TCF7L1 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the shipping method and destination. Please consult your local distributors for specific delivery times.
Synonyms
bHLHb21 antibody; HMG box transcription factor 3 antibody; HMG box transcription factor antibody; OTTMUSP00000023419 antibody; T cell factor 3 antibody; TCF 3 antibody; TCF-3 antibody; TCF3 antibody; Tcf7l1 antibody; TF7L1_HUMAN antibody; Transcription factor 7 like 1 (T-cell specific, HMG-box) antibody; Transcription factor 7 like 1 antibody; Transcription factor 7-like 1 antibody
Target Names
Uniprot No.

Target Background

Function
TCF7L1 is a transcription factor that participates in the Wnt signaling pathway. It binds to DNA and acts as a repressor in the absence of β-catenin (CTNNB1), and as an activator in its presence. TCF7L1 is crucial for the terminal differentiation of epidermal cells, the formation of keratohyalin granules, and the development of the epidermis's barrier function. It also down-regulates NQO1, leading to increased resistance to mitomycin C.
Gene References Into Functions
  1. HIPK2 up-regulates transcription by phosphorylating TCF3, a transcriptional repressor, but inhibits transcription by phosphorylating LEF1, a transcriptional activator. PMID: 21285352
  2. TCF3 is partially responsible for the butyrate-resistant phenotype of colorectal cancer cells, as this DNA-binding protein suppresses the hyperinduction of Wnt activity by butyrate. PMID: 23063976
  3. Research findings identify TCF3 as a key regulator of tumor growth and initiation. PMID: 23090119
  4. TCF3, a novel positive regulator of osteogenesis, plays a critical role in miR-17 modulating the diverse effect of canonical Wnt signaling in different microenvironments. PMID: 23492770
  5. The TCF/TLE tetramer complex promotes structural transitions of chromatin to mediate repression. PMID: 24596249
  6. These results indicate that a dynamic interplay of TCF transcription factors governs MYC gene expression in colorectal cancers. PMID: 25659031
  7. TCF7L1, a Wnt pathway repressor, buffers CTNNB1/TCF target gene expression to promote CRC growth PMID: 27333864
  8. TCF7L1 plays a significant role in maintaining hESC pluripotency, which has implications for human development during gastrulation PMID: 29361574
  9. Through transcriptome profiling and combined gain- and loss-of-function studies, researchers identified LCN2 as a major downstream effector of TCF7L1 that drives tumor growth. PMID: 28467300
  10. Researchers report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects PMID: 26764381
  11. No difference in nuclear beta-catenin signal intensity was found, which may be caused by an alteration in the Wnt pathway in microsatellite stable sporadic tumors by unknown mechanisms leading to lower TCF-3, 4 protein expression. PMID: 20532534

Show More

Hide All

Database Links

HGNC: 11640

OMIM: 604652

KEGG: hsa:83439

STRING: 9606.ENSP00000282111

UniGene: Hs.516297

Protein Families
TCF/LEF family
Subcellular Location
Nucleus.
Tissue Specificity
Detected in hair follicles and skin keratinocytes, and at lower levels in stomach epithelium.

Q&A

What is TCF7L1 and what cellular functions does it regulate?

TCF7L1 (Transcription factor 7-like 1) is a protein encoded by the TCF7L1 gene in humans, also known as TCF-3, TCF3, or HMG box transcription factor 3. This protein participates as a key mediator in the Wnt signaling pathway, functioning as a DNA-binding transcriptional regulator. TCF7L1 acts primarily as a repressor in the absence of β-catenin (CTNNB1) and converts to an activator when β-catenin is present . The protein is approximately 62.6 kilodaltons in mass and has orthologs in several species including canine, porcine, monkey, mouse, and rat .

From a functional perspective, TCF7L1 is necessary for critical developmental processes including:

  • Terminal differentiation of epidermal cells

  • Formation of keratohyalin granules

  • Development of barrier function in the epidermis

  • Down-regulation of NQO1, leading to increased mitomycin c resistance

Recent research has identified TCF7L1 as an essential factor in hypothalamo-pituitary axis development, where it functions as a transcriptional repressor regulating hypothalamic signals involved in pituitary formation .

What does HRP conjugation mean in the context of TCF7L1 antibodies and how does it enhance experimental applications?

HRP (Horseradish Peroxidase) conjugation refers to the covalent attachment of the enzyme horseradish peroxidase to an antibody targeting TCF7L1. This conjugation provides a direct detection system through enzymatic activity rather than requiring secondary antibody binding. When HRP encounters its substrate (typically TMB, DAB, or luminol-based reagents), it catalyzes a reaction producing colorimetric, chemiluminescent, or fluorescent signals.

The HRP conjugation offers several methodological advantages:

  • Eliminates the need for secondary antibody incubation, reducing experimental time and potential cross-reactivity

  • Provides enhanced sensitivity for detection of low-abundance proteins

  • Enables direct quantification through enzymatic signal amplification

  • Reduces background noise in assays like ELISA by eliminating non-specific binding from secondary antibodies

For TCF7L1 research specifically, HRP-conjugated antibodies (such as product code CSB-PA884626OB01HU) are optimized for ELISA applications where direct detection provides cleaner results for quantitative analysis of TCF7L1 expression levels .

What are the optimal conditions for using TCF7L1 Antibody, HRP conjugated in ELISA experiments?

When utilizing TCF7L1 Antibody, HRP conjugated (such as CSB-PA884626OB01HU) for ELISA applications, the following optimized protocol parameters are recommended:

Sample Preparation:

  • For cell/tissue lysates: Extract proteins using RIPA buffer supplemented with protease inhibitors

  • For serum/plasma: Dilute 1:5 to 1:20 in blocking buffer to minimize matrix effects

ELISA Protocol Optimization:

  • Coating: Use 1-5 μg/ml of capture antibody in carbonate buffer (pH 9.6) overnight at 4°C

  • Blocking: 2-3% BSA in PBS for 1-2 hours at room temperature

  • Sample incubation: 100 μl/well, 1-2 hours at room temperature or overnight at 4°C

  • TCF7L1 Antibody, HRP conjugated: Dilute 1:500 to 1:2000 in blocking buffer (optimal dilution should be determined empirically)

  • Substrate development: TMB substrate for 15-30 minutes at room temperature

  • Stop reaction: 2M H₂SO₄

  • Read absorbance at 450 nm with 570 nm reference wavelength

Optimization Table for TCF7L1 Antibody, HRP Conjugated ELISA:

ParameterStandard ConditionOptimization RangeNotes
Antibody Dilution1:10001:500 - 1:5000Higher concentrations for low-abundance samples
Incubation Time1 hour30 min - overnightLonger incubation may improve sensitivity
Incubation TemperatureRoom temp.4°C - 37°CLower temperature reduces background
Wash BufferPBS + 0.05% Tween-200.01% - 0.1% Tween-20Higher detergent reduces non-specific binding
Substrate Incubation15 min5 - 30 minMonitor to prevent signal saturation

The working dilution and optimal conditions should be determined for each specific experimental setup to ensure reproducible results.

How can I ensure specificity and reduce background when using TCF7L1 Antibody, HRP conjugated in complex tissue samples?

When working with complex tissue samples, ensuring specificity and reducing background with TCF7L1 Antibody, HRP conjugated requires methodological adjustments:

Pre-analytical considerations:

  • Tissue preparation: Ensure complete homogenization and adequate protein extraction

  • Pre-clearing: Incubate lysates with protein A/G beads to remove non-specifically binding proteins

  • Pre-absorption: Consider pre-absorbing the antibody with recombinant TCF7L1 protein to confirm specificity

Experimental modifications:

  • Blocking optimization: Use 5% non-fat dry milk or 3-5% BSA with 0.1-0.3% Triton X-100 to reduce non-specific binding

  • Additional washing steps: Increase wash frequency (5-7 times) and duration (5 minutes each)

  • Signal amplification control: Include gradient dilutions of recombinant TCF7L1 protein as a standard curve

Validation approaches:

  • Knockout/knockdown controls: Include TCF7L1-depleted samples as negative controls

  • Peptide competition: Pre-incubate antibody with immunizing peptide to confirm binding specificity

  • Cross-validation: Compare results with unconjugated TCF7L1 antibodies (CSB-PA884626OA01HU) using a secondary detection system

For highly complex samples like brain tissue where TCF7L1 plays a role in hypothalamo-pituitary axis development, adding 0.1% SDS to your wash buffer can help reduce non-specific interactions while maintaining specific binding .

What are the most common technical issues when using TCF7L1 Antibody, HRP conjugated, and how can they be addressed?

Researchers commonly encounter several technical challenges when working with TCF7L1 Antibody, HRP conjugated. Here's a systematic approach to identifying and resolving these issues:

Problem: Weak or No Signal

  • Possible causes:

    • Insufficient antigen in sample

    • Antibody degradation

    • Suboptimal antibody concentration

    • Inactive HRP enzyme

  • Solutions:

    • Verify TCF7L1 expression in samples via alternative methods (qPCR)

    • Perform Bradford/BCA assay to ensure adequate protein concentration

    • Use freshly prepared antibody dilutions and avoid freeze-thaw cycles

    • Test antibody activity using positive control samples (e.g., samples from tissues known to express TCF7L1)

    • Increase antibody concentration or extend incubation time

Problem: High Background Signal

  • Possible causes:

    • Insufficient blocking

    • Cross-reactivity with related proteins

    • Excessive antibody concentration

    • Inadequate washing

  • Solutions:

    • Optimize blocking by testing different agents (milk, BSA, serum) and concentrations

    • Increase washing frequency and duration

    • Titrate antibody to determine optimal working concentration

    • Add 0.1-0.5% Tween-20 to wash buffer to reduce non-specific binding

Problem: Inconsistent Results

  • Possible causes:

    • Batch-to-batch variation

    • Sample degradation

    • Inconsistent experimental conditions

  • Solutions:

    • Use the same lot number for all experiments when possible

    • Include internal controls in each experiment

    • Standardize all protocol steps including incubation times and temperatures

    • Document and maintain consistent laboratory conditions

How should I interpret variations in TCF7L1 detection between different experimental systems?

Variations in TCF7L1 detection across experimental systems require careful interpretation and methodological consideration. These variations may reflect biological differences rather than technical issues.

Cross-system analysis framework:

  • Expression level differences:

    • TCF7L1 expression varies significantly across tissue types and developmental stages

    • Hypothalamic and pituitary tissues show distinct expression patterns compared to epidermal tissues

    • Document relative expression levels normalized to appropriate housekeeping genes

  • Post-translational modifications:

    • TCF7L1 undergoes phosphorylation and other modifications that affect antibody recognition

    • Consider using phosphorylation-specific antibodies for comparative analyses

    • Different cell types may process TCF7L1 differently, affecting epitope availability

  • Protein interactions:

    • TCF7L1 functions differ depending on β-catenin interaction status

    • Binding partners may mask antibody epitopes in tissue-specific manner

    • Consider using denaturation or epitope retrieval methods for consistent detection

  • Interpretation guidelines:

    • Always include positive controls from tissues known to express TCF7L1

    • Use multiple antibodies targeting different epitopes to confirm results

    • Consider the specific isoforms present in your experimental system

  • Variant analysis:

    • Genetic variants like p.R92P and p.R400Q can affect antibody binding

    • These variants show reduced repressing activity compared to wild-type TCF7L1

    • Consider sequencing TCF7L1 in your experimental system to identify potential variants

When comparing results between different systems (e.g., in vitro cell cultures vs. tissue samples), differences in detection may reflect physiologically relevant modulation of TCF7L1 activity rather than technical issues.

How can TCF7L1 Antibody, HRP conjugated be used to investigate Wnt signaling pathway dynamics in development and disease?

TCF7L1 Antibody, HRP conjugated provides a valuable tool for dissecting the complex dynamics of the Wnt signaling pathway in developmental processes and disease states. Here are advanced methodological approaches:

Developmental studies:

  • Temporal profiling of TCF7L1 repressor activity:

    • Quantify TCF7L1 levels across developmental time points using standardized ELISA protocols

    • Correlate with β-catenin localization to determine repressor-to-activator transition points

    • Map developmental switches in gene expression programs controlled by TCF7L1

  • Tissue-specific Wnt pathway modulation:

    • Compare TCF7L1 levels between tissues with active vs. inactive Wnt signaling

    • Identify tissue-specific co-factors through co-immunoprecipitation followed by mass spectrometry

    • Analyze how TCF7L1 levels correlate with differentiation markers in developing tissues

Disease-related applications:

  • Cancer research:

    • Quantify TCF7L1/β-catenin ratios in tumor samples vs. normal tissues

    • Correlate TCF7L1 levels with tumor progression and treatment response

    • Monitor changes in TCF7L1 expression during epithelial-mesenchymal transition

  • Developmental disorders:

    • Screen patient samples with hypothalamo-pituitary axis defects for TCF7L1 expression abnormalities

    • Evaluate how TCF7L1 variants (p.R92P and p.R400Q) affect protein function

    • Develop diagnostic algorithms based on TCF7L1 expression patterns

Advanced experimental designs:

  • ChIP-seq integration:

    • Use TCF7L1 Antibody to perform chromatin immunoprecipitation

    • Identify genome-wide binding sites and correlate with repression/activation patterns

    • Integrate with β-catenin binding data to map pathway dynamics

  • Single-cell analysis:

    • Develop protocols for detecting TCF7L1 in fixed cells using flow cytometry

    • Correlate with other Wnt pathway components at single-cell resolution

    • Track cell fate decisions in real-time based on TCF7L1 levels

These approaches allow researchers to move beyond simple detection and quantification into mechanistic understanding of TCF7L1's role in development and disease.

What are the methodological considerations for investigating TCF7L1 variants associated with congenital hypopituitarism?

Investigating TCF7L1 variants associated with congenital hypopituitarism requires specialized methodological approaches to characterize functional impacts. Recent research has identified two significant missense variants (p.R92P and p.R400Q) in human TCF7L1 that exhibit reduced repressing activity compared to wild-type TCF7L1 .

Experimental design considerations:

  • Variant-specific antibody validation:

    • Test TCF7L1 Antibody, HRP conjugated for equivalent binding to wild-type and variant proteins

    • Establish standard curves for each variant to ensure accurate quantification

    • Consider epitope location relative to variant positions (R92P and R400Q)

  • Functional characterization protocol:

    • Repressor activity assay:

      • Utilize dual-luciferase reporter systems with TCF/LEF binding sites

      • Compare repression levels between wild-type and variant TCF7L1

      • Quantify relative repression as percentage of wild-type activity

    • DNA binding analysis:

      • Perform electrophoretic mobility shift assays (EMSA) with purified proteins

      • Determine binding affinity constants for target DNA sequences

      • Assess competition with β-catenin for DNA binding sites

  • In vivo rescue experiments:

    • Inject mRNA encoding wild-type or variant TCF7L1 into zebrafish models

    • Assess phenotypic rescue in tcf7l1a/tcf7l1b-deficient zebrafish

    • Quantify developmental outcomes using standardized scoring systems

Data analysis framework:

ParameterWild-type TCF7L1p.R92P Variantp.R400Q VariantAnalytical Method
Repressor Activity100% (reference)Reduced (~60-80%)Reduced (~40-70%)Luciferase assay
DNA BindingHigh affinityAltered specificityReduced affinityEMSA, ChIP-seq
Protein StabilityStandard half-lifeMay be alteredMay be alteredPulse-chase analysis
In vivo FunctionNormal HP axis developmentPartial rescuePartial rescueZebrafish model

When designing studies to investigate these variants, it's crucial to include both positive controls (wild-type protein) and negative controls (known non-functional mutants) to calibrate the sensitivity of your detection systems .

How can I combine TCF7L1 Antibody, HRP conjugated with other molecular techniques to comprehensively study Wnt pathway regulation?

Integrating TCF7L1 Antibody, HRP conjugated with complementary molecular techniques creates a powerful approach for comprehensive analysis of Wnt pathway regulation. Here's a methodological framework for such integration:

Transcriptional regulation analysis:

  • ChIP-seq/ChIP-qPCR integration:

    • Use TCF7L1 Antibody for chromatin immunoprecipitation

    • Follow with next-generation sequencing or qPCR of target promoters

    • Compare TCF7L1 binding profiles in different activation states

    • Correlate with histone modifications (H3K27ac, H3K4me3, H3K27me3)

  • RNA-seq correlation:

    • Perform RNA-seq after TCF7L1 knockdown/overexpression

    • Correlate changes in gene expression with TCF7L1 binding sites

    • Identify direct vs. indirect transcriptional targets

    • Validate key targets using reporter assays

Protein interaction networks:

  • Co-immunoprecipitation (Co-IP):

    • Use TCF7L1 Antibody to pull down protein complexes

    • Identify interaction partners by mass spectrometry

    • Validate key interactions with reverse Co-IP

    • Map interaction dynamics during Wnt pathway activation/inhibition

  • Proximity ligation assay (PLA):

    • Combine TCF7L1 Antibody with antibodies against potential partners

    • Visualize and quantify interactions in situ

    • Track spatial dynamics of interactions during development

Functional analysis integration:

  • CRISPR-based approaches:

    • Generate TCF7L1 knockout/knockin cell lines

    • Perform phenotypic rescue with wild-type or variant TCF7L1

    • Quantify rescue efficiency using TCF7L1 Antibody, HRP conjugated

    • Correlate functional outcomes with pathway regulation

  • Live-cell imaging:

    • Develop protocols to track TCF7L1 binding dynamics in real-time

    • Correlate with β-catenin nuclear translocation

    • Measure temporal aspects of target gene activation

These integrated approaches allow researchers to move beyond static measurements to understand the dynamic regulation of Wnt signaling through TCF7L1.

What are the critical controls and validation steps needed when using TCF7L1 Antibody, HRP conjugated in multiplexed assays?

When incorporating TCF7L1 Antibody, HRP conjugated into multiplexed assays, rigorous controls and validation steps are essential to ensure data integrity and interpretability. Here is a comprehensive validation framework:

Essential controls for multiplexed assays:

  • Antibody specificity validation:

    • Knockout/knockdown controls: Test antibody in TCF7L1-null or knockdown samples

    • Peptide competition: Pre-incubate antibody with immunizing peptide

    • Cross-reactivity assessment: Test against related family members (TCF7L2, TCF7)

    • Isotype control: Include matched isotype IgG as negative control

  • Signal validation:

    • Signal-to-noise ratio determination: Calculate minimum detection threshold

    • Dynamic range analysis: Create standard curves with recombinant TCF7L1

    • Cross-platform validation: Compare results with alternative methods (Western blot, immunofluorescence)

    • Inter-assay calibrators: Include reference standards across all experiments

  • Multiplexing-specific controls:

    • Spectral overlap correction: Perform single-color controls to establish compensation matrices

    • Blocking verification: Test for potential cross-reactivity between detection systems

    • Sequential detection: Validate that detection order doesn't affect signal intensity

    • Antibody interference testing: Ensure antibodies don't sterically hinder each other's binding

Validation protocol for TCF7L1 Antibody in multiplexed assays:

Validation StepMethodAcceptance CriteriaTroubleshooting
SpecificityWestern blot comparisonSingle band at 62.6 kDaOptimize antibody dilution
SensitivityTitration seriesDetection limit < 10 ng/mlIncrease incubation time
Linear rangeStandard curveR² > 0.98 over 2 logsAdjust working concentration
ReproducibilityCV calculationCV < 15% between replicatesStandardize protocol steps
Multiplexing compatibilitySequential vs. simultaneous< 20% signal differenceAdjust antibody concentration

Analytical considerations:

  • Data normalization: Use appropriate housekeeping proteins as internal controls

  • Statistical analysis: Apply methods that account for inter-assay variation

  • Batch effects: Include control samples across experimental batches

  • Quantitative reporting: Report results relative to standard curves rather than raw signal

Implementing these validation steps ensures reliable and interpretable results when using TCF7L1 Antibody, HRP conjugated in complex multiplexed experimental designs.

How can TCF7L1 Antibody, HRP conjugated be utilized to investigate pathological conditions associated with hypothalamo-pituitary axis dysfunction?

TCF7L1 Antibody, HRP conjugated offers valuable opportunities for investigating pathological conditions linked to hypothalamo-pituitary (HP) axis dysfunction. Recent research has established TCF7L1 as a critical determinant in HP axis development, making it a promising target for studying related disorders .

Methodological approach for clinical specimens:

  • Patient sample analysis:

    • Screening protocol: Develop standardized ELISA for TCF7L1 quantification in patient samples

    • Sample types: Analyze expression in pituitary tissue biopsies, cerebrospinal fluid, or blood

    • Reference ranges: Establish normal vs. pathological TCF7L1 levels in different age groups

    • Variant detection: Couple with genetic screening for TCF7L1 variants (p.R92P, p.R400Q, etc.)

  • Genotype-phenotype correlation:

    • Clinical parameters: Correlate TCF7L1 levels with endocrine function tests

    • Imaging correlation: Link expression levels with MRI findings of pituitary abnormalities

    • Growth metrics: Associate TCF7L1 variant status with growth parameters in children

    • Treatment response: Monitor changes in TCF7L1 activity during hormone replacement therapy

Experimental disease models:

  • In vitro models:

    • Generate patient-derived induced pluripotent stem cells (iPSCs)

    • Differentiate into hypothalamic and pituitary lineages

    • Compare TCF7L1 expression and function between patient and control cells

    • Test response to Wnt pathway modulators

  • Animal models:

    • Analyze tcf7l1a/tcf7l1b-deficient zebrafish for HP axis phenotypes

    • Generate conditional TCF7L1 knockout mice specific to hypothalamic/pituitary tissues

    • Perform rescue experiments with wild-type vs. variant TCF7L1

    • Monitor developmental milestones and endocrine parameters

Integration with clinical data:

Clinical ParameterMeasurement MethodCorrelation with TCF7L1Analysis Approach
Growth hormone deficiencyStimulation testInverse correlationRegression analysis
Pituitary morphologyMRI volumetricsSize correlates with expression3D reconstruction
Multiple hormone deficiencyHormone panelVariant-specific patternsCluster analysis
Development timingGrowth chartsDelayed milestonesLongitudinal tracking

This integrated approach allows researchers to establish mechanistic links between TCF7L1 dysfunction and clinical manifestations of HP axis disorders .

What are the methodological considerations for using TCF7L1 Antibody, HRP conjugated in high-throughput screening for modulators of Wnt signaling?

Using TCF7L1 Antibody, HRP conjugated in high-throughput screening (HTS) to identify modulators of Wnt signaling requires careful methodological considerations to ensure robust and reproducible results. Here is a comprehensive approach:

Assay development and optimization:

  • Miniaturization protocol:

    • Adapt standard ELISA protocols to 384- or 1536-well formats

    • Optimize reagent volumes (10-25 μl per well)

    • Validate Z-factor (>0.5) to ensure assay robustness

    • Establish automated liquid handling parameters

  • Signal detection optimization:

    • Select appropriate HRP substrates for HTS (chemiluminescent preferred)

    • Establish signal stability window (30-60 minutes optimal)

    • Determine minimal detection threshold and upper limit of linearity

    • Optimize plate reader settings (integration time, gain)

  • Controls and normalization:

    • Include positive controls (known Wnt activators like CHIR99021)

    • Include negative controls (Wnt inhibitors like IWP-2)

    • Include neutral controls (DMSO vehicle)

    • Develop robust normalization algorithms to adjust for plate effects

Screening workflow design:

  • Primary screen:

    • Measure TCF7L1 levels after compound treatment

    • Alternative: measure TCF7L1 repressor activity using reporter systems

    • Use single-concentration screening (typically 10 μM)

    • Set hit criteria (typically >3 standard deviations from control mean)

  • Confirmation and dose-response:

    • Retest hits in triplicate

    • Perform 8-10 point dose-response curves

    • Calculate EC50/IC50 values

    • Eliminate compounds with poor curve fits

  • Secondary assays:

    • Confirm Wnt pathway modulation using orthogonal assays

    • Assess effects on TCF7L1-DNA binding

    • Evaluate changes in TCF7L1-β-catenin interaction

    • Test effects on downstream target genes

Data analysis framework:

Analysis StageMethodsMetricsSuccess Criteria
Quality ControlPlate uniformityCV < 20%Pass ≥90% of plates
Hit Selection%inhibition or %activationZ-score ≥3Hit rate 0.1-1%
Dose Response4-parameter logistic fitR² > 0.9EC50 in nM-μM range
Counter-screenCell viability assay>80% viabilityExclude cytotoxic compounds

Implementation considerations:

  • Batch preparation of TCF7L1 Antibody, HRP conjugated to minimize lot-to-lot variation

  • Automated liquid handling systems to ensure consistency

  • Barcoded plates for tracking and data management

  • Integrated data analysis pipeline for rapid hit identification

This methodological framework enables efficient identification of compounds that modulate TCF7L1 function in the Wnt signaling pathway, potentially leading to novel therapeutics for developmental disorders and cancer.

How might emerging technologies enhance the utility of TCF7L1 Antibody, HRP conjugated for studying transcriptional regulation mechanisms?

Emerging technologies present exciting opportunities to enhance the utility of TCF7L1 Antibody, HRP conjugated for investigating transcriptional regulation mechanisms. These advancements can provide unprecedented insights into TCF7L1 function in both normal development and disease states.

Single-cell technologies:

  • Single-cell proteomics:

    • Adapt TCF7L1 Antibody, HRP conjugated for mass cytometry (CyTOF)

    • Develop protocols for single-cell western blotting

    • Correlate TCF7L1 levels with cell state markers at single-cell resolution

    • Map heterogeneity in TCF7L1 expression across developmental gradients

  • Spatial transcriptomics integration:

    • Combine TCF7L1 protein detection with spatial transcriptomics

    • Map spatial relationships between TCF7L1 activity and target gene expression

    • Identify tissue microenvironments with active Wnt signaling

    • Correlate with developmental boundaries and signaling gradients

Advanced imaging approaches:

  • Super-resolution microscopy:

    • Adapt TCF7L1 Antibody for STORM or PALM imaging

    • Resolve sub-nuclear localization of TCF7L1 binding sites

    • Track dynamics of TCF7L1-β-catenin interactions at nanometer scale

    • Visualize changes in chromatin organization at TCF7L1 binding sites

  • Live-cell imaging technologies:

    • Develop non-perturbing methods to track TCF7L1 binding in living cells

    • Monitor real-time changes in TCF7L1 localization during Wnt activation

    • Correlate with dynamic changes in target gene expression

    • Measure binding kinetics at individual genomic loci

Integrative multi-omics approaches:

  • Proteogenomic integration:

    • Correlate TCF7L1 binding sites (ChIP-seq) with proteome changes

    • Map post-translational modifications affecting TCF7L1 function

    • Identify protein interaction networks specific to developmental contexts

    • Link genetic variants to altered TCF7L1 function

  • Systems biology modeling:

    • Develop quantitative models of TCF7L1 activity in Wnt signaling

    • Simulate effects of TCF7L1 variants on signaling dynamics

    • Predict compensatory mechanisms in TCF7L1 dysfunction

    • Model therapeutic interventions to restore normal signaling

These emerging technologies will transform our understanding of TCF7L1's role in transcriptional regulation, providing both basic biological insights and potential therapeutic targets for developmental disorders.

What are the potential applications of TCF7L1 Antibody, HRP conjugated in studying the molecular basis of congenital hypopituitarism and related disorders?

The discovery of TCF7L1's critical role in hypothalamo-pituitary axis development opens new avenues for investigating congenital hypopituitarism and related disorders. TCF7L1 Antibody, HRP conjugated can serve as a valuable tool in these investigations through several innovative applications.

Diagnostic and screening applications:

  • Patient stratification protocol:

    • Develop standardized ELISA to quantify TCF7L1 in patient samples

    • Create diagnostic algorithms incorporating TCF7L1 levels and clinical parameters

    • Establish cutoff values for different forms of congenital hypopituitarism

    • Correlate TCF7L1 function with severity of hormonal deficiencies

  • Variant-specific detection:

    • Generate antibodies specific to common TCF7L1 variants (p.R92P, p.R400Q)

    • Develop immunoassays to distinguish variant proteins from wild-type

    • Create multiplexed panels to detect multiple variants simultaneously

    • Correlate variant protein expression with functional outcomes

Mechanistic research applications:

  • Developmental pathway mapping:

    • Track TCF7L1 expression during critical windows of HP axis development

    • Identify co-factors that modulate TCF7L1 function in hypothalamic tissues

    • Map signaling cascades downstream of TCF7L1 in pituitary progenitors

    • Characterize epigenetic changes associated with TCF7L1 binding

  • Disease modeling:

    • Generate patient-derived organoids of hypothalamic and pituitary tissues

    • Quantify TCF7L1 expression and activity in these 3D models

    • Test pharmaceutical interventions to rescue TCF7L1 function

    • Validate findings in animal models of congenital hypopituitarism

Therapeutic development framework:

  • Target identification:

    • Screen for proteins that modulate TCF7L1 activity in HP axis development

    • Identify druggable nodes in TCF7L1-regulated pathways

    • Map TCF7L1 interaction partners as potential therapeutic targets

    • Characterize tissue-specific regulators of TCF7L1 expression

  • Functional restoration strategies:

    • Develop approaches to restore wild-type activity to variant TCF7L1

    • Screen for compounds that stabilize TCF7L1-DNA interactions

    • Test gene therapy approaches to correct TCF7L1 mutations

    • Evaluate cell-based therapies for severe deficiency cases

Research impact assessment:

Research DomainCurrent StatusFuture PotentialMethodological Approach
Genetic diagnosisLimited variant knowledgeComprehensive variant databaseHigh-throughput functional screening
Disease mechanismsBasic understandingDetailed molecular pathwaysIntegrative multi-omics
Therapeutic targetsFew identifiedMultiple intervention pointsSystems biology modeling
Patient stratificationClinical presentation onlyMolecular classificationMachine learning algorithms

The application of TCF7L1 Antibody, HRP conjugated in these research directions can significantly advance our understanding of congenital hypopituitarism and ultimately lead to improved diagnostic and therapeutic approaches for affected patients .

What are the key considerations researchers should remember when selecting and using TCF7L1 Antibody, HRP conjugated for their studies?

Researchers planning to incorporate TCF7L1 Antibody, HRP conjugated into their experimental workflows should consider several critical factors to ensure optimal results:

Selection criteria:

  • Epitope specificity: Choose antibodies targeting epitopes away from known variant sites (p.R92P, p.R400Q) unless specifically studying these variants

  • Validation documentation: Select products with comprehensive validation data (Western blot, immunoprecipitation, ELISA)

  • Species reactivity: Ensure compatibility with your experimental model (human, mouse, etc.)

  • HRP conjugation quality: Verify enzyme activity and conjugation stability data

  • Lot-to-lot consistency: Request information on quality control measures between production lots

Experimental design considerations:

  • Appropriate controls: Include positive controls (samples known to express TCF7L1), negative controls (TCF7L1-null samples), and isotype controls

  • Protocol optimization: Develop standardized protocols with empirically determined antibody concentrations, incubation times, and detection parameters

  • Cross-validation: Validate key findings using alternative detection methods or antibodies targeting different epitopes

  • Storage and handling: Follow manufacturer recommendations for storage conditions, avoid freeze-thaw cycles, and prepare fresh working dilutions

Data interpretation framework:

  • Signal specificity: Verify that detected signals correspond to the expected molecular weight (approximately 62.6 kDa for TCF7L1)

  • Quantitative analysis: Establish standard curves using recombinant TCF7L1 protein for accurate quantification

  • Biological context: Interpret results in the context of Wnt signaling status and developmental stage

  • Statistical rigor: Apply appropriate statistical methods with adequate sample sizes to ensure reproducibility

By carefully considering these factors, researchers can maximize the utility of TCF7L1 Antibody, HRP conjugated in their studies of development, disease, and signaling pathways.

How can researchers integrate findings from TCF7L1 studies into broader understanding of developmental disorders and potential therapeutic approaches?

Integrating findings from TCF7L1 studies into a broader understanding of developmental disorders requires a multidisciplinary approach that connects molecular mechanisms to clinical manifestations and therapeutic opportunities. Here's a framework for such integration:

Translational research pathway:

  • From molecular mechanism to phenotype:

    • Map the effects of TCF7L1 dysfunction on gene regulatory networks

    • Connect disrupted pathways to cellular behaviors (proliferation, differentiation, migration)

    • Link cellular changes to tissue-level abnormalities

    • Correlate tissue defects with clinical manifestations

  • Clinical correlation framework:

    • Develop standardized protocols for TCF7L1 assessment in patient samples

    • Create comprehensive databases linking TCF7L1 variants to clinical phenotypes

    • Establish international registries for rare TCF7L1-associated disorders

    • Generate predictive models for disease progression based on molecular data

  • Therapeutic development pipeline:

    • Target identification:

      • Map druggable nodes in TCF7L1-regulated pathways

      • Identify compensatory mechanisms that could be therapeutically enhanced

      • Develop screening platforms for compounds affecting TCF7L1 function

      • Prioritize targets based on tissue specificity and safety profiles

    • Precision medicine approaches:

      • Classify patients based on TCF7L1 variant functional consequences

      • Develop variant-specific therapeutic strategies

      • Design combination therapies targeting multiple pathway components

      • Establish biomarkers for treatment response monitoring

Integration with other developmental pathways:

PathwayIntersection with TCF7L1Research ApproachTherapeutic Potential
Sonic HedgehogCross-talk during forebrain developmentDual pathway modulationCombined targeting
NotchCooperative effects on progenitor maintenanceSynthetic genetic screeningSynergistic intervention
BMP/TGF-βAntagonistic interactions in patterningPathway balance restorationCompensatory modulation
FGFCoordinate regulation of HP axis developmentIntegrated signaling analysisSequential targeting

Implementation in clinical research:

  • Biomarker development: Establish TCF7L1 as a diagnostic and prognostic marker for developmental disorders

  • Clinical trial design: Stratify patients based on TCF7L1 status for targeted interventions

  • Therapeutic monitoring: Use TCF7L1 activity as a surrogate endpoint for treatment efficacy

  • Preventive strategies: Identify at-risk populations through TCF7L1 screening

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.