QTRT1 Human

Queuine TRNA-Ribosyltransferase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

  • Gene: QTRT1 is located on chromosome 19p13.2, spanning ~12 kb with 10 exons .

  • Protein: The recombinant QTRT1 protein is a 428-amino-acid polypeptide (molecular mass: 46.7 kDa) produced in E. coli. It includes a 25-amino-acid N-terminal His-tag for purification .

  • Catalytic Activity: QTRT1 forms a heterodimer with QTRT2 (accessory subunit) to function as tRNA-guanine transglycosylase (TGT). The enzyme requires Zn²⁺ for catalysis and operates via a double-displacement mechanism .

PropertyDetails
Molecular Weight46.7 kDa (recombinant)
Subunit CompositionCatalytic subunit (QTRT1) + non-catalytic subunit (QTRT2)
Metal DependencyZn²⁺-dependent enzymatic activity
Substrate SpecificitytRNAs with GU(N) anticodons (Asp, Asn, His, Tyr)

tRNA Modification and Translational Regulation

QTRT1’s primary role is modifying tRNAs to optimize codon-anticodon interactions. The queuosine modification enhances:

  • Translation Accuracy: Reduces mistranslation at wobble positions .

  • Cellular Stress Response: Modulates protein synthesis under stress conditions .

Interactions and Pathways

  • Protein Interactions: QTRT1 interacts with QTRT2 (0.999 confidence score) to form active TGT .

  • Pathway Involvement: Linked to epithelial-mesenchymal transition (EMT), axoneme assembly, and androgen response via GSEA/GO analyses .

Lung Adenocarcinoma (LUAD)

DatasetSample SizeSurvival OutcomeP-value
GSE72094 (OS)398High QTRT1 → Poor OS0.0033
TCGA (DFS)502High QTRT1 → Shorter DFS0.0036
TCGA (OS)502High QTRT1 → Shorter OS0.0022

Breast Cancer

  • QTRT1 Knockout: Suppresses cell proliferation, migration, and tumor growth. Alters tight junctions and microbiome composition in vivo .

Metabolic Regulation

  • Lipid Metabolism: Hepatic QTRT1 deficiency reduces de novo lipogenesis (DNL) and mitigates atherosclerosis in mice .

  • Mechanism: QTRT1 regulates odorant-binding protein 2A (OBP2A), a modulator of lipid synthesis .

Therapeutic Targeting

  • Cancer Therapy: Inhibiting QTRT1 may suppress tumor growth and metastasis, particularly in breast cancer .

  • Atherosclerosis: Hepatic QTRT1 inhibition reduces hyperlipidemia and plaque burden in mice .

Product Specs

Introduction
Queuine TRNA-Ribosyltransferase 1, also called QTRT1, belongs to the queuine tRNA-ribosyltransferase family. This enzyme interacts with QTRTD1 to form a functional queuine tRNA-ribosyltransferase complex. QTRT1 is responsible for replacing guanine with queuine at the wobble position of tRNAs containing GUN anticodons (specifically, tRNA-Asp, tRNA-Asn, tRNA-His, and tRNA-Tyr). This process results in the formation of the hypermodified nucleoside queuosine (Q), which has the chemical structure 7-(((4,5-cis-dihydroxy-2-cyclopenten-1-yl)amino)methyl)-7-deazaguanosine.
Description
Recombinant human QTRT1, expressed in E. coli, is a single, non-glycosylated polypeptide chain. This protein comprises 428 amino acids (specifically, amino acids 1 through 403), resulting in a molecular weight of 46.7 kDa. The recombinant QTRT1 protein is engineered with a 25 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
The product is a clear, sterile-filtered solution.
Formulation
The QTRT1 protein is supplied in a solution with a concentration of 0.25 mg/ml. The solution contains phosphate buffered saline (pH 7.4), 30% glycerol, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), the product should be stored at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To ensure stability during long-term storage, the addition of a carrier protein such as HSA or BSA (0.1%) is recommended. It is important to avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity
The purity of the QTRT1 protein is determined by SDS-PAGE analysis and is guaranteed to be greater than 80.0%.
Synonyms
Queuine TRNA-Ribosyltransferase 1, TRNA-Guanine Transglycosylase, TGT, Guanine Insertion Enzyme, EC 2.4.2.29, TGUT, Queuine TRNA-Ribosyltransferase, TGT, Catalytic Subunit, TGT, 43-KD Subunit, FP3235, Queuine tRNA-ribosyltransferase.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEFMAGAA TQASLESAPR IMRLVAECSR SRARAGELWL PHGTVATPVF MPVGTQATMK GITTEQLDAL GCRICLGNTY HLGLRPGPEL IQKANGLHGF MNWPHNLLTD SGGFQMVSLV SLSEVTEEGV RFRSPYDGNE TLLSPEKSVQ IQNALGSDII MQLDDVVSST VTGPRVEEAM YRSIRWLDRC IAAHQRPDKQ NLFAIIQGGL DADLRATCLE EMTKRDVPGF AIGGLSGGES KSQFWRMVAL STSRLPKDKP RYLMGVGYAT DLVVCVALGC DMFDCVFPTR TARFGSALVP TGNLQLRKKV FEKDFGPIDP ECTCPTCQKH SRAFLHALLH SDNTAALHHL TVHNIAYQLQ LMSAVRTSIV EKRFPDFVRD FMGAMYGDPT LCPTWATDAL ASVGITLG.

Q&A

What is QTRT1 and what is its primary function in human cells?

QTRT1, also known as tRNA-guanine transglycosylase (TGT), is the catalytic subunit of the tRNA-guanine transglycosylase complex located on chromosome 19p13.2. It contains 10 exons spanning approximately 12 kb . The primary molecular function of QTRT1 is catalyzing the exchange of guanine (G) with queuine (Q) at position 34 of specific tRNAs, resulting in hypermodified transfer RNAs . This post-transcriptional modification occurs specifically in tRNAs that code for asparagine, aspartic acid, histidine, and tyrosine .

Unlike bacterial TGT enzymes which function as homodimers, the active eukaryotic TGT enzyme is a heterodimer consisting of a catalytic QTRT1 subunit and a non-catalytic QTRT2 subunit . QTRT1 is primarily located in the mitochondrion and plays a critical role in tRNA modification by synthesizing the 7-deazaguanosine queuosine .

How does QTRT1 differ structurally and functionally between humans and bacteria?

The structural and functional differences between human and bacterial QTRT1 are significant:

  • Quaternary structure: The human TGT is a heterodimer consisting of the catalytic QTRT1 subunit and the non-catalytic QTRT2 subunit, whereas bacterial TGT functions as a homodimer .

  • Substrate specificity: While both human and bacterial enzymes modify tRNAs at position 34, bacterial TGT incorporates a queuine precursor (preQ1), whereas human QTRT1 directly incorporates queuine .

  • Reaction mechanism: The bacterial TGT enzyme forms a covalent tRNA-enzyme reaction intermediate through nucleophilic attack by an aspartate residue (Asp280), followed by replacement of guanine with preQ1 . The human QTRT1 likely operates through a similar mechanism but with different substrate specificities.

  • Clinical relevance: Bacterial TGT has been identified as a virulence factor in pathogens like Shigella, where its absence reduces pathogenicity . Human QTRT1, conversely, has been implicated in cancer progression .

What experimental methods are most effective for measuring QTRT1 expression levels?

Several experimental approaches have proven effective for measuring QTRT1 expression:

  • Transcriptional level analysis:

    • RT-qPCR for targeted gene expression analysis

    • RNA-seq for genome-wide expression profiling, as used in the TCGA datasets referenced in studies

    • Microarray analysis, as employed in GEO datasets (GSE10072, GSE49996, GSE63384, GSE72094)

  • Protein level detection:

    • Western blotting, which has successfully shown that QTRT1 expression in mitochondria of human LUAD A549 cells is higher than in normal human bronchial epithelial 16HBE cells

    • Immunohistochemistry (IHC), which has demonstrated significantly higher positive expression of QTRT1 in LUAD compared to normal lung tissues

  • Methylation analysis:

    • Methylation arrays for analyzing QTRT1 methylation status, as employed in the GSE49996 and GSE63384 datasets

When selecting a method, researchers should consider their specific research question, available sample types, and whether they need to examine expression at the RNA or protein level.

What is the relationship between QTRT1 expression and clinical outcomes in lung adenocarcinoma?

Studies have revealed significant correlations between QTRT1 expression and clinical outcomes in lung adenocarcinoma (LUAD):

This data strongly suggests that QTRT1 can serve as a novel biomarker for the prognosis of LUAD patients.

How does QTRT1 interact with known oncogenic pathways in cancer development?

The interaction of QTRT1 with oncogenic pathways appears to be complex and multifaceted:

  • Relationship with established oncogenes: Studies have shown that QTRT1, along with EGFR and KRAS mutations, significantly impacts survival outcomes in LUAD. Multivariate analysis revealed:

    • QTRT1 (HR=1.37, P=0.020), EGFR (HR=1.32, P=0.034), and KRAS (HR=1.37, P=0.016) were all significantly related to DFS in LUAD patients

    • QTRT1 (HR=1.27, P=0.044) and KRAS (HR=1.38, P=0.006) were closely associated with OS

  • Biological pathways: Gene Set Enrichment Analysis (GSEA) and Gene Ontology (GO) term enrichment analysis indicated that QTRT1 expression is significantly correlated with:

    • Axoneme assembly

    • Androgen response

    • Epithelial-mesenchymal transition (EMT)

  • tRNA modification: As QTRT1 is involved in tRNA modification, its dysregulation may impact protein synthesis and cell signaling networks that contribute to cancer progression. Specific tRNAs have been related to human breast cancer, colon adenocarcinoma, and human lung cancer .

What are the challenges in interpreting conflicting data on QTRT1 expression across different cancer types?

Researchers face several challenges when interpreting QTRT1 expression data across different cancer types:

  • Tissue-specific effects: QTRT1 may exhibit different roles across tissue types. While it shows increased expression in LUAD , its expression patterns and effects may differ in other cancers, requiring tissue-specific analyses.

  • Methodological differences: Various studies employ different methodologies (RNA-seq, microarray, IHC, western blot), making direct comparisons challenging. Standardization of techniques and analysis pipelines is crucial for consistent interpretation.

  • Confounding factors: Clinical variables like age, gender, smoking status, and comorbidities may influence QTRT1 expression and function. The relationship between QTRT1 expression and clinical features showed no significant correlation with age (P=0.342), gender (P=0.756), or cancer stage (P=0.509) , but these factors should still be considered in analysis.

  • Causality vs. correlation: Determining whether altered QTRT1 expression is a cause or consequence of cancer development remains difficult. Functional studies with knockdown or overexpression models are needed to establish causative relationships.

  • Integration of multi-omics data: Combining expression, methylation, mutation, and clinical data presents analytical challenges. Researchers should employ integrated bioinformatics approaches to better understand the complex role of QTRT1 in cancer.

What are the optimal methods for studying QTRT1 enzymatic activity in vitro?

To effectively study QTRT1 enzymatic activity in vitro, researchers should consider these methodologies:

  • Recombinant protein production:

    • Express recombinant human QTRT1 in appropriate expression systems (E. coli, insect cells, or mammalian cells)

    • Co-express QTRT1 with QTRT2 to form the functional heterodimeric complex

    • Purify using affinity chromatography followed by size exclusion chromatography

  • Enzyme activity assays:

    • Radioisotope-based assays using [³H]-guanine or [³H]-queuine to measure incorporation into tRNA substrates

    • HPLC or LC-MS based methods to detect modified nucleosides in tRNA

    • In vitro transcription to generate specific tRNA substrates (tRNAAsp, tRNAAsn, tRNAHis, and tRNATyr)

  • Structural studies:

    • X-ray crystallography to determine QTRT1 structure alone or in complex with substrates

    • Cryo-EM for studying the QTRT1-QTRT2 heterodimer complex

    • Molecular dynamics simulations to understand conformational changes during catalysis

  • Binding assays:

    • Surface plasmon resonance (SPR) to measure binding kinetics between QTRT1 and tRNA or queuine

    • Isothermal titration calorimetry (ITC) to determine thermodynamic parameters of binding

    • Fluorescence-based assays to monitor conformational changes upon substrate binding

The crystal structure of human QTRT1 has been reported , providing valuable insights for structure-based studies of enzymatic mechanism and inhibitor design.

How can researchers effectively knockdown or overexpress QTRT1 for functional studies?

Several strategies can be employed for manipulating QTRT1 expression in experimental systems:

  • RNA interference (RNAi) approaches:

    • siRNA transfection for transient knockdown

    • shRNA expression vectors for stable knockdown

    • Design multiple siRNA/shRNA sequences targeting different regions of QTRT1 mRNA to ensure specificity

    • Validate knockdown efficiency at both mRNA (RT-qPCR) and protein (western blot) levels

  • CRISPR-Cas9 genome editing:

    • Generate QTRT1 knockout cell lines using CRISPR-Cas9

    • Create point mutations to study specific functional domains

    • Use CRISPR interference (CRISPRi) for transcriptional repression

    • Employ CRISPR activation (CRISPRa) for targeted overexpression

  • Overexpression systems:

    • Construct expression vectors with constitutive promoters (CMV, EF1α) for high expression

    • Use inducible expression systems (Tet-On/Off) for controlled expression

    • Consider epitope tags (FLAG, HA, His) for detection and purification

    • Validate overexpression by RT-qPCR and western blotting

  • Delivery methods:

    • Lipid-based transfection for most cell lines

    • Electroporation for difficult-to-transfect cells

    • Viral vectors (lentivirus, adenovirus) for stable integration or primary cells

    • Nanoparticle-based delivery for in vivo applications

When conducting these studies in lung adenocarcinoma contexts, A549 cells have been previously used as an appropriate model system, while 16HBE cells serve as normal controls .

What are the key considerations for designing clinical studies to evaluate QTRT1 as a biomarker?

When designing clinical studies to evaluate QTRT1 as a biomarker, researchers should consider:

Based on previous research with 1,012 LUAD samples and 112 normal controls , researchers should aim for similarly robust sample sizes to ensure statistical power.

How should researchers design experiments to investigate the relationship between QTRT1 and tRNA modification in cancer cells?

When investigating the relationship between QTRT1 and tRNA modification in cancer cells, consider the following experimental design:

  • Comparative profiling of tRNA modifications:

    • LC-MS/MS analysis of tRNA modifications in cancer vs. normal cells

    • Specific focus on queuosine levels in tRNAAsp, tRNAAsn, tRNAHis, and tRNATyr

    • Northern blot analysis with probes specific for modified/unmodified tRNAs

    • Next-generation sequencing approaches tailored for tRNA analysis

  • Manipulation of QTRT1 expression:

    • Generate QTRT1 knockdown and overexpression models in relevant cancer cell lines

    • Analyze changes in tRNA modification profiles following QTRT1 modulation

    • Assess downstream effects on protein synthesis using puromycin incorporation or ribosome profiling

    • Measure translational fidelity using reporter constructs

  • Functional consequences assessment:

    • Analyze changes in cancer hallmarks (proliferation, migration, invasion, apoptosis)

    • Correlate phenotypic changes with alterations in tRNA modification profiles

    • Conduct rescue experiments by providing exogenous queuine to QTRT1-deficient cells

    • Investigate effects on specific signaling pathways implicated in cancer progression

  • In vivo models:

    • Develop xenograft models with QTRT1-modulated cancer cells

    • Generate conditional QTRT1 knockout mouse models

    • Analyze tumor growth, metastasis, and response to therapy

    • Examine tRNA modification profiles in tumor samples

  • Clinical correlation:

    • Analyze patient-derived samples for QTRT1 expression and tRNA modification profiles

    • Correlate findings with clinical outcomes and treatment responses

    • Consider using patient-derived xenografts (PDX) or organoids for translational studies

These approaches should be implemented with appropriate controls and validated in multiple cell lines to ensure robustness of findings.

What control conditions are essential when studying QTRT1 methylation patterns?

When investigating QTRT1 methylation patterns, the following control conditions are essential:

  • Sample-related controls:

    • Matched normal-tumor tissue pairs from the same patient to account for individual variability

    • Technical replicates to assess method reliability

    • Different tissue types to determine tissue-specific methylation patterns

    • Cell lines with known methylation profiles as reference standards

  • Technical controls for methylation analysis:

    • Fully methylated DNA standards (e.g., SssI-treated DNA)

    • Fully unmethylated DNA standards (e.g., whole-genome amplified DNA)

    • Negative controls (water blanks, no-template controls)

    • Spike-in controls with known methylation levels

  • Validation using multiple methods:

    • Bisulfite sequencing for single-base resolution methylation analysis

    • Methylation-specific PCR targeting specific CpG sites

    • Pyrosequencing for quantitative methylation assessment

    • Methylation arrays for genome-wide profiling

  • Functional validation:

    • Treatment with DNA methyltransferase inhibitors (e.g., 5-aza-2'-deoxycytidine)

    • Correlation of methylation changes with expression changes

    • Reporter assays to assess the impact of methylation on promoter activity

    • CRISPR-based epigenetic editing to manipulate methylation at specific sites

Previous studies have shown marked downregulation in QTRT1 methylation in LUAD compared to normal tissues , so appropriate controls are critical for accurate interpretation of results.

How can researchers design experiments to elucidate the mechanism of QTRT1's impact on cancer progression?

To elucidate the mechanisms by which QTRT1 impacts cancer progression, researchers should design experiments that address the following aspects:

  • Pathway analysis and interaction studies:

    • Conduct RNA-seq and proteomics after QTRT1 modulation to identify affected pathways

    • Perform co-immunoprecipitation to identify protein interaction partners

    • Use proximity labeling techniques (BioID, APEX) to map the QTRT1 interactome

    • Employ ChIP-seq to identify transcription factors regulating QTRT1 expression

  • Cell-based functional assays:

    • Assess effects of QTRT1 modulation on:

      • Cell proliferation and cell cycle progression

      • Migration and invasion capabilities

      • Anchorage-independent growth

      • Response to standard chemotherapeutics

    • Conduct rescue experiments with wild-type vs. catalytically inactive QTRT1

  • Codon-specific translation effects:

    • Analyze codon usage in cancer-related genes

    • Perform ribosome profiling to assess translation efficiency at specific codons

    • Use reporter constructs with varying codon usage to test translation efficiency

    • Investigate mistranslation rates using mass spectrometry-based proteomics

  • Investigation of specific pathways identified in previous studies:

    • Focus on axoneme assembly, androgen response, and epithelial-mesenchymal transition pathways previously linked to QTRT1

    • Analyze changes in key EMT markers after QTRT1 modulation

    • Investigate androgen receptor signaling in relation to QTRT1 function

    • Examine cytoskeletal reorganization and cell motility

  • In vivo metastasis models:

    • Tail vein injection to assess lung colonization

    • Orthotopic implantation models for more physiologically relevant tumor growth

    • CRISPR-based in vivo screens to identify synthetic lethal interactions with QTRT1

    • Patient-derived xenograft models to validate findings in human tumors

What statistical approaches are most appropriate for analyzing QTRT1 expression data in relation to patient outcomes?

When analyzing QTRT1 expression data in relation to patient outcomes, researchers should consider these statistical approaches:

  • Survival analysis techniques:

    • Kaplan-Meier analysis with log-rank test for comparing survival curves between high and low QTRT1 expression groups

    • Cox proportional hazards regression for univariate and multivariate analyses

    • Competing risk analysis when multiple outcome events are possible

    • Time-dependent ROC curve analysis to evaluate the predictive accuracy of QTRT1 expression

  • Determination of optimal cutoff values:

    • Maximal statistical significance approach (minimal p-value)

    • X-tile analysis for determination of optimal cutpoint

    • Recursive partitioning methods

    • Quartile or median-based cutoffs for robustness

  • Adjustment for confounding variables:

    • Multivariate Cox regression including established prognostic factors

    • Propensity score matching to reduce selection bias

    • Stratified analysis across different clinical subgroups

    • Interaction analysis to identify effect modifiers

  • Validation strategies:

    • Internal validation using bootstrapping or cross-validation

    • External validation in independent cohorts

    • Meta-analysis combining multiple datasets

    • Sensitivity analysis with varying cutoff values

Statistical MethodAdvantagesLimitationsApplication in QTRT1 Research
Kaplan-Meier with log-rankSimple visualization, easily interpretableCannot adjust for covariatesUsed to compare OS and DFS between high and low QTRT1 expression groups
Cox proportional hazardsAllows for multivariate analysisAssumes proportional hazardsUsed to calculate HR for QTRT1 (HR=1.27, P=0.044 for OS)
ROC curve analysisEvaluates diagnostic/prognostic valueMay oversimplify time-dependent relationshipsUseful for determining optimal QTRT1 expression cutoff
Meta-analysisIncreases statistical powerHeterogeneity between studiesCan combine findings across GEO and TCGA datasets

Previous studies divided datasets like GSE72094 (n=398) and TCGA (n=502) into high and low QTRT1 expression groups using median cutoffs, which provided statistically significant survival differences .

How should researchers integrate multi-omics data to comprehensively understand QTRT1's role in cancer?

Integration of multi-omics data provides a more comprehensive understanding of QTRT1's role in cancer:

  • Integration strategies:

    • Correlation-based approaches linking expression, methylation, and clinical data

    • Network-based methods to identify functional modules involving QTRT1

    • Machine learning approaches to identify patterns across multi-omics datasets

    • Pathway enrichment analysis across multiple data types

  • Data types to integrate:

    • Transcriptomics: RNA-seq or microarray for expression profiling

    • Epigenomics: DNA methylation, histone modifications, chromatin accessibility

    • Proteomics: Protein expression, post-translational modifications

    • Metabolomics: Queuosine levels and related metabolites

    • Clinical data: Survival outcomes, treatment response, pathological features

  • Analytical frameworks:

    • Multi-Omics Factor Analysis (MOFA) to identify factors explaining variation

    • Similarity Network Fusion (SNF) to create integrated patient networks

    • iCluster for integrative clustering of cancer subtypes

    • DIABLO for multi-omics biomarker discovery

  • Visualization approaches:

    • Heatmaps with hierarchical clustering across omics layers

    • Circos plots for circular visualization of genomic data

    • Network diagrams showing interactions between different data types

    • Multi-dimensional scaling plots for sample relationships

  • Validation of findings:

    • Functional validation of computational predictions

    • Cross-validation across multiple cohorts

    • Integration with public knowledge bases

    • Experimental verification of key predictions

Studies have already begun this integration by examining both QTRT1 expression and methylation in relation to clinical features and survival outcomes in LUAD , finding complementary patterns of increased expression and decreased methylation.

What are the potential pitfalls in interpreting QTRT1 expression changes across different experimental platforms?

Researchers should be aware of several potential pitfalls when interpreting QTRT1 expression changes across different experimental platforms:

  • Platform-specific biases:

    • Microarray vs. RNA-seq differences in dynamic range and sensitivity

    • Batch effects within and between platforms

    • Probe/primer design differences affecting specificity

    • Normalization methods influencing relative expression values

  • Sample preparation variations:

    • RNA quality differences (RIN values) affecting reliability

    • FFPE vs. fresh-frozen tissue preservation methods

    • Cell culture conditions affecting baseline expression

    • Tissue heterogeneity and tumor purity differences

  • Analytical challenges:

    • Different statistical methods for defining differential expression

    • Varying cutoff criteria for significance (p-value, fold change)

    • Correction for multiple testing (FDR, Bonferroni)

    • Batch correction algorithms introducing artifacts

  • Biological considerations:

    • Splice variant detection differences between platforms

    • Cellular localization not captured by bulk RNA analysis

    • Post-transcriptional regulation not reflected in mRNA levels

    • Temporal variations in expression not captured in single timepoint analyses

  • Verification strategies:

    • Validate findings across multiple platforms

    • Confirm RNA expression changes at protein level

    • Use orthogonal methods (e.g., RT-qPCR to validate RNA-seq)

    • Examine expression in multiple model systems

What are the promising therapeutic approaches targeting QTRT1 or its pathway?

Several promising therapeutic approaches could be developed targeting QTRT1 or its pathway:

  • Direct enzyme inhibition:

    • Structure-based design of small molecule inhibitors targeting the QTRT1 active site

    • Development of nucleoside analogs that compete with queuine

    • Allosteric inhibitors targeting the QTRT1-QTRT2 interaction interface

    • Covalent inhibitors targeting conserved catalytic residues

  • RNA-based therapeutics:

    • siRNA or antisense oligonucleotides for QTRT1 knockdown

    • CRISPR-Cas13 for targeted RNA degradation

    • miRNA modulators affecting QTRT1 expression

    • Splice-switching oligonucleotides if relevant splice variants are identified

  • Combination approaches:

    • Synergistic combinations with standard chemotherapeutics

    • Combinations with immunotherapy based on pathway analysis

    • Synthetic lethality approaches with EGFR or KRAS inhibitors, given their established connections

    • Targeted degradation using PROTACs (Proteolysis Targeting Chimeras)

  • Biomarker-guided therapy:

    • Use of QTRT1 expression as a stratification marker for existing therapies

    • Development of companion diagnostics for QTRT1-targeted therapies

    • Monitoring QTRT1 expression/methylation as response markers

    • Liquid biopsy applications for non-invasive monitoring

  • Novel approaches:

    • Targeting queuine metabolism or transport

    • Modulation of downstream effectors identified in pathway analyses

    • Repurposing of existing drugs that affect tRNA modification pathways

    • Nutritional approaches based on queuine bioavailability

Given the established role of QTRT1 as a risk factor for LUAD progression and its association with survival outcomes , therapeutic targeting represents a promising avenue for future research.

What key questions remain unresolved about the molecular mechanisms of QTRT1 in human disease?

Despite advances in QTRT1 research, several key questions remain unresolved:

  • Mechanistic questions:

    • How does QTRT1-mediated tRNA modification specifically affect translation of cancer-related genes?

    • What is the precise mechanism by which QTRT1 influences epithelial-mesenchymal transition and other cancer-related pathways?

    • How does QTRT1 interact with the mitochondrial proteome and influence mitochondrial function?

    • What is the functional significance of QTRT1 methylation in regulating its expression?

  • Regulatory questions:

    • What transcription factors and epigenetic mechanisms control QTRT1 expression?

    • How is QTRT1 activity regulated post-translationally?

    • What signals modulate the formation and activity of the QTRT1-QTRT2 complex?

    • How does cellular queuine availability influence QTRT1 function?

  • Clinical questions:

    • Does QTRT1 expression or activity differ across cancer subtypes or stages?

    • Can QTRT1 serve as a predictive marker for specific therapies beyond its prognostic value?

    • Does QTRT1 contribute to therapy resistance mechanisms?

    • Is QTRT1 involved in other human diseases beyond cancer?

  • Evolutionary and comparative questions:

    • Why has the eukaryotic TGT evolved as a heterodimer compared to the bacterial homodimer?

    • How conserved is QTRT1 function across species and what can be learned from model organisms?

    • What is the evolutionary advantage of queuine modification in specific tRNAs?

  • Methodological questions:

    • What are the best approaches to specifically measure queuosine-modified tRNAs in clinical samples?

    • How can QTRT1 activity (rather than just expression) be accurately assessed in tissues?

Addressing these questions will require interdisciplinary approaches combining structural biology, biochemistry, cell biology, and clinical research.

How might advanced technologies like single-cell analysis advance our understanding of QTRT1 function?

Advanced technologies, particularly single-cell approaches, offer tremendous potential for advancing QTRT1 research:

  • Single-cell RNA sequencing (scRNA-seq):

    • Reveal cell-type specific expression patterns of QTRT1 within heterogeneous tumors

    • Identify rare cell populations with distinctive QTRT1 expression

    • Track changes in QTRT1 expression during cancer progression or treatment

    • Correlate QTRT1 with cell states (proliferative, invasive, stem-like)

  • Spatial transcriptomics:

    • Map QTRT1 expression within the tumor microenvironment

    • Correlate spatial expression patterns with histopathological features

    • Identify regional heterogeneity of QTRT1 expression

    • Relate QTRT1 expression to microenvironmental factors

  • Single-cell epigenomics:

    • Characterize cell-specific methylation patterns of the QTRT1 locus

    • Map chromatin accessibility at the QTRT1 gene regulatory regions

    • Identify epigenetic mechanisms controlling QTRT1 expression heterogeneity

    • Correlate epigenetic states with expression levels

  • Advanced proteomics:

    • Single-cell proteomics to measure QTRT1 protein levels in individual cells

    • Proximity labeling to map the QTRT1 interactome in specific cellular contexts

    • Mass spectrometry imaging to visualize QTRT1 distribution in tissues

    • Targeted proteomics to quantify modified tRNAs in small samples

  • CRISPR-based functional genomics:

    • Single-cell CRISPR screens to identify genetic interactions with QTRT1

    • Perturb-seq combining genetic perturbation with scRNA-seq readout

    • CRISPR activation/inhibition libraries to modulate QTRT1 expression

    • Base editing to introduce specific mutations and assess functional consequences

These technologies could help address the current limitations in understanding cell-specific roles of QTRT1 in heterogeneous tissues and provide new insights into its contribution to cancer progression not captured by bulk tissue analysis used in existing studies .

Product Science Overview

Enzymatic Function

QTRT1 catalyzes the exchange of guanine with queuine in the tRNA molecule. The reaction can be summarized as follows:

[tRNA]guanine+queuine[tRNA]queuine+guanine[tRNA]-guanine + queuine \rightleftharpoons [tRNA]-queuine + guanine

This reaction is essential for the proper functioning of tRNAs during protein synthesis, as the presence of queuine enhances the accuracy and efficiency of translation .

Biological Significance

The queuosine modification is critical for the proper decoding of genetic information. It has been shown to influence codon-anticodon interactions, thereby affecting the fidelity of protein synthesis. In humans, the queuosine modification is dependent on the gut microbiome, as queuine is a product of microbial metabolism. The enzyme QTRT1, encoded by the QTRT1 gene, facilitates the incorporation of queuine into tRNA .

Structural Insights

QTRT1 belongs to the family of glycosyltransferases, specifically the pentosyltransferases. Structural studies have revealed that this enzyme has a complex architecture that allows it to recognize and modify its tRNA substrates. As of late 2007, multiple structures of this enzyme class have been solved, providing insights into its catalytic mechanism .

Clinical Relevance

Mutations in the QTRT1 gene have been associated with various diseases, including complement component 9 deficiency and spastic paraplegia 26, autosomal recessive . Understanding the function and structure of QTRT1 is therefore important for developing potential therapeutic interventions for these conditions.

Research and Applications

Recombinant QTRT1 is used in research to study the role of queuosine modification in tRNA function and its impact on cellular processes. It is also utilized in biochemical assays to investigate the enzyme’s activity and its interaction with tRNA substrates.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.