This antibody oxidizes medium and long chain aldehydes into non-toxic fatty acids.
ALDH3B2, also known as ALDH8, is a 385 amino acid protein belonging to the aldehyde dehydrogenase family. It functions primarily in the pathway of alcohol metabolism by catalyzing the NADP+-dependent conversion of aldehydes into acids. ALDH3B2 is predominantly expressed in salivary gland tissue and is one of several mammalian ALDH3 genes involved in peroxidic and fatty aldehyde metabolism . The gene encoding ALDH3B2 maps to human chromosome 11, which contains approximately 1,400 genes and comprises nearly 4% of the human genome . Understanding ALDH3B2's normal physiological function provides critical context for interpreting its pathological roles in various disease states.
ALDH3B2 antibodies are immunoglobulins specifically designed to recognize and bind to the ALDH3B2 protein. High-quality ALDH3B2 antibodies typically detect the endogenous protein at its molecular weight of approximately 43 kDa. When selecting an ALDH3B2 antibody for research, consider factors such as antibody type (monoclonal vs. polyclonal), species reactivity, and validated applications. Polyclonal antibodies against ALDH3B2, such as those commercially available, generally show >95% purity by SDS-PAGE analysis . These antibodies can recognize various epitopes on the ALDH3B2 protein, making them particularly useful for immunohistochemistry and Western blot applications in research settings.
ALDH3B2 plays a significant role in promoting cholangiocarcinoma progression through multiple mechanisms. Research has demonstrated that ALDH3B2 enhances cell proliferation and clone formation by promoting the G1/S phase transition in the cell cycle . Additionally, ALDH3B2 contributes to the metastatic potential of cholangiocarcinoma cells by facilitating cell migration, invasion, and epithelial-mesenchymal transition (EMT) . Knockdown studies have shown that inhibition of ALDH3B2 expression reduces these aggressive cancer phenotypes and restrains tumor metastasis in vivo . At the molecular level, ALDH3B2 appears to exert these effects through regulation of ITGB1 (Integrin β1) expression and by modulating the phosphorylation of downstream signaling molecules including c-Jun, p38, and ERK . These findings establish ALDH3B2 as an important oncogenic driver in cholangiocarcinoma pathogenesis.
ALDH3B2 and ITGB1 (Integrin β1) demonstrate a significant functional relationship in cholangiocarcinoma progression. Research has revealed that patients with high expression of ALDH3B2 also exhibit high expression of ITGB1 in iCCA, pCCA, and dCCA at both mRNA and protein levels . Mechanistically, knockdown of ALDH3B2 leads to downregulation of ITGB1 expression and inhibits the phosphorylation of c-Jun, p38, and ERK in the downstream signaling pathway . Importantly, knockdown of ITGB1 counteracts the promoting effects of ALDH3B2 overexpression on cell proliferation, migration, and invasion . This interdependence suggests a regulatory axis where ALDH3B2 enhances cancer progression partly through ITGB1 upregulation. Furthermore, the combination of ITGB1 and ALDH3B2 as a dual biomarker demonstrates superior performance in predicting patient prognosis compared to either marker alone , indicating their synergistic role in cancer progression.
For detecting ALDH3B2 expression in tissue samples, immunohistochemistry (IHC) is the gold standard method. When performing IHC for ALDH3B2, tissue samples should be properly fixed in formalin, embedded in paraffin, and sectioned at 4-5 μm thickness. Antigen retrieval is critical and typically involves heat-induced epitope retrieval in citrate buffer (pH 6.0). For antibody selection, validated polyclonal antibodies that recognize endogenous ALDH3B2 protein should be used at optimized dilutions . Visualization systems with high sensitivity, such as DAB (3,3'-diaminobenzidine) chromogen, provide clear detection of ALDH3B2 expression. To assess expression levels, a standardized scoring system that considers both staining intensity and percentage of positive cells should be employed. This typically involves categorizing expression as low or high based on predetermined cutoff values . Additionally, quantitative PCR (qPCR) can serve as a complementary method for measuring ALDH3B2 mRNA expression, particularly when comparing paired tumor and non-tumor tissues .
Researchers can modulate ALDH3B2 expression in cellular models through several complementary approaches. For overexpression studies, transfection of expression plasmids containing the ALDH3B2 coding sequence under a strong promoter (such as CMV) is effective. Lentiviral or adenoviral vectors can also be employed for stable expression, particularly in difficult-to-transfect cell lines. For knockdown experiments, RNA interference (RNAi) using siRNA or shRNA targeting specific regions of ALDH3B2 mRNA has proven successful . The CRISPR-Cas9 gene editing system represents an advanced approach for complete knockout of ALDH3B2. When designing knockdown experiments, researchers should target multiple regions of the ALDH3B2 sequence to minimize off-target effects and validate knockdown efficiency through both mRNA (qPCR) and protein (Western blot) analyses . For functional studies, it's recommended to establish both overexpression and knockdown models in the same cell lines to comprehensively assess the biological effects of ALDH3B2 modulation.
ALDH3B2 belongs to the ALDH3 subfamily of the larger ALDH family, which includes ALDH3A1, ALDH3A2, and ALDH3B1. While all ALDH enzymes catalyze the oxidation of aldehydes to their corresponding acids, their tissue distribution, substrate preferences, and roles in cancer biology differ considerably. Unlike ALDH1, which is widely studied as a cancer stem cell marker and involved in cyclophosphamide resistance, or ALDH2, which is essential in ethanol metabolism and lipid peroxidation, ALDH3B2 appears to have more specialized functions . In cholangiocarcinoma, ALDH3B2 demonstrates stronger associations with tumor progression and patient prognosis compared to its close relative ALDH3B1, which shows upregulation in tumors but lacks prognostic significance . This functional divergence suggests distinct regulatory mechanisms and downstream effectors despite structural similarities. ALDH3B2's unique role in promoting cell proliferation through cell cycle regulation and enhancing metastatic potential through EMT underscores its specific importance in cancer biology that distinguishes it from other ALDH family members .
Given ALDH3B2's role in promoting proliferation, migration, invasion, and EMT in cholangiocarcinoma, it represents a promising therapeutic target. Direct inhibition of ALDH3B2 enzymatic activity could potentially reduce cancer cell growth and metastasis. Although specific ALDH3B2 inhibitors are not yet well-developed, research on other ALDH family members suggests that direct or indirect ALDH inhibition can reduce cell proliferation, invasion, and increase drug sensitization . Alternatively, targeting the ALDH3B2-ITGB1 axis could provide a novel therapeutic approach, given the demonstrated interdependence of these proteins in promoting cancer progression . RNA interference strategies specifically targeting ALDH3B2 might be developed into therapeutic tools, as knockdown studies have already demonstrated effectiveness in reducing cancer cell aggressiveness in vitro and in vivo . Furthermore, the prognostic value of ALDH3B2 suggests its potential utility in precision medicine approaches, where high ALDH3B2-expressing tumors might benefit from more aggressive treatment regimens or specific targeted therapies.
While direct evidence linking ALDH3B2 to drug resistance is still emerging, several mechanisms can be hypothesized based on its functions and the known roles of other ALDH family members. ALDH enzymes generally contribute to drug resistance by detoxifying reactive aldehydes generated by certain chemotherapeutic agents. ALDH1, for instance, dehydrogenates cyclophosphamide intermediates, conferring resistance to this drug . By similar mechanisms, ALDH3B2 might detoxify specific chemotherapeutic agents through its aldehyde-metabolizing activity. Additionally, ALDH3B2's promotion of the epithelial-mesenchymal transition (EMT) could indirectly contribute to drug resistance, as EMT is associated with reduced sensitivity to various chemotherapeutic agents . ALDH3B2's role in activating signaling pathways involving c-Jun, p38, and ERK phosphorylation could also modulate cellular responses to therapy, as these pathways regulate apoptosis, survival, and stress responses . Furthermore, if ALDH3B2 contributes to a cancer stem cell-like phenotype, as suggested by its role in promoting cell proliferation and invasion, this could further enhance therapeutic resistance through mechanisms associated with cancer stem cells.
Selecting appropriate ALDH3B2 antibodies requires careful consideration of several factors. First, determine whether monoclonal or polyclonal antibodies best suit the research application - monoclonal antibodies offer higher specificity for a single epitope, while polyclonal antibodies provide stronger signals by recognizing multiple epitopes . Second, verify the antibody's species reactivity, ensuring it recognizes ALDH3B2 from your experimental model (human, mouse, rat) . Third, confirm the antibody has been validated for your specific application (Western blot, IHC, flow cytometry, etc.) . For validation, perform Western blot analysis to confirm the antibody detects a single band at the expected molecular weight of 43 kDa . Additionally, use positive and negative controls, including tissues known to express ALDH3B2 (salivary gland) and those with low expression, respectively. Knockdown or knockout models serve as excellent negative controls . For cross-reactivity assessment, test the antibody against recombinant proteins of closely related ALDH family members, particularly ALDH3B1 . Finally, compare results across different detection methods (protein vs. mRNA) to ensure consistency in expression patterns.
Discrepancies between ALDH3B2 mRNA and protein expression can arise from various biological and technical factors. Biologically, post-transcriptional mechanisms including mRNA stability, microRNA regulation, translational efficiency, and protein stability can cause divergence between transcript and protein levels. When encountering such contradictions, researchers should first validate the specificity of both assays - for mRNA detection, primer specificity should be confirmed through sequencing of PCR products or using alternative primer sets targeting different regions of ALDH3B2 transcript . For protein detection, antibody specificity should be revalidated as discussed in question 5.1. Temporal considerations are also important, as mRNA levels may change more rapidly than protein levels in response to stimuli. In cancer research specifically, tumor heterogeneity can contribute to discrepancies when different sample portions are used for RNA and protein analyses . To resolve contradictions, researchers should employ multiple methodologies for both RNA (qPCR, RNA-seq) and protein (Western blot, IHC, flow cytometry) detection, analyze larger sample sizes, and potentially explore the role of post-transcriptional regulators like microRNAs that might explain the disconnect between transcription and translation of ALDH3B2.
The selection of experimental models for ALDH3B2 research should align with specific research objectives. For in vitro studies, cholangiocarcinoma cell lines with detectable ALDH3B2 expression provide relevant models, as ALDH3B2's role has been well-documented in this cancer type . When selecting cell lines, researchers should first screen for baseline ALDH3B2 expression levels to enable both overexpression and knockdown studies. Primary cell cultures derived from patient tumors offer advantages in terms of clinical relevance but may present challenges in terms of limited lifespan and variability. For in vivo investigations, xenograft models using ALDH3B2-modulated cell lines can assess effects on tumor growth and metastasis . Patient-derived xenografts (PDXs) maintain tumor heterogeneity and microenvironment interactions, providing a more physiologically relevant system. Genetically engineered mouse models (GEMMs) with ALDH3B2 overexpression or knockout, particularly in tissues where it's implicated in cancer (such as biliary tissue), could offer insights into its role in tumor initiation and progression. Finally, organoid cultures bridge the gap between 2D cell culture and in vivo models, allowing the study of ALDH3B2 in more complex tissue architectures while maintaining experimental accessibility.