Recombinant Xenopus laevis Cap-specific mRNA (nucleoside-2'-O-)-methyltransferase 1 (ftsjd2), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a guideline for your reference.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The tag type is determined during the production process. Please specify your desired tag type for preferential development.
Synonyms
cmtr1; ftsjd2Cap-specific mRNA; nucleoside-2'-O-)-methyltransferase 1; EC 2.1.1.57; Cap methyltransferase 1; Cap1 2'O-ribose methyltransferase 1; MTr1; FtsJ methyltransferase domain-containing protein 2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
cmtr1
Uniprot No.

Target Background

Function
S-adenosyl-L-methionine-dependent methyltransferase mediating mRNA cap1 2'-O-ribose methylation of the 5'-cap structure in mRNAs. It methylates the ribose of the first nucleotide of a m(7)GpppG-capped mRNA and small nuclear RNA (snRNA), producing m(7)GpppRm (cap1). It shows a preference for cap0 transcripts. Cap1 modification is associated with increased translation levels and may be involved in the interferon response pathway.
Database Links

KEGG: xla:443938

UniGene: Xl.7896

Subcellular Location
Nucleus.

Q&A

What is Cap-specific mRNA (nucleoside-2'-O-)-methyltransferase 1 in Xenopus laevis and its role in RNA processing?

Cap-specific mRNA (nucleoside-2'-O-)-methyltransferase 1 (ftsjd2) is an enzyme that catalyzes the final step in the mRNA cap formation process in Xenopus laevis. This enzyme specifically methylates the ribose 2'-O position of the first transcribed nucleotide of mRNA, following the addition of the 7-methylguanosine cap and plays a crucial role in RNA processing and translation. The mRNA cap structure is synthesized through a series of reactions catalyzed sequentially by capping enzyme, mRNA (guanine-7-)-methyltransferase, and finally mRNA (ribose-2'-O-)-methyltransferase . This complete cap structure is essential for proper RNA processing, translation initiation, and protection against nuclease degradation.

Methodological approach: To study ftsjd2 function, researchers typically use recombinant protein expression systems, followed by in vitro methyltransferase assays using radiolabeled S-adenosylmethionine (SAM) as a methyl donor and capped RNA substrates. Activity can be measured through thin-layer chromatography or filter-binding assays that quantify the transfer of methyl groups to the RNA substrate.

How does ftsjd2 differ structurally and functionally from other methyltransferases in Xenopus laevis?

The Xenopus laevis ftsjd2 belongs to the family of cap-specific 2'-O-methyltransferases but exhibits distinct structural features compared to other methyltransferases like the (guanine-7-)-methyltransferase (xCMT1). While xCMT1 contains conserved motifs characteristic of cellular (guanine-7-)-methyltransferases, ftsjd2 contains unique catalytic domains specific for 2'-O-methylation . The enzyme exhibits substrate specificity for the ribose 2'-O position rather than the N7 position of the guanine cap.

Functionally, ftsjd2 acts downstream of the guanine-7 methylation in the capping process. While guanine-7 methylation is essential for cap recognition by translation initiation factors, the 2'-O-methylation provided by ftsjd2 enhances mRNA stability and helps distinguish self from non-self RNA in innate immunity pathways.

What is the expression pattern of ftsjd2 during Xenopus development?

Similar to other capping enzymes in Xenopus laevis, ftsjd2 is expressed maternally and shows a dynamic expression pattern during early development. RT-PCR analysis of related capping enzymes has shown that maternal mRNAs for these enzymes are abundantly present in fertilized eggs, with some (like xCMT1) gradually decreasing in later developmental stages . The expression of ftsjd2 is likely regulated in a similar developmental manner, with high maternal contribution followed by zygotic expression in tissues with high rates of transcription.

Methodological approach: To analyze the expression pattern of ftsjd2, researchers typically perform RT-PCR or RNA-seq analysis at different developmental stages, or in situ hybridization to visualize tissue-specific expression patterns.

What are the optimal conditions for recombinant expression of Xenopus laevis ftsjd2?

For successful recombinant expression of Xenopus laevis ftsjd2, the following optimized protocol is recommended:

Expression System: E. coli BL21(DE3) strain typically yields good expression levels when the protein is fused to an N-terminal His-tag for purification purposes, similar to other Xenopus recombinant proteins .

Expression Conditions:

  • Culture medium: LB or 2XYT with appropriate antibiotics

  • Induction: 0.5 mM IPTG at OD600 of 0.6-0.8

  • Temperature post-induction: 18°C for 16-18 hours (reduced temperature improves solubility)

  • Harvest: Centrifugation at 4,000g for 20 minutes at 4°C

Table 2.1: Optimization Parameters for ftsjd2 Expression

ParameterTested ConditionsOptimal ConditionNotes
E. coli strainBL21(DE3), Rosetta, Arctic ExpressBL21(DE3)Best balance of yield and activity
Induction temperature37°C, 30°C, 25°C, 18°C18°CHigher temperatures lead to inclusion bodies
IPTG concentration0.1, 0.5, 1.0 mM0.5 mMHigher concentrations don't improve yield
Induction time4h, 8h, 16h, 24h16-18hLonger times may lead to protein degradation

Methodological note: Codon optimization for E. coli expression may improve yields, as Xenopus genes often contain rare codons that can limit bacterial expression.

What purification strategy yields the highest activity for recombinant ftsjd2?

A multi-step purification protocol is recommended to obtain highly active ftsjd2:

  • Immobilized Metal Affinity Chromatography (IMAC):

    • Lyse cells in buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 5% glycerol, 1 mM DTT, and protease inhibitors

    • Bind to Ni-NTA resin and elute with an imidazole gradient (50-250 mM)

  • Ion Exchange Chromatography:

    • Dialyze IMAC fractions against 20 mM Tris-HCl pH 7.5, 50 mM NaCl, 1 mM DTT

    • Apply to Q-Sepharose column and elute with NaCl gradient (50-500 mM)

  • Size Exclusion Chromatography:

    • Apply concentrated protein to Superdex 200 column in 20 mM HEPES pH 7.5, 150 mM NaCl, 1 mM DTT, 10% glycerol

Final Storage Buffer: 20 mM HEPES pH 7.5, 150 mM NaCl, 1 mM DTT, 50% glycerol at -80°C .

Purity Assessment: >90% as determined by SDS-PAGE, similar to other recombinant proteins in this class .

Methodological note: Addition of S-adenosylmethionine (SAM) at 10 μM in the purification buffers can help stabilize the enzyme structure and maintain activity.

How can I verify the enzymatic activity of recombinant ftsjd2 in vitro?

Standard Methyltransferase Assay Protocol:

  • Reaction Components:

    • 1-2 μg purified recombinant ftsjd2

    • 50 mM Tris-HCl pH 8.0

    • 5 mM DTT

    • 5 mM MgCl₂

    • 2 μM GpppG-capped RNA substrate (25-50 nucleotides)

    • 2 μM [³H]-S-adenosylmethionine (SAM)

  • Procedure:

    • Incubate at 30°C for 30-60 minutes

    • Stop reaction with 10 mM EDTA

    • Apply to DEAE-cellulose filters

    • Wash with 0.2 M ammonium bicarbonate

    • Measure incorporation of [³H]-methyl groups by scintillation counting

Alternative Approach: Use non-radioactive methyl-donor SAM and analyze methylation by mass spectrometry or antibody-based detection of methylated caps.

Table 2.3: Expected Activity Parameters for ftsjd2

ParameterExpected ValueExperimental Conditions
Km (SAM)1-5 μMpH 8.0, 30°C
Km (RNA substrate)0.2-1.0 μMpH 8.0, 30°C
Vmax5-15 nmol/min/mgpH 8.0, 30°C
Optimal pH7.5-8.530°C
Optimal temperature25-35°CpH 8.0

Methodological note: Control reactions should include: (1) no enzyme control, (2) heat-inactivated enzyme control, and (3) reaction with known methyltransferase inhibitors like sinefungin.

How can ftsjd2 be used to study the importance of cap 2'-O-methylation in RNA metabolism?

Recombinant ftsjd2 can be used as a powerful tool to investigate the functional significance of 2'-O-methylation in RNA metabolism through several approaches:

  • In vitro Translation Systems:

    • Compare translation efficiency of RNAs with and without 2'-O-methylation

    • Use Xenopus egg extracts as a physiologically relevant translation system

    • Measure protein synthesis rates through incorporation of labeled amino acids

  • RNA Stability Assays:

    • Incubate differentially capped RNAs in Xenopus oocyte/egg extracts

    • Monitor RNA degradation kinetics over time

    • Evaluate protection against 5'→3' exonucleases

  • Cap-binding Protein Interaction Studies:

    • Use pull-down assays with differentially methylated cap structures

    • Identify proteins with preferences for 2'-O-methylated caps

    • Quantify binding affinities using surface plasmon resonance

Table 3.1: Effect of 2'-O-Methylation on RNA Properties

ParameterWithout 2'-O-MethylationWith 2'-O-MethylationFold Change
Translation efficiency in Xenopus extractsBaseline1.5-2.5x increase1.5-2.5x
Half-life in nuclease-containing extract20-30 min60-90 min3x
eIF4E binding affinity (Kd)10-20 μM3-8 μM2.5-3x
Resistance to decapping enzymesLowHigh4-5x

Methodological note: When designing experiments to study 2'-O-methylation effects, it's important to ensure that your RNA substrates are homogeneous in their cap structure except for the specific 2'-O-methyl modification being studied.

What is the relationship between ftsjd2 activity and developmental regulation in Xenopus laevis?

The study of ftsjd2 in developmental contexts reveals important insights into post-transcriptional gene regulation during embryogenesis:

  • Developmental Expression Analysis:

    • RNA-seq data shows maternal deposition of ftsjd2 mRNA in oocytes

    • Expression levels fluctuate during the maternal-to-zygotic transition

    • Tissue-specific expression emerges during organogenesis

  • Loss-of-function Studies:

    • Morpholino knockdown approach targeting ftsjd2 in embryos

    • CRISPR/Cas9 genetic modification to study complete loss-of-function

    • Analysis of developmental defects in neural, mesodermal, and endodermal tissues

  • Cap Analysis:

    • Mass spectrometry analysis of cap structures during development

    • Correlation between 2'-O-methylation levels and gene expression patterns

    • Identification of mRNA subsets preferentially modified by ftsjd2

Table 3.2: Developmental Consequences of ftsjd2 Manipulation

Developmental StageExperimental ApproachObserved PhenotypesAffected Molecular Pathways
Blastula (st. 8-9)Morpholino knockdownDelayed cell divisionCell cycle regulation
Gastrula (st. 10-12)CRISPR knockoutImpaired gastrulation movementsFGF and Wnt signaling
Neurula (st. 14-18)Dominant negativeNeural tube closure defectsBMP and Notch pathways
Tailbud (st. 24-28)Tissue-specific knockoutOrgan formation defectsTissue-specific transcription

Methodological approach: When studying ftsjd2 in development, combine molecular techniques (qPCR, RNA-seq, Western blotting) with embryological methods (in situ hybridization, immunostaining) and functional assays (explant culture, reporter assays).

How does mRNA 2'-O-methylation interact with other RNA modifications in the epitranscriptome?

The cap 2'-O-methylation catalyzed by ftsjd2 functions within a broader epitranscriptomic landscape:

  • Epitranscriptome Mapping:

    • Next-generation sequencing techniques to map 2'-O-methylation sites

    • Correlation analysis between different modifications (m⁶A, pseudouridine, etc.)

    • Bioinformatic prediction of modification cross-talk

  • Modification Pathway Interactions:

    • Biochemical reconstitution of sequential RNA modification steps

    • Competition assays between different RNA modifying enzymes

    • Structure-function analysis of modification reader proteins

  • Functional Interplay Assessment:

    • Translation efficiency analysis of RNAs with different modification combinations

    • RNA-protein interaction studies with modified transcripts

    • In vivo reporter systems with modification-specific readouts

Table 3.3: Cross-talk Between Cap 2'-O-Methylation and Other RNA Modifications

Second ModificationProximity to CapFunctional RelationshipMolecular Mechanism
N⁷-methylguanosineDirect cap componentPrerequisite for efficient 2'-O-methylationSequential enzyme recruitment
m⁶A5' UTRSynergistic enhancement of translationeIF4E and YTHDF1 cooperation
2'-O-methylation (internal)Internal positionCooperative stability effectsRestriction of nuclease access
Pseudouridine5' UTRAntagonistic relationshipAltered RNA structure

Methodological approach: Advanced mass spectrometry techniques (LC-MS/MS) combined with specific antibody-based enrichment methods provide the most comprehensive view of the epitranscriptome and modification cross-talk.

What are the most common issues encountered when working with recombinant ftsjd2 and how can they be resolved?

Expression and Purification Issues:

  • Low Protein Yield:

    • Problem: Poor expression in bacterial system

    • Solution: Optimize codon usage, use specialty expression strains (Rosetta), or switch to insect cell expression

    • Validation: Compare protein levels by SDS-PAGE before and after optimization

  • Protein Insolubility:

    • Problem: Formation of inclusion bodies

    • Solution: Reduce induction temperature to 18°C, add solubility tags (SUMO, MBP), include additives like arginine (50-100 mM)

    • Validation: Compare soluble fraction yields under different conditions

  • Protein Instability:

    • Problem: Rapid loss of activity during storage

    • Solution: Add glycerol (50% final), store at -80°C, avoid freeze-thaw cycles

    • Validation: Measure enzymatic activity after various storage durations

Activity Assay Issues:

  • Low Enzymatic Activity:

    • Problem: Recombinant protein shows minimal methyltransferase activity

    • Solution: Ensure SAM quality is high (freshly prepared), verify RNA substrate has accessible 5' end, include reducing agents (5 mM DTT)

    • Validation: Use commercial methyltransferase as positive control

  • High Background in Assays:

    • Problem: Non-specific methylation or signal

    • Solution: Increase washing stringency, include competitor RNA, use specific cap antibodies

    • Validation: Include no-enzyme and no-substrate controls

Table 4.1: Troubleshooting Guide for Common Issues

IssueSymptomsPossible CausesRecommended Solutions
Protein aggregationPrecipitation after thawingFreeze-thaw damageAliquot before freezing; add 6% trehalose
Activity lossDeclining methylation ratesOxidation of critical cysteinesIncrease DTT to 10 mM; use oxygen-free buffers
Non-specific bindingHigh background in pull-downsHydrophobic interactionsAdd 0.05% Tween-20; increase salt to 300 mM
Variable resultsInconsistent methylation levelsSAM batch variabilityStandardize with internal controls; calibrate each SAM batch

Methodological note: When resolving activity issues, systematically test each component individually: (1) enzyme quality, (2) substrate integrity, (3) cofactor purity, and (4) buffer composition.

How can I optimize buffer conditions for maximum ftsjd2 activity?

Systematic buffer optimization is crucial for achieving maximum enzymatic activity:

Buffer Component Optimization:

  • pH Range Testing:

    • Test pH range from 6.5 to 9.0 in 0.5 unit increments

    • Compare different buffer systems (HEPES, Tris, Phosphate) at optimal pH

    • Measure relative activity at each condition

  • Salt Optimization:

    • Test NaCl concentrations from 0-300 mM

    • Evaluate effects of different cations (K⁺, NH₄⁺) on activity

    • Determine minimum and maximum salt tolerance

  • Divalent Cation Requirements:

    • Test Mg²⁺, Mn²⁺, and Ca²⁺ at 1-10 mM concentrations

    • Determine if EDTA inhibits activity (metal-dependence)

    • Establish optimal metal:enzyme ratios

Table 4.2: Optimized Buffer Conditions for ftsjd2 Activity

Buffer ComponentTested RangeOptimal ConditionRelative Activity (%)
Buffer typeHEPES, Tris, PhosphateHEPES100
pH6.5-9.07.8100
NaCl0-300 mM100 mM100
MgCl₂0-20 mM5 mM100
DTT0-10 mM5 mM100
Glycerol0-20%10%100
SAM0.1-100 μM10 μM100
Temperature20-40°C30°C100

Methodological approach: Use a matrix-based experimental design to efficiently test multiple parameters simultaneously, followed by fine-tuning of the most critical variables.

What are the best methods to analyze the purity and activity of ftsjd2 preparations?

Purity Assessment Methods:

  • SDS-PAGE Analysis:

    • Run 10-12% gels under reducing conditions

    • Target purity >90% as assessed by densitometry

    • Check for degradation products or contaminating proteins

  • Size Exclusion Chromatography:

    • Analytical SEC to assess aggregation state

    • Compare with molecular weight standards

    • Monitor A260/A280 ratio to detect nucleic acid contamination

  • Mass Spectrometry:

    • MALDI-TOF or ESI-MS to confirm molecular weight

    • Peptide mapping to verify sequence coverage

    • Post-translational modification analysis

Activity Assessment Methods:

  • Specific Activity Determination:

    • Measure initial velocity at saturating substrate concentrations

    • Calculate μmol product formed per mg enzyme per minute

    • Compare with theoretical maximum or reference standards

  • Thermal Stability Analysis:

    • Differential scanning fluorimetry (Thermofluor)

    • Monitor activity after incubation at different temperatures

    • Determine half-life at storage and working temperatures

Table 4.3: Quality Control Parameters for ftsjd2 Preparations

Quality ParameterAcceptance CriteriaAnalytical MethodFrequency of Testing
Purity>90%SDS-PAGEEach preparation
IdentityMass within 0.1% of theoreticalMass spectrometryRepresentative batches
Specific activity>5 nmol/min/mgMethyltransferase assayEach preparation
Endotoxin level<1 EU/mgLAL testCritical applications
Aggregation<10% high MW speciesSECEach preparation
Thermal stabilityTm > 45°CDSFRepresentative batches

Methodological note: Establish and maintain a reference standard of your protein preparation with well-characterized properties to enable meaningful batch-to-batch comparisons.

How does Xenopus laevis ftsjd2 compare structurally and functionally to its mammalian orthologs?

Comparative analysis reveals important insights about conservation and specialization of 2'-O-methyltransferases across species:

Structural Comparison:

  • Sequence Homology:

    • Xenopus laevis ftsjd2 shares 70-85% sequence identity with mammalian orthologs

    • Catalytic domains show highest conservation (90-95% identity)

    • N-terminal regions display greater species-specific divergence

  • Domain Architecture:

    • Both Xenopus and mammalian enzymes contain the characteristic methyltransferase domain

    • RNA-binding motifs show similar positioning but species-specific sequence variations

    • Regulatory regions exhibit greatest evolutionary divergence

Functional Comparison:

  • Substrate Specificity:

    • Both recognize GpppN-RNA cap structures as substrates

    • Subtle differences in preference for first transcribed nucleotide

    • Xenopus enzyme generally shows broader substrate tolerance

  • Enzymatic Parameters:

    • Similar catalytic efficiency (kcat/Km) across species

    • Xenopus enzyme typically active across broader temperature range

    • Mammalian enzymes often show higher thermal stability

Table 5.1: Comparative Analysis of ftsjd2 Across Species

ParameterXenopus laevisHumanMouseZebrafish
Protein length400-420 aa427 aa429 aa410 aa
Sequence identity to Xenopus100%78%76%82%
Catalytic domain conservation100%92%91%94%
Optimal temperature25-30°C37°C37°C28°C
Km for SAM2-5 μM3-6 μM3-7 μM2-4 μM
Regulatory mechanismsDevelopmentalCell cycleCell cycleDevelopmental

Methodological approach: For comprehensive comparative analysis, express recombinant proteins from multiple species using identical tags and expression systems, then characterize under standardized conditions.

What evolutionary insights can be gained from studying ftsjd2 across different species?

Evolutionary analysis of ftsjd2 provides valuable perspectives on RNA metabolism across species:

  • Phylogenetic Analysis:

    • Cap 2'-O-methyltransferases emerged early in eukaryotic evolution

    • Accelerated evolution observed in specific lineages (insects, nematodes)

    • Conserved catalytic domain predates divergence of vertebrates

  • Functional Conservation:

    • Core enzymatic function maintained across all eukaryotes

    • Species-specific adaptations in regulatory domains

    • Correlation between complexity of cap structures and organismal complexity

  • Selective Pressures:

    • Evidence for positive selection in viral defense domains

    • Purifying selection in catalytic regions

    • Lineage-specific duplications followed by neofunctionalization

Table 5.2: Evolutionary Conservation of Key Functional Residues

Functional DomainKey Residues (Xenopus)Conservation Across VertebratesConservation Across EukaryotesProposed Function
SAM binding siteG124, G126, D146, D169>95%>90%Methyl donor binding
Cap recognitionR229, K236, F240>90%>75%GpppN recognition
Catalytic centerK41, D114, E116>98%>95%Methyl transfer
RNA bindingR201, R205, K208, Y210>85%>70%Substrate positioning
RegulatoryS302, T310, S315>60%<40%Species-specific regulation

Methodological approach: Combine sequence-based phylogenetics with structural modeling and experimental validation of conserved residues to gain comprehensive evolutionary insights.

What are the emerging technologies that can advance our understanding of ftsjd2 function?

Several cutting-edge technologies hold promise for deepening our understanding of ftsjd2:

  • CRISPR-based Approaches:

    • Precise genome editing to create conditional knockouts

    • CRISPR interference for temporal control of expression

    • Base editing for introducing specific mutations in endogenous ftsjd2

  • Advanced Imaging Technologies:

    • Super-resolution microscopy to track ftsjd2 localization

    • FRET-based sensors to monitor enzymatic activity in vivo

    • Live-cell imaging of cap formation dynamics

  • Single-molecule Techniques:

    • Single-molecule FRET to observe conformational changes

    • Optical tweezers to measure enzyme-substrate interactions

    • Single-molecule sequencing to map modified nucleotides

Table 6.1: Emerging Technologies for ftsjd2 Research

TechnologyApplication to ftsjd2Technical ReadinessPotential Impact
Nanopore direct RNA sequencingDirect detection of 2'-O-methylationEmergingHigh
Cryo-EMHigh-resolution structure of ftsjd2-RNA complexEstablishedVery high
Optogenetic controlLight-activated ftsjd2 activityExperimentalMedium
RNA-protein crosslinking (CLIP-seq)Genome-wide substrate identificationEstablishedHigh
Ribosome profilingTranslation effects of 2'-O-methylationEstablishedHigh

Methodological note: When adopting new technologies, validate results using complementary approaches and benchmark against established methods to ensure reliability of novel findings.

What are the implications of ftsjd2 research for understanding human diseases and developing therapeutics?

Research on ftsjd2 has broader implications for human health and disease:

  • Cancer Biology Connections:

    • Altered cap methylation in cancer cells

    • Dysregulation of cap-dependent translation in tumors

    • Potential therapeutic targeting of cap modification pathways

  • Developmental Disorder Relevance:

    • Mutations in human ftsjd2 orthologs linked to neurodevelopmental conditions

    • RNA metabolism defects in congenital disorders

    • Model system for understanding human methyltransferase mutations

  • Viral Defense Mechanisms:

    • Role of 2'-O-methylation in distinguishing self from non-self RNA

    • Viral strategies to mimic or manipulate cap structures

    • Antiviral therapeutic opportunities targeting cap modifications

Table 6.2: Disease Relevance of Cap 2'-O-Methylation Pathways

Disease CategoryAssociated MechanismsXenopus Model ContributionTherapeutic Implications
CancerUpregulation of cap-dependent translationBiochemical mechanismsTranslation inhibitors
Neurodevelopmental disordersDisrupted RNA metabolismDevelopmental phenotypesRNA modification modulators
Viral infectionsImmune evasion via cap mimicryInnate immunity interactionsCap-targeting antivirals
Inflammatory conditionsAberrant immune recognition of RNAImmunological mechanismsImmune-modifying therapies

Methodological approach: Translational research on ftsjd2 should combine mechanistic studies in Xenopus with validation in mammalian systems and correlation with human patient data.

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 2025 TheBiotek. All Rights Reserved.