Recombinant Triticum aestivum Protein RAFTIN 1A (RAFTIN1A)

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 purchase method and location. Please contact your local distributor for precise delivery estimates. Note: All proteins are shipped with standard 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. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on various 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
Tag type is determined during the manufacturing process. The tag type is determined during production. To request a specific tag, please inform us; we will prioritize its development.
Synonyms
RAFTIN1A; Protein RAFTIN 1A; TaRAFTIN1a; BURP domain-containing protein 1A
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
21-389
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Triticum aestivum (Wheat)
Target Names
RAFTIN1A
Target Protein Sequence
QLGHAAPATA EVFWRAVLPH SPLPDAVLRL LKQPAAGVEL LTEATSFVRD AEDRPPFDYR DYSRSPPDDE PSKSTGAASG ARDFDYDDYS GGDKLRGAAS GARDFDYDDY SGADKLRGAT DEYKAPSSSL AGNGASMARG GKAETTTVFF HEEAVRVGKR LPFRFPPATP AALGFLPRQV ADSVPFTTAA LPGVLATFGV ASDSATVASM EATLRACESP TIAGESKFCA TSLEALVERA MEVLGTRDIR PVTSTLPRAG APLQTYTVRS VRPVEGGPVF VACHDEAYPY TVYRCHTTGP SRAYMVDMEG ARGGDAVTIA TVCHTDTSLW NPEHVSFKLL GTKPGGTPVC HLMPYGHIIW AKNVNRSPA
Uniprot No.

Target Background

Function
Essential for pollen development. Likely synthesized in the tapetum, packaged within Ubisch bodies, and transported to microspores at specific developmental stages.
Database Links

UniGene: Ta.56095

Tissue Specificity
Specifically expressed in anthers, in the tapetum and microspores (at protein level).

Q&A

What is RAFTIN1A and what is its biological function in wheat?

RAFTIN1A (also known as Protein RAFTIN 1A or BURP domain-containing protein 1A) is a protein found in Triticum aestivum (common wheat) that plays a critical role in pollen development. The protein is primarily synthesized in the tapetum, packaged in Ubisch bodies, and transported at appropriate developmental stages to the microspores . Research indicates that RAFTIN1A is part of a larger network of proteins involved in wheat reproductive development.

The protein contains approximately 389 amino acids and belongs to the BURP domain-containing protein family . The biological significance of RAFTIN1A lies in its essential function in male reproductive development in wheat, making it an important target for crop improvement research and understanding plant reproduction mechanisms.

How does RAFTIN1A differ from other similar proteins like mammalian RAFT1?

Despite similar nomenclature, RAFTIN1A from Triticum aestivum should not be confused with mammalian RAFT1 (Rapamycin and FKBP12 Target 1), which is a completely different protein with distinct functions. Mammalian RAFT1 is a 245 kDa protein that interacts with FKBP12 in a rapamycin-dependent manner and shows homology to yeast TOR proteins . This mammalian protein is involved in immunosuppressant mechanisms and cell signaling pathways.

In contrast, wheat RAFTIN1A functions in plant reproductive development and does not share structural or functional similarities with mammalian RAFT1. When designing experiments or interpreting literature, researchers should be careful to distinguish between these distinct proteins to avoid confusion in experimental design and data interpretation.

What are the optimal expression systems for producing recombinant RAFTIN1A?

Research has demonstrated successful expression of recombinant RAFTIN1A in prokaryotic systems, particularly using E. coli. The most effective expression system reported in the literature is E. coli strain ArcticExpress (DE3) from Agilent Technologies . This strain was shown to produce higher yields compared to other E. coli strains like BL21(DE3) and Rosetta-gami 2(DE3) due to its ability to fold proteins at lower temperatures, which is particularly beneficial for plant proteins that might otherwise form inclusion bodies.

The expression protocol involves:

  • Transforming E. coli ArcticExpress (DE3) with a plasmid containing the Rht-D1a gene

  • Growing cells to OD600 of 0.6 at 37°C

  • Reducing temperature to 13°C and incubating for 30 minutes

  • Inducing expression with 500 μM IPTG

  • Continuing expression for 16 hours

This low-temperature expression strategy significantly improves the solubility and correct folding of RAFTIN1A, allowing for more efficient purification.

What is the recommended purification protocol for obtaining high-purity RAFTIN1A?

For successful purification of recombinant RAFTIN1A, affinity chromatography has proven to be highly effective. The recommended protocol based on published research includes:

  • Construct a 6xHis-tagged RAFTIN1A expression vector (e.g., using pET-28c vector)

  • Express in ArcticExpress (DE3) E. coli cells as described above

  • Perform single-step affinity purification using nickel or cobalt-based resins

  • Elute the protein using an imidazole gradient

Research has shown that "Due to strong expression in ArcticExpress (DE3), it was possible to purify RHT-D1A to homogeneity by only single-step affinity purification" . This approach yields protein of sufficient purity for most applications, including antibody generation.

For researchers requiring ultra-high purity, additional purification steps such as size exclusion chromatography or ion exchange chromatography may be employed, though these were not found necessary in published protocols for standard applications.

How can researchers generate effective polyclonal antibodies against RAFTIN1A?

Generation of polyclonal antibodies against RAFTIN1A has been successfully accomplished using the following methodology:

  • Expression and Purification: Express and purify soluble 6xHis-tagged RAFTIN1A protein as described earlier.

  • Immunization Protocol:

    • Use New Zealand white rabbits as hosts

    • Inject purified recombinant RAFTIN1A according to standard immunization schedules

    • Collect antiserum after sufficient immunization period

  • Antibody Purification:

    • Perform ammonium sulfate precipitation of antiserum

    • Follow with affinity chromatography on protein A-agarose beads

This approach has been demonstrated to yield polyclonal antibodies with high specificity and sensitivity for RAFTIN1A detection in various applications.

What validation methods should be used to confirm antibody specificity for RAFTIN1A?

To ensure the specificity and reliability of anti-RAFTIN1A antibodies, comprehensive validation is essential. Published research demonstrates that effective validation includes:

  • Western Blot Analysis: Test the antibody against both recombinant RAFTIN1A and wheat seedling extracts. The antibody should recognize a protein with a molecular mass close to the predicted molecular weight of the endogenous RAFTIN1A protein .

  • Immunoprecipitation: Verify that the antibody can successfully immunoprecipitate RAFTIN1A from plant tissue extracts.

  • Response to Physiological Conditions: Confirm that the antibody can detect changes in RAFTIN1A levels in response to relevant stimuli. For example, research has shown that "RHT-D1A disappears in response to exogenous and endogenous gibberellic acid" , providing a useful control condition.

  • Cross-Reactivity Testing: Evaluate potential cross-reactivity with other related proteins by testing the antibody against control samples lacking RAFTIN1A or containing related proteins.

Proper validation ensures that experimental results obtained using these antibodies are reliable and reproducible, which is crucial for publication-quality research.

What experimental design approaches are most suitable for studying RAFTIN1A function in wheat development?

When designing experiments to study RAFTIN1A function in wheat development, several experimental design approaches should be considered:

  • Randomized Block Design: This approach is particularly useful when studying RAFTIN1A in different wheat varieties or under different treatment conditions. As described in the literature, a randomized block design helps "prevent factors not accounted for by the experimenter from affecting the response variable" . For example, when studying RAFTIN1A expression across different wheat cultivars, blocking by cultivar would control for genetic variation.

  • Factorial Experimental Design: This design is optimal when investigating the effects of multiple factors on RAFTIN1A expression or function. It "involves collecting data on all possible combinations of the factors" . For instance, studying how both environmental conditions and developmental stages affect RAFTIN1A expression would benefit from this approach.

  • Time-series Experimental Design: Since RAFTIN1A is involved in developmental processes, time-series designs are valuable for tracking expression changes throughout pollen development stages.

The specific design choice should be guided by the research questions and available resources. Statistical power analysis should be conducted to determine appropriate sample sizes for detecting expected effect sizes.

How can researchers effectively control for variables when studying RAFTIN1A in different wheat varieties?

Controlling for variables is crucial when studying RAFTIN1A across different wheat varieties to ensure valid and reproducible results. Recommended approaches include:

  • Use of Isogenic Lines: When possible, near-isogenic lines differing only in RAFTIN1A alleles provide the strongest control for genetic background effects.

  • Blocking Design Implementation: As described in research literature, "blocking is used in order to enable comparisons among the treatments to be made within blocks of homogeneous experimental units" . For wheat variety studies, this might involve:

    • Grouping varieties by genetic relatedness

    • Controlling for developmental stage when sampling

    • Accounting for environmental conditions by using growth chambers with standardized conditions

  • Standardized Tissue Collection: Collect tissue samples at precisely defined developmental stages across all varieties, preferably using both morphological and molecular markers to confirm developmental equivalence.

  • Internal Controls: Include consistent internal reference genes or proteins across all varieties to normalize expression data. This is particularly important when comparing RAFTIN1A levels between varieties with different genetic backgrounds.

  • Technical Replication: Employ sufficient technical replicates (minimum of three) for all molecular analyses to account for measurement variability.

By systematically controlling for these variables, researchers can more confidently attribute observed differences to varietal effects rather than confounding factors.

What methodologies are recommended for studying RAFTIN1A protein interactions in wheat?

To investigate RAFTIN1A protein interactions in wheat, several complementary approaches are recommended:

  • Co-Immunoprecipitation (Co-IP): Using validated anti-RAFTIN1A antibodies, researchers can immunoprecipitate RAFTIN1A along with its interacting partners from wheat anther or pollen extracts. This technique has proven effective with RAFTIN1A as demonstrated in previous studies .

  • Yeast Two-Hybrid (Y2H) Screening: This approach can identify direct protein-protein interactions. The RAFTIN1A coding sequence can be cloned into appropriate Y2H vectors and screened against wheat cDNA libraries to identify potential interacting partners.

  • Proximity-Based Labeling: Techniques such as BioID or APEX2 proximity labeling, where RAFTIN1A is fused to a biotin ligase, can identify proteins in close proximity to RAFTIN1A in planta.

  • Bioinformatic Prediction and Validation: The STRING database already lists potential functional partners of RAFTIN1A . These predictions can guide targeted validation experiments. The database indicates interactions with several serine/threonine-protein phosphatases and uncharacterized proteins with scores ranging from 0.590 to 0.721, as shown in the table below:

Protein IDDescriptionInteraction Score
A0A3B5Y1M2Uncharacterized protein0.721
A0A3B5YYW5Uncharacterized protein0.721
A0A3B5ZX64Uncharacterized protein0.721
A0A3B5XWS0Serine/threonine-protein phosphatase0.708
A0A3B5YTD5Serine/threonine-protein phosphatase0.708
A0A3B6KCU5Serine/threonine-protein phosphatase0.708
A0A3B6LFS8Serine/threonine-protein phosphatase0.708
A0A3B6MLJ6Serine/threonine-protein phosphatase0.708
A0A3B6NQZ1Uncharacterized protein0.590
A0A3B6PP66Uncharacterized protein0.590

These predicted interactions suggest RAFTIN1A may be involved in phosphorylation-dependent signaling pathways during pollen development.

How can researchers differentiate between direct and indirect protein interactions with RAFTIN1A?

Distinguishing between direct and indirect protein interactions is crucial for understanding RAFTIN1A's functional mechanism. Recommended methodologies include:

  • In Vitro Binding Assays:

    • Pull-down assays using purified recombinant RAFTIN1A and candidate interacting proteins

    • Surface Plasmon Resonance (SPR) to quantify binding kinetics

    • Isothermal Titration Calorimetry (ITC) to determine binding thermodynamics

    These techniques can confirm direct physical interactions and provide quantitative binding parameters.

  • Domain Mapping Experiments:

    • Generate truncated versions of RAFTIN1A to identify specific interaction domains

    • Perform site-directed mutagenesis of key residues to disrupt specific interactions

    • Use these modified proteins in binding assays to verify direct interaction sites

  • Cross-Linking Studies:

    • Chemical cross-linking followed by mass spectrometry (XL-MS) can capture direct interaction interfaces

    • Photo-cross-linking with modified amino acids at specific positions can verify proximity of interaction partners

  • Förster Resonance Energy Transfer (FRET):

    • Express RAFTIN1A and candidate interacting proteins as fluorescent protein fusions

    • FRET signal indicates proteins are within 10 nm of each other, supporting direct interaction

  • Control Experiments:

    • Include non-interacting protein controls

    • Perform competition assays with unlabeled proteins

    • Test interactions in the presence of inhibitors or blocking peptides

By combining multiple complementary approaches, researchers can build strong evidence for direct versus indirect interactions, creating a more accurate model of RAFTIN1A's interactome.

How can RAFTIN1A research contribute to understanding wheat reproductive development?

RAFTIN1A research has significant potential to advance our understanding of wheat reproductive development through several research avenues:

  • Developmental Expression Profiling: Tracking RAFTIN1A expression throughout anther and pollen development can identify critical developmental windows where the protein functions. This can be accomplished using the validated antibodies described earlier combined with developmental staging of wheat reproductive tissues.

  • Functional Genomics Approaches: CRISPR/Cas9-mediated mutation or RNAi-based silencing of RAFTIN1A can reveal its specific roles in pollen development. Phenotypic analysis of these modified plants would provide insights into reproductive development processes.

  • Hormone Response Studies: Research has shown that "RHT-D1A disappears in response to exogenous and endogenous gibberellic acid" . Further investigation into how RAFTIN1A responds to various plant hormones could elucidate regulatory mechanisms in reproductive development.

  • Comparative Analysis Across Wheat Varieties: Studying RAFTIN1A sequence and expression variations across diverse wheat germplasm could identify correlations with reproductive fitness traits, potentially identifying valuable alleles for breeding programs.

  • Stress Response Investigation: Analyzing how environmental stresses affect RAFTIN1A expression and function could explain aspects of stress-induced male sterility in wheat, a significant agricultural concern.

These research directions can provide fundamental insights into wheat reproductive biology while potentially contributing to applied breeding objectives for improving wheat reproductive resilience.

What methods are effective for studying RAFTIN1A's response to hormonal signals like gibberellic acid?

Research has demonstrated that RAFTIN1A responds to gibberellic acid (GA), making this an important aspect to investigate . Effective methodologies for studying these hormone responses include:

  • Hormone Treatment Time-Course Experiments:

    • Treat wheat seedlings or anther cultures with different concentrations of GA

    • Collect samples at multiple time points post-treatment

    • Analyze RAFTIN1A protein levels using western blot with validated antibodies

    • Document the degradation kinetics and dose-response relationships

  • Protein Stability Assays:

    • Cycloheximide chase experiments to determine if GA affects RAFTIN1A protein stability

    • In vitro degradation assays using plant extracts supplemented with GA

    • Ubiquitination assays to determine if GA promotes RAFTIN1A ubiquitination

  • Transcriptional Regulation Analysis:

    • RT-qPCR to measure RAFTIN1A transcript levels in response to GA

    • Promoter-reporter constructs to visualize transcriptional responses in planta

    • Chromatin immunoprecipitation (ChIP) to identify transcription factors binding to the RAFTIN1A promoter in response to GA

  • Protein Localization Studies:

    • Immunolocalization or fluorescent protein fusions to track RAFTIN1A subcellular distribution before and after GA treatment

    • Cell fractionation followed by western blotting to quantify redistribution between cellular compartments

  • Interaction Partner Dynamics:

    • Co-immunoprecipitation before and after GA treatment to identify hormone-dependent changes in the RAFTIN1A interactome

    • Proximity labeling experiments with and without GA treatment to capture dynamic protein interaction networks

These methodologies, when applied systematically, can reveal the molecular mechanisms through which GA regulates RAFTIN1A, providing insights into hormone-responsive developmental pathways in wheat reproduction.

What are the best practices for analyzing and presenting RAFTIN1A expression data?

When analyzing and presenting RAFTIN1A expression data, researchers should follow these best practices:

  • Statistical Analysis Approaches:

    • Apply appropriate statistical tests based on experimental design (t-tests, ANOVA, or non-parametric alternatives)

    • Account for multiple comparisons when analyzing expression across many conditions

    • Include power analysis to justify sample sizes

    • Report effect sizes along with p-values to indicate biological significance

  • Data Visualization:

    • Present expression data using appropriate charts (bar graphs for discrete comparisons, line graphs for time-course data)

    • Include individual data points alongside means and error bars to show distribution

    • Use consistent color schemes and formatting throughout publications

    • Include representative western blot images alongside quantification

  • Controls and Normalization:

    • Always include appropriate loading controls for western blots

    • Normalize RAFTIN1A expression to stable reference proteins

    • For transcript analysis, validate reference genes for stability across experimental conditions

  • Results Reporting:

    • "The results section should simply state the findings, without bias or interpretation, and arranged in a logical sequence"

    • "The results section should always be written in the past tense"

    • "Be concise, using non-textual elements, such as figures and tables, if appropriate, to present results more effectively"

    • "Avoid providing data that is not critical to answering the research question"

  • Integration with Other Data Types:

    • Present RAFTIN1A expression in context with related proteins or processes

    • Consider using heat maps or network visualizations for complex datasets

    • When appropriate, integrate expression data with functional or phenotypic data

Following these practices ensures that RAFTIN1A expression data is presented in a clear, unbiased, and scientifically rigorous manner that facilitates interpretation and reproducibility.

How should researchers address contradictory findings in RAFTIN1A research?

Addressing contradictory findings is an essential aspect of scientific research. When encountering conflicting results in RAFTIN1A studies, researchers should:

  • Methodological Comparison:

    • Thoroughly analyze differences in experimental protocols (expression systems, purification methods, antibody sources)

    • Consider differences in sample preparation, detection methods, and reagent quality

    • Reproduce both contradictory methods in parallel to directly compare results

  • Biological Context Examination:

    • Evaluate differences in wheat varieties, developmental stages, or growth conditions

    • Consider tissue-specific effects that might explain different outcomes

    • Assess whether contradictions might represent genuine biological variability

  • Statistical Reassessment:

    • Review statistical analyses for appropriate tests and power

    • Consider whether contradictions might be explained by sampling variation

    • Re-analyze raw data when available using consistent statistical approaches

  • Resolution Approaches:

    • Design experiments specifically to address the contradiction

    • Employ multiple complementary techniques to triangulate findings

    • Collaborate with groups reporting contradictory results when possible

  • Transparent Reporting:

    • Acknowledge contradictions openly in publications

    • Present alternative interpretations of data

    • Suggest specific experiments that could resolve contradictions in future work

  • Consortium Approaches:

    • For persistent contradictions, consider coordinated multi-laboratory studies using standardized protocols

    • Develop community standards for RAFTIN1A research methods

By addressing contradictions systematically and transparently, researchers can advance RAFTIN1A research while maintaining scientific integrity and contributing to the field's progress rather than confusion.

What emerging technologies could advance RAFTIN1A functional studies?

Several cutting-edge technologies hold promise for advancing RAFTIN1A functional studies:

  • CRISPR/Cas9 Gene Editing:

    • Create precise modifications to RAFTIN1A in wheat

    • Generate knockout lines, domain-specific deletions, or tagged versions of the endogenous protein

    • Study effects on pollen development with unprecedented specificity

  • Single-Cell Transcriptomics and Proteomics:

    • Analyze RAFTIN1A expression in specific cell types within the anther

    • Map expression patterns at unprecedented resolution during development

    • Identify cell-type specific interaction partners

  • Cryo-Electron Microscopy (Cryo-EM):

    • Determine high-resolution structures of RAFTIN1A alone and in complex with interacting partners

    • Gain insights into functional domains and mechanistic details

    • Guide structure-based functional studies

  • Proximity-Dependent Labeling Proteomics:

    • Techniques like BioID, APEX2, or TurboID can reveal the dynamic RAFTIN1A interactome in living plant cells

    • Identify transient interactions that might be missed by traditional co-immunoprecipitation

  • Optical and Super-Resolution Microscopy:

    • Track RAFTIN1A dynamics in living cells with techniques like lattice light-sheet microscopy

    • Visualize subcellular localization with nanometer precision using PALM/STORM microscopy

    • Perform live-cell FRET experiments to monitor protein interactions in real-time

  • Proteogenomics Integration:

    • Combine genomic, transcriptomic, and proteomic data to build comprehensive models of RAFTIN1A function

    • Identify regulatory networks controlling RAFTIN1A expression and activity

  • Synthetic Biology Approaches:

    • Create synthetic variants of RAFTIN1A with altered properties

    • Design orthogonal systems to probe specific functional hypotheses

    • Engineer RAFTIN1A with controllable activity for developmental studies

These technologies, especially when used in combination, could transform our understanding of RAFTIN1A's role in wheat reproduction and potentially reveal applications for crop improvement.

How can computational biology contribute to RAFTIN1A research?

Computational biology offers powerful approaches to advance RAFTIN1A research:

  • Structural Prediction and Analysis:

    • Predict RAFTIN1A's 3D structure using AlphaFold2 or RoseTTAFold

    • Identify functional domains and potential binding sites

    • Simulate protein dynamics to understand conformational changes

    • Model protein-protein interactions with predicted partners

  • Comparative Genomics:

    • Analyze RAFTIN1A evolution across plant species

    • Identify conserved regions likely critical for function

    • Discover lineage-specific adaptations in the Triticum genus

    • Map natural variation in RAFTIN1A sequences across wheat varieties

  • Network Analysis:

    • Construct protein-protein interaction networks centered on RAFTIN1A

    • Identify hub proteins and key pathways connected to RAFTIN1A function

    • Predict functional associations from co-expression data

    • Integrate multiple data types (transcriptomic, proteomic, metabolomic) to build comprehensive models

  • Machine Learning Applications:

    • Predict phenotypic outcomes from RAFTIN1A sequence variations

    • Identify patterns in large-scale expression datasets that correlate with RAFTIN1A activity

    • Develop models to predict RAFTIN1A responses to environmental factors

  • Systems Biology Modeling:

    • Develop mathematical models of the regulatory pathways involving RAFTIN1A

    • Simulate the effects of perturbations to predict experimental outcomes

    • Identify control points in the system that might be targets for manipulation

  • Data Integration Platforms:

    • Tools like MADIBA can be adapted for "visualising implicated genes on KEGG metabolic pathways; their chromosomal localisations; putative common transcriptional regulatory elements"

    • Integrate diverse experimental data types into coherent models

By leveraging these computational approaches, researchers can generate testable hypotheses, guide experimental design, and extract deeper insights from experimental data related to RAFTIN1A function in wheat.

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.