TDRD12 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of TDRD12 Antibody

TDRD12 Antibody is an immunological reagent designed to detect the Tudor Domain Containing 12 (TDRD12) protein, a key regulator in germ cell development and transposon silencing. This antibody is primarily used in research to study TDRD12’s role in spermatogenesis, piRNA biogenesis, and its potential links to cancer and stem cell biology.

Applications in Research

TDRD12 antibodies are employed in diverse experimental contexts:

Western Blotting

  • Purpose: Quantify TDRD12 protein levels in testicular extracts or cancer cell lines.

  • Key Findings:

    • TDRD12 (~130 kDa) is highly expressed in mouse testis during postnatal development (P14–P28) .

    • Knockdown of TDRD12 in embryonal carcinoma cells reduces PIWIL2 protein levels, suggesting functional interdependence .

Immunofluorescence

  • Purpose: Map subcellular localization of TDRD12.

  • Key Findings:

    • Acrosome Localization: TDRD12 co-localizes with lectin-PNA in the acrosome of round spermatids during spermatogenesis .

    • No Overlap with TDRD1: TDRD12 does not co-localize with TDRD1 in spermatocytes, indicating distinct roles in piRNA biogenesis .

ELISA

  • Purpose: Analyze TDRD12 expression in serum or tissue lysates.

  • Key Findings:

    • TDRD12 antibodies (e.g., CSB-PA707440LA01HU) are validated for ELISA in human samples .

Role in Spermatogenesis

  • Acrosome Formation: TDRD12 is essential for acrosome development in spermatids, critical for sperm-egg interaction .

  • Transposon Silencing: TDRD12 mutants show derepressed retrotransposons (e.g., LINE1, IAP), linked to male infertility .

Cancer and Stem Cell Biology

  • Expression in Tumors: TDRD12 is enriched in colon adenocarcinoma and embryonal carcinoma cells, suggesting oncogenic potential .

  • Stem Cell Regulation: TDRD12 may maintain stemness in cancer cells by regulating retroelements .

Mechanistic Insights

  • piRNA Biogenesis: TDRD12 interacts with MILI (PIWIL2) and TDRD1 in a complex that produces secondary piRNAs, distinct from primary piRNA pathways .

  • Domain Importance: The second Tudor domain of Bombyx mori TDRD12 is critical for binding primary piRNA-bound Piwi proteins .

Challenges and Future Directions

  • Specificity: Cross-reactivity with related TDRD family proteins (e.g., TDRD1, TDRD9) requires careful validation .

  • Therapeutic Potential: TDRD12’s role in cancer stem cells and transposon regulation warrants exploration in targeted therapies .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timeframes.
Synonyms
ECAT8 antibody; ES cell associated transcript 8 antibody; ES cell-associated transcript 8 protein antibody; TDR12_HUMAN antibody; Tdrd12 antibody; Tudor domain containing 12 antibody; Tudor domain-containing protein 12 antibody
Target Names
TDRD12
Uniprot No.

Target Background

Function
TDRD12 Antibody is a probable ATP-binding RNA helicase that plays a crucial role during spermatogenesis. Its primary function is to repress transposable elements, preventing their mobilization and ensuring the integrity of the germline. TDRD12 achieves this through its involvement in the piRNA metabolic process. This process mediates the repression of transposable elements during meiosis by forming complexes composed of piRNAs and Piwi proteins. These complexes control the methylation and subsequent repression of transposons. TDRD12 is also involved in the secondary piRNAs metabolic process. It acts via the PET complex, a multiprotein complex essential for the secondary piRNAs metabolic process. The PET complex facilitates the loading of PIWIL4 piRNAs, triggered by the slicing activity of PIWIL2.
Database Links

HGNC: 25044

KEGG: hsa:91646

STRING: 9606.ENSP00000390621

UniGene: Hs.646351

Q&A

Abstract

This document provides comprehensive answers to frequently asked questions about Tudor Domain Containing 12 (TDRD12) antibodies in research contexts. Based on academic literature and scientific databases, these FAQs address both fundamental concepts and advanced applications of TDRD12 antibodies in reproductive biology, molecular genetics, and developmental studies. Researchers will find methodological guidance, troubleshooting tips, and critical insights for experimental design.

Fundamental Research Questions

What is TDRD12 and why is it important for reproductive biology research?

TDRD12 (Tudor Domain Containing 12) is a protein essential for mammalian germ cell development and male fertility. It functions in the piRNA (Piwi-interacting RNA) pathway, which plays a crucial role in silencing transposable elements in the germ line.

Structurally, TDRD12 contains two tudor domains and a DEAD box, making it a unique member of the TDRD family . The importance of TDRD12 is highlighted by multiple studies showing that:

  • Male mice lacking functional TDRD12 exhibit infertility

  • TDRD12 is essential for secondary piRNA biogenesis

  • It interacts with MILI (a Piwi protein) and associates with primary piRNAs

  • Loss of TDRD12 function causes derepression of retrotransposons

  • In humans, genetic variants in TDRD12 are associated with LINE1 derepression in spermatogonia and azoospermia

Understanding TDRD12 provides insights into fundamental mechanisms of germ cell development, transposon control, and causes of male infertility.

What are the expression patterns of TDRD12 in different tissues?

TDRD12 shows a highly specific expression pattern, which is important to consider when designing experiments with TDRD12 antibodies:

  • Tissue specificity: TDRD12 is predominantly expressed in the testis, with some studies detecting low expression in the heart

  • Developmental timeline: In mice, TDRD12 mRNA is detected in the testis from postnatal day 0 (P0) and remains high through P28

  • Protein expression: TDRD12 protein levels peak at P14 during postnatal development and then decrease at P21

  • Cell-type specificity: TDRD12 expression is initially detected in primary spermatocytes at 2 weeks of development, but later becomes localized to the acrosome of round spermatids

This specific expression pattern makes TDRD12 antibodies particularly valuable for studying spermatogenesis and testicular development.

How do you select an appropriate TDRD12 antibody for your research?

When selecting a TDRD12 antibody, consider these critical factors:

  • Species reactivity: Ensure the antibody recognizes your species of interest. Commercial antibodies are available for human, mouse, cow, horse, and pig TDRD12

  • Epitope location: Different antibodies target different regions of TDRD12. For example:

    • C-terminal antibodies (AA 991-1164)

    • Mid-region antibodies (AA 289-338)

    • N-terminal region antibodies

  • Validation status: Check if the antibody has been validated for your application:

    • Western blot validation is common for TDRD12 antibodies

    • IHC and ELISA applications require specific validation

  • Application compatibility: Different experimental techniques require antibodies with specific properties:

ApplicationRecommended Antibody TypeImportant Considerations
Western BlotUnconjugated polyclonalValidated against specific regions
IHC/IFFITC-conjugated or unconjugatedFixation method compatibility
Co-IPHigh-specificity unconjugatedLow cross-reactivity
ELISAHRP-conjugatedOptimal working dilution needed
  • Clonality: Most available TDRD12 antibodies are polyclonal, which may provide better sensitivity but potentially lower specificity

Methodological Applications

What are the optimal protocols for TDRD12 antibody use in immunofluorescence of testicular tissues?

Based on published research methodologies, here is an optimized protocol for TDRD12 immunofluorescence in testicular tissues:

  • Tissue preparation:

    • Fix testicular tissue in 4% paraformaldehyde (PFA) for 4-6 hours

    • Cryoprotect in 30% sucrose solution

    • Embed and section at 8-10 μm thickness

  • Antigen retrieval:

    • Incubate sections in 10 mM sodium citrate buffer (pH 6.0) at 95°C for 10 minutes

    • Cool to room temperature for 30 minutes

  • Blocking and permeabilization:

    • Permeabilize with 0.2% Triton X-100 in PBS for 15 minutes

    • Block with 3% BSA/10% normal goat serum in PBS for 1 hour at room temperature

  • Primary antibody incubation:

    • Dilute TDRD12 antibody to 1:100-1:500 in blocking solution

    • Incubate overnight at 4°C in a humidified chamber

    • For co-localization studies, combine with other markers:

      • Anti-DDX4 (VASA) for germ cells

      • Anti-TDRD1 for spermatocytes

      • Lectin-PNA for acrosome structure

  • Secondary antibody incubation:

    • Wash 3× with PBS (5 minutes each)

    • Incubate with fluorophore-conjugated secondary antibody (1:500) for 1 hour at room temperature

    • Include DAPI (1:1000) for nuclear counterstaining

  • Mounting and imaging:

    • Mount with anti-fade mounting medium

    • Image using confocal microscopy with appropriate filters

Note: TDRD12 has been observed specifically in the acrosome of round spermatids and not co-localized with TDRD1 or DDX4, which is important for interpreting results .

How can TDRD12 antibodies be used to study piRNA pathway components through co-immunoprecipitation?

Co-immunoprecipitation (co-IP) with TDRD12 antibodies can reveal important interactions within the piRNA pathway. Based on published methodologies :

  • Lysate preparation:

    • Homogenize fresh or flash-frozen testicular tissue in IP buffer (20 mM HEPES pH 7.4, 150 mM NaCl, 0.1% NP-40, 2 mM MgCl₂, 1 mM DTT)

    • Include protease inhibitors and RNase inhibitors if RNA interactions are being studied

    • Clear lysate by centrifugation at 14,000 × g for 15 minutes at 4°C

  • Antibody coupling:

    • Pre-clear lysate with Protein A/G beads for 1 hour at 4°C

    • Couple TDRD12 antibody to Protein A/G beads (typically 2-5 μg antibody per experiment)

    • For control, use equivalent amount of non-specific IgG from the same species

  • Immunoprecipitation:

    • Incubate pre-cleared lysate with antibody-coupled beads overnight at 4°C with gentle rotation

    • To assess RNA-dependent interactions, split samples and treat one with RNase A (100 μg/ml) for 30 minutes at room temperature

  • Washing and elution:

    • Wash beads 4-5 times with IP buffer

    • Elute proteins by boiling in SDS-sample buffer or use gentler elution with glycine (pH 2.5) for downstream applications

  • Detection methods:

    • Western blot to identify known interacting proteins (MILI, TDRD1)

    • Mass spectrometry for unbiased identification of partners

    • Small RNA sequencing of co-purified RNAs to identify associated piRNAs

Key findings from TDRD12 co-IP experiments:

  • TDRD12 co-immunoprecipitates with MILI and TDRD1

  • The MILI interaction is partially RNA-dependent, as RNase treatment reduces recovery

  • TDRD12 complexes contain piRNAs with a length profile (~26 nt) similar to MILI-bound piRNAs

  • Unlike the MILI-TDRD1 interaction, which is mediated by symmetrically dimethylated arginine (sDMA) recognition, TDRD12 interactions appear to be sDMA-independent

What are the best practices for using TDRD12 antibodies in Western blot analysis?

For successful Western blot detection of TDRD12, follow these research-validated practices:

  • Sample preparation:

    • Extract proteins using RIPA buffer with protease inhibitors

    • For testicular tissue, homogenize thoroughly and sonicate briefly to shear genomic DNA

    • Use appropriate protein quantification method (BCA or Bradford)

  • Gel electrophoresis considerations:

    • TDRD12 is a large protein (~132.6 kDa) , requiring:

      • 4-20% gradient gels for optimal resolution

      • Longer running times to ensure proper separation

      • Efficient transfer to membrane (preferably overnight at low voltage)

  • Membrane and blocking:

    • Nitrocellulose membranes are recommended

    • Block with 5% non-fat milk in TBST (TBS with 0.1% Tween-20)

  • Antibody incubation:

    • Primary antibody dilution: 1:1000 to 1:2000 in TBST with 5% milk

    • Incubate overnight at 4°C

    • Secondary antibody: HRP-conjugated anti-rabbit IgG (1:5000)

    • Include α-tubulin (~55 kDa) as loading control

  • Detection and troubleshooting:

    • Use ECL-based detection systems

    • Expected band size: ~130 kDa for full-length TDRD12

    • Mutant forms may show shifted band sizes or absence of signal

  • Validation controls:

    • Include positive control (testis extracts)

    • Use TDRD12 knockout/knockdown samples as negative controls when available

    • For developmental studies, compare samples from different postnatal days (P0, P7, P14, P21, P28)

Note: In mice, TDRD12 protein is highly detected at P14 and then decreases at P21, which should be considered when interpreting developmental expression patterns .

Advanced Research Applications

How can TDRD12 antibodies be used to study male infertility factors?

TDRD12 antibodies offer valuable tools for investigating male infertility, particularly in cases involving transposon derepression and piRNA pathway defects:

  • Clinical sample analysis:

    • TDRD12 expression can be assessed in testicular biopsies from infertile patients

    • Immunohistochemistry can reveal altered localization patterns

    • Recent research has identified homozygous variants [(Leu1053Phefs4)];[(Leu1053Phefs4)] in TDRD12 in patients with azoospermia

  • Experimental approaches:

    • Expression profiling: Compare TDRD12 levels between fertile and infertile individuals

    • Localization studies: Assess acrosome localization in round spermatids

    • Mutation screening: Identify TDRD12 variants using antibodies specific to common mutant epitopes

  • Molecular mechanisms investigation:

    • Use TDRD12 antibodies to examine piRNA pathway components in infertile samples

    • Assess transposon activation by co-staining for TDRD12 and LINE1 ORF1p

    • Examine DNA methylation at retrotransposon promoters in samples with altered TDRD12 expression

  • Translational applications:

    • Develop diagnostic assays for TDRD12-related infertility

    • Use TDRD12 antibodies in combination with other piRNA pathway markers to create comprehensive screening panels

Research findings from murine models provide important insights:

  • TDRD12-deficient mice show activation of LINE1 and IAP retrotransposons

  • DNA methylation of L1 and IAP promoters is reduced in spermatogonia from TDRD12 mutants (53% and 61% respectively compared to ~90% in controls)

  • TDRD12 mutations specifically affect repeat element methylation while genomic imprinted loci remain unaffected

How do you resolve contradictory data regarding TDRD12 localization using different antibodies?

Resolving contradictory TDRD12 localization data requires systematic methodological investigation:

  • Epitope mapping analysis:

    • Different antibodies recognize distinct regions of TDRD12, potentially explaining discrepancies

    • Compare antibodies targeting different domains:

      • N-terminal region antibodies

      • Tudor domain-specific antibodies

      • C-terminal antibodies

    • Generate an epitope map to understand which regions are accessible in different cellular compartments

  • Fixation and permeabilization optimization:

    • Compare multiple fixation methods:

      • Paraformaldehyde (4%) for general fixation

      • Methanol for better nuclear protein detection

      • Acetone for membrane protein preservation

    • Test different permeabilization protocols to ensure antibody accessibility

  • Validation with multiple approaches:

    • Combine immunofluorescence with biochemical fractionation

    • Use GFP-tagged TDRD12 constructs to confirm localization patterns

    • Employ super-resolution microscopy for precise localization

  • Developmental timing consideration:

    • Published research shows that TDRD12 localization changes during development:

      • At 2 weeks: detected in cytoplasm of primary spermatocytes

      • At 4+ weeks: localizes to acrosome of round spermatids

    • These temporal changes may explain conflicting reports if developmental stage is not carefully controlled

  • Species-specific differences:

    • Compare localization patterns between species:

      • Mouse TDRD12 localizes to acrosome in spermatids

      • In insects like Bombyx mori, BmTdrd12 interacts with Siwi in a complex

      • Drosophila TDRD12 ortholog interacts with Vreteno in ovarian germline cells

When facing contradictory data, it is recommended to use multiple antibodies targeting different regions of TDRD12 in parallel experiments, while carefully controlling for developmental stage and fixation conditions.

What are the key considerations for studying TDRD12's role in piRNA biogenesis using antibody-based approaches?

Investigating TDRD12's role in piRNA biogenesis requires careful experimental design with antibodies:

  • Target selection for co-immunoprecipitation:

    • TDRD12 interacts with specific piRNA pathway components:

      • MILI (primary piRNA-binding Piwi protein)

      • TDRD1 (Tudor domain protein)

      • piRNAs of ~26 nt length

    • These interactions should be validated in your experimental system

  • RNA-protein interaction analysis:

    • TDRD12-MILI interaction is partially RNA-dependent (RNase-sensitive)

    • Design experiments with and without RNase treatment to distinguish:

      • Direct protein-protein interactions

      • RNA-mediated associations

  • Developmental timing is critical:

    • TDRD12's role in piRNA biogenesis is most evident during specific developmental windows:

      • Studies in mice focus on embryonic day 18.5 (E18.5) and postnatal day 0 (P0) testes when MILI and MIWI2 are coexpressed

      • Secondary piRNA biogenesis defects can be assessed at these timepoints

  • Functional readouts:

    • Use multiple measures to assess TDRD12's role:

      • MIWI2 loading: TDRD12 is essential for loading MIWI2 with piRNAs

      • Nuclear localization of MIWI2: Unloaded MIWI2 remains cytoplasmic in TDRD12 mutants

      • Ping-pong signature: 10-nt overlap between 5' ends of MILI- and MIWI2-bound piRNAs is absent in TDRD12 mutants

      • Transposon derepression: L1 and IAP elements become activated in absence of functional TDRD12

  • Technical considerations for antibody-based approaches:

    • Use stringent wash conditions for specificity

    • Include appropriate controls (IgG, knockout/knockdown samples)

    • Consider epitope masking in protein complexes

    • Validate findings with orthogonal approaches (genetic models, tagged protein expression)

Key finding to note: While TDRD12 is essential for secondary piRNA biogenesis (MIWI2-bound piRNAs), primary piRNA biogenesis (MILI-bound piRNAs) appears normal in TDRD12 mutants, indicating a specific role in the secondary processing pathway .

Troubleshooting and Quality Control

How do you validate TDRD12 antibody specificity for cross-species studies?

Ensuring TDRD12 antibody specificity across species requires comprehensive validation:

  • Sequence homology analysis:

    • Align TDRD12 sequences from target species to identify:

      • Conserved regions for cross-reactive antibodies

      • Variable regions that may affect antibody binding

    • Example: Antibodies targeting the C-terminal region may show cross-reactivity with predicted binding to cow (86%), horse (86%), and pig (86%) TDRD12

  • Experimental validation approaches:

    • Western blot validation:

      • Test antibody against tissue lysates from multiple species

      • Verify expected molecular weight differences

      • Include positive controls (e.g., testis extracts) from each species

    • Peptide competition assays:

      • Pre-incubate antibody with immunizing peptide

      • Signal should be blocked in specific binding

    • Knockout/knockdown controls:

      • Use TDRD12-deficient samples when available

      • CRISPR-edited cell lines or tissues

      • RNAi-treated samples with confirmed knockdown

  • Species-specific considerations:

    • Mouse studies: Validated antibodies have been generated using the peptide SQRPNEKPLRLTEKKDC (amino acids 318-334)

    • Human studies: Antibodies targeting AA 991-1164 (C-terminal) or AA 289-338 have been validated

    • Model organisms: For zebrafish studies, custom antibodies may be required due to limited commercial options

  • Orthogonal validation:

    • Confirm results using multiple antibodies targeting different epitopes

    • Correlate protein detection with mRNA expression data

    • Use tagged TDRD12 constructs to validate antibody performance

Remember that different TDRD12 orthologs may have distinct subcellular localizations and functions across species:

  • Mouse TDRD12: Acrosome in spermatids

  • Bombyx mori BmTdrd12: Complex with Siwi but not Ago3

  • Drosophila TDRD12 ortholog: Interacts with Vreteno in germline cells

What are common pitfalls when using TDRD12 antibodies and how can they be addressed?

Researchers should be aware of these common challenges when working with TDRD12 antibodies:

  • Background and non-specific binding:

    • Issue: High background in testicular tissues due to abundant RNA-binding proteins

    • Solution:

      • Increase blocking time/concentration (5% BSA or 10% normal serum)

      • Include 0.1-0.3M NaCl in wash buffers to reduce non-specific interactions

      • Use monovalent Fab fragments instead of complete IgG

  • Cross-reactivity with related tudor domains:

    • Issue: False positives due to antibody recognition of conserved tudor domains in other proteins

    • Solution:

      • Choose antibodies targeting unique regions outside tudor domains

      • Validate with peptide competition assays

      • Confirm with knockout/knockdown controls

  • Variable results across developmental stages:

    • Issue: TDRD12 expression and localization change dramatically during development

    • Solution:

      • Carefully stage samples (embryonic, postnatal days, adult)

      • Be aware that TDRD12 peaks at P14 and shifts from spermatocytes to spermatid acrosome

      • Include developmental marker controls

  • Protein degradation:

    • Issue: TDRD12 is susceptible to proteolytic degradation

    • Solution:

      • Use fresh samples or flash-freeze immediately

      • Include multiple protease inhibitors in extraction buffers

      • Process samples quickly and maintain cold temperatures

  • Epitope masking in complexes:

    • Issue: TDRD12 exists in complexes with MILI and other factors that may obscure epitopes

    • Solution:

      • Try multiple antibodies targeting different regions

      • Test different extraction conditions (detergent types/concentrations)

      • Consider cross-linking prior to immunoprecipitation for transient interactions

  • Antibody batch variation:

    • Issue: Polyclonal antibodies show batch-to-batch variation

    • Solution:

      • Purchase larger antibody lots for long-term studies

      • Re-validate each new lot against a standard sample

      • Consider generating monoclonal antibodies for critical applications

For Western blot applications specifically, use gradient gels (4-20%) to effectively resolve the large TDRD12 protein (~132.6 kDa) and include both positive controls (testis extracts) and negative controls in each experiment .

Emerging Research Directions

How can TDRD12 antibodies facilitate studies on non-canonical functions beyond piRNA biogenesis?

While TDRD12's role in piRNA biogenesis is well-established, emerging research points to potential additional functions that can be investigated using antibody-based approaches:

  • Acrosome-related functions:

    • TDRD12 localizes to the acrosome in round spermatids, unlike other piRNA pathway components

    • This distinct localization suggests functions beyond piRNA biogenesis

    • Research applications:

      • Co-immunoprecipitation with acrosomal proteins

      • Investigation of acrosome biogenesis in TDRD12 mutants

      • Temporal correlation between TDRD12 expression and acrosome formation

  • RNA regulation beyond piRNAs:

    • TDRD12 contains a DEAD box typical of RNA helicases

    • This domain might function in RNA metabolism beyond piRNA pathways

    • Experimental approaches:

      • RNA immunoprecipitation followed by sequencing (RIP-seq) to identify non-piRNA targets

      • In vitro RNA helicase assays with immunopurified TDRD12

      • Analysis of mRNA expression changes in TDRD12-deficient cells

  • Potential roles in cell cycle regulation:

    • Tudor domain proteins often have multiple cellular functions

    • TDRD12 expression changes during development may correlate with specific cell cycle phases

    • Research strategies:

      • Co-staining with cell cycle markers

      • Flow cytometry analysis of TDRD12 expression across cell cycle

      • Chromatin immunoprecipitation to identify potential genomic targets

  • Species-specific functions:

    • Drosophila TDRD12 ortholog interacts with Vreteno in follicle cells

    • Zebrafish Tdrd12 is essential for germ cell development

    • Cross-species comparisons could reveal evolutionary adaptations in TDRD12 function

    • Approaches:

      • Comparative immunoprecipitation studies across species

      • Rescue experiments with TDRD12 orthologs from different species

      • Domain-specific functional analysis

  • Potential roles in somatic tissues:

    • While predominantly expressed in testis, low TDRD12 expression has been detected in heart tissue

    • This suggests possible functions in non-germline contexts

    • Investigation methods:

      • Highly sensitive immunodetection in non-gonadal tissues

      • Single-cell analysis to identify rare TDRD12-expressing cells in somatic tissues

      • Conditional knockout studies focusing on tissues with low expression

For all these non-canonical studies, using multiple antibodies targeting different TDRD12 domains and including appropriate controls are essential for reliable results.

What are the latest methodological advances in TDRD12 antibody applications for studying spermatogenesis?

Recent methodological advances have expanded the utility of TDRD12 antibodies in spermatogenesis research:

  • Single-cell approaches:

    • Combining TDRD12 antibodies with single-cell technologies:

      • Single-cell mass cytometry (CyTOF) for multi-parameter analysis

      • Imaging mass cytometry for spatial resolution of TDRD12 expression

      • scRNA-seq paired with protein detection (CITE-seq) to correlate TDRD12 protein with transcriptome profiles

  • Super-resolution microscopy applications:

    • Nanoscale localization of TDRD12 within cellular compartments:

      • STORM or PALM microscopy for precise acrosomal localization

      • Two-color super-resolution to visualize TDRD12 interactions with piRNA pathway components

      • 3D reconstruction of TDRD12 distribution during spermatid development

  • Proximity labeling techniques:

    • Identifying TDRD12 interaction partners in their native cellular context:

      • BioID or TurboID fusion proteins to biotinylate proximal proteins

      • APEX2-based proximity labeling for temporal control

      • Split-BioID for detecting specific protein-protein interactions

  • CRISPR-based approaches:

    • Combining CRISPR technology with antibody-based detection:

      • CRISPR screening followed by TDRD12 immunostaining to identify regulators

      • Endogenous tagging of TDRD12 for live cell imaging

      • Domain-specific mutations to dissect functional regions

  • In situ detection advancements:

    • Multiplexed detection of TDRD12 with other markers:

      • Highly multiplexed immunofluorescence (cyclic immunofluorescence)

      • Combined immunofluorescence and FISH to correlate protein localization with RNA expression

      • Clearing-based 3D imaging of intact testicular tissue with TDRD12 immunolabeling

  • Translational applications in clinical research:

    • Using TDRD12 antibodies in patient samples:

      • Immunohistochemistry panels for diagnosing specific forms of male infertility

      • Correlation of TDRD12 expression patterns with genetic variants

      • Development of non-invasive detection methods for TDRD12-related biomarkers

These methodological advances offer researchers powerful new tools to investigate TDRD12's role in normal and pathological spermatogenesis, potentially leading to improved diagnostics and therapeutic approaches for male infertility.

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.