POLRMT is a 139 kDa enzyme encoded by the POLRMT gene (NCBI Gene ID: 5442) and localized to mitochondria . It transcribes mitochondrial DNA (mtDNA) to produce RNA primers for mtDNA replication and mitochondrial transcripts essential for oxidative phosphorylation . Structurally, POLRMT comprises four domains: the N-terminal extension, pentatricopeptide repeat (PPR), N-terminal, and C-terminal domains . Dysregulation of POLRMT is linked to cancers, including lung, breast, and osteosarcoma, due to its role in sustaining mitochondrial metabolism .
POLRMT antibodies are widely used in molecular biology and cancer research. Below are validated applications and protocols:
Additional Applications: ELISA, CUT&RUN, and immunocytochemistry (ICC) .
Non-Small Cell Lung Cancer (NSCLC):
Osteosarcoma (OS):
Skin Squamous Cell Carcinoma (SCC):
POLRMT mutations impair mitochondrial transcription, leading to neurological defects and classical mitochondrial disease phenotypes .
| Parameter | Details |
|---|---|
| Host/Isotype | Rabbit/IgG |
| Reactivity | Human (tested), Mouse (predicted) |
| Immunogen | POLRMT fusion protein (AA 1–1230) |
| Observed MW | 130–139 kDa |
| Storage | -20°C in PBS with 0.02% sodium azide |
| Parameter | Details |
|---|---|
| Target Region | Center (AA 42–230) |
| Applications | WB, IF, ICC, IHC (paraffin), CUT&RUN |
| Clonality | Polyclonal |
| Purification | Antigen-affinity chromatography |
IMT1 (POLRMT Inhibitor): Blocks mitochondrial transcription, reduces ATP production, and suppresses tumor growth in colorectal cancer (CRC) models .
2-C-Methyladenosine: Inhibits POLRMT activity, inducing apoptosis in acute myeloid leukemia (AML) cells .
POLRMT is a promising therapeutic target due to its oncogenic role in multiple cancers. Inhibitors like IMT1 and genetic silencing strategies (shRNA, CRISPR/Cas9) show robust anti-tumor efficacy in preclinical models .
POLRMT is the mitochondrial RNA polymerase responsible for transcribing the mitochondrial genome, which encodes essential components of the oxidative phosphorylation process. It functions as part of a transcription initiation complex that includes mitochondrial transcription factor A (TFAM) and mitochondrial transcription factor B2 (TFB2M) . Beyond its primary role in transcription, POLRMT also possesses DNA primase activity, catalyzing the synthesis of short RNA primers necessary for initiating lagging-strand DNA synthesis from the origin of light-strand DNA replication (OriL) . The importance of POLRMT in research has grown significantly due to its emerging role in various cancers, with studies showing that POLRMT is overexpressed in multiple cancer types including osteosarcoma, skin squamous cell carcinoma, and prostate cancer . This overexpression appears to promote cancer cell growth, proliferation, and invasion, suggesting POLRMT as a potential therapeutic target.
POLRMT antibodies have been validated for multiple research applications that facilitate the study of this protein in various experimental contexts. According to available data, commercial POLRMT antibodies (such as the rabbit polyclonal antibody ab228576) have been validated for Western blotting (WB), which allows for the detection and semi-quantification of POLRMT in protein lysates . Additionally, these antibodies are suitable for immunohistochemistry on paraffin-embedded tissues (IHC-P), enabling the visualization of POLRMT expression patterns in tissue sections while preserving morphological context . For cellular localization studies, POLRMT antibodies can be used in immunocytochemistry/immunofluorescence (ICC/IF) applications, providing insights into the subcellular distribution of the protein . While most commercially available POLRMT antibodies have been validated with human samples, researchers should carefully examine cross-reactivity information when working with other species, as reactivity may vary depending on sequence homology and antibody characteristics.
Proper validation of POLRMT antibodies is essential for generating reliable research data. A comprehensive validation approach should include multiple complementary techniques. First, researchers should perform Western blotting using positive control samples known to express POLRMT (such as cancer cell lines with verified POLRMT expression) alongside negative controls where POLRMT expression has been suppressed through shRNA knockdown or CRISPR/Cas9 knockout approaches . The expected molecular weight of human POLRMT is approximately 140 kDa, and validation should confirm a single specific band at this size. Additionally, immunocytochemistry should show the expected mitochondrial localization pattern, which can be confirmed by co-staining with established mitochondrial markers. For more rigorous validation, researchers can compare antibody specificity across different lots and compare results from different antibodies targeting distinct epitopes of POLRMT. Expression patterns detected by the antibody should also be consistent with mRNA expression data from qRT-PCR analyses, as demonstrated in multiple studies where POLRMT protein levels correlated with mRNA expression levels .
Several technical challenges may arise when using POLRMT antibodies for Western blotting. Given POLRMT's relatively high molecular weight (approximately 140 kDa), researchers often encounter difficulties with efficient protein transfer from gel to membrane. To address this issue, it is advisable to use a lower percentage gel (6-8%) and extend the transfer time or employ a wet transfer system rather than semi-dry transfer. Additionally, due to the predominantly mitochondrial localization of POLRMT, standard whole-cell lysis buffers may not optimally extract the protein. Using specialized mitochondrial extraction protocols or lysis buffers containing higher detergent concentrations can improve yields. Non-specific binding can also pose challenges, particularly with polyclonal antibodies. This can be minimized by optimizing blocking conditions (typically 5% non-fat milk or BSA) and including additional washing steps. When troubleshooting weak signals, researchers should consider that POLRMT expression levels vary significantly across different cell types and tissues, with cancer cells often showing higher expression than normal cells . Therefore, loading sufficient protein (typically 30-50 μg of total protein) and using enhanced chemiluminescence detection systems may be necessary for optimal results.
Investigating the mitochondrial transcription initiation complex requires sophisticated methodologies that leverage POLRMT antibodies in conjunction with other techniques. Co-immunoprecipitation (Co-IP) experiments using POLRMT antibodies can isolate the native transcription initiation complex, allowing for the identification of interacting proteins such as TFAM and TFB2M . For these experiments, researchers should use cell lysis conditions that preserve protein-protein interactions (typically containing low concentrations of non-ionic detergents like NP-40 or Triton X-100). When coupled with mass spectrometry, this approach can identify novel interaction partners beyond the known components. Chromatin immunoprecipitation (ChIP) assays using POLRMT antibodies are valuable for examining POLRMT binding to mitochondrial DNA promoters in vivo. This technique requires careful optimization of crosslinking conditions, as the mitochondrial genome has unique properties compared to nuclear DNA. Recent structural studies have revealed specific interactions between POLRMT and the light-strand promoter (LSP), particularly at positions -4 to +2 of the template strand . To investigate these specific interactions, researchers can combine POLRMT antibodies with proximity ligation assays (PLA) or use them in super-resolution microscopy techniques like STORM or PALM to visualize the spatial organization of the transcription complex within mitochondria with nanometer precision.
Contradictory findings regarding POLRMT expression across different studies can stem from multiple methodological factors that researchers should systematically address. First, researchers should employ multiple POLRMT antibodies targeting different epitopes to verify consistency in expression patterns, as antibody specificity variations can lead to discrepant results. When comparing POLRMT levels between studies, it is crucial to normalize expression data to appropriate reference genes or proteins that remain stable under the experimental conditions being tested. Mitochondrial content varies substantially between different cell types and tissues, which can confound POLRMT expression analyses if not properly normalized to mitochondrial markers such as TOMM20 or mitochondrial DNA copy number . Technical variations in sample preparation, particularly in how mitochondrial fractions are isolated, can significantly impact detected POLRMT levels. For tissues and tumors with heterogeneous cell populations, laser capture microdissection can isolate specific cell types before analysis to prevent dilution effects from non-target cells. When reconciling contradictory findings between mRNA and protein expression, researchers should consider post-transcriptional regulation mechanisms, as several studies have demonstrated cases where POLRMT protein levels do not directly correlate with mRNA abundance, suggesting complex regulatory processes beyond transcriptional control .
Optimizing POLRMT antibodies for immunohistochemistry (IHC) in cancer research requires addressing several technical considerations specific to this application. Antigen retrieval methods significantly impact POLRMT detection in formalin-fixed, paraffin-embedded (FFPE) tissues, with heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) showing optimal results for most POLRMT antibodies. Researchers should empirically determine the optimal retrieval conditions for each antibody and tissue type. The mitochondrial localization of POLRMT presents unique challenges, as mitochondrial density varies across cancer types and cellular states. To address this variability, researchers should include co-staining with mitochondrial markers to normalize POLRMT signals to mitochondrial content. For quantitative analyses, automated image analysis algorithms can be employed to measure staining intensity relative to mitochondrial markers, enabling more objective comparisons across samples. In cancer tissues with heterogeneous POLRMT expression, multiplexed immunofluorescence approaches allow simultaneous detection of POLRMT alongside cancer markers, enabling cell type-specific analysis. Studies have demonstrated that POLRMT expression is significantly upregulated in skin squamous cell carcinoma and osteosarcoma compared to adjacent normal tissues , making standardized scoring systems essential for consistent reporting across research groups.
Sophisticated methodologies combining antibody-based detection with functional assays provide powerful insights into POLRMT biology. Proximity-dependent biotin identification (BioID) or APEX2 proximity labeling, when coupled with POLRMT antibodies for verification, can map the proximal protein environment of POLRMT in living cells, revealing transient interaction partners that might be missed by conventional co-immunoprecipitation approaches. Researchers can employ Förster resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) using fluorescently tagged antibody fragments to visualize POLRMT interactions with components like TFAM and TFB2M in real-time within living cells. For investigating POLRMT's catalytic activity, researchers can combine immunodepletion using POLRMT antibodies with in vitro transcription assays using mitochondrial promoter templates, allowing assessment of how specific factors influence POLRMT function. Recent structural studies have identified critical domains in POLRMT, including the pentatricopeptide repeat domain (residues 218-368) and the intercalating hairpin (residues 605-624), which interact with specific regions of mitochondrial DNA promoters . Domain-specific POLRMT antibodies can be used to block these interactions selectively, providing insights into domain-specific functions without the need for genetic manipulation of the protein.
Designing robust POLRMT knockdown or knockout experiments requires careful planning and validation to ensure specificity and reliability of results. For shRNA-mediated knockdown, researchers should design multiple shRNA constructs targeting different regions of the POLRMT transcript and validate them individually. As demonstrated in osteosarcoma studies, effective shRNAs can achieve greater than 95% knockdown of POLRMT mRNA, with corresponding reductions in protein levels . For CRISPR/Cas9-mediated knockout, at least two different guide RNA sequences should be designed to minimize off-target effects. The knockout efficiency should be verified at both the mRNA level using qRT-PCR and at the protein level using Western blotting with validated POLRMT antibodies . Since complete POLRMT knockout may be lethal in some cell types due to its essential role in mitochondrial function, inducible systems like Tet-On/Off or conditional Cre-loxP approaches offer better control over the timing and extent of gene silencing. All genetic manipulation experiments require appropriate controls, including scrambled shRNA or non-targeting CRISPR guides. Functional validation of POLRMT knockdown/knockout should assess mitochondrial DNA content, mitochondrial transcript levels, and expression of respiratory chain complex components, as these parameters directly reflect POLRMT activity . Additionally, assessment of cellular phenotypes such as proliferation (EdU incorporation), migration (Transwell assays), and viability (CCK-8 assays) provides comprehensive characterization of the biological consequences of POLRMT depletion.
Accurate quantification of POLRMT levels requires tailored methodologies depending on the experimental context and research questions. For absolute quantification of POLRMT protein, quantitative Western blotting using purified recombinant POLRMT protein as a standard curve reference provides the most precise measurements. This approach requires careful validation of antibody specificity and linear detection range. When analyzing POLRMT expression across different tissues or cell types, normalization to both total protein load (using stain-free technology or housekeeping proteins) and mitochondrial content markers (such as VDAC or TOMM20) is essential to account for variations in mitochondrial abundance . For high-throughput screening applications, ELISA-based methods using validated POLRMT antibodies offer greater sample processing capacity, though these typically require pair-matched antibodies recognizing different epitopes. In tissue sections, quantitative immunohistochemistry with automated image analysis software can measure POLRMT expression while preserving spatial context. This approach benefits from machine learning algorithms to distinguish cell types and subcellular compartments. For single-cell analyses, flow cytometry or mass cytometry (CyTOF) with metal-conjugated POLRMT antibodies enables quantification of POLRMT levels in thousands of individual cells, revealing population heterogeneity that might be masked in bulk analyses. These approaches are particularly valuable for heterogeneous samples like tumors, where POLRMT expression may vary substantially between different cell populations.
Investigating interactions between POLRMT and other components of the mitochondrial transcription machinery requires specialized techniques that preserve and detect protein complexes. Co-immunoprecipitation (Co-IP) using POLRMT antibodies followed by Western blotting for TFAM and TFB2M provides direct evidence of physical association between these proteins . For more sensitive detection of native complexes, Blue Native PAGE combined with Western blotting can preserve and resolve intact mitochondrial transcription complexes. Mutagenesis studies have identified critical interaction domains, such as the tether helix (residues 122-145) of POLRMT that interacts with TFAM, and the lever loop (residues 588-604) that interacts with TFB2M . To study these domain-specific interactions, researchers can generate domain deletion or point mutation constructs and assess complex formation using co-immunoprecipitation with POLRMT antibodies. Structural studies using cryo-electron microscopy have revealed that the transcription initiation complex involves DNA bending by approximately 55° between upstream and downstream DNA duplexes, stabilized by interactions with both POLRMT and TFB2M . To investigate these structural dynamics in cells, Förster resonance energy transfer (FRET) between fluorescently labeled proteins can detect conformational changes during complex assembly and activity. For mapping the complete interactome of POLRMT beyond the known components, proximity-dependent biotinylation approaches like BioID or APEX2, followed by streptavidin pulldown and mass spectrometry, can identify proteins that transiently interact with POLRMT during different phases of transcription initiation and elongation.
Using POLRMT antibodies in cancer biomarker studies requires rigorous standardization and validation approaches that account for the complexities of clinical samples. Tissue microarray (TMA) analysis with standardized immunohistochemistry protocols allows efficient screening of POLRMT expression across large patient cohorts while minimizing batch effects. When developing POLRMT as a potential diagnostic biomarker, researchers should establish clear scoring criteria based on staining intensity and percentage of positive cells, ideally using automated image analysis software to minimize subjective interpretation. Studies have shown significant POLRMT upregulation in skin squamous cell carcinoma, osteosarcoma, and prostate cancer tissues compared to matched normal tissues , suggesting diagnostic potential. For prognostic applications, POLRMT expression data should be correlated with clinicopathological parameters and survival outcomes using appropriate statistical methods like Kaplan-Meier analysis and multivariate Cox regression models. Multiplexed immunofluorescence or mass cytometry approaches allow simultaneous detection of POLRMT alongside established cancer markers and immune cell markers, providing context for interpretation within the tumor microenvironment. When developing liquid biopsy applications, researchers can explore detection of POLRMT in circulating tumor cells or extracellular vesicles using highly sensitive immunoassays. For all biomarker applications, validation across multiple independent cohorts with diverse patient populations is essential before clinical implementation.
Non-specific binding is a common challenge when working with POLRMT antibodies that can compromise experimental results if not properly addressed. Several strategic approaches can minimize this issue across different applications. For Western blotting, optimization of blocking conditions is essential, with 5% BSA in TBST often providing better results than milk-based blockers, particularly for phospho-specific antibodies. Additionally, increasing the concentration of detergent (0.1-0.3% Tween-20) in wash buffers and extending wash times can substantially reduce background. For immunoprecipitation experiments, pre-clearing lysates with protein A/G beads before adding the POLRMT antibody removes proteins that might non-specifically bind to the beads. When performing immunohistochemistry or immunofluorescence, including an additional blocking step with 10% serum from the same species as the secondary antibody source can dramatically reduce non-specific signals. For POLRMT antibodies with persistent non-specific binding, epitope-tagged POLRMT expression systems (where commercially available antibodies against the tag can be used) provide an alternative approach with potentially improved specificity. When analyzing results, researchers should always include appropriate controls, such as POLRMT knockdown or knockout samples, to definitively identify specific bands or signals . Additionally, comparing results from multiple POLRMT antibodies targeting different epitopes can help distinguish true POLRMT signals from non-specific artifacts.
Detecting low POLRMT expression presents technical challenges that require specialized approaches to enhance sensitivity without compromising specificity. For Western blotting, signal amplification systems such as enhanced chemiluminescence (ECL) Plus or Super Signal West Femto can significantly improve detection limits compared to standard ECL reagents. Additionally, concentrating protein samples through immunoprecipitation before Western blotting can enrich for POLRMT, making low expression levels more detectable. When analyzing tissues with low POLRMT expression by immunohistochemistry, tyramide signal amplification (TSA) systems can amplify signals by up to 100-fold while maintaining spatial resolution. This approach has proven particularly useful for detecting low-abundance mitochondrial proteins in tissue sections. For mRNA detection, digital droplet PCR offers superior sensitivity compared to conventional qRT-PCR for quantifying low-abundance POLRMT transcripts, with the ability to detect single-molecule differences. When analyzing cell populations with heterogeneous POLRMT expression, single-cell techniques like flow cytometry with highly sensitive fluorophores or mass cytometry (CyTOF) can identify rare cell populations with altered POLRMT expression that might be masked in bulk analyses. When interpreting results from samples with low POLRMT expression, researchers should carefully establish detection thresholds based on validated positive and negative controls to distinguish true positive signals from background noise .
Accurately visualizing the mitochondrial localization of POLRMT requires specialized techniques that account for the complex and dynamic nature of mitochondria across different cell types. Super-resolution microscopy techniques such as Structured Illumination Microscopy (SIM), Stimulated Emission Depletion (STED), or Photoactivated Localization Microscopy (PALM) overcome the diffraction limit of conventional microscopy, allowing visualization of POLRMT within the intricate mitochondrial ultrastructure. These approaches can resolve individual nucleoids within mitochondria where POLRMT functions. For live-cell imaging of POLRMT dynamics, researchers can use cell lines stably expressing fluorescently tagged POLRMT constructs, though careful validation is needed to ensure that the tag doesn't interfere with localization or function. When performing immunofluorescence with POLRMT antibodies, co-staining with multiple mitochondrial markers targeting different submitochondrial compartments (such as TOMM20 for outer membrane, cytochrome c for intermembrane space, and mtSSB for nucleoids) provides context for POLRMT localization within the organelle. For tissue sections, particularly from tissues with high autofluorescence, spectral unmixing approaches or use of far-red fluorophores can improve signal-to-noise ratios. Electron microscopy immunogold labeling with POLRMT antibodies offers the highest resolution for localization studies, capable of pinpointing POLRMT at the nucleoid level, though this technique requires specialized sample preparation and highly specific antibodies suitable for post-embedding immunogold labeling.
Distinguishing between specific POLRMT isoforms or post-translational modifications requires sophisticated methodological approaches that provide high resolution at the protein level. Two-dimensional gel electrophoresis followed by Western blotting with POLRMT antibodies can separate protein variants based on both molecular weight and isoelectric point, revealing potential post-translational modifications that alter charge. For more comprehensive characterization, immunoprecipitation of POLRMT followed by mass spectrometry analysis can identify specific post-translational modifications such as phosphorylation, acetylation, or ubiquitination, along with their exact locations within the protein sequence. When specific modifications have been identified, researchers can generate or source modification-specific antibodies that selectively recognize POLRMT with particular post-translational modifications. These specialized antibodies can then be used in standard applications like Western blotting or immunofluorescence to study the abundance and localization of modified POLRMT forms. For distinguishing between splice variants, RT-PCR with primers spanning potential splice junctions, followed by sequencing, can identify which isoforms are expressed in particular tissues or cell types. Additionally, isoform-specific antibodies targeting unique epitopes present in specific splice variants provide a direct approach for protein-level detection. When studying how modifications affect POLRMT function, researchers can combine immunoprecipitation using modification-specific antibodies with in vitro transcription assays to assess how particular modifications impact enzymatic activity.