GIPC2 (GIPC PDZ domain-containing family member 2) is a tumor suppressor protein encoded by the GIPC2 gene located at chromosome 1p31.1 . It belongs to the GIPC family, which includes GIPC1 and GIPC3, and functions as an adaptor protein with a central PDZ domain for protein-protein interactions . GIPC2 is primarily expressed in adrenal, kidney, and colon tissues and plays critical roles in endocytosis, cell signaling, and tumor suppression . Its dysregulation is implicated in cancers such as pheochromocytomas (PPGLs), colon adenocarcinoma (COAD), and prostate cancer .
GIPC2 exhibits tissue-specific expression patterns, with high levels in normal adrenal medulla, renal tubules, and gastrointestinal tract . Its localization varies between cytoplasmic and nuclear compartments, depending on the tissue and cellular context .
GIPC2’s role in cancer is context-dependent:
GIPC2 promotes metastasis via interaction with Frizzled class receptor 7 (Fzd7), enhancing cell migration without affecting proliferation .
GIPC2 regulates key signaling pathways and transcriptional networks:
GIPC2 serves as a biomarker with divergent implications:
GIPC2 is leveraged in preclinical and translational studies:
Recombinant Protein: Used in functional assays (e.g., proliferation, apoptosis) .
Epigenetic Targeting: DNA methyltransferase inhibitors (e.g., 5-AZA) restore GIPC2 expression in PPGL models .
Immune Therapy: High GIPC2 expression in COAD predicts responsiveness to checkpoint inhibitors (e.g., PD-1/PD-L1) .
GIPC2 is a member of the GIPC family of proteins containing PDZ domains. It encodes a PDZ domain-containing adaptor protein with preferential expression in adrenal tissues. The protein is predominantly expressed in the nucleus, though it can also be found in other cellular compartments. GIPC2 contains structural motifs that enable protein-protein interactions, particularly through its PDZ domain, which facilitates binding with various signaling proteins that regulate cellular processes including proliferation and differentiation .
GIPC2 shows tissue-specific expression patterns with particularly high expression in normal adrenal and digestive tract tissues. In colorectal tissues, immunohistochemical analysis has revealed significant GIPC2 protein expression in normal colon epithelium. The Human Protein Atlas database confirms this distribution pattern, with notable expression in normal colon tissues compared to other tissue types . This tissue-specific expression pattern suggests specialized functional roles in these tissues that may be disrupted during pathological processes.
Based on current research, GIPC2 appears to function in several cellular processes including:
Cell cycle regulation through interaction with cell cycle proteins such as p27
Suppression of MAPK/Erk signaling pathways
Involvement in tight junction organization and epithelial cell signaling
Regulation of microvillus organization
Participation in DNA replication and mitosis-associated processes
Potential modulation of immune cell infiltration within tumor microenvironments
GIPC2 expression varies significantly across cancer types, with distinct patterns emerging:
Downregulated in:
Colorectal cancer/colon adenocarcinoma (COAD): Significantly reduced expression compared to normal colon tissues
Pheochromocytoma (PCC): Reduced genomic copy number and expression in sporadic cases
Upregulated in:
For comprehensive evaluation of GIPC2 expression in research settings, multiple complementary approaches should be employed:
Transcriptomic analysis: RNA-seq or qRT-PCR to quantify mRNA expression levels
Genomic analysis: SNP arrays to identify copy number variations
Protein detection: Western blotting and immunohistochemistry to visualize protein expression and localization
Methylation analysis: MassARRAY EpiTYPER assay and methylation-specific PCR to detect epigenetic modifications of the GIPC2 promoter region
Researchers should consider using multiple methods, as studies have shown GIPC2 regulation occurs at genomic, transcriptomic, and epigenetic levels . For the most reliable results, validation across different techniques is strongly recommended.
GIPC2 expression demonstrates significant correlations with clinical parameters in multiple cancer types:
In colorectal cancer, GIPC2 expression correlates significantly with:
In colon adenocarcinoma, GIPC2 expression is significantly associated with:
GIPC2 appears to interact with multiple signaling pathways, with effects that vary by tissue context:
Cell cycle regulation pathways: GIPC2 influences cell cycle checkpoints, DNA replication, G1-S transition, G2-M checkpoints, and mitotic processes
MAPK/Erk pathway: In pheochromocytoma, GIPC2 suppresses activation of MAPK/Erk pathways without affecting pAKT or mTOR pathways
Epithelial cell signaling: GIPC2 co-expressed genes are enriched in epithelial cell signaling, tight junction formation, and peroxisome function
Wnt signaling: In prostate cancer, GIPC2 interacts with Fzd7, suggesting involvement in Wnt signaling pathways that promote metastasis
Gene set enrichment analysis has identified significant enrichment of GIPC2-related pathways in cell cycle checkpoints, DNA replication, retinoblastoma gene in cancer, and sister chromatid separation pathways .
GIPC2 operates through protein-protein interactions mediated primarily through its PDZ domain:
NONO interaction: In pheochromocytoma, GIPC2 interacts with NONO (a nucleoprotein with known roles in cell cycle regulation), which was demonstrated through mass spectrometry and confirmed with reciprocal interaction studies
Top co-expressed proteins: Analysis of GIPC2 co-expression networks reveals strong correlations with EPCAM, LRRC8D, EPB41L4B, ACSL5, and CDS1 in colon adenocarcinoma
Fzd7 interaction: In prostate cancer, GIPC2 has been reported to interact with Fzd7, potentially promoting cancer metastasis
p27 regulation: GIPC2 appears to regulate the cell cycle gene p27, suggesting a mechanism for its tumor suppressor function in some contexts
These diverse interactions help explain the context-specific functions of GIPC2 across different tissue types and disease states.
For comprehensive investigation of GIPC2's molecular mechanisms, researchers should consider multiple experimental approaches:
Protein interaction studies:
Co-immunoprecipitation coupled with mass spectrometry to identify binding partners
Proximity ligation assays to confirm interactions in situ
Yeast two-hybrid screening for novel interactors
Functional studies:
GIPC2 overexpression and siRNA knockdown in appropriate cell lines (e.g., PC12, hPheo for pheochromocytoma, CRC cell lines for colorectal cancer)
CRISPR/Cas9-mediated gene editing to create cellular models
Xenograft models to evaluate GIPC2's role in tumor formation and progression
Pathway analysis:
Phospho-protein arrays to detect changes in signaling pathway activation
RNA-seq followed by GSEA for pathway enrichment analysis
ChIP-seq to identify potential transcriptional targets
These complementary approaches provide a comprehensive assessment of GIPC2's mechanistic roles in both normal and pathological contexts.
Multiple lines of evidence support GIPC2's function as a tumor suppressor, particularly in certain cancer types:
GIPC2 appears to modulate cell cycle progression through several mechanisms:
Cyclin-dependent kinase inhibition: GIPC2 induces p27, a cyclin-dependent kinase inhibitor that blocks cell cycle progression
MAPK/Erk pathway suppression: GIPC2 inhibits the activation of MAPK/Erk pathways, which are known to promote cell proliferation
Cell cycle checkpoint regulation: Gene set enrichment analysis shows GIPC2 expression is associated with pathways controlling cell cycle checkpoints, particularly G1-S and G2-M transitions
DNA replication control: GIPC2 influences DNA replication pathways, potentially affecting S-phase progression
Mitotic regulation: GIPC2 expression is associated with pathways controlling mitotic metaphase, anaphase, and sister chromatid separation
These mechanisms collectively contribute to GIPC2's apparent anti-proliferative effects in certain cellular contexts.
Developing therapeutic approaches targeting GIPC2 presents several significant challenges:
Context-dependent expression: GIPC2 functions as a tumor suppressor in some cancers (colorectal, pheochromocytoma) but appears upregulated in metastatic prostate cancer, suggesting potentially opposing roles in different contexts
Protein-protein interaction targeting: As GIPC2 functions through protein-protein interactions rather than enzymatic activity, developing small molecule inhibitors is more challenging
Delivery mechanisms: Restoring GIPC2 expression in cancers where it's downregulated would require gene therapy approaches that face delivery and expression control challenges
Specificity concerns: GIPC2 belongs to a family of related proteins, raising potential off-target effects when developing targeting strategies
Pathway complexity: GIPC2 interacts with multiple pathways and proteins, making it difficult to predict downstream effects of its modulation
Any therapeutic approach would need to account for tissue-specific effects and ensure that intervention doesn't promote unwanted effects in tissues where GIPC2 may play different roles .
The relationship between GIPC2 expression and cancer metastasis appears to be cancer-type dependent:
Several factors may explain GIPC2's seemingly contradictory roles across cancer types:
Tissue-specific interaction partners: GIPC2 interacts with different proteins depending on the cellular context (NONO in pheochromocytoma, Fzd7 in prostate cancer)
Pathway specificity: The dominant signaling pathways affected by GIPC2 vary by tissue (MAPK/Erk in pheochromocytoma, potential Wnt signaling in prostate cancer)
Genetic context: The broader mutational landscape of each cancer type likely influences how GIPC2 alterations affect cellular behavior
Epigenetic regulation: Different patterns of promoter methylation and other epigenetic modifications may result in context-specific GIPC2 regulation
Splice variants: Potential tissue-specific splice variants of GIPC2 might have different functional properties
Understanding these context-dependent mechanisms requires tissue-specific research approaches rather than generalizing findings across cancer types.
When faced with conflicting data about GIPC2 across different experimental models, researchers should:
Carefully validate experimental systems: Ensure cell lines and animal models accurately reflect the cancer type being studied
Consider tissue context: Recognize that GIPC2 may have fundamentally different roles in different tissues
Account for genetic background: Document the full genetic context of experimental models, as other mutations may influence GIPC2 function
Use complementary approaches: Employ both in vitro and in vivo models, and validate findings across multiple experimental systems
Utilize patient-derived models: When possible, use patient-derived xenografts or organoids to better recapitulate the complexity of human tumors
Perform detailed molecular profiling: Characterize GIPC2 expression, localization, interaction partners, and pathway activation in each model
Consider temporal aspects: Examine GIPC2's role across different stages of cancer progression within the same model
This methodical approach can help resolve apparent contradictions and develop a more nuanced understanding of GIPC2's context-dependent functions.
Recent research has revealed significant associations between GIPC2 expression and immune cell infiltration, particularly in colon adenocarcinoma:
Positive correlation with immune infiltration: High GIPC2 expression in colon adenocarcinoma is associated with increased infiltration of several immune cell types
Specific immune cell types affected: Analysis using the CIBERSORT algorithm demonstrated that plasma B cells, resting CD4+ memory T cells, activated CD4+ memory T cells, activated myeloid dendritic cells, and activated mast cells were present in significantly higher proportions in tumors with high GIPC2 expression
Immune checkpoint association: Significant differences in the expression of immune checkpoint-associated genes (HAVCR2, LAG3, PDCD1, and SIGLEC15) between high and low GIPC2 expression groups were observed
These findings suggest GIPC2 may influence the tumor immune microenvironment, potentially affecting immunotherapy responsiveness.
To comprehensively investigate GIPC2's influence on tumor immune microenvironment, researchers should consider these methodological approaches:
Computational methods:
CIBERSORT algorithm for deconvolution of immune cell types from bulk RNA-seq data
Gene set enrichment analysis (GSEA) to identify immune-related pathways associated with GIPC2 expression
Correlation analysis between GIPC2 and immune checkpoint genes
Laboratory techniques:
Multiplex immunofluorescence to visualize and quantify immune cell populations in relation to GIPC2 expression
Flow cytometry for detailed immune cell profiling in models with GIPC2 modulation
Single-cell RNA sequencing to characterize both immune and tumor cell populations
Co-culture experiments with immune cells and cancer cells with varying GIPC2 expression
Functional assays:
T cell activation and cytotoxicity assays in the presence of GIPC2-modulated cancer cells
Cytokine profiling to assess immune signaling changes
In vivo models evaluating tumor growth and immune infiltration with GIPC2 modulation
These complementary approaches provide a comprehensive assessment of how GIPC2 may influence the complex interactions between tumor cells and the immune system.
Based on current evidence, GIPC2 expression shows potential as a biomarker for immunotherapy response, particularly in colon adenocarcinoma:
Immune checkpoint gene correlation: Significant associations between GIPC2 expression and immune checkpoint genes (HAVCR2, LAG3, PDCD1, SIGLEC15) suggest potential relevance to checkpoint inhibitor therapy
Immune cell infiltration: The positive correlation between high GIPC2 expression and increased immune cell infiltration suggests GIPC2 may identify "hot" tumors more likely to respond to immunotherapy
Favorable prognosis association: High GIPC2 expression correlates with better patient outcomes in colon adenocarcinoma, which could indicate immune-mediated tumor control
Potential mechanism identification: Understanding GIPC2's role in immune regulation could identify novel therapeutic targets or combination strategies
Further research is needed to validate GIPC2 as an immunotherapy response biomarker, including prospective clinical studies correlating GIPC2 expression with response to various immunotherapeutic approaches.
Several promising therapeutic strategies targeting GIPC2-related pathways are emerging:
Protein-protein interaction inhibitors: Developing small molecules or peptides that disrupt specific interactions between GIPC2 and its binding partners (e.g., NONO in pheochromocytoma or Fzd7 in prostate cancer)
Epigenetic modifiers: Using DNA methyltransferase inhibitors to reverse GIPC2 promoter hypermethylation in cancers where it acts as a tumor suppressor
Gene therapy approaches: Delivering functional GIPC2 to tumors with reduced expression, potentially using tumor-targeting nanoparticles or viral vectors
Combination strategies: Combining GIPC2-targeted therapies with immune checkpoint inhibitors, particularly in contexts where GIPC2 expression correlates with immune cell infiltration
Pathway-specific interventions: Targeting downstream effectors in GIPC2-regulated pathways, such as cell cycle components or MAPK/Erk signaling molecules
Preclinical data has identified potential inhibitors able to bind to WNT receptor complexes that may prevent prostate cancer metastasis, which could be relevant given GIPC2's reported interaction with Fzd7 .
Single-cell technologies offer several advantages for unraveling GIPC2's complex functions:
Cellular heterogeneity resolution: Single-cell RNA sequencing can reveal cell-type specific expression patterns of GIPC2 within complex tissues and tumor microenvironments
Co-expression network analysis: Identifying cell-specific co-expression patterns can reveal context-dependent GIPC2 interaction networks
Spatial transcriptomics: Mapping GIPC2 expression in spatial context can reveal relationships to tissue architecture and microenvironmental factors
Clonal evolution tracking: Following GIPC2 expression changes during tumor progression and treatment response at clonal resolution
Integrated multi-omics: Combining single-cell transcriptomics with proteomics and epigenomics to comprehensively characterize GIPC2 regulation
These approaches could help resolve contradictory findings regarding GIPC2's role in different cancer types by identifying specific cellular contexts where it functions as either a tumor suppressor or promoter.
Several critical knowledge gaps should be prioritized in future GIPC2 research:
Tissue-specific mechanisms: More detailed investigation of why GIPC2 functions differently across tissue types and cancer contexts
Structural biology: Crystal structures of GIPC2 in complex with key interaction partners would facilitate targeted therapeutic development
In vivo models: Development of conditional knockout or overexpression models to study GIPC2 function in physiologically relevant contexts
Clinical validation: Prospective studies validating GIPC2's prognostic and predictive value in larger, diverse patient cohorts
Therapeutic targeting: Development and testing of compounds that modulate GIPC2 expression or function
Immune regulatory mechanisms: Deeper investigation of how GIPC2 influences immune cell infiltration and function in the tumor microenvironment
Epigenetic regulation: Comprehensive characterization of the mechanisms controlling GIPC2 expression across different tissues and disease states Addressing these knowledge gaps will require collaborative efforts across structural biology, molecular oncology, immunology, and clinical research disciplines.
The GIPC2 gene is located on chromosome 1p31.1 and consists of six exons . The protein encoded by this gene is composed of 315 amino acids and contains a central PDZ domain . This domain is crucial for the protein’s function, as it mediates interactions with other proteins. GIPC2 shares 62% amino acid identity with GIPC1, another member of the GIPC family, and 59.6% identity with Kermit, the Xenopus ortholog of GIPC1 .
GIPC2 is expressed in various tissues, with moderate expression levels detected in the adult human kidney, pancreas, liver, and spleen . It is also expressed throughout the gastrointestinal tract, from the stomach to the rectum, with higher expression levels in the ascending colon . The protein is located in extracellular exosomes, which are small vesicles released into the extracellular environment and play a role in cell-to-cell communication .
GIPC2 is involved in several cellular processes, primarily through its interactions with other proteins. One of its notable interactions is with the insulin-like growth factor 1 receptor (IGF1R), which plays a role in regulating growth and development . GIPC2 has been shown to interact with eukaryotic initiation factor 6 (EIF6), modulating its expression in a dose-dependent manner to regulate eye morphogenesis in Xenopus . This interaction highlights the importance of GIPC2 in developmental processes.
Mutations or dysregulation of the GIPC2 gene have been associated with certain diseases. For example, GIPC2 has been linked to pheochromocytoma, a type of tumor that arises from the adrenal gland . Understanding the role of GIPC2 in these diseases can provide insights into potential therapeutic targets and strategies.
Recombinant GIPC2 refers to the protein that has been genetically engineered and produced in a laboratory setting. This recombinant protein is used in various research applications to study its function, interactions, and potential therapeutic uses. By using recombinant GIPC2, researchers can gain a deeper understanding of its role in cellular processes and disease mechanisms.