TIAF1 (TGFB1-induced anti-apoptotic factor 1), also known as MYO18A, is a 12 kDa protein encoded by the TIAF1 gene (Entrez Gene ID: 9220) located on chromosome 2p13 . Initially identified as a TGF-β1-inducible factor, TIAF1 regulates apoptosis, cell proliferation, and stress responses. Its roles span anti-apoptotic functions in fibroblasts to pro-apoptotic effects in monocytic cells, with involvement in neurodegenerative diseases and cancer .
TIAF1 interacts with Smad4 to modulate TGF-β signaling and regulates p53-mediated apoptosis through nuclear translocation . Its LCD facilitates protein-protein interactions, enabling aggregation under stress .
Anti-apoptotic: Inhibits TNF-α cytotoxicity by blocking TRADD/FADD/RIPK1 signaling in fibroblasts .
Pro-apoptotic: Induces apoptosis in monocytic U937 cells via p53/Cip1/p21 upregulation and ERK suppression .
TIAF1 binds Smad4, influencing Smad-dependent promoter activity. Knockdown of TIAF1 leads to spontaneous nuclear Smad accumulation and TGF-β1-mediated gene activation .
TIAF1 aggregation under TGF-β1 induces APP dephosphorylation, promoting Aβ generation in vitro .
TIAF1 upregulation in early tumors may decline in metastatic stages, suggesting a dual role in tumor suppression and progression .
TIAF1 (TGFB1-induced anti-apoptotic factor 1) is a protein encoded by the TIAF1 gene in humans. It is also known by several aliases including MAJN and SPR210, with an NCBI Gene ID of 9220 . This protein was initially identified as a TGF-β1-induced factor with anti-apoptotic properties. The human TIAF1 protein is characterized by its relatively small size, functioning as a regulatory protein in multiple cellular pathways .
TIAF1 has demonstrated significant evolutionary conservation, with homologs identified in multiple species including bovine forms (cow TIAF1, gene ID: 101903486) . This conservation suggests fundamental biological importance across mammalian species. The protein's molecular structure contains domains that facilitate its interactions with multiple binding partners, contributing to its diverse functional capabilities.
TIAF1 engages in several critical protein-protein interactions that define its functional roles. Confirmed interaction partners include:
These interaction networks position TIAF1 at the intersection of multiple critical cellular processes. When investigating these interactions experimentally, co-immunoprecipitation with specific antibodies like SAB1412775 (monoclonal anti-TIAF1) or HPA051129 (polyclonal anti-TIAF1) can be employed for protein complex isolation and analysis .
TIAF1 demonstrates tissue-specific expression patterns that are further altered in pathological conditions. Based on analysis of expression databases and clinical studies:
TIAF1 shows differential expression in neural tissues, with specific patterns during brain development according to Allen Brain Atlas data .
The protein is significantly upregulated in activated helper T lymphocytes (TH2) during chronic kidney and liver allograft rejection, suggesting immunological functions .
Expression analysis across multiple datasets indicates that TIAF1 has at least 1,297 functional associations with biological entities spanning 7 categories, extracted from 44 datasets .
For experimental determination of TIAF1 expression:
RNA-seq or qRT-PCR can quantify transcript levels using gene-specific primers
Immunohistochemistry with antibodies like HPA051129 allows visualization in tissue sections
Western blot analysis provides quantitative protein expression data across sample types
TIAF1 exhibits a complex relationship with TGF-β signaling through multiple mechanisms:
TIAF1 physically interacts with Smad4, a central mediator of canonical TGF-β signaling, blocking SMAD-dependent promoter activation when overexpressed .
Knockdown experiments using siRNA against TIAF1 result in spontaneous accumulation of Smad proteins in the nucleus and activation of SMAD-governed promoters, confirming its inhibitory role in this pathway .
Most notably, TGF-β1 can induce TIAF1 self-aggregation through a non-canonical mechanism that operates independently of the type II TGF-β receptor, representing an alternative signaling pathway .
Smad4 has been demonstrated to interrupt TIAF1 self-aggregation, establishing a regulatory feedback mechanism within this signaling axis .
For researchers investigating this pathway, experimental approaches should include:
Luciferase reporter assays with SMAD binding elements to assess transcriptional effects
Subcellular fractionation to monitor Smad nuclear translocation
Protein aggregation assays to detect TIAF1 self-assembly following TGF-β1 stimulation
Co-expression studies of TIAF1 and Smad4 to examine their functional interdependence
TIAF1 demonstrates contextual duality in apoptotic regulation, functioning as:
An anti-apoptotic factor that inhibits the cytotoxic effects of TNF-alpha, TRADD, and FADD under normal conditions, protecting cells from death receptor-mediated apoptosis .
Paradoxically, when TIAF1 undergoes self-aggregation, it converts to a pro-apoptotic factor, inducing cell death through caspase-dependent mechanisms .
A critical component in p53- and WOX1-mediated apoptotic pathways, with suppression of TIAF1 by siRNA preventing UV irradiation-mediated p53 phosphorylation and nuclear translocation .
To investigate these dual roles, researchers should consider:
Time-course analyses following apoptotic stimuli with measurement of TIAF1 aggregation status
Caspase activity assays in contexts of TIAF1 overexpression, knockdown, and aggregation
Co-localization studies with death receptor components and p53 pathway elements
Mutational analyses to identify domains responsible for anti- versus pro-apoptotic functions
While direct evidence for TIAF1 in inflammatory signaling is limited in the provided search results, several connections can be inferred:
TIAF1 interactions with JAK3 suggest potential involvement in cytokine signaling cascades that regulate inflammatory responses .
The upregulation of TIAF1 in TH2 lymphocytes during allograft rejection points to roles in adaptive immunity and potential inflammatory regulation .
Though a genetic association study did not demonstrate strong evidence for TIAF1 involvement in Crohn's disease susceptibility, this does not exclude functional roles in inflammatory conditions .
Given these connections, researchers investigating inflammatory aspects should:
Examine TIAF1 expression and aggregation status in inflammatory cell populations
Assess cytokine production profiles in TIAF1-manipulated systems
Investigate potential post-translational modifications of TIAF1 during inflammatory activation
Consider TIAF1's relationship with NF-κB and other inflammatory transcription factors
TIAF1 has emerged as a significant factor in Alzheimer's disease (AD) pathogenesis through several mechanisms:
TIAF1 aggregates have been identified in hippocampal tissues of both non-demented individuals and AD patients, with significantly higher prevalence in AD cases (48% versus 17% in non-demented controls), suggesting TIAF1 aggregation may precede amyloid β (Aβ) plaque formation .
Mechanistic studies reveal that TGF-β-regulated TIAF1 aggregation initiates a cascade of events leading to:
Polymerized TIAF1 physically interacts with amyloid fibrils, potentially stabilizing and promoting plaque formation in vivo .
This temporal relationship is visualized in the following progression model:
| Stage | Process | Cellular Events |
|---|---|---|
| 1 | TGF-β signaling activation | Environmental/cellular stress triggers pathway |
| 2 | TIAF1 self-aggregation | Occurs independently of type II TGF-β receptor |
| 3 | APP dephosphorylation | Modification at Thr668 position |
| 4 | APP processing alteration | Increased AICD and Aβ generation |
| 5 | Amyloid fibril formation | Production of amyloidogenic species |
| 6 | TIAF1-amyloid interactions | Stabilization of plaques and disease progression |
These findings position TIAF1 as a potential upstream mediator in AD pathogenesis, worthy of consideration as both a biomarker and therapeutic target.
When investigating TIAF1 in neurodegenerative diseases, researchers should employ multiple complementary approaches:
For detection of TIAF1 aggregates in neural tissues:
For examining TIAF1-amyloid interactions:
Co-immunoprecipitation of TIAF1 with Aβ species
Proximity ligation assays in tissue sections or cell models
In vitro binding studies with purified components
Electron microscopy to visualize physical interactions
For modeling TIAF1 pathogenic mechanisms:
Transgenic animal models with TIAF1 overexpression or conditional knockout
Neural cell cultures with manipulated TIAF1 expression
Patient-derived iPSCs differentiated into relevant neural cell types
Organoid models to capture complex cellular interactions
For therapeutic targeting:
Small molecule screening against TIAF1 aggregation
Peptide inhibitors of TIAF1-amyloid interactions
Antisense oligonucleotides to modulate TIAF1 expression
When designing these experiments, researchers should account for the temporal aspects of TIAF1 involvement, potentially focusing on early-stage processes before overt amyloid pathology.
TIAF1 demonstrates complex roles in cancer biology through multiple mechanisms:
TIAF1 shows aggregation-dependent control of tumor progression and metastasis, with its aggregation state potentially serving as a molecular switch .
The protein plays essential roles in p53- and WOX1-mediated cell death pathways, which are frequently dysregulated in cancer .
The duality of TIAF1 as both an anti-apoptotic factor under normal conditions and a potential pro-apoptotic factor when aggregated suggests context-dependent functions in tumorigenesis .
These observations raise several research questions for oncology investigations:
Does TIAF1 aggregation status correlate with cancer stage or prognosis?
How do cancer cells manipulate TIAF1 to evade apoptosis?
Are there cancer-specific mutations or modifications of TIAF1?
Could targeting TIAF1 aggregation represent a therapeutic strategy?
Methodologically, researchers should consider:
Comprehensive profiling of TIAF1 expression and aggregation across cancer types
Analysis of TIAF1 in paired normal-tumor samples to identify alterations
Correlation of TIAF1 status with p53 pathway functionality
Functional studies in cancer cell lines and animal models
Several approaches have been validated for modulating TIAF1 expression in research contexts:
When designing knockdown experiments, researchers should:
Include appropriate non-targeting controls
Validate knockdown efficiency at both mRNA (qRT-PCR) and protein (Western blot) levels
Monitor cellular phenotypes, particularly in relation to TGF-β signaling and apoptosis
Consider potential compensatory mechanisms in long-term studies
Commercial reagents mentioned in the search results include MISSION esiRNA targeting human TIAF1 (EHU221441) , which can provide efficient knockdown for experimental applications.
TIAF1 detection presents several technical challenges that researchers should address:
The protein's potential for self-aggregation can affect antibody accessibility and epitope recognition, necessitating specialized extraction methods.
TIAF1's relatively small size and potential post-translational modifications require careful selection of detection reagents and conditions.
The distinction between monomeric, oligomeric, and aggregated forms requires specific methodological approaches.
To overcome these challenges:
For Western blotting: Consider both reducing and non-reducing conditions to capture different forms
For immunohistochemistry: Optimize antigen retrieval methods for aggregated proteins
For aggregation detection: Employ filter retardation assays as used in hippocampal studies
For quantification: Consider ELISA approaches with capture and detection antibody pairs
Commercial antibodies validated for TIAF1 detection include:
SAB1412775: Monoclonal Anti-TIAF1 antibody (mouse), validated for ELISA and Western blot
HPA051129: Polyclonal Anti-TIAF1 antibody (rabbit), validated for immunohistochemistry
These can be paired with PEP-1581, a synthetic peptide corresponding to 16 amino acids near the amino terminus of human TIAF1, as a blocking peptide for specificity controls
To investigate TIAF1's context-dependent functions (anti-apoptotic versus pro-apoptotic):
Time-course experiments following specific stimuli:
Track TIAF1 aggregation status using biochemical separation
Monitor corresponding cellular phenotypes (survival versus death)
Assess activation of downstream pathways
Structure-function analyses:
Generate domain-specific mutants to dissect regions responsible for each function
Create aggregation-prone versus aggregation-resistant variants
Perform rescue experiments in TIAF1-depleted systems
Single-cell approaches:
Utilize live-cell imaging with fluorescently-tagged TIAF1
Correlate TIAF1 aggregation status with cellular outcomes
Implement single-cell transcriptomics to identify divergent pathway activation
Stress-response studies:
Expose cells to varied stressors (UV, oxidative stress, TGF-β)
Monitor TIAF1 conformational changes and interactome dynamics
Correlate with downstream pathway activation (apoptotic versus survival)
These approaches should be combined with careful controls and quantitative metrics to capture the transitional states of TIAF1 function.
The observation that TGF-β1 can induce TIAF1 self-aggregation independent of the canonical type II TGF-β receptor presents a significant mechanistic puzzle . Several hypothetical pathways warrant investigation:
Direct TGF-β1-TIAF1 interaction:
Is there direct binding between TGF-β1 and TIAF1?
Could this represent an alternative signaling mechanism?
What structural domains mediate this potential interaction?
Alternative receptor utilization:
Does TGF-β1 signal through non-canonical receptors to influence TIAF1?
Are there co-receptors or adaptor proteins facilitating this process?
How does this pathway intersect with canonical TGF-β signaling?
Cellular stress as an intermediary:
Does TGF-β1 induce cellular stress that subsequently promotes TIAF1 aggregation?
What stress response pathways might converge on TIAF1?
How do environmental alterations influence this process?
Experimental approaches to address these questions include:
In vitro reconstitution with purified components to test direct interactions
Receptor knockout systems to verify independence from canonical pathway
Proteomic analysis of TIAF1 complexes formed under TGF-β1 stimulation
Structural studies of TIAF1 conformational changes following various stimuli
The discovery that TIAF1 aggregates appear in both non-demented and AD hippocampi, with higher prevalence in AD, suggests a potential temporal progression . Several critical questions emerge:
Does TIAF1 aggregation truly precede Aβ deposition in vivo, and by what time frame?
What triggers the transition from benign to pathogenic TIAF1 aggregation?
How does aggregated TIAF1 mechanistically lead to APP dephosphorylation?
What phosphatases are recruited or activated in this process?
To address these questions, researchers should consider:
Longitudinal studies in animal models with temporal mapping of TIAF1 and Aβ pathology
Analysis of preclinical AD biomarkers in relation to TIAF1 aggregation status
Proteomic identification of TIAF1 aggregate-associated phosphatases
Phosphorylation site-specific mutants of APP to confirm the critical sites
The finding that TIAF1-positive samples containing Aβ aggregates comprise 17% of non-demented versus 48% of AD hippocampi suggests TIAF1 aggregation as a potential early biomarker worthy of further investigation as both a diagnostic tool and therapeutic target .
Given TIAF1's potential as a therapeutic target, developing screening platforms for TIAF1 aggregation modulators represents an important research direction:
Assay development considerations:
Recombinant TIAF1 production and purification systems
Fluorescence-based aggregation detection methods (ThT/ThS binding)
Cell-based reporters of TIAF1 aggregation status
Methods to distinguish between beneficial and pathological modulation
Compound library selection:
Known modulators of protein aggregation
Natural products with neuroprotective properties
Repurposing candidates from other aggregation-related diseases
Fragment-based approaches for novel chemical matter
Validation pipeline design:
Secondary assays for mechanism confirmation
Cellular models of TIAF1-mediated pathology
Selectivity assessments against other aggregation-prone proteins
Early ADME and blood-brain barrier penetration studies
Advanced screening technologies:
Surface plasmon resonance for direct binding studies
Nuclear magnetic resonance for structural insights
High-content imaging for cellular phenotypes
Label-free aggregation detection methods
By establishing robust screening platforms, researchers can accelerate the discovery of chemical probes and potential therapeutic candidates targeting TIAF1's role in neurodegenerative and potentially other diseases.
Transforming Growth Factor Beta 1 (TGFB1) is a multifunctional cytokine that plays a crucial role in regulating various cellular processes, including cell growth, differentiation, apoptosis, and immune responses. TGFB1 is known for its ability to induce anti-apoptotic factors, which are essential for cell survival and tissue homeostasis. One such factor is the TGFB1-Induced Anti-Apoptotic Factor 1 (TIAF1), a protein that has garnered significant attention for its role in preventing apoptosis in various cell types.
The recombinant human TGFB1-Induced Anti-Apoptotic Factor 1 is typically produced using recombinant DNA technology. The protein is expressed in suitable host cells, such as E. coli or mammalian cells, and subsequently purified to obtain a high-quality product. The TIAF1 solution often contains 20mM Tris-HCl buffer (pH 8.0), 0.4M Urea, and 10% glycerol .
TGFB1 exerts its effects by binding to specific receptors on the cell surface, leading to the activation of intracellular signaling pathways. One of the key pathways involved is the SMAD signaling pathway. Upon TGFB1 binding, the receptors phosphorylate SMAD2 and SMAD3 proteins, which then form a complex with SMAD4. This complex translocates to the nucleus, where it regulates the expression of target genes, including those involved in anti-apoptotic processes .
The primary function of TIAF1 is to inhibit apoptosis, thereby promoting cell survival. This is particularly important in tissues that are prone to damage or stress, such as the brain, liver, and immune system. TIAF1 achieves this by modulating the expression of various apoptotic and anti-apoptotic genes. For instance, it can upregulate the expression of anti-apoptotic proteins like Bcl-2 and downregulate pro-apoptotic proteins like Bax .
The anti-apoptotic properties of TIAF1 have significant clinical implications. In the context of neurodegenerative diseases, such as Alzheimer’s and Parkinson’s, enhancing TIAF1 activity could potentially protect neurons from apoptosis, thereby slowing disease progression. Similarly, in cancer therapy, targeting TIAF1 could help in sensitizing cancer cells to apoptosis, making them more susceptible to treatment .
Ongoing research is focused on understanding the detailed mechanisms by which TIAF1 exerts its anti-apoptotic effects. Studies are also exploring the potential therapeutic applications of TIAF1 in various diseases. For example, researchers are investigating the use of TIAF1 in combination with other therapeutic agents to enhance its efficacy and reduce potential side effects .