TPD52L1, also known as hD53, is a member of the tumor protein D52 (TPD52) family. This protein contains a coiled-coil domain that enables it to form homo- or hetero-dimers with other TPD52 family members . TPD52L1 plays significant roles in:
Cell proliferation pathways
Calcium signaling mechanisms
Apoptotic regulation through interaction with mitogen-activated protein kinase kinase kinase 5 (MAP3K5/ASK1)
Unlike its family member TPD52, TPD52L1 does not appear to significantly participate in lipid storage or lipid droplet formation in cells . Multiple alternatively spliced transcript variants of TPD52L1 have been identified, though the complete characterization of some variants remains ongoing .
TPD52L1 antibodies have been validated for multiple experimental applications with specific recommended parameters:
Application | Recommended Dilution | Notes |
---|---|---|
Western Blot (WB) | 1:500-1:1000 | Validated in MCF-7 cells |
Immunoprecipitation (IP) | 0.5-4.0 μg per 1.0-3.0 mg protein | Validated in MCF-7 cells |
Immunohistochemistry (IHC) | 1:50-1:500 | Validated in human prostate cancer tissue |
Immunofluorescence (IF)/ICC | 1:20-1:200 | Validated in MCF-7 cells |
ELISA | 1:8000 (for some antibodies) | Application-specific |
The specific application determines the optimal working dilution, and researchers should conduct preliminary experiments to establish the ideal conditions for their specific experimental system .
TPD52L1 exhibits a notable discrepancy between its calculated and observed molecular weights:
Calculated molecular weight: 22 kDa (based on amino acid sequence)
Observed molecular weight in experiments: 25 kDa (in Western blot analyses)
This difference is likely attributable to post-translational modifications. Additionally, different isoforms resulting from alternative splicing demonstrate varying molecular weights:
When analyzing Western blot results, researchers should anticipate these variations and optimize gel resolution accordingly.
For optimal TPD52L1 detection in formalin-fixed, paraffin-embedded tissues, appropriate antigen retrieval is critical. The following methods are recommended:
Primary method: Heat-induced epitope retrieval using TE buffer pH 9.0
Immerse slides in TE buffer and heat to 95-98°C for 15-20 minutes
Allow gradual cooling before proceeding with IHC protocol
Alternative method: Citrate buffer pH 6.0
May be employed if the primary method yields suboptimal results
Follow similar heating parameters as above
When working with human prostate cancer tissue (a positive control for TPD52L1), TE buffer at pH 9.0 has been demonstrated to produce reliable results . The method selection may depend on the specific antibody clone and tissue type being examined.
Based on extensive validation data, the following positive controls are recommended:
Cell lines:
Tissue samples:
When establishing a new experimental system, these samples serve as appropriate positive controls to verify antibody performance. For negative controls, researchers should consider using cell lines with confirmed low TPD52L1 expression or employing siRNA knockdown in positive cell lines.
Proper storage and handling are essential for maintaining antibody performance over time:
Storage temperature: -20°C is optimal for long-term stability
Typical formulation: PBS with 0.02% sodium azide and 50% glycerol (pH 7.3)
For aliquoting practices:
Always ensure proper temperature maintenance during shipping and handling to preserve antibody functionality.
Despite belonging to the same protein family, TPD52L1 and TPD52 demonstrate distinct functional roles in lipid metabolism:
TPD52 expression substantially increases neutral lipid storage and lipid droplet formation in cultured cells (approximately 4-10 fold increase in lipid droplet numbers compared to control cells)
In contrast, TPD52L1-expressing cell lines show lipid droplet staining patterns comparable to control cells, suggesting minimal involvement in enhanced lipid storage
This functional distinction is further supported by protein interaction studies:
TPD52 directly interacts with ADRP (also known as PLIN2), a lipid droplet-associated protein
No direct interaction has been detected between TPD52L1 and ADRP in yeast two-hybrid system analyses
When designing comparative studies of TPD52 family proteins, researchers should account for these isoform-specific functional differences and employ standardized lipid quantification methods.
TPD52L1 has been identified as an interactor with mitogen-activated protein kinase kinase kinase 5 (MAP3K5/ASK1), a central regulator of stress-induced apoptosis. Current research indicates that:
TPD52L1 positively regulates MAP3K5-induced apoptotic pathways
The interaction likely involves the coiled-coil domain of TPD52L1
This function represents a distinct role from other TPD52 family members
For researchers investigating these apoptotic mechanisms, several experimental approaches should be considered:
Co-immunoprecipitation assays to confirm the interaction in specific cell types
Knockdown/knockout studies to evaluate effects on MAP3K5 signaling
Phosphorylation analysis to determine if TPD52L1 is a substrate of MAP3K5
Apoptosis assays (caspase activation, PARP cleavage, Annexin V staining) with and without TPD52L1 expression
The precise mechanisms underlying TPD52L1's enhancement of MAP3K5-induced apoptosis represent an important area for ongoing investigation.
TPD52L1 undergoes alternative splicing, producing multiple transcript variants with distinct functional properties. To effectively detect and distinguish these variants, researchers can employ several complementary approaches:
RT-PCR with isoform-specific primers:
Design primers spanning exon-exon junctions unique to each variant
Use nested PCR for low-abundance transcripts
Quantitative real-time PCR provides relative expression levels
Western blotting:
Isoform-specific antibodies:
When reporting research findings, clear specification of which isoforms were detected and the methods used for distinction is essential, as different isoforms may possess distinct functional properties.
The discrepancy between calculated molecular weight (22 kDa) and observed migration pattern (25 kDa) can be attributed to several factors:
Post-translational modifications:
Phosphorylation: TPD52 family proteins undergo phosphorylation, adding approximately 80 Da per phosphate group
Potential glycosylation, though not extensively reported for TPD52L1
Protein structure and composition:
Proportion of acidic amino acids can cause anomalous migration on SDS-PAGE
The coiled-coil domain may affect SDS binding efficiency
Technical considerations:
To address this discrepancy experimentally:
Use multiple molecular weight markers
Consider running dephosphorylation controls (treat lysates with phosphatase)
Compare migration patterns under reducing and non-reducing conditions
Understanding the basis for this difference may provide insights into TPD52L1's post-translational regulation in different cellular contexts.
Researchers encountering weak signal when detecting TPD52L1 by Western blot can implement several optimization strategies:
Sample preparation enhancement:
Enrich for TPD52L1 using subcellular fractionation
Use comprehensive protease inhibitor cocktails during lysis
Consider phosphatase inhibitors if phosphorylated forms are of interest
Protein loading optimization:
Detection system improvements:
Employ more sensitive detection methods (enhanced chemiluminescence systems)
Consider fluorescent secondary antibodies for quantitative analysis
Try biotin-streptavidin amplification systems
Antibody optimization:
When implementing these approaches, modify one parameter at a time to identify the specific factor limiting signal intensity. Document successful optimization strategies for reproducibility.
Thorough validation of antibody specificity is essential for reliable experimental outcomes. For TPD52L1 antibody, several complementary approaches are recommended:
Genetic manipulation approaches:
siRNA or shRNA knockdown of TPD52L1 should proportionally reduce signal intensity
CRISPR/Cas9 knockout provides a definitive negative control
Overexpression systems can confirm expected molecular weight patterns
Peptide competition assays:
Pre-incubate antibody with immunizing peptide
Should abolish specific signal while non-specific binding remains
Multiple antibody validation:
Utilize antibodies raised against different epitopes of TPD52L1
Consistent results across different antibodies increase confidence in specificity
Cross-reactivity assessment:
Test against recombinant TPD52, TPD52L1, and TPD52L2 proteins
Ensures the antibody discriminates between family members
Orthogonal methods:
Comprehensive documentation of validation experiments should be maintained to support the reliability of research findings involving TPD52L1 antibody.
Given TPD52L1's roles in various cellular processes through protein-protein interactions, several complementary methodological approaches are recommended:
Co-immunoprecipitation (Co-IP):
Yeast two-hybrid (Y2H) screening:
GST pulldown assays:
Proximity-ligation assays (PLA):
Allows visualization of protein interactions in situ
Provides spatial information about interaction sites within cells
When studying TPD52L1 interactions, consider that the protein may form both homo- and hetero-dimers with TPD52 family members, potentially complicating interpretation of results. Multiple approaches should be employed to confirm interactions.
As TPD52L1 continues to be investigated in various cellular processes, several emerging research directions warrant consideration:
Development of isoform-specific antibodies to better distinguish between alternatively spliced variants
Exploration of TPD52L1's potential roles in cancer progression through comparative studies of normal and malignant tissues
Investigation of post-translational modifications of TPD52L1 and their functional significance
Further characterization of the regulatory mechanisms controlling TPD52L1 expression and activity
Tumor Protein D52 L1 (TPD52L1) is a member of the Tumor Protein D52 (TPD52) family, which is known for its role in cellular proliferation, survival, and vesicle trafficking. The TPD52 family consists of several proteins, including TPD52, TPD52L1, TPD52L2, and TPD52L3, which share a conserved coiled-coil domain that facilitates protein-protein interactions .
TPD52L1 is expressed in various tissues and has been implicated in several types of cancer, including breast, prostate, and ovarian cancers. The overexpression of TPD52L1 has been associated with tumorigenesis, suggesting that it plays a critical role in cancer development and progression . Studies have shown that TPD52L1 can interact with other proteins involved in cell cycle regulation and apoptosis, further highlighting its importance in cancer biology .
Mouse Anti-Human TPD52L1 antibodies are monoclonal antibodies generated by immunizing mice with human TPD52L1 protein. These antibodies are highly specific and can be used in various applications, including Western blotting, immunohistochemistry, and flow cytometry . The primary purpose of these antibodies is to detect and quantify TPD52L1 expression in human tissues and cell lines, aiding in cancer research and diagnostics .
The production of Mouse Anti-Human TPD52L1 antibodies involves several steps:
These antibodies are stored at -20°C to maintain their stability and activity .
The use of Mouse Anti-Human TPD52L1 antibodies has several important applications in cancer research: