CDC37L1 (also known as CDC37B or HARC) is a co-chaperone protein that contains 337 amino acids and is structurally similar to CDC37. Unlike its analog CDC37 (which promotes tumor progression), CDC37L1 functions as a tumor suppressor in several cancer types. Research shows that CDC37L1 expression is decreased in high-grade gastric cancer tissues compared to low-grade tissues, and lower expression correlates with poor patient survival rates . CDC37L1 contributes to the regulation of HSP90 function and appears to inhibit cancer cell proliferation and migration through various mechanisms, most notably through CDK6 reduction .
CDC37L1 antibodies have been validated for multiple laboratory applications:
| Application | Recommended Dilution | Sample Types |
|---|---|---|
| Western Blot (WB) | 1:1000-1:5000 | Cell lysates, tissue homogenates |
| Immunohistochemistry (IHC) | 1:50-1:500 | FFPE tissue sections |
| ELISA | 1:40000 | Serum, plasma, cell culture supernatants |
For optimal results, these antibodies have been successfully tested on various human samples including HeLa cells, Jurkat cells, MCF-7 cells, U2OS cells, and human kidney and liver tissues . When performing IHC, antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0 is recommended for best results .
CDC37L1 antibodies typically come in liquid form in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide at pH 7.3 . For optimal stability:
Store antibodies at -20°C where they remain stable for approximately one year after shipment
For smaller antibody quantities (≤20μl), aliquoting is generally unnecessary for -20°C storage
For larger volumes, consider creating single-use aliquots to avoid repeated freeze-thaw cycles
Upon receipt of new antibodies, some manufacturers recommend storing at -80°C for longest shelf-life
Always centrifuge briefly before opening the antibody vial to ensure collection of all liquid
Proper controls are essential for validating CDC37L1 antibody specificity:
Positive controls: Use cell lines known to express CDC37L1 such as HeLa, Jurkat, MCF-7, or U2OS cells
Negative controls: Consider primary antibody omission controls and isotype controls
Knockdown/knockout validation: Implement CDC37L1 siRNA knockdown or CRISPR/Cas9 knockout samples as gold-standard specificity controls
Molecular weight verification: Confirm detection at the expected molecular weight of approximately 39 kDa
Tissue expression patterns: Compare results with known CDC37L1 expression patterns (expressed in kidney and liver tissues)
Researchers should validate each new lot of antibody in their specific experimental system before use in critical experiments.
For optimal Western blot detection of CDC37L1:
Sample preparation:
Use RIPA buffer with protease inhibitors for efficient extraction
Load 20-40 μg of total protein per lane
Gel electrophoresis:
Transfer conditions:
Semi-dry or wet transfer systems work well
Transfer at 100V for 90 minutes or 30V overnight in cold room
Blocking and antibody incubation:
Signal detection:
Based on published research using CDC37L1 antibodies in tissue microarrays :
Antigen retrieval:
Antibody dilution and incubation:
Scoring system for expression levels:
Evaluate both staining intensity (weak, moderate, strong) and percentage of positive cells
Use two independent pathologists for scoring to reduce subjective bias
Consider automated image analysis systems for quantitative assessment
Controls and reporting:
CDC37L1 antibodies can be utilized in multiple sophisticated approaches to investigate its mechanistic role in cancer:
Co-immunoprecipitation (Co-IP):
Chromatin immunoprecipitation (ChIP):
Investigate whether CDC37L1 associates with chromatin-bound complexes
May help elucidate its role in transcriptional regulation
Proximity ligation assay (PLA):
Visualize protein-protein interactions in situ
Particularly useful for confirming interactions between CDC37L1 and potential partners like HSP90
Immunofluorescence microscopy:
Track subcellular localization changes during cancer progression
Combine with other markers to assess co-localization with client proteins
Proteomic approaches:
Use antibody-based enrichment followed by mass spectrometry
Can identify novel CDC37L1-associated proteins in different cancer contexts
These approaches have helped establish CDC37L1's role in inhibiting gastric cancer cell proliferation and migration through CDK6 reduction .
CDC37L1 functions as a co-chaperone for HSP90, potentially regulating its function . This relationship can be investigated using:
In vitro binding assays:
Pull-down assays using recombinant proteins
Surface plasmon resonance to measure binding kinetics
Isothermal titration calorimetry for thermodynamic parameters
Cellular assays:
Co-IP with CDC37L1 antibodies followed by HSP90 detection
Manipulation of CDC37L1 levels (overexpression/knockdown) to assess impact on HSP90 client protein stability
Assess whether CDC37L1 competes with CDC37 for HSP90 binding
Functional assays:
ATPase activity assays to determine if CDC37L1 affects HSP90's enzymatic function
Client protein folding assays in the presence/absence of CDC37L1
HSP90 inhibitor sensitivity in cells with varying CDC37L1 expression
Research has shown that CDC37L1 strengthens the binding between HSP90 and certain client proteins like PPIA (peptidylprolyl isomerase A), suggesting it may selectively modulate HSP90's chaperone function toward specific client proteins .
CDC37L1 has been implicated in drug resistance mechanisms, particularly regarding sorafenib resistance in hepatocellular carcinoma (HCC):
Expression correlation with therapy response:
Mechanistic insights:
MicroRNA regulation:
Potential therapeutic targets:
Targeting CDC37L1 or its regulatory miRNAs might be a strategy to overcome sorafenib resistance
Combined inhibition of HSP90 and modulation of CDC37L1 levels could sensitize resistant tumors
When encountering conflicting data about CDC37L1's role across cancer types:
Methodological considerations:
Different antibodies may have varying specificities and sensitivities
Compare antibody clones, dilutions, and detection methods used across studies
Evaluate sample preparation techniques and scoring systems in IHC studies
Biological context differences:
Genetic background influences:
Analyze whether genetic alterations in related pathways modify CDC37L1 function
Consider oncogenic drivers present in different tumor types (e.g., RAS mutations)
Technical validation approaches:
Use multiple detection methods (IHC, WB, qPCR) to confirm expression findings
Employ functional studies (knockdown/overexpression) to validate biological effects
Consider spatial heterogeneity in tumors that might affect sampling
Data integration strategies:
Correlate findings with public databases (e.g., TCGA) for broader context
Meta-analysis approaches may help resolve contradictions
Single-cell analysis can reveal heterogeneity masked in bulk tissue studies
Research has revealed specific patterns of CDC37L1 expression across cancer progression:
Understanding these patterns has important implications for using CDC37L1 as a prognostic biomarker and potential therapeutic target.
CDC37L1 antibodies enable several innovative screening strategies:
Antibody-drug conjugate (ADC) development:
High-throughput compound screening:
CDC37L1 antibodies can be used in cell-based assays to screen for compounds that modulate its expression or function
Changes in CDC37L1 levels or localization following compound treatment can be detected by immunofluorescence or high-content imaging
PROTAC (Proteolysis Targeting Chimera) development:
CDC37L1 antibodies can help validate degradation of target proteins in PROTAC development
Monitor changes in CDC37L1 client proteins following targeted degradation approaches
Synthetic lethal interactions:
Combination therapy evaluation:
CDC37L1 antibodies can monitor expression changes during combination treatments
Particularly relevant for combinations of HSP90 inhibitors with other targeted therapies
Advanced techniques for investigating CDC37L1's role in cell cycle regulation include:
Live-cell imaging approaches:
Fluorescently tagged CDC37L1 to track localization during cell cycle progression
FRET/BRET-based biosensors to detect interactions with cell cycle proteins in real-time
Mass spectrometry-based interactomics:
BioID or APEX proximity labeling to identify cell cycle-specific interaction partners
Quantitative proteomics to measure changes in the CDC37L1 interactome across cell cycle phases
Functional genomics screens:
Cell cycle synchronization studies:
Examine CDC37L1 expression and function in synchronized cell populations
Flow cytometry analysis combined with CDC37L1 antibody staining to correlate with cell cycle phases
Flow cytometry has revealed that CDC37L1 knockdown leads to increased cells in S phase, an effect inhibited by CDK4/6 inhibitor Palbociclib
CDK activity assays:
In vitro kinase assays to assess CDC37L1's impact on CDK activity
Phospho-specific antibodies to measure CDK substrate phosphorylation
These advanced approaches are revealing CDC37L1's mechanistic role in regulating cell proliferation through modulation of CDK6 and potentially other cell cycle proteins.