional conformation enables selective binding to specific glycoprotein structures, which explains its specialized role in glycoprotein processing. Researchers investigating FUCA2's structure should employ X-ray crystallography or cryo-electron microscopy to determine its precise molecular architecture and compare it with other fucosidases to understand functional differences .

What are the known genetic variants of FUCA2 and their functional implications?

Several genetic variants of FUCA2 have been identified through genome-wide association studies and targeted sequencing approaches. These polymorphisms can affect enzyme activity, substrate specificity, and protein stability. When studying FUCA2 variants, researchers should consider employing whole-exome sequencing to identify rare variants and single nucleotide polymorphisms (SNPs) that might influence enzyme function. Functional assays measuring enzymatic activity of different variants can provide insights into how these genetic differences translate to phenotypic variations. Some variants have been associated with altered cancer susceptibility and progression, particularly in hepatocellular carcinoma, suggesting that genetic screening of FUCA2 variants might have prognostic value in cancer research .

How can researchers investigate FUCA2's role in immune infiltration in cancer microenvironments?

To investigate FUCA2's role in immune infiltration within cancer microenvironments, researchers should employ a multi-faceted approach combining computational and experimental methods. Computationally, tools such as CIBERSORT, TIMER 2.0, and TISIDB can be used to analyze the correlation between FUCA2 expression and various immune cell infiltrates (B cells, T cells, macrophages, dendritic cells, and NK cells) from transcriptomic data. The Spearman correlation coefficient is particularly useful for quantifying these relationships. Experimentally, single-cell RNA sequencing of tumor samples can provide high-resolution mapping of FUCA2 expression across different cell types within the tumor microenvironment. Immunohistochemistry and flow cytometry with appropriate markers can validate computational findings by visualizing the spatial relationship between FUCA2-expressing cells and immune infiltrates. Additionally, co-culture experiments with FUCA2-expressing tumor cells and immune cells can elucidate functional interactions that influence immune response .

In which cancer types is FUCA2 overexpressed, and what are the methodological approaches to study its pan-cancer expression patterns?

FUCA2 overexpression has been documented across multiple cancer types. Analysis using platforms such as GEPIA2 and TIMER 2.0 has revealed significant upregulation in breast, esophageal, lung, gastric, liver, colon, and pancreatic cancers. Specifically, paired analysis of tumor versus adjacent normal tissues has confirmed FUCA2 upregulation in UCEC (uterine corpus endometrial carcinoma), STAD (stomach adenocarcinoma), PRAD (prostate adenocarcinoma), LUAD (lung adenocarcinoma), LIHC (liver hepatocellular carcinoma), KIRP (kidney renal papillary cell carcinoma), HNSC (head and neck squamous cell carcinoma), ESCA (esophageal carcinoma), KIRC (kidney renal clear cell carcinoma), CHOL (cholangiocarcinoma), CESC (cervical squamous cell carcinoma), BLCA (bladder urothelial carcinoma), and BRCA (breast invasive carcinoma). Interestingly, FUCA2 is downregulated in KICH (kidney chromophobe). To study pan-cancer expression patterns, researchers should employ a systematic approach using multiple databases (TCGA, GTEx, CPTAC) and analysis platforms. Differential expression analysis should be performed using tools like DESeq2 or limma, with appropriate normalization methods to account for batch effects across different datasets .

What signaling pathways are associated with FUCA2 expression and function in cancer?

Gene Set Enrichment Analysis (GSEA) has identified several key signaling pathways associated with FUCA2 expression in cancer. These predominantly involve immune regulation mechanisms, including adaptive and innate immune systems, neutrophil degranulation, and cytokine signaling. To investigate these pathways, researchers should employ GSEA using the R package "clusterprofiler" with appropriate gene set databases such as MSigDB Hallmark gene sets. The analysis should focus on pathways showing positive Normalized Enrichment Scores (NES) with adjusted p-values < 0.05. For validation, researchers can perform pathway perturbation experiments using inhibitors of key pathway components followed by assessment of FUCA2 expression and function. Additionally, Gene Set Variation Analysis (GSVA) can provide sample-specific pathway scores to understand heterogeneity in pathway activation across different tumors. When interpreting results, researchers should consider that pathways with consistent enrichment across multiple cancer types are likely to represent core mechanisms of FUCA2 function .

What is the relationship between FUCA2 and Helicobacter pylori adhesion, and how can this be experimentally investigated?

FUCA2 has been identified as essential for Helicobacter pylori adhesion to human gastric cancer cells, suggesting a critical role in host-pathogen interactions that may influence gastric cancer development. This relationship can be experimentally investigated through several complementary approaches. Bacterial adhesion assays using fluorescently labeled H. pylori strains on gastric cancer cell lines with manipulated FUCA2 expression (through siRNA knockdown, CRISPR knockout, or overexpression) can quantify the direct impact of FUCA2 on bacterial binding. Co-immunoprecipitation experiments can identify specific H. pylori adhesins that interact with FUCA2 or FUCA2-modified glycoproteins. Glycan microarrays containing various fucosylated structures can determine the specific glycan epitopes involved in this interaction. For in vivo relevance, researchers can analyze clinical samples for correlations between FUCA2 expression, H. pylori infection status, and gastric cancer progression. Additionally, transgenic mouse models with altered FUCA2 expression can be used to study the impact on H. pylori colonization and subsequent gastric pathology .

What are the optimal methods for measuring FUCA2 enzymatic activity in clinical samples?

The optimal methods for measuring FUCA2 enzymatic activity in clinical samples involve both spectrofluorometric and chromatographic techniques. The most sensitive approach utilizes 4-methylumbelliferyl-α-L-fucopyranoside as a fluorogenic substrate, which releases the fluorescent 4-methylumbelliferone upon hydrolysis by FUCA2. This assay should be performed at pH 5.5-6.0 to distinguish FUCA2 activity from FUCA1, which shows optimal activity at more acidic pH (around 4.5-5.0). For more specific analysis, researchers should perform immunoprecipitation with FUCA2-specific antibodies before activity assays to separate it from other fucosidases. When working with plasma samples, it's critical to add appropriate protease inhibitors immediately after collection to prevent enzyme degradation. For tissue samples, careful homogenization in non-denaturing buffers is essential. To ensure accuracy, standard curves using purified recombinant FUCA2 should be included, and activity should be normalized to total protein concentration. Additionally, high-performance liquid chromatography (HPLC) or ultra-performance liquid chromatography (UPLC) coupled with mass spectrometry can be employed to analyze the natural substrate processing by quantifying the released fucose or defucosylated glycoprotein products .

What multi-omics approaches can be used to comprehensively study FUCA2's role in cancer?

To comprehensively study FUCA2's role in cancer, researchers should implement integrated multi-omics approaches that combine genomic, transcriptomic, proteomic, glycomic, and functional data. For genomic analysis, whole-genome or targeted sequencing can identify FUCA2 mutations, copy number variations, and regulatory polymorphisms. RNA sequencing provides transcriptome-wide context for FUCA2 expression patterns and co-expressed genes that may function in the same pathways. Proteomics using liquid chromatography-mass spectrometry (LC-MS/MS) can quantify FUCA2 protein levels and post-translational modifications. Glycomics approaches, including mass spectrometry-based glycan profiling and lectin arrays, can identify changes in the fucosylation landscape due to FUCA2 activity. Integration of these multi-omics data can be achieved using computational tools like DIABLO and NOLAS, which employ different strategies for data integration. DIABLO uses a sparse generalized canonical correlation analysis (sGCCA) to identify correlated variables across multiple omics datasets, while NOLAS employs Singular Value Decomposition (SVD) and follows a middle integration strategy to extract latent variables that capture distinct biological insights. When designing multi-omics studies, researchers should carefully preprocess data to ensure compatibility between different platforms, employ appropriate normalization methods, and validate key findings using targeted experimental approaches .

What are the advantages and limitations of using DIABLO versus NOLAS for FUCA2 multi-omics data integration?

For FUCA2 research specifically, the choice between these methods should depend on research objectives. If the goal is biomarker discovery with strong predictive modeling, DIABLO may be preferable. If the focus is on identifying core biological mechanisms with stringent statistical significance, NOLAS might be more appropriate. Researchers should consider performing appropriate preprocessing, including filtering datasets, normalization, and handling missing values, before applying either method. Validation using independent cohorts is essential regardless of the chosen integration method .

What are the best experimental models for studying FUCA2 function in cancer progression?

The optimal experimental models for studying FUCA2 function in cancer progression include both in vitro and in vivo systems, each with specific advantages for addressing different research questions. For in vitro studies, researchers should consider: (1) Cell line models: Cancer cell lines with varying FUCA2 expression levels provide a foundation for mechanistic studies. CRISPR-Cas9-mediated knockout, siRNA knockdown, or overexpression systems in these lines enable functional characterization. Multiple cell lines should be used to account for genetic heterogeneity. (2) Patient-derived organoids: These three-dimensional cultures better recapitulate tumor architecture and heterogeneity than monolayer cultures, offering a more physiologically relevant system for studying FUCA2's impact on tumor growth and drug response. (3) Co-culture systems: Combining cancer cells with immune cells or stromal components allows investigation of FUCA2's role in tumor microenvironment interactions.

For in vivo models, researchers should consider: (1) Patient-derived xenografts (PDXs): These maintain the heterogeneity and many characteristics of the original tumor, making them valuable for studying FUCA2's role in tumor growth and metastasis. (2) Genetically engineered mouse models (GEMMs): Conditional FUCA2 knockout or overexpression in specific tissues allows study of FUCA2's role in cancer initiation and progression in an immunocompetent setting. (3) Orthotopic models: Implanting cancer cells directly into the organ of origin provides a more relevant microenvironment than subcutaneous models.

When designing experiments, researchers should include appropriate controls, such as isogenic cell lines differing only in FUCA2 status, and consider time-course analyses to capture dynamic effects of FUCA2 on cancer progression .

How should researchers approach FUCA2 gene expression manipulation for functional studies?

When manipulating FUCA2 gene expression for functional studies, researchers should employ a systematic approach considering the specific research question, cell type, and desired duration of effect. For transient knockdown, siRNA or shRNA approaches are effective, with optimal transfection conditions determined empirically for each cell line. At least three independent siRNA sequences targeting different regions of FUCA2 mRNA should be tested to confirm specificity of observed phenotypes. For stable knockdown or knockout, CRISPR-Cas9 systems with appropriate guide RNA design (minimizing off-target effects) are recommended, with verification of editing efficiency by sequencing and protein loss by Western blot.

For overexpression studies, researchers should consider using inducible expression systems (such as Tet-On) to control FUCA2 expression levels and timing, particularly important since non-physiological overexpression may cause artifacts. Expression vectors should contain the full-length human FUCA2 cDNA with appropriate epitope tags for detection, while maintaining enzymatic activity. Both wild-type FUCA2 and catalytically inactive mutants (created by site-directed mutagenesis of active site residues) should be included to distinguish between enzymatic and potential scaffolding functions of FUCA2.

For all gene manipulation approaches, comprehensive validation is essential: (1) qRT-PCR to confirm changes at mRNA level, (2) Western blotting to verify protein level changes, (3) enzymatic activity assays to confirm functional consequences, and (4) rescue experiments where appropriate to confirm specificity. Researchers should also consider potential compensatory mechanisms, particularly upregulation of other fucosidases like FUCA1 in response to FUCA2 manipulation .

What considerations are important when designing clinical studies to validate FUCA2 as a biomarker?

When designing clinical studies to validate FUCA2 as a biomarker, researchers must address several critical considerations to ensure robust and clinically relevant results. Sample size calculation should be performed based on preliminary data and expected effect sizes, with power analysis ensuring sufficient statistical strength. Patient cohort selection requires careful stratification based on disease stage, treatment history, and relevant clinical parameters, with matched case-control design when appropriate. Multi-center validation is essential to account for institutional variations in sample collection and patient populations.

For specimen collection and processing, standardized protocols must be established for sample timing (relation to treatment, disease progression), collection methods, processing, and storage conditions to ensure reproducibility and minimize pre-analytical variables. Both tissue (primary tumor, metastases, adjacent normal) and liquid biopsies (blood, serum, plasma) should be considered, with matched samples where possible.

Analytical validation requires determining assay specificity (distinguishing FUCA2 from other fucosidases), sensitivity (lower limits of detection), precision (intra- and inter-assay variability), and reproducibility across different laboratories. Both FUCA2 protein levels (ELISA, immunohistochemistry) and enzymatic activity should be measured when possible.

Clinical validation should assess FUCA2's performance as a diagnostic, prognostic, or predictive biomarker using appropriate statistical methods. For diagnostic applications, ROC curve analysis with calculation of sensitivity, specificity, positive and negative predictive values is essential. For prognostic value, Kaplan-Meier survival analysis with multivariable Cox regression accounting for established prognostic factors should be performed. Integration with existing clinical biomarkers and comparison to current gold standards is crucial for establishing FUCA2's added clinical value .

How can researchers resolve contradictory findings about FUCA2's role across different cancer types?

Resolving contradictory findings about FUCA2's role across different cancer types requires a systematic multi-faceted approach. First, researchers should conduct a comprehensive meta-analysis of existing studies, stratifying by cancer type, methodology, sample size, and population demographics to identify patterns in contradictions. Critical evaluation of methodological differences is essential—variations in FUCA2 detection methods (antibodies, activity assays), reference genes for normalization, or statistical approaches can lead to apparently contradictory results.

Biological context differences should be considered, as FUCA2 may have tissue-specific functions due to differential glycosylation patterns and substrate availability across tissues. The cancer stage-specific analysis is critical, as FUCA2's role may evolve during cancer progression—early protective effects might shift to promoting metastasis in advanced stages. Genetic background variations, including polymorphisms in FUCA2 or interacting genes, might explain differential effects across populations.

To experimentally address contradictions, researchers should design cross-cancer studies using consistent methodology across multiple cancer types, ideally performed in the same laboratory. Isogenic cell line panels representing multiple cancer types manipulated for FUCA2 expression allow direct comparison of phenotypic effects. Single-cell approaches can identify cell type-specific effects that might be masked in bulk tissue analysis. Finally, detailed pathway analysis comparing FUCA2-associated signaling across cancer types can reveal context-dependent molecular mechanisms. When publishing findings, researchers should explicitly discuss contradictions with existing literature and provide potential explanations based on methodological and biological differences .

What statistical approaches are most appropriate for analyzing FUCA2 expression in correlation with patient outcomes?

Cox proportional hazards regression should be employed for both univariate and multivariate analyses, incorporating established prognostic factors to determine if FUCA2 provides independent prognostic information. The proportional hazards assumption should be verified using Schoenfeld residuals. For time-varying effects of FUCA2, extended Cox models with time-dependent covariates may be necessary.

For categorical outcomes (treatment response, disease recurrence), logistic regression models are appropriate, with goodness-of-fit assessed using Hosmer-Lemeshow tests and model performance evaluated using ROC curve analysis. When analyzing FUCA2 as a continuous variable, restricted cubic splines can capture non-linear relationships with outcomes.

For all analyses, appropriate correction for multiple testing is essential when examining FUCA2 alongside other potential biomarkers. Sample size considerations are critical—power calculations should ensure sufficient events (particularly for survival analyses) to detect clinically meaningful differences. Validation in independent cohorts is necessary to confirm findings, and sensitivity analyses should be performed to assess the robustness of results to different analytical choices .

What are the key considerations when interpreting FUCA2 expression data across different omics platforms?

The relationship between transcript and protein levels requires careful consideration, as post-transcriptional regulation may result in discordance between mRNA and protein expression. Correlation analysis between FUCA2 mRNA and protein levels can identify potential regulatory mechanisms. Additionally, researchers should distinguish between FUCA2 expression and enzymatic activity, as post-translational modifications may affect function without changing expression levels.

Tissue heterogeneity introduces complexity in bulk tissue analyses—variations in cellular composition can confound FUCA2 expression measurements. Computational deconvolution methods or single-cell approaches can help address this issue. When interpreting FUCA2 expression in the context of gene signatures or pathway activities, researchers should consider both statistical significance and biological relevance, with pathway enrichment analyses providing context for FUCA2's role.

For clinical applications, researchers must establish threshold values for "high" versus "low" FUCA2 expression that are clinically meaningful and reproducible across platforms. The development of standardized assays with established reference ranges is essential for translational applications .

Table 1: FUCA2 Expression Across Cancer Types Compared to Normal Tissues

Cancer TypeAbbreviationFUCA2 Expression StatusStatistical SignificanceReference Database
Uterine Corpus Endometrial CarcinomaUCECUpregulatedP<0.001TCGA
Stomach AdenocarcinomaSTADUpregulatedP<0.001TCGA
Prostate AdenocarcinomaPRADUpregulatedP<0.001TCGA
Lung AdenocarcinomaLUADUpregulatedP<0.001TCGA/CPTAC
Liver Hepatocellular CarcinomaLIHCUpregulatedP=1.62×10^-12TCGA
Kidney Renal Papillary Cell CarcinomaKIRPUpregulatedP<0.001TCGA
Head and Neck Squamous Cell CarcinomaHNSCUpregulatedP<0.001TCGA
Esophageal CarcinomaESCAUpregulatedP<0.001TCGA
Kidney Renal Clear Cell CarcinomaKIRCUpregulatedP<0.001TCGA
CholangiocarcinomaCHOLUpregulatedP<0.001TCGA
Cervical Squamous Cell CarcinomaCESCUpregulatedP<0.001TCGA
Bladder Urothelial CarcinomaBLCAUpregulatedP<0.001TCGA
Breast Invasive CarcinomaBRCAUpregulatedP<0.001TCGA
Kidney ChromophobeKICHDownregulatedP<0.05TCGA

Table 2: Prognostic Value of FUCA2 in Hepatocellular Carcinoma

ParameterValue95% Confidence IntervalP-valueAnalysis Method
Hazard Ratio (FUCA2 high vs. low)1.741.31-2.30<0.001Univariate Cox regression
M classification HR1.251.00-1.560.0478Univariate Cox regression
Residual tumor HR1.301.01-1.680.0453Univariate Cox regression
Median OS (FUCA2 low expression)Significantly longer-<0.001Kaplan-Meier analysis
Median OS (FUCA2 high expression)Significantly shorter-<0.001Kaplan-Meier analysis
FUCA2 expression cutoff value4.791648 (50th percentile)--TCGA database analysis

What are the most promising therapeutic strategies targeting FUCA2 in cancer?

The most promising therapeutic strategies targeting FUCA2 in cancer encompass several complementary approaches. Small molecule inhibitors of FUCA2 enzymatic activity represent a direct approach—structure-based drug design utilizing crystallographic data of FUCA2's active site can identify selective inhibitors that spare other fucosidases. High-throughput screening of chemical libraries against recombinant FUCA2, followed by medicinal chemistry optimization, can identify lead compounds with appropriate pharmacokinetic and safety profiles. For cancers where FUCA2 is overexpressed, antibody-based approaches offer potential—humanized monoclonal antibodies against FUCA2 could be developed for targeted delivery of cytotoxic agents (antibody-drug conjugates) or for immune recruitment (bispecific antibodies).

RNA interference therapeutics using siRNA or shRNA encapsulated in nanoparticles with tumor-targeting capabilities could achieve selective FUCA2 knockdown in tumors. The emerging CRISPR-based therapeutics might eventually allow for gene editing approaches targeting FUCA2 in cancer cells. Given FUCA2's role in immune regulation pathways, combination strategies with immunotherapies such as immune checkpoint inhibitors merit investigation, particularly in cancers where FUCA2 expression correlates with immune infiltration patterns.

For clinical development, researchers should prioritize cancer types showing strong correlation between FUCA2 expression and poor outcomes, establish appropriate biomarkers for patient selection, and design early-phase clinical trials with pharmacodynamic endpoints to confirm target engagement before proceeding to efficacy studies .

How might advances in glycomics technologies enhance our understanding of FUCA2 biology?

Advances in glycomics technologies are poised to substantially enhance our understanding of FUCA2 biology through several innovative approaches. High-resolution mass spectrometry with improved sensitivity now enables comprehensive mapping of fucosylated glycans in complex biological samples, allowing researchers to identify specific FUCA2 substrates with unprecedented detail. When combined with stable isotope labeling, these techniques can track dynamic changes in fucosylation patterns in response to FUCA2 modulation, providing insights into the enzyme's in vivo activity.

Glycan imaging technologies, including metabolic glycan labeling and glycan-specific probes, now allow visualization of fucosylated glycans in living cells and tissues, enabling spatial and temporal analysis of FUCA2 activity in physiological contexts. Single-cell glycomics approaches can reveal cell-type specific effects of FUCA2 that may be masked in bulk analyses, particularly important in heterogeneous tumor microenvironments.

Glycoproteomics technologies combining glycan analysis with proteomics can identify specific proteins affected by FUCA2 activity, linking glycan changes to functional proteome alterations. Advanced glycan arrays containing structurally defined fucosylated glycans enable high-throughput analysis of binding preferences and substrate specificities for FUCA2.

Computational glycobiology, including machine learning approaches trained on glycomics datasets, can predict functional consequences of altered fucosylation patterns due to FUCA2 dysregulation. These computational models, when integrated with multi-omics data, can generate testable hypotheses about FUCA2's role in complex biological processes.

For researchers entering this field, collaboration between glycobiologists and cancer biologists will be essential to fully leverage these technologies and translate glycomics findings into mechanistic insights and potential therapeutic strategies .

What are the key knowledge gaps in understanding FUCA2's role in non-cancerous pathological conditions?

Despite significant research on FUCA2 in cancer, major knowledge gaps exist regarding its role in non-cancerous pathological conditions. In infectious diseases, while FUCA2's importance in Helicobacter pylori adhesion is established, its potential involvement in other bacterial, viral, or parasitic infections remains largely unexplored. Researchers should investigate how FUCA2-mediated modification of host glycans affects pathogen recognition and immune response across different infectious agents.

In inflammatory disorders, the role of FUCA2 in regulating immune cell function and inflammatory signaling requires systematic investigation. Given GSEA results linking FUCA2 to immune regulation pathways, its potential contribution to autoimmune diseases, chronic inflammation, and inflammatory bowel diseases merits exploration. Flow cytometry and single-cell analysis of immune populations with manipulated FUCA2 expression could reveal its impact on immune cell function and cytokine production.

In metabolic disorders, limited information exists about FUCA2's potential involvement in diabetes, obesity, or metabolic syndrome. Researchers should investigate whether FUCA2-mediated glycan modifications affect insulin signaling, adipocyte function, or metabolic regulation. Animal models with tissue-specific FUCA2 modulation in metabolic tissues could provide valuable insights.

In neurodegenerative diseases, where aberrant glycosylation has been implicated, FUCA2's potential contribution remains unexamined. Researchers should investigate FUCA2 expression and function in neuronal tissues and its potential role in protein aggregation characteristic of conditions like Alzheimer's and Parkinson's diseases.

Product Science Overview

Structure and Production

The recombinant form of this enzyme is produced using Chinese Hamster Ovary (CHO) cells. The human plasma alpha-L-fucosidase/FUCA2 protein is expressed with an N-terminal 6-His tag, which facilitates its purification . The recombinant protein has a predicted molecular mass of approximately 52 kDa, and it is typically supplied as a 0.2 μm filtered solution in Tris and NaCl .

Function and Activity

Fucosidase Alpha-L-2 Plasma is essential for the hydrolysis of fucose residues from glycoproteins and glycolipids. This activity is critical for various biological processes, including cell-cell interaction, signal transduction, and immune response . The enzyme’s activity is measured by its ability to cleave a fluorogenic substrate, 4-methylumbelliferyl-alpha-L-fucopyranoside, with a specific activity of over 550 pmol/min/μg .

Clinical Significance

The enzyme represents 10-20% of the total cellular fucosidase activity in human plasma . It is particularly important for the adhesion of Helicobacter pylori to human gastric cancer cells, highlighting its role in pathogenic interactions . Additionally, mutations in the FUCA2 gene have been associated with developmental and epileptic encephalopathies .

Storage and Stability

The recombinant enzyme is stable for six months from the date of receipt when stored at -20 to -70°C. It is important to avoid repeated freeze-thaw cycles to maintain its activity .

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.