RNASEH1 is a ribonuclease that cleaves the RNA strand in RNA-DNA hybrids, playing roles in transcription termination, DNA replication, and R-loop resolution . The HRP-conjugated antibody binds specifically to RNASEH1, marking it for detection in assays. HRP catalyzes colorimetric reactions, such as the oxidation of tetramethylbenzidine (TMB), generating measurable signals in ELISA .
This antibody is optimized for sensitivity and specificity, avoiding cross-reactivity with other proteins .
The HRP-conjugated antibody enables precise quantification of RNASEH1 levels in cell lysates or biological samples. Its use is critical in studies investigating RNASEH1 expression in diseases linked to RNA-DNA hybrid instability, such as genomic disorders or cancer .
| Feature | HRP-Conjugated (PACO58874) | Non-HRP (e.g., ab229078) |
|---|---|---|
| Primary Use | ELISA | WB, IHC-P |
| Signal Detection | Enzymatic amplification | Fluorescent/chromogenic |
| Reactivity | Human | Human, Zebrafish |
| Cited Publications | Not explicitly reported | 2 publications |
RNASEH1 is essential for resolving R-loops, which are implicated in genomic instability and transcriptional stress . While GFP-dRNH1 (a catalytically inactive RNASEH1 variant) is used to visualize RNA-DNA hybrids, the HRP-conjugated antibody quantifies RNASEH1 levels, aiding in studies of its regulation under pathological conditions .
RNASEH1 accumulation correlates with chromosome size-dependent DNA replication timing in organisms like Leishmania, suggesting its role in coordinating replication with transcription . The HRP-conjugated antibody could elucidate RNASEH1’s dynamics during cell cycle progression.
Specificity: Unlike GFP-dRNH1, which binds RNA-DNA hybrids directly, this antibody detects RNASEH1 protein, not its activity .
Cross-Reactivity: While optimized for human samples, cross-reactivity with other species (e.g., mouse, rat) is not confirmed .
Alternative Probes: S9.6 antibody and GFP-dRNH1 are used for hybrid detection, but the HRP-conjugated antibody focuses on protein quantification .
RNASEH1 (Ribonuclease H1) is an endonuclease that specifically degrades the RNA component of RNA-DNA hybrids. It belongs to the RNase H family and contains one RNase H domain . The human RNASEH1 gene is located on chromosome 2p25.3, with pseudogenes found on chromosomes 17p11.2 and 1q .
RNASEH1 plays several crucial biological roles:
It degrades RNA-DNA hybrids that form during replication and repair processes, which could lead to DNA instability if not processed .
It participates in RNA polymerase II (RNAp II) transcription termination by degrading R-loop RNA-DNA hybrid formation at G-rich pause sites located downstream of the poly(A) site .
It is essential for mitochondrial R-loop processing, transcription, and mitochondrial DNA replication .
It is necessary for the activity of antisense oligonucleotides (ASOs) designed to serve as RNase H1 substrates .
Eukaryotic RNase H1 has evolved a Hybrid Binding Domain that confers processivity and affinity for the substrate, making it larger and more complex than its prokaryotic counterparts .
Based on the technical data available, RNASEH1 Antibody, HRP conjugated is suitable for the following applications:
| Application | Suitability | Recommended Dilution |
|---|---|---|
| ELISA | Primary application for HRP-conjugated format | Varies by product |
| Western Blot (WB) | Suitable with HRP conjugate | 1:500-1:50000 |
| Immunohistochemistry (IHC) | Suitable with HRP conjugate | 1:200-1:6000 |
For non-HRP conjugated RNASEH1 antibodies, additional applications include:
The HRP conjugation eliminates the need for secondary antibody detection, making the antibody particularly suitable for direct detection in applications such as ELISA, Western blot, and IHC .
According to multiple antibody product specifications:
This consistency between calculated and observed molecular weights indicates that RNASEH1 typically does not undergo significant post-translational modifications that would substantially alter its migration pattern on SDS-PAGE gels. When performing Western blot analysis with RNASEH1 antibodies, researchers should expect to observe a clear band at approximately 32 kDa .
Validation data from various antibody suppliers confirms detection of RNASEH1 in multiple cell lines, including:
HeLa cells (human cervical adenocarcinoma)
A549 cells (human lung carcinoma)
HEK-293 cells (human embryonic kidney)
U2OS cells (human osteosarcoma)
Based on manufacturer specifications, the recommended storage and handling conditions for RNASEH1 Antibody, HRP conjugated are:
It is important to minimize freeze-thaw cycles to preserve antibody functionality. Follow manufacturer-specific recommendations, as formulations may vary slightly between suppliers .
Validating the specificity of RNASEH1 antibody in R-loop detection experiments requires multiple experimental controls:
Enzymatic Digestion Controls:
Treat samples with E. coli RNase H1 prior to immunoprecipitation or immunostaining. As demonstrated in knockout mouse studies, "the immunoprecipitated samples from the cKO mice digested with the E. coli enzyme showed S7 R-loop levels comparable to the cControl samples" . This approach confirms that detected signals are indeed from RNA-DNA hybrids.
Genetic Controls:
Use cells with RNASEH1 knockout or knockdown as reference points. The liver-specific RNase H1 knockout mice described in the literature provide an excellent model system .
Compare signal intensity between wild-type and RNASEH1-depleted samples to establish baseline differences in R-loop levels.
Antibody Cross-Validation:
Compare results with the S9.6 monoclonal antibody, which specifically binds DNA-RNA hybrids . Testing whether both RNASEH1 and S9.6 antibody signals diminish after RNase H1 treatment provides robust validation.
As described in one study, researchers tested "the specificity of the S9.6 antibody's ability to detect R-loops using RNaseH1," hypothesizing that "the RnaseH1 would digest the R-loop and that the S9.6 signal would be diminished" .
Immunofluorescence Controls:
Include appropriate negative controls (primary antibody omission, isotype controls).
Use cells with known R-loop phenotypes as positive controls. For example, one study showed that "shPTEN increases the nuclear intensity of the S9.6 signal" .
These validation strategies ensure that signals detected with RNASEH1 antibody accurately represent R-loops in experimental systems, reducing the risk of misinterpreting artifacts or non-specific binding.
RNASEH1 functions differently in nuclear and mitochondrial compartments, with distinct roles that reflect the specialized needs of these cellular locations:
Mitochondrial RNASEH1 Functions:
Nuclear RNASEH1 Functions:
RNASEH1 appears to be particularly critical in mitochondria, as evidenced by the severe mitochondrial dysfunction in knockout models leading to "mitochondrial dysfunction, loss of mitochondria and apoptosis" . This differential importance may guide experimental design when targeting RNASEH1 in specific cellular compartments.
Based on published research, several experimental approaches effectively demonstrate RNASEH1's essential role in antisense oligonucleotide (ASO) activity:
Genetic Manipulation Models:
Knockout Mouse Systems: Both constitutive and inducible liver-specific RNase H1 knockout mice have demonstrated that "ASOs designed to serve as substrates for RNase H1 were inactive in the hepatocytes from the RNase H1 knockout mice and in vivo, demonstrating that RNase H1 is necessary for the activity of DNA-like ASOs" .
Overexpression Studies: "Overexpression of human RNase H1 in mouse liver increased the potency of a DNA-like ASO targeting Fas after intravenous administration" , providing complementary evidence to knockout studies.
In Vitro Cellular Assays:
Dose-Response Transfection Experiments: "Potencies ranging from the sub-nM to low-nM were observed in both iControl and cControl hepatocytes transfected with the various ASOs. Conversely, no ASO activity was observed in the hepatocytes from either the cKO mice or tamoxifen-treated iKO mice" .
Target mRNA Quantification: Measuring target mRNA levels after ASO treatment in cells with varying levels of RNASEH1 expression.
In Vivo ASO Activity Assessment:
These approaches collectively provide comprehensive evidence that "RNase H1 is necessary for the activity of such ASOs" , enabling researchers to fully characterize the relationship between RNASEH1 and antisense oligonucleotide efficacy in various experimental systems.
Recent research has revealed the existence of "distinct populations of RNA:DNA hybrids that are differently sensitive to RnhA" . RNASEH1 antibodies can help distinguish between these hybrid populations through several methodological approaches:
Differential Enzymatic Sensitivity:
Treat samples with recombinant RNase H1 before immunoprecipitation or staining with S9.6 or RNASEH1 antibodies.
Recent findings showed that "RnhA had no impact on R-loops mapped by the DRIP or Cut&Tag methods" , suggesting some RNA-DNA hybrids are resistant to RNase H1 digestion.
This differential sensitivity can be exploited to categorize hybrids into RNase H1-sensitive and RNase H1-resistant populations.
Genomic Location-Specific Analysis:
Following immunoprecipitation of RNA/DNA hybrids, use PCR with region-specific primers to identify hybrids at particular genomic locations.
In mitochondrial studies, researchers found that "the signals for S7 R-loop using the CSB III PCR primer set targeting the region adjacent to the S7 RNA were higher compared to the downstream OH PCR primer set" , demonstrating how this approach can distinguish positionally distinct hybrids.
Subcellular Fractionation:
Isolate mitochondrial and nuclear fractions before immunoprecipitation to distinguish between compartment-specific hybrids.
Nuclear R-loops often form during transcription elongation, while mitochondrial R-loops like the S7 R-loop have specific functions in mtDNA replication .
Combined Antibody Approaches:
Use both RNASEH1 and S9.6 antibodies in parallel experiments to detect potentially different subsets of RNA-DNA hybrids.
The specificity of S9.6 antibody can be tested "using RNaseH1" to determine if all detected hybrids are equally sensitive to RNase H1 digestion .
These methodological approaches enable researchers to differentiate between RNA-DNA hybrid populations based on their genomic location, cellular compartment, and enzymatic sensitivity, providing deeper insights into their diverse functional roles.
Recent research has revealed critical insights into the relationship between RNASEH1, replication fork progression, and genomic stability:
RNASEH1 and Replication Fork Dynamics:
"The over-expression of RNase H1 enables the progression of replication forks under stress and safeguards genome stability" . This suggests that RNASEH1 plays a protective role during DNA replication.
Using "an innovative strategy to rapidly deliver ready-made Escherichia coli RNase H1 (RnhA) in live cells," researchers showed that "RnhA is enriched in the vicinity of active replication forks and that it rescues replication fork progression in different stress conditions" .
Unexpected R-loop Relationship:
Surprisingly, research showed that "RnhA had no impact on R-loops mapped by the DRIP or Cut&Tag methods" . This challenges the conventional model that RNASEH1 overexpression removes R-loops whose formation would hinder replication fork progression.
These findings reveal "the existence of distinct populations of RNA:DNA hybrids that are differently sensitive to RnhA and invites new interpretations of well-established observations" .
Mitochondrial Genome Stability:
In mitochondria, RNASEH1 is critical for genome stability. Studies in knockout mice showed that absence of RNASEH1 leads to "reduced mitochondrial encoded DNA and mRNA levels, suggesting impaired mitochondrial R-loop processing, transcription and mitochondrial DNA replication" .
These changes resulted in "mitochondrial dysfunction with marked changes in mitochondrial fusion, fission, morphology and transcriptional changes reflective of mitochondrial damage and stress" .
Alternative Mechanisms:
The finding that RNase H1 can support replication fork progression without resolving all R-loops suggests that:
Some RNA-DNA hybrids may actually facilitate rather than hinder replication
RNASEH1 might process only specific types of replication-blocking structures
RNASEH1 could have functions beyond R-loop resolution that promote replication fork progression
This evolving understanding of RNASEH1's role in replication and genomic stability has significant implications for research into DNA damage repair, cancer biology, and diseases associated with genome instability.
The optimal working dilutions for RNASEH1 Antibody, HRP conjugated vary by application and should be empirically determined for each specific experimental system. Based on product specifications, the following ranges serve as starting points:
Important Technical Considerations:
"It is recommended that this reagent should be titrated in each testing system to obtain optimal results" .
For Western blots, expect to detect a band at approximately 32 kDa, as this is the observed molecular weight of RNASEH1 .
For IHC applications, the search results indicate "suggested antigen retrieval with TE buffer pH 9.0; (*) Alternatively, antigen retrieval may be performed with citrate buffer pH 6.0" .
The HRP conjugation eliminates the need for secondary antibody incubation, reducing protocol time and potentially decreasing background signal.
For optimal results, perform preliminary experiments with dilution series to determine the optimal concentration for your specific experimental conditions and sample types.
To ensure reliable and interpretable results when using RNASEH1 Antibody, HRP conjugated, the following controls should be included:
Antibody Validation Controls:
Positive Control Samples: Include cell lines known to express RNASEH1, such as HeLa, A549, or HEK-293 cells .
Negative Control Samples: When possible, include RNASEH1 knockout or knockdown samples. Studies have used "liver-specific constitutive and inducible RNase H1 knockout mice" to generate negative control tissues.
Primary Antibody Omission: Include samples treated with all reagents except the primary antibody to assess background from the detection system.
Isotype Control: Include samples treated with a non-specific HRP-conjugated antibody of the same isotype (usually rabbit IgG) to assess non-specific binding.
Functional Validation Controls:
Enzymatic Digestion: Treat replicate samples with E. coli RNase H1 prior to antibody application to confirm specificity for RNA-DNA hybrids. As demonstrated in one study, samples "digested with the E. coli enzyme showed S7 R-loop levels comparable to the cControl samples" .
RNase A Treatment: Include RNase A-treated samples to distinguish between RNA-DNA hybrids and other RNA structures.
S9.6 Antibody Parallel Detection: The S9.6 antibody specifically recognizes RNA-DNA hybrids and can provide complementary validation .
Technical Controls:
Loading Controls: For Western blots, include detection of housekeeping proteins (e.g., β-actin, GAPDH) to ensure equal loading across samples.
Dilution Series: For quantitative applications, include a dilution series of the positive control sample to establish the linear detection range.
Substrate-Only Control: Include wells with HRP substrate but no antibody to assess substrate auto-oxidation.
Implementation of these controls will help validate the specificity of RNASEH1 Antibody, HRP conjugated in your experimental system and ensure the reliability of your results.
RNASEH1 antibody offers several methodological approaches to investigate mitochondrial dysfunction through its role in mitochondrial DNA maintenance and R-loop processing:
Immunohistochemistry for Tissue Analysis:
RNASEH1 antibodies can be used to examine RNASEH1 distribution in tissues with suspected mitochondrial pathology at dilutions of 1:1500-1:6000 .
Comparative analysis between healthy and affected tissues can reveal abnormal RNASEH1 expression or localization patterns associated with mitochondrial dysfunction.
Immunofluorescence for Subcellular Localization:
Use RNASEH1 antibodies at dilutions of 1:50-1:500 for immunofluorescence to visualize its subcellular distribution.
Co-staining with mitochondrial markers (e.g., TOMM20, COX IV) can assess RNASEH1's association with mitochondria in normal versus dysfunctional states.
Alterations in RNASEH1 mitochondrial localization may indicate impaired mitochondrial function.
R-loop Detection in Mitochondria:
Use RNASEH1 antibodies alongside S9.6 antibody to detect mitochondrial R-loops.
Studies have shown that RNASEH1 deficiency leads to "elevated levels of the S7 R-loop" in mitochondria.
PCR analysis of immunoprecipitated material can specifically target mitochondrial regions: "PCR primers targeting the CSB III or OH regions of the mitochondrial genome, which are complementary to or immediately downstream of the S7 RNA" .
Correlation with Mitochondrial Dynamics Markers:
Combine RNASEH1 detection with markers of mitochondrial fusion (MFN1/2, OPA1), fission (DRP1, FIS1), and mitophagy (PINK1, Parkin).
Research has shown that RNASEH1 deficiency leads to "marked changes in mitochondrial fusion, fission, morphology" .
Analysis of Mitochondrial DNA and Transcripts:
Correlate RNASEH1 levels with mtDNA copy number and mitochondrial transcript levels.
RNASEH1 knockout models showed "reduced mitochondrial encoded DNA and mRNA levels, suggesting impaired mitochondrial transcription" .
Functional Mitochondrial Assays:
Correlate RNASEH1 expression/localization with functional parameters (e.g., oxygen consumption, ATP production, membrane potential).
RNASEH1 deficiency causes "mitochondrial dysfunction" with "transcriptional changes reflective of mitochondrial damage and stress" .
These methodological approaches using RNASEH1 antibodies can provide valuable insights into the relationship between RNASEH1 dysfunction and mitochondrial pathology, potentially identifying novel therapeutic targets for mitochondrial diseases.
RNASEH1-AS1, a long non-coding RNA transcribed from the antisense strand of the RNASEH1 gene, has emerged as a significant factor in cancer research with potential diagnostic and therapeutic implications:
RNASEH1-AS1 as a Cancer Biomarker:
Impact on Tumor Immune Microenvironment:
RNASEH1-AS1 shows an inverse association "with the infiltration of most immune cell types, including plasmacytoid dendritic cells (pDC), B cells and neutrophils" .
RNASEH1 antibodies can be used in multiplex immunofluorescence studies to examine co-localization of RNASEH1 with immune cell markers in the tumor microenvironment.
Molecular Pathways and Co-expression Networks:
RNASEH1-AS1 has "a total of 1109 positively co-expressed genes (PCEGs)" in HCC that are mainly related to "RNA processing, ribosome biogenesis, transcription and histone acetylation" .
The top 10 hub genes in this network "were all highly expressed in HCC and positively correlated with histological grade" .
RNASEH1 antibodies can be used in ChIP experiments to investigate how RNASEH1 protein binding to chromatin relates to expression of these co-expressed genes.
Functional Effects in Cancer Cells:
Experimental validation revealed that "RNASEH1-AS1 was significantly elevated in HCC tissues and several cell lines, and its knockdown could suppress the proliferation, migration, and invasion of HCC cells" .
RNASEH1 antibodies can be used to monitor whether RNASEH1-AS1 modulation affects RNASEH1 protein levels or localization.
Prognostic Risk Model Development:
"A risk model was constructed based on four RNASEH1-AS1-related hub genes (EIF4A3, WDR12, DKC1, and NAT10) with good prognostic predictive potential" .
RNASEH1 antibodies could contribute to multiparameter analyses examining correlations between RNASEH1 protein and these hub genes in patient samples.
Genomic Stability in Cancer:
Given RNASEH1's role in R-loop resolution and that "the over-expression of RNase H1 enables the progression of replication forks under stress and safeguards genome stability" , RNASEH1 antibodies can help investigate how genomic instability in cancer relates to RNASEH1 function.
By utilizing RNASEH1 antibodies in these research contexts, investigators can gain deeper insights into the complex relationships between RNASEH1 protein, its antisense transcript RNASEH1-AS1, and cancer pathogenesis, potentially leading to novel diagnostic and therapeutic approaches.
Despite its utility in various applications, researchers may encounter several challenges when working with RNASEH1 Antibody, HRP conjugated. Here are common issues and their solutions:
High Background Signal:
Challenge: Non-specific background staining can mask specific RNASEH1 detection.
Solution: Increase blocking time (5% BSA or milk in TBST for 1-2 hours), optimize antibody dilution (begin with manufacturer recommendations and test higher dilutions), and include 0.1-0.3% Tween-20 in wash buffers to reduce non-specific binding .
Weak or Absent Signal:
Challenge: Failure to detect RNASEH1 despite appropriate experimental conditions.
Solution: Verify protein expression in your sample using positive control cells (HeLa, A549, or HEK-293 cells are recommended) . Ensure proper antigen retrieval for IHC (using "TE buffer pH 9.0 or citrate buffer pH 6.0" ). Consider using signal enhancement systems compatible with HRP detection.
Multiple Bands in Western Blot:
Challenge: Detection of additional bands besides the expected 32 kDa RNASEH1 band.
Solution: Optimize protein extraction protocols to minimize degradation or post-translational modifications. Use freshly prepared samples and include protease inhibitors. Compare with knockout/knockdown controls to identify which band represents specific RNASEH1 signal .
Inconsistent Results Between Experiments:
Challenge: Variability in signal intensity or patterns between experimental replicates.
Solution: Standardize all experimental conditions, including sample preparation, antibody dilution, incubation times, and detection parameters. Create detailed protocols with specific timing. Consider preparing larger batches of working dilutions to use across multiple experiments.
Cross-Reactivity Issues:
Challenge: Potential cross-reactivity with related proteins (e.g., RNase H2).
Solution: Validate specificity using genetic models (RNASEH1 knockout cells or tissues as described in the literature ). Test antibody reaction with recombinant RNASEH1 and related proteins to confirm specificity.
HRP Activity Loss:
Challenge: Reduced enzymatic activity of the HRP conjugate over time.
Solution: Store antibody according to manufacturer recommendations (-20°C with 50% glycerol) . Avoid repeated freeze-thaw cycles by preparing appropriate aliquots. Check expiration date and consider replacing antibody if activity is significantly reduced despite proper storage.
Excessive Signal Development Speed:
Challenge: Overly rapid signal development in HRP substrate causing oversaturation.
Solution: Dilute antibody further, reduce substrate incubation time, or consider using substrate with slower kinetics. For Western blots, capture images at multiple exposure times to find optimal signal-to-background ratio.
By applying these troubleshooting strategies, researchers can optimize their use of RNASEH1 Antibody, HRP conjugated and obtain reliable, reproducible results across various experimental applications.
RNASEH1 antibody provides valuable tools to investigate the complex relationship between R-loops and replication stress through several methodological approaches:
Proximity-Based Analysis at Replication Forks:
Recent research revealed that "RnhA is enriched in the vicinity of active replication forks and that it rescues replication fork progression in different stress conditions" .
RNASEH1 antibodies can be used in proximity ligation assays (PLA) or ChIP experiments to detect RNASEH1 association with replication machinery components (e.g., PCNA, MCM proteins) at sites of replication stress.
Replication Stress Induction Experiments:
Treat cells with replication stress-inducing agents (e.g., hydroxyurea, aphidicolin, camptothecin).
Use RNASEH1 antibodies to track changes in RNASEH1 localization and abundance at different time points after stress induction.
Correlate these changes with markers of replication stress (γH2AX, RPA phosphorylation) and R-loop formation (S9.6 antibody staining).
Cell Cycle-Specific Analysis:
Synchronize cells at different cell cycle phases or use flow cytometry to separate populations.
Employ RNASEH1 antibodies in immunofluorescence or Western blot analysis to determine how RNASEH1 levels and localization change throughout the cell cycle, particularly during S phase.
Correlate with R-loop dynamics across the cell cycle using S9.6 antibody staining.
Chromatin Fractionation Studies:
Perform biochemical fractionation to separate chromatin-bound proteins from soluble nuclear fractions.
Use RNASEH1 antibodies to analyze how replication stress affects RNASEH1 recruitment to chromatin.
This approach can reveal whether RNASEH1 is actively recruited to chromatin under stress conditions to resolve R-loops.
Distinguishing Different Types of RNA-DNA Hybrids:
Recent findings showed that "RnhA had no impact on R-loops mapped by the DRIP or Cut&Tag methods" , suggesting "the existence of distinct populations of RNA:DNA hybrids that are differently sensitive to RnhA" .
RNASEH1 antibodies can be used alongside different R-loop detection methods (DRIP, S9.6 immunostaining, etc.) to distinguish between these hybrid populations and their relationship to replication stress.
Functional Rescue Experiments:
In cells experiencing replication stress, test whether overexpression of RNASEH1 rescues replication fork progression.
Use RNASEH1 antibodies to confirm increased protein levels in overexpression experiments.
This approach can help determine if R-loop resolution is sufficient to overcome replication stress in your experimental system.
These methodological approaches using RNASEH1 antibodies can provide significant insights into how R-loops contribute to replication stress and how cells resolve these structures to maintain genomic integrity during DNA replication.
Recent findings have opened several promising research directions regarding RNASEH1 function, presenting opportunities for researchers to make significant contributions to this evolving field:
Distinct RNA-DNA Hybrid Populations:
A groundbreaking study revealed "the existence of distinct populations of RNA:DNA hybrids that are differently sensitive to RnhA" . This challenges conventional models of R-loop biology.
Research opportunity: Characterize these distinct hybrid populations using comparative approaches (DRIP vs. Cut&Tag vs. S9.6 immunostaining) and correlate with genetic requirements for their formation and resolution.
Methodological approach: Apply RNASEH1 antibodies in combination with different hybrid detection methods across various cellular contexts.
Replication Fork Progression Mechanisms:
Research showed that "RnhA rescues replication fork progression in different stress conditions" but surprisingly "had no impact on R-loops mapped by the DRIP or Cut&Tag methods" .
Research opportunity: Identify the specific mechanisms by which RNASEH1 promotes replication fork progression independent of bulk R-loop resolution.
Methodological approach: Use RNASEH1 antibodies in proximity-based proteomics to identify novel interaction partners specifically at replication forks.
RNASEH1-AS1 in Cancer Biology:
The lncRNA RNASEH1-AS1 has been identified as having "good diagnostic and prognostic values for HCC" and shows inverse association "with the infiltration of most immune cell types" .
Research opportunity: Investigate whether RNASEH1-AS1 directly regulates RNASEH1 protein levels or function, and how this relationship impacts cancer progression.
Methodological approach: Combine RNASEH1 antibody detection with RNASEH1-AS1 modulation in cancer models.
Mitochondrial R-loop Regulation:
RNASEH1 has been shown to be "essential for mitochondrial R-loop processing, transcription and mitochondrial DNA replication" .
Research opportunity: Characterize the specific mitochondrial R-loop structures regulated by RNASEH1 and their roles in mitochondrial genome maintenance.
Methodological approach: Apply RNASEH1 antibodies in mitochondrial ChIP-seq and R-loop mapping experiments.
Therapeutic Antisense Oligonucleotide Development: