TTC32 Human

Tetratricopeptide Repeat Domain 32 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Protein Characteristics

  • Genomic Location: Chromosome 2 (2p24.1), spanning ~12 kb .

  • Protein Length: 130 amino acids, with TPR domains mediating interactions in cilia formation and protein complexes .

  • Immunogen Sequence: YEAMDDYTSAIEVQPNFEVPYYNRGLILYRLGYFDDALEDFKKVLDLNPGFQDATLSLKQTILDKEE .

  • Antibody Applications: Validated for immunohistochemistry (1:50–1:200) and immunofluorescence (0.25–2 μg/mL) .

Genetic Associations with Coronary Artery Disease (CAD)

TTC32 is part of the TTC32-WDR35 gene cluster, implicated in CAD through genome-wide association studies (GWAS). Key findings include:

Table 1: Genetic Variants in TTC32-WDR35 and CAD Risk

SNPGenotypeOdds Ratio (OR)Association with CAD RiskPlasma HDL ImpactSource
rs721932CG0.68 (0.54–0.86)Reduced risk (P = .001)↓ HDL (P = .004)
rs12617744AA0.62 (0.42–0.93)Male-specific risk reduction↓ HDL (P = .009)
  • rs721932: Linked to fewer vascular lesions (P = .048) and CAD progression (P = .028) .

  • rs12617744: Associated with left circumflex artery disease (P = .027) and male-specific CAD risk .

DNA Methylation in Diabetes

TTC32 mRNA methylation patterns in diabetic fibroblasts persist despite normoglycemic conditions, suggesting "metabolic memory" linked to wound healing defects .

Expression Quantitative Trait Loci (eQTLs)

SNPs rs2278528, rs7594214, and rs721932 influence TTC32 expression in arterial tissues, indicating regulatory roles in vascular health .

mRNA Expression Profiles

Data from the Human Protein Atlas show variable TTC32 expression across tissues:

Table 2: TTC32 mRNA Expression (Normalized TPM)

TissueMax Subtype nTPMSource
Brain0.6–1.2
Liver0.3–0.7
Kidney0.2–0.5
Spleen0.1–0.3

Clinical and Therapeutic Implications

  • CAD Severity: TTC32-WDR35 variants correlate with HDL levels and vascular lesion counts, suggesting a role in lipid metabolism .

  • Diabetes Complications: Epigenetic changes in TTC32 may contribute to impaired wound healing in diabetic foot ulcers .

  • Ciliary Dysfunction: TPR domain proteins like TTC32 are critical for cilia formation; mutations may link to skeletal disorders .

Product Specs

Introduction
TTC32, a protein containing three tetratricopeptide repeat (TPR) motifs, plays a crucial role in protein-protein interactions. TPR motifs, typically arranged in groups of 3 to 16, create structural scaffolds that facilitate the assembly of multiprotein complexes. Proteins with TPR domains are involved in diverse cellular processes. Examples include cell cycle regulation (e.g., anaphase-promoting complex subunits cdc16, cdc23, and cdc27), immune response (e.g., NADPH oxidase subunit p67-phox and hsp90-binding immunophilins), transcriptional regulation (e.g., transcription factors), protein transport (e.g., peroxisomal import protein PEX5 and mitochondrial import proteins), and protein kinase regulation (e.g., PKR protein kinase inhibitor).
Description
Recombinant human TTC32, expressed in E. coli, is a single polypeptide chain with a molecular weight of 19.7 kDa. This protein comprises 174 amino acids, including the 151 amino acids of TTC32 and a 23 amino acid His-tag fused to the N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
TTC32 is supplied as a 0.5 mg/ml solution in 20mM Tris-HCl buffer (pH 8.0), 0.15M NaCl, 2mM DTT, and 20% glycerol.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Tetratricopeptide Repeat Domain 32, Tetratricopeptide Repeat Protein 32, TPR Repeat Protein 32.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMEGQRQE SHATLTLAQA HFNNGEYAEA EALYSAYIRR CACAASSDES PGSKCSPEDL ATAYNNRGQI KYFRVDFYEA MDDYTSAIEV QPNFEVPYYN RGLILYRLGY FDDALEDFKK VLDLNPGFQD ATLSLKQTIL DKEEKQRRNV AKNY.

Q&A

What is the basic structure and function of TTC32 in humans?

TTC32 (tetratricopeptide repeat domain 32) is a 151 amino acid protein that contains three tetratricopeptide repeat (TPR) motifs. Each TPR motif is a degenerate, 34 amino acid sequence with a characteristic helix-turn-helix shape that stacks with other TPR repeats to achieve specific ligand binding capabilities . The protein's primary function appears to be mediating protein-protein interactions in various cellular pathways, which is consistent with the general role of TPR-containing proteins .

The TTC32 gene is located on human chromosome 2p22.3, a region that makes up part of the second largest human chromosome, comprising approximately 8% of the human genome . The protein has a calculated molecular weight of 17 kDa, though it has been observed to run between 17-30 kDa in experimental conditions, suggesting potential post-translational modifications .

What experimental approaches are commonly used to detect TTC32 protein in human samples?

Several validated experimental approaches are available for detecting TTC32 in human samples:

TechniqueValidated DilutionSample TypesNotes
Western Blot (WB)1:200-1:1000Human, mouseDetects at 17-30 kDa range
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg total proteinMouse testis tissueUseful for protein-protein interaction studies
Immunohistochemistry (IHC)1:50-1:500Mouse testis tissueBest with TE buffer pH 9.0 for antigen retrieval
ELISASample-dependentHuman, mouseRequires optimization per system

For optimal results, antibody titration is recommended in each specific testing system . When performing IHC, antigen retrieval with TE buffer at pH 9.0 is suggested, though citrate buffer at pH 6.0 can serve as an alternative .

In which human tissues is TTC32 predominantly expressed?

Based on available data from multiple expression databases, TTC32 shows variable expression across human tissues. Notable expression has been documented in:

  • Brain tissues - The Allen Brain Atlas indicates differential expression of TTC32 in both adult and developing human brain tissues

  • Testis tissue - Experimental validation of antibodies shows detectable levels in testis tissue, which has been used as a positive control for detection methods

The gene has 2,953 functional associations with biological entities spanning 8 categories extracted from 58 datasets, suggesting its involvement in multiple cellular processes across various tissue types . Expression databases including the Allen Brain Atlas Developing Human Brain Tissue and Prenatal Human Brain Tissue profiles suggest developmental regulation of TTC32 expression .

What methodological approaches should be used to study TTC32 protein-protein interactions?

When investigating TTC32 protein-protein interactions, researchers should employ a multi-method approach:

  • Immunoprecipitation followed by mass spectrometry:

    • Use validated antibodies (0.5-4.0 μg for 1.0-3.0 mg of total protein lysate)

    • Cross-link protein complexes if studying transient interactions

    • Perform LC-MS/MS analysis on eluted proteins to identify binding partners

  • Yeast two-hybrid screening:

    • Create bait constructs using full-length TTC32 and truncation variants to identify domain-specific interactions

    • Screen against human cDNA libraries from tissues with high TTC32 expression

    • Validate candidate interactions using co-immunoprecipitation and co-localization studies

  • Structural characterization of interactions:

    • The helix-turn-helix shape of TPR domains stacks to achieve ligand binding specificity

    • Consider X-ray crystallography or Cryo-EM approaches to determine binding interfaces

    • Mutational analysis of key residues in the TPR motifs can identify critical interaction points

  • Functional validation:

    • siRNA knockdown approaches (commercially available)

    • CRISPR-Cas9 gene editing to create functional mutants

    • Rescue experiments with wild-type vs. mutant protein expression

These methods should be applied iteratively, with initial candidates from screening approaches validated through multiple orthogonal techniques.

How can researchers distinguish between TTC32 isoforms in transcriptome analysis?

Distinguishing between TTC32 isoforms requires a sophisticated approach combining short-read and long-read sequencing technologies:

  • Hybrid sequencing approach:

    • Combine second-generation (short-read) and third-generation (long-read) sequencing, similar to methods used for comprehensive transcriptome characterization

    • Short reads provide high accuracy but cannot reliably identify full-length isoforms

    • Long reads can capture full-length transcripts but have higher error rates

    • Integration of both approaches provides high confidence isoform identification

  • Statistical modeling for isoform reconstruction:

    • Develop models for how different read types sample from underlying isoforms

    • Implement algorithms that can infer isoforms based on these models

    • Combine information from both full-length long reads and partial transcript fragments

    • Utilize the statistical power of short reads (typically an order of magnitude more abundant)

  • Methodological validation:

    • RT-PCR with isoform-specific primers spanning exon junctions

    • Nanopore direct RNA sequencing to confirm full-length isoforms

    • Sanger sequencing of cloned full-length cDNAs

  • Data analysis considerations:

    • Apply error correction to long reads using short read data

    • Map reads to reference genome using splice-aware aligners

    • Employ transcript assembly algorithms that can integrate heterogeneous data types

This hybrid approach significantly improves both sensitivity and specificity in isoform identification compared to methods relying solely on short-read sequencing .

What are the recommended protocols for studying TTC32 gene expression in relation to human disease models?

When studying TTC32 gene expression in human disease contexts, researchers should follow these methodological guidelines:

  • Expression profiling in patient cohorts:

    • Perform RNA-Seq or qPCR analysis on patient-derived samples

    • Compare expression levels across disease stages and in matched controls

    • Consider tissue-specific expression patterns identified in databases such as Allen Brain Atlas

    • Evaluate TTC32 expression in context with related genes in the TPR family

  • Cell line models:

    • Select appropriate cell lines based on CCLE Cell Line Gene Expression Profiles

    • Create knockdown/knockout models using validated siRNAs or CRISPR-Cas9

    • Perform rescue experiments with wild-type and mutant constructs

    • Assess downstream pathway effects using RNA-Seq or proteomics

  • Survival analysis approach:

    • Follow methodologies similar to those used in cancer cohort studies

    • Calculate hazard ratios (HR) to determine prognostic value

    • Perform multivariate analysis to control for confounding factors

    • Validate findings across independent cohorts

  • Functional validation in model systems:

    • Consider using mouse models where TTC32 expression has been validated

    • Design experiments with appropriate statistical power

    • Include both male and female animals to detect sex-specific effects

    • Correlate findings with human data using orthologous gene analysis

When interpreting results, researchers should account for the multiple functional associations TTC32 has across biological pathways and its potential involvement in protein-protein interaction networks characteristic of TPR-containing proteins .

How should researchers optimize antibody-based detection of TTC32 in human samples?

Optimizing antibody-based detection of TTC32 requires careful consideration of several methodological aspects:

  • Western blot optimization:

    • Start with the recommended dilution range (1:200-1:1000) and titrate to determine optimal concentration

    • Include positive control (mouse testis tissue)

    • Expect protein detection between 17-30 kDa range

    • Use gradient gels (10-20%) to ensure optimal separation

    • Consider membrane type (PVDF vs. nitrocellulose) and blocking conditions

    • Test multiple antibodies targeting different epitopes if available

  • Immunohistochemistry protocol refinement:

    • Begin with 1:50-1:500 dilution range

    • Test both recommended antigen retrieval methods:

      • Primary: TE buffer pH 9.0

      • Alternative: Citrate buffer pH 6.0

    • Optimize incubation time and temperature

    • Include positive and negative controls in each experiment

    • Consider automated staining platforms for consistency

  • Immunoprecipitation enhancement:

    • Use 0.5-4.0 μg antibody for 1.0-3.0 mg of total protein lysate

    • Optimize lysis buffer composition to preserve protein-protein interactions

    • Consider crosslinking approaches for transient interactions

    • Pre-clear lysates to reduce non-specific binding

    • Validate results with reverse IP when possible

  • ELISA development:

    • Determine optimal coating concentration

    • Test various blocking agents (BSA, milk, commercial blockers)

    • Perform checkerboard titration of primary and secondary antibodies

    • Validate with recombinant protein standards

    • Include sample dilution curves to ensure linearity

For all methods, researchers should validate antibody specificity using knockdown/knockout controls and consider lot-to-lot variability when ordering new antibody preparations.

What considerations are important when designing experiments to study the role of TTC32 in protein-protein interaction networks?

When designing experiments to investigate TTC32's role in protein-protein interaction networks, researchers should consider:

  • Structural basis for interactions:

    • The TPR motif's helix-turn-helix shape stacks with other TPR repeats to achieve ligand binding specificity

    • TTC32 contains three TPR repeats, which may create a superhelix structure similar to other TPR proteins

    • Design experiments that can probe the structural requirements for binding:

      • Point mutations in key residues

      • Domain deletion constructs

      • Peptide competition assays

  • Network analysis approach:

    • Integrate TTC32 interaction data with existing protein-protein interaction databases

    • Consider the 2,953 functional associations spanning 8 biological categories

    • Use network visualization tools to identify hub proteins and potential pathways

    • Implement clustering algorithms to identify functional modules

  • Tissue-specific interaction dynamics:

    • Design experiments relevant to tissues with validated TTC32 expression:

      • Brain tissues (developing and adult)

      • Testis tissue

    • Consider developmental timing based on expression data from Allen Brain Atlas

    • Compare interaction networks across different cellular contexts

  • Technical considerations:

    • Include appropriate controls for protein-protein interaction studies:

      • Non-specific IgG controls for immunoprecipitation

      • Empty vector controls for overexpression studies

      • Scrambled siRNA for knockdown experiments

    • Address potential issues with protein solubility and preservation of native interactions

    • Consider the impact of tags (His, FLAG, GST) on protein folding and interactions

    • Implement quantitative approaches (SILAC, TMT labeling) for interaction dynamics

  • Validation through orthogonal methods:

    • Confirm key interactions through multiple techniques:

      • Co-immunoprecipitation

      • Proximity ligation assays

      • FRET/BRET approaches

      • In vitro binding assays with purified components

These methodological considerations will help ensure robust and reproducible results when studying TTC32's role in protein-protein interaction networks.

How can researchers integrate TTC32 expression data with broader genomic datasets?

To effectively integrate TTC32 expression data with broader genomic datasets, researchers should implement the following methodological approaches:

  • Multi-omics data integration:

    • Correlate TTC32 expression with:

      • Genomic variations (SNPs, CNVs)

      • Epigenomic features (methylation, histone modifications)

      • Proteomic datasets

      • Metabolomic profiles

    • Implement computational pipelines that can handle heterogeneous data types

    • Consider both linear and non-linear relationships between data types

  • Functional enrichment analysis:

    • Leverage TTC32's 2,953 functional associations across 8 biological categories

    • Perform pathway analysis using databases such as:

      • KEGG

      • Reactome

      • Gene Ontology

    • Identify enriched biological processes, molecular functions, and cellular components

    • Consider tissue-specific pathway databases for context-relevant analysis

  • Co-expression network analysis:

    • Build co-expression networks from RNA-Seq data

    • Identify gene modules that co-express with TTC32

    • Calculate network centrality measures to determine TTC32's position in the network

    • Compare network structures across different tissues and conditions

  • Cross-species comparative approaches:

    • Compare expression patterns and functional associations across model organisms

    • Identify evolutionarily conserved regulatory mechanisms

    • Leverage data from mouse models where TTC32 has been characterized

    • Use orthology mapping to translate findings between species

  • Visualization and exploration tools:

    • Implement interactive visualizations for multi-dimensional data exploration

    • Use dimensionality reduction techniques (PCA, t-SNE, UMAP) for data representation

    • Develop custom R/Python scripts for integrated analysis workflows

    • Consider machine learning approaches for pattern identification

These integration strategies will help researchers place TTC32 expression data within a broader biological context and generate hypotheses about its functional roles in different cellular environments.

What statistical approaches are recommended for analyzing TTC32 expression changes in disease states?

When analyzing TTC32 expression changes in disease states, researchers should employ rigorous statistical methodologies:

  • Differential expression analysis:

    • For RNA-Seq data:

      • Apply appropriate normalization methods (TPM, FPKM, or count-based normalization)

      • Use established packages (DESeq2, edgeR, limma-voom)

      • Control for batch effects and confounding variables

    • For proteomics data:

      • Apply specialized normalization methods for mass spectrometry data

      • Account for missing values appropriately

    • Calculate fold changes, p-values, and adjusted p-values (FDR)

  • Survival analysis methods:

    • Calculate hazard ratios (HR) to assess association with outcomes

    • Example approach: analyzing TTC32 expression in relation to survival outcomes, similar to methods used for other genes (HR calculation with p-value determination)

    • Implement Kaplan-Meier curves for visual representation

    • Conduct multivariate Cox regression to control for covariates

    • Perform stratified analyses based on clinical variables

  • Machine learning approaches:

    • Develop predictive models incorporating TTC32 expression

    • Use cross-validation to assess model performance

    • Compare multiple algorithms (random forests, support vector machines, neural networks)

    • Implement feature selection to identify key variables

  • Meta-analysis techniques:

    • Combine results across multiple independent cohorts

    • Account for between-study heterogeneity

    • Use random-effects models when appropriate

    • Assess publication bias and study quality

  • Power analysis and sample size determination:

    • Calculate minimum sample sizes needed to detect clinically meaningful differences

    • Consider effect sizes observed in preliminary studies

    • Account for multiple testing when determining significance thresholds

    • Implement simulation-based approaches for complex study designs

These statistical approaches will ensure robust, reproducible analysis of TTC32 expression changes in disease contexts, providing a solid foundation for biological interpretation and clinical translation.

What emerging technologies could advance our understanding of TTC32 function in human biology?

Several cutting-edge technologies show promise for advancing our understanding of TTC32 function:

  • Single-cell multi-omics approaches:

    • Single-cell RNA-Seq to map TTC32 expression across cell types

    • Spatial transcriptomics to understand tissue-specific expression patterns

    • CITE-seq for simultaneous protein and RNA quantification

    • Single-cell ATAC-seq to correlate chromatin accessibility with TTC32 expression

    • These approaches would provide unprecedented resolution of TTC32's role in heterogeneous tissues, particularly in brain tissues where expression has been documented

  • Advanced protein structure determination methods:

    • AlphaFold2 and RoseTTAFold for in silico structure prediction

    • Cryo-electron microscopy for visualizing TTC32 protein complexes

    • Hydrogen-deuterium exchange mass spectrometry to map binding interfaces

    • These methods could elucidate how the three TPR repeats in TTC32 form helix-turn-helix structures and achieve ligand binding specificity

  • CRISPR-based functional genomics:

    • CRISPR interference/activation for precise modulation of TTC32 expression

    • Base editing and prime editing for introducing specific mutations

    • CRISPR screens to identify synthetic lethal interactions

    • These approaches would complement existing siRNA methods with greater specificity and versatility

  • Organoid and advanced cell culture systems:

    • Brain organoids to study TTC32 in neurodevelopment

    • Testis organoids to investigate function in reproductive biology

    • Patient-derived organoids to study disease-specific effects

    • These models would provide physiologically relevant systems to study TTC32 in tissues where it shows expression

  • Computational approaches:

    • Network medicine algorithms to place TTC32 in disease modules

    • Deep learning for integrating multi-modal data

    • Advanced molecular dynamics simulations of TPR domain interactions

    • These computational tools could leverage TTC32's extensive functional associations to generate testable hypotheses

By combining these emerging technologies, researchers can develop a comprehensive understanding of TTC32's molecular function, tissue-specific roles, and potential contributions to human disease.

How might understanding TTC32 contribute to translational research applications?

Understanding TTC32 function could contribute to translational research in several key areas:

  • Biomarker development:

    • Expression analysis methods similar to those used in cancer cohorts could identify prognostic or predictive value of TTC32

    • Tissue-specific expression patterns documented in brain and testis tissues suggest potential utility in neurological or reproductive disorders

    • Methodological approach:

      • Validate expression in patient cohorts using qPCR or digital droplet PCR

      • Correlate with clinical outcomes using survival analysis techniques

      • Develop standardized assays for clinical implementation

  • Therapeutic target identification:

    • The protein-protein interaction function of TTC32 through its TPR domains presents opportunities for targeted intervention

    • Structure-based drug design approaches:

      • Virtual screening against binding pockets

      • Fragment-based lead discovery

      • Peptidomimetic design targeting specific interaction surfaces

    • Small molecule modulators of protein-protein interactions

  • Genetic testing applications:

    • TTC32's location on chromosome 2p22.3 places it in a region associated with various genetic disorders

    • Integration with genomic medicine approaches:

      • Include in targeted gene panels for relevant conditions

      • Analyze in whole exome/genome sequencing data

      • Correlate genetic variants with expression changes and clinical phenotypes

  • Disease modeling:

    • Leverage knowledge of TTC32 expression patterns in brain tissues for neurological disease models

    • Methods for functional validation:

      • Patient-derived iPSCs differentiated into relevant cell types

      • CRISPR-engineered cell lines and animal models

      • Organoid systems mimicking tissue-specific environments

  • Precision medicine applications:

    • Patient stratification based on TTC32 expression or genetic variants

    • Tailored therapeutic approaches for specific molecular subtypes

    • Resistance mechanism identification in therapeutic settings

Product Science Overview

Introduction

Tetratricopeptide Repeat Domain 32 (TTC32) is a protein encoded by the TTC32 gene in humans. This protein is part of a larger family of proteins characterized by the presence of tetratricopeptide repeat (TPR) motifs. TPR motifs are structural motifs that consist of a degenerate 34 amino acid tandem repeat, which are found in a wide variety of proteins and play a crucial role in mediating protein-protein interactions .

Structure and Function

The TPR motif is typically composed of a pair of antiparallel alpha helices. These motifs are found in tandem arrays of 3–16 motifs, forming scaffolds that mediate protein-protein interactions and often the assembly of multiprotein complexes . The TPR domain usually folds into a single, linear solenoid structure, which is essential for its function in various cellular processes.

Proteins containing TPR motifs are involved in a wide range of biological functions, including the regulation of the cell cycle, protein folding, and the assembly of protein complexes. For example, TPR motifs are found in the anaphase-promoting complex (APC) subunits, NADPH oxidase subunit p67-phox, hsp90-binding immunophilins, transcription factors, and mitochondrial import proteins .

TTC32 Gene

The TTC32 gene, also known as Tetratricopeptide Repeat Domain 32, is a protein-coding gene. Gene Ontology (GO) annotations related to this gene include identical protein binding . An important paralog of this gene is ST13 . The TTC32 gene is located on chromosome 2 and has several aliases, including TPR Repeat Protein 32 .

Applications and Research

Human recombinant TTC32 is used in various research applications to study protein-protein interactions and the assembly of multiprotein complexes. Understanding the structure and function of TTC32 can provide insights into its role in cellular processes and its potential implications in diseases.

Quick Inquiry

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