HLTF is a SWI/SNF family protein encoded by the HLTF gene on human chromosome 3. It functions as a:
Ubiquitin ligase for proliferating cell nuclear antigen (PCNA) polyubiquitination
Key regulator of genome stability through G-quadruplex (G4) suppression and replication stress response
Hepatocellular Carcinoma (HCC):
| Experimental Model | Key Result | Citation |
|---|---|---|
| Xenograft tumors | HLTF silencing ↓ tumor volume by 60% | |
| Orthotopic models | HLTF overexpression ↑ lung metastases | |
| Patient tissues | High HLTF linked to 5-year survival <30% |
G-quadruplex Regulation:
Replication Stress Response:
HLTF's three critical domains govern its roles:
| Domain | Function | Impact of Mutation |
|---|---|---|
| HIRAN | DNA binding | Disrupts replication fork reversal |
| RING Finger | Ubiquitin ligase activity | Impairs PCNA modification |
| ATPase | G4 resolution & ALT suppression | ↑ Telomeric RNA-DNA hybrids |
These domains make HLTF antibody essential for studying structure-function relationships in DNA repair mechanisms .
HLTF is a multifunctional protein that serves primarily as a DNA translocase involved in post-replication DNA repair. HLTF has several critical functions:
Fork reversal activity: HLTF promotes replication fork remodeling during replication stress, preventing other mechanisms of replication stress tolerance in cancer cells . EM studies have confirmed HLTF as a bona fide fork reversal protein in human cells, with HLTF-KO cell lines exhibiting a significant 2-3 fold reduction in reversed fork frequency .
Ubiquitin ligase activity: Similar to yeast Rad5, HLTF functions as a ubiquitin ligase that promotes the polyubiquitination of proliferating cell nuclear antigen (PCNA) at its Lys-164 residue . This activity is crucial for error-free post-replication repair of damaged DNA.
Interaction with DNA repair proteins: HLTF physically interacts with multiple DNA repair proteins including the Rad6-Rad18 and Mms2-Ubc13 ubiquitin-conjugating enzyme complexes . It also interacts with mismatch repair (MMR) proteins, with human HLTF shown to directly interact with MSH2 .
HLTF has a molecular weight of approximately 130 kDa on SDS-PAGE gels . When working with HLTF antibodies:
The immunoprecipitated HLTF typically runs at the predicted molecular weight of 116 kDa
Non-immunoprecipitated HLTF in input lanes may run at a slightly higher molecular weight and show several additional bands
This pattern is consistent with manufacturer specifications for commercial HLTF antibodies
HLTF is expressed in various cell types including:
Primary human macrophages (MDMs), with expression detected at day 4 or day 7 following differentiation of freshly isolated monocytes
Multiple tissues in human and animal models, though expression levels vary significantly
Interestingly, HLTF expression in non-dividing cells like macrophages suggests potential functions beyond its established role in S-phase DNA repair .
To detect HLTF's role in replication stress, several experimental approaches are recommended:
DNA fiber analysis to assess replication dynamics:
Pulse-label cells with IdU followed by CldU in the presence of replication stressors (e.g., 50μM HU or MMC)
In wild-type cells, replication tracts shorten by ~30% upon drug treatment
HLTF-KO cells exhibit unrestrained fork progression (no shortening of tracts)
This phenotype can be observed in various cell types including cancer cell lines (K562) and non-cancerous cells (RPE1)
Detection of fork reversal by electron microscopy:
Isolate replication intermediates after exposing cells to replication stressors
Use in vivo psoralen crosslinking and EM analysis
Quantify reversed fork structures (typically ~23% of replication intermediates in wild-type cells treated with HU)
HLTF-KO cells show 2-3 fold reduction in reversed fork frequency
S1 nuclease assay for discontinuous replication:
Treat cells with replication stressors (e.g., 50μM HU)
Incubate permeabilized cells with/without S1 nuclease
S1 treatment specifically shortens replication tracts in HLTF-KO cells under stress conditions, indicating discontinuous replication with ssDNA gaps
Comprehensive controls are essential when investigating HLTF function:
Genetic controls:
HLTF-KO cell lines (multiple clones recommended to rule out off-target effects)
HLTF-HIRAN domain mutants (to distinguish effects of different HLTF domains)
Complementation with wild-type or mutant HLTF expression constructs
Double knockout models (e.g., HLTF-PRIMPOL-dKO) to study pathway dependencies
Experimental controls for immunoprecipitation:
DNase treatment controls to distinguish DNA-mediated from direct protein-protein interactions
Empty vector controls for tagged-protein expression systems
Negative control antibodies for immunoprecipitation
Input samples (5-10% of lysate) to verify protein expression levels
Technical validation:
siRNA-resistant HLTF constructs for rescue experiments
Multiple HLTF antibodies targeting different epitopes
Cell cycle synchronization to account for cell cycle-dependent effects
HLTF belongs to a small family of DNA translocases including SMARCAL1 and ZRANB3 that catalyze fork regression activity. To distinguish their functions:
Protein-specific interactions:
HLTF selectively interacts with MSH2 but not MLH1
SHPRH interacts with MLH1 but not MSH2
These differential interactions persist after DNase treatment, confirming specific protein-protein interactions
Functional assays:
MNNG sensitivity: SHPRH-KO cells show moderate resistance to alkylating agents, whereas HLTF-KO cells remain sensitive
Double knockout (HLTF/SHPRH-dKO) shows similar resistance to SHPRH-KO alone, suggesting non-redundant functions
Domain-specific assays:
The HIRAN domain of HLTF is required for restraining replication fork progression
HLTF's RING domain mediates its ubiquitin ligase activity
Domain-specific mutations can help differentiate the multiple functions of HLTF
When investigating HLTF in cancer contexts, researchers should consider:
Expression analysis:
Experimental models:
Cell line-derived xenograft (CDX) models with controlled HLTF status are valuable
For colorectal cancer studies, HCT116-derived models with shRNA knockdown of HLTF have been established
Both the tumor cells and tumor microenvironment (TME) should be considered, as HLTF status in both compartments affects tumor biology
Response to therapy:
HLTF-deficient cells show reduced DNA damage signaling (ATR/ATM) after replication stress
HLTF loss can affect sensitivity to chemotherapeutic agents
Combined analysis of HLTF with other DNA repair factors provides more comprehensive insights into therapy response
Several factors can contribute to variable HLTF detection patterns:
Post-translational modifications:
HLTF undergoes various modifications that can alter its migration pattern
Ubiquitination of HLTF (particularly in HIV-1 Vpr-expressing cells) can generate additional bands
Phosphorylation status may vary depending on cellular context
Protein degradation mechanisms:
HLTF can be targeted for proteasomal degradation by viral proteins like HIV-1 Vpr
Endogenous degradation pathways may generate cleavage products
Use of protease inhibitors during sample preparation is critical
Technical recommendations:
Freshly prepared samples typically yield cleaner results
Include positive controls from cells known to express HLTF
Compare results using antibodies targeting different HLTF epitopes
Verify specificity using HLTF-knockout lysates as negative controls
For successful HLTF co-immunoprecipitation studies:
Sample preparation:
Nuclear fractionation is recommended as HLTF primarily localizes to the nucleus
Use of 1:50 dilution of HLTF antibody has been validated for immunoprecipitation
Include DNase treatment controls to distinguish DNA-mediated from direct protein interactions
Buffer conditions:
Salt concentration is critical: too high may disrupt interactions, too low increases nonspecific binding
Mild detergents (0.1% NP-40) help maintain protein complex integrity
Consider crosslinking approaches for transient interactions
Validation approaches:
Reciprocal IP (pull down with antibody against interaction partner)
Both endogenous and tagged-protein approaches provide complementary evidence
Follow interactions under different conditions (e.g., DNA damage, cell cycle phases)
When studying HLTF's impact on cell cycle progression:
Common pitfalls:
Cell type-specific effects: HLTF loss impacts different cell types differently
Failure to account for cell synchronization: HLTF's effects are most pronounced during S-phase
Incomplete pathway analysis: HLTF functions within complex DNA repair networks
Recommended approaches:
Quantitative image-based cytometry (QIBC) for cell cycle analysis provides superior resolution compared to standard flow cytometry
Dual pulse labeling (e.g., EdU/BrdU) allows tracking of cell progression through S-phase
Combined analysis of DNA content and replication markers provides more complete cell cycle assessment
Key considerations:
HLTF-KO cells progress faster through S-phase under replication stress conditions
This accelerated progression depends partly on the primase PRIMPOL
Cell cycle differences emerge after replication stress induction, not under normal conditions
HLTF contains several functional domains with distinct roles:
HIRAN domain mutations:
ATPase domain mutations:
The DE557,558AA ATPase mutant lacks translocase activity but retains protein interaction capabilities
This mutation abolishes fork regression activity but not protein complex formation
Useful for separating enzymatic from scaffolding functions of HLTF
RING domain mutations:
Affect HLTF's ubiquitin ligase activity
Can be used to study PCNA polyubiquitination independently of fork remodeling
Important for understanding pathway-specific functions
To effectively study HLTF's functions at replication forks:
In vitro fork regression assays:
Purified HLTF protein can process model replication fork structures
HLTF concertedly unwinds and anneals the nascent and parental strands without exposing extended single-stranded regions
SSB proteins do not affect HLTF-mediated fork regression, unlike conventional helicase assays
Chromatin association studies:
iPOND (isolation of Proteins On Nascent DNA) to identify HLTF recruitment to replication forks
SIRF (in situ analysis of protein interactions at DNA replication forks) for visualization of HLTF at individual forks
ChIP-seq approaches for genome-wide analysis of HLTF binding sites
Protein-protein interaction mapping:
HLTF interacts with key replication and repair factors including PCNA, Rad18, Mms2, and Ubc13
These interactions occur constitutively and are not significantly affected by DNA damage induction
Both endogenous and tagged protein approaches provide complementary information
The literature contains seemingly contradictory findings about HLTF in cancer:
Tumor suppressor evidence:
HLTF is frequently silenced by hypermethylation in colorectal cancer
HLTF promotes error-free DNA repair, consistent with tumor suppressor function
Oncogenic properties:
HLTF is upregulated in hepatocellular carcinoma and promotes tumor progression
HLTF-deleted cancer cells show increased sensitivity to certain DNA damaging agents
HLTF can activate specific signaling pathways like ERK/MAPK in HCC
Reconciliation approaches:
Tissue-specific effects: HLTF may have different roles depending on tissue context
Stage-specific functions: HLTF may be important for initial genome stability but problematic in established cancers
Pathway dependencies: HLTF's function may depend on the status of other DNA repair pathways
Consider both tumor cells and the tumor microenvironment, as HLTF status in both can affect cancer progression
Researchers have multiple options for modulating HLTF expression:
Working with HLTF in primary samples presents unique challenges:
Primary cell isolation and culture:
HLTF is expressed in primary human macrophages and can be studied following monocyte differentiation
Expression should be verified as it may vary with differentiation stage (e.g., detectable at day 4 or 7 post-differentiation)
Primary lymphocytes and dendritic cells may also be relevant for studying HLTF's immune functions
Tissue sample analysis:
Immunohistochemistry for HLTF requires careful optimization and validation
FFPE samples can be used with appropriate antigen retrieval methods
Spatial transcriptomics provides valuable information on positional gene patterns within intact tissue samples
Multi-omics approaches:
Combining RNAseq with species-specific mapping and spatial transcriptomics provides comprehensive insights
Proteomics approaches (e.g., 2D DIGE, MALDI-TOF/TOF mass spectrometry) complement transcriptomic data
Integrated analysis yields holistic understanding of HLTF's role in complex biological processes
To assess HLTF's impact on DNA repair mechanisms:
Replication stress tolerance:
Colony formation assays following treatment with replication stressors (HU, MMS, MMC)
HLTF-deficient cells show reduced DSB formation and increased survival upon replication stress
HLTF loss confers mild resistance to alkylating agents like MNNG, but not to the level of MMR-deficient cells
DNA damage signaling:
Western blot analysis of ATR/ATM activation markers following high-dose HU treatment (3mM)
HLTF-KO cells show reduced DNA damage signaling compared to wild-type cells
Analysis of γH2AX foci formation provides cellular resolution of DNA damage response
Pathway-specific assays:
PRIMPOL-dependent mechanisms can be assessed in HLTF-KO cells using PRIMPOL knockdown/knockout
REV1-dependent translesion synthesis in HLTF-HIRAN mutants requires specific TLS polymerase inhibition/depletion
Combined genetic approaches (double knockouts, domain-specific mutations) help dissect pathway dependencies