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) .
TTC32 is part of the TTC32-WDR35 gene cluster, implicated in CAD through genome-wide association studies (GWAS). Key findings include:
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 .
TTC32 mRNA methylation patterns in diabetic fibroblasts persist despite normoglycemic conditions, suggesting "metabolic memory" linked to wound healing defects .
SNPs rs2278528, rs7594214, and rs721932 influence TTC32 expression in arterial tissues, indicating regulatory roles in vascular health .
Data from the Human Protein Atlas show variable TTC32 expression across tissues:
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 .
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 .
Several validated experimental approaches are available for detecting TTC32 in human samples:
Technique | Validated Dilution | Sample Types | Notes |
---|---|---|---|
Western Blot (WB) | 1:200-1:1000 | Human, mouse | Detects at 17-30 kDa range |
Immunoprecipitation (IP) | 0.5-4.0 μg for 1.0-3.0 mg total protein | Mouse testis tissue | Useful for protein-protein interaction studies |
Immunohistochemistry (IHC) | 1:50-1:500 | Mouse testis tissue | Best with TE buffer pH 9.0 for antigen retrieval |
ELISA | Sample-dependent | Human, mouse | Requires 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 .
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 .
When investigating TTC32 protein-protein interactions, researchers should employ a multi-method approach:
Immunoprecipitation followed by mass spectrometry:
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:
Functional validation:
These methods should be applied iteratively, with initial candidates from screening approaches validated through multiple orthogonal techniques.
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 .
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:
Survival analysis approach:
Functional validation in model systems:
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 .
Optimizing antibody-based detection of TTC32 requires careful consideration of several methodological aspects:
Western blot optimization:
Immunohistochemistry protocol refinement:
Immunoprecipitation enhancement:
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.
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:
Technical considerations:
Include appropriate controls for protein-protein interaction studies:
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.
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:
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.
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.
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:
Organoid and advanced cell culture systems:
Computational approaches:
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.
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:
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
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 .
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 .
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 .
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.