Prostate Cancer: RARRES1 suppresses invasion and colony-forming ability in prostate cancer cells by inhibiting stem cell (SC) properties. Its expression is induced by all-trans retinoic acid (atRA) in differentiated cells .
Renal Cell Carcinoma (KIRC): RARRES1 interacts with ICAM1 to activate M1 macrophages, reducing tumor viability and promoting apoptosis .
DNA Methylation: Silenced via promoter hypermethylation in prostate, breast, and nasopharyngeal cancers .
RARRES1 depletion reprograms glucose metabolism, shifting cells from aerobic glycolysis to glucose-dependent lipid synthesis .
Modulates mTOR and SIRT1 pathways to induce autophagy, a key process in energy homeostasis .
Correlates with macrophage infiltration in KIRC and regulates cell adhesion pathways (e.g., ICAM1) .
Enhances immune cell recruitment (B cells, neutrophils) in tumor microenvironments .
Retinoic Acid Therapy: atRA upregulates RARRES1, suppressing invasion and promoting differentiation in prostate cancer .
Metabolic Targeting: RARRES1 depletion increases detyrosinated tubulin, altering mitochondrial membrane potential and AMPK activation .
Retinoic acid receptor responder protein 1 isoform 1, TIG1, Retinoic acid receptor responder protein 1, RAR-responsive protein TIG1, RARRES1, Recombinant Human Retinoic Acid Receptor Responder 1.
RARRES1 is a tumor suppressor protein whose expression is frequently suppressed in various tumor cells. In normal tissues, it functions as a key regulator of cell adhesion processes, which is evident from Gene Ontology (GO) biological process analyses showing that RARRES1-correlated genes are most significantly enriched in pathways related to regulation of cell adhesion . Additionally, RARRES1 plays important roles in protein maturation, negative regulation of hydrolase activity, and protein hydroxylation as revealed by Metascape analysis of RARRES1 and its 284 related genes . The protein primarily exerts suppressive effects on cellular invasion and migration in normal epithelial tissues, acting as a barrier to malignant transformation.
RARRES1 expression is regulated through multiple mechanisms, with epigenetic control being particularly important. Studies have demonstrated that RARRES1 expression can be altered through both genetic alterations and epigenetic regulation, contributing to its varied expression patterns in different tissue contexts . Retinoic acid has been shown to induce RARRES1 expression, as suggested by its name (Retinoic Acid Receptor Responder 1). The regulation of RARRES1 appears to be tissue-specific, with different regulatory networks controlling its expression in various cell types. In cancer progression, methylation of the RARRES1 promoter frequently leads to its silencing, which suggests that demethylating agents might potentially restore its tumor suppressive functions.
RARRES1 expression in human tissues can be detected through several methodological approaches:
Immunohistochemistry (IHC): As demonstrated in the research on renal cell carcinoma (RCC), RARRES1 can be detected using specific antibodies (such as HPA003892, Sigma Aldrich) at appropriate dilutions (1:250) . The immunostaining results can be categorized as membranous, cytoplasmic, or negative .
Quantitative PCR (qPCR): Used to measure RARRES1 mRNA expression levels, as shown in studies examining the effects of RARRES1 overexpression in renal carcinoma cells .
Western Blotting: For protein-level detection and quantification.
Tissue Microarray Analysis: Used for high-throughput analysis of RARRES1 expression across multiple tissue samples, with cores typically taken from representative tumor areas of different morphology and/or nuclear grade .
Bioinformatic Analysis: Tools like GEPIA and TIMER are used to analyze correlation between RARRES1 and other genes or immune cell infiltration levels .
When performing IHC for RARRES1, it's recommended to use normal foetal and adult kidney samples as positive controls, and omission of primary antibody serves as negative control .
RARRES1 expression shows complex correlations with patient survival that vary by cancer type. In kidney renal clear cell carcinoma (KIRC), research has revealed a negative correlation between RARRES1 expression and patient survival time . This seemingly paradoxical finding—where a tumor suppressor gene correlates with poorer outcomes—may be explained by the complexity of the tumor microenvironment.
As explained by researchers, this apparent contradiction might occur because "tumors are abnormal organs composed of multiple cell types and extracellular matrix rather than simply clones of cancer cells." In KIRC cases, high RARRES1 expression may indicate advanced tumor malignancy, where RARRES1 is actively recruiting macrophages to suppress tumor growth, but the suppressive effects are insufficient to counteract the aggressive malignancy .
In contrast, studies in other cancer types like prostate cancer and triple-negative breast cancer have identified RARRES1 as an invasion suppressor, potentially with more straightforward positive correlations with survival .
RARRES1 demonstrates significant relationships with immune cell infiltration in tumor microenvironments, particularly in KIRC:
RARRES1 expression shows a negative correlation with tumor purity in KIRC, suggesting higher levels in samples with greater immune cell infiltration .
Strong positive correlations exist between RARRES1 expression and infiltration of multiple immune cell types, including:
More specifically, RARRES1 expression significantly correlates with:
Among these relationships, the correlation with macrophages is notably the strongest, suggesting a particular importance of macrophage interactions in RARRES1's tumor suppressive functions.
Distinguishing between direct tumor-suppressive effects of RARRES1 and its immune-mediated effects requires a multi-faceted experimental approach:
In vitro monoculture experiments: Researchers should first test the direct effects of RARRES1 overexpression or knockdown on cancer cell lines in isolation, measuring parameters like proliferation, apoptosis, migration, and invasion without immune cell presence.
Co-culture systems: As demonstrated in the KIRC studies, Transwell co-culture systems can be employed to examine the interactions between RARRES1-expressing tumor cells and immune cells without direct contact .
Mechanistic blocking experiments: Selective blocking of specific pathways (such as ICAM1-Mac-1 interaction) can help determine which effects are dependent on immune cell interactions versus intrinsic to the tumor cells .
Immunodeficient mouse models: Comparing the behavior of RARRES1-modulated tumors in immunocompetent versus immunodeficient mice can help differentiate immune-dependent from immune-independent effects.
Pathway analysis: Comprehensive gene expression profiling after RARRES1 modulation, with and without immune cells present, can identify distinct signaling pathways involved in direct versus immune-mediated effects.
In KIRC research, scientists used a combination of these approaches to demonstrate that RARRES1 exerts antitumor effects primarily by promoting ICAM1 expression and subsequent M1 macrophage activation, rather than through direct tumor cell inhibition alone .
RARRES1 plays a crucial role in regulating cell adhesion processes, which has significant implications for cancer invasion and metastasis:
Regulation of adhesion-related genes: Gene Ontology analysis revealed that the biological process most significantly enriched with RARRES1-correlated genes is "regulation of cell adhesion" . This pathway includes 26 genes such as ADA, BCL2, and importantly, ICAM1 .
ICAM1 upregulation: RARRES1 overexpression in renal carcinoma cells (Caki-1) significantly increases ICAM1 expression at both mRNA and protein levels . This upregulation represents a critical mechanism by which RARRES1 influences cell adhesion.
Effect on cell-cell interactions: By enhancing ICAM1 expression, RARRES1 modifies how tumor cells interact with immune cells, particularly macrophages through the ICAM1-Mac-1 binding interaction .
The implications for cancer invasion are substantial since cell adhesion is a critical first step in cancer metastasis. By maintaining proper cell adhesion, RARRES1 may help prevent detachment of tumor cells from the primary site, potentially limiting their invasive and metastatic capabilities. This is consistent with RARRES1's documented role as an invasion suppressor in prostate cancer and triple-negative breast cancer .
Furthermore, by enhancing ICAM1-Mac-1 interactions, RARRES1 facilitates macrophage recognition and attack of tumor cells, creating an additional barrier to cancer progression through immune surveillance enhancement.
The molecular mechanism of interaction between RARRES1 and ICAM1 involves a regulatory relationship where RARRES1 enhances ICAM1 expression, which subsequently influences immune cell interactions. The current research suggests this is a gene regulatory relationship rather than a direct protein-protein interaction.
To experimentally verify this mechanism, researchers have employed several approaches:
RARRES1 overexpression systems: Using lentiviral vectors to overexpress RARRES1 in renal carcinoma cells (Caki-1), researchers demonstrated increased ICAM1 expression at both mRNA and protein levels .
qPCR analysis: Measurement of ICAM1 mRNA expression in RARRES1-overexpressing cells confirmed the upregulation of ICAM1 transcription .
ELISA: Quantification of ICAM1 protein in cell supernatants showed significantly increased secretion after RARRES1 overexpression in RCC cells .
Co-immunoprecipitation (Co-IP): To examine the functional consequences of RARRES1-induced ICAM1 upregulation, researchers performed Co-IP assays to assess the binding between ICAM1 and Mac-1 (CD11b/CD18) on macrophages. This revealed that RARRES1 overexpression in RCC cells promoted the interaction between ICAM1 and Mac-1 .
Coculture systems: Transwell coculture of RARRES1-overexpressing RCC cells with M1-polarized THP-1 macrophages allowed observation of the functional consequences of altered ICAM1 expression .
Additional methods that could further elucidate this mechanism include:
ChIP assays to determine if RARRES1 influences ICAM1 promoter activity
Luciferase reporter assays with ICAM1 promoter constructs
siRNA knockdown of potential intermediate signaling molecules to identify the pathway connecting RARRES1 to ICAM1 expression
While ICAM1 is the most well-documented cell adhesion molecule interacting with the RARRES1 pathway in KIRC, gene correlation and functional analyses suggest several other adhesion-related molecules may be involved in RARRES1 signaling networks:
ADA and BCL2: These were identified alongside ICAM1 in the group of 26 genes included in the GO biological process "regulation of cell adhesion" that correlate with RARRES1 expression .
CMTM7, PLAUR, and IL23A: These genes showed significant positive correlations with both RARRES1 expression and macrophage infiltration in KIRC tissues, suggesting their potential involvement in RARRES1-mediated adhesion and immune cell interaction networks .
Mac-1 (CD11b/CD18) complex: While expressed on macrophages rather than tumor cells, this integrin receptor for ICAM1 represents a critical component of the RARRES1-influenced adhesion pathway .
Additional research would be beneficial to:
Perform proteomic analyses of RARRES1-overexpressing cells to identify altered expression of other adhesion molecules
Conduct functional screening using siRNA libraries targeting adhesion molecules to identify those that influence RARRES1's tumor suppressive effects
Investigate whether RARRES1 affects expression of cadherins, selectins, or other integrins that play established roles in cancer cell adhesion and metastasis
Optimal experimental models for studying RARRES1 function in human cancers should be selected based on research objectives and specific cancer types. Based on current research approaches, these models include:
Cell line models:
For kidney cancer: Caki-1 cells have been successfully used to study RARRES1 overexpression effects
THP-1 monocytic cells (differentiated to macrophages) provide valuable immune cell interaction models
Other cell lines representing cancers where RARRES1 functions as an invasion suppressor (prostate, triple-negative breast cancer) should be considered for comparative studies
Co-culture systems:
Transwell co-culture systems allow examination of paracrine interactions without direct cell contact
3D co-culture models provide more physiological representations of tumor-immune interactions
Animal models:
Xenograft models with RARRES1-modulated cancer cells in immunodeficient mice
Syngeneic mouse models with intact immune systems for studying immune interactions
Genetically engineered mouse models with conditional RARRES1 knockout/overexpression
Patient-derived models:
Patient-derived xenografts (PDXs) maintain tumor heterogeneity
Patient-derived organoids allow study of RARRES1 in more complex 3D structures
Ex vivo culture of tumor slices with preserved microenvironment
Bioinformatic approaches:
Tools like TIMER, GEPIA, and UALCAN databases for correlation analyses
Analysis of TCGA datasets for different cancer types to identify cancer-specific patterns
For kidney cancer specifically, the combination of Caki-1 cells (for RARRES1 overexpression), THP-1-derived macrophages (for immune interactions), and Transwell co-culture systems has proven effective for mechanistic studies .
Accurate quantification of RARRES1-mediated immune cell recruitment and activation requires a multi-parameter approach:
In vitro migration assays:
Transwell migration assays to quantify immune cell chemotaxis toward RARRES1-expressing or control cancer cells
Time-lapse microscopy to track immune cell movement in real-time
Flow cytometry:
Cytokine/chemokine profiling:
ELISA or multiplex assays to measure secreted factors
qPCR for cytokine/chemokine gene expression analysis
Protein interaction assays:
Functional readouts:
In vivo immune monitoring:
Immunohistochemistry of tumor sections to quantify immune infiltrates
Flow cytometry of dissociated tumors to characterize immune populations
In vivo imaging of fluorescently labeled immune cells
Single-cell approaches:
Single-cell RNA sequencing to identify transcriptional changes in immune cells
CyTOF (mass cytometry) for high-dimensional phenotyping of immune cells
In the context of RARRES1-ICAM1-macrophage interactions, researchers have effectively used qPCR to measure CD86 expression (M1 marker), Co-IP to assess ICAM1-Mac-1 binding, and functional assays to measure the impact on tumor cell viability and apoptosis .
Several computational approaches can help predict novel interactions and functions of RARRES1 in human disease:
Co-expression network analysis:
Pearson correlation analysis of RARRES1 with other genes across tumor samples can identify functionally related genes, as demonstrated in KIRC studies where 164 positively and 120 negatively correlated genes were identified
Weighted gene co-expression network analysis (WGCNA) to identify modules of genes with similar expression patterns
Pathway enrichment analysis:
Protein-protein interaction (PPI) networks:
Immune infiltration correlation analysis:
Multi-omics integration:
Integration of transcriptomic, proteomic, and epigenomic data to build comprehensive models
Correlation of RARRES1 expression with mutation profiles, copy number alterations, and methylation patterns
Machine learning approaches:
Supervised learning to predict patient outcomes based on RARRES1 expression and related features
Unsupervised clustering to identify patient subgroups with distinct RARRES1-related patterns
Deep learning to identify complex patterns in imaging data related to RARRES1 expression
Text mining and knowledge graphs:
Natural language processing of scientific literature to extract relationships related to RARRES1
Construction of knowledge graphs connecting RARRES1 to diseases, pathways, and drugs
These computational approaches, particularly when integrated, can generate testable hypotheses about novel RARRES1 functions and interactions that can then be validated experimentally.
RARRES1 expression patterns show significant potential for patient stratification in precision oncology, particularly in kidney cancer:
Prognostic stratification:
Immune response prediction:
Strong correlations between RARRES1 expression and immune cell infiltration (particularly macrophages, B cells, neutrophils, and dendritic cells) suggest RARRES1 could predict immunotherapy response
Patients with high RARRES1 expression might benefit more from immunotherapies that enhance macrophage-mediated tumor killing
Treatment selection guidance:
Combination therapy design:
Patients with altered RARRES1-ICAM1 pathway might benefit from combination therapies targeting both tumor cells and enhancing immune cell activation
RARRES1 status could help identify patients who would benefit from macrophage-targeting therapies
Monitoring disease progression:
Serial assessment of RARRES1 expression could potentially serve as a biomarker for monitoring disease progression and treatment response
To implement RARRES1-based stratification in clinical practice, standardized assessment methods (such as the immunohistochemical approach described in the RCC studies) would need to be validated in larger, prospective clinical trials .
Several approaches can potentially restore RARRES1 expression in cancers where it is suppressed:
Epigenetic modulation:
DNA methyltransferase inhibitors (DNMTi) like 5-azacytidine or decitabine could potentially reverse hypermethylation of the RARRES1 promoter
Histone deacetylase inhibitors (HDACi) might enhance chromatin accessibility at the RARRES1 locus
Retinoid therapy:
Given that RARRES1 is a retinoic acid receptor responder, treatment with retinoids (vitamin A derivatives) like all-trans retinoic acid (ATRA) could potentially induce RARRES1 expression
Synthetic retinoids or selective retinoic acid receptor modulators might offer more targeted approaches
Gene therapy approaches:
Small molecule screening:
High-throughput screening to identify compounds that specifically induce RARRES1 expression
Drug repurposing studies to identify approved drugs that might incidentally increase RARRES1 levels
Indirect pathway modulation:
Targeting upstream regulators of RARRES1 expression
Inhibiting pathways that suppress RARRES1 expression
Combined approaches:
Simultaneous targeting of RARRES1 and downstream effectors like ICAM1 to enhance therapeutic efficacy
Combining RARRES1 restoration with immune checkpoint inhibitors to potentiate anti-tumor immune responses
Experimental validation of these approaches would require demonstration not only of restored RARRES1 expression but also confirmation of functional outcomes, such as enhanced ICAM1 expression, increased macrophage activation, and ultimately, reduced tumor growth.
The RARRES1-ICAM1-macrophage axis presents several promising avenues for novel immunotherapeutic strategies:
Enhancing RARRES1 expression:
Development of small molecules or biologics that induce RARRES1 expression
Epigenetic modifiers targeting RARRES1 promoter methylation
Gene therapy approaches to deliver functional RARRES1
Boosting ICAM1-Mac-1 interactions:
Development of agonistic antibodies that enhance ICAM1-Mac-1 binding
Engineering of ICAM1 variants with increased affinity for Mac-1
Local delivery of recombinant ICAM1 to tumor sites
Macrophage-targeted approaches:
Combination therapies:
RARRES1/ICAM1 enhancement plus immune checkpoint inhibitors
Combining macrophage-targeting strategies with T-cell-focused immunotherapies
Sequential therapy to first promote M1 polarization, then enhance ICAM1-Mac-1 binding
Tumor microenvironment modulation:
Strategies to overcome immunosuppressive factors that might dampen RARRES1-induced macrophage activation
Targeting the extracellular matrix to facilitate macrophage infiltration and contact with tumor cells
Monitoring and companion diagnostics:
Development of biomarkers to identify patients likely to respond to therapies targeting this axis
Real-time monitoring of macrophage polarization and activation during treatment
Research has demonstrated that "the interaction of RARRES1 with ICAM1 modulating macrophages may be a new target for immunotherapy of kidney renal clear cell carcinoma" . This approach could be particularly valuable since macrophage-based therapies that augment macrophage functionalities with antitumor activity represent an emerging area in cancer immunotherapy .
The paradoxical finding that RARRES1 functions as a tumor suppressor yet correlates with poorer survival in some cancers like KIRC presents a significant interpretive challenge. Researchers can address this contradiction through several approaches:
Context-dependent analysis:
Stratify patients by disease stage, molecular subtype, and treatment history to determine if RARRES1's prognostic significance varies across contexts
Analyze whether RARRES1's correlation with survival is modified by other factors (e.g., immune infiltration levels, mutation status of other genes)
Mechanistic dissection:
Investigate whether RARRES1 expression in poor-prognosis tumors represents a failed compensatory mechanism, as suggested by researchers who noted that "high expression of RARRES1 may indicate high degree of tumor malignancy and RARRES1 is recruiting more macrophages to suppress tumor. But the tumor microenvironment is complex, the tumor suppressor effects of RARRES1 may fail to counteract malignant tumor"
Examine whether RARRES1's function differs qualitatively (not just quantitatively) in advanced versus early-stage tumors
Time-course studies:
Analyze RARRES1 expression changes during disease progression to determine if its role evolves over time
Investigate whether initially protective RARRES1-driven immune responses might eventually lead to immune exhaustion or adaptation
Functional heterogeneity analysis:
Determine if RARRES1's effects vary across different regions of heterogeneous tumors
Examine single-cell data to identify if specific cell populations within tumors respond differently to RARRES1
Alternative splicing and post-translational modifications:
Investigate whether advanced tumors express functionally distinct RARRES1 isoforms or variants
Analyze whether post-translational modifications alter RARRES1's function in advanced disease
Spatial context analysis:
Employ spatial transcriptomics or multiplexed immunohistochemistry to understand how RARRES1's effects depend on its location within the tumor microenvironment
Analyze whether RARRES1's proximity to specific immune populations affects its function
Understanding this paradox will likely require integrating multiple approaches and may ultimately reveal that RARRES1 represents a marker of aggressive disease that simultaneously attempts (but fails) to mount an effective anti-tumor response.
Designing experiments to study RARRES1-mediated macrophage activation requires careful attention to several technical considerations:
Macrophage source and polarization:
Coculture system design:
Direct contact versus Transwell systems (allowing soluble factor exchange without cellular contact)
Ratio of tumor cells to macrophages (typically 1:1 in initial studies)
Duration of coculture (optimized to observe activation without exhaustion)
RARRES1 expression modulation:
Controls and blocking experiments:
Readout selection:
Timing considerations:
Temporal analysis of macrophage activation and subsequent tumor cell effects
Sequential vs. simultaneous manipulation of pathway components
Physiological relevance:
Oxygen tension (normoxic vs. hypoxic conditions)
Inclusion of other stromal components (fibroblasts, extracellular matrix)
3D versus 2D culture systems
Technical validation:
Reproducibility across different cell lines and macrophage sources
Confirmation of key findings using multiple complementary techniques
Verification in more complex systems (e.g., ex vivo tissue cultures, animal models)
Following these considerations will help ensure robust, reproducible results that accurately reflect the biological processes of RARRES1-mediated macrophage activation.
Single-cell analysis techniques offer powerful approaches to dissect RARRES1 function in heterogeneous tumor samples:
Single-cell RNA sequencing (scRNA-seq):
Reveals cell-type-specific expression patterns of RARRES1 and related genes
Identifies distinct cellular populations that express or respond to RARRES1
Maps transcriptional changes in immune cells (especially macrophages) in RARRES1-high versus RARRES1-low tumor regions
Constructs cell-type-specific gene regulatory networks involving RARRES1
Single-cell proteomics:
Mass cytometry (CyTOF) to simultaneously measure multiple proteins at single-cell resolution
Allows correlation of RARRES1 expression with activation states of multiple immune cell types
Enables detection of rare cell populations that might be critical for RARRES1 function
Spatial transcriptomics and proteomics:
Visium, Slide-seq, or MERFISH to map RARRES1 expression within the spatial context of tumors
Correlation of RARRES1 expression with immune cell localization and activation state
Identification of spatial relationships between RARRES1-expressing cells and ICAM1+ or Mac-1+ cells
Single-cell multiomics:
CITE-seq (combining transcriptomics with surface protein measurement)
Single-cell ATAC-seq to examine chromatin accessibility at the RARRES1 locus across cell types
Integration of genomic, transcriptomic, and epigenomic data from the same cells
Lineage tracing:
Tracking the fate of RARRES1-expressing cells over time in model systems
Determining whether RARRES1 expression changes during tumor evolution
Live cell imaging at single-cell resolution:
Real-time visualization of interactions between RARRES1-expressing tumor cells and macrophages
Monitoring of dynamic processes like macrophage migration, contact duration, and tumor cell killing
Computational analysis approaches:
Trajectory analysis to map cellular states related to RARRES1 expression
Cell-cell communication analysis to identify signaling between RARRES1+ cells and immune populations
Integration of single-cell data with bulk tissue outcomes for clinical correlation
These single-cell approaches can help resolve apparently contradictory findings by revealing how RARRES1 functions differently across distinct cellular populations within the same tumor, potentially explaining the complex relationship between RARRES1 expression and clinical outcomes.
RARRES1 is located on chromosome 3q25 and is adjacent to the Latexin (LXN) gene . The gene is known to produce multiple transcript variants encoding distinct isoforms . The protein encoded by RARRES1 is involved in various cellular processes, including the regulation of fatty acid metabolism and the alpha-tubulin tyrosination cycle .
The expression of RARRES1 is upregulated by retinoic acid receptors and tazarotene, a topical retinoid used in the treatment of psoriasis and acne . However, the expression of this gene is found to be downregulated in several cancers, including prostate cancer, due to the methylation of its promoter and CpG island .
RARRES1 has been identified as a tumor suppressor and plays a significant role in metabolic reprogramming of epithelial cells . It regulates fatty acid metabolism by inhibiting the cytoplasmic carboxypeptidase AGBL2, which may influence the alpha-tubulin tyrosination cycle . In epithelial cells, depletion of RARRES1 leads to an increase in lipid synthesis and a switch from aerobic glycolysis to glucose-dependent de novo lipogenesis (DNL) . This metabolic shift provides an advantage to cells during starvation by increasing fatty acid availability for mitochondrial respiration .
RARRES1 is differentially expressed in various metabolic diseases, such as hepatic steatosis, hyperinsulinemia, and obesity . Its expression is also contextually correlated with the expression of fatty acid metabolism genes and fatty acid-regulated transcription factors . The gene’s hypermethylation and subsequent loss of expression have been observed in multiple cancers, making it a potential target for cancer therapy .
The role of RARRES1 in regulating fatty acid metabolism and its tumor suppressor function opens up new avenues for research and therapeutic interventions. Targeting RARRES1 and its associated pathways could provide novel strategies for treating cancers and metabolic diseases with impaired fatty acid metabolism .