TGFBI (Transforming Growth Factor Beta Induced) is a 68 kDa extracellular matrix (ECM) protein encoded by the TGFBI gene located on human chromosome 5q31 . It is induced by TGF-β signaling and plays critical roles in cell adhesion, migration, and collagen binding through its RGD (Arg-Gly-Asp) motif . Primarily expressed in the cornea, TGFBI is also found in tissues such as bone, cartilage, and skin . Mutations in TGFBI are strongly associated with hereditary corneal dystrophies, while dysregulated expression correlates with cancer progression and immune modulation .
Over 60 TGFBI mutations are linked to corneal dystrophies, characterized by progressive vision loss due to amyloid or non-amyloid deposits . Common mutations include:
These mutations disrupt TGFBI folding, leading to pathogenic aggregation in the cornea .
TGFBI expression varies across cancers, influencing prognosis and therapy response:
In ovarian cancer, TGFBI loss due to promoter hypermethylation is linked to chemoresistance . Conversely, TGFBI secreted by peritoneal cells enhances ovarian cancer adhesion and metastasis .
Mutation Hotspots: Codons 124 and 555 are frequent mutation sites in corneal dystrophies .
Epigenetic Silencing: Hypermethylation of the TGFBI promoter occurs in 40% of ovarian cancers, correlating with advanced stage .
Immune Modulation: TGFBI expression inversely correlates with tumor mutational burden (TMB) and microsatellite instability (MSI) in colorectal and lung cancers .
Ocular Therapies: Gene-editing approaches (e.g., CRISPR/Cas9) are being explored to correct TGFBI mutations .
Cancer Therapeutics: TGFBI’s dual role complicates targeting. Inhibitors of TGFBI-integrin interactions (e.g., RGD-blocking peptides) show promise in metastatic cancers .
Biomarker Potential: TGFBI levels in serum or tumor tissue may predict immunotherapy response, particularly in cancers with high stromal infiltration .
TGFBI (Transforming growth factor-beta-induced protein), also known as βig-H3, is an extracellular matrix protein induced by TGFβ1 and secreted by many cell types. It functions primarily as a structural and signaling protein that binds to collagens type I, II, and IV, contributing to extracellular matrix organization. At the cellular level, TGFBI mediates cell-matrix interactions by binding to integrins on the cell surface, thereby influencing cellular adhesion, migration, and signaling processes. Research approaches to study these interactions include co-immunoprecipitation assays to identify binding partners, cell adhesion assays to evaluate functional outcomes, and knockout/knockdown experiments to assess the physiological significance of these interactions .
TGFBI expression is primarily induced by TGFβ1 at the transcriptional level, though regulation varies across different tissue contexts. To investigate this regulation, researchers typically employ promoter analysis using reporter gene assays, chromatin immunoprecipitation (ChIP) to identify transcription factor binding, and expression analysis under different stimulatory conditions. Interestingly, TGFBI expression patterns differ significantly between normal tissues and cancerous contexts, suggesting context-specific regulatory mechanisms. Databases such as TCGA, GEO, and Oncomine provide valuable resources for analyzing TGFBI expression across different tissues and disease states .
TGFBI engages in multiple molecular interactions that dictate its biological functions:
Extracellular matrix binding: TGFBI interacts with collagens type I, II, and IV, contributing to matrix organization and stability
Integrin binding: Acts as a ligand for various integrin receptors on cell surfaces
Signaling pathway activation: Triggers intracellular signaling, particularly through the AKT and mTORC1 pathways
The STRING database analysis reveals numerous TGFBI-binding proteins that form a complex protein-protein interaction network. These interactions can be visualized using network analysis tools and validated through techniques such as co-immunoprecipitation, proximity ligation assays, and surface plasmon resonance .
Based on current research, several model systems have proven valuable for TGFBI research:
Cellular models:
Primary human cells (fibroblasts, epithelial cells)
Cancer cell lines with manipulation of TGFBI expression
Pancreatic islet cells for diabetes-related research
Animal models:
TGFBI knockout mice: Studies show these mice are normoglycemic under basal conditions but exhibit compromised islet survival and function in vitro
Conditional knockout models for tissue-specific analysis
Disease-specific models: Streptozotocin-induced diabetes models reveal increased susceptibility in TGFBI KO mice
When designing experiments, researchers should consider both loss-of-function (knockout/knockdown) and gain-of-function (overexpression/recombinant protein) approaches to comprehensively assess TGFBI's role in specific biological contexts .
Detecting and quantifying TGFBI requires multiple complementary approaches:
Protein expression analysis:
Western blotting for semi-quantitative analysis
ELISA for quantitative measurement in biological fluids
Immunohistochemistry for spatial localization within tissues
mRNA expression analysis:
qRT-PCR for targeted quantification
RNA-seq for genome-wide expression patterns
In situ hybridization for spatial distribution
High-throughput approaches:
Public databases like GEPIA2 provide comparative analysis of TGFBI expression between cancer and normal tissues
The "Expression Analysis-Box Plots" module allows visualization with statistical parameters (p-value <0.01, log2 fold change >1)
Methodology selection should depend on the specific research question, with consideration of sensitivity, specificity, and spatial information requirements .
Multiple bioinformatic approaches have proven valuable for TGFBI research:
Approach | Tools/Databases | Applications |
---|---|---|
Expression analysis | TCGA, GTEx, Oncomine | Compare TGFBI levels across normal and disease tissues |
Survival analysis | GEPIA2, PrognoScan | Correlate TGFBI with patient outcomes |
Protein interactions | STRING database | Identify TGFBI-binding proteins |
Pathway analysis | DAVID, KEGG | Determine biological processes associated with TGFBI |
Correlation analysis | TIMER2.0 | Explore associations with immune cell infiltration |
Integrative approaches that combine multiple data types can provide comprehensive insights. For example, researchers have used the "Similar Gene Detection" module of GEPIA2 to obtain TGFBI-correlated genes, then conducted pathway enrichment analysis using DAVID to identify biological functions .
TGFBI shows remarkably diverse associations with cancer prognosis, varying by cancer type:
These conflicting patterns suggest context-dependent functions of TGFBI. Methodologically, these associations can be analyzed using Kaplan-Meier survival analysis with log-rank tests and Cox regression models to calculate hazard ratios with 95% confidence intervals .
TGFBI exhibits significant correlations with tumor microenvironment (TME) components across cancer types:
Immune cell infiltration correlations:
Positive associations: neutrophils, monocytes, macrophages, cancer-associated fibroblasts (CAFs), and myeloid-derived suppressor cells (MDSCs)
Negative associations: B cells, T follicular helper (Tfh) cells, and CD8+ T cells
Tumor stemness associations:
TGFBI correlates with tumor stemness scores (RNAss and DNAss), which are associated with tumor dedifferentiation
Evidence suggests TGFBI may support cancer stem cell growth and progression to metastasis
Stromal interactions:
Significant correlations with stromal cell infiltration based on ESTIMATE algorithm analysis
These relationships vary by cancer type, with notable differences in ovarian cancer, prostate adenocarcinoma, uveal melanoma, and thyroid carcinoma. Researchers can investigate these associations using bioinformatic tools like TIMER2.0 and experimental approaches such as single-cell RNA sequencing .
TGFBI expression shows significant correlations with genomic features that predict immunotherapy response:
Tumor Mutational Burden (TMB):
Negative correlation in colon adenocarcinoma (COAD), head and neck squamous cell carcinoma (HNSC), bladder cancer (BLCA), and lung squamous cell carcinoma (LUSC)
This suggests tumors with high mutation rates tend to have lower TGFBI expression
Microsatellite Instability (MSI):
Negative correlation in lung squamous cell carcinoma (LUSC), lung adenocarcinoma (LUAD), colon adenocarcinoma (COAD), and cholangiocarcinoma (CHOL)
MSI is associated with deficient mismatch repair, which leads to accumulated DNA mutations
These findings suggest TGFBI may influence genetic stability or the response to genomic instability. Given that both TMB and MSI are predictive biomarkers for immunotherapy response, TGFBI expression might serve as an independent predictor for immunotherapy efficacy across cancer types .
Multiple lines of evidence establish TGFBI as a diabetes risk gene:
Genetic evidence:
Human genetic studies identified three single-nucleotide polymorphisms (SNPs) in the TGFBI gene and vicinity significantly associated with type 1 diabetes risk
One SNP was significantly associated with type 2 diabetes risk
Functional evidence from animal models:
TGFBI knockout mice showed:
Compromised islet survival and function in vitro
Inferior islet transplantation outcomes compared to wild-type
Increased susceptibility to streptozotocin-induced diabetes
Molecular evidence:
Recombinant TGFBI and transgenic TGFBI overexpression promote both islet survival and function
TGFBI activates key survival pathways in islets, including AKT and mTORC1 signaling
These findings collectively suggest that TGFBI plays a protective role in pancreatic islet biology, and its disruption increases diabetes susceptibility .
TGFBI influences pancreatic islet function through several key signaling pathways:
AKT signaling pathway:
Phosphoprotein array analysis identified AKT1S1 (a molecule linking AKT and mTORC1) as modulated by TGFBI
TGFBI stimulation upregulates phosphorylation of AKT itself
mTORC1 signaling:
TGFBI stimulation increases phosphorylation of:
RPS6 (ribosomal protein S6)
EIF4EBP1 (eukaryotic translation initiation factor 4E-binding protein 1)
These are key downstream effectors of mTORC1 signaling
Functional validation:
Chemical inhibition of AKT activity modulates islet survival
siRNA knockdown of AKT1S1, RPS6, and EIF4EBP1 affects islet survival
These interventions confirm the relevance of these pathways in TGFBI-mediated effects
These molecular mechanisms suggest that TGFBI promotes islet survival and function by activating pro-survival and protein synthesis pathways, providing a mechanistic basis for its role as a diabetes risk gene .
Based on current research findings, several therapeutic strategies targeting TGFBI for diabetes could be considered:
Augmentation approaches:
Recombinant TGFBI protein administration to enhance islet survival and function
Gene therapy to increase local TGFBI expression in pancreatic tissue
Small molecules that mimic TGFBI's effects on downstream signaling
Protective strategies for islet transplantation:
Ex vivo TGFBI treatment of islets before transplantation
TGFBI-expressing scaffolds or encapsulation materials
Genetic modification of donor islets to overexpress TGFBI
Targeted approaches for specific polymorphisms:
Personalized interventions based on patient genotypes at TGFBI-associated SNPs
Allele-specific therapeutics to correct or bypass effects of risk variants
Combination therapies:
TGFBI-based interventions combined with immunomodulatory approaches for type 1 diabetes
Integration with metabolic modulators for type 2 diabetes
Each approach requires careful validation to ensure efficacy and safety, particularly given TGFBI's complex roles in multiple tissues and disease contexts .
TGFBI has been implicated in cardiovascular pathophysiology, particularly in myocardial fibrosis and atrial fibrillation:
Myocardial fibrosis connection:
TGFBI binds to collagens and participates in extracellular matrix organization
Its induction by TGFβ1, a known pro-fibrotic factor, suggests involvement in fibrotic processes
Atrial fibrillation association:
Research indicates TGFBI may contribute to the pathophysiological basis of atrial fibrillation
Microarray analysis of left atrial tissue samples from atrial fibrillation patients has been used to investigate TGFBI coexpression patterns
Molecular mechanisms:
Bioinformatic analysis identified TGFBI coexpression genes enriched in biological processes, cellular components, molecular functions, and KEGG pathways relevant to cardiac function
Protein-protein interaction networks help illuminate how TGFBI interacts with other cardiac-relevant factors
While current evidence establishes correlations, further functional studies are needed to definitively characterize TGFBI's role in cardiac pathophysiology and determine whether it represents a viable therapeutic target for cardiac fibrosis and arrhythmias .
To investigate TGFBI's role in cardiac tissue, researchers should consider these complementary approaches:
Expression analysis in clinical samples:
Comparative analysis of TGFBI levels in healthy versus diseased cardiac tissue
Correlation with fibrosis markers and electrical conduction parameters
Spatial mapping using immunohistochemistry to determine cellular localization
In vitro models:
Primary cardiac fibroblast cultures with TGFBI manipulation
Cardiomyocyte-fibroblast co-culture systems to assess cell-cell interactions
Engineered heart tissues with controlled TGFBI expression
Animal models:
Cardiac-specific TGFBI knockout or overexpression
Fibrosis induction models (e.g., angiotensin II infusion, transaortic constriction)
Electrophysiological phenotyping for arrhythmia susceptibility
Molecular studies:
Identification of cardiac-specific TGFBI-binding partners
TGFBI-dependent signaling pathway analysis in cardiac cells
Integration with other known fibrosis and arrhythmia pathways
Systems biology approaches:
Multi-omics analysis of TGFBI effects on cardiac transcriptome, proteome, and metabolome
Network analysis to position TGFBI within cardiac disease pathways
Machine learning to identify TGFBI-associated phenotypes from clinical data
These approaches can help establish whether TGFBI actively drives cardiac pathology or represents a secondary response to cardiac injury .
Understanding TGFBI's interactions with established mediators of cardiac fibrosis requires investigation of several potential mechanisms:
TGFβ signaling pathway integration:
As a TGFβ-induced protein, TGFBI likely functions downstream of canonical TGFβ signaling
Potential feedback mechanisms where TGFBI modulates TGFβ receptor availability or activity
Cross-talk with Smad-dependent and Smad-independent TGFβ pathways
Extracellular matrix interactions:
TGFBI binds to collagens that are abundant in fibrotic cardiac tissue
Potential role in organizing matrix architecture during fibrotic remodeling
Interaction with matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs)
Inflammatory mediator interactions:
TGFBI's associations with immune cell populations suggest potential roles in cardiac inflammation
Interaction with inflammatory cytokines that drive cardiac fibrosis
Modulation of macrophage polarization in the injured heart
Integrin signaling:
TGFBI's known interactions with integrins may influence mechanotransduction in cardiac cells
Potential enhancement or inhibition of integrin-mediated fibrotic signaling
Role in myofibroblast differentiation and activation
Methodologically, these interactions can be studied through co-immunoprecipitation, proximity ligation assays, FRET-based interaction studies, and genetic epistasis experiments in animal models .
Single-cell technologies offer unprecedented opportunities to dissect TGFBI's role in complex tissues:
Cell type-specific expression mapping:
Single-cell RNA sequencing (scRNA-seq) can identify which specific cell populations express TGFBI
This reveals potential cellular sources and targets of TGFBI in disease contexts
Particularly valuable in heterogeneous tissues like tumors or the pancreatic islet
Receptor-ligand interaction networks:
Computational tools applied to scRNA-seq data can predict TGFBI-receptor interactions across different cell types
This enables mapping of potential TGFBI signaling networks within complex tissue microenvironments
Cellular trajectory analysis:
Pseudotime analysis can track how TGFBI expression changes during cellular differentiation or disease progression
Reveals temporal dynamics of TGFBI function in developmental or pathological processes
Spatial context:
Spatial transcriptomics can map TGFBI expression within tissue architecture
Critical for understanding TGFBI's role in cell-cell communication and tissue organization
Multi-modal analysis:
Simultaneous protein and RNA detection can validate TGFBI expression at multiple levels
CITE-seq approaches can link TGFBI expression with cell surface phenotypes
These approaches could significantly advance our understanding of how TGFBI functions within the complex cellular ecosystems of tumors, pancreatic islets, and fibrotic tissues .
Systems biology offers powerful frameworks for contextualizing TGFBI within broader disease networks:
Network biology approaches:
Protein-protein interaction (PPI) networks: The search results mention using STRING database to identify the top 50 TGFBI-binding proteins
Gene regulatory networks: Integrating transcription factors controlling TGFBI with its downstream targets
Pathway cross-talk analysis: Examining how TGFBI-associated pathways interact with other disease-relevant processes
Multi-omics integration:
Correlating TGFBI genomic variants with transcriptomic, proteomic, and phenotypic data
Identifying molecular signatures associated with TGFBI expression across diseases
Methods like MOFA (Multi-Omics Factor Analysis) or DIABLO can be applied
Machine learning applications:
Predictive modeling of disease outcomes based on TGFBI and related biomarkers
Feature importance analysis to determine TGFBI's relative contribution to disease processes
Patient stratification based on TGFBI-associated molecular profiles
Comparative systems approaches:
Cross-disease analysis to identify common and distinct roles of TGFBI
Evolutionary conservation analysis of TGFBI networks
Drug repurposing opportunities based on TGFBI pathway modulation
These integrative approaches can help position TGFBI within the complex molecular landscape of diseases, potentially identifying novel therapeutic targets or biomarker combinations .
Post-translational modifications (PTMs) likely play crucial roles in regulating TGFBI function, though this area remains underexplored:
Potential PTMs affecting TGFBI:
Glycosylation: As an extracellular protein, TGFBI may undergo N- and O-linked glycosylation
Phosphorylation: Potential regulation by extracellular kinases
Proteolytic processing: Cleavage may generate fragments with distinct functions
Cross-linking: Possible transglutaminase-mediated modifications
Functional implications:
Altered binding affinity for collagens, integrins, or other partners
Modified stability or half-life in the extracellular environment
Generation of bioactive fragments with distinct signaling properties
Changed immunogenicity or antigenicity
Methodological approaches:
Mass spectrometry-based proteomics to identify and quantify PTMs
Site-directed mutagenesis of potential modification sites
In vitro enzymatic modification followed by functional assays
Antibodies specific to modified forms of TGFBI
Disease relevance:
PTM patterns may differ between normal and pathological states
Disease-specific modifications could serve as biomarkers
Targeting enzymes that modify TGFBI might offer therapeutic opportunities
Investigation of TGFBI PTMs represents an important frontier that could explain context-dependent functions and reveal new regulatory mechanisms .
The contradictory roles of TGFBI observed across different studies require careful interpretation:
The diverse associations of TGFBI with prognosis across cancer types exemplify this complexity, with high expression predicting poor outcomes in some cancers but favorable outcomes in others .
When analyzing TGFBI associations with clinical outcomes, researchers should consider these statistical approaches:
Survival analysis methodology:
Kaplan-Meier method to visualize survival differences
Log-rank tests for statistical significance assessment
Cox proportional hazards models for hazard ratio calculation with confidence intervals
Testing of proportional hazards assumptions
Expression categorization approaches:
Median-based dichotomization (high/low) as used in many TGFBI studies
Quartile or percentile-based categorization for dose-response assessment
Continuous variable analysis when appropriate
Sensitivity analysis with different cutoff values
Multivariate adjustments:
Inclusion of established prognostic factors as covariates
Assessment for potential confounding and effect modification
Stratified analysis by important clinical variables
Multiple testing considerations:
Appropriate correction methods (Benjamini-Hochberg, Bonferroni)
Clear reporting of both unadjusted and adjusted p-values
Pre-specified hypothesis testing versus exploratory analysis
Validation approaches:
Internal validation (bootstrapping, cross-validation)
External validation in independent cohorts
Comparison with previously published results
The search results demonstrate these approaches, particularly in analyzing TGFBI's prognostic significance across different cancer types and survival endpoints .
Comparing TGFBI function across different model systems requires careful methodological approaches:
Standardization strategies:
Consistent TGFBI manipulation methods across models
Matched genetic backgrounds in animal models
Comparable experimental conditions and readouts
Uniform statistical approaches and reporting
Cross-species considerations:
Sequence homology analysis between human and model organism TGFBI
Functional domain conservation assessment
Species-specific binding partner identification
Consideration of differences in tissue architecture and physiology
Translation between in vitro and in vivo findings:
Validation of key in vitro observations in appropriate animal models
Ex vivo approaches using primary tissues to bridge the gap
Organoid models to better recapitulate tissue complexity
Careful interpretation of differences between simplified and complex systems
Integration with human data:
Correlation of experimental findings with human genetic associations
Validation in patient-derived samples when possible
Comparison with disease-specific gene expression signatures
Assessment of clinical relevance of experimental endpoints
The search results illustrate this challenge, with findings from TGFBI knockout mice showing compromised islet function that correlates with human genetic associations between TGFBI variants and diabetes risk .
Transforming Growth Factor Beta Induced (TGFBI), also known as βig-H3 or keratoepithelin, is an extracellular matrix protein induced by Transforming Growth Factor Beta (TGF-β). It plays a crucial role in various physiological and pathological processes, including cell growth, differentiation, apoptosis, and extracellular matrix production .
TGFBI is a secreted protein that interacts with various components of the extracellular matrix. It contains four fasciclin-1 (FAS1) domains, which are involved in cell adhesion and interaction with integrins. TGFBI is known to bind to integrins such as α3β1, αvβ3, and αvβ5, mediating cell adhesion and migration .
TGFBI has diverse biological functions, including:
TGFBI has significant clinical implications in various diseases: