TINAGL1, or Tubulointerstitial Nephritis Antigen Like 1, is a protein encoded by the TINAGL1 gene in humans. This protein is similar in sequence to tubulointerstitial nephritis antigen, a secreted glycoprotein recognized by antibodies in some types of immune-related tubulointerstitial nephritis . TINAGL1 has been studied for its roles in various diseases, including cancer, where it exhibits both tumor-suppressive and oncogenic properties depending on the cancer type.
Gene Information:
Protein Information:
TINAGL1 is expressed in various tissues, including endothelial cells and smooth muscle cells in the brain . Its expression pattern suggests a role in vascular biology and potentially in other cellular processes.
TINAGL1 has been shown to inhibit TNBC progression and metastasis by binding to epidermal growth factor receptor (EGFR) and integrin β1 subunit (ITGB1), thereby blocking key signaling pathways involved in tumor growth and spread . High expression of TINAGL1 is associated with better prognosis in TNBC patients, suggesting its potential as a therapeutic agent .
In contrast to its tumor-suppressive role in TNBC, TINAGL1 promotes hepatocellular carcinogenesis by activating the TGF-β signaling pathway and increasing VEGF secretion, leading to enhanced proliferation, survival, and metastasis of HCC cells . This indicates that TINAGL1 can have context-dependent roles in cancer.
TINAGL1 is also associated with colorectal adenocarcinoma, although detailed mechanisms and its specific role in this cancer type are less well-defined compared to TNBC and HCC .
| Cancer Type | Role of TINAGL1 | Mechanism | Clinical Implication |
|---|---|---|---|
| TNBC | Tumor suppressor | Inhibits EGFR and integrin signaling | Potential therapeutic agent |
| HCC | Oncogene | Activates TGF-β/Smad3/VEGF axis | Potential biomarker and therapeutic target |
| Colorectal Adenocarcinoma | Associated with disease | Less defined | Requires further research |
TINAGL1 (Tubulointerstitial Nephritis Antigen Like 1) is a secreted glycoprotein that functions as an extracellular matrix protein involved in cell adhesion, proliferation, migration, and differentiation . It shares sequence similarity with tubulointerstitial nephritis antigen, which is recognized by antibodies in certain immune-related tubulointerstitial nephritis cases .
From a functional perspective, TINAGL1 demonstrates cysteine-type peptidase activity and scavenger receptor activity, though it is classified as a non-catalytic peptidase C1 family protein . Research indicates it may be implicated in adrenocortical zonation and mechanisms for repressing CYP11B1 gene expression in adrenocortical cells . The protein has been identified as having involvement in multiple physiological systems, with particularly notable roles emerging in reproductive biology and potential implications in several pathological conditions.
TINAGL1 expression patterns vary significantly across tissue types, with emerging research highlighting its presence in human granulosa cells of the ovary . Expression regulation appears to be tissue-specific and potentially hormone-responsive, as evidenced by research in mouse models showing TINAGL1 functions as a serum biomarker that fluctuates throughout the estrous cycle, with peak expression during the estrous phase .
The gene is associated with multiple transcript variants encoding different isoforms, with at least three identified variants according to current research . These different isoforms may contribute to tissue-specific functions and regulatory mechanisms. The molecular weight of the protein is approximately 52 kDa with 467 amino acids, consistent with observations in immunoblotting experiments .
Gene regulation may involve microRNA-mediated mechanisms, as TINAGL1 has been identified as a potential target for multiple microRNAs, suggesting complex post-transcriptional regulatory networks .
For reliable TINAGL1 detection in human tissue samples, immunological techniques have demonstrated significant efficacy. Specifically:
Immunofluorescence (IF): Validated antibodies such as the Rabbit Polyclonal TINAGL1 antibody (CL647-12077) have shown excellent reactivity with human samples in immunofluorescence applications. For optimal results, recommended dilution ranges are 1:50-1:500 for immunofluorescence on paraffin-embedded tissues (IF-P) .
Enzyme-linked Immunosorbent Assay (ELISA): Commercial ELISA kits have been successfully employed to quantify TINAGL1 protein levels in human follicular fluid, as demonstrated in research studying its relationship to ovarian reserve .
Quantitative Real-time PCR (qPCR): For gene expression analysis, qPCR following RNA extraction and cDNA amplification has proven effective for quantifying TINAGL1 mRNA expression in human cells, such as granulosa cells recovered from follicular fluid .
The choice of method should be tailored to the specific research question, with consideration of whether protein localization, quantification, or gene expression data is most relevant to the investigation.
| Antibody Type | Product Example | Host/Isotype | Validated Applications | Reactivity | Conjugation |
|---|---|---|---|---|---|
| Polyclonal | CL647-12077 | Rabbit/IgG | Immunofluorescence (IF-P) | Human, mouse, rat | CoraLite® Plus 647 Fluorescent Dye |
This antibody has been specifically validated for immunofluorescence on paraffin-embedded tissues (IF-P) with a recommended dilution range of 1:50-1:500. It recognizes the full TINAGL1 protein (52 kDa) and has demonstrated specific staining in human liver cancer tissue .
When selecting antibodies for TINAGL1 research, considerations should include:
Target application (Western blot, immunohistochemistry, immunofluorescence, etc.)
Species cross-reactivity requirements
Conjugation needs (fluorescent tags vs. unconjugated primary antibodies)
Mono vs. polyclonal properties depending on specificity requirements
Researchers should validate antibodies in their specific experimental conditions, as performance can vary based on tissue type, fixation methods, and detection systems .
Recent research has identified TINAGL1 as a potentially critical factor in human reproductive biology, particularly in ovarian function. A 2024 study (the first to demonstrate TINAGL1 expression in human granulosa cells) found that TINAGL1 concentration was significantly lower in the follicular fluid of patients with diminished ovarian reserve (DOR) compared to control patients without infertility or those with polycystic ovarian syndrome (PCOS) .
The protein's importance in reproduction is further supported by evidence from animal models. TINAGL1 knockout mice have been documented to be infertile, suggesting a fundamental role in normal reproductive function . Additionally, TINAGL1 functions as a serum biomarker that fluctuates throughout the mouse estrous cycle, with peak expression during the estrous phase, indicating hormone-responsive regulation .
These findings collectively suggest that TINAGL1 may serve as a novel biomarker for ovarian reserve assessment and potentially play a mechanistic role in female fertility. The correlation between decreased TINAGL1 levels and diminished ovarian reserve could provide new insights into the molecular basis of certain forms of female infertility.
Based on recent methodological approaches in TINAGL1 ovarian research, investigators should consider the following experimental design elements:
Sample Collection Protocol:
Collect follicular fluid during oocyte retrieval procedures from well-defined patient populations (e.g., DOR patients, PCOS patients, and controls undergoing fertility preservation or with partners with male factor infertility only)
Isolate granulosa cells from follicular fluid using standardized separation techniques
Ensure proper demographic and clinical data collection for meaningful stratification and analysis
Protein Expression Analysis:
Quantify TINAGL1 protein levels in follicular fluid using validated commercial ELISA kits
Consider analyzing TINAGL1 in matched serum samples to determine if circulating levels correlate with follicular fluid concentrations
Gene Expression Analysis:
Extract RNA from isolated granulosa cells using standardized protocols
Perform cDNA synthesis and qPCR with validated TINAGL1-specific primers
Include appropriate housekeeping genes as internal controls
Functional Studies:
Consider in vitro modulation of TINAGL1 expression in cultured granulosa cells (knockdown or overexpression)
Assess impact on cell proliferation, steroidogenesis, and response to gonadotropins
Evaluate interactions with extracellular matrix components and potential receptor-mediated signaling
A comprehensive approach combining these methods would provide insights into both the potential utility of TINAGL1 as a biomarker and its functional significance in ovarian physiology and pathology .
TINAGL1 has emerging significance in cancer biology with notable associations to colorectal adenocarcinoma. According to GeneCards data, colorectal adenocarcinoma is among the diseases associated with TINAGL1 gene dysregulation . While the search results don't provide extensive details about the specific mechanisms in colorectal cancer, research in other cancer models offers relevant insights.
In breast cancer research, TINAGL1 has been studied in the context of tumor microenvironment interactions. Studies have indicated that TINAGL1 can bind to EGFR and integrins on tumor cells, which in the 4T1 murine breast cancer model resulted in downregulation of cancer proliferation and metastasis pathways . This suggests that TINAGL1 may function as a matricellular protein that influences cancer cell signaling and potentially tumor progression.
For researchers investigating TINAGL1 in colorectal cancer, examination of:
Expression patterns in tumor versus normal mucosa
Correlation with disease stage and progression
Potential interactions with known colorectal cancer signaling pathways (such as Wnt/β-catenin)
Its utility as a prognostic or predictive biomarker
would be valuable approaches to better understand its role in this specific cancer type.
When investigating TINAGL1 in disease states, researchers should consider implementing the following methodological approaches:
Tissue Expression Analysis:
Immunohistochemistry/immunofluorescence to evaluate TINAGL1 expression patterns in diseased versus normal tissues
Tissue microarrays for high-throughput screening across multiple patient samples
Laser capture microdissection to isolate specific cell populations for more precise expression analysis
Functional Studies:
Gene knockdown or overexpression in relevant cell lines to assess phenotypic effects
Co-immunoprecipitation to identify protein-protein interactions relevant to disease mechanisms
Receptor binding assays to characterize interactions with EGFR, integrins, or other potential binding partners
Clinical Correlation:
Analysis of TINAGL1 levels in patient biospecimens (tissue, serum, or other biological fluids)
Correlation with clinicopathological parameters and patient outcomes
Multivariate analysis to assess independent prognostic value
Animal Models:
Development or utilization of conditional tissue-specific knockout models to assess TINAGL1 function in relevant disease contexts
Xenograft studies with TINAGL1-manipulated cells to evaluate effects on tumor growth and metastasis
These approaches should be tailored to the specific disease context and research question. Researchers should be mindful that TINAGL1's matricellular nature suggests both structural roles and signaling functions that may be tissue-specific and context-dependent .
TINAGL1 demonstrates complex interactions with various proteins and signaling pathways that are beginning to be elucidated. Based on current research:
TINAGL1 has been shown to interact with cell surface receptors, notably EGFR (epidermal growth factor receptor) and integrins in cancer models. These interactions appear to influence downstream signaling, potentially affecting proliferation and metastasis pathways . This suggests TINAGL1 may function as a ligand or co-factor in receptor-mediated signaling.
The protein's extracellular matrix (ECM) association indicates interactions with other ECM components that may modulate cell adhesion, migration, and tissue organization. Its structure suggests potential interactions with elements of the basement membrane, which would be consistent with its role in cell-matrix communication .
In adrenocortical cells, TINAGL1 has been implicated in mechanisms repressing CYP11B1 gene expression, suggesting involvement in transcriptional regulation pathways, though the exact signaling intermediates remain to be fully characterized .
For advanced investigation of TINAGL1 interaction networks, researchers should consider:
Proteomics approaches to identify binding partners
Phosphoproteomic analysis to determine effects on signaling cascades
Transcriptomic profiling following TINAGL1 modulation to identify downstream effects
Chromatin immunoprecipitation studies if nuclear interactions are suspected
Creating effective TINAGL1 knockout or knockdown models presents several technical challenges that researchers should consider when designing experiments:
Complete Knockout Challenges:
Embryonic lethality or severe developmental defects may complicate generation of viable knockout models, as suggested by TINAGL1's importance in embryonic development including heart and adrenal glands
Infertility in knockout mice makes breeding and maintaining knockout lines difficult, potentially requiring heterozygous breeding schemes
Tissue-Specific and Inducible Systems:
Development of conditional knockout systems (Cre-loxP) is recommended to circumvent embryonic lethality
Temporal control using inducible systems (e.g., tetracycline-controlled transcriptional activation) allows study of TINAGL1 function at specific developmental stages or in adult tissues
RNAi and CRISPR Approaches:
siRNA or shRNA approaches may achieve incomplete knockdown, necessitating validation of knockdown efficiency at both mRNA and protein levels
CRISPR-Cas9 editing requires careful gRNA design to minimize off-target effects
Verification of knockout by sequencing and functional assays is essential
Compensatory Mechanisms:
Related proteins (particularly paralog TINAG) may compensate for TINAGL1 loss, potentially masking phenotypes
Analysis of compensatory changes in gene expression should be incorporated into experimental design
Delivery Methods:
For in vivo manipulation, determining appropriate delivery vectors (viral vectors, nanoparticles) for tissue-specific targeting
Cell type-specific promoters may be necessary for targeted manipulation in complex tissues
Researchers should incorporate appropriate controls and validation steps to ensure the specificity and efficacy of their TINAGL1 manipulation strategy.
Several promising translational applications of TINAGL1 research are emerging in clinical medicine:
Biomarker for Ovarian Reserve Assessment:
The discovery that TINAGL1 levels are decreased in the follicular fluid of patients with diminished ovarian reserve (DOR) suggests significant potential as a novel biomarker for fertility assessment . This could provide an additional tool for clinicians evaluating patients with fertility concerns, potentially offering earlier or more accurate detection of DOR than current methods.
Cancer Diagnostics and Therapeutics:
TINAGL1's association with colorectal adenocarcinoma and its demonstrated ability to bind EGFR and integrins, influencing cancer proliferation and metastasis pathways , indicates potential applications in:
Diagnostic or prognostic stratification of cancer patients
Therapeutic targeting of TINAGL1-mediated pathways
Development of TINAGL1-based compounds that could disrupt tumor progression
Reproductive Medicine Interventions:
The critical role of TINAGL1 in fertility, supported by evidence that TINAGL1 knockout mice are infertile , suggests potential therapeutic approaches for certain forms of infertility through:
Modulation of TINAGL1 expression or activity
Development of recombinant TINAGL1 supplementation strategies
Screening for TINAGL1 genetic variants in unexplained infertility cases
Extracellular Matrix-Related Disorders:
As an extracellular matrix protein involved in cell adhesion, proliferation, migration, and differentiation , TINAGL1 may have relevance to conditions involving ECM dysfunction, potentially including fibrotic diseases or certain developmental disorders.
Future translational research should focus on validating these potential applications through larger clinical studies and deeper mechanistic investigations.
Several methodological advances would significantly accelerate our understanding of TINAGL1's physiological functions:
Advanced Imaging Techniques:
Implementation of super-resolution microscopy for precise localization of TINAGL1 within tissues and subcellular compartments
Live-cell imaging with fluorescently tagged TINAGL1 to track dynamics in real-time
Correlative light and electron microscopy to connect TINAGL1 localization with ultrastructural context
Single-Cell Analysis:
Single-cell RNA sequencing to identify cell populations expressing TINAGL1 and characterize heterogeneity in expression patterns
Single-cell proteomics to detect TINAGL1 protein levels and post-translational modifications at the individual cell level
Spatial transcriptomics to map TINAGL1 expression within the architectural context of tissues
Structural Biology Approaches:
Cryo-electron microscopy or X-ray crystallography of TINAGL1 to determine precise three-dimensional structure
Structural studies of TINAGL1 in complex with binding partners to elucidate interaction mechanisms
Molecular dynamics simulations to predict functional domains and binding interfaces
High-Throughput Functional Screening:
CRISPR activation/inhibition screens to identify genes that modify TINAGL1 function
Protein-protein interaction screens to map the complete TINAGL1 interactome
Small molecule screening to identify compounds that modulate TINAGL1 expression or activity
Integrative Multi-Omics:
Combined analysis of transcriptomics, proteomics, and metabolomics data following TINAGL1 modulation
Integration of epigenetic profiling to understand TINAGL1 regulation
Systems biology approaches to position TINAGL1 within broader biological networks
These methodological advances would provide a more comprehensive understanding of TINAGL1's diverse functions across different physiological contexts and potentially reveal new applications in both basic science and clinical medicine.
TINAGL1, also known as Tubulointerstitial Nephritis Antigen-Related Protein, is a glycoprotein that belongs to the peptidase C1 family. The mature human TINAGL1 protein contains several distinct domains, including an SMB (somatomedin B) domain, a vWFC (von Willebrand factor type C) domain, and a nonenzymatic peptidase C1A region . These structural features contribute to its functional versatility.
TINAGL1 is implicated in a variety of biological processes. It is known to play a role in the zonal differentiation of adrenocortical cells, where it is referred to as adrenocortical zonation factor 1 (AZ-1) or lipocalin 7 . Additionally, TINAGL1 has been associated with the regulation of gene expression in adrenocortical cells, particularly in repressing the expression of the CYP11B1 gene .
TINAGL1 has been studied extensively in the context of cancer. It has been proposed to both protect against cancer and contribute to pathological abnormalities in tumors . For instance, in diffuse-type gastric cancer (DGC), TINAGL1 secreted by cancer-associated fibroblasts (CAFs) has been shown to promote tumor progression by inducing the phosphorylation of focal adhesion kinase (FAK) in cancer cells . This interaction enhances the migration and tumorigenesis of DGC cells, making TINAGL1 a potential therapeutic target for this type of cancer.
The recombinant form of TINAGL1 is produced using advanced biotechnological methods to ensure high purity and activity. This form is particularly useful in research and therapeutic applications, as it allows for the detailed study of TINAGL1’s functions and interactions in a controlled environment. Recombinant TINAGL1 is utilized in various assays and experimental setups to investigate its role in different biological processes and diseases .