Recombinant Human Netrin receptor UNC5A (UNC5A), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.

Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.

Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our default glycerol concentration is 50%, provided as a reference for customers.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.

Tag type determination occurs during production. If a specific tag type is required, please inform us for preferential development.

Synonyms
FLJ16449; KIAA1976; Netrin receptor UNC 5A; Netrin receptor UNC5A; Netrin receptor Unc5h1; Protein unc-5 homolog 1; Protein unc-5 homolog A; Unc 5 homolog 1; Unc 5 homolog A; Unc 5 netrin receptor A; Unc 5a; Unc5 a; Unc5 homolog 1; Unc5 homolog A; Unc5a; UNC5A_HUMAN; Unc5h1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
UNC5A
Uniprot No.

Target Background

Function

Netrin receptor essential for axon guidance. It functions within the netrin signaling pathway, promoting neurite outgrowth in response to netrin-1 (NTN1). In developing nervous systems, it mediates axon repulsion in growth cones responding to netrin. This repulsion may involve DCC association, triggering repulsion signaling. Furthermore, it acts as a dependence receptor, inducing apoptosis in the absence of netrin ligand.

Gene References Into Functions
  1. Data suggest a cooperative relationship between the unfolded protein response (UPR) and UNC5A depletion, potentially contributing to liver cancer development and growth. PMID: 29277614
  2. UNC5A inhibits autophagy and promotes HCV restriction by impacting virion infectivity, independent of cell death and involving DAPK. PMID: 28783179
  3. FANCC interferes with UNC5A's apoptotic functions, suggesting a potential role in developmental processes through UNC5A association. PMID: 24676280
  4. UNC5A, a potential tumor suppressor, is implicated in predicting response to chemotherapy-induced DNA damage and regulating cell death in bladder cancer. PMID: 24737586
  5. UNC5A is a p53 transcriptional target involved in p53-dependent apoptosis. PMID: 20372800
  6. UNC5A may function as a tumor suppressor, inhibiting tumor spread beyond netrin-1-rich regions by inducing apoptosis. PMID: 12655055
  7. UNC5 orthologs regulate apoptosis in cancer cells through a novel mechanism, encompassing both induction and suppression. (Review) PMID: 15573119
Database Links

HGNC: 12567

OMIM: 607869

KEGG: hsa:90249

STRING: 9606.ENSP00000332737

UniGene: Hs.33191

Protein Families
Unc-5 family
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Membrane raft. Cell projection, neuron projection.

Q&A

What is UNC5A and what is its primary function?

UNC5A (Unc-5 netrin receptor A) is a type-I transmembrane receptor located on the 5q35 chromosomal region that functions primarily as a receptor for the netrin family of proteins, including netrin-1 and netrin-3. UNC5A plays key roles in neuronal development and differentiation, particularly in axon guidance . The most notable functional feature of UNC5A is its classification as a dependence receptor, meaning its function depends on the availability of its ligands. In the absence of netrin-1, UNC5A exists in a monomeric state where the intracellular region is cleaved by caspases, releasing the C-terminal portion that initiates apoptotic pathways. Conversely, when netrin-1 is present, UNC5A forms dimers that transmit signals promoting cell survival, migration, and differentiation . This dual functionality makes UNC5A particularly interesting in both developmental biology and cancer research contexts.

How does UNC5A relate to other members of the UNC5 receptor family?

The UNC5 gene family consists of four related genes: UNC5A, UNC5B, UNC5C, and UNC5D. All encode type-I transmembrane receptors for netrin-1 and share fundamental structural and functional similarities . All UNC5 receptors function as dependence receptors, transducing positive cell proliferation and survival signals when bound to netrin-1 but inducing caspase-dependent apoptosis in the absence of their ligand . Despite these similarities, each UNC5 family member exhibits distinct expression patterns and may be regulated through different mechanisms. For example, while UNC5A, UNC5B, and UNC5C are all frequently downregulated in colorectal cancer, they may be silenced through different epigenetic mechanisms . Additionally, UNC5A, UNC5B, and UNC5D are transcriptionally regulated by the tumor suppressor p53 and are involved in p53-dependent apoptosis in response to DNA damage induced by conventional chemotherapeutic drugs like doxorubicin .

What are the recommended methodologies for studying UNC5A expression in tumor tissues?

When investigating UNC5A expression in tumor tissues, researchers should employ multiple complementary techniques to ensure robust results. For mRNA expression analysis, RT-qPCR remains the gold standard, as demonstrated in studies examining UNC5A expression in colorectal cancer . For protein-level analysis, immunohistochemistry (IHC) provides valuable spatial information about UNC5A expression patterns within tissue samples. When conducting IHC, proper antibody validation is crucial, including positive and negative controls .
For more comprehensive analysis, RNA sequencing allows examination of UNC5A expression across multiple cancer types simultaneously. This approach was used in pan-cancer analyses that revealed differential UNC5A expression patterns across various tumor types . Additionally, since epigenetic mechanisms play a significant role in UNC5A regulation, bisulfite sequencing or methylation-specific PCR should be considered to assess the methylation status of the UNC5A promoter. When analyzing results, integrating UNC5A expression data with clinical parameters, such as tumor stage, grade, and patient outcomes, provides more meaningful insights into its biological significance .

What experimental models are most appropriate for investigating UNC5A function?

For in vitro studies, human cancer cell lines with varying levels of endogenous UNC5A expression offer valuable experimental models. HCT116 colorectal cancer cells have been successfully used to study UNC5A regulation by O-GlcNAcylation and the PRC2 complex . For gain-of-function studies, researchers can use expression vectors containing UNC5A cDNA, while loss-of-function studies can be conducted using siRNA or CRISPR-Cas9 gene editing technology to knockdown or knockout UNC5A expression, respectively .
For in vivo investigations, genetically engineered mouse models provide physiologically relevant systems. AOM/DSS-treated mice represent an established model for studying colorectal carcinogenesis and have been used to investigate UNC5A expression in relation to dietary factors . When designing these models, researchers should consider the potential impact of different diets, as high carbohydrate diets have been shown to influence UNC5A expression in colon tissues . Additionally, patient-derived xenograft (PDX) models, where patient tumor samples are implanted into immunodeficient mice, can recapitulate the heterogeneity of human tumors and provide insights into UNC5A function in a physiologically relevant microenvironment.

What is the prognostic significance of UNC5A expression in different cancer types?

The prognostic significance of UNC5A expression varies substantially across different cancer types, making it crucial for researchers to consider cancer-specific contexts. In head and neck squamous cell carcinoma (HNSC) and prostate adenocarcinoma (PRAD), patients with UNC5A mutations demonstrate poorer progression-free survival (PFS) . Cox regression analysis has identified high UNC5A expression as a risk factor for disease-free interval (DFI) in lung squamous cell carcinoma (LUSC) and testicular germ cell tumors (TGCT), but as a protective factor for pheochromocytoma and paraganglioma (PCPG) and lower-grade glioma (LGG) .
For disease-specific survival (DSS), high UNC5A levels correlate with worse outcomes in LUSC, kidney renal clear cell carcinoma (KIRC), and uveal melanoma (UVM), but better outcomes in LGG . Similarly, for progression-free interval (PFI), elevated UNC5A expression represents a risk factor for KIRC, adrenocortical carcinoma (ACC), and UVM, but a protective factor for LGG and PCPG . These divergent associations highlight the context-dependent role of UNC5A in cancer progression and emphasize the importance of cancer-specific approaches when considering UNC5A as a prognostic biomarker. Researchers should integrate UNC5A expression data with other clinical parameters and molecular features for more accurate prognostic assessment.

How does UNC5A expression correlate with tumor immunity parameters?

UNC5A expression demonstrates significant correlations with multiple tumor immunity parameters, positioning it as a potential immunotherapy target. Research has established associations between UNC5A expression and tumor mutation burden (TMB), neoantigen load, tumor microenvironment (TME) composition, and microsatellite instability (MSI) . These correlations vary across cancer types, indicating cancer-specific immunological implications.
UNC5A expression also correlates with various immunomodulators, including chemokines, receptors, major histocompatibility complex (MHC) components, immunoinhibitors, and immunostimulators . These relationships suggest UNC5A may influence immune cell recruitment and activation within the tumor microenvironment. Furthermore, connections between UNC5A expression and immune checkpoint genes have been observed, potentially affecting immunotherapy response . Methodologically, researchers investigating these relationships should use comprehensive approaches like Spearman correlation analysis for evaluating associations between UNC5A expression and immune parameters. The strength of correlations should be classified as weak (r < 0.3), moderate (0.3 ≤ r < 0.6), or strong (r ≥ 0.6) to properly interpret biological significance .

What epigenetic mechanisms regulate UNC5A expression in cancer cells?

UNC5A expression is regulated by complex epigenetic mechanisms, particularly in cancer contexts. DNA methylation plays a crucial role, with hypermethylation of the UNC5A promoter leading to transcriptional silencing in various cancers . The degree of UNC5A promoter methylation correlates with its expression across multiple cancer types, as revealed by Spearman correlation analysis . DNA methyltransferases, including DNMT1, DNMT2, DNMT3a, and DNMT3b, are implicated in this process, though their relative contributions may vary across cancer types .
Histone modifications, particularly those mediated by the Polycomb Repressive Complex 2 (PRC2), represent another key epigenetic regulatory mechanism for UNC5A. The PRC2 complex, which includes EZH2 as its catalytic subunit, deposits repressive H3K27me3 marks on the UNC5A promoter, leading to transcriptional silencing . Experimental approaches to study these mechanisms include chromatin immunoprecipitation (ChIP) assays to detect PRC2 component binding to the UNC5A promoter, luciferase reporter assays to assess promoter activity under various epigenetic conditions, and treatments with epigenetic inhibitors like DNA methyltransferase inhibitors or EZH2 inhibitors to evaluate their effects on UNC5A re-expression . Understanding these epigenetic mechanisms provides potential therapeutic opportunities through epigenetic modifiers that could restore UNC5A expression and its tumor-suppressive functions.

How does O-GlcNAcylation influence UNC5A expression and function?

O-GlcNAcylation, a dynamic post-translational modification influenced by nutritional status, plays a significant role in regulating UNC5A expression. This modification involves the addition of N-acetylglucosamine to serine or threonine residues of proteins by O-GlcNAc transferase (OGT) . Research has demonstrated that O-GlcNAcylation levels increase in response to high carbohydrate diets and in colorectal tumors compared to normal tissues . Importantly, inverse correlations have been observed between O-GlcNAcylation levels and UNC5A expression, suggesting a regulatory relationship .
Mechanistically, O-GlcNAcylation influences UNC5A expression by regulating the PRC2 complex. O-GlcNAcylation is required for PRC2-mediated repression of UNC5A, as demonstrated by experiments where OGT knockdown or inhibition prevented PRC2-induced UNC5A downregulation . When studying this phenomenon, researchers should employ multiple complementary approaches. These include OGT knockdown using siRNA or inhibition using compounds like Ac5S-GlcNAc, followed by assessment of UNC5A expression at both mRNA and protein levels . Additionally, chromatin immunoprecipitation (ChIP) assays can determine how O-GlcNAcylation affects PRC2 recruitment to the UNC5A promoter. Integrating these approaches provides comprehensive insights into how nutritional factors influence UNC5A expression through O-GlcNAcylation-dependent epigenetic mechanisms.

How does the interaction between UNC5A and Netrin-1 influence downstream signaling pathways?

The interaction between UNC5A and its ligand Netrin-1 significantly impacts multiple downstream signaling pathways, with context-dependent outcomes. In glioma cells, Netrin-1 binding to UNC5A activates the NF-κB signaling pathway, particularly through promoting p65 ser536 phosphorylation, which subsequently upregulates c-Myc expression and enhances cell proliferation . This pro-tumorigenic effect is UNC5A-dependent, as suppression of UNC5A significantly inhibits NF-κB activation and reduces cell proliferation .
Beyond NF-κB signaling, the UNC5A/Netrin-1 interaction influences other pathways relevant to cancer biology. When investigating these signaling mechanisms, researchers should employ a comprehensive approach including phosphorylation analysis of key signaling molecules, gene expression profiling to identify downstream targets, and functional assays to assess biological outcomes. Experiments should include both gain-of-function approaches (e.g., Netrin-1 treatment) and loss-of-function approaches (e.g., UNC5A knockdown or Netrin-1/UNC5A interaction inhibitors) . Additionally, since UNC5A can function as a dependence receptor, researchers should compare signaling events in Netrin-1-rich versus Netrin-1-depleted conditions to fully understand the dual nature of UNC5A-mediated signaling.

How might UNC5A serve as a therapeutic target in cancer treatment?

UNC5A presents multiple avenues for therapeutic intervention in cancer treatment, stemming from its dual role as both a dependence receptor and signaling molecule. The most direct approach leverages UNC5A's pro-apoptotic function in the absence of Netrin-1. Since many cancers upregulate Netrin-1 to prevent UNC5A-mediated apoptosis, disrupting this interaction could restore UNC5A's tumor-suppressive properties . Netrin-1/UNC5A interaction inhibitors (such as TRAP-netrin UNC5A) have shown promise in potentiating doxorubicin-induced cell death, suggesting potential for combination therapies .
Another therapeutic strategy involves restoring UNC5A expression in cancers where it has been epigenetically silenced. Since O-GlcNAcylation influences PRC2-mediated repression of UNC5A, targeting the OGT/EZH2 interaction could potentially reactivate UNC5A expression . This approach might enhance the efficacy of conventional chemotherapy in colorectal cancer and other malignancies where UNC5A downregulation contributes to disease progression. Additionally, given UNC5A's associations with tumor immunity parameters, including immune checkpoint genes, it may inform immunotherapy strategies . Researchers investigating these therapeutic approaches should use comprehensive methodologies, including in vitro drug sensitivity assays, combination studies with conventional chemotherapeutics, and in vivo efficacy studies in appropriate animal models to evaluate both antitumor effects and potential toxicities.

What is the potential of UNC5A as a biomarker for cancer diagnosis and prognosis?

UNC5A demonstrates considerable potential as a biomarker for cancer diagnosis and prognosis, though its utility varies by cancer type. Systematic analyses have revealed associations between UNC5A expression and multiple clinically relevant parameters, including tumor mutation burden (TMB), microsatellite instability (MSI), and immune infiltration . These associations suggest UNC5A could serve as a pan-cancer biomarker with implications for treatment selection and outcome prediction.
The prognostic value of UNC5A depends on cancer type, as high UNC5A expression correlates with worse outcomes in some cancers (LUSC, KIRC, UVM) but better outcomes in others (LGG) . This context-dependence necessitates cancer-specific approaches when evaluating UNC5A as a prognostic marker. Methodologically, researchers should employ multivariate Cox regression analysis to assess UNC5A's independent prognostic value while controlling for established clinicopathological factors . Kaplan-Meier survival analysis with logrank tests provides visualization of survival differences between patient groups with high versus low UNC5A expression . Additionally, integrating UNC5A with other molecular markers may enhance prognostic accuracy, potentially leading to the development of UNC5A-based prognostic signatures. For diagnostic applications, researchers should evaluate UNC5A's sensitivity and specificity in distinguishing between normal and cancer tissues, as well as between different cancer subtypes, using receiver operating characteristic (ROC) curve analysis.

How does UNC5A contribute to cancer cell plasticity and metastasis?

UNC5A's role in cancer cell plasticity and metastasis represents a frontier in cancer research worthy of focused investigation. Current evidence suggests UNC5A may influence epithelial-to-mesenchymal transition (EMT), a process crucial for metastatic spread. This hypothesis stems from UNC5A's involvement in cell adhesion, migration, and invasion—key processes in EMT and metastasis . When studying these phenomena, researchers should employ a comprehensive approach including in vitro migration and invasion assays with UNC5A-modulated cells, along with analysis of EMT markers (E-cadherin, vimentin, Snail, etc.) at both mRNA and protein levels.
For more advanced insights, researchers should investigate UNC5A-dependent signaling pathways that might influence cell plasticity, such as TGF-β, Wnt/β-catenin, and Notch signaling. In vivo metastasis models, including tail vein injection and orthotopic implantation approaches, provide physiologically relevant systems for studying UNC5A's impact on metastatic potential. Additionally, single-cell RNA sequencing of heterogeneous tumor populations with varying UNC5A expression could reveal subpopulation-specific roles in tumor cell plasticity. Researchers should also explore potential interactions between UNC5A and the tumor microenvironment, as stromal and immune cells might influence UNC5A-mediated phenotypes through paracrine signaling or direct cell-cell interactions.

What role does UNC5A play in modulating the tumor microenvironment and immune evasion?

UNC5A's influence on the tumor microenvironment (TME) and immune evasion mechanisms represents a complex area requiring sophisticated research approaches. Evidence indicates associations between UNC5A expression and various immunomodulators, immune checkpoint genes, and immune cell infiltration patterns . These relationships suggest UNC5A may influence the recruitment, activation, and function of immune cells within the TME, potentially affecting antitumor immunity and response to immunotherapy. When investigating these relationships, researchers should employ multi-dimensional approaches. Flow cytometry and immunohistochemistry can assess immune cell populations in UNC5A-high versus UNC5A-low tumors, while RNA sequencing and protein arrays can identify UNC5A-dependent immunomodulatory factors. Co-culture experiments with cancer cells and immune cells (T cells, macrophages, dendritic cells) under conditions of UNC5A modulation provide insights into direct immune regulation. For in vivo studies, syngeneic mouse models with UNC5A-modified tumors allow evaluation of immune infiltration and response to immunotherapies like immune checkpoint inhibitors. Additionally, researchers should investigate whether UNC5A-dependent signaling affects expression of immune evasion markers like PD-L1 or secretion of immunosuppressive cytokines. Understanding these mechanisms could identify opportunities for therapeutic intervention that combines UNC5A targeting with immunotherapy approaches. Spatial transcriptomics and multiplex immunofluorescence would provide valuable insights into the spatial relationships between UNC5A-expressing cells and immune cell populations within the tumor microenvironment.

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.